1
|
Socała K, Jakubiec M, Abram M, Mlost J, Starowicz K, Kamiński RM, Ciepiela K, Andres-Mach M, Zagaja M, Metcalf CS, Zawadzki P, Wlaź P, Kamiński K. TRPV1 channel in the pathophysiology of epilepsy and its potential as a molecular target for the development of new antiseizure drug candidates. Prog Neurobiol 2024; 240:102634. [PMID: 38834133 DOI: 10.1016/j.pneurobio.2024.102634] [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: 10/25/2023] [Revised: 04/26/2024] [Accepted: 05/28/2024] [Indexed: 06/06/2024]
Abstract
Identification of transient receptor potential cation channel, subfamily V member 1 (TRPV1), also known as capsaicin receptor, in 1997 was a milestone achievement in the research on temperature sensation and pain signalling. Very soon after it became evident that TRPV1 is implicated in a wide array of physiological processes in different peripheral tissues, as well as in the central nervous system, and thereby could be involved in the pathophysiology of numerous diseases. Increasing evidence suggests that modulation of TRPV1 may also affect seizure susceptibility and epilepsy. This channel is localized in brain regions associated with seizures and epilepsy, and its overexpression was found both in animal models of seizures and in brain samples from epileptic patients. Moreover, modulation of TRPV1 on non-neuronal cells (microglia, astrocytes, and/or peripheral immune cells) may have an impact on the neuroinflammatory processes that play a role in epilepsy and epileptogenesis. In this paper, we provide a comprehensive and critical overview of currently available data on TRPV1 as a possible molecular target for epilepsy management, trying to identify research gaps and future directions. Overall, several converging lines of evidence implicate TRPV1 channel as a potentially attractive target in epilepsy research but more studies are needed to exploit the possible role of TRPV1 in seizures/epilepsy and to evaluate the value of TRPV1 ligands as candidates for new antiseizure drugs.
Collapse
Affiliation(s)
- Katarzyna Socała
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, Lublin PL 20-033, Poland.
| | - Marcin Jakubiec
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| | - Michał Abram
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| | - Jakub Mlost
- Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Cracow PL 31-343, Poland
| | - Katarzyna Starowicz
- Department of Neurochemistry, Maj Institute of Pharmacology, Polish Academy of Sciences, Smętna 12, Cracow PL 31-343, Poland
| | - Rafał M Kamiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| | - Katarzyna Ciepiela
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland; Selvita S.A., Bobrzyńskiego 14, Cracow PL 30-348, Poland
| | - Marta Andres-Mach
- Department of Experimental Pharmacology, Institute of Rural Health, Jaczewskiego 2, Lublin PL 20-090, Poland
| | - Mirosław Zagaja
- Department of Experimental Pharmacology, Institute of Rural Health, Jaczewskiego 2, Lublin PL 20-090, Poland
| | - Cameron S Metcalf
- Department of Pharmacology and Toxicology, University of Utah, Salt Lake City, UT 84112, USA
| | - Przemysław Zawadzki
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| | - Piotr Wlaź
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Akademicka 19, Lublin PL 20-033, Poland
| | - Krzysztof Kamiński
- Department of Medicinal Chemistry, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, Cracow PL 30-688, Poland
| |
Collapse
|
2
|
Devinsky O, Jones NA, Cunningham MO, Jayasekera BAP, Devore S, Whalley BJ. Cannabinoid treatments in epilepsy and seizure disorders. Physiol Rev 2024; 104:591-649. [PMID: 37882730 DOI: 10.1152/physrev.00049.2021] [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: 03/25/2022] [Revised: 10/17/2023] [Accepted: 10/22/2023] [Indexed: 10/27/2023] Open
Abstract
Cannabis has been used to treat convulsions and other disorders since ancient times. In the last few decades, preclinical animal studies and clinical investigations have established the role of cannabidiol (CBD) in treating epilepsy and seizures and support potential therapeutic benefits for cannabinoids in other neurological and psychiatric disorders. Here, we comprehensively review the role of cannabinoids in epilepsy. We briefly review the diverse physiological processes mediating the central nervous system response to cannabinoids, including Δ9-tetrahydrocannabinol (Δ9-THC), cannabidiol, and terpenes. Next, we characterize the anti- and proconvulsive effects of cannabinoids from animal studies of acute seizures and chronic epileptogenesis. We then review the clinical literature on using cannabinoids to treat epilepsy, including anecdotal evidence and case studies as well as the more recent randomized controlled clinical trials that led to US Food and Drug Administration approval of CBD for some types of epilepsy. Overall, we seek to evaluate our current understanding of cannabinoids in epilepsy and focus future research on unanswered questions.
Collapse
Affiliation(s)
- Orrin Devinsky
- Department of Neurology, NYU Grossman School of Medicine, New York, New York, United States
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, New York, United States
- Department of Psychiatry, NYU Grossman School of Medicine, New York, New York, United States
| | | | - Mark O Cunningham
- Discipline of Physiology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - B Ashan P Jayasekera
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Department of Neurosurgery, Royal Victoria Hospital, Newcastle upon Tyne, United Kingdom
| | - Sasha Devore
- Department of Neurology, NYU Grossman School of Medicine, New York, New York, United States
| | | |
Collapse
|
3
|
Yang F, Sivils A, Cegielski V, Singh S, Chu XP. Transient Receptor Potential (TRP) Channels in Pain, Neuropsychiatric Disorders, and Epilepsy. Int J Mol Sci 2023; 24:ijms24054714. [PMID: 36902145 PMCID: PMC10003176 DOI: 10.3390/ijms24054714] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 02/21/2023] [Accepted: 02/22/2023] [Indexed: 03/05/2023] Open
Abstract
Pharmacomodulation of membrane channels is an essential topic in the study of physiological conditions and disease status. Transient receptor potential (TRP) channels are one such family of nonselective cation channels that have an important influence. In mammals, TRP channels consist of seven subfamilies with a total of twenty-eight members. Evidence shows that TRP channels mediate cation transduction in neuronal signaling, but the full implication and potential therapeutic applications of this are not entirely clear. In this review, we aim to highlight several TRP channels which have been shown to mediate pain sensation, neuropsychiatric disorders, and epilepsy. Recent findings suggest that TRPM (melastatin), TRPV (vanilloid), and TRPC (canonical) are of particular relevance to these phenomena. The research reviewed in this paper validates these TRP channels as potential targets of future clinical treatment and offers patients hope for more effective care.
Collapse
|
4
|
Manna SSS. Dual effects of anandamide in the antiepileptic activity of diazepam in pentylenetetrazole-induced seizures in mice. Behav Pharmacol 2022; 33:527-541. [PMID: 36094027 DOI: 10.1097/fbp.0000000000000700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The prototype endocannabinoid, anandamide activates both CB 1 and transient receptor potential vanilloid type 1 channels (TRPV1) receptor at different concentrations. At high concentrations, anandamide-mediated TRPV1 effects are opposite to its effects at low concentrations via CB 1 receptor. Thus, synaptic concentrations of anandamide govern the neuronal activity and consequently might affect the response of a drug. This study was undertaken to investigate the influence of high and low doses of anandamide on the anticonvulsant action of diazepam on the subcutaneous dose of pentylenetetrazole (PTZ) in Swiss mice weighing 20-25 g. Results revealed that intracerebroventricular administration of capsazepine (a TRPV1 antagonist: 1, 10, or 100 µg/mouse) and the low doses (10 µg/mouse) of anandamide, AM404 (anandamide transport inhibitor), or URB597 (fatty acid amide hydrolase inhibitor) augmented the anticonvulsant effect of diazepam. Conversely, higher dose of anandamide, AM404, URB597 (100 µg/mouse) as well as capsaicin (a TRPV1 agonist: 1, 10, or 100 µg/mouse) attenuated the protective effect of diazepam against PTZ-induced seizures. Thus, this study demonstrates that the effects of diazepam may be augmented by activating CB 1 receptors or dampened via TRPV1 receptors. The findings of the present study can be extrapolated to understand the use of TRPV1 blockers alone or in combination of benzodiazepines in the treatment of benzodiazepines-refractory status epilepticus, a condition associated with maladaptive trafficking of synaptic gamma-aminobutyric acid and glutamate receptors. However, potential clinical applications are needed to further support such preclinical studies.
Collapse
|
5
|
Zirotti Rosenberg A, Méndez-Ruette M, Gorziglia M, Alzerreca B, Cabello J, Kaufmann S, Rambousek L, Iturriaga Jofré A, Wyneken U, Lafourcade CA. Behavioral and Molecular Responses to Exogenous Cannabinoids During Pentylenetetrazol-Induced Convulsions in Male and Female Rats. Front Mol Neurosci 2022; 15:868583. [PMID: 36147210 PMCID: PMC9488559 DOI: 10.3389/fnmol.2022.868583] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 05/13/2022] [Indexed: 11/13/2022] Open
Abstract
Epilepsy is a disabling, chronic brain disease,affecting ~1% of the World’s population, characterized by recurrent seizures (sudden, uncontrolled brain activity), which may manifest with motor symptoms (e.g., convulsions) or non-motor symptoms. Temporal lobe epilepsies (TLE) compromising the hippocampus are the most common form of focal epilepsies. Resistance in ~1/3 of epileptic patients to the first line of treatment, i.e., antiepileptic drugs (AEDs), has been an important motivation to seek alternative treatments. Among these, the plant Cannabis sativa (commonly known as marihuana) or compounds extracted from it (cannabinoids) have gained widespread popularity. Moreover, sex differences have been proposed in epilepsy syndromes and in cannabinoid action. In the hippocampus, cannabinoids interact with the CB1R receptor whose membrane levels are regulated by β-Arrestin2, a protein that promotes its endocytosis and causes its downregulation. In this article, we evaluate the modulatory role of WIN 55,212-2 (WIN), a synthetic exogenous cannabinoid on behavioral convulsions and on the levels of CB1R and β-Arrestin2 in female and male adolescent rats after a single injection of the proconvulsant pentylenetetrazol (PTZ). As epilepsies can have a considerable impact on synaptic proteins that regulate neuronal toxicity, plasticity, and cognition, we also measured the levels of key proteins markers of excitatory synapses, in order to examine whether exogenous cannabinoids may prevent such pathologic changes after acute seizures. We found that the exogenous administration of WIN prevented convulsions of medium severity in females and males and increased the levels of phosphorylated CaMKII in the hippocampus. Furthermore, we observed a higher degree of colocalization between CB1R and β-Arrestin2 in the granule cell layer.
Collapse
Affiliation(s)
| | - Maxs Méndez-Ruette
- Centro de investigación e innovación Biomédica (CiiB), Laboratorio de Neurociencias, Universidad de Los Andes, Santiago, Chile
| | - Mario Gorziglia
- Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | | | - Javiera Cabello
- Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Sofía Kaufmann
- Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
| | - Lukas Rambousek
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | | | - Ursula Wyneken
- Facultad de Medicina, Universidad de Los Andes, Santiago, Chile
- IMPACT, Center of Interventional Medicine for Precision and Advanced Cellular Therapy, Santiago, Chile
| | - Carlos A. Lafourcade
- Department of Biological Sciences, Xi’an Jiaotong-Liverpool University (XJTLU), Suzhou, China
- *Correspondence: Carlos A. Lafourcade
| |
Collapse
|
6
|
Llanos MA, Enrique N, Sbaraglini ML, Garofalo FM, Talevi A, Gavernet L, Martín P. Structure-Based Virtual Screening Identifies Novobiocin, Montelukast, and Cinnarizine as TRPV1 Modulators with Anticonvulsant Activity In Vivo. J Chem Inf Model 2022; 62:3008-3022. [PMID: 35696534 DOI: 10.1021/acs.jcim.2c00312] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The transient receptor potential vanilloid 1 (TRPV1) receptor is a nonselective cation channel, known to be involved in the regulation of many important physiological and pathological processes. In the last few years, it has been proposed as a promising target to develop novel anticonvulsant compounds. However, thermoregulatory effects associated with the channel inhibition have hampered the path for TRPV1 antagonists to become marketed drugs. In this regard, we conducted a structure-based virtual screening campaign to find potential TRPV1 modulators among approved drugs, which are known to be safe and thermally neutral. To this end, different docking models were developed and validated by assessing their pose and score prediction powers. Novobiocin, montelukast, and cinnarizine were selected from the screening as promising candidates for experimental testing and all of them exhibited nanomolar inhibitory activity. Moreover, the in vivo profiles showed promising results in at least one of the three models of seizures tested.
Collapse
Affiliation(s)
- Manuel A Llanos
- Departamento de Ciencias Biológicas and Laboratorio de Investigación y Desarrollo de Bioactivos (LIDeB), UNLP, Facultad de Ciencias Exactas, La Plata Buenos Aires (B1900ADU), Argentina
| | - Nicolás Enrique
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP, CONICET─Universidad Nacional de la Plata), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata Buenos Aires (B1900BJW), Argentina
| | - María L Sbaraglini
- Departamento de Ciencias Biológicas and Laboratorio de Investigación y Desarrollo de Bioactivos (LIDeB), UNLP, Facultad de Ciencias Exactas, La Plata Buenos Aires (B1900ADU), Argentina
| | - Federico M Garofalo
- Departamento de Ciencias Biológicas and Laboratorio de Investigación y Desarrollo de Bioactivos (LIDeB), UNLP, Facultad de Ciencias Exactas, La Plata Buenos Aires (B1900ADU), Argentina
| | - Alan Talevi
- Departamento de Ciencias Biológicas and Laboratorio de Investigación y Desarrollo de Bioactivos (LIDeB), UNLP, Facultad de Ciencias Exactas, La Plata Buenos Aires (B1900ADU), Argentina
| | - Luciana Gavernet
- Departamento de Ciencias Biológicas and Laboratorio de Investigación y Desarrollo de Bioactivos (LIDeB), UNLP, Facultad de Ciencias Exactas, La Plata Buenos Aires (B1900ADU), Argentina
| | - Pedro Martín
- Instituto de Estudios Inmunológicos y Fisiopatológicos (IIFP, CONICET─Universidad Nacional de la Plata), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata Buenos Aires (B1900BJW), Argentina
| |
Collapse
|
7
|
Beneficial Effects of Capsaicin in Disorders of the Central Nervous System. Molecules 2022; 27:molecules27082484. [PMID: 35458680 PMCID: PMC9029810 DOI: 10.3390/molecules27082484] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 01/13/2023] Open
Abstract
Capsaicin is a natural compound found in chili peppers and is used in the diet of many countries. The important mechanism of action of capsaicin is its influence on TRPV1 channels in nociceptive sensory neurons. Furthermore, the beneficial effects of capsaicin in cardiovascular and oncological disorders have been described. Many recent publications show the positive effects of capsaicin in animal models of brain disorders. In Alzheimer’s disease, capsaicin reduces neurodegeneration and memory impairment. The beneficial effects of capsaicin in Parkinson’s disease and depression have also been described. It has been found that capsaicin reduces the area of infarction and improves neurological outcomes in animal models of stroke. However, both proepileptic and antiepileptic effects of capsaicin in animal models of epilepsy have been proposed. These contradictory results may be caused by the fact that capsaicin influences not only TRPV1 channels but also different molecular targets such as voltage-gated sodium channels. Human studies show that capsaicin may be helpful in treating stroke complications such as dysphagia. Additionally, this compound exerts pain-relieving effects in migraine and cluster headaches. The purpose of this review is to discuss the mechanisms of the beneficial effects of capsaicin in disorders of the central nervous system.
Collapse
|
8
|
Khaksar S, Salimi M, Zeinoddini H, Naderi N. The Role of the Possible Receptors and Intracellular Pathways in Protective Effect of Exogenous Anandamide in Kindling Model of Epilepsy. Neurochem Res 2022; 47:1226-1242. [PMID: 35112235 DOI: 10.1007/s11064-021-03517-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 12/01/2021] [Accepted: 12/24/2021] [Indexed: 11/24/2022]
Abstract
In this research, the involvement of CB1 and TRPV1 receptors in the possible protective effects of anandamide were investigated in the kindling model of epilepsy. The basolateral amygdala of the rat brain was chosen to put stimulating electrodes. Semi-rapid kindling was induced by a repetitive sub-threshold stimulation for 5-9 consecutive days. There were seven groups, six of which were kindled and used for drug testing by intracerebroventricular (i.c.v.) microinjection. (i) Sham, (ii) control group received vehicles, (iii) anandamide (AEA; 100 ng/rat), (iv) capsazepine (TRPV1 antagonist; 100 ng/rat), (v) AM251 (CB1 antagonist; 100 ng/rat), (vi) AM251 + anandamide, and (vii) capsazepine + anandamide. The after-discharge duration, seizure duration, and stage five duration were measured in rats. Moreover, the expressions of the extracellular signal-regulated kinase (ERK) and the cAMP responsive element binding (CREB) proteins in the hippocampus were also studied. The anandamide-treated group showed a significant decrease in seizure scores, while no change was shown in seizure scores in the capsazepine- and AM251-treated groups compared with the control group. Co-administrations of either capsazepine + AEA or AM251 + AEA attenuated the protective effect of AEA against seizure. Furthermore, the group received AEA showed a decrease in the expressions of CREB and p-CREB possibly through the activation of the CB1 and TRPV1 receptors. Activation of CB1 and TRPV1 receptors might be involved in AEA anticonvulsant effect in kindling model of epilepsy. This effect could be due to suppression of CREB phosphorylation in hippocampal neurons.
Collapse
Affiliation(s)
- Sepideh Khaksar
- Department of Plant Sciences, Biological Sciences, Alzahra University, Tehran, Iran
| | - Mona Salimi
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Hadi Zeinoddini
- Department of Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, No. 2660, Vali-e-Asr Ave, 1996835113, Tehran, Iran
| | - Nima Naderi
- Department of Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, No. 2660, Vali-e-Asr Ave, 1996835113, Tehran, Iran. .,Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Sugaya Y, Kano M. Endocannabinoid-Mediated Control of Neural Circuit Excitability and Epileptic Seizures. Front Neural Circuits 2022; 15:781113. [PMID: 35046779 PMCID: PMC8762319 DOI: 10.3389/fncir.2021.781113] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/29/2021] [Indexed: 01/11/2023] Open
Abstract
Research on endocannabinoid signaling has greatly advanced our understanding of how the excitability of neural circuits is controlled in health and disease. In general, endocannabinoid signaling at excitatory synapses suppresses excitability by inhibiting glutamate release, while that at inhibitory synapses promotes excitability by inhibiting GABA release, although there are some exceptions in genetically epileptic animal models. In the epileptic brain, the physiological distributions of endocannabinoid signaling molecules are disrupted during epileptogenesis, contributing to the occurrence of spontaneous seizures. However, it is still unknown how endocannabinoid signaling changes during seizures and how the redistribution of endocannabinoid signaling molecules proceeds during epileptogenesis. Recent development of cannabinoid sensors has enabled us to investigate endocannabinoid signaling in much greater spatial and temporal details than before. Application of cannabinoid sensors to epilepsy research has elucidated activity-dependent changes in endocannabinoid signaling during seizures. Furthermore, recent endocannabinoid research has paved the way for the clinical use of cannabidiol for the treatment of refractory epilepsy, such as Dravet syndrome, Lennox-Gastaut syndrome and tuberous sclerosis complex. Cannabidiol significantly reduces seizures and is considered to have comparable tolerability to conventional antiepileptic drugs. In this article, we introduce recent advances in research on the roles of endocannabinoid signaling in epileptic seizures and discuss future directions.
Collapse
Affiliation(s)
- Yuki Sugaya
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, Japan
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, Japan
- *Correspondence: Masanobu Kano,
| |
Collapse
|
10
|
Martínez-Aguirre C, Cinar R, Rocha L. Targeting Endocannabinoid System in Epilepsy: For Good or for Bad. Neuroscience 2021; 482:172-185. [PMID: 34923038 DOI: 10.1016/j.neuroscience.2021.12.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/29/2021] [Accepted: 12/09/2021] [Indexed: 02/07/2023]
Abstract
Epilepsy is a neurological disorder with a high prevalence worldwide. Several studies carried out during the last decades indicate that the administration of cannabinoids as well as the activation of the endocannabinoid system (ECS) represent a therapeutic strategy to control epilepsy. However, there are controversial studies indicating that activation of ECS results in cell damage, inflammation and neurotoxicity, conditions that facilitate the seizure activity. The present review is focused to present findings supporting this issue. According to the current discrepancies, it is relevant to elucidate the different effects induced by the activation of ECS and determine the conditions under which it facilitates the seizure activity.
Collapse
Affiliation(s)
| | - Resat Cinar
- Section on Fibrotic Disorders, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Rockville, USA
| | - Luisa Rocha
- Department of Pharmacobiology, Center for Research and Advanced Studies, Mexico City, Mexico.
| |
Collapse
|
11
|
Asth L, Iglesias LP, De Oliveira AC, Moraes MFD, Moreira FA. Exploiting cannabinoid and vanilloid mechanisms for epilepsy treatment. Epilepsy Behav 2021; 121:106832. [PMID: 31839498 DOI: 10.1016/j.yebeh.2019.106832] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 11/25/2019] [Accepted: 11/25/2019] [Indexed: 12/28/2022]
Abstract
This review focuses on the possible roles of phytocannabinoids, synthetic cannabinoids, endocannabinoids, and "transient receptor potential cation channel, subfamily V, member 1" (TRPV1) channel blockers in epilepsy treatment. The phytocannabinoids are compounds produced by the herb Cannabis sativa, from which Δ9-tetrahydrocannabinol (Δ9-THC) is the main active compound. The therapeutic applications of Δ9-THC are limited, whereas cannabidiol (CBD), another phytocannabinoid, induces antiepileptic effects in experimental animals and in patients with refractory epilepsies. Synthetic CB1 agonists induce mixed effects, which hamper their therapeutic applications. A more promising strategy focuses on compounds that increase the brain levels of anandamide, an endocannabinoid produced on-demand to counteract hyperexcitability. Thus, anandamide hydrolysis inhibitors might represent a future class of antiepileptic drugs. Finally, compounds that block the TRPV1 ("vanilloid") channel, a possible anandamide target in the brain, have also been investigated. In conclusion, the therapeutic use of phytocannabinoids (CBD) is already in practice, although its mechanisms of action remain unclear. Endocannabinoid and TRPV1 mechanisms warrant further basic studies to support their potential clinical applications. This article is part of the Special Issue "NEWroscience 2018".
Collapse
Affiliation(s)
- Laila Asth
- Graduate School in Physiology and Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Lia P Iglesias
- Graduate School in Neurosciences, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Antônio C De Oliveira
- Graduate School in Physiology and Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil; Graduate School in Neurosciences, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil; Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Marcio F D Moraes
- Graduate School in Physiology and Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil; Graduate School in Neurosciences, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil; Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil
| | - Fabrício A Moreira
- Graduate School in Physiology and Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil; Graduate School in Neurosciences, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil; Department of Pharmacology, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Brazil.
| |
Collapse
|
12
|
Emerging Roles of Cannabinoids and Synthetic Cannabinoids in Clinical Experimental Models. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1264:47-65. [PMID: 33332003 DOI: 10.1007/978-3-030-57369-0_4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In recent years, an increasing number of investigations has demonstrated the therapeutic potential of molecules targeting the endocannabinoid system. Cannabinoids of endogenous, phytogenic, and synthetic nature have been assessed in a wide variety of disease models ranging from neurological to metabolic disorders. Even though very few compounds of this type have already reached the market, numerous preclinical and clinical studies suggest that cannabinoids are suitable drugs for the clinical management of diverse pathologies.In this chapter, we will provide an overview of the endocannabinoid system under certain physiopathological conditions, with a focus on neurological, oncologic, and metabolic disorders. Cannabinoids evaluated as potential therapeutic agents in experimental models with an emphasis in the most successful chemical entities and their perspectives towards the clinic will be discussed.
Collapse
|
13
|
Insights into Potential Targets for Therapeutic Intervention in Epilepsy. Int J Mol Sci 2020; 21:ijms21228573. [PMID: 33202963 PMCID: PMC7697405 DOI: 10.3390/ijms21228573] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/04/2020] [Accepted: 11/11/2020] [Indexed: 02/06/2023] Open
Abstract
Epilepsy is a chronic brain disease that affects approximately 65 million people worldwide. However, despite the continuous development of antiepileptic drugs, over 30% patients with epilepsy progress to drug-resistant epilepsy. For this reason, it is a high priority objective in preclinical research to find novel therapeutic targets and to develop effective drugs that prevent or reverse the molecular mechanisms underlying epilepsy progression. Among these potential therapeutic targets, we highlight currently available information involving signaling pathways (Wnt/β-catenin, Mammalian Target of Rapamycin (mTOR) signaling and zinc signaling), enzymes (carbonic anhydrase), proteins (erythropoietin, copine 6 and complement system), channels (Transient Receptor Potential Vanilloid Type 1 (TRPV1) channel) and receptors (galanin and melatonin receptors). All of them have demonstrated a certain degree of efficacy not only in controlling seizures but also in displaying neuroprotective activity and in modifying the progression of epilepsy. Although some research with these specific targets has been done in relation with epilepsy, they have not been fully explored as potential therapeutic targets that could help address the unsolved issue of drug-resistant epilepsy and develop new antiseizure therapies for the treatment of epilepsy.
Collapse
|
14
|
Lourenço DM, Ribeiro-Rodrigues L, Sebastião AM, Diógenes MJ, Xapelli S. Neural Stem Cells and Cannabinoids in the Spotlight as Potential Therapy for Epilepsy. Int J Mol Sci 2020; 21:E7309. [PMID: 33022963 PMCID: PMC7582633 DOI: 10.3390/ijms21197309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 09/28/2020] [Accepted: 09/30/2020] [Indexed: 01/18/2023] Open
Abstract
Epilepsy is one of the most common brain diseases worldwide, having a huge burden in society. The main hallmark of epilepsy is the occurrence of spontaneous recurrent seizures, having a tremendous impact on the lives of the patients and of their relatives. Currently, the therapeutic strategies are mostly based on the use of antiepileptic drugs, and because several types of epilepsies are of unknown origin, a high percentage of patients are resistant to the available pharmacotherapy, continuing to experience seizures overtime. Therefore, the search for new drugs and therapeutic targets is highly important. One key aspect to be targeted is the aberrant adult hippocampal neurogenesis (AHN) derived from Neural Stem Cells (NSCs). Indeed, targeting seizure-induced AHN may reduce recurrent seizures and shed some light on the mechanisms of disease. The endocannabinoid system is a known modulator of AHN, and due to the known endogenous antiepileptic properties, it is an interesting candidate for the generation of new antiepileptic drugs. However, further studies and clinical trials are required to investigate the putative mechanisms by which cannabinoids can be used to treat epilepsy. In this manuscript, we will review how cannabinoid-induced modulation of NSCs may promote neural plasticity and whether these drugs can be used as putative antiepileptic treatment.
Collapse
Affiliation(s)
- Diogo M. Lourenço
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (D.M.L.); (L.R.-R.); (A.M.S.); (M.J.D.)
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Leonor Ribeiro-Rodrigues
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (D.M.L.); (L.R.-R.); (A.M.S.); (M.J.D.)
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Ana M. Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (D.M.L.); (L.R.-R.); (A.M.S.); (M.J.D.)
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Maria J. Diógenes
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (D.M.L.); (L.R.-R.); (A.M.S.); (M.J.D.)
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Sara Xapelli
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal; (D.M.L.); (L.R.-R.); (A.M.S.); (M.J.D.)
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisboa, Portugal
| |
Collapse
|
15
|
Functional Crosstalk between CB and TRPV1 Receptors Protects Nigrostriatal Dopaminergic Neurons in the MPTP Model of Parkinson's Disease. J Immunol Res 2020; 2020:5093493. [PMID: 33062722 PMCID: PMC7539109 DOI: 10.1155/2020/5093493] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 08/31/2020] [Accepted: 09/08/2020] [Indexed: 01/28/2023] Open
Abstract
The present study examined whether crosstalk between cannabinoid (CB) and transient potential receptor vanilloid type 1 (TRPV1) could contribute to the survival of nigrostriatal dopamine neurons in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of Parkinson's disease (PD). MPTP induced a significant loss of nigrostriatal dopamine neurons and glial activation in the substantia nigra (SN) and striatum (STR) as visualized by tyrosine hydroxylase (TH) or macrophage antigen complex-1 (MAC-1) or glial fibrillary acidic protein (GFAP) immunocytochemistry, respectively. RT-PCR analysis shows the upregulation of inducible nitric oxide synthase, interleukin-1β, and tumor necrosis factor-α in microglia in the SN in vivo, indicating the activation of the inflammatory system. By contrast, treatment with capsaicin (a specific TRPV1 agonist) increased the survival of dopamine neurons in the SN and their fibers and dopamine levels in the STR in MPTP mice. Capsaicin neuroprotection is accompanied by inhibiting MPTP-induced glial activation and production of inflammatory cytokines. Treatment with AM251 and AM630 (CB1/2 antagonists) abolished capsaicin-induced beneficial effects, indicating the existence of a functional crosstalk between CB and TRPV1. Moreover, treatment with anandamide (an endogenous agonist for both CB and TRVP1) rescued nigrostriatal dopamine neurons and reduced gliosis-derived neuroinflammatory responses in MPTP mice. These results suggest that the cannabinoid and vanilloid system may be beneficial for the treatment of neurodegenerative diseases, such as PD, that are associated with neuroinflammation.
Collapse
|
16
|
Roles of TRP Channels in Neurological Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:7289194. [PMID: 32963700 PMCID: PMC7492880 DOI: 10.1155/2020/7289194] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Accepted: 07/02/2020] [Indexed: 11/17/2022]
Abstract
Transient receptor potential (TRP) proteins consist of a superfamily of cation channels that have been involved in diverse physiological processes in the brain as well as in the pathogenesis of neurological disease. TRP channels are widely expressed in the brain, including neurons and glial cells, as well as in the cerebral vascular endothelium and smooth muscle. Members of this channel superfamily show a wide variety of mechanisms ranging from ligand binding to voltage, physical, and chemical stimuli, implying the promising therapeutic potential of TRP in neurological diseases. In this review, we focus on the physiological functions of TRP channels in the brain and the pathological roles in neurological disorders to explore future potential neuroprotective strategies.
Collapse
|
17
|
Jacobs JA, Sehgal A. Anandamide Metabolites Protect against Seizures through the TRP Channel Water Witch in Drosophila melanogaster. Cell Rep 2020; 31:107710. [PMID: 32492422 PMCID: PMC9161705 DOI: 10.1016/j.celrep.2020.107710] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 02/26/2020] [Accepted: 05/07/2020] [Indexed: 02/07/2023] Open
Abstract
Endocannabinoids protect against seizures, but their mechanism of action is still unclear, as they can have effects independent of known cannabinoid receptors. Using Drosophila melanogaster, which lacks canonical cannabinoid receptors, we report that the endocannabinoids anandamide and 2-arachidonoylglycerol protect against seizures in multiple fly seizure models. Surprisingly, inhibition of anandamide catabolism renders flies insensitive to protection by anandamide, indicating that anandamide metabolites are responsible for seizure protection. Consistent with this finding, arachidonic acid, a direct metabolite of anandamide, protects against seizures. To identify downstream effectors, we test for a role of transient receptor potential (TRP) channels and find that the TRPV1 antagonist capsazepine blocks the protective effect of anandamide. Also, a targeted genetic screen of TRP channels identifies water witch as a mediator of protection by anandamide. Using a Drosophila model, we reveal the role of arachidonic acid in seizure protection and identify a cannabinoid-receptor-1/2-independent mechanism of endocannabinoid seizure protection.
Collapse
Affiliation(s)
- Jack A Jacobs
- Pharmacology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Amita Sehgal
- Pharmacology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Chronobiology and Sleep Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Howard Hughes Medical Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
18
|
Stasiulewicz A, Znajdek K, Grudzień M, Pawiński T, Sulkowska JI. A Guide to Targeting the Endocannabinoid System in Drug Design. Int J Mol Sci 2020; 21:ijms21082778. [PMID: 32316328 PMCID: PMC7216112 DOI: 10.3390/ijms21082778] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 04/07/2020] [Accepted: 04/14/2020] [Indexed: 12/11/2022] Open
Abstract
The endocannabinoid system (ECS) is one of the most crucial systems in the human organism, exhibiting multi-purpose regulatory character. It is engaged in a vast array of physiological processes, including nociception, mood regulation, cognitive functions, neurogenesis and neuroprotection, appetite, lipid metabolism, as well as cell growth and proliferation. Thus, ECS proteins, including cannabinoid receptors and their endogenous ligands’ synthesizing and degrading enzymes, are promising therapeutic targets. Their modulation has been employed in or extensively studied as a treatment of multiple diseases. However, due to a complex nature of ECS and its crosstalk with other biological systems, the development of novel drugs turned out to be a challenging task. In this review, we summarize potential therapeutic applications for ECS-targeting drugs, especially focusing on promising synthetic compounds and preclinical studies. We put emphasis on modulation of specific proteins of ECS in different pathophysiological areas. In addition, we stress possible difficulties and risks and highlight proposed solutions. By presenting this review, we point out information pivotal in the spotlight of ECS-targeting drug design, as well as provide an overview of the current state of knowledge on ECS-related pharmacodynamics and show possible directions for needed research.
Collapse
Affiliation(s)
- Adam Stasiulewicz
- Department of Drug Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland; (M.G.); (T.P.)
- Interdisciplinary Laboratory of Biological Systems Modelling, Centre of New Technologies, University of Warsaw, Banacha 2c, 02-097 Warsaw, Poland;
- Correspondence: (A.S.); (J.I.S.)
| | - Katarzyna Znajdek
- Interdisciplinary Laboratory of Biological Systems Modelling, Centre of New Technologies, University of Warsaw, Banacha 2c, 02-097 Warsaw, Poland;
- Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland
| | - Monika Grudzień
- Department of Drug Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland; (M.G.); (T.P.)
| | - Tomasz Pawiński
- Department of Drug Chemistry, Faculty of Pharmacy, Medical University of Warsaw, Banacha 1, 02-097 Warsaw, Poland; (M.G.); (T.P.)
| | - Joanna I. Sulkowska
- Interdisciplinary Laboratory of Biological Systems Modelling, Centre of New Technologies, University of Warsaw, Banacha 2c, 02-097 Warsaw, Poland;
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
- Materials and Process Simulation Center, California Institute of Technology, Pasadena, CA 91125, USA
- Correspondence: (A.S.); (J.I.S.)
| |
Collapse
|
19
|
Günaydın C, Arslan G, Bilge SS. Proconvulsant effect of trans-cinnamaldehyde in pentylenetetrazole-induced kindling model of epilepsy: The role of TRPA1 channels. Neurosci Lett 2020; 721:134823. [PMID: 32035165 DOI: 10.1016/j.neulet.2020.134823] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/28/2020] [Accepted: 02/04/2020] [Indexed: 12/27/2022]
Abstract
The transient receptor potential ankyrin 1 (TRPA1), a member of the TRP superfamily, is widely distributed in the central nervous system (CNS) and plays an important role in pain and inflammation. However, no data has been reported regarding the effects of TRPA1 on epileptic seizures. Thus, this study was designed to investigate the sub-chronic effect of trans-cinnamaldehyde (TCA), an agonist of TRPA1, in pentylenetetrazole (PTZ) induced kindling model via electrocorticography (ECoG). Furthermore, the expressions of cAMP response element binding protein (CREB), brain-derived neurotrophic factor (BDNF), and NMDA receptor subunit NR2B were measured using Western blotting. Rats were kindled by intraperitoneal (i.p.) PTZ (35 mg/kg) injections. After electrode implantation and healing period, 10 and 30 mg/kg TCA was given i.p. for 14 consecutive days. On the next day, ECoG recordings were obtained after the injection of PTZ (35 mg/kg, i.p.), and twenty-four hours later, rats were decapitated for molecular analyses. TCA, at a dose of 30 mg/kg, decreased the first myoclonic jerk latency and increased seizure duration and total spike activity. Additionally, both doses of TCA enhanced CREB, BDNF, and NR2B expressions, which were increased by the kindling. The evidence from this study suggests that long term activation of TRPA1 channels causes an exacerbated seizure activity. Moreover, PTZ-induced increases in CREB, BDNF, and NR2B levels were enhanced by the repeated administrations of TCA.
Collapse
Affiliation(s)
- Caner Günaydın
- Department of Pharmacology, School of Medicine, Ondokuz Mayis University, Samsun, Turkey
| | - Gökhan Arslan
- Department of Physiology, School of Medicine, Ondokuz Mayis University, Samsun, Turkey.
| | - S Sırrı Bilge
- Department of Pharmacology, School of Medicine, Ondokuz Mayis University, Samsun, Turkey
| |
Collapse
|
20
|
Saliba SW, Bonifacino T, Serchov T, Bonanno G, de Oliveira ACP, Fiebich BL. Neuroprotective Effect of AM404 Against NMDA-Induced Hippocampal Excitotoxicity. Front Cell Neurosci 2019; 13:566. [PMID: 31920563 PMCID: PMC6932953 DOI: 10.3389/fncel.2019.00566] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 12/06/2019] [Indexed: 11/13/2022] Open
Abstract
Different studies have demonstrated that inflammation and alterations in glutamate neurotransmission are two events contributing to the pathophysiology of neurodegenerative or neurological disorders. There are evidences that N-arachidonoylphenolamine (AM404), a cannabinoid system modulator and paracetamol metabolite, modulates inflammation and exerts neuroprotective effects on Huntington's (HD) and Parkinson's diseases (PD), and ischemia. However, the effects of AM404 on the production of inflammatory mediators and excitotoxicity in brain tissue stimulated with N-methyl-D-aspartic acid (NMDA) are not elucidated. In this present study, we investigated the effects of AM404 on the production of inflammatory mediators and neuronal cell death induced by NMDA in organotypic hippocampal slices cultures (OHSC) using qPCR, western blot (WB), and immunohistochemistry. Moreover, to comprehend the mechanism of excitotoxicity, we evaluated the effects of AM404 on glutamate release in hippocampal synaptosomes and the NMDA-induced calcium responses in acute hippocampal slices. Our results showed that AM404 led to a significant decrease in cell death induced by NMDA, through a mechanism possibly involving the reduction of glutamate release and the calcium ions responses. Furthermore, it decreased the expression of the interleukin (IL)-1β. This study provides new significant insights about the anti-inflammatory and neuroprotection effects of AM404 on NMDA-induced excitotoxicity. To understand the effects of AM404 in these processes might contribute to the therapeutic potential of AM404 in diseases with involvement of neuroinflammation and neurodegeneration and might lead to a possible future treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Soraya Wilke Saliba
- Neuroimmunology and Neurochemistry Research Group, Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tiziana Bonifacino
- Unit of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy
| | - Tsvetan Serchov
- Laboratory of Stereotaxy and Interventional Neuroscience, Department of Stereotactic and Functional Neurosurgery, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Giambattista Bonanno
- Unit of Pharmacology and Toxicology and Center of Excellence for Biomedical Research, Department of Pharmacy, School of Medical and Pharmaceutical Sciences, University of Genoa, Genoa, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Ospedale Policlinico San Martino, Genoa, Italy
| | | | - Bernd L Fiebich
- Neuroimmunology and Neurochemistry Research Group, Department of Psychiatry and Psychotherapy, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
21
|
Cannabinoids and the expanded endocannabinoid system in neurological disorders. Nat Rev Neurol 2019; 16:9-29. [DOI: 10.1038/s41582-019-0284-z] [Citation(s) in RCA: 320] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2019] [Indexed: 12/13/2022]
|
22
|
Gambino G, Rizzo V, Giglia G, Ferraro G, Sardo P. Cannabinoids, TRPV and nitric oxide: the three ring circus of neuronal excitability. Brain Struct Funct 2019; 225:1-15. [PMID: 31792694 DOI: 10.1007/s00429-019-01992-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 11/22/2019] [Indexed: 12/13/2022]
Abstract
Endocannabinoid system is considered a relevant player in the regulation of neuronal excitability, since it contributes to maintaining the balance of the synaptic ionic milieu. Perturbations to bioelectric conductances have been implicated in the pathophysiological processes leading to hyperexcitability and epileptic seizures. Cannabinoid influence on neurosignalling is exerted on classic receptor-mediated mechanisms or on further molecular targets. Among these, transient receptor potential vanilloid (TRPV) are ionic channels modulated by cannabinoids that are involved in the transduction of a plethora of stimuli and trigger fundamental downstream pathways in the post-synaptic site. In this review, we aim at providing a brief summary of the most recent data about the cross-talk between cannabinoid system and TRPV channels, drawing attention on their role on neuronal hyperexcitability. Then, we aim to unveil a plausible point of interaction between these neural signalling systems taking into consideration nitric oxide, a gaseous molecule inducing profound modifications to neural performances. From this novel perspective, we struggle to propose innovative cellular mechanisms in the regulation of hyperexcitability phenomena, with the goal of exploring plausible CB-related mechanisms underpinning epileptic seizures.
Collapse
Affiliation(s)
- Giuditta Gambino
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy.
| | - Valerio Rizzo
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy
| | - Giuseppe Giglia
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy
| | - Giuseppe Ferraro
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy
| | - Pierangelo Sardo
- Department of Experimental Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Sezione di Fisiologia Umana G. Pagano, University of Palermo, Corso Tukory 129, Palermo, Italy
| |
Collapse
|
23
|
Barrett KT, Roy A, Rivard KB, Wilson RJ, Scantlebury MH. Vagal TRPV1 activation exacerbates thermal hyperpnea and increases susceptibility to experimental febrile seizures in immature rats. Neurobiol Dis 2018; 119:172-189. [DOI: 10.1016/j.nbd.2018.08.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Revised: 07/20/2018] [Accepted: 08/07/2018] [Indexed: 12/22/2022] Open
|
24
|
TRPV1 mediates the anticonvulsant effects of acetaminophen in mice. Epilepsy Res 2018; 145:153-159. [DOI: 10.1016/j.eplepsyres.2018.06.016] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 06/19/2018] [Accepted: 06/30/2018] [Indexed: 12/16/2022]
|
25
|
Sugaya Y, Kano M. Control of excessive neural circuit excitability and prevention of epileptic seizures by endocannabinoid signaling. Cell Mol Life Sci 2018; 75:2793-2811. [PMID: 29737364 PMCID: PMC11105219 DOI: 10.1007/s00018-018-2834-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/23/2018] [Accepted: 05/02/2018] [Indexed: 12/12/2022]
Abstract
Progress in research on endocannabinoid signaling has greatly advanced our understanding of how it controls neural circuit excitability in health and disease. In general, endocannabinoid signaling at excitatory synapses suppresses seizures by inhibiting glutamate release. In contrast, endocannabinoid signaling promotes seizures by inhibiting GABA release at inhibitory synapses. The physiological distribution of endocannabinoid signaling molecules becomes disrupted with the development of epileptic focus in patients with mesial temporal lobe epilepsy and in animal models of experimentally induced epilepsy. Augmentation of endocannabinoid signaling can promote the development of epileptic focus at initial stages. However, at later stages, increased endocannabinoid signaling delays it and suppresses spontaneous seizures. Thus, the regulation of endocannabinoid signaling at specific synapses that cause hyperexcitability during particular stages of disease development may be effective for treating epilepsy and epileptogenesis.
Collapse
Affiliation(s)
- Yuki Sugaya
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Tokyo, 113-0033, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, 113-0033, Japan
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, 7-3-1 Hongo, Tokyo, 113-0033, Japan.
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, Tokyo, 113-0033, Japan.
| |
Collapse
|
26
|
Khan AA, Shekh-Ahmad T, Khalil A, Walker MC, Ali AB. Cannabidiol exerts antiepileptic effects by restoring hippocampal interneuron functions in a temporal lobe epilepsy model. Br J Pharmacol 2018; 175:2097-2115. [PMID: 29574880 PMCID: PMC5979781 DOI: 10.1111/bph.14202] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Revised: 12/13/2017] [Accepted: 02/27/2018] [Indexed: 12/29/2022] Open
Abstract
Background and Purpose A non‐psychoactive phytocannabinoid, cannabidiol (CBD), shows promising results as an effective potential antiepileptic drug in some forms of refractory epilepsy. To elucidate the mechanisms by which CBD exerts its anti‐seizure effects, we investigated its effects at synaptic connections and on the intrinsic membrane properties of hippocampal CA1 pyramidal cells and two major inhibitory interneurons: fast spiking, parvalbumin (PV)‐expressing and adapting, cholecystokinin (CCK)‐expressing interneurons. We also investigated whether in vivo treatment with CBD altered the fate of CCK and PV interneurons using immunohistochemistry. Experimental Approach Electrophysiological intracellular whole‐cell recordings combined with neuroanatomy were performed in acute brain slices of rat temporal lobe epilepsy in in vivo (induced by kainic acid) and in vitro (induced by Mg2+‐free solution) epileptic seizure models. For immunohistochemistry experiments, CBD was administered in vivo (100 mg·kg−1) at zero time and 90 min post status epilepticus, induced with kainic acid. Key Results Bath application of CBD (10 μM) dampened excitability at unitary synapses between pyramidal cells but enhanced inhibitory synaptic potentials elicited by fast spiking and adapting interneurons at postsynaptic pyramidal cells. Furthermore, CBD restored impaired membrane excitability of PV, CCK and pyramidal cells in a cell type‐specific manner. These neuroprotective effects of CBD were corroborated by immunohistochemistry experiments that revealed a significant reduction in atrophy and death of PV‐ and CCK‐expressing interneurons after CBD treatment. Conclusions and Implications Our data suggest that CBD restores excitability and morphological impairments in epileptic models to pre‐epilepsy control levels through multiple mechanisms to reinstate normal network function.
Collapse
Affiliation(s)
| | | | | | | | - Afia B Ali
- UCL School of Pharmacy, London, WC1N 1AX, UK
| |
Collapse
|
27
|
Carletti F, Gambino G, Rizzo V, Ferraro G, Sardo P. Neuronal nitric oxide synthase is involved in CB/TRPV1 signalling: Focus on control of hippocampal hyperexcitability. Epilepsy Res 2017; 138:18-25. [DOI: 10.1016/j.eplepsyres.2017.09.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/15/2017] [Accepted: 09/29/2017] [Indexed: 12/29/2022]
|
28
|
Cho SJ, Vaca MA, Miranda CJ, N'Gouemo P. Inhibition of transient potential receptor vanilloid type 1 suppresses seizure susceptibility in the genetically epilepsy-prone rat. CNS Neurosci Ther 2017; 24:18-28. [PMID: 29105300 DOI: 10.1111/cns.12770] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Revised: 09/25/2017] [Accepted: 09/26/2017] [Indexed: 11/30/2022] Open
Abstract
AIMS Intracellular calcium plays an important role in neuronal hyperexcitability that leads to seizures. One calcium influx route of interest is the transient receptor potential vanilloid type 1 (TRPV1) channel. Here, we evaluated the effects of capsazepine (CPZ), a potent blocker of TRPV1 channels on acoustically evoked seizures (audiogenic seizures, AGS) in the genetically epilepsy-prone rat (GEPR-3), a model of inherited epilepsy. METHODS Male and female GEPR-3s were used. For the acute CPZ treatment study, GEPR-3s were tested for AGS susceptibility before and after treatment with various doses of CPZ (0, 1, 3, and 10 mg/kg; ip). For semichronic CPZ treatment study, GEPR-3s were tested for AGS susceptibility before and after 5-day CPZ treatment at the dose of 1 mg/kg (ip). The prevalence, latency, and severity of AGS were recorded and analyzed. RESULTS We found that acute CPZ pretreatment reduced the seizure severity in male GEPR-3s; the effect was dose-dependent. In female GEPR-3s, however, CPZ treatment completely suppressed the seizure susceptibility. Furthermore, semichronic CPZ treatment suppressed seizure susceptibility in female GEPR-3s, but only reduced the seizure severity in male GEPR-3s. CONCLUSIONS These findings suggest that the TRPV1 channel is a promising molecular target for seizure suppression, with female GEPR-3s exhibiting higher sensitivity than male GEPR-3s.
Collapse
Affiliation(s)
- Sue J Cho
- Department of Pediatrics, Georgetown University Medical Center, Washington, DC, USA
| | - Michelle A Vaca
- Department of Pediatrics, Georgetown University Medical Center, Washington, DC, USA
| | - Clive J Miranda
- Department of Pediatrics, Georgetown University Medical Center, Washington, DC, USA
| | - Prosper N'Gouemo
- Department of Pediatrics, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
29
|
Rosenberg EC, Patra PH, Whalley BJ. Therapeutic effects of cannabinoids in animal models of seizures, epilepsy, epileptogenesis, and epilepsy-related neuroprotection. Epilepsy Behav 2017; 70:319-327. [PMID: 28190698 PMCID: PMC5651410 DOI: 10.1016/j.yebeh.2016.11.006] [Citation(s) in RCA: 138] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2016] [Accepted: 11/03/2016] [Indexed: 12/21/2022]
Abstract
The isolation and identification of the discrete plant cannabinoids in marijuana revived interest in analyzing historical therapeutic claims made for cannabis in clinical case studies and anecdotes. In particular, sources as old as the 11th and 15th centuries claimed efficacy for crude marijuana extracts in the treatment of convulsive disorders, prompting a particularly active area of preclinical research into the therapeutic potential of plant cannabinoids in epilepsy. Since that time, a large body of literature has accumulated describing the effects of several of the >100 individual plant cannabinoids in preclinical models of seizures, epilepsy, epileptogenesis, and epilepsy-related neuroprotection. We surveyed the literature for relevant reports of such plant cannabinoid effects and critically reviewed their findings. We found that acute CB1R agonism in simple models of acute seizures in rodents typically produces anti-convulsant effects whereas CB1R antagonists exert converse effects in the same models. However, when the effects of such ligands are examined in more complex models of epilepsy, epileptogenesis and neuroprotection, a less simplistic narrative emerges. Here, the complex interactions between (i) brain regions involved in a given model, (ii) relative contributions of endocannabinoid signaling to modulation of synaptic transmission in such areas, (iii) multi-target effects, (iv) cannabinoid type 1 and type 2 receptor signaling interactions and, (v) timing, (vi) duration and (vii) localization of ligand administration suggest that there is both anti-epileptic therapeutic potential and a pro-epileptic risk in up- and down-regulation of endocannabinoid signaling in the central nervous system. Factors such receptor desensitization and specific pharmacology of ligands used (e.g. full vs partial agonists and neutral antagonists vs inverse agonists) also appear to play an important role in the effects reported. Furthermore, the effects of several plant cannabinoids, most notably cannabidiol (CBD) and cannabidavarin (CBDV), in models of seizures, epilepsy, epileptogenesis, and neuroprotection are less ambiguous, and consistent with reports of therapeutically beneficial effects of these compounds in clinical studies. However, continued paucity of firm information regarding the therapeutic molecular mechanism of CBD/CBDV highlights the continued need for research in this area in order to identify as yet under-exploited targets for drug development and raise our understanding of treatment-resistant epilepsies. The recent reporting of positive results for cannabidiol treatment in two Phase III clinical trials in treatment-resistant epilepsies provides pivotal evidence of clinical efficacy for one plant cannabinoid in epilepsy. Moreover, risks and/or benefits associated with the use of unlicensed Δ9-THC containing marijuana extracts in pediatric epilepsies remain poorly understood. Therefore, in light of these paradigm-changing clinical events, the present review's findings aim to drive future drug development for newly-identified targets and indications, identify important limitations of animal models in the investigation of plant cannabinoid effects in the epilepsies, and focuses future research in this area on specific, unanswered questions regarding the complexities of endocannabinoid signaling in epilepsy. This article is part of a Special Issue titled Cannabinoids and Epilepsy.
Collapse
Affiliation(s)
- Evan C. Rosenberg
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Langone Medical Center, New York, NY 10016, USA
| | - Pabitra H. Patra
- Department of Pharmacy, School of Chemistry, Food & Nutritional Sciences and Pharmacy, University of Reading, Whiteknights, Reading, Berkshire RG6 6AP, UK
| | - Benjamin J. Whalley
- Department of Pharmacy, School of Chemistry, Food & Nutritional Sciences and Pharmacy, University of Reading, Whiteknights, Reading, Berkshire RG6 6AP, UK,Corresponding author: (B.J. Whalley)
| |
Collapse
|
30
|
Shubina L, Aliev R, Kitchigina V. Endocannabinoid-dependent protection against kainic acid-induced long-term alteration of brain oscillations in guinea pigs. Brain Res 2017; 1661:1-14. [DOI: 10.1016/j.brainres.2017.02.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 02/02/2017] [Accepted: 02/03/2017] [Indexed: 01/12/2023]
|
31
|
Manjarrez-Marmolejo J, Franco-Pérez J. Gap Junction Blockers: An Overview of their Effects on Induced Seizures in Animal Models. Curr Neuropharmacol 2017; 14:759-71. [PMID: 27262601 PMCID: PMC5050393 DOI: 10.2174/1570159x14666160603115942] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 02/26/2016] [Accepted: 04/21/2016] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Gap junctions are clusters of intercellular channels allowing the bidirectional pass of ions directly into the cytoplasm of adjacent cells. Electrical coupling mediated by gap junctions plays a role in the generation of highly synchronized electrical activity. The hypersynchronous neuronal activity is a distinctive characteristic of convulsive events. Therefore, it has been postulated that enhanced gap junctional communication is an underlying mechanism involved in the generation and maintenance of seizures. There are some chemical compounds characterized as gap junction blockers because of their ability to disrupt the gap junctional intercellular communication. OBJECTIVE Hence, the aim of this review is to analyze the available data concerning the effects of gap junction blockers specifically in seizure models. RESULTS Carbenoxolone, quinine, mefloquine, quinidine, anandamide, oleamide, heptanol, octanol, meclofenamic acid, niflumic acid, flufenamic acid, glycyrrhetinic acid and retinoic acid have all been evaluated on animal seizure models. In vitro, these compounds share anticonvulsant effects typically characterized by the reduction of both amplitude and frequency of the epileptiform activity induced in brain slices. In vivo, gap junction blockers modify the behavioral parameters related to seizures induced by 4-aminopyridine, pentylenetetrazole, pilocarpine, penicillin and maximal electroshock. CONCLUSION Although more studies are still required, these molecules could be a promising avenue in the search for new pharmaceutical alternatives for the treatment of epilepsy.
Collapse
Affiliation(s)
| | - Javier Franco-Pérez
- Laboratory of Physiology of Reticular Formation, National Institute of Neurology and Neurosurgery, M.V.S. Insurgentes Sur 3877, Col. La Fama, C.P. 14269, Mexico D.F., Mexico
| |
Collapse
|
32
|
Carletti F, Gambino G, Rizzo V, Ferraro G, Sardo P. Involvement of TRPV1 channels in the activity of the cannabinoid WIN 55,212-2 in an acute rat model of temporal lobe epilepsy. Epilepsy Res 2016; 122:56-65. [PMID: 26970948 DOI: 10.1016/j.eplepsyres.2016.02.005] [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: 09/28/2015] [Revised: 01/18/2016] [Accepted: 02/09/2016] [Indexed: 10/21/2022]
Abstract
The exogenous cannabinoid agonist WIN 55,212-2, (R)-(+)-[2,3-dihydro-5-methyl-3-(4-morpholinylmethyl) pyrrolo[1,2,3-de]-1,4-benzoxazin-6-Yl]-1-naphthalenylmethanone (WIN), has revealed to play a role on modulating the hyperexcitability phenomena in the hippocampus. Cannabinoid-mediated mechanisms of neuroprotection have recently been found to imply the modulation of transient receptor potential vanilloid 1 (TRPV1), a cationic channel subfamily that regulate synaptic excitation. In our study, we assessed the influence of pharmacological manipulation of TRPV1 function, alone and on WIN antiepileptic activity, in the Maximal Dentate Activation (MDA) acute model of temporal lobe epilepsy. Our results showed that the TRPV1 agonist, capsaicin, increased epileptic outcomes; whilst antagonizing TRPV1 with capsazepine exerts a protective role on paroxysmal discharge. When capsaicin is co-administered with WIN effective dose of 10mg/kg is able to reduce its antiepileptic strength, especially on the triggering of MDA response. Accordingly, capsazepine at the protective dose of 2mg/kg managed to potentiate WIN antiepileptic effects, when co-treated. Moreover, WIN subeffective dose of 5mg/kg was turned into effective when capsazepine comes into play. This evidence suggests that systemic administration of TRPV1-active drugs influences electrically induced epilepsy, with a noticeable protective activity for capsazepine. Furthermore, results from the pharmacological interaction with WIN support an interplay between cannabinoid and TRPV1 signaling that could represent a promising approach for a future pharmacological strategy to challenge hyperexcitability-based diseases.
Collapse
Affiliation(s)
- Fabio Carletti
- Department of "Biomedicina Sperimentale e Neuroscienze Cliniche" (Bio.Ne.C.), "Sezione di Fisiologia umana G. Pagano", University of Palermo, Corso Tukory 129-90134 Palermo, Italy.
| | - Giuditta Gambino
- Department of "Biomedicina Sperimentale e Neuroscienze Cliniche" (Bio.Ne.C.), "Sezione di Fisiologia umana G. Pagano", University of Palermo, Corso Tukory 129-90134 Palermo, Italy.
| | - Valerio Rizzo
- Department of "Biomedicina Sperimentale e Neuroscienze Cliniche" (Bio.Ne.C.), "Sezione di Fisiologia umana G. Pagano", University of Palermo, Corso Tukory 129-90134 Palermo, Italy.
| | - Giuseppe Ferraro
- Department of "Biomedicina Sperimentale e Neuroscienze Cliniche" (Bio.Ne.C.), "Sezione di Fisiologia umana G. Pagano", University of Palermo, Corso Tukory 129-90134 Palermo, Italy.
| | - Pierangelo Sardo
- Department of "Biomedicina Sperimentale e Neuroscienze Cliniche" (Bio.Ne.C.), "Sezione di Fisiologia umana G. Pagano", University of Palermo, Corso Tukory 129-90134 Palermo, Italy.
| |
Collapse
|
33
|
Su LY, Li H, Lv L, Feng YM, Li GD, Luo R, Zhou HJ, Lei XG, Ma L, Li JL, Xu L, Hu XT, Yao YG. Melatonin attenuates MPTP-induced neurotoxicity via preventing CDK5-mediated autophagy and SNCA/α-synuclein aggregation. Autophagy 2016; 11:1745-59. [PMID: 26292069 DOI: 10.1080/15548627.2015.1082020] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Autophagy is involved in the pathogenesis of neurodegenerative diseases including Parkinson disease (PD). However, little is known about the regulation of autophagy in neurodegenerative process. In this study, we characterized aberrant activation of autophagy induced by neurotoxin 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) and demonstrated that melatonin has a protective effect on neurotoxicity. We found an excessive activation of autophagy in monkey brain tissues and C6 cells, induced by MPTP, which is mediated by CDK5 (cyclin-dependent kinase 5). MPTP treatment significantly reduced total dendritic length and dendritic complexity of cultured primary cortical neurons and melatonin could reverse this effect. Decreased TH (tyrosine hydroxylase)-positive cells and dendrites of dopaminergic neurons in the substantia nigra pars compacta (SNc) were observed in MPTP-treated monkeys and mice. Along with decreased TH protein level, we observed an upregulation of CDK5 and enhanced autophagic activity in the striatum of mice with MPTP injection. These changes could be salvaged by melatonin treatment or knockdown of CDK5. Importantly, melatonin or knockdown of CDK5 reduced MPTP-induced SNCA/α-synuclein aggregation in mice, which is widely thought to trigger the pathogenesis of PD. Finally, melatonin or knockdown of CDK5 counteracted the PD phenotype in mice induced by MPTP. Our findings uncover a potent role of CDK5-mediated autophagy in the pathogenesis of PD, and suggest that control of autophagic pathways may provide an important clue for exploring potential target for novel therapeutics of PD.
Collapse
Affiliation(s)
- Ling-Yan Su
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China.,b Kunming College of Life Science; University of Chinese Academy of Sciences ; Kunming, Yunnan , China
| | - Hao Li
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China.,b Kunming College of Life Science; University of Chinese Academy of Sciences ; Kunming, Yunnan , China
| | - Li Lv
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China.,b Kunming College of Life Science; University of Chinese Academy of Sciences ; Kunming, Yunnan , China
| | - Yue-Mei Feng
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China
| | - Guo-Dong Li
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China.,b Kunming College of Life Science; University of Chinese Academy of Sciences ; Kunming, Yunnan , China.,d School of Life Science; Anhui University ; Hefei, Anhui , China
| | - Rongcan Luo
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China
| | - He-Jiang Zhou
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China
| | - Xiao-Guang Lei
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China
| | - Liang Ma
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China.,b Kunming College of Life Science; University of Chinese Academy of Sciences ; Kunming, Yunnan , China
| | - Jia-Li Li
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China.,c Kunming Primate Research Center of the Chinese Academy of Sciences; Kunming Institute of Zoology; Chinese Academy of Sciences ; Kunming, Yunnan , China
| | - Lin Xu
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China.,e CAS Center for Excellence in Brain Science and Intelligence Technology; Chinese Academy of Sciences ; Shanghai , China
| | - Xin-Tian Hu
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China.,c Kunming Primate Research Center of the Chinese Academy of Sciences; Kunming Institute of Zoology; Chinese Academy of Sciences ; Kunming, Yunnan , China.,e CAS Center for Excellence in Brain Science and Intelligence Technology; Chinese Academy of Sciences ; Shanghai , China
| | - Yong-Gang Yao
- a Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences; Kunming Institute of Zoology ; Kunming, Yunnan , China.,b Kunming College of Life Science; University of Chinese Academy of Sciences ; Kunming, Yunnan , China.,c Kunming Primate Research Center of the Chinese Academy of Sciences; Kunming Institute of Zoology; Chinese Academy of Sciences ; Kunming, Yunnan , China.,e CAS Center for Excellence in Brain Science and Intelligence Technology; Chinese Academy of Sciences ; Shanghai , China
| |
Collapse
|
34
|
de Carvalho CR, Hoeller AA, Franco PLC, Martini APS, Soares FMS, Lin K, Prediger RD, Whalley BJ, Walz R. The cannabinoid CB2 receptor-specific agonist AM1241 increases pentylenetetrazole-induced seizure severity in Wistar rats. Epilepsy Res 2016; 127:160-167. [PMID: 27608434 DOI: 10.1016/j.eplepsyres.2016.08.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 08/02/2016] [Accepted: 08/12/2016] [Indexed: 11/25/2022]
Abstract
The potential efficacy of cannabinoid receptor ligands for the treatment of epilepsy remains controversial; cannabis components that act via cannabinoid type 1 (CB1) receptors produce anticonvulsant effects in animal models despite treatment with the CB receptor agonist reliably inducing convulsions in various species. Moreover, the potential role of cannabinoid receptor type 2 (CB2) to modulate seizures remains under-investigated. This study assessed the effects of the selective CB2 receptor agonist, AM1241, on pentylenetetrazole (PTZ)-induced seizures in rats. A stereotactically placed guide cannula was surgically implanted into the right lateral ventricle in adult Wistar rats which, 5-6days later, received an acute intracerebroventricular (i.c.v.) microinfusion of AM1241 (0.01, 1 or 10μg/2μl or vehicle) 5min before intraperitoneal (i.p.) injection of PTZ (70mg/kg). Rats were observed for 30min and the seizure severity behavior measured using a modified Racine's scale. Additional groups of rats were pretreated with a single low dose of the selective CB2 receptor antagonist, AM630 (dose 1mg/kg; i.p.), or vehicle, 30min prior to i.c.v. microinfusion of AM1241 (1μg/2μl). AM1241 administration significantly increased tonic-clonic seizure incidence and severity while also decreasing the onset of generalized seizures (AM1241 1 and 10μg/2μl). Pretreatment with AM630 prevented the proconvulsant effects of AM1241. This study shows, for the first time, that selective activation of CB2 receptors can increase generalized seizure susceptibility and suggests that pathological hyperexcitability phenomena can be differentially regulated by targeting CB1 and CB2 receptors.
Collapse
Affiliation(s)
- Cristiane R de Carvalho
- Programa de Pós- graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Centro de Neurociências Aplicadas, Hospital Universitário, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.
| | - Alexandre A Hoeller
- Centro de Neurociências Aplicadas, Hospital Universitário, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Programa de Pós- graduação em Ciências Médicas, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Pedro L C Franco
- Centro de Neurociências Aplicadas, Hospital Universitário, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Athos P S Martini
- Centro de Neurociências Aplicadas, Hospital Universitário, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Flávia M S Soares
- Centro de Neurociências Aplicadas, Hospital Universitário, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Programa de Pós- graduação em Ciências Médicas, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Katia Lin
- Centro de Neurociências Aplicadas, Hospital Universitário, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Programa de Pós- graduação em Ciências Médicas, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Serviço de Neurologia, Departamento de Clínica Médica, Hospital Universitário, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Rui D Prediger
- Centro de Neurociências Aplicadas, Hospital Universitário, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Departamento de Farmacologia, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| | - Benjamin J Whalley
- School of Chemistry, Food and Nutritional Sciences, and Pharmacy, The University of Reading, Reading, Berkshire RG6 6AP, UK
| | - Roger Walz
- Programa de Pós- graduação em Neurociências, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Centro de Neurociências Aplicadas, Hospital Universitário, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Programa de Pós- graduação em Ciências Médicas, Centro de Ciências da Saúde, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil; Serviço de Neurologia, Departamento de Clínica Médica, Hospital Universitário, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil
| |
Collapse
|
35
|
Expression of TRPV1 Receptors Increased in Hippocampus Following Pentylenetetrazole-Induced Kindling in Male Rats. ARCHIVES OF NEUROSCIENCE 2016. [DOI: 10.5812/archneurosci.35459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
36
|
Iannotti FA, Di Marzo V, Petrosino S. Endocannabinoids and endocannabinoid-related mediators: Targets, metabolism and role in neurological disorders. Prog Lipid Res 2016; 62:107-28. [DOI: 10.1016/j.plipres.2016.02.002] [Citation(s) in RCA: 235] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 01/25/2016] [Accepted: 02/26/2016] [Indexed: 12/19/2022]
|
37
|
Saffarzadeh F, Eslamizade M, Mousavi S, Abraki S, Hadjighassem M, Gorji A. TRPV1 receptors augment basal synaptic transmission in CA1 and CA3 pyramidal neurons in epilepsy. Neuroscience 2016; 314:170-8. [DOI: 10.1016/j.neuroscience.2015.11.045] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 10/06/2015] [Accepted: 11/19/2015] [Indexed: 01/13/2023]
|
38
|
Abstract
Cannabis has been used for centuries to treat seizures. Recent anecdotal reports, accumulating animal model data, and mechanistic insights have raised interest in cannabis-based antiepileptic therapies. In this study, we review current understanding of the endocannabinoid system, characterize the pro- and anticonvulsive effects of cannabinoids [e.g., Δ9-tetrahydrocannabinol and cannabidiol (CBD)], and highlight scientific evidence from pre-clinical and clinical trials of cannabinoids in epilepsy. These studies suggest that CBD avoids the psychoactive effects of the endocannabinoid system to provide a well-tolerated, promising therapeutic for the treatment of seizures, while whole-plant cannabis can both contribute to and reduce seizures. Finally, we discuss results from a new multicenter, open-label study using CBD in a population with treatment-resistant epilepsy. In all, we seek to evaluate our current understanding of cannabinoids in epilepsy and guide future basic science and clinical studies.
Collapse
Affiliation(s)
- Evan C Rosenberg
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Langone Medical Center, New York, NY, 10016, USA
| | - Richard W Tsien
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Langone Medical Center, New York, NY, 10016, USA
| | - Benjamin J Whalley
- School of Pharmacy, The University of Reading, Whiteknights, Reading, RG6 6AP, UK
| | - Orrin Devinsky
- Department of Neurology, Comprehensive Epilepsy Center, New York University School of Medicine, New York, NY, 10016, UK.
| |
Collapse
|
39
|
Ibeas Bih C, Chen T, Nunn AVW, Bazelot M, Dallas M, Whalley BJ. Molecular Targets of Cannabidiol in Neurological Disorders. Neurotherapeutics 2015; 12:699-730. [PMID: 26264914 PMCID: PMC4604182 DOI: 10.1007/s13311-015-0377-3] [Citation(s) in RCA: 384] [Impact Index Per Article: 42.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Cannabis has a long history of anecdotal medicinal use and limited licensed medicinal use. Until recently, alleged clinical effects from anecdotal reports and the use of licensed cannabinoid medicines are most likely mediated by tetrahydrocannabinol by virtue of: 1) this cannabinoid being present in the most significant quantities in these preparations; and b) the proportion:potency relationship between tetrahydrocannabinol and other plant cannabinoids derived from cannabis. However, there has recently been considerable interest in the therapeutic potential for the plant cannabinoid, cannabidiol (CBD), in neurological disorders but the current evidence suggests that CBD does not directly interact with the endocannabinoid system except in vitro at supraphysiological concentrations. Thus, as further evidence for CBD's beneficial effects in neurological disease emerges, there remains an urgent need to establish the molecular targets through which it exerts its therapeutic effects. Here, we conducted a systematic search of the extant literature for original articles describing the molecular pharmacology of CBD. We critically appraised the results for the validity of the molecular targets proposed. Thereafter, we considered whether the molecular targets of CBD identified hold therapeutic potential in relevant neurological diseases. The molecular targets identified include numerous classical ion channels, receptors, transporters, and enzymes. Some CBD effects at these targets in in vitro assays only manifest at high concentrations, which may be difficult to achieve in vivo, particularly given CBD's relatively poor bioavailability. Moreover, several targets were asserted through experimental designs that demonstrate only correlation with a given target rather than a causal proof. When the molecular targets of CBD that were physiologically plausible were considered for their potential for exploitation in neurological therapeutics, the results were variable. In some cases, the targets identified had little or no established link to the diseases considered. In others, molecular targets of CBD were entirely consistent with those already actively exploited in relevant, clinically used, neurological treatments. Finally, CBD was found to act upon a number of targets that are linked to neurological therapeutics but that its actions were not consistent withmodulation of such targets that would derive a therapeutically beneficial outcome. Overall, we find that while >65 discrete molecular targets have been reported in the literature for CBD, a relatively limited number represent plausible targets for the drug's action in neurological disorders when judged by the criteria we set. We conclude that CBD is very unlikely to exert effects in neurological diseases through modulation of the endocannabinoid system. Moreover, a number of other molecular targets of CBD reported in the literature are unlikely to be of relevance owing to effects only being observed at supraphysiological concentrations. Of interest and after excluding unlikely and implausible targets, the remaining molecular targets of CBD with plausible evidence for involvement in therapeutic effects in neurological disorders (e.g., voltage-dependent anion channel 1, G protein-coupled receptor 55, CaV3.x, etc.) are associated with either the regulation of, or responses to changes in, intracellular calcium levels. While no causal proof yet exists for CBD's effects at these targets, they represent the most probable for such investigations and should be prioritized in further studies of CBD's therapeutic mechanism of action.
Collapse
Affiliation(s)
- Clementino Ibeas Bih
- School of Chemistry, Food and Nutritional Sciences, and Pharmacy, University of Reading, Whiteknights, Reading, RG6 6AP, UK
| | - Tong Chen
- School of Chemistry, Food and Nutritional Sciences, and Pharmacy, University of Reading, Whiteknights, Reading, RG6 6AP, UK
| | | | - Michaël Bazelot
- School of Chemistry, Food and Nutritional Sciences, and Pharmacy, University of Reading, Whiteknights, Reading, RG6 6AP, UK
- GW Pharmaceuticals Ltd, Sovereign House, Vision Park, Chivers Way, Histon, Cambridge, CB24 9BZ, UK
| | - Mark Dallas
- School of Chemistry, Food and Nutritional Sciences, and Pharmacy, University of Reading, Whiteknights, Reading, RG6 6AP, UK
| | - Benjamin J Whalley
- School of Chemistry, Food and Nutritional Sciences, and Pharmacy, University of Reading, Whiteknights, Reading, RG6 6AP, UK.
| |
Collapse
|
40
|
Alterations in CA1 pyramidal neuronal intrinsic excitability mediated by Ih channel currents in a rat model of amyloid beta pathology. Neuroscience 2015; 305:279-92. [PMID: 26254243 DOI: 10.1016/j.neuroscience.2015.07.087] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 07/28/2015] [Accepted: 07/31/2015] [Indexed: 01/05/2023]
Abstract
Amyloid beta (Aβ) accumulation plays an important role in the pathogenesis of Alzheimer's disease (AD) by changing the neuronal excitability. However, the cellular mechanisms by which accumulation of Aβ affects intrinsic neuronal properties are not well understood. The effect of bilateral intra-frontal cortex Aβ (1-42) peptide injection on the intrinsic excitability of hippocampal CA1 pyramidal neurons with particular focus on the contribution of hyperpolarization-activated (Ih) channel currents was examined using whole-cell patch-clamp recording. Passive avoidance memory impairment and morphological changes in rats receiving intra-frontal Aβ treatment were observed, which was associated with significant changes both in passive and active intrinsic electrical membrane properties of CA1 pyramidal neurons. Electrophysiological recording showed a significant decrease in neuronal excitability associated with an augmentation in the first spike after-hyperpolarization (AHP) amplitude. In addition, the depolarizing sag voltage was altered in neurons recorded from Aβ-treated group. In voltage-clamp condition, a hyperpolarizing activated inward current sensitive to ZD7288 and capsaicin was significantly increased in neurons from Aβ-treated rats. The Ih current density was increased and the activation curve was shifted toward less negative potential in the Aβ-treated group as compared to control group. The enhancing effect of Aβ treatment on Ih current was confirmed by showing upregulation of the mRNA of HCN1 channel in the CA1 pyramidal layer of hippocampi. These findings suggest the contribution of Ih and possibly TRPV1 channel currents to the changes induced by Aβ treatment in the intrinsic membrane properties, which, in turn, may provide therapeutic targets for treatment of AD.
Collapse
|
41
|
Saffarzadeh F, Eslamizade MJ, Ghadiri T, Modarres Mousavi SM, Hadjighassem M, Gorji A. Effects of TRPV1 on the hippocampal synaptic plasticity in the epileptic rat brain. Synapse 2015; 69:375-83. [PMID: 25967571 DOI: 10.1002/syn.21825] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Revised: 04/04/2015] [Accepted: 04/20/2015] [Indexed: 12/27/2022]
Abstract
Temporal lobe epilepsy is often presented by medically intractable recurrent seizures due to dysfunction of temporal lobe structures, mostly the temporomesial structures. The role of transient receptor potential vaniloid 1 (TRPV1) activity on synaptic plasticity of the epileptic brain tissues was investigated. We studied hippocampal TRPV1 protein content and distribution in the hippocampus of epileptic rats. Furthermore, the effects of pharmacologic modulation of TRPV1 receptors on field excitatory postsynaptic potentials have been analyzed after induction of long term potentiation (LTP) in the hippocampal CA1 and CA3 areas after 1 day (acute phase) and 3 months (chronic phase) of pilocarpine-induced status epilepticus (SE). A higher expression of TRPV1 protein in the hippocampus as well as a higher distribution of this channel in CA1 and CA3 areas in both acute and chronic phases of pilocarpine-induced SE was observed. Activation of TRPV1 using capsaicin (1 µM) enhanced LTP induction in CA1 region in non-epileptic rats. Inhibition of TRPV1 by capsazepine (10 µM) did not affect LTP induction in non-epileptic rats. In acute phase of SE, activation of TRPV1 enhanced LTP in both CA1 and CA3 areas but TRPV1 inhibition did not affect LTP. In chronic phase of SE, application of TRPV1 antagonist enhanced LTP induction in CA1 and CA3 regions but TRPV1 activation had no effect on LTP. These findings indicate that a higher expression of TRPV1 in epileptic conditions is accompanied by a functional impact on the synaptic plasticity in the hippocampus. This suggests TRPV1 as a potential target in treatment of seizure attacks.
Collapse
Affiliation(s)
- Fatemeh Saffarzadeh
- Department of Neuroscience, School of Medical Advanced Studies, Iran University of Medical Sciences, Tehran, Iran.,Shefa Neuroscience Research Center, Khatam-Al-Anbia Hospital, Tehran, Iran
| | - Mohammad J Eslamizade
- Department of Neuroscience, School of Medical Advanced Studies, Iran University of Medical Sciences, Tehran, Iran.,Shefa Neuroscience Research Center, Khatam-Al-Anbia Hospital, Tehran, Iran.,Department of Physiology and Neurophysiology Research Center, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Tahereh Ghadiri
- Shefa Neuroscience Research Center, Khatam-Al-Anbia Hospital, Tehran, Iran
| | | | - Mahmoudreza Hadjighassem
- Department of Neuroscience, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Gorji
- Shefa Neuroscience Research Center, Khatam-Al-Anbia Hospital, Tehran, Iran.,Klinik Und Poliklinik Für Neurochirurgie, Department of Neurology, Epilepsy Research Center, Westfälische Wilhelms-Universität Münster, Germany
| |
Collapse
|
42
|
Nazıroğlu M, Övey İ. Involvement of apoptosis and calcium accumulation through TRPV1 channels in neurobiology of epilepsy. Neuroscience 2015; 293:55-66. [DOI: 10.1016/j.neuroscience.2015.02.041] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 02/15/2015] [Accepted: 02/20/2015] [Indexed: 12/20/2022]
|
43
|
Socała K, Nieoczym D, Pieróg M, Wlaź P. α-Spinasterol, a TRPV1 receptor antagonist, elevates the seizure threshold in three acute seizure tests in mice. J Neural Transm (Vienna) 2015; 122:1239-47. [PMID: 25764210 PMCID: PMC4540766 DOI: 10.1007/s00702-015-1391-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Accepted: 03/05/2015] [Indexed: 12/17/2022]
Abstract
α-Spinasterol is a plant-derived compound which was reported to act as a selective antagonist for the transient receptor potential vanilloid 1 (TRPV1) receptor. Several studies revealed that the TRPV1 receptors might modulate seizure activity in animal models of seizures and epilepsy. The aim of the present study was to investigate the effect of α-spinasterol on the seizure threshold in three acute models of seizures, i.e., in the intravenous (i.v.) pentylenetetrazole (PTZ) seizure test, in the maximal electroshock seizure threshold (MEST) test and in the model of psychomotor seizures induced by 6 Hz stimulation in mice. Our results revealed significant anticonvulsant effect of α-spinasterol in all the used seizure tests. In the i.v. PTZ test, statistically significant elevation was noted in case of the threshold for myoclonic twitches (doses of 0.1–1 mg/kg) and generalized clonus seizures (doses of 0.5 and 1 mg/kg) but not for tonic seizures. The studied TRPV1 antagonist also increased the threshold for tonic hindlimb extension in the MEST (doses of 0.5 and 1 mg/kg) and 6 Hz psychomotor seizure (doses of 0.1 and 0.5 mg/kg) tests in mice. Furthermore, α-spinasterol did not produce any significant impairment of motor coordination (assessed in the chimney test) and muscular strength (investigated in the grip-strength test) and it did not provoke significant changes in body temperature in mice. Based on the results of our study and the fact that α-spinasterol is characterized by good blood–brain permeability, we postulate further investigation of this compound to precisely evaluate mechanism of its anticonvulsant action and opportunity of its usage in clinical practice.
Collapse
Affiliation(s)
- Katarzyna Socała
- Department of Animal Physiology, Institute of Biology and Biochemistry, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033, Lublin, Poland,
| | | | | | | |
Collapse
|
44
|
Jia YF, Li YC, Tang YP, Cao J, Wang LP, Yang YX, Xu L, Mao RR. Interference of TRPV1 function altered the susceptibility of PTZ-induced seizures. Front Cell Neurosci 2015; 9:20. [PMID: 25713512 PMCID: PMC4322730 DOI: 10.3389/fncel.2015.00020] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 01/13/2015] [Indexed: 01/21/2023] Open
Abstract
Transient receptor potential vanilloid 1 (TRPV1) is widely distributed in the central nervous system (CNS) including hippocampus, and regulates the balance of excitation and inhibition in CNS, which imply its important role in epilepsy. We used both pharmacological manipulations and transgenic mice to disturb the function of TRPV1 and then studied the effects of these alterations on the susceptibility of pentylenetetrazol (PTZ)-induced seizures. Our results showed that systemic administration of TRPV1 agonist capsaicin (CAP, 40 mg/kg) directly induced tonic-clonic seizures (TCS) without PTZ induction. The severity of seizure was increased in lower doses of CAP groups (5 and 10 mg/kg), although the latency to TCS was delayed. On the other hand, systemic administration of TRPV1 antagonist capsazepine (CPZ, 0.05 and 0.5 mg/kg) and TRPV1 knockout mice exhibited delayed latency to TCS and reduced mortality. Furthermore, hippocampal administration of CPZ (10 and 33 nmol/μL/side) was firstly reported to increase the latency to TCS, decrease the maximal grade of seizure and mortality. It is worth noting that decreased susceptibility of PTZ-induced seizures was observed in hippocampal TRPV1 overexpression mice and hippocampal CAP administration (33 nmol/μL/side), which is opposite from results of systemic agonist CAP. Our findings suggest that the systemic administration of TRPV1 antagonist may be a novel therapeutic target for epilepsy, and alteration of hippocampal TRPV1 function exerts a critical role in seizure susceptibility.
Collapse
Affiliation(s)
- Yun-Fang Jia
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Disease, and Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences Yunnan, China ; Kunming College of Life Science, University of Chinese Academy of Sciences Beijing, China
| | - Ying-Chao Li
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Disease, and Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences Yunnan, China ; School of Life Sciences, University of Science and Technology of China Hefei, China
| | - Yan-Ping Tang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Disease, and Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences Yunnan, China ; Kunming College of Life Science, University of Chinese Academy of Sciences Beijing, China
| | - Jun Cao
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Disease, and Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences Yunnan, China ; Kunming College of Life Science, University of Chinese Academy of Sciences Beijing, China
| | - Li-Ping Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Disease, and Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences Yunnan, China ; Kunming College of Life Science, University of Chinese Academy of Sciences Beijing, China
| | - Yue-Xiong Yang
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Disease, and Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences Yunnan, China ; Kunming College of Life Science, University of Chinese Academy of Sciences Beijing, China
| | - Lin Xu
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Disease, and Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences Yunnan, China ; Kunming College of Life Science, University of Chinese Academy of Sciences Beijing, China
| | - Rong-Rong Mao
- Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences & Yunnan Province, and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Disease, and Laboratory of Learning and Memory, Kunming Institute of Zoology, Chinese Academy of Sciences Yunnan, China ; Kunming College of Life Science, University of Chinese Academy of Sciences Beijing, China
| |
Collapse
|
45
|
Attenuation of kainic acid-induced status epilepticus by inhibition of endocannabinoid transport and degradation in guinea pigs. Epilepsy Res 2015; 111:33-44. [PMID: 25769371 DOI: 10.1016/j.eplepsyres.2015.01.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 11/26/2014] [Accepted: 01/14/2015] [Indexed: 02/08/2023]
Abstract
Status epilepticus (SE) is a medical emergency associated with a high rate of mortality if not treated promptly. Exogenous and endogenous cannabinoids have been shown to possess anticonvulsant properties both in vivo and in vitro. Here we study the influence of endocannabinoid metabolism on the development of kainic acid-induced SE in guinea pigs. For this purpose, the inhibitors of endocannabinoid transport, AM404, and enzymatic (fatty acid amide hydrolase) degradation, URB597, were applied. Cannabinoid CB1 receptor antagonist, AM251, was also tested. Animal behavior as well as local electric field potentials in four structures: medial septum, hippocampus, entorhinal cortex and amygdala were analyzed when AM404 (120nmol), URB597 (4.8nmol) or AM251 (20nmol) were administrated alone or together with 0.4μg of kainic acid. All substances were injected i.c.v. AM404, URB597 or AM251 administered alone did not alter markedly local field potentials of all four studied structures in the long-term compared with their basal activity. AM404 and URB597 significantly alleviated kainic acid-induced SE, decreasing behavioral manifestations, duration of seizure events and SE in general without changing the amplitude of local field potentials. AM251 did not produce distinct effects on SE in terms of our experimental paradigm. There was no apparent change of the seizure initiation pattern when kainic acid was coadministrated with AM404, URB597 or AM251. The present study provides electrophysiologic and behavioral evidences that inhibition of endocannabinoid metabolism plays a protective role against kainic acid-induced SE and may be employed for therapeutic purposes. Further investigations of the influences of cannabinoid-related compounds on SE genesis and especially epileptogenesis are required.
Collapse
|
46
|
Abstract
The antiepileptic potential of Cannabis sativa preparations has been historically recognized. Recent changes in legal restrictions and new well-documented cases reporting remarkably strong beneficial effects have triggered an upsurge in exploiting medical marijuana in patients with refractory epilepsy. Parallel research efforts in the last decade have uncovered the fundamental role of the endogenous cannabinoid system in controlling neuronal network excitability raising hopes for cannabinoid-based therapeutic approaches. However, emerging data show that patient responsiveness varies substantially, and that cannabis administration may sometimes even exacerbate seizures. Qualitative and quantitative chemical variability in cannabis products and personal differences in the etiology of seizures, or in the pathological reorganization of epileptic networks, can all contribute to divergent patient responses. Thus, the consensus view in the neurologist community is that drugs modifying the activity of the endocannabinoid system should first be tested in clinical trials to establish efficacy, safety, dosing, and proper indication in specific forms of epilepsies. To support translation from anecdote-based practice to evidence-based therapy, the present review first introduces current preclinical and clinical efforts for cannabinoid- or endocannabinoid-based epilepsy treatments. Next, recent advances in our knowledge of how endocannabinoid signaling limits abnormal network activity as a central component of the synaptic circuit-breaker system will be reviewed to provide a framework for the underlying neurobiological mechanisms of the beneficial and adverse effects. Finally, accumulating evidence demonstrating robust synapse-specific pathophysiological plasticity of endocannabinoid signaling in epileptic networks will be summarized to gain better understanding of how and when pharmacological interventions may have therapeutic relevance.
Collapse
Affiliation(s)
- István Katona
- Momentum Laboratory of Molecular Neurobiology, Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony u. 43, Budapest, 1083, Hungary.
| |
Collapse
|
47
|
Epilepsy But Not Mobile Phone Frequency (900 MHz) Induces Apoptosis and Calcium Entry in Hippocampus of Epileptic Rat: Involvement of TRPV1 Channels. J Membr Biol 2014; 248:83-91. [DOI: 10.1007/s00232-014-9744-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2014] [Accepted: 10/23/2014] [Indexed: 11/24/2022]
|
48
|
Ghazizadeh V, Nazıroğlu M. Electromagnetic radiation (Wi-Fi) and epilepsy induce calcium entry and apoptosis through activation of TRPV1 channel in hippocampus and dorsal root ganglion of rats. Metab Brain Dis 2014; 29:787-99. [PMID: 24792079 DOI: 10.1007/s11011-014-9549-9] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 04/14/2014] [Indexed: 11/28/2022]
Abstract
Incidence rates of epilepsy and use of Wi-Fi worldwide have been increasing. TRPV1 is a Ca(2+) permeable and non-selective channel, gated by noxious heat, oxidative stress and capsaicin (CAP). The hyperthermia and oxidant effects of Wi-Fi may induce apoptosis and Ca(2+) entry through activation of TRPV1 channel in epilepsy. Therefore, we tested the effects of Wi-Fi (2.45 GHz) exposure on Ca(2+) influx, oxidative stress and apoptosis through TRPV1 channel in the murine dorsal root ganglion (DRG) and hippocampus of pentylentetrazol (PTZ)-induced epileptic rats. Rats in the present study were divided into two groups as controls and PTZ. The PTZ groups were divided into two subgroups namely PTZ + Wi-Fi and PTZ + Wi-Fi + capsazepine (CPZ). The hippocampal and DRG neurons were freshly isolated from the rats. The DRG and hippocampus in PTZ + Wi-Fi and PTZ + Wi-Fi + CPZ groups were exposed to Wi-Fi for 1 hour before CAP stimulation. The cytosolic free Ca(2+), reactive oxygen species production, apoptosis, mitochondrial membrane depolarization, caspase-3 and -9 values in hippocampus were higher in the PTZ group than in the control although cell viability values decreased. The Wi-Fi exposure induced additional effects on the cytosolic Ca(2+) increase. However, pretreatment of the neurons with CPZ, results in a protection against epilepsy-induced Ca(2+) influx, apoptosis and oxidative damages. In results of whole cell patch-clamp experiments, treatment of DRG with Ca(2+) channel antagonists [thapsigargin, verapamil + diltiazem, 2-APB, MK-801] indicated that Wi-Fi exposure induced Ca(2+) influx via the TRPV1 channels. In conclusion, epilepsy and Wi-Fi in our experimental model is involved in Ca(2+) influx and oxidative stress-induced hippocampal and DRG death through activation of TRPV1 channels, and negative modulation of this channel activity by CPZ pretreatment may account for the neuroprotective activity against oxidative stress.
Collapse
Affiliation(s)
- Vahid Ghazizadeh
- Neuroscience Research Center, University of Suleyman Demirel, 32260, Isparta, Isparta, Turkey
| | | |
Collapse
|
49
|
Distinct modulation of the endocannabinoid system upon kainic acid-induced in vivo seizures and in vitro epileptiform bursting. Mol Cell Neurosci 2014; 62:1-9. [PMID: 25064144 DOI: 10.1016/j.mcn.2014.07.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/06/2014] [Accepted: 07/23/2014] [Indexed: 12/11/2022] Open
Abstract
There is clear evidence on the neuroprotective role of the endocannabinoid (eCB) signaling cascade in various models of epilepsy. In particular, increased levels of eCBs protect against kainic acid (KA)-induced seizures. However, the molecular mechanisms underlying this effect and its age-dependence are still unknown. To clarify this issue, we investigated which step of the biosynthetic and catabolic pathways of the eCBs may be responsible for the eCB-mediated neuroprotection in the hippocampus of P14 and P56-70 KA-treated rats. We found that both anandamide and N-palmitoylethanolamine, together with their biosynthetic enzyme significantly increased in the hippocampus of younger KA-treated rats, while decreasing in adults. In contrast, the levels of the other major eCB, 2-arachidonoylglycerol, similar to its biosynthetic enzyme, were higher in the hippocampus of P56-70 compared to P14 rats. In line with these data, extracellular field recordings in CA1 hippocampus showed that enhancement of endogenous AEA and 2-AG significantly counteracted KA-induced epileptiform bursting in P56-70 and P14 rats, respectively. On the contrary, while the CB1R antagonist SR141716 per se did not affect the population spike, it did worsen KA-induced bursts, confirming increased eCB tone upon KA treatment. Altogether these data indicate an age-specific alteration of the eCB system caused by KA and provide insights for the protective mechanism of the cannabinoid system against epileptiform discharges.
Collapse
|
50
|
Vilela LR, Medeiros DC, de Oliveira ACP, Moraes MF, Moreira FA. Anticonvulsant effects of N-arachidonoyl-serotonin, a dual fatty acid amide hydrolase enzyme and transient receptor potential vanilloid type-1 (TRPV1) channel blocker, on experimental seizures: the roles of cannabinoid CB1 receptors and TRPV1 channels. Basic Clin Pharmacol Toxicol 2014; 115:330-4. [PMID: 24674273 DOI: 10.1111/bcpt.12232] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Accepted: 03/04/2014] [Indexed: 01/21/2023]
Abstract
Selective blockade of anandamide hydrolysis, through the inhibition of the FAAH enzyme, has anticonvulsant effects, which are mediated by CB1 receptors. Anandamide, however, also activates TRPV1 channels, generally with an opposite outcome on neuronal modulation. Thus, we suggested that the dual FAAH and TRPV1 blockade with N-arachidonoyl-serotonin (AA-5-HT) would be efficacious in inhibiting pentylenetetrazole (PTZ)-induced seizures in mice. We also investigated the contribution of CB1 activation and TRPV1 blockade to the overt effect of AA-5-HT. In the first experiment, injection of AA-5-HT (0.3-3.0 mg/kg) delayed the onset and reduced the duration of PTZ (60 mg)-induced seizures in mice. These effects were reversed by pre-treatment with the CB1 antagonist, AM251 (1.0-3.0 mg/kg). Finally, we observed that administration of the selective TRPV1 antagonist, SB366791 (0.1-1 mg/kg), did not entirely mimic AA-5-HT effects. In conclusion, AA-5-HT alleviates seizures in mice, an effect inhibited by CB1 antagonism, but not completely mimicked by TRPV1 blockage, indicating that the overall effect of AA-5-HT seems to depend mainly on CB1 receptors. This may represent a new strategy for the development of drugs against seizures, epilepsies and related syndromes.
Collapse
Affiliation(s)
- Luciano R Vilela
- Graduate School in Neuroscience, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | | | | | | | | |
Collapse
|