1
|
Abdel-Aal RA, Meligy FY, Maghraby N, Sayed N, Mohamed Ashry IES. Comparing levetiracetam and zonisamide effects on rivastigmine anti-Alzheimer's activity in aluminum chloride-induced Alzheimer's-like disease in rats: Impact on α7 nicotinic acetylcholine receptors and amyloid β. Brain Res 2025; 1855:149573. [PMID: 40096940 DOI: 10.1016/j.brainres.2025.149573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 02/02/2025] [Accepted: 03/13/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND AND AIM Alzheimer's disease (AD) is the most progressive form of neurodegenerative disease, which severely impairs cognitive function. The leading class of drugs used to treat AD is acetylcholinesterase inhibitors (AChE-Is) as Rivastigmine (RIVA), partially ameliorate its cognitive symptoms. Since epilepsy is a common comorbidity with AD, we explored the potential that new the antiepileptic drugs; Levetiracetam (LEV) and Zonisamide (ZNS) may possess an additional therapeutic benefit to RIVA in AlCl3-induced AD rat model. MATERIALS AND METHODS AlCl3 was used to provoke AD in rats which were then supplemented with treatment drugs for 2 weeks. Treated groups were: Control, AlCl3, RIVA, LEV, RIVA + LEV, ZNS and RIVA + ZNS. Then, the behavioral tests; passive avoidance (PA), Morris water maze (MWM) and novel object recognition (NOR) were conducted to assess cognitive behavior and memory. The Hippocampal Aβ assembly was thoroughly examined by histopathology and ELISA. α7 Nicotinic ACh receptors' (α7nAChRs) expression was assessed immunohistochemically and by real-time quantitative polymerase chain reaction (qPCR). Caspase 3 expression was also assessed by real-time qPCR in hippocampal tissues. RESULTS AlCl3 administration impaired memory and cognitive functions in rats, augmented hippocampal Aβ deposition, with subsequent neurodegeneration and α7nAChRs down-regulation. LEV, but not ZNS, administration significantly mitigated AlCl3-induced cognitive impairment probably through suppression of amyloid β (Aβ) deposition, enhancement of neurogenesis and α7nAChRs expression. When combined to RIVA, ZNS treatment negatively affected cognition possibly through its impact on hippocampal Aβ and subsequent neuronal damage. CONCLUSION Although our results indicated that neither LEV nor ZNS provided any extra benefit to cognitive enhancements in AD rats receiving rivastigmine, LEV demonstrated positive effects individually while ZNS had negative effects when combined with RIVA. As a result, this study suggests the use of LEV rather than ZNS for managing epilepsy in patients with AD given that Alzheimer's and epilepsy can coexist.
Collapse
Affiliation(s)
- Raafat A Abdel-Aal
- Pharmacology Department, Faculty of Medicine, Assiut University, Assiut 71515, Egypt.
| | - Fatma Y Meligy
- Department of Restorative Dentistry and Basic Medical Sciences, Faculty of Dentistry, University of Petra, Amman 11196, Jordan; Histology and Cell Biology Department, Faculty of Medicine, Assiut University, Assiut 71515, Egypt.
| | - Nashwa Maghraby
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Assiut University, Assiut 71515, Egypt.
| | - Nehal Sayed
- Pharmacology Department, Faculty of Medicine, Assiut University, Assiut 71515, Egypt.
| | | |
Collapse
|
2
|
Altinok FA, Petrella M, Masi A, Borruto AM, Ciccocioppo R, Ozturk Y. Exploring the supraspinal antihyperalgesic effects of levetiracetam in the rat model of chronic constriction injury. Can J Physiol Pharmacol 2025. [PMID: 40245838 DOI: 10.1139/cjpp-2024-0302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
Neuropathic pain severely impacts quality of life and effective treatments are needed. To address this, the present study investigated the antihyperalgesic mechanisms of levetiracetam administered at the supraspinal level, together with its effects on ion channel activities. The ventral posterolateral nucleus of the thalamus was selected as the location for micro-injection. Thermal hyperalgesia and mechanical allodynia were assessed via in vivo experiments using the Hargreave's and e-Von Frey apparatus, respectively. Levetiracetam displayed statistically meaningful time and dose-dependent effects in the chronic constriction injury model, with statistical probability values less than 0.05. It was discovered that the antihyperalgesic effects were more pronounced in mechanical allodynia. Electrophysiological studies conducted through whole-cell patch clamp recordings indicated that levetiracetam tended to activate or increase the permeability of one or more channels for ion flow that are active only at hyperpolarized membrane potentials (-130 to -90 mV), suggesting the potential participation of hyperpolarization-activated cyclic nucleotide-gated, inwardly-rectifying K+, or G protein-gated inwardly-rectifying K+ channels. The findings could guide future drug development studies towards levetiracetam and its derivatives as effective treatments for neuropathic pain.
Collapse
Affiliation(s)
- Feyza Alyu Altinok
- Department of Pharmacology, Faculty of Pharmacy, Anadolu University, 26470 Eskisehir, Turkey
| | - Michele Petrella
- Department of Pharmacological Sciences and Experimental Medicine, University of Camerino, 62032 Camerino, Italy
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, S-581 85, Linköping, Sweden
| | - Alessio Masi
- Department of Neuroscience, Psychology, Drug Research and Child's Health, NEUROFARBA, Università di Firenze, 50139 Firenze, Italy
| | - Anna Maria Borruto
- Department of Pharmacological Sciences and Experimental Medicine, University of Camerino, 62032 Camerino, Italy
- Center for Social and Affective Neuroscience, Department of Biomedical and Clinical Sciences, Linköping University, S-581 85, Linköping, Sweden
| | - Roberto Ciccocioppo
- Department of Pharmacological Sciences and Experimental Medicine, University of Camerino, 62032 Camerino, Italy
| | - Yusuf Ozturk
- Department of Pharmacology, Faculty of Pharmacy, Istanbul Aydin University, 34295 Istanbul, Turkey
| |
Collapse
|
3
|
Hogg JA, Cousin MA. Control of Synaptotagmin-1 Trafficking by SV2A-Mechanism and Consequences for Presynaptic Function and Dysfunction. J Neurochem 2025; 169:e16308. [PMID: 39853744 PMCID: PMC11758464 DOI: 10.1111/jnc.16308] [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: 11/13/2024] [Revised: 12/17/2024] [Accepted: 01/03/2025] [Indexed: 01/26/2025]
Abstract
Synaptic vesicle protein 2A (SV2A) is an abundant synaptic vesicle cargo with an as yet unconfirmed role in presynaptic function. It is also heavily implicated in epilepsy, firstly being the target of the leading anti-seizure medication levetiracetam and secondly with loss of function mutations culminating in human disease. A range of potential presynaptic functions have been proposed for SV2A; however its interaction with the calcium sensor for synchronous neurotransmitter release, synaptotagmin-1 (Syt1), has received particular attention over the past decade. In this review we will assess the evidence that the primary role of SV2A is to control the expression and localisation of Syt1 at the presynapse. This will integrate biochemical, cell biological and physiological studies where the interaction, trafficking and functional output of Syt1 is altered by SV2A. The potential for SV2A-dependent epilepsy to be a result of dysfunctional Syt1 expression and localisation is also discussed. Finally, a series of key open questions will be posed that require resolution before a definitive role for SV2A in Syt1 function in health and disease can be confirmed.
Collapse
Affiliation(s)
- James A. Hogg
- Centre for Discovery Brain Sciences, Hugh Robson Building, George Square, University of EdinburghEdinburghScotlandUK
- Simons Initiative for the Developing Brain, Hugh Robson Building, George SquareUniversity of EdinburghEdinburghScotlandUK
- Muir Maxwell Epilepsy CentreHugh Robson Building, George Square, University of EdinburghEdinburghScotlandUK
| | - Michael A. Cousin
- Centre for Discovery Brain Sciences, Hugh Robson Building, George Square, University of EdinburghEdinburghScotlandUK
- Simons Initiative for the Developing Brain, Hugh Robson Building, George SquareUniversity of EdinburghEdinburghScotlandUK
- Muir Maxwell Epilepsy CentreHugh Robson Building, George Square, University of EdinburghEdinburghScotlandUK
| |
Collapse
|
4
|
Thompson JC, Levis Rabi M, Novoa M, Nash KR, Joly-Amado A. Evaluating the Efficacy of Levetiracetam on Non-Cognitive Symptoms and Pathology in a Tau Mouse Model. Biomedicines 2024; 12:2891. [PMID: 39767797 PMCID: PMC11727630 DOI: 10.3390/biomedicines12122891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 01/16/2025] Open
Abstract
Background/Objectives: Alzheimer's disease (AD) is marked by amyloid-β plaques and hyperphosphorylated tau neurofibrillary tangles (NFTs), leading to cognitive decline and debilitating non-cognitive symptoms. This study aimed to evaluate compounds from four different classes in a short-term (7-day) study using transgenic tau mice to assess their ability to reduce non-cognitive symptoms. The best candidate was then evaluated for longer exposure to assess non-cognitive symptoms, cognition, and pathology. Methods: Tg4510 mice, expressing mutated human tau (P301L), were administered with levetiracetam, methylphenidate, diazepam, and quetiapine for 7 days at 6 months old, when pathology and cognitive deficits are established. Drugs were given in the diet, and non-cognitive symptoms were evaluated using metabolic cages. Levetiracetam was chosen for longer exposure (3 months) in 3-month-old Tg4510 mice and non-transgenic controls to assess behavior and pathology. Results: After 3 months of diet, levetiracetam mildly reduced tau pathology in the hippocampus but did not improve cognition in Tg4510 mice. Interestingly, it influenced appetite, body weight, anxiety-like behavior, and contextual fear memory in non-transgenic animals but not in Tg4510 mice. Conclusions: While levetiracetam has shown benefits in amyloid deposition models, it had limited effects on tau pathology and behavior in an animal model of tau deposition, which is crucial for AD context. The differential effects on non-transgenic versus Tg4510 mice warrant further investigation.
Collapse
Affiliation(s)
| | | | | | | | - Aurelie Joly-Amado
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, 12901 Bruce B Downs Blvd, Tampa, FL 33612, USA; (J.C.T.); (M.L.R.); (M.N.); (K.R.N.)
| |
Collapse
|
5
|
Rao NR, DeGulis O, Nomura T, Lee S, Hark TJ, Dynes JC, Dexter EX, Dulewicz M, Ge J, Upadhyay A, Fornasiero EF, Vassar R, Hanrieder J, Contractor A, Savas JN. Levetiracetam prevents Aβ 42 production through SV2a-dependent modulation of App processing in Alzheimer's disease models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620698. [PMID: 39554163 PMCID: PMC11565754 DOI: 10.1101/2024.10.28.620698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
In Alzheimer's disease (AD), amyloid-beta (Aβ) peptides are produced by proteolytic cleavage of the amyloid precursor protein (APP), which can occur during synaptic vesicle (SV) cycling at presynapses. Precisely how amyloidogenic APP processing may impair presynaptic proteostasis and how to therapeutically target this process remains poorly understood. Using App knock-in mouse models of early Aβ pathology, we found proteins with hampered degradation accumulate at presynaptic sites. At this mild pathological stage, amyloidogenic processing leads to accumulation of Aβ42 inside SVs. To explore if targeting SVs modulates Aβ accumulation, we investigated levetiracetam (Lev), a SV-binding small molecule drug that has shown promise in mitigating AD-related pathologies despite its mechanism of action being unclear. We discovered Lev reduces Aβ42 levels by decreasing amyloidogenic processing of APP in a SV2a-dependent manner. Lev corrects SV protein levels and cycling, which results in increased surface localization of APP, where it favors processing via the non-amyloidogenic pathway. Using metabolic stable isotopes and mass spectrometry we confirmed that Lev prevents the production of Aβ42 in vivo. In transgenic mice with aggressive pathology, electrophysiological and immunofluorescent microscopy analyses revealed that Lev treatment reduces SV cycling and minimizes synapse loss. Finally, we found that human Down syndrome brains with early Aβ pathology, have elevated levels of presynaptic proteins, confirming a comparable presynaptic deficit in human brains. Taken together, we report a mechanism that highlights the therapeutic potential of Lev to modify the early stages of AD and represent a promising strategy to prevent Aβ42 pathology before irreversible damage occurs.
Collapse
Affiliation(s)
- Nalini R. Rao
- Department of Neurology, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - Olivia DeGulis
- Department of Neurology, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - Toshihiro Nomura
- Department of Neuroscience, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - SeungEun Lee
- Department of Neurology, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - Timothy J. Hark
- Department of Neurology, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - Justin C. Dynes
- Department of Neurology, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - Emily X. Dexter
- Department of Neurology, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - Maciej Dulewicz
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg; Mölndal, Sweden
| | - Junyue Ge
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg; Mölndal, Sweden
| | - Arun Upadhyay
- Department of Neurology, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - Eugenio F. Fornasiero
- Department of Neuro- and Sensory Physiology, University Medical Center Göttingen, Göttingen, Germany
| | - Robert Vassar
- Department of Neurology, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - Jörg Hanrieder
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, University of Gothenburg; Mölndal, Sweden
- Department of Neurodegenerative disease, Queen Square Institute of Neurology, University College London, London, UK
| | - Anis Contractor
- Department of Neuroscience, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| | - Jeffrey N. Savas
- Department of Neurology, Northwestern University Feinberg School of Medicine; Chicago, IL USA
| |
Collapse
|
6
|
Czapińska-Ciepiela EK, Łuszczki J, Czapiński P, Czuczwar SJ, Lasoń W. Presynaptic antiseizure medications - basic mechanisms and clues for their rational combinations. Pharmacol Rep 2024; 76:623-643. [PMID: 38776036 PMCID: PMC11294404 DOI: 10.1007/s43440-024-00603-7] [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: 02/22/2024] [Revised: 05/09/2024] [Accepted: 05/11/2024] [Indexed: 08/02/2024]
Abstract
Among clinically highly efficient antiseizure medications (ASMs) there are modifiers of the presynaptic release machinery. Of them, levetiracetam and brivaracetam show a high affinity to the synaptic vesicle protein type 2 A (SV2A), whereas pregabalin and gabapentin are selective ligands for the α2δ1 subunits of the voltage-gated calcium channels. In this paper, we present recent progress in understanding the significance of presynaptic release machinery in the neurochemical mechanisms of epilepsy and ASMs. Furthermore, we discuss whether the knowledge of the basic mechanisms of the presynaptically acting ASMs might help establish a rational polytherapy for drug-resistant epilepsy.
Collapse
Affiliation(s)
| | - Jarogniew Łuszczki
- Department of Occupational Medicine, Medical University of Lublin, 20-090, Lublin, Poland
| | - Piotr Czapiński
- Epilepsy and Migraine Treatment Center, 31-209, Kraków, Poland
| | - Stanisław J Czuczwar
- Department of Pathophysiology, Medical University of Lublin, 20-090, Lublin, Poland
| | - Władysław Lasoń
- Maj Institute of Pharmacology, Department of Experimental Neuroendocrinology, Polish Academy of Sciences, 31-343, Kraków, Poland.
| |
Collapse
|
7
|
Wu PP, Cao BR, Tian FY, Gao ZB. Development of SV2A Ligands for Epilepsy Treatment: A Review of Levetiracetam, Brivaracetam, and Padsevonil. Neurosci Bull 2024; 40:594-608. [PMID: 37897555 PMCID: PMC11127901 DOI: 10.1007/s12264-023-01138-2] [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: 03/27/2023] [Accepted: 08/16/2023] [Indexed: 10/30/2023] Open
Abstract
Epilepsy is a common neurological disorder that is primarily treated with antiseizure medications (ASMs). Although dozens of ASMs are available in the clinic, approximately 30% of epileptic patients have medically refractory seizures; other limitations in most traditional ASMs include poor tolerability and drug-drug interactions. Therefore, there is an urgent need to develop alternative ASMs. Levetiracetam (LEV) is a first-line ASM that is well tolerated, has promising efficacy, and has little drug-drug interaction. Although it is widely accepted that LEV acts through a unique therapeutic target synaptic vesicle protein (SV) 2A, the molecular basis of its action remains unknown. Even so, the next-generation SV2A ligands against epilepsy based on the structure of LEV have achieved clinical success. This review highlights the research and development (R&D) process of LEV and its analogs, brivaracetam and padsevonil, to provide ideas and experience for the R&D of novel ASMs.
Collapse
Affiliation(s)
- Peng-Peng Wu
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Bi-Rong Cao
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fu-Yun Tian
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
| | - Zhao-Bing Gao
- Center for Neurological and Psychiatric Research and Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan, 528400, China.
| |
Collapse
|
8
|
Williams NC, Morgan LA, Friedman J, Siegler J. Ketamine Efficacy for Management of Status Epilepticus: Considerations for Prehospital Clinicians. Air Med J 2024; 43:84-89. [PMID: 38490790 DOI: 10.1016/j.amj.2023.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/10/2023] [Accepted: 09/21/2023] [Indexed: 03/17/2024]
Abstract
Current first-line therapies for seizure management recommend benzodiazepines, which target gamma-aminobutyric acid type A channels to stop the seizure activity. However, seizures may be refractory to traditional first-line therapies, transitioning into status epilepticus and becoming resistant to gamma-aminobutyric acid type A augmenting drugs. Although there are other antiseizure medications available for clinicians to use in the intensive care unit, these options can be less readily available outside of the intensive care unit and entirely absent in the prehospital setting. Instead, patients frequently receive multiple doses of first-line agents with increased risk of hemodynamic or airway collapse. Ketamine is readily available in the prehospital setting and emergency department, has well-established antiseizure effects with a favorable safety profile, and is a drug often used for several other indications. This article aimed to explore the utilization of ketamine for seizure management in the prehospital setting, reviewing seizure pathophysiology, established treatment mechanisms of action and pharmacokinetics, and potential benefits of early ketamine use in status epilepticus.
Collapse
Affiliation(s)
| | - Lindsey A Morgan
- Department of Neurology, Division of Pediatric Neurology, University of Washington, Seattle, WA; Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, WA
| | | | - Jeffrey Siegler
- Department of Emergency Medicine, Washington University School of Medicine, St Louis, MO
| |
Collapse
|
9
|
Diaz-Villegas V, Pichardo-Macías LA, Juárez-Méndez S, Ignacio-Mejía I, Cárdenas-Rodríguez N, Vargas-Hernández MA, Mendoza-Torreblanca JG, Zamudio SR. Changes in the Dentate Gyrus Gene Expression Profile Induced by Levetiracetam Treatment in Rats with Mesial Temporal Lobe Epilepsy. Int J Mol Sci 2024; 25:1690. [PMID: 38338984 PMCID: PMC10855401 DOI: 10.3390/ijms25031690] [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: 12/16/2023] [Revised: 01/24/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
Temporal lobe epilepsy (TLE) is one of the most common forms of focal epilepsy. Levetiracetam (LEV) is an antiepileptic drug whose mechanism of action at the genetic level has not been fully described. Therefore, the aim of the present work was to evaluate the relevant gene expression changes in the dentate gyrus (DG) of LEV-treated rats with pilocarpine-induced TLE. Whole-transcriptome microarrays were used to obtain the differential genetic profiles of control (CTRL), epileptic (EPI), and EPI rats treated for one week with LEV (EPI + LEV). Quantitative RT-qPCR was used to evaluate the RNA levels of the genes of interest. According to the results of the EPI vs. CTRL analysis, 685 genes were differentially expressed, 355 of which were underexpressed and 330 of which were overexpressed. According to the analysis of the EPI + LEV vs. EPI groups, 675 genes were differentially expressed, 477 of which were downregulated and 198 of which were upregulated. A total of 94 genes whose expression was altered by epilepsy and modified by LEV were identified. The RT-qPCR confirmed that LEV treatment reversed the increased expression of Hgf mRNA and decreased the expression of the Efcab1, Adam8, Slc24a1, and Serpinb1a genes in the DG. These results indicate that LEV could be involved in nonclassical mechanisms involved in Ca2+ homeostasis and the regulation of the mTOR pathway through Efcab1, Hgf, SLC24a1, Adam8, and Serpinb1a, contributing to reduced hyperexcitability in TLE patients.
Collapse
Affiliation(s)
- Veronica Diaz-Villegas
- Departamento de Fisiología, Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Mexico City 07738, Mexico; (V.D.-V.); (L.A.P.-M.)
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Mexico City 04530, Mexico;
| | - Luz Adriana Pichardo-Macías
- Departamento de Fisiología, Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Mexico City 07738, Mexico; (V.D.-V.); (L.A.P.-M.)
| | - Sergio Juárez-Méndez
- Laboratorio de Oncología Experimental, Instituto Nacional de Pediatría, Secretaría de Salud, Mexico City 04530, Mexico;
| | - Iván Ignacio-Mejía
- Laboratorio de Medicina Traslacional, Escuela Militar de Graduados de Sanidad, Universidad del Ejército y Fuerza Aérea, Mexico City 11200, Mexico;
| | - Noemí Cárdenas-Rodríguez
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Mexico City 04530, Mexico;
| | - Marco Antonio Vargas-Hernández
- Subdirección de Investigación, Escuela Militar de Graduados de Sanidad, Universidad del Ejército y Fuerza Aérea, Mexico City 11200, Mexico;
| | | | - Sergio R. Zamudio
- Departamento de Fisiología, Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Mexico City 07738, Mexico; (V.D.-V.); (L.A.P.-M.)
| |
Collapse
|
10
|
Cavichioli AM, Santos-Silva T, Grace AA, Guimarães FS, Gomes FV. Levetiracetam Attenuates Adolescent Stress-induced Behavioral and Electrophysiological Changes Associated With Schizophrenia in Adult Rats. Schizophr Bull 2023; 49:68-77. [PMID: 35988039 PMCID: PMC9810001 DOI: 10.1093/schbul/sbac106] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
BACKGROUND AND HYPOTHESIS Stress during adolescence is a major risk factor for schizophrenia. We have found previously in rats that adolescent stress caused, in adulthood, behavioral changes and enhanced ventral tegmental area (VTA) dopamine system activity, which were associated with dysregulation of the excitatory-inhibitory (E/I) balance in the ventral hippocampus (vHip). Levetiracetam, an anticonvulsant drug, regulates the release of neurotransmitters, including glutamate, via SV2A inhibition. It also modulates parvalbumin interneuron activity via Kv3.1 channels. Therefore, levetiracetam could ameliorate deficits in the E/I balance. We tested whether levetiracetam attenuate the adolescent stress-induced behavioral changes, vHip hyperactivity, and enhanced VTA dopamine system activity in adult rats. STUDY DESIGN Male Sprague-Dawley rats were subjected to a combination of daily footshock (postnatal day [PD] 31-40), and three 1 h-restraint stress sessions (at PD31, 32, and 40). In adulthood (PD62), animals were tested for anxiety responses (elevated plus-maze and light-dark box), social interaction, and cognitive function (novel object recognition test). The activity of vHip pyramidal neurons and VTA dopamine neurons was also recorded. STUDY RESULTS Adolescent stress produced anxiety-like responses and impaired sociability and cognitive function. Levetiracetam (10 mg/kg) reversed these changes. Levetiracetam also reversed the increased VTA dopamine neuron population activity and the enhanced firing rate of vHip pyramidal neurons induced by adolescent stress. CONCLUSIONS These findings suggest that levetiracetam attenuates the adverse outcomes associated with schizophrenia caused by stress during adolescence.
Collapse
Affiliation(s)
- Andreza M Cavichioli
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Thamyris Santos-Silva
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Anthony A Grace
- Departments of Neuroscience, Psychiatry, and Psychology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Francisco S Guimarães
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Felipe V Gomes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
11
|
Castro PA, Pinto-Borguero I, Yévenes GE, Moraga-Cid G, Fuentealba J. Antiseizure medication in early nervous system development. Ion channels and synaptic proteins as principal targets. Front Pharmacol 2022; 13:948412. [PMID: 36313347 PMCID: PMC9614143 DOI: 10.3389/fphar.2022.948412] [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: 05/19/2022] [Accepted: 09/05/2022] [Indexed: 12/04/2022] Open
Abstract
The main strategy for the treatment of epilepsy is the use of pharmacological agents known as antiseizure medication (ASM). These drugs control the seizure onset and improves the life expectancy and quality of life of patients. Several ASMs are contraindicated during pregnancy, due to a potential teratogen risk. For this reason, the pharmacological treatments of the pregnant Women with Epilepsy (WWE) need comprehensive analyses to reduce fetal risk during the first trimester of pregnancy. The mechanisms by which ASM are teratogens are still under study and scientists in the field, propose different hypotheses. One of them, which will be addressed in this review, corresponds to the potential alteration of ASM on ion channels and proteins involved in relevant signaling and cellular responses (i.e., migration, differentiation) during embryonic development. The actual information related to the action of ASM and its possible targets it is poorly understood. In this review, we will focus on describing the eventual presence of some ion channels and synaptic proteins of the neurotransmitter signaling pathways present during early neural development, which could potentially interacting as targets of ASM. This information leads to elucidate whether these drugs would have the ability to affect critical signaling during periods of neural development that in turn could explain the fetal malformations observed by the use of ASM during pregnancy.
Collapse
Affiliation(s)
- Patricio A. Castro
- Laboratory of Physiology and Pharmacology for Neural Development, LAND, Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
- *Correspondence: Patricio A. Castro,
| | - Ingrid Pinto-Borguero
- Laboratory of Physiology and Pharmacology for Neural Development, LAND, Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Gonzalo E. Yévenes
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Gustavo Moraga-Cid
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| | - Jorge Fuentealba
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Universidad de Concepción, Concepción, Chile
| |
Collapse
|
12
|
Rossi R, Arjmand S, Bærentzen SL, Gjedde A, Landau AM. Synaptic Vesicle Glycoprotein 2A: Features and Functions. Front Neurosci 2022; 16:864514. [PMID: 35573314 PMCID: PMC9096842 DOI: 10.3389/fnins.2022.864514] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 04/05/2022] [Indexed: 01/05/2023] Open
Abstract
In recent years, the field of neuroimaging dramatically moved forward by means of the expeditious development of specific radioligands of novel targets. Among these targets, the synaptic vesicle glycoprotein 2A (SV2A) is a transmembrane protein of synaptic vesicles, present in all synaptic terminals, irrespective of neurotransmitter content. It is involved in key functions of neurons, focused on the regulation of neurotransmitter release. The ubiquitous expression in gray matter regions of the brain is the basis of its candidacy as a marker of synaptic density. Following the development of molecules derived from the structure of the anti-epileptic drug levetiracetam, which selectively binds to SV2A, several radiolabeled markers have been synthetized to allow the study of SV2A distribution with positron emission tomography (PET). These radioligands permit the evaluation of in vivo changes of SV2A distribution held to be a potential measure of synaptic density in physiological and pathological conditions. The use of SV2A as a biomarker of synaptic density raises important questions. Despite numerous studies over the last decades, the biological function and the expressional properties of SV2A remain poorly understood. Some functions of SV2A were claimed, but have not been fully elucidated. While the expression of SV2A is ubiquitous, stronger associations between SV2A and Υ amino butyric acid (GABA)-ergic rather than glutamatergic synapses were observed in some brain structures. A further issue is the unclear interaction between SV2A and its tracers, which reflects a need to clarify what really is detected with neuroimaging tools. Here, we summarize the current knowledge of the SV2A protein and we discuss uncertain aspects of SV2A biology and physiology. As SV2A expression is ubiquitous, but likely more strongly related to a certain type of neurotransmission in particular circumstances, a more extensive knowledge of the protein would greatly facilitate the analysis and interpretation of neuroimaging results by allowing the evaluation not only of an increase or decrease of the protein level, but also of the type of neurotransmission involved.
Collapse
Affiliation(s)
- Rachele Rossi
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Shokouh Arjmand
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Simone Larsen Bærentzen
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Albert Gjedde
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Neuroscience, University of Copenhagen, Copenhagen, Denmark.,Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Anne M Landau
- Translational Neuropsychiatry Unit, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.,Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
13
|
Contreras-García IJ, Cárdenas-Rodríguez N, Romo-Mancillas A, Bandala C, Zamudio SR, Gómez-Manzo S, Hernández-Ochoa B, Mendoza-Torreblanca JG, Pichardo-Macías LA. Levetiracetam Mechanisms of Action: From Molecules to Systems. Pharmaceuticals (Basel) 2022; 15:475. [PMID: 35455472 PMCID: PMC9030752 DOI: 10.3390/ph15040475] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 04/06/2022] [Accepted: 04/06/2022] [Indexed: 11/16/2022] Open
Abstract
Epilepsy is a chronic disease that affects millions of people worldwide. Antiepileptic drugs (AEDs) are used to control seizures. Even though parts of their mechanisms of action are known, there are still components that need to be studied. Therefore, the search for novel drugs, new molecular targets, and a better understanding of the mechanisms of action of existing drugs is still crucial. Levetiracetam (LEV) is an AED that has been shown to be effective in seizure control and is well-tolerable, with a novel mechanism of action through an interaction with the synaptic vesicle protein 2A (SV2A). Moreover, LEV has other molecular targets that involve calcium homeostasis, the GABAergic system, and AMPA receptors among others, that might be integrated into a single mechanism of action that could explain the antiepileptogenic, anti-inflammatory, neuroprotective, and antioxidant properties of LEV. This puts it as a possible multitarget drug with clinical applications other than for epilepsy. According to the above, the objective of this work was to carry out a comprehensive and integrative review of LEV in relation to its clinical uses, structural properties, therapeutical targets, and different molecular, genetic, and systemic action mechanisms in order to consider LEV as a candidate for drug repurposing.
Collapse
Affiliation(s)
| | - Noemí Cárdenas-Rodríguez
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico;
| | - Antonio Romo-Mancillas
- Laboratorio de Diseño Asistido por Computadora y Síntesis de Fármacos, Facultad de Química, Universidad Autónoma de Querétaro, Centro Universitario, Querétaro 76010, Mexico;
| | - Cindy Bandala
- Neurociencia Básica, Instituto Nacional de Rehabilitación LGII, Secretaría de Salud, Ciudad de México 14389, Mexico;
- Escuela Superior de Medicina, Instituto Politécnico Nacional, Ciudad de México 11340, Mexico
| | - Sergio R. Zamudio
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 07738, Mexico;
| | - Saúl Gómez-Manzo
- Laboratorio de Bioquímica Genética, Instituto Nacional de Pediatría, Secretaría de Salud, Ciudad de México 04530, Mexico;
| | - Beatriz Hernández-Ochoa
- Laboratorio de Inmunoquímica, Hospital Infantil de México Federico Gómez, Secretaría de Salud, Ciudad de México 06720, Mexico;
| | | | - Luz Adriana Pichardo-Macías
- Departamento de Fisiología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México 07738, Mexico;
| |
Collapse
|
14
|
Efficacy of levetiracetam in STXBP1 encephalopathy with different phenotypic and genetic spectra. Seizure 2021; 95:64-74. [PMID: 35007884 DOI: 10.1016/j.seizure.2021.12.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 12/08/2021] [Accepted: 12/15/2021] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE Syntaxin binding protein 1 (STXBP1) plays an important role in the release of synaptic vesicles. STXBP1-related encephalopathy is a brain dysfunction caused by STXBP1 variation. Levetiracetam (LEV) exerts antiepileptic effects by binding to synaptic vesicle protein 2A (SV2A). This study aimed to analyze the prognosis of LEV treatment of STXBP1 encephalopathy (STXBP1-E) and the correlation among genotype, phenotype, and LEV efficacy. METHODS Patients with pathogenic STXBP1 variants were collected from multiple centers, and their clinical history, video electroencephalogram (vEEG) characteristics, imaging examination data, and anti-seizure medication (ASM) history were systematically analyzed. The ASMs related to the prognosis were explored. RESULTS Forty patients with STXBP1-E were enrolled in this study. The detailed ASM usage of 37 patients was recorded without intervening in ASM selection. At the endpoint of six months treatment, the results of Fisher's exact test showed that in all ASMs, LEV affected the prognosis of patients with STXBP1-E. LEV was effective in improving the partial remission rate but did not achieve seizure freedom. However, LEV monotherapy could achieve seizure freedom in patients with other early-onset epileptic and encephalopathy. For refractory West syndrome (WS) or Ohtahara syndrome (OS), LEV combined with other ASMs could improve the seizure remission rate. CONCLUSION LEV increased the seizure reduction rate and improved the vEEG characteristics in patients with STXBP1-E, but not seizure freedom. LEV combined with other ASMs could increase the seizure reduction rate, especially for refractory WS or OS. Thus, LEV could be considered after identifying the pathogenicity of STXBP1 variants.
Collapse
|
15
|
Li L, Chen GD, Salvi R. Effect of antiepileptic drug levetiracetam on cochlear function. Hear Res 2021; 415:108396. [PMID: 34903423 DOI: 10.1016/j.heares.2021.108396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Levetiracetam (LEV, 5-100 mg/kg) has been shown to prevent audiogenic seizures in a dose-dependent manner. This chemical is known to bind to synaptic vesicle protein 2A and inhibit l-type calcium channels, affecting neurotransmitter release. We hypothesize that the drug prevents audiogenic seizures partially by affecting cochlear neural response. METHODS To test this hypothesis, rats were given 1000, 500, 50, or 0 mg/kg (saline control) LEV-injection. Distortion product otoacoustic emissions (DPOAE), reflecting outer hair cell (OHC) function, and cochlear compound action potentials (CAP), reflecting cochlear neural output, were recorded and compared pre- and post-LEV. RESULTS 1000 mg/kg LEV-injection did not significantly affect DPOAE. The high dose LEV-injection, however, significantly reduced CAP amplitude resulting threshold shift (TS), prolonged CAP latency, and enhanced CAP forward masking. CAP latency and forward masking were significantly affected at the 500 mg/kg dose, but CAP-TS remained unchanged after LEV-injection. Interestingly, CAP latency wassignificantly prolonged, at least at the low stimulation levels, although the amplitude of CAP remained constant after a clinical dose of LEV-injection (50 mg/kg). DISCUSSION Since the clinical dose of LEV-injection does not reduce CAP amplitude, the reduction of cochlear neural output is unlikely to be the underlying mechanism of LEV in the treatment of audiogenic seizure. The delayed cochlear neural response may be partially related to the prevention of audiogenic seizure. However, neuropharmacological changes in the central nervous system must play a major role in the treatment of audiogenic seizure, as it does in the treatment of focal epilepsy.
Collapse
Affiliation(s)
- Li Li
- Center for Hearing and Deafness, SUNY at Buffalo, Buffalo, NY 14214, USA
| | - Guang-Di Chen
- Center for Hearing and Deafness, SUNY at Buffalo, Buffalo, NY 14214, USA.
| | - Richard Salvi
- Center for Hearing and Deafness, SUNY at Buffalo, Buffalo, NY 14214, USA
| |
Collapse
|
16
|
Ni R. Positron Emission Tomography in Animal Models of Alzheimer's Disease Amyloidosis: Translational Implications. Pharmaceuticals (Basel) 2021; 14:1179. [PMID: 34832961 PMCID: PMC8623863 DOI: 10.3390/ph14111179] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 11/13/2021] [Accepted: 11/15/2021] [Indexed: 12/30/2022] Open
Abstract
Animal models of Alzheimer's disease amyloidosis that recapitulate cerebral amyloid-beta pathology have been widely used in preclinical research and have greatly enabled the mechanistic understanding of Alzheimer's disease and the development of therapeutics. Comprehensive deep phenotyping of the pathophysiological and biochemical features in these animal models is essential. Recent advances in positron emission tomography have allowed the non-invasive visualization of the alterations in the brain of animal models and in patients with Alzheimer's disease. These tools have facilitated our understanding of disease mechanisms and provided longitudinal monitoring of treatment effects in animal models of Alzheimer's disease amyloidosis. In this review, we focus on recent positron emission tomography studies of cerebral amyloid-beta accumulation, hypoglucose metabolism, synaptic and neurotransmitter receptor deficits (cholinergic and glutamatergic system), blood-brain barrier impairment, and neuroinflammation (microgliosis and astrocytosis) in animal models of Alzheimer's disease amyloidosis. We further propose the emerging targets and tracers for reflecting the pathophysiological changes and discuss outstanding challenges in disease animal models and future outlook in the on-chip characterization of imaging biomarkers towards clinical translation.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, ETH & University of Zurich, 8093 Zurich, Switzerland;
- Institute for Regenerative Medicine, University of Zurich, 8952 Zurich, Switzerland
| |
Collapse
|
17
|
Dell'Aquila J, Soti V. Treating Status Epilepticus: Phenytoin Versus Levetiracetam. Cureus 2021; 13:e18515. [PMID: 34659919 PMCID: PMC8492029 DOI: 10.7759/cureus.18515] [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] [Accepted: 10/05/2021] [Indexed: 11/25/2022] Open
Abstract
For decades, phenytoin has been the drug of choice for the treatment of epilepsy but also the second-line treatment for status epilepticus (SE). However, newer antiepileptic drugs (AEDs) have emerged as safer alternatives for the suppression of seizures. Consequently, phenytoin has recently fallen under scrutiny in the research world, prompting many studies to compare its efficacy to these other drugs, most notably levetiracetam. Levetiracetam is a second-generation AED, which is gaining wide clinical use as the second-line agent in treating SE patients. This review focuses on several clinical studies that have directly compared the effectiveness of phenytoin and levetiracetam in suppressing SE seizure activity. Additionally, this review highlights several advantages of using levetiracetam over phenytoin in this clinical context.
Collapse
Affiliation(s)
| | - Varun Soti
- Pharmacology and Therapeutics, Lake Erie College of Osteopathic Medicine, Elmira, USA
| |
Collapse
|
18
|
Contreras-García IJ, Gómez-Lira G, Phillips-Farfán BV, Pichardo-Macías LA, García-Cruz ME, Chávez-Pacheco JL, Mendoza-Torreblanca JG. Synaptic Vesicle Protein 2A Expression in Glutamatergic Terminals Is Associated with the Response to Levetiracetam Treatment. Brain Sci 2021; 11:brainsci11050531. [PMID: 33922424 PMCID: PMC8145097 DOI: 10.3390/brainsci11050531] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/24/2021] [Accepted: 03/24/2021] [Indexed: 12/05/2022] Open
Abstract
Synaptic vesicle protein 2A (SV2A), the target of the antiepileptic drug levetiracetam (LEV), is expressed ubiquitously in all synaptic terminals. Its levels decrease in patients and animal models of epilepsy. Thus, changes in SV2A expression could be a critical factor in the response to LEV. Epilepsy is characterized by an imbalance between excitation and inhibition, hence SV2A levels in particular terminals could also influence the LEV response. SV2A expression was analyzed in the epileptic hippocampus of rats which responded or not to LEV, to clarify if changes in SV2A alone or together with glutamatergic or GABAergic markers may predict LEV resistance. Wistar rats were administered saline (control) or pilocarpine to induce epilepsy. These groups were subdivided into untreated or LEV-treated groups. All epileptic rats were video-monitored to assess their number of seizures. Epileptic rats with an important seizure reduction (>50%) were classified as responders. SV2A, vesicular γ-aminobutyric acid transporter and vesicular glutamate transporter (VGLUT) expression were assessed by immunostaining. SV2A expression was not modified during epilepsy. However, responders showed ≈55% SV2A-VGLUT co-expression in comparison with the non-responder group (≈40%). Thus, SV2A expression in glutamatergic terminals may be important for the response to LEV treatment.
Collapse
Affiliation(s)
- Itzel Jatziri Contreras-García
- Área de Neurociencias, Biología de la Reproducción, Unidad Iztapalapa, Universidad Autónoma Metropolitana, Ciudad de México 09340, Mexico;
| | - Gisela Gómez-Lira
- Departamento de Farmacobiología, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México 14330, Mexico;
| | - Bryan Víctor Phillips-Farfán
- Laboratorio de Nutrición Experimental, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico;
| | - Luz Adriana Pichardo-Macías
- Departamento de Fisiología, Instituto Politécnico Nacional, Escuela Nacional de Ciencias Biológicas, Ciudad de México 07738, Mexico;
| | - Mercedes Edna García-Cruz
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico;
| | - Juan Luis Chávez-Pacheco
- Laboratorio de Farmacología, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico;
| | - Julieta G. Mendoza-Torreblanca
- Laboratorio de Neurociencias, Subdirección de Medicina Experimental, Instituto Nacional de Pediatría, Ciudad de México 04530, Mexico;
- Correspondence: ; Tel.: +52-55-840900 (ext. 1425) or +52-55-21836345
| |
Collapse
|
19
|
Cawthorne C, Maguire P, Mercier J, Sciberras D, Serdons K, Bormans G, de Hoon J, Van Laere K, Koole M. Human biodistribution and dosimetry of [ 11C]-UCB-J, a PET radiotracer for imaging synaptic density. EJNMMI Phys 2021; 8:37. [PMID: 33891195 PMCID: PMC8065069 DOI: 10.1186/s40658-021-00384-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 04/13/2021] [Indexed: 11/10/2022] Open
Abstract
RATIONALE [11C]-UCB-J is an emerging tool for the noninvasive measurement of synaptic vesicle density in vivo. Here, we report human biodistribution and dosimetry estimates derived from sequential whole-body PET using two versions of the OLINDA dosimetry program. METHODS Sequential whole-body PET scans were performed in 3 healthy subjects for 2 h after injection of 254 ± 77 MBq [11C]-UCB-J. Volumes of interest were drawn over relevant source organs to generate time-activity curves and calculate time-integrated activity coefficients, with effective dose coefficients calculated using OLINDA 2.1 and compared to values derived from OLINDA 1.1 and those recently reported in the literature. RESULTS [11C]-UCB-J administration was safe and showed mixed renal and hepatobiliary clearance, with largest organ absorbed dose coefficients for the urinary bladder wall and small intestine (21.7 and 23.5 μGy/MBq, respectively). The average (±SD) effective dose coefficient was 5.4 ± 0.7 and 5.1 ± 0.8 μSv/MBq for OLINDA versions 1.1 and 2.1 respectively. Doses were lower than previously reported in the literature using either software version. CONCLUSIONS A single IV administration of 370 MBq [11C]-UCB-J corresponds to an effective dose of less than 2.0 mSv, enabling multiple PET examinations to be carried out in the same subject. TRIAL REGISTRATION EudraCT number: 2016-001190-32. Registered 16 March 2016, no URL available for phase 1 trials.
Collapse
Affiliation(s)
- Christopher Cawthorne
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | | | | | | | - Kim Serdons
- Division of Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmaceutical Research, KU Leuven, Leuven, Belgium
| | - Jan de Hoon
- Center for Clinical Pharmacology, University Hospitals Leuven, Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.,Division of Nuclear Medicine, University Hospitals Leuven, Leuven, Belgium
| | - Michel Koole
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
20
|
[Efficacy and safety of levetiracetam versus phenytoin as second-line drugs for the treatment of children with convulsive status epilepticus: a Meta analysis]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2021; 23. [PMID: 33840407 PMCID: PMC8050550 DOI: 10.7499/j.issn.1008-8830.2010066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
OBJECTIVE To systematically evaluate the efficacy and safety of levetiracetam (LEV) versus phenytoin (PHT) as second-line drugs for the treatment of convulsive status epilepticus (CSE) in children. METHODS English and Chinese electronic databases were searched for the randomized controlled trials comparing the efficacy and safety of LEV and PHT as second-line drugs for the treatment of childhood CSE. RevMan 5.3 software was used for data analysis. RESULTS Seven studies with 1 434 children were included. The Meta analysis showed that compared with the PHT group, the LEV group achieved a significantly higher control rate of CSE (RR=1.12, 95%CI:1.00-1.24, P=0.05), but there was no significant difference between the two groups in the recurrence rate of epilepsy within 24 hours (RR=0.82, 95%CI:0.22-3.11, P=0.77) and the rate of further antiepileptic drug therapy (RR=0.97, 95%CI:0.64-1.45, P=0.87). There was no significant difference in the incidence rate of adverse events between the two groups (RR=0.77, 95%CI:0.55-1.09, P=0.15). CONCLUSIONS LEV has a better clinical effect than PHT in the treatment of children with CSE and does not increase the incidence rate of adverse events.
Collapse
|
21
|
Calame DG, Herman I, Riviello JJ. A de novo heterozygous rare variant in SV2A causes epilepsy and levetiracetam-induced drug-resistant status epilepticus. Epilepsy Behav Rep 2021; 15:100425. [PMID: 33554103 PMCID: PMC7844124 DOI: 10.1016/j.ebr.2020.100425] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/23/2020] [Accepted: 12/28/2020] [Indexed: 11/01/2022] Open
Abstract
SV2A encodes a neuronal synaptic vesicle glycoprotein essential for neurotransmitter release. Altered SV2A function leads to epilepsy in animal models, yet only two reports of human variants have linked SV2A to syndromic drug-resistant epileptic encephalopathies and epilepsy. SV2A is also the binding site for the commonly used antiseizure medication levetiracetam (LEV). However, information about how rare SV2A variants influence LEV response is lacking. Here, we report a two-year-old child with new-onset epilepsy found to have a de novo heterozygous rare variant in SV2A (NM_014849.5:c.1978G>A;p.Gly660Arg) who developed refractory status epilepticus after escalation of LEV treatment for initial baseline seizure control. This report provides additional evidence that monoallelic pathogenic SV2A variants cause epilepsy and that genetic variation in SV2A could lead to paradoxical seizure worsening when treated with LEV.
Collapse
Affiliation(s)
- Daniel G Calame
- Department of Pediatrics, Section of Pediatric Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, TX 77030, United States
| | - Isabella Herman
- Department of Pediatrics, Section of Pediatric Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, TX 77030, United States
| | - James J Riviello
- Department of Pediatrics, Section of Pediatric Neurology and Developmental Neuroscience, Baylor College of Medicine, Houston, TX 77030, United States
| |
Collapse
|
22
|
Salinas-Marín R, Villanueva-Cabello TM, Martínez-Duncker I. Biology of Proteoglycans and Associated Glycosaminoglycans. COMPREHENSIVE GLYCOSCIENCE 2021:63-102. [DOI: 10.1016/b978-0-12-819475-1.00065-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
23
|
Stout K, Dunn A, Hoffman C, Miller GW. The Synaptic Vesicle Glycoprotein 2: Structure, Function, and Disease Relevance. ACS Chem Neurosci 2019; 10:3927-3938. [PMID: 31394034 PMCID: PMC11562936 DOI: 10.1021/acschemneuro.9b00351] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The synaptic vesicle glycoprotein 2 (SV2) family is comprised of three paralogues: SV2A, SV2B, and SV2C. In vertebrates, SV2s are 12-transmembrane proteins present on every secretory vesicle, including synaptic vesicles, and are critical to neurotransmission. Structural and functional studies suggest that SV2 proteins may play several roles to promote proper vesicular function. Among these roles are their potential to stabilize the transmitter content of vesicles, to maintain and orient the releasable pool of vesicles, and to regulate vesicular calcium sensitivity to ensure efficient, coordinated release of the transmitter. The SV2 family is highly relevant to human health in a number of ways. First, SV2A plays a role in neuronal excitability and as such is the specific target for the antiepileptic drug levetiracetam. SV2 proteins also act as the target by which potent neurotoxins, particularly botulinum, gain access to neurons and exert their toxicity. Both SV2B and SV2C are increasingly implicated in diseases such as Alzheimer's disease and Parkinson's disease. Interestingly, despite decades of intensive research, their exact function remains elusive. Thus, SV2 proteins are intriguing in their potentially diverse roles within the presynaptic terminal, and several recent developments have enhanced our understanding and appreciation of the protein family. Here, we review the structure and function of SV2 proteins as well as their relevance to disease and therapeutic development.
Collapse
Affiliation(s)
- Kristen Stout
- Department of Physiology, Northwestern University, Feinberg School of Medicine, Chicago, Illinois, United States
| | - Amy Dunn
- The Jackson Laboratory, Bar Harbor, Maine, United States
| | - Carlie Hoffman
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, Georgia, United States
| | - Gary W. Miller
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, New York, United States
| |
Collapse
|
24
|
Ciruelas K, Marcotulli D, Bajjalieh SM. Synaptic vesicle protein 2: A multi-faceted regulator of secretion. Semin Cell Dev Biol 2019; 95:130-141. [PMID: 30826548 DOI: 10.1016/j.semcdb.2019.02.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 01/11/2019] [Accepted: 02/21/2019] [Indexed: 01/01/2023]
Abstract
Synaptic Vesicle Protein 2 (SV2) comprises a recently evolved family of proteins unique to secretory vesicles that undergo calcium-regulated exocytosis. In this review we consider SV2s' structural features, evolution, and function and discuss its therapeutic potential as the receptors for an expanding class of drugs used to treat epilepsy and cognitive decline.
Collapse
Affiliation(s)
- Kristine Ciruelas
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Daniele Marcotulli
- Department of Pharmacology, University of Washington, Seattle, WA, United States
| | - Sandra M Bajjalieh
- Department of Pharmacology, University of Washington, Seattle, WA, United States.
| |
Collapse
|
25
|
Differential expression of SV2A in hippocampal glutamatergic and GABAergic terminals during postnatal development. Brain Res 2019; 1715:73-83. [PMID: 30905653 DOI: 10.1016/j.brainres.2019.03.021] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Revised: 03/08/2019] [Accepted: 03/20/2019] [Indexed: 01/15/2023]
Abstract
The function of synaptic vesicle protein 2A (SV2A) has not been clearly identified, although it has an essential role in normal neurotransmission. Changes in SV2A expression have been linked to several diseases that could implicate an imbalance between excitation and inhibition, such as epilepsy. Although it is known that SV2A expression is necessary for survival, SV2A expression and its relationship with γ-aminobutyric acid (GABA) and glutamate neurotransmitter systems along development has not been addressed. This report follows SV2A expression levels in the rat hippocampus and their association with glutamatergic and GABAergic terminals along postnatal development. Total SV2A expression was assessed by real time PCR and western blot, while immunofluorescence was used to identify SV2A protein in the different hippocampal layers and its co-localization with GABA or glutamate vesicular transporters. SV2A was dynamically regulated along development and its association with GABA or glutamate transporters varied in the different hippocampal layers. In the principal cells layers (granular and pyramidal), SV2A protein was preferentially localized to GABAergic terminals, while in the hilus and stratum lucidum SV2A was associated mainly to glutamatergic terminals. Although SV2A was ubiquitously expressed in the entire hippocampus, it established a differential association with excitatory or inhibitory terminals, which could contribute to the maturation of excitatory/inhibitory balance.
Collapse
|
26
|
Klein P, Diaz A, Gasalla T, Whitesides J. A review of the pharmacology and clinical efficacy of brivaracetam. Clin Pharmacol 2018; 10:1-22. [PMID: 29403319 PMCID: PMC5783144 DOI: 10.2147/cpaa.s114072] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Brivaracetam (BRV; Briviact) is a new antiepileptic drug (AED) approved for adjunctive treatment of focal (partial-onset) seizures in adults. BRV is a selective, high-affinity ligand for synaptic vesicle 2A (SV2A) with 15- to 30-fold higher affinity than levetiracetam, the first AED acting on SV2A. It has high lipid solubility and rapid brain penetration, with engagement of the target molecule, SV2A, within minutes of administration. BRV has potent broad-spectrum antiepileptic activity in animal models. Phase I studies indicated BRV was well tolerated and showed a favorable pharmacokinetic profile over a wide dose range following single (10-1,000 mg) and multiple (200-800 mg/day) oral dosing. Three pivotal Phase III studies have demonstrated promising efficacy and a good safety and tolerability profile across doses of 50-200 mg/day in the adjunctive treatment of refractory focal seizures. Long-term data indicate that the response to BRV is sustained, with good tolerability and retention rate. BRV is highly effective in patients experiencing secondarily generalized tonic-clonic seizures. Safety data to date suggest a favorable psychiatric adverse effect profile in controlled studies, although limited postmarketing data are available. BRV is easy to use, with no titration and little drug-drug interaction. It can be initiated at target dose with no titration. Efficacy is seen on day 1 of oral use in a significant percentage of patients. Intravenous administration in a 2-minute bolus and 15-minute infusion is well tolerated. Here, we review the pharmacology, pharmacokinetics, and clinical data of BRV.
Collapse
Affiliation(s)
- Pavel Klein
- Mid-Atlantic Epilepsy and Sleep Center, Bethesda, MD, USA
| | - Anyzeila Diaz
- Neurology Patient Value Unit, UCB Pharma, Smyrna, GA, USA
| | - Teresa Gasalla
- Neurology Patient Value Unit, UCB Pharma, Monheim am Rhein, Germany
| | | |
Collapse
|
27
|
Zhang L, Wang C, Li W. A meta-analysis of randomized controlled trials on levetiracetam in the treatment of pediatric patients with epilepsy. Neuropsychiatr Dis Treat 2018; 14:769-779. [PMID: 29559784 PMCID: PMC5857150 DOI: 10.2147/ndt.s151413] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE To evaluate clinical efficacy, safety, and tolerability of levetiracetam as mono- or adjunctive therapy in the treatment of children and adolescents with epilepsy. MATERIALS AND METHODS We performed a meta-analysis of randomized controlled trials published from January 2007 to December 2016 in the databases Web of Science, Medline, Embase, Cochrane Library, and PubMed, Bing, Baidu, Google Scholar, Chinese National Knowledge Infrastructure (CNKI), and Wanfang Data. All of the studies eligible were compared for the efficacy, safety, and tolerability of levetiracetam with other antiepileptic drugs (AEDs) in epilepsy. RESULTS Thirteen randomized controlled trials on a total of 1,013 patients met the inclusion criteria in present study. Compared with other AEDs (oxcarbazepine, valproate, sulthiame, carbamazepine, and placebo), we found that levetiracetam had a comparable seizure-free rate (RR 1.16, 95% CI 1.03-1.31; P=0.30). Regarding seizure-frequency reduction ≥50% from baseline, levetiracetam also seemed equivalent to other AEDs (RR 1.08, 95% CI 1.01-1.16; P=0.35). In spite of patients treated with levetiracetam having a lower incidence of side effects compared with patients treated with other AEDs (RR 0.90, 95% CI 0.77-1.06), the difference between them was minute and not statistically significant (P=0.22). CONCLUSION Based on this meta-analysis, it seemed that levetiracetam had comparable effects concerning efficacy, tolerability, and adverse events. Nevertheless, 13 studies were insufficient to draw a conclusion that levetiracetam is effective as mono- and adjunctive therapy for all types of epilepsy syndromes and seizures. Larger-sample and more well-designed trials are needed to justify the widespread use of levetiracetam in the treatment of children and adolescents.
Collapse
Affiliation(s)
- Lanlan Zhang
- Department of Pediatric Neurology, Yancheng Maternal and Child Health Hospital
| | - Chengzhong Wang
- Department of Pediatric Neurology, Yancheng Maternal and Child Health Hospital
| | - Wei Li
- Department of Medical Imaging, Jiangsu Vocational College of Medicine, Yancheng, China
| |
Collapse
|
28
|
Dunn AR, Hoffman CA, Stout KA, Ozawa M, Dhamsania RK, Miller GW. Immunochemical analysis of the expression of SV2C in mouse, macaque and human brain. Brain Res 2017; 1702:85-95. [PMID: 29274878 DOI: 10.1016/j.brainres.2017.12.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 11/30/2017] [Accepted: 12/19/2017] [Indexed: 11/17/2022]
Abstract
The synaptic vesicle glycoprotein 2C (SV2C) is an undercharacterized protein with enriched expression in phylogenetically old brain regions. Its precise role within the brain is unclear, though various lines of evidence suggest that SV2C is involved in the function of synaptic vesicles through the regulation of vesicular trafficking, calcium-induced exocytosis, or synaptotagmin function. SV2C has been linked to multiple neurological disorders, including Parkinson's disease and psychiatric conditions. SV2C is expressed in various cell types-primarily dopaminergic, GABAergic, and cholinergic cells. In mice, it is most highly expressed in nuclei within the basal ganglia, though it is unknown if this pattern of expression is consistent across species. Here, we use a custom SV2C-specific antiserum to describe localization within the brain of mouse, nonhuman primate, and human, including cell-type localization. We found that the immunoreactivity with this antiserum is consistent with previously-published antibodies, and confirmed localization of SV2C in the basal ganglia of rodent, rhesus macaque, and human. We observed strongest expression of SV2C in the substantia nigra, ventral tegmental area, dorsal striatum, pallidum, and nucleus accumbens of each species. Further, we demonstrate colocalization between SV2C and markers of dopaminergic, GABAergic, and cholinergic neurons within these brain regions. SV2C has been increasingly linked to dopamine and basal ganglia function. These antisera will be an important resource moving forward in our understanding of the role of SV2C in vesicle dynamics and neurological disease.
Collapse
Affiliation(s)
- Amy R Dunn
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Carlie A Hoffman
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Kristen A Stout
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Minagi Ozawa
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Rohan K Dhamsania
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States
| | - Gary W Miller
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, United States; Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, United States.
| |
Collapse
|
29
|
Luchian R, Vinţeler E, Chiş C, Vasilescu M, Leopold N, Prates Ramalho JP, Chiş V. Conformational Preference and Spectroscopical Characteristics of the Active Pharmaceutical Ingredient Levetiracetam. J Pharm Sci 2017; 106:3564-3573. [PMID: 28842298 DOI: 10.1016/j.xphs.2017.08.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 08/01/2017] [Accepted: 08/14/2017] [Indexed: 11/19/2022]
Abstract
The analysis of the possible conformers and the conformational change between solid and liquid states of a particular drug molecule are mandatory not only for describing reliably its spectroscopical properties but also for understanding the interaction with the receptor and its mechanism of action. Therefore, here we investigated the free-energy conformational landscape of levetiracetam (LEV) in gas phase as well as in water and ethanol, aiming to describe the 3-dimensional structure and energetic stability of its conformers. Twenty-two unique conformers were identified, and their energetic stability was determined at density functional theory B3LYP/6-31+G(2d,2p) level of theory. The 6 most stable monomers in water, within a relative free-energy window of 0.71 kcal mol-1 and clearly separated in energy from the remaining subset of 16 conformers, as well as the 3 most stable dimers were then used to compute the Boltzmann populations-averaged UV-Vis and NMR spectra of LEV. The conformational landscape in solution is distinctly different from that corresponding to gas phase, particularly due to the relative orientations of the butanamide group. Aiming to clarify the stability of the possible dimers of LEV, we also investigated computationally the structure of a set of 11 nonhydrated and hydrated homochiral hydrogen-bonded LEV dimers.
Collapse
Affiliation(s)
- Raluca Luchian
- Faculty of Physics, Babeş-Bolyai University, 1 Kogălniceanu, RO-400084 Cluj-Napoca, Romania
| | - Emil Vinţeler
- Faculty of Physics, Babeş-Bolyai University, 1 Kogălniceanu, RO-400084 Cluj-Napoca, Romania
| | - Cosmina Chiş
- Pediatric Neurology Department, Children Emergency Hospital, Cluj-Napoca, Romania
| | - Mihai Vasilescu
- Faculty of Physics, Babeş-Bolyai University, 1 Kogălniceanu, RO-400084 Cluj-Napoca, Romania
| | - Nicolae Leopold
- Faculty of Physics, Babeş-Bolyai University, 1 Kogălniceanu, RO-400084 Cluj-Napoca, Romania
| | - João P Prates Ramalho
- Department of Chemistry, School of Science and Technology, University of Évora, Rua Romão Ramalho, 59, 7000-671 Évora, Portugal; CGE-Centro de Geofisica de Evora, University of Évora, Rua Romão Ramalho, 59, 7000-671 Évora, Portugal
| | - Vasile Chiş
- Faculty of Physics, Babeş-Bolyai University, 1 Kogălniceanu, RO-400084 Cluj-Napoca, Romania.
| |
Collapse
|
30
|
Bartholome O, Van den Ackerveken P, Sánchez Gil J, de la Brassinne Bonardeaux O, Leprince P, Franzen R, Rogister B. Puzzling Out Synaptic Vesicle 2 Family Members Functions. Front Mol Neurosci 2017; 10:148. [PMID: 28588450 PMCID: PMC5438990 DOI: 10.3389/fnmol.2017.00148] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 05/02/2017] [Indexed: 01/18/2023] Open
Abstract
Synaptic vesicle proteins 2 (SV2) were discovered in the early 80s, but the clear demonstration that SV2A is the target of efficacious anti-epileptic drugs from the racetam family stimulated efforts to improve understanding of its role in the brain. Many functions have been suggested for SV2 proteins including ions or neurotransmitters transport or priming of SVs. Moreover, several recent studies highlighted the link between SV2 and different neuronal disorders such as epilepsy, Schizophrenia (SCZ), Alzheimer's or Parkinson's disease. In this review article, we will summarize our present knowledge on SV2A function(s) and its potential role(s) in the pathophysiology of various brain disorders.
Collapse
Affiliation(s)
- Odile Bartholome
- Laboratory of Nervous System Disorders and Therapy, GIGA-Neurosciences, University of LiègeLiège, Belgium
| | | | - Judit Sánchez Gil
- Laboratory of Nervous System Disorders and Therapy, GIGA-Neurosciences, University of LiègeLiège, Belgium
| | | | - Pierre Leprince
- Laboratory of Nervous System Disorders and Therapy, GIGA-Neurosciences, University of LiègeLiège, Belgium
| | - Rachelle Franzen
- Laboratory of Nervous System Disorders and Therapy, GIGA-Neurosciences, University of LiègeLiège, Belgium
| | - Bernard Rogister
- Laboratory of Nervous System Disorders and Therapy, GIGA-Neurosciences, University of LiègeLiège, Belgium.,Department of Neurology, Centre Hospitalier Universitaire de Liège (CHU), University of LiègeLiège, Belgium
| |
Collapse
|
31
|
Wood MD, Gillard M. Evidence for a differential interaction of brivaracetam and levetiracetam with the synaptic vesicle 2A protein. Epilepsia 2016; 58:255-262. [DOI: 10.1111/epi.13638] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2016] [Indexed: 01/31/2023]
|
32
|
Löscher W, Gillard M, Sands ZA, Kaminski RM, Klitgaard H. Synaptic Vesicle Glycoprotein 2A Ligands in the Treatment of Epilepsy and Beyond. CNS Drugs 2016; 30:1055-1077. [PMID: 27752944 PMCID: PMC5078162 DOI: 10.1007/s40263-016-0384-x] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The synaptic vesicle glycoprotein SV2A belongs to the major facilitator superfamily (MFS) of transporters and is an integral constituent of synaptic vesicle membranes. SV2A has been demonstrated to be involved in vesicle trafficking and exocytosis, processes crucial for neurotransmission. The anti-seizure drug levetiracetam was the first ligand to target SV2A and displays a broad spectrum of anti-seizure activity in various preclinical models. Several lines of preclinical and clinical evidence, including genetics and protein expression changes, support an important role of SV2A in epilepsy pathophysiology. While the functional consequences of SV2A ligand binding are not fully elucidated, studies suggest that subsequent SV2A conformational changes may contribute to seizure protection. Conversely, the recently discovered negative SV2A modulators, such as UCB0255, counteract the anti-seizure effect of levetiracetam and display procognitive properties in preclinical models. More broadly, dysfunction of SV2A may also be involved in Alzheimer's disease and other types of cognitive impairment, suggesting potential novel therapies for levetiracetam and its congeners. Furthermore, emerging data indicate that there may be important roles for two other SV2 isoforms (SV2B and SV2C) in the pathogenesis of epilepsy, as well as other neurodegenerative diseases. Utilization of recently developed SV2A positron emission tomography ligands will strengthen and reinforce the pharmacological evidence that SV2A is a druggable target, and will provide a better understanding of its role in epilepsy and other neurological diseases, aiding in further defining the full therapeutic potential of SV2A modulation.
Collapse
Affiliation(s)
- Wolfgang Löscher
- Department of Pharmacology, Toxicology and Pharmacy, University of Veterinary Medicine Hannover, Bünteweg 17, 30559, Hannover, Germany.
- Center for Systems Neuroscience, Hannover, Germany.
| | | | | | | | | |
Collapse
|
33
|
Mariani E, Frabetti F, Tarozzi A, Pelleri MC, Pizzetti F, Casadei R. Meta-Analysis of Parkinson's Disease Transcriptome Data Using TRAM Software: Whole Substantia Nigra Tissue and Single Dopamine Neuron Differential Gene Expression. PLoS One 2016; 11:e0161567. [PMID: 27611585 PMCID: PMC5017670 DOI: 10.1371/journal.pone.0161567] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 08/08/2016] [Indexed: 01/21/2023] Open
Abstract
The understanding of the genetic basis of the Parkinson's disease (PD) and the correlation between genotype and phenotype has revolutionized our knowledge about the pathogenetic mechanisms of neurodegeneration, opening up exciting new therapeutic and neuroprotective perspectives. Genomic knowledge of PD is still in its early stages and can provide a good start for studies of the molecular mechanisms that underlie the gene expression variations and the epigenetic mechanisms that may contribute to the complex and characteristic phenotype of PD. In this study we used the software TRAM (Transcriptome Mapper) to analyse publicly available microarray data of a total of 151 PD patients and 130 healthy controls substantia nigra (SN) samples, to identify chromosomal segments and gene loci differential expression. In particular, we separately analyzed PD patients and controls data from post-mortem snap-frozen SN whole tissue and from laser microdissected midbrain dopamine (DA) neurons, to better characterize the specific DA neuronal expression profile associated with the late-stage Parkinson's condition. The default "Map" mode analysis resulted in 10 significantly over/under-expressed segments, mapping on 8 different chromosomes for SN whole tissue and in 4 segments mapping on 4 different chromosomes for DA neurons. In conclusion, TRAM software allowed us to confirm the deregulation of some genomic regions and loci involved in key molecular pathways related to neurodegeneration, as well as to provide new insights about genes and non-coding RNA transcripts not yet associated with the disease.
Collapse
Affiliation(s)
- Elisa Mariani
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Flavia Frabetti
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Andrea Tarozzi
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
| | - Maria Chiara Pelleri
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Fabrizio Pizzetti
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna, Italy
| | - Raffaella Casadei
- Department for Life Quality Studies, University of Bologna, Rimini, Italy
- * E-mail:
| |
Collapse
|
34
|
Zou H, Hurwitz M, Fowler L, Wagner AK. Abbreviated levetiracetam treatment effects on behavioural and histological outcomes after experimental TBI. Brain Inj 2016; 29:78-85. [PMID: 25255156 DOI: 10.3109/02699052.2014.955528] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE Long-term prophylactic treatment with levetiracetam (LEV) has multiple neuroprotective effects in a traumatic brain injury (TBI) rat model. Although a rational time-frame of seizure prophylactic treatment with LEV for after TBI is not well established, clinical prophylaxis with LEV often includes treatment duration similar to clinical treatment guidelines with Phenytoin. Thus, this study investigated the effects of abbreviated LEV treatment on behavioural function and histological evidence of neuroprotection. RESEARCH DESIGN Pre-clinical trial of abbreviated LEV dosing in an experimental model of TBI Methods: After either controlled cortical impact (CCI) injury or sham surgery, rats received three 50 mg kg(-1) doses over 24 hours or vehicle. After injury/sham surgery, beam performance, spatial learning, contusion volume size and hippocampal neuron survival were assessed. RESULTS Abbreviated LEV did not improve motor or cognitive performance after TBI. Further, abbreviated LEV did not improve hippocampal neuron sparing or contusion volumes compared with vehicle controls. CONCLUSIONS Together with previous work assessing daily LEV treatment, these results suggest that longer-term therapy may be required to confer beneficial effects within these domains. These findings may guide (1) future experimental studies assessing minimal effective dosing for neuroprotection and anti-epileptogenesis and (2) treatment guideline updates for seizure prophylaxis post-TBI.
Collapse
Affiliation(s)
- Huichao Zou
- a Department of Physical Medicine and Rehabilitation and
| | | | | | | |
Collapse
|
35
|
Gordon SL, Cousin MA. The iTRAPs: Guardians of Synaptic Vesicle Cargo Retrieval During Endocytosis. Front Synaptic Neurosci 2016; 8:1. [PMID: 26903854 PMCID: PMC4746236 DOI: 10.3389/fnsyn.2016.00001] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 01/25/2016] [Indexed: 11/30/2022] Open
Abstract
The reformation of synaptic vesicles (SVs) during endocytosis is essential for the maintenance of neurotransmission in central nerve terminals. Newly formed SVs must be generated with the correct protein cargo in the correct stoichiometry to be functional for exocytosis. Classical clathrin adaptor protein complexes play a key role in sorting and clustering synaptic vesicle cargo in this regard. However it is becoming increasingly apparent that additional “fail-safe” mechanisms exist to ensure the accurate retrieval of essential cargo molecules. For example, the monomeric adaptor proteins AP180/CALM and stonin-2 are required for the efficient retrieval of synaptobrevin II (sybII) and synaptotagmin-1 respectively. Furthermore, recent studies have revealed that sybII and synaptotagmin-1 interact with other SV cargoes to ensure a high fidelity of retrieval. These cargoes are synaptophysin (for sybII) and SV2A (for synaptotagmin-1). In this review, we summarize current knowledge regarding the retrieval mechanisms for both sybII and synaptotagmin-1 during endocytosis. We also define and set criteria for a new functional group of SV molecules that facilitate the retrieval of their interaction partners. We have termed these molecules intrinsic trafficking partners (iTRAPs) and we discuss how the function of this group impacts on presynaptic performance in both health and disease.
Collapse
Affiliation(s)
- Sarah L Gordon
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne Parkville, VIC, Australia
| | - Michael A Cousin
- Centre for Integrative Physiology, University of Edinburgh Edinburgh, UK
| |
Collapse
|
36
|
Dilena R, Striano P, Traverso M, Viri M, Cristofori G, Tadini L, Barbieri S, Romeo A, Zara F. Dramatic effect of levetiracetam in early-onset epileptic encephalopathy due to STXBP1 mutation. Brain Dev 2016; 38:128-31. [PMID: 26212315 DOI: 10.1016/j.braindev.2015.07.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 07/04/2015] [Accepted: 07/12/2015] [Indexed: 11/25/2022]
Abstract
BACKGROUND Syntaxin Binding Protein 1 (STXBP1) mutations determine a central neurotransmission dysfunction through impairment of the synaptic vesicle release, thus causing a spectrum of phenotypes varying from syndromic and non-syndromic epilepsy to intellectual disability of variable degree. Among the antiepileptic drugs, levetiracetam has a unique mechanism of action binding SV2A, a glycoprotein of the synaptic vesicle release machinery. PATIENT DESCRIPTION We report a 1-month-old boy manifesting an epileptic encephalopathy with clonic seizures refractory to phenobarbital, pyridoxine and phenytoin that presented a dramatic response to levetiracetam with full epilepsy control and EEG normalization. Genetic analysis identified a novel de novo heterozygous mutation (c.[922A>T]p.[Lys308(∗)]) in the STXBP1 gene that severely affects the protein. CONCLUSIONS The observation of a dramatic efficacy of levetiracetam in a case of STXBP1 epileptic encephalopathy refractory to other antiepileptic drugs and considerations regarding the specific mechanism of action of levetiracetam modulating the same system affected by STXBP1 mutations support the hypothesis that this drug may be able to reverse specifically the disease epileptogenic abnormalities. Further clinical observations and laboratory studies are needed to confirm this hypothesis and eventually lead to consider levetiracetam as the first choice treatment of patients with suspected or confirmed STXBP1-related epilepsies.
Collapse
Affiliation(s)
- Robertino Dilena
- Unit of Clinical Neurophysiology, Department of Neuroscience and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy.
| | - Pasquale Striano
- Pediatric Neurology and Muscular Diseases Unit, Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, Institute "G. Gaslini", University of Genova, Genoa, Italy
| | - Monica Traverso
- Pediatric Neurology and Muscular Diseases Unit, Laboratory of Neurogenetics, Institute "G. Gaslini", Genoa, Italy
| | - Maurizio Viri
- Pediatric Neurology Unit and Epilepsy Center, Department of Neuroscience, "Fatebenefratelli e Oftalmico" Hospital, Milan, Italy
| | - Gloria Cristofori
- Neonatal Intensive Care Unit, Department of Clinical Sciences and Community Health, University of Milan, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Laura Tadini
- Unit of Clinical Neurophysiology, Department of Neuroscience and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sergio Barbieri
- Unit of Clinical Neurophysiology, Department of Neuroscience and Mental Health, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Antonino Romeo
- Pediatric Neurology Unit and Epilepsy Center, Department of Neuroscience, "Fatebenefratelli e Oftalmico" Hospital, Milan, Italy
| | - Federico Zara
- Pediatric Neurology and Muscular Diseases Unit, Laboratory of Neurogenetics, Institute "G. Gaslini", Genoa, Italy
| |
Collapse
|
37
|
Yang X, Meehan AL, Rothman SM, Dubinsky JM. Seletracetam enhances short term depression in vitro. Epilepsy Res 2015; 117:17-22. [DOI: 10.1016/j.eplepsyres.2015.08.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 03/20/2015] [Accepted: 08/06/2015] [Indexed: 12/01/2022]
|
38
|
Homozygous Mutation in Synaptic Vesicle Glycoprotein 2A Gene Results in Intractable Epilepsy, Involuntary Movements, Microcephaly, and Developmental and Growth Retardation. Pediatr Neurol 2015; 52:642-6.e1. [PMID: 26002053 DOI: 10.1016/j.pediatrneurol.2015.02.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 02/06/2015] [Accepted: 02/11/2015] [Indexed: 11/23/2022]
Abstract
BACKGROUND Synaptic vesicle protein 2A (SV2a) is the binding site of the antiepileptic drug levetiracetam and the only known synaptic vesicle target of an epilepsy medication. To date, no pathogenic mutation in SV2A, which is the gene encoding synaptic vesicle glycoprotein 2A, has been identified in humans. We report a homozygous mutation in the SV2A gene in a patient with intractable epilepsy. METHODS We investigated a patient with intractable epilepsy, involuntary movements, microcephaly, and developmental and growth retardation. Both parents were multiply consanguineous and an earlier-born brother of the proband had a similar course and died at 7 months of age. Detailed clinical history, imaging, electroencephalograph and metabolic testing were obtained. Full exome sequencing was performed using genomic DNA isolated from the patient and both parents. RESULTS Exome sequencing identified a homozygous arginine to glutamine mutation in amino acid position 383 (R383Q) in exon 5 of the SV2A gene. Both parents were carriers for the R383Q variant, suggesting that R383Q is a recessive mutation. There were no other candidate alterations in the exome that could explain the phenotype in the proband. The amino acid arginine at position 383 of SV2a gene is evolutionally conserved throughout vertebrates. R383Q change is not observed in known healthy cohorts, exome databases, or the Database of Single Nucleotide Polymorphisms. The R383Q mutation is located in the second adenine binding domain in SV2a protein and may alter adenine nucleotides binding to SV2a. CONCLUSION Our report provides the elusive evidence that an SV2A mutation can be a cause of epilepsy in humans. Levetiracetam, which binds to SV2A, was not effective as an antiepileptic medication. The location of the mutation in our patient supports an important role of adenine nucleotides binding in SV2A function.
Collapse
|
39
|
Overlapping functions of stonin 2 and SV2 in sorting of the calcium sensor synaptotagmin 1 to synaptic vesicles. Proc Natl Acad Sci U S A 2015; 112:7297-302. [PMID: 26015569 PMCID: PMC4466747 DOI: 10.1073/pnas.1501627112] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Brain function depends on neurotransmission, and alterations in this process are linked to neurological disorders. Neurotransmitter release requires the rapid recycling of synaptic vesicles (SVs) by endocytosis. How synapses maintain the molecular composition of SVs during recycling is poorly understood. We demonstrate that overlapping functions of two completely distinct proteins, the vesicle protein SV2A/B and the adaptor stonin 2, mediate endocytic sorting of the vesicular calcium sensor synaptotagmin 1. As SV2A is the target of the commonly used antiepileptic drug levetiracetam and is linked to late onset Alzheimer’s disease, our findings bear implications for the treatment of neurological and neurodegenerative disorders. Neurotransmission involves the calcium-regulated exocytic fusion of synaptic vesicles (SVs) and the subsequent retrieval of SV membranes followed by reformation of properly sized and shaped SVs. An unresolved question is whether each SV protein is sorted by its own dedicated adaptor or whether sorting is facilitated by association between different SV proteins. We demonstrate that endocytic sorting of the calcium sensor synaptotagmin 1 (Syt1) is mediated by the overlapping activities of the Syt1-associated SV glycoprotein SV2A/B and the endocytic Syt1-adaptor stonin 2 (Stn2). Deletion or knockdown of either SV2A/B or Stn2 results in partial Syt1 loss and missorting of Syt1 to the neuronal surface, whereas deletion of both SV2A/B and Stn2 dramatically exacerbates this phenotype. Selective missorting and degradation of Syt1 in the absence of SV2A/B and Stn2 impairs the efficacy of neurotransmission at hippocampal synapses. These results indicate that endocytic sorting of Syt1 to SVs is mediated by the overlapping activities of SV2A/B and Stn2 and favor a model according to which SV protein sorting is guarded by both cargo-specific mechanisms as well as association between SV proteins.
Collapse
|
40
|
Abstract
BACKGROUND Levetiracetam, a second-generation anti-epileptic drug (AED) with a good efficacy and safety profile, is licensed as monotherapy for adults and children older than 16 years with focal seizures with or without secondary generalization. However, it is increasingly being used off-label in younger children. OBJECTIVES We critically reviewed the available evidence and discuss the present status of levetiracetam monotherapy in children 0-16 years old. DATA SOURCES We systematically searched the literature using PubMed, Web of Science and Embase up to August 2014 for articles on levetiracetam monotherapy in children. Keywords were levetiracetam, monotherapy and child*. The titles and abstracts of 532 articles were evaluated by AW, of which 480 were excluded. The full texts of the other 52 articles were assessed for relevance. RESULTS We covered one review, one opinion statement and 32 studies in this review, including four randomized controlled trials, ten open-label prospective studies, eight retrospective studies, and ten case reports. The formal evidence for levetiracetam monotherapy in children is minimal: it is potentially efficacious or effective as initial monotherapy in children with benign epilepsy with centrotemporal spikes. In all of the published studies, however, efficacy and tolerability of levetiracetam seemed to be good and comparable to other AEDs. CONCLUSION The data of 32 studies on levetiracetam monotherapy in children were insufficient to confirm that levetiracetam is effective as initial monotherapy for different types of seizures and/or epilepsy syndromes. There is still an urgent need for well designed trials to justify the widespread use of levetiracetam monotherapy in children of all ages.
Collapse
Affiliation(s)
- Amerins Weijenberg
- Department of Neurology, University Medical Centre Groningen, University of Groningen, AB 51, P.O. Box 30 001, 9700 RB Groningen, The Netherlands
| | - Oebele F. Brouwer
- Department of Neurology, University Medical Centre Groningen, University of Groningen, AB 51, P.O. Box 30 001, 9700 RB Groningen, The Netherlands
| | - Petra M. C. Callenbach
- Department of Neurology, University Medical Centre Groningen, University of Groningen, AB 51, P.O. Box 30 001, 9700 RB Groningen, The Netherlands
| |
Collapse
|
41
|
Vogl C, Tanifuji S, Danis B, Daniels V, Foerch P, Wolff C, Whalley BJ, Mochida S, Stephens GJ. Synaptic vesicle glycoprotein 2A modulates vesicular release and calcium channel function at peripheral sympathetic synapses. Eur J Neurosci 2014; 41:398-409. [PMID: 25484265 DOI: 10.1111/ejn.12799] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Revised: 11/03/2014] [Accepted: 11/05/2014] [Indexed: 12/01/2022]
Abstract
Synaptic vesicle glycoprotein (SV)2A is a transmembrane protein found in secretory vesicles and is critical for Ca(2+) -dependent exocytosis in central neurons, although its mechanism of action remains uncertain. Previous studies have proposed, variously, a role of SV2 in the maintenance and formation of the readily releasable pool (RRP) or in the regulation of Ca(2+) responsiveness of primed vesicles. Such previous studies have typically used genetic approaches to ablate SV2 levels; here, we used a strategy involving small interference RNA (siRNA) injection to knockdown solely presynaptic SV2A levels in rat superior cervical ganglion (SCG) neuron synapses. Moreover, we investigated the effects of SV2A knockdown on voltage-dependent Ca(2+) channel (VDCC) function in SCG neurons. Thus, we extended the studies of SV2A mechanisms by investigating the effects on vesicular transmitter release and VDCC function in peripheral sympathetic neurons. We first demonstrated an siRNA-mediated SV2A knockdown. We showed that this SV2A knockdown markedly affected presynaptic function, causing an attenuated RRP size, increased paired-pulse depression and delayed RRP recovery after stimulus-dependent depletion. We further demonstrated that the SV2A-siRNA-mediated effects on vesicular release were accompanied by a reduction in VDCC current density in isolated SCG neurons. Together, our data showed that SV2A is required for correct transmitter release at sympathetic neurons. Mechanistically, we demonstrated that presynaptic SV2A: (i) acted to direct normal synaptic transmission by maintaining RRP size, (ii) had a facilitatory role in recovery from synaptic depression, and that (iii) SV2A deficits were associated with aberrant Ca(2+) current density, which may contribute to the secretory phenotype in sympathetic peripheral neurons.
Collapse
|
42
|
Niespodziany I, André VM, Leclère N, Hanon E, Ghisdal P, Wolff C. Brivaracetam differentially affects voltage-gated sodium currents without impairing sustained repetitive firing in neurons. CNS Neurosci Ther 2014; 21:241-51. [PMID: 25444522 PMCID: PMC4359682 DOI: 10.1111/cns.12347] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 10/03/2014] [Accepted: 10/05/2014] [Indexed: 11/29/2022] Open
Abstract
Aims Brivaracetam (BRV) is an antiepileptic drug in Phase III clinical development. BRV binds to synaptic vesicle 2A (SV2A) protein and is also suggested to inhibit voltage‐gated sodium channels (VGSCs). To evaluate whether the effect of BRV on VGSCs represents a relevant mechanism participating in its antiepileptic properties, we explored the pharmacology of BRV on VGSCs in different cell systems and tested its efficacy at reducing the sustained repetitive firing (SRF). Methods Brivaracetam investigations on the voltage‐gated sodium current (INa) were performed in N1E‐155 neuroblastoma cells, cultured rat cortical neurons, and adult mouse CA1 neurons. SRF was measured in cultured cortical neurons and in CA1 neurons. All BRV (100–300 μM) experiments were performed in comparison with 100 μM carbamazepine (CBZ). Results Brivaracetam and CBZ reduced INa in N1E‐115 cells (30% and 40%, respectively) and primary cortical neurons (21% and 47%, respectively) by modulating the fast‐inactivated state of VGSCs. BRV, in contrast to CBZ, did not affect INa in CA1 neurons and SRF in cortical and CA1 neurons. CBZ consistently inhibited neuronal SRF by 75–93%. Conclusions The lack of effect of BRV on SRF in neurons suggests that the reported inhibition of BRV on VGSC currents does not contribute to its antiepileptic properties.
Collapse
|
43
|
Abstract
Voltage-gated sodium channels initiate action potentials in brain neurons, mutations in sodium channels cause inherited forms of epilepsy, and sodium channel blockers-along with other classes of drugs-are used in therapy of epilepsy. A mammalian voltage-gated sodium channel is a complex containing a large, pore-forming α subunit and one or two smaller β subunits. Extensive structure-function studies have revealed many aspects of the molecular basis for sodium channel structure, and X-ray crystallography of ancestral bacterial sodium channels has given insight into their three-dimensional structure. Mutations in sodium channel α and β subunits are responsible for genetic epilepsy syndromes with a wide range of severity, including generalized epilepsy with febrile seizures plus (GEFS+), Dravet syndrome, and benign familial neonatal-infantile seizures. These seizure syndromes are treated with antiepileptic drugs that offer differing degrees of success. The recent advances in understanding of disease mechanisms and sodium channel structure promise to yield improved therapeutic approaches.
Collapse
Affiliation(s)
- William A Catterall
- Department of Pharmacology, University of Washington, Seattle, Washington 98195-7280;
| |
Collapse
|
44
|
Effects of Levetiracetam on Aggregation and Fusion of Membranes of Synaptic Vesicles in a Cell-Free Exocytosis Model. NEUROPHYSIOLOGY+ 2014. [DOI: 10.1007/s11062-014-9416-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
45
|
|
46
|
Levetiracetam resistance: Synaptic signatures & corresponding promoter SNPs in epileptic hippocampi. Neurobiol Dis 2013; 60:115-25. [DOI: 10.1016/j.nbd.2013.08.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 08/22/2013] [Accepted: 08/27/2013] [Indexed: 01/16/2023] Open
|
47
|
Hill-Burns EM, Singh N, Ganguly P, Hamza TH, Montimurro J, Kay DM, Yearout D, Sheehan P, Frodey K, Mclear JA, Feany MB, Hanes SD, Wolfgang WJ, Zabetian CP, Factor SA, Payami H. A genetic basis for the variable effect of smoking/nicotine on Parkinson's disease. THE PHARMACOGENOMICS JOURNAL 2013; 13:530-7. [PMID: 23032990 PMCID: PMC3538110 DOI: 10.1038/tpj.2012.38] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 07/27/2012] [Accepted: 08/20/2012] [Indexed: 11/09/2022]
Abstract
Prior studies have established an inverse association between cigarette smoking and the risk of developing Parkinson's disease (PD), and currently, the disease-modifying potential of the nicotine patch is being tested in clinical trials. To identify genes that interact with the effect of smoking/nicotine, we conducted genome-wide interaction studies in humans and in Drosophila. We identified SV2C, which encodes a synaptic-vesicle protein in PD-vulnerable substantia nigra (P=1 × 10(-7) for gene-smoking interaction on PD risk), and CG14691, which is predicted to encode a synaptic-vesicle protein in Drosophila (P=2 × 10(-11) for nicotine-paraquat interaction on gene expression). SV2C is biologically plausible because nicotine enhances the release of dopamine through synaptic vesicles, and PD is caused by the depletion of dopamine. Effect of smoking on PD varied by SV2C genotype from protective to neutral to harmful (P=5 × 10(-10)). Taken together, cross-validating evidence from humans and Drosophila suggests SV2C is involved in PD pathogenesis and it might be a useful marker for pharmacogenomics studies involving nicotine.
Collapse
Affiliation(s)
- Erin M. Hill-Burns
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Navjot Singh
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Prabarna Ganguly
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Taye H. Hamza
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Jennifer Montimurro
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Denise M. Kay
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Dora Yearout
- VA Puget Sound Health Care System and Department of Neurology, University of Washington, Seattle, WA, USA
| | - Patricia Sheehan
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Kevin Frodey
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Julie A. Mclear
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Mel B. Feany
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Steven D. Hanes
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY, USA
- Division of Infectious Disease Wadsworth Center, New York State Department of Health, Albany, NY, United States, USA
- Department of Biomedical Science, School of Public Health, State University of New York, Albany, NY, USA
- Department of Biochemistry and Molecular Biology, SUNY-Upstate Medical University, Syracuse, NY, USA
| | - William J. Wolfgang
- Division of Infectious Disease Wadsworth Center, New York State Department of Health, Albany, NY, United States, USA
- Department of Biomedical Science, School of Public Health, State University of New York, Albany, NY, USA
| | - Cyrus P. Zabetian
- VA Puget Sound Health Care System and Department of Neurology, University of Washington, Seattle, WA, USA
| | - Stewart A. Factor
- Department of Neurology, Emory University School of Medicine, Atlanta, GA, USA
| | - Haydeh Payami
- Division of Genetics, Wadsworth Center, New York State Department of Health, Albany, NY, USA
- Department of Biomedical Science, School of Public Health, State University of New York, Albany, NY, USA
| |
Collapse
|
48
|
Bankstahl M, Bankstahl JP, Löscher W. Pilocarpine-induced epilepsy in mice alters seizure thresholds and the efficacy of antiepileptic drugs in the 6-Hertz psychomotor seizure model. Epilepsy Res 2013; 107:205-16. [DOI: 10.1016/j.eplepsyres.2013.09.014] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2013] [Revised: 09/02/2013] [Accepted: 09/27/2013] [Indexed: 12/25/2022]
|
49
|
Nensa FM, Neumann MHD, Schrötter A, Przyborski A, Mastalski T, Susdalzew S, Looβe C, Helling S, El Magraoui F, Erdmann R, Meyer HE, Uszkoreit J, Eisenacher M, Suh J, Guénette SY, Röhner N, Kögel D, Theiss C, Marcus K, Müller T. Amyloid beta a4 precursor protein-binding family B member 1 (FE65) interactomics revealed synaptic vesicle glycoprotein 2A (SV2A) and sarcoplasmic/endoplasmic reticulum calcium ATPase 2 (SERCA2) as new binding proteins in the human brain. Mol Cell Proteomics 2013; 13:475-88. [PMID: 24284412 DOI: 10.1074/mcp.m113.029280] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
FE65 is a cytosolic adapter protein and an important binding partner of amyloid precursor protein. Dependent on Thr668 phosphorylation in amyloid precursor protein, which influences amyloidogenic amyloid precursor protein processing, FE65 undergoes nuclear translocation, thereby transmitting a signal from the cell membrane to the nucleus. As this translocation may be relevant in Alzheimer disease, and as FE65 consists of three protein-protein interaction domains able to bind and affect a variety of other proteins and downstream signaling pathways, the identification of the FE65 interactome is of central interest in Alzheimer disease research. In this study, we identified 121 proteins as new potential FE65 interacting proteins in a pulldown/mass spectrometry approach using human post-mortem brain samples as protein pools for recombinantly expressed FE65. Co-immunoprecipitation assays further validated the interaction of FE65 with the candidates SV2A and SERCA2. In parallel, we investigated the whole cell proteome of primary hippocampal neurons from FE65/FE65L1 double knockout mice. Notably, the validated FE65 binding proteins were also found to be differentially abundant in neurons derived from the FE65 knockout mice relative to wild-type control neurons. SERCA2 is an important player in cellular calcium homeostasis, which was found to be up-regulated in double knockout neurons. Indeed, knock-down of FE65 in HEK293T cells also evoked an elevated sensitivity to thapsigargin, a stressor specifically targeting the activity of SERCA2. Thus, our results suggest that FE65 is involved in the regulation of intracellular calcium homeostasis. Whereas transfection of FE65 alone caused a typical dot-like phenotype in the nucleus, co-transfection of SV2A significantly reduced the percentage of FE65 dot-positive cells, pointing to a possible role for SV2A in the modulation of FE65 intracellular targeting. Given that SV2A has a signaling function at the presynapse, its effect on FE65 intracellular localization suggests that the SV2A/FE65 interaction might play a role in synaptic signal transduction.
Collapse
Affiliation(s)
- Fabian M Nensa
- Functional Proteomics, Medizinisches Proteom-Center, Ruhr-University Bochum, D-44801 Bochum, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Harada S, Tanaka S, Takahashi Y, Matsumura H, Shimamoto C, Nakano T, Kuwabara H, Sawabe Y, Nakahari T. Inhibition of Ca(2+)-regulated exocytosis by levetiracetam, a ligand for SV2A, in antral mucous cells of guinea pigs. Eur J Pharmacol 2013; 721:185-92. [PMID: 24076180 DOI: 10.1016/j.ejphar.2013.09.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 09/14/2013] [Accepted: 09/19/2013] [Indexed: 11/18/2022]
Abstract
Levtiracetam (Lev), an inhibitor of SV2A (synaptic vesicle protein A2), affected the ATP-dependent priming of Ca(2+)-regulated exocytosis in antral mucous cells of guinea pig. In antral mucous cells, the Ca(2+)-regulated exocytosis, which is activated by acetylcholine (ACh), consists of an initial peak that declines rapidly (initial phase) followed by a second slower decline (late phase). Dinitrophenol (DNP), which depletes ATP, inhibits the ATP-dependent priming. DNP abolished the initial phase by reducing the number of primed granules, Lev decreased the frequency of initial phase, but not in the presence of DNP. Moreover, 8-bromoguanosine 3'5'-cyclic monophosphate (8BrcGMP) accelerates the ATP-dependent priming. 8BrcGMP enhances the frequency of initial phase by increasing the number of primed granule. Lev added prior to 8BrcGMP addition decreased the frequency of initial phase, but Lev added after 8BrcGMP addition did not. Thus, Lev affected the granules in the process of priming, but it did not affect the granules already primed. Lev did not affect [Ca(2+)]i in unstimulated or ACh-stimulated antral mucous cells. Immunohistochemistry and western blotting demonstrated that SV2A exists in antral mucous cells. The results suggest that SV2A plays an essential role in maintaining the process of ATP-dependent priming in antral mucous cells. In conclusion, Lev decreases the frequency of Ca(2+)-regulated exocytosis the number of primed granules by inhibiting SV2A functions, leading to a decrease in antral mucous cells.
Collapse
Affiliation(s)
- Saeko Harada
- Nakahari Project of Central Research Laboratory, Osaka Medical College, Takatsuki 569-8686, Japan; Laboratory of Pharmacotherapy, Osaka University of Pharmaceutical Sciences, Takatsuki 569-1094, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|