1
|
Antonucci F, Bozzi Y. Action of Botulinum Neurotoxin E Type in Experimental Epilepsies. Toxins (Basel) 2023; 15:550. [PMID: 37755976 PMCID: PMC10536604 DOI: 10.3390/toxins15090550] [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: 07/21/2023] [Revised: 08/09/2023] [Accepted: 08/26/2023] [Indexed: 09/28/2023] Open
Abstract
Botulinum neurotoxins (BoNTs) are zinc endopeptidases produced by the Clostridium genus of anerobic bacteria, largely known for their ability to cleave synaptic proteins, leading to neuromuscular paralysis. In the central nervous system, BoNTs are known to block the release of glutamate neurotransmitter, and for this reason, researchers explored the possible therapeutic action in disorders characterized by neuronal hyperactivity, such as epilepsy. Thus, using multidisciplinary approaches and models of experimental epilepsy, we investigated the pharmacological potential of BoNT/E serotype. In this review, written in memory of Prof. Matteo Caleo, a pioneer in these studies, we go back over the hypotheses and experimental approaches that led us to the conclusion that intrahippocampal administration of BoNT/E (i) displays anticonvulsant effects if prophylactically delivered in a model of acute generalized seizures; (ii) does not have any antiepileptogenic action after the induction of status epilepticus; (iii) reduces frequency of spontaneous seizures in a model of recurrent seizures if delivered during the chronic phase but in a transient manner. Indeed, the control on spontaneous seizures stops when BoNT/E effects are off (few days), thus limiting its pharmacological potential in humans.
Collapse
Affiliation(s)
- Flavia Antonucci
- Department of Medical Biotechnology and Translational Medicine (BIOMETRA), University of Milan, via Fratelli Cervi 93, 20054 Milan, Italy
- CNR Institute of Neuroscience, via Raoul Follereau 3, 20854 Vedano al Lambro, Italy
- CIMeC-Center for Mind/Brain Sciences, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, Italy
| | - Yuri Bozzi
- CIMeC-Center for Mind/Brain Sciences, University of Trento, Piazza della Manifattura 1, 38068 Rovereto, Italy
- CNR Institute of Neuroscience, via Giuseppe Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
2
|
Drexel M, Sperk G. Seizure-induced overexpression of NPY induces epileptic tolerance in a mouse model of spontaneous recurrent seizures. Front Mol Neurosci 2022; 15:974784. [PMID: 36311021 PMCID: PMC9608171 DOI: 10.3389/fnmol.2022.974784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/26/2022] [Indexed: 11/21/2022] Open
Abstract
Epileptic seizures result in pronounced over-expression of neuropeptide Y (NPY). In vivo and in vitro studies revealed that NPY exerts potent anticonvulsive actions through presynaptic Y2 receptors by suppressing glutamate release from principal neurons. We now investigated whether seizure-induced over-expression of NPY contributes to epileptic tolerance induced by preceding seizures. We used a previously established animal model based on selective inhibition of GABA release from parvalbumin (PV)-containing interneurons in the subiculum in mice. The animals present spontaneous recurrent seizures (SRS) and clusters of interictal spikes (IS). The frequency of SRS declined after five to six weeks, indicating development of seizure tolerance. In interneurons of the subiculum and sector CA1, SRS induced over-expression of NPY that persisted there for a prolonged time despite of a later decrease in SRS frequency. In contrast to NPY, somatostatin was not overexpressed in the respective axon terminals. Contrary to interneurons, NPY was only transiently expressed in mossy fibers. To demonstrate a protective function of endogenous, over-expressed NPY, we injected the selective NPY-Y2 receptor antagonist JNJ 5207787 simultaneously challenging the mice by a low dose of pentylenetetrazol (PTZ, 30 or 40 mg/kg, i.p.). In control mice, neither PTZ nor PTZ plus JNJ 5207787 induced convulsions. In mice with silenced GABA/PV neurons, PTZ alone only modestly enhanced EEG activity. When we injected JNJ 5207787 together with PTZ (either dose) the number of seizures, however, became significantly increased. In addition, in the epileptic mice CB1 receptor immunoreactivity was reduced in terminal areas of basket cells pointing to reduced presynaptic inhibition of GABA release from these neurons. Our experiments demonstrate that SRS result in overexpression of NPY in hippocampal interneurons. NPY overexpression persists for several weeks and may be related to later decreasing SRS frequency. Injection of the Y2 receptor antagonist JNJ 5207787 prevents this protective action of NPY only when release of the peptide is triggered by injection of PTZ and induces pronounced convulsions. Thus, over-expressed NPY released “on demand” by seizures may help terminating acute seizures and may prevent from recurrent epileptic activity.
Collapse
|
3
|
Kohek SRB, Foresti ML, Blanco MM, Cavarsan CF, da Silva CS, Mello LE. Anxious Profile Influences Behavioral and Immunohistological Findings in the Pilocarpine Model of Epilepsy. Front Pharmacol 2021; 12:640715. [PMID: 34025410 PMCID: PMC8132119 DOI: 10.3389/fphar.2021.640715] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 03/22/2021] [Indexed: 12/01/2022] Open
Abstract
Anxiety and epilepsy have a complex bidirectional relationship, where a depressive/anxious condition is a factor that can trigger seizures which in turn can aggravate the depressive/anxious condition. In addition, brain structures such as the hippocampus and amygdala might have a critical relevance in both epilepsy and anxiety. The aim of the present work was to investigate the influence of different anxious profiles to epileptogenesis. Initially, animals were screened through the elevated plus-maze anxiety test, and then seizure development was evaluated using the pilocarpine model of epilepsy. There were no differences in the susceptibility to status epilepticus, mortality rate or frequency of spontaneous recurrent seizures between animals characterized as anxious as compared to the non-anxious animals. Next, we evaluated immunohistological patterns related to seizures and anxiety in various related brain areas. Despite a decrease in the density of neuropeptide Y and parvalbumin expression in epileptic animals, those presenting greater neuropeptide Y immunoreactivity in various brain regions, also showed higher spontaneous recurrent seizures frequency. Differences on the anxious profile showed to interfere with some of these findings in some regions. In addition, animals that were injected with pilocarpine, but did not develop status epilepticus, had behavioral and neuroanatomical alterations as compared to control animals, indicating its importance as an additional tool for investigating the heterogeneity of the epileptogenic response after an initial insult. This study allowed to better understand the association between anxiety and temporal lobe epilepsy and might allow for therapeutic targets to be developed to minimize the negative impacts associated with it.
Collapse
Affiliation(s)
| | | | | | - Clarissa Fantin Cavarsan
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, University of Rhode Island, Kingston, RI, United States.,George and Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI, United States
| | | | - Luiz E Mello
- Physiology Department, Universidade Federal de São Paulo, São Paulo, Brazil.,Instituto D' Or de Pesquisa e Ensino, Rio de Janeiro, Brazil
| |
Collapse
|
4
|
Anticonvulsant mechanisms of the ketogenic diet and caloric restriction. Epilepsy Res 2020; 168:106499. [PMID: 33190066 DOI: 10.1016/j.eplepsyres.2020.106499] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/16/2020] [Accepted: 10/27/2020] [Indexed: 01/29/2023]
Abstract
Many treatments have been proposed to control epileptic seizures, such as the ketogenic diet and caloric restriction. However, seizure control has not yet been improved completely in all patients. Probably, due to the lack of understanding regarding this neurological disorder pathogenesis or pathophysiology, including its molecular approach. Currently, there is not much information about the molecular processes and genes involved, and their relation to the possible beneficial effects of diet therapy on epilepsy. The ketogenic diet and caloric restriction are implicated in potential anti-seizure mechanisms related to the gut microbiome, metabolic pathways, hormones and neurotransmitters, mitochondria improvement, a role in inflammation, and oxidative stress, among others. In this review, we pretend to describe the molecular mechanism and the possible genes involved in the different ketogenic diet and caloric restriction mechanisms of action described to decrease neural excitability and, therefore, epileptic seizures, especially when conventional treatment is not enough to achieve control of epilepsy.
Collapse
|
5
|
Clinical value of a set of neuropeptides in term and preterm neonates with seizures: Brain derived neurotrophic factor, galanin and neuropeptide Y. J Clin Neurosci 2020; 74:168-174. [PMID: 32098713 DOI: 10.1016/j.jocn.2020.02.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Accepted: 02/10/2020] [Indexed: 12/26/2022]
Abstract
The aim of our study to investigate clinical value of a set of neuropeptides (brain derived neurotrophic factor-BDNF, galanin and neuropeptide Y-NPY) in critically ill neonates. A total of 53 neonates (preterm: 26, term: 27) evaluated with lumbar pucture for etiologic evaluation were consequtively included into the study. Serum and CSF levels of the neuropeptides were measured in the first 48 h of life. All infants were prospectively followed for prognostic outcome (survival and neurodevelopmental) at the first year of life. The study cohort was categorized into four groups with respect to seizure development; preterm neonates with or without seizure and term neonates with or without seizure. Mean CSF levels of NPY (pg/ml) were significantly higher in term neonates with than those without seizures (389.76 vs. 122.66) and galanin (3.31 vs. 1.55) respectively. Term neonates with seizures had significantly higher serum levels of NPY (ng/mL) as compared with neonates without seizures (54.00 vs. 9.10). No significant difference was noted in serum and CSF levels for the set of neuropeptides in neonates with respect to prognostic outcome. Serum NPY and CSF NPY and galanin levels have a potential role for detection of clinical seizures in term neonates.
Collapse
|
6
|
Tulke S, Haas CA, Häussler U. Expression of brain‐derived neurotrophic factor and structural plasticity in the dentate gyrus and
CA
2 region correlate with epileptiform activity. Epilepsia 2019; 60:1234-1247. [DOI: 10.1111/epi.15540] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 04/17/2019] [Accepted: 04/22/2019] [Indexed: 01/29/2023]
Affiliation(s)
- Susanne Tulke
- Experimental Epilepsy Research Department of Neurosurgery Medical Center - University of Freiburg Faculty of Medicine University of Freiburg Freiburg im Breisgau Germany
- Faculty of Biology University of Freiburg Freiburg im Breisgau Germany
| | - Carola A. Haas
- Experimental Epilepsy Research Department of Neurosurgery Medical Center - University of Freiburg Faculty of Medicine University of Freiburg Freiburg im Breisgau Germany
- BrainLinks‐BrainTools Cluster of Excellence University of Freiburg Freiburg im Breisgau Germany
| | - Ute Häussler
- Experimental Epilepsy Research Department of Neurosurgery Medical Center - University of Freiburg Faculty of Medicine University of Freiburg Freiburg im Breisgau Germany
- BrainLinks‐BrainTools Cluster of Excellence University of Freiburg Freiburg im Breisgau Germany
| |
Collapse
|
7
|
Pottoo FH, Tabassum N, Javed MN, Nigar S, Rasheed R, Khan A, Barkat MA, Alam MS, Maqbool A, Ansari MA, Barreto GE, Ashraf GM. The Synergistic Effect of Raloxifene, Fluoxetine, and Bromocriptine Protects Against Pilocarpine-Induced Status Epilepticus and Temporal Lobe Epilepsy. Mol Neurobiol 2019; 56:1233-1247. [PMID: 29881945 DOI: 10.1007/s12035-018-1121-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/11/2018] [Indexed: 10/14/2022]
Abstract
The present antiepileptic drugs pose several problems in the management of seizures owing to their meager neuroprotective potential, adverse effects on bone, detrimental effects on cognitive function, chronic toxicity, drug interactions, side effects including aggression, agitation, and irritability and sometimes exacerbation of seizures. We followed up progressive preclinical investigation in mice against pilocarpine (PILO)-induced status epilepticus (SE) and temporal lobe epilepsy (TLE). To determine the response of raloxifene (RF) (4 and 8 mg/kg), fluoxetine (FT) (14 and 22 mg/kg), bromocriptine (BC) (6 and 10 mg/kg), and their low-dose combinations, oral treatment was scheduled for 28 days followed by PILO (300 mg/kg, i.p). The response was stalked for intensive behavioral monitoring of convulsions, hippocampal neuropeptide Y (NPY), and oxidative stress discernment along with histomorphological studies. The resultant data confirmed the therapeutic potential of triple drug combination of raloxifene (4 mg/kg) with fluoxetine (14 mg/kg) and bromocriptine (6 mg/kg) compared to monotherapy with raloxifene (4 mg/kg), and bromocriptine (6 mg/kg) as otherwise monotherapy with fluoxetine (14 mg/kg) was ineffective to suppress convulsions; an effect better than sodium valproate (300 mg/kg), a standard AED, was validated. Most profoundly, PILO-induced compensatory increases in hippocampal NPY levels (20.01%), which was escalated (100%) with the triple drug combination. The same pattern of results was superseded for oxidative stress indices and neuronal damage. The results for the first time demonstrate the propitious role of triple drug combination in the management of SE and TLE. Therapeutically, this enhancing profile of drugs fosters a safer and more effective drug-combination regimen. Graphical abstract.
Collapse
Affiliation(s)
- Faheem Hyder Pottoo
- Department of Pharmaceutical Sciences, Faculty of Applied Sc. and Tech., University of Kashmir, Srinagar, India.
| | - Nahida Tabassum
- Department of Pharmaceutical Sciences, Faculty of Applied Sc. and Tech., University of Kashmir, Srinagar, India.
| | - Md Noushad Javed
- Department of Pharmaceutics, School of Pharmaceutical Sciences and Research, Jamia Hamdard, New Delhi, India
| | - Shah Nigar
- Department of Pharmaceutical Sciences, Faculty of Applied Sc. and Tech., University of Kashmir, Srinagar, India
| | - Rouqia Rasheed
- Department of Pharmaceutical Sciences, Faculty of Applied Sc. and Tech., University of Kashmir, Srinagar, India
| | - Ayash Khan
- Department of Pharmaceutical Sciences, Faculty of Applied Sc. and Tech., University of Kashmir, Srinagar, India
| | - Md Abul Barkat
- Department of Pharmacy, School of Medical and Allied Sciences, K.R. Mangalam University, Gurugram, India
| | - Md Sabir Alam
- Department of Pharmacy, School of Medical and Allied Sciences, K.R. Mangalam University, Gurugram, India
| | - Amir Maqbool
- Department of Zoology, Govt. College for Women, M. A. Road, Srinagar, India
| | - Mohammad Azam Ansari
- Department of Epidemic Disease Research, Institute of Research and Medical Consultations (IRMC), Imam Abdulrahman Bin Faisal University, Dammam, 31441, Saudi Arabia
| | - George E Barreto
- Departamento de Nutrición y Bioquímica, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá D.C., Colombia
- Instituto de Ciencias Biomédicas, Universidad Autónoma de Chile, Santiago, Chile
| | - Ghulam Md Ashraf
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
8
|
Gene therapy mediated seizure suppression in Genetic Generalised Epilepsy: Neuropeptide Y overexpression in a rat model. Neurobiol Dis 2018; 113:23-32. [DOI: 10.1016/j.nbd.2018.01.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Revised: 01/07/2018] [Accepted: 01/22/2018] [Indexed: 02/01/2023] Open
|
9
|
Differential Effect of Neuropeptides on Excitatory Synaptic Transmission in Human Epileptic Hippocampus. J Neurosci 2015; 35:9622-31. [PMID: 26134645 DOI: 10.1523/jneurosci.3973-14.2015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Development of novel disease-modifying treatment strategies for neurological disorders, which at present have no cure, represents a major challenge for today's neurology. Translation of findings from animal models to humans represents an unresolved gap in most of the preclinical studies. Gene therapy is an evolving innovative approach that may prove useful for clinical applications. In animal models of temporal lobe epilepsy (TLE), gene therapy treatments based on viral vectors encoding NPY or galanin have been shown to effectively suppress seizures. However, how this translates to human TLE remains unknown. A unique possibility to validate these animal studies is provided by a surgical therapeutic approach, whereby resected epileptic tissue from temporal lobes of pharmacoresistant patients are available for neurophysiological studies in vitro. To test whether NPY and galanin have antiepileptic actions in human epileptic tissue as well, we applied these neuropeptides directly to human hippocampal slices in vitro. NPY strongly decreased stimulation-induced EPSPs in dentate gyrus and CA1 (up to 30 and 55%, respectively) via Y2 receptors, while galanin had no significant effect. Receptor autoradiographic binding revealed the presence of both NPY and galanin receptors, while functional receptor binding was only detected for NPY, suggesting that galanin receptor signaling may be impaired. These results underline the importance of validating findings from animal studies in human brain tissue, and advocate for NPY as a more appropriate candidate than galanin for future gene therapy trials in pharmacoresistant TLE patients.
Collapse
|
10
|
Gorter JA, van Vliet EA, Lopes da Silva FH. Which insights have we gained from the kindling and post-status epilepticus models? J Neurosci Methods 2015; 260:96-108. [PMID: 25842270 DOI: 10.1016/j.jneumeth.2015.03.025] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 03/23/2015] [Accepted: 03/24/2015] [Indexed: 11/24/2022]
Abstract
Experimental animal epilepsy research got a big boost since the discovery that daily mild and short (seconds) tetanic stimulations in selected brain regions led to seizures with increasing duration and severity. This model that was developed by Goddard (1967) became known as the kindling model for epileptogenesis and has become a widely used model for temporal lobe epilepsy with complex partial seizures. During the late ninety-eighties the number of publications related to electrical kindling reached its maximum. However, since the kindling procedure is rather labor intensive and animals only develop spontaneous seizures (epilepsy) after hundreds of stimulations, research has shifted toward models in which the animals exhibit spontaneous seizures after a relatively short latent period. This led to post-status epilepticus (SE) models in which animals experience SE after injection of pharmacological compounds (e.g. kainate or pilocarpine) or via electrical stimulation of (limbic) brain regions. These post-SE models are the most widely used models in epilepsy research today. However, not all aspects of mesial temporal lobe epilepsy (MTLE) are reproduced and the widespread brain damage is often a caricature of the situation in the patient. Therefore, there is a need for models that can better replicate the disease. Kindling, although already a classic model, can still offer valid clues in this context. In this paper, we review different aspects of the kindling model with emphasis on experiments in the rat. Next, we review characteristic properties of the post-SE models and compare the neuropathological, electrophysiological and molecular differences between kindling and post-SE epilepsy models. Finally, we shortly discuss the advantages and disadvantages of these models.
Collapse
Affiliation(s)
- Jan A Gorter
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands.
| | - Erwin A van Vliet
- Department of (Neuro)Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Fernando H Lopes da Silva
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands; Instituto Superior Técnico, University of Lisbon, 1049-001 Lisbon, Portugal
| |
Collapse
|
11
|
Amini E, Rezaei M, Mohamed Ibrahim N, Golpich M, Ghasemi R, Mohamed Z, Raymond AA, Dargahi L, Ahmadiani A. A Molecular Approach to Epilepsy Management: from Current Therapeutic Methods to Preconditioning Efforts. Mol Neurobiol 2014; 52:492-513. [PMID: 25195699 DOI: 10.1007/s12035-014-8876-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2014] [Accepted: 08/25/2014] [Indexed: 01/16/2023]
Abstract
Epilepsy is the most common and chronic neurological disorder characterized by recurrent unprovoked seizures. The key aim in treating patients with epilepsy is the suppression of seizures. An understanding of focal changes that are involved in epileptogenesis may therefore provide novel approaches for optimal treatment of the seizure. Although the actual pathogenesis of epilepsy is still uncertain, recently growing lines of evidence declare that microglia and astrocyte activation, oxidative stress and reactive oxygen species (ROS) production, mitochondria dysfunction, and damage of blood-brain barrier (BBB) are involved in its pathogenesis. Impaired GABAergic function in the brain is probably the most accepted hypothesis regarding the pathogenesis of epilepsy. Clinical neuroimaging of patients and experimental modeling have demonstrated that seizures may induce neuronal apoptosis. Apoptosis signaling pathways are involved in the pathogenesis of several types of epilepsy such as temporal lobe epilepsy (TLE). The quality of life of patients is seriously affected by treatment-related problems and also by unpredictability of epileptic seizures. Moreover, the available antiepileptic drugs (AED) are not significantly effective to prevent epileptogenesis. Thus, novel therapies that are proficient to control seizure in people who are suffering from epilepsy are needed. The preconditioning method promises to serve as an alternative therapeutic approach because this strategy has demonstrated the capability to curtail epileptogenesis. For this reason, understanding of molecular mechanisms underlying brain tolerance induced by preconditioning is crucial to delineate new neuroprotective ways against seizure damage and epileptogenesis. In this review, we summarize the work to date on the pathogenesis of epilepsy and discuss recent therapeutic strategies in the treatment of epilepsy. We will highlight that novel therapy targeting such as preconditioning process holds great promise. In addition, we will also highlight the role of gene reprogramming and mitochondrial biogenesis in the preconditioning-mediated neuroprotective events.
Collapse
Affiliation(s)
- Elham Amini
- Department of Medicine, Universiti Kebangsaan Malaysia Medical Centre, Cheras, Kuala Lumpur, Malaysia
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Taksande BG, Kotagale NR, Gawande DY, Bharne AP, Chopde CT, Kokare DM. Neuropeptide Y in the central nucleus of amygdala regulates the anxiolytic effect of agmatine in rats. Eur Neuropsychopharmacol 2014; 24:955-63. [PMID: 24461723 DOI: 10.1016/j.euroneuro.2013.12.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/11/2013] [Revised: 11/15/2013] [Accepted: 12/02/2013] [Indexed: 10/25/2022]
Abstract
In the present study, modulation of anxiolytic action of agmatine by neuropeptide Y (NPY) in the central nucleus of amygdala (CeA) is evaluated employing Vogel's conflict test (VCT) in rats. The intra-CeA administration of agmatine (0.6 and 1.2µmol/rat), NPY (10 and 20pmol/rat) or NPY Y1/Y5 receptors agonist [Leu(31), Pro(34)]-NPY (30 and 60pmol/rat) significantly increased the number of punished drinking licks following 15min of treatment. Combination treatment of subeffective dose of NPY (5pmol/rat) or [Leu(31), Pro(34)]-NPY (15pmol/rat) and agmatine (0.3µmol/rat) produced synergistic anxiolytic-like effect. However, intra-CeA administration of selective NPY Y1 receptor antagonist, BIBP3226 (0.25 and 0.5mmol/rat) produced anxiogenic effect. In separate set of experiment, pretreatment with BIBP3226 (0.12mmol/rat) reversed the anxiolytic effect of agmatine (0.6µmol/rat). Furthermore, we evaluated the effect of intraperitoneal injection of agmatine (40mg/kg) on NPY-immunoreactivity in the nucleus accumbens shell (AcbSh), lateral part of bed nucleus of stria terminalis (BNSTl) and CeA. While agmatine treatment significantly decreased the fibers density in BNSTl, increase was noticed in AcbSh. In addition, agmatine reduced NPY-immunoreactive cells in the AcbSh and CeA. Immunohistochemical data suggest the enhanced transmission of NPY from the AcbSh and CeA. Taken together, this study suggests that agmatine produced anxiolytic effect which might be regulated via modulation of NPYergic system particularly in the CeA.
Collapse
Affiliation(s)
- Brijesh G Taksande
- Division of Neuroscience, Department of Pharmacology, Shrimati Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur (M.S.) 441002, India
| | - Nandkishor R Kotagale
- Division of Neuroscience, Department of Pharmacology, Shrimati Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur (M.S.) 441002, India
| | - Dinesh Y Gawande
- Division of Neuroscience, Department of Pharmacology, Shrimati Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur (M.S.) 441002, India
| | - Ashish P Bharne
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440033, India
| | - Chandrabhan T Chopde
- Division of Neuroscience, Department of Pharmacology, Shrimati Kishoritai Bhoyar College of Pharmacy, New Kamptee, Nagpur (M.S.) 441002, India
| | - Dadasaheb M Kokare
- Department of Pharmaceutical Sciences, Rashtrasant Tukadoji Maharaj Nagpur University, Nagpur 440033, India.
| |
Collapse
|
13
|
Clynen E, Swijsen A, Raijmakers M, Hoogland G, Rigo JM. Neuropeptides as targets for the development of anticonvulsant drugs. Mol Neurobiol 2014; 50:626-46. [PMID: 24705860 PMCID: PMC4182642 DOI: 10.1007/s12035-014-8669-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 02/27/2014] [Indexed: 11/04/2022]
Abstract
Epilepsy is a common neurological disorder characterized by recurrent seizures. These seizures are due to abnormal excessive and synchronous neuronal activity in the brain caused by a disruption of the delicate balance between excitation and inhibition. Neuropeptides can contribute to such misbalance by modulating the effect of classical excitatory and inhibitory neurotransmitters. In this review, we discuss 21 different neuropeptides that have been linked to seizure disorders. These neuropeptides show an aberrant expression and/or release in animal seizure models and/or epilepsy patients. Many of these endogenous peptides, like adrenocorticotropic hormone, angiotensin, cholecystokinin, cortistatin, dynorphin, galanin, ghrelin, neuropeptide Y, neurotensin, somatostatin, and thyrotropin-releasing hormone, are able to suppress seizures in the brain. Other neuropeptides, such as arginine-vasopressine peptide, corticotropin-releasing hormone, enkephalin, β-endorphin, pituitary adenylate cyclase-activating polypeptide, and tachykinins have proconvulsive properties. For oxytocin and melanin-concentrating hormone both pro- and anticonvulsive effects have been reported, and this seems to be dose or time dependent. All these neuropeptides and their receptors are interesting targets for the development of new antiepileptic drugs. Other neuropeptides such as nesfatin-1 and vasoactive intestinal peptide have been less studied in this field; however, as nesfatin-1 levels change over the course of epilepsy, this can be considered as an interesting marker to diagnose patients who have suffered a recent epileptic seizure.
Collapse
Affiliation(s)
- Elke Clynen
- Biomedical Research Institute BIOMED, Hasselt University, Martelarenlaan 42, 3500, Hasselt, Belgium,
| | | | | | | | | |
Collapse
|
14
|
Xu X, Guo F, He Q, Cai X, Min D, Wang Q, Wang S, Tian L, Cai J, Zhao Y. Altered expression of neuropeptide Y, Y1 and Y2 receptors, but not Y5 receptor, within hippocampus and temporal lobe cortex of tremor rats. Neuropeptides 2014; 48:97-105. [PMID: 24444822 DOI: 10.1016/j.npep.2013.12.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Revised: 12/18/2013] [Accepted: 12/22/2013] [Indexed: 01/24/2023]
Abstract
As an endogenous inhibitor of glutamate-mediated synaptic transmission in mammalian central nervous system, neuropeptide Y (NPY) plays a crucial role in regulating homeostasis of neuron excitability. Loss of balance between excitatory and inhibitory neurotransmission is thought to be a chief mechanism of epileptogenesis. The abnormal expression of NPY and its receptors observed following seizures have been demonstrated to be related to the production of epilepsy. The tremor rat (TRM) is a hereditary epileptic animal model. So far, there is no report concerning whether NPY and its receptors may be involved in TRM pathogenesis. In this study, we focused on the expression of NPY and its three receptor subtypes: Y1R, Y2R and Y5R in the TRM brain. We first found the expression of NPY in TRM hippocampus and temporal lobe cortex was increased compared with control (Wistar) rats. The mRNA and protein expression of Y1R was down-regulated in hippocampus but up-regulated in temporal lobe cortex, whereas Y2R expression was significantly increased in both areas. There was no significant change of Y5R expression in either area. The immunohistochemistry data showed that Y1R, Y2R, Y5R were present throughout CA1, CA3, dentate gyrus (DG) and the entorhinal cortex which is included in the temporal lobe cortex of TRM. In conclusion, our results showed the altered expression of NPY, Y1R and Y2R but not Y5R in hippocampus and temporal lobe cortex of TRM brain. This abnormal expression may be associated with the generation of epileptiform activity and provide a candidate target for treatment of genetic epilepsy.
Collapse
Affiliation(s)
- Xiaoxue Xu
- Biochip Center, College of Basic Medicine, China Medical University, Shenyang 110001, China; Department of Neurology, The First Hospital of China Medical University, Shenyang 110001, China
| | - Feng Guo
- Department of Pharmaceutical Toxicology, School of Pharmaceutical Science, China Medical University, Shenyang 110001, China
| | - Qun He
- Biochip Center, College of Basic Medicine, China Medical University, Shenyang 110001, China
| | - Xinze Cai
- Central Lab, The First Hospital of China Medical University, Shenyang 110001, China
| | - Dongyu Min
- Experiment Center of Traditional Chinese Medicine, The Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 110032, China
| | - Qianhui Wang
- Department of Pharmaceutical Toxicology, School of Pharmaceutical Science, China Medical University, Shenyang 110001, China
| | - Shaocheng Wang
- Biochip Center, College of Basic Medicine, China Medical University, Shenyang 110001, China
| | - Liu Tian
- Biochip Center, College of Basic Medicine, China Medical University, Shenyang 110001, China
| | - Jiqun Cai
- Department of Pharmaceutical Toxicology, School of Pharmaceutical Science, China Medical University, Shenyang 110001, China
| | - Yujie Zhao
- Biochip Center, College of Basic Medicine, China Medical University, Shenyang 110001, China.
| |
Collapse
|
15
|
Marx M, Haas CA, Häussler U. Differential vulnerability of interneurons in the epileptic hippocampus. Front Cell Neurosci 2013; 7:167. [PMID: 24098270 PMCID: PMC3787650 DOI: 10.3389/fncel.2013.00167] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 09/07/2013] [Indexed: 11/30/2022] Open
Abstract
The loss of hippocampal interneurons has been considered as one reason for the onset of temporal lobe epilepsy (TLE) by shifting the excitation-inhibition balance. Yet, there are many different interneuron types which show differential vulnerability in the context of an epileptogenic insult. We used the intrahippocampal kainate (KA) mouse model for TLE in which a focal, unilateral KA injection induces status epilepticus (SE) followed by development of granule cell dispersion (GCD) and hippocampal sclerosis surrounding the injection site but not in the intermediate and temporal hippocampus. In this study, we characterized the loss of interneurons with respect to septotemporal position and to differential vulnerability of interneuron populations. To this end, we performed intrahippocampal recordings of the initial SE, in situ hybridization for glutamic acid decarboxylase 67 (GAD67) mRNA and immunohistochemistry for parvalbumin (PV) and neuropeptide Y (NPY) in the early phase of epileptogenesis at 2 days and at 21 days after KA injection, when recurrent epileptic activity and GCD have fully developed. We show that SE extended along the entire septotemporal axis of both hippocampi, but was stronger at distant sites than at the injection site. There was an almost complete loss of interneurons surrounding the injection site and expanding to the intermediate hippocampus already at 2 days but increasing until 21 days after KA. Furthermore, we observed differential vulnerability of PV- and NPY-expressing cells: while the latter were lost at the injection site but preserved at intermediate sites, PV-expressing cells were gone even at sites more temporal than GCD. In addition, we found upregulation of GAD67 mRNA expression in dispersed granule cells and of NPY staining in ipsilateral granule cells and ipsi- and contralateral mossy fibers. Our data thus indicate differential survival capacity of interneurons in the epileptic hippocampus and compensatory plasticity mechanisms depending on the hippocampal position.
Collapse
Affiliation(s)
- Markus Marx
- Experimental Epilepsy Research, Department of Neurosurgery, University of Freiburg Freiburg, Germany
| | | | | |
Collapse
|
16
|
Elms J, Powell KL, van Raay L, Dedeurwaerdere S, O’Brien TJ, Morris MJ. Long-term valproate treatment increases brain neuropeptide Y expression and decreases seizure expression in a genetic rat model of absence epilepsy. PLoS One 2013; 8:e73505. [PMID: 24039965 PMCID: PMC3767750 DOI: 10.1371/journal.pone.0073505] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Accepted: 07/23/2013] [Indexed: 11/19/2022] Open
Abstract
The mechanisms by which valproate, one of the most widely prescribed anti-epileptic drugs, suppresses seizures have not been fully elucidated but may involve up-regulation of neuropeptide Y (NPY). We investigated the effects of valproate treatment in Genetic Absence Epilepsy Rats from Strasbourg (GAERS) on brain NPY mRNA expression and seizure control. GAERS were administered either valproate (42 mg.kg−1 hr−1) or saline continuously for 5 days. Electroencephalograms were recorded for 24 hrs on treatment days 1, 3 and 5 and the percentage of time spent in seizure activity was analysed. NPY mRNA expression was measured in different brain regions using qPCR. Valproate treatment suppressed seizures by 80% in GAERS (p<0.05) and increased NPY mRNA expression in the thalamus (p<0.05) compared to saline treatment. These results demonstrate that long-term valproate treatment results in an upregulation of thalamic expression of NPY implicating this as a potential contributor to the mechanism by which valproate suppresses absence seizures.
Collapse
Affiliation(s)
- Johanna Elms
- Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, Australia
- The Department of Medicine, The University of Melbourne, Royal Melbourne Hospital, Melbourne, Australia
| | - Kim L. Powell
- The Department of Medicine, The University of Melbourne, Royal Melbourne Hospital, Melbourne, Australia
| | - Leena van Raay
- The Department of Medicine, The University of Melbourne, Royal Melbourne Hospital, Melbourne, Australia
| | | | - Terence J. O’Brien
- The Department of Medicine, The University of Melbourne, Royal Melbourne Hospital, Melbourne, Australia
| | - Margaret J. Morris
- Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, Australia
- * E-mail:
| |
Collapse
|
17
|
|
18
|
Zhu XB, Wang YB, Chen O, Zhang DQ, Zhang ZH, Cao AH, Huang SY, Sun RP. Characterization of the expression of macrophage inflammatory protein-1α (MIP-1α) and C-C chemokine receptor 5 (CCR5) after kainic acid-induced status epilepticus (SE) in juvenile rats. Neuropathol Appl Neurobiol 2012; 38:602-16. [DOI: 10.1111/j.1365-2990.2012.01251.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
19
|
Wu G, Feder A, Wegener G, Bailey C, Saxena S, Charney D, Mathé AA. Central functions of neuropeptide Y in mood and anxiety disorders. Expert Opin Ther Targets 2012; 15:1317-31. [PMID: 21995655 DOI: 10.1517/14728222.2011.628314] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Neuropeptide Y (NPY) is a highly conserved neuropeptide belonging to the pancreatic polypeptide family. Its potential role in the etiology and pathophysiology of mood and anxiety disorders has been extensively studied. NPY also has effects on feeding behavior, ethanol intake, sleep regulation, tissue growth and remodeling. Findings from animal studies have delineated the physiological and behavioral effects mediated by specific NPY receptor subtypes, of which Y1 and Y2 are the best understood. AREAS COVERED Physiological roles and alterations of the NPYergic system in anxiety disorders, depression, posttraumatic stress disorder (PTSD), alcohol dependence and epilepsy. For each disorder, studies in animal models and human investigations are outlined and discussed, focusing on behavior, neurophysiology, genetics and potential for novel treatment targets. EXPERT OPINION The wide implications of NPY in psychiatric disorders such as depression and PTSD make the NPYergic system a promising target for the development of novel therapeutic interventions. These include intranasal NPY administration, currently under study, and the development of agonists and antagonists targeting NPY receptors. Therefore, we are proposing that via this mode of administration, NPY might exert CNS therapeutic actions without untoward systemic effects. Future work will show if this is a feasible approach.
Collapse
Affiliation(s)
- Gang Wu
- Karolinska Institutet-Clinical Neuroscience, Stockholm, Sweden
| | | | | | | | | | | | | |
Collapse
|
20
|
Ledri M, Sørensen AT, Erdelyi F, Szabo G, Kokaia M. Tuning afferent synapses of hippocampal interneurons by neuropeptide Y. Hippocampus 2011; 21:198-211. [DOI: 10.1002/hipo.20740] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
21
|
Goto EM, Silva MDP, Perosa SR, Argañaraz GA, Pesquero JB, Cavalheiro EA, Naffah-Mazzacoratti MG, Teixeira VPC, Silva JA. Akt pathway activation and increased neuropeptide Y mRNA expression in the rat hippocampus: implications for seizure blockade. Neuropeptides 2010; 44:169-76. [PMID: 20064661 DOI: 10.1016/j.npep.2009.12.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 12/09/2009] [Accepted: 12/10/2009] [Indexed: 01/01/2023]
Abstract
The aim of this study was to analyze the expression of survival-related molecules such Akt and integrin-linked kinase (ILK) to evaluate Akt pathway activation in epileptogenesis process. Furthermore, was also investigated the mRNA expression of neuropeptide Y, a considered antiepileptic neuropeptide, in the pilocarpine-induced epilepsy. Male Wistar rats were submitted to the pilocarpine model of epilepsy. Hippocampi were removed 6h (acute phase), 12h (late acute), 5d (silent) and 60d (chronic) after status epilepticus (SE) onset, and from animals that received pilocarpine but did not develop SE (partial group). Hippocampi collected were used to specify mRNA expression using Real-Time PCR. Immunohistochemistry assay was employed to place ILK distribution in the hippocampus and Western blot technique was used to determine Akt activation level. A decrease in ILK mRNA content was found during acute (0.39+/-0.03) and chronic (0.48+/-0.06) periods when compared to control group (0.87+/-0.10). Protein levels of ILK were also diminished during both periods. Partial group showed increased ILK mRNA expression (0.80+/-0.06) when compared with animals in the acute stage. Silent group had ILK mRNA and immunoreactivity similar to control group. Western blot assay showed an augmentation in Akt activation in silent period (0.52+/-0.03) in comparison with control group (0.44+/-0.01). Neuropeptide Y mRNA expression increased in the partial group (1.67+/-0.22) and in the silent phase (1.45+/-0.29) when compared to control group (0.36+/-0.12). Results suggest that neuropeptide Y (as anticonvulsant) might act in protective mechanisms occurred during epileptic phenomena. Together with ILK expression and Akt activation, these molecules could be involved in hippocampal neuroprotection in epilepsy.
Collapse
Affiliation(s)
- Eduardo M Goto
- Pathology Department, Universidade Federal de São Paulo, UNIFESP, Brazil
| | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Lin EJD, Lin S, Aljanova A, During MJ, Herzog H. Adult-onset hippocampal-specific neuropeptide Y overexpression confers mild anxiolytic effect in mice. Eur Neuropsychopharmacol 2010; 20:164-75. [PMID: 19781916 DOI: 10.1016/j.euroneuro.2009.08.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Revised: 07/09/2009] [Accepted: 08/18/2009] [Indexed: 01/29/2023]
Abstract
The anticonvulsive properties of neuropeptide Y (NPY) are opening up opportunity for the development of NPY gene transfer as a therapy for epilepsy. In order to pursue the potential clinical translation of this approach, the effects of somatic NPY gene transfer on other hippocampal functions need to be assessed. The present study characterized the behavioral effects of recombinant adeno-associated viral vector (rAAV)-mediated hippocampal NPY overexpression in adult male mice and also Y1 receptor knockout mice. In wild-type mice, there were no obvious adverse effects on the general health, motor function and cognition following rAAV-NPY treatment. Moreover, hippocampal NPY overexpression induced a moderate anxiolytic effect in the open field test and elevated plus maze. Intriguingly, the treatment also increased depressive-like behavior in the tail suspension test. Elevated hippocampal NPY levels in the absence of Y1 signalling had no effects on anxiety or cognition and actually improved the depressive-like phenotype observed in the wild-type mice treated with rAAV-NPY.
Collapse
Affiliation(s)
- En-Ju Deborah Lin
- Neurobiology Program, Garvan Institute of Medical Research, Sydney, Australia.
| | | | | | | | | |
Collapse
|
23
|
Affiliation(s)
- Paula Elyse Schauwecker
- Department of Pharmacology and Pharmaceutical Sciences, USC School of Pharmacy, Los Angeles, California, USA.
| |
Collapse
|
24
|
Kondo A, Shingo T, Yasuhara T, Kuramoto S, Kameda M, Kikuchi Y, Matsui T, Miyoshi Y, Agari T, Borlongan CV, Date I. Erythropoietin exerts anti-epileptic effects with the suppression of aberrant new cell formation in the dentate gyrus and upregulation of neuropeptide Y in seizure model of rats. Brain Res 2009; 1296:127-36. [DOI: 10.1016/j.brainres.2009.08.025] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Revised: 08/02/2009] [Accepted: 08/05/2009] [Indexed: 11/15/2022]
|
25
|
Kalev-Zylinska ML, Symes W, Young D, During MJ. Knockdown and overexpression of NR1 modulates NMDA receptor function. Mol Cell Neurosci 2009; 41:383-96. [PMID: 19394426 DOI: 10.1016/j.mcn.2009.04.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2008] [Revised: 03/24/2009] [Accepted: 04/17/2009] [Indexed: 01/17/2023] Open
Abstract
The N-methyl-d-aspartate receptor (NMDAR) is critically involved in learning and memory, neuronal survival, as well as neuroexcitotoxicity and seizures. We hypothesize that even mild reductions in the numbers of hippocampal NMDARs could impair learning and memory, whereas increasing receptor activity would facilitate learning but reduce seizure threshold. We developed novel gene transfer strategies assisted by an adeno-associated viral vector 1/2 to bi-directionally modulate expression levels of the NR1 protein in rat hippocampus. Functional consequences of the altered NR1 expression were examined in the acute seizure model, and on normal processes of fear memory and neurogenesis. We found that knocking down NR1 protected against seizures at the expense of impaired learning, as predicted. Paradoxically, NR1 overexpression not only increased fear memory and neurogenesis, but also delayed onset of more severe seizures. In conclusion, the observed consequences of NR1 knockdown and overexpression underscore NMDAR requirement for neuronal plasticity, and are in agreement with its dichotomous functions.
Collapse
Affiliation(s)
- Maggie L Kalev-Zylinska
- Department of Molecular Medicine and Pathology, The University of Auckland, Auckland, New Zealand
| | | | | | | |
Collapse
|
26
|
Estradiol facilitates the release of neuropeptide Y to suppress hippocampus-dependent seizures. J Neurosci 2009; 29:1457-68. [PMID: 19193892 DOI: 10.1523/jneurosci.4688-08.2009] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
About one-third of women with epilepsy have a catamenial seizure pattern, in which seizures fluctuate with the menstrual cycle. Catamenial seizures occur more frequently when the ratio of circulating estradiol to progesterone is high, suggesting that estradiol is proconvulsant. We used adult female rats to test how estradiol-induced suppression of GABAergic inhibition in the hippocampus affects behavioral seizures induced by kainic acid. As expected, estradiol decreased the latency to initiate seizures, indicating increased seizure susceptibility. At the same time, however, estradiol also shortened the duration of late-stage seizures, indicating decreased seizure severity. Additional analyses showed that the decrease in seizure severity was attributable to greater release of the anticonvulsant neuropeptide, neuropeptide Y (NPY). First, blocking hippocampal NPY during seizures eliminated the estradiol-induced decrease in seizure duration. Second, light and electron microscopic studies indicated that estradiol increases the potentially releasable pool of NPY in inhibitory presynaptic boutons and facilitates the release of NPY from inhibitory boutons during seizures. Finally, the presence of estrogen receptor-alpha on large dense-core vesicles (LDCVs) in the hippocampus suggests that estradiol could facilitate neuropeptide release by acting directly on LDCVs themselves. Understanding how estradiol regulates NPY-containing LDCVs could point to molecular targets for novel anticonvulsant therapies.
Collapse
|
27
|
Sørensen AT, Nikitidou L, Ledri M, Lin EJD, During MJ, Kanter-Schlifke I, Kokaia M. Hippocampal NPY gene transfer attenuates seizures without affecting epilepsy-induced impairment of LTP. Exp Neurol 2008; 215:328-33. [PMID: 19038255 DOI: 10.1016/j.expneurol.2008.10.015] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Revised: 10/25/2008] [Accepted: 10/29/2008] [Indexed: 10/21/2022]
Abstract
Recently, hippocampal neuropeptide Y (NPY) gene therapy has been shown to effectively suppress both acute and chronic seizures in animal model of epilepsy, thus representing a promising novel antiepileptic treatment strategy, particularly for patients with intractable mesial temporal lobe epilepsy (TLE). However, our previous studies show that recombinant adeno-associated viral (rAAV)-NPY treatment in naive rats attenuates long-term potentiation (LTP) and transiently impairs hippocampal learning process, indicating that negative effect on memory function could be a potential side effect of NPY gene therapy. Here we report how rAAV vector-mediated overexpression of NPY in the hippocampus affects rapid kindling, and subsequently explore how synaptic plasticity and transmission is affected by kindling and NPY overexpression by field recordings in CA1 stratum radiatum of brain slices. In animals injected with rAAV-NPY, we show that rapid kindling-induced hippocampal seizures in vivo are effectively suppressed as compared to rAAV-empty injected (control) rats. Six to nine weeks later, basal synaptic transmission and short-term synaptic plasticity are unchanged after rapid kindling, while LTP is significantly attenuated in vitro. Importantly, transgene NPY overexpression has no effect on short-term synaptic plasticity, and does not further compromise LTP in kindled animals. These data suggest that epileptic seizure-induced impairment of memory function in the hippocampus may not be further affected by rAAV-NPY treatment, and may be considered less critical for clinical application in epilepsy patients already experiencing memory disturbances.
Collapse
Affiliation(s)
- Andreas T Sørensen
- Experimental Epilepsy Group, Wallenberg Neuroscience Center, BMC A-11, Lund University Hospital, Lund, Sweden
| | | | | | | | | | | | | |
Collapse
|
28
|
Sosulina L, Schwesig G, Seifert G, Pape HC. Neuropeptide Y activates a G-protein-coupled inwardly rectifying potassium current and dampens excitability in the lateral amygdala. Mol Cell Neurosci 2008; 39:491-8. [PMID: 18790060 DOI: 10.1016/j.mcn.2008.08.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2008] [Revised: 08/01/2008] [Accepted: 08/13/2008] [Indexed: 11/26/2022] Open
Abstract
Neuropeptide Y (NPY) reduces anxiety-related behavior in various animal models. Since activity in the lateral amygdala (LA) seems crucial for fear expression of behavior, we studied the mechanisms of NPY in LA projection neurons using whole-cell patch-clamp recordings in slices of the rat amygdala in vitro. Application of NPY activated a membrane K(+) current with inwardly rectifying properties in 92% of tested neurons. Pharmacological properties were indicative of mediation via Y1 receptors. Nonhydrolyzable analogues of guanine nucleotides and SCH23390 blocked the NPY-activated current. Single-cell RT-PCR demonstrated expression of G-protein-coupled inwardly rectifying K(+) channel (GIRK) subunits GIRK1, GIRK2 and GIRK3, suggesting mediation of the NPY response through GIRK type channels. The NPY-activated current depressed action potential firing in LA projection neurons, through membrane hyperpolarization and decreased input resistance. Functionally, the dampening of excitability in projection neurons of the amygdala may contribute to the decrease in anxiogenic behavior during action of NPY.
Collapse
Affiliation(s)
- Ludmila Sosulina
- Institut für Physiologie I, Westfälische Wilhelms-Universität Münster, Münster, Germany
| | | | | | | |
Collapse
|
29
|
Abstract
Although often overshadowed by factors influencing seizure initiation, seizure termination is a critical step in the return to the interictal state. Understanding the mechanisms contributing to seizure termination could potentially identify novel targets for anticonvulsant drug development and may also highlight the pathophysiological processes contributing to seizure initiation. In this article, we review known physiological mechanisms contributing to seizure termination and discuss additional mechanisms that are likely to be relevant even though specific data are not yet available. This review is organized according to successively increasing "size scales"-from membranes to synapses to networks to circuits. We first discuss mechanisms of seizure termination acting at the shortest distances and affecting the excitable membranes of neurons in the seizure onset zone. Next we consider the contributions of ensembles of neurons and glia interacting at intermediate distances within the region of the seizure onset zone. Lastly, we consider the contribution of brain nuclei, such as the substantia nigra pars reticulata (SNR), that are capable of modulating seizures and exert their influence over the seizure onset zone (and neighboring areas) from a relatively great-in neuroanatomical terms-distance. It is our hope that the attention to the mechanisms contributing to seizure termination will stimulate novel avenues of epilepsy research and will contribute to improved patient care.
Collapse
Affiliation(s)
- Fred A Lado
- The Saul R. Korey Department of Neurology, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, NY 10461, USA.
| | | |
Collapse
|
30
|
Noè F, Pool AH, Nissinen J, Gobbi M, Bland R, Rizzi M, Balducci C, Ferraguti F, Sperk G, During MJ, Pitkänen A, Vezzani A. Neuropeptide Y gene therapy decreases chronic spontaneous seizures in a rat model of temporal lobe epilepsy. Brain 2008; 131:1506-15. [DOI: 10.1093/brain/awn079] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
31
|
Mouri G, Jimenez-Mateos E, Engel T, Dunleavy M, Hatazaki S, Paucard A, Matsushima S, Taki W, Henshall DC. Unilateral hippocampal CA3-predominant damage and short latency epileptogenesis after intra-amygdala microinjection of kainic acid in mice. Brain Res 2008; 1213:140-51. [PMID: 18455706 DOI: 10.1016/j.brainres.2008.03.061] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2008] [Revised: 03/18/2008] [Accepted: 03/18/2008] [Indexed: 02/08/2023]
Abstract
Mesial temporal lobe epilepsy is the most common, intractable seizure disorder in adults. It is associated with an asymmetric pattern of hippocampal neuron loss within the endfolium (hilus and CA3) and CA1, with limited pathology in extra-hippocampal regions. We previously developed a model of focally-evoked seizure-induced neuronal death using intra-amygdala kainic acid (KA) microinjection and characterized the acute hippocampal pathology. Here, we sought to characterize the full extent of hippocampal and potential extra-hippocampal damage in this model, and the temporal onset of epileptic seizures. Seizure damage assessed at four stereotaxic levels by FluoroJade B staining was most prominent in ipsilateral hippocampal CA3 where it extended from septal to temporal pole. Minor but significant neuronal injury was present in ipsilateral CA1. Extra-hippocampal neuronal damage was generally limited in extent and restricted to the lateral septal nucleus, injected amygdala and select regions of neocortex ipsilateral to the seizure elicitation side. Continuous surface EEG recorded with implanted telemetry units in freely-moving mice detected spontaneous, epileptic seizures by five days post-KA in all mice. Epileptic seizure number averaged 1-4 per day. Hippocampi from epileptic mice 15 days post-KA displayed unilateral CA3 lesions, astrogliosis and increased neuropeptide Y immunoreactivity suggestive of mossy fiber rearrangement. These studies characterize a mouse model of unilateral hippocampal-dominant neuronal damage and short latency epileptogenesis that may be suitable for studying the cell and molecular pathogenesis of human mesial temporal lobe epilepsy.
Collapse
Affiliation(s)
- Genshin Mouri
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin, 2, Ireland
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Sørensen AT, Kanter-Schlifke I, Carli M, Balducci C, Noe F, During MJ, Vezzani A, Kokaia M. NPY gene transfer in the hippocampus attenuates synaptic plasticity and learning. Hippocampus 2008; 18:564-74. [DOI: 10.1002/hipo.20415] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
33
|
Meurs A, Clinckers R, Ebinger G, Michotte Y, Smolders I. Sigma 1 receptor-mediated increase in hippocampal extracellular dopamine contributes to the mechanism of the anticonvulsant action of neuropeptide Y. Eur J Neurosci 2007; 26:3079-92. [PMID: 18005069 DOI: 10.1111/j.1460-9568.2007.05911.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The potent anticonvulsant properties of neuropeptide Y (NPY) are generally attributed to a Y2 receptor-mediated inhibition of glutamatergic synaptic transmission. Independent studies have shown that NPY increases brain dopamine content, possibly via interaction with sigma 1 receptors. Recently, we showed that increased extracellular hippocampal dopamine attenuates pilocarpine-induced limbic seizures via activation of hippocampal D2 receptors. Our aim in this study was to elucidate the role of increased hippocampal dopamine in the mechanism of the anticonvulsant action of NPY and to investigate the involvement of Y2 and sigma 1 receptors in this process. Limbic seizures were evoked in freely moving rats by intrahippocampal administration of pilocarpine via a microdialysis probe. NPY was administered intracerebroventricularly, intrahippocampally via the microdialysis probe, or coadministered intrahippocampally with the D2 receptor antagonist remoxipride, the Y2 receptor antagonist BIIE0246 or the sigma 1 receptor antagonist BD1047. Changes in hippocampal extracellular dopamine were monitored, and behavioural changes indicative of seizure activity were scored. Intracerebroventricular (10 nmol/3 microL) and intrahippocampal (20-50 microm) NPY administration increased hippocampal dopamine and attenuated pilocarpine-induced seizures. Hippocampal D2 receptor blockade (4 microm remoxipride) reversed the anticonvulsant effect of NPY. Y2 receptor blockade (1 microm BIIE0246) reversed the anticonvulsant effect of NPY but did not prevent NPY-induced increases in hippocampal dopamine. Sigma 1 receptor blockade (10 microm BD1047) abolished NPY-induced increases in hippocampal dopamine and reversed the anticonvulsant effect of NPY. Our results indicate that NPY-induced increases in hippocampal dopamine are mediated via sigma 1 receptors and contribute to the anticonvulsant effect of NPY via increased activation of hippocampal D2 receptors. This novel mechanism of anticonvulsant action of NPY is separate from, and may be complementary to, the well established Y2 receptor-mediated inhibition of hippocampal excitability.
Collapse
Affiliation(s)
- Alfred Meurs
- Department of Neurology, U. Z. Brussel, Laarbeeklaan 101,1090 Brussels, Belgium
| | | | | | | | | |
Collapse
|
34
|
Foti S, Haberman RP, Samulski RJ, McCown TJ. Adeno-associated virus-mediated expression and constitutive secretion of NPY or NPY13-36 suppresses seizure activity in vivo. Gene Ther 2007; 14:1534-6. [PMID: 17713567 PMCID: PMC3557464 DOI: 10.1038/sj.gt.3303013] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neuropeptide Y (NPY) is a 36-amino-acid peptide that attenuates seizure activity following direct infusion or adeno-associated virus (AAV)-mediated expression in the central nervous system. However, NPY activates all NPY receptor subtypes, potentially causing unwanted side effects. NPY13-36 is a C-terminal peptide fragment of NPY that primarily activates the NPY Y2 receptor, thought to mediate the antiseizure activity. Therefore, we investigated if recombinant adeno-associated virus-mediated expression and constitutive secretion of NPY or NPY13-36 could alter limbic seizure sensitivity. Rats received bilateral piriform cortex infusions of AAV vectors that express and constitutively secrete full-length NPY (AAV-FIB-NPY) or NPY13-36 (AAV-FIB-NPY13-36). Control rats received no infusion, as we have previously shown that vectors expressing and secreting reporter genes like GFP (AAV-FIB-EGFP), as well as vectors expressing peptides that lack secretion sequences (AAV-GAL) have no effect on seizures. One week later, all animals received kainic acid (10 mg kg(-1), intraperitoneally), and the latencies to wet dog shakes and limbic seizure behaviors were determined. Although both control and vector-treated rats developed wet dog shake behaviors with similar latencies, the latencies to class III and class IV limbic seizures were significantly prolonged in both NPY- and NPY13-36-treated groups. Thus, AAV-mediated expression and constitutive secretion of NPY and NPY13-36 is effective in attenuating limbic seizures, and provides a platform for delivering therapeutic peptide fragments with increased receptor selectivity.
Collapse
Affiliation(s)
- S Foti
- Curriculum in Neurobiology, University of North Carolina School of Medicine, Chapel Hill, NC, USA
| | - RP Haberman
- Department of Psychological and Brain Sciences, Johns Hopkins University, Baltimore, MD, USA
| | - RJ Samulski
- UNC Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - TJ McCown
- UNC Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
35
|
Morris MJ, Gannan E, Stroud LM, Beck-Sickinger AG, O'Brien TJ. Neuropeptide Y suppresses absence seizures in a genetic rat model primarily through effects on Y2 receptors. Eur J Neurosci 2007; 25:1136-43. [PMID: 17331209 DOI: 10.1111/j.1460-9568.2007.05348.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Neuropeptide Y (NPY) potently suppresses absence seizures in a model of genetic generalized epilepsy, genetic absence epilepsy rats of Strasbourg (GAERS). Here we investigated the Y-receptor subtype(s) on which NPY exerts this anti-absence effect. A dual in vivo approach was used: the cumulative duration of seizures was quantified in adult male GAERS in 90-min electroencephalogram recordings following intracerebroventricular (i.c.v.) injection of: (i) subtype-selective agonists of Y1 ([Leu31Pro34]NPY, 2.5 nmol), Y2 (Ac[Leu(28,31)]NPY24-36, 3 nmol), Y5 receptors [hPP1(-17),Ala31,Aib32]NPY, 4 nmol), NPY (3 nmol) or vehicle; and following (ii) i.c.v. injection of antagonists of Y1 (BIBP3226, 20 nmol), Y2 (BIIE0246, 20 nmol) and Y5 (NPY5RA972, 20 nmol) receptors or vehicle, followed by NPY (3 nmol). Injection of the Y1- and Y5-selective agonists resulted in significantly less mean seizure suppression (37.4% and 53.9%, respectively) than NPY (83.2%; P < 0.05), while the Y2 agonist had similar effects to NPY (62.3% suppression, P = 0.57). Food intake was not increased following injection of the Y2 agonist, while significant increases in food intake were seen following NPY and the other Y-subtype agonists. Compared with vehicle, NPY injection suppressed seizures following the Y1 and Y5 antagonists (45.3% and 80.1%, respectively, P < 0.05), but not following the Y2 antagonist (5.1% suppression, P = 0.46). We conclude that NPY Y2 receptors are more important than Y1 and Y5 receptors in mediating the effect of NPY to suppress absence seizures in a genetic rat model. Y2 receptor agonists may represent targets for novel drugs against genetic generalized epilepsies without resulting in appetite stimulation.
Collapse
Affiliation(s)
- Margaret J Morris
- Department of Physiology and Pharmacology, University of New South Wales, Kensington, New South Wales, Australia, 2052.
| | | | | | | | | |
Collapse
|
36
|
Xapelli S, Silva AP, Ferreira R, Malva JO. Neuropeptide Y can rescue neurons from cell death following the application of an excitotoxic insult with kainate in rat organotypic hippocampal slice cultures. Peptides 2007; 28:288-94. [PMID: 17212973 DOI: 10.1016/j.peptides.2006.09.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2006] [Accepted: 09/22/2006] [Indexed: 10/23/2022]
Abstract
In the present work we investigated the neuroprotective role of neuropeptide Y (NPY) after an excitotoxic insult in rat organotypic hippocampal slice cultures. Exposure of 2 week-old rat hippocampal slice cultures to 12muM kainate (KA) for 24h induced neuronal death in dentate gyrus (DG) granular cell layer, CA1 and CA3 pyramidal cell layers, as quantified by cellular propidium iodide (PI) uptake. The activation of Y(1) or Y(2) receptors 30min after starting the exposure to the excitotoxic insult with kainate resulted in neuroprotection by reducing the PI uptake in DG, CA1 and CA3 cell layers. The use of Y(1) or Y(2) receptors antagonists, BIBP3226 (1muM) or BIIE0246 (1muM), resulted in the loss of the neuroprotection induced by the activation of Y(1) or Y(2) receptors, respectively, in all hippocampal subfields. Taken together these results suggest that activation of NPY Y(1) or Y(2) receptors activates neuroprotective pathways that are able to rescue neurons from excitotoxic cell death.
Collapse
Affiliation(s)
- S Xapelli
- Center for Neuroscience and Cell Biology of Coimbra, 3004-517 Coimbra, Portugal
| | | | | | | |
Collapse
|
37
|
Nadler JV, Tu B, Timofeeva O, Jiao Y, Herzog H. Neuropeptide Y in the recurrent mossy fiber pathway. Peptides 2007; 28:357-64. [PMID: 17204350 PMCID: PMC1853293 DOI: 10.1016/j.peptides.2006.07.026] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2006] [Accepted: 07/14/2006] [Indexed: 10/23/2022]
Abstract
In the epileptic brain, hippocampal dentate granule cells become synaptically interconnected through the sprouting of mossy fibers. This new circuitry is expected to facilitate epileptiform discharge. Prolonged seizures induce the long-lasting neoexpression of neuropeptide Y (NPY) in mossy fibers. NPY is released spontaneously from recurrent mossy fiber terminals, reduces glutamate release from those terminals by activating presynaptic Y2 receptors, and depresses granule cell epileptiform activity dependent on the recurrent pathway. These effects are much greater in rats than in C57BL/6 mice, despite apparently equivalent mossy fiber sprouting and neoexpression of NPY. This species difference can be explained by contrasting changes in the expression of mossy fiber Y2 receptors; seizures upregulate Y2 receptors in rats but downregulate them in mice. The recurrent mossy fiber pathway may synchronize granule cell discharge more effectively in humans and mice than in rats, due to its lower expression of either NPY (humans) or Y2 receptors (mice).
Collapse
Affiliation(s)
- J Victor Nadler
- Department of Pharmacology and Cancer Biology and Department of Neurobiology, Box 3813, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | | | | | |
Collapse
|
38
|
Silva AP, Lourenço J, Xapelli S, Ferreira R, Kristiansen H, Woldbye DPD, Oliveira CR, Malva JO. Protein kinase C activity blocks neuropeptide Y-mediated inhibition of glutamate release and contributes to excitability of the hippocampus in status epilepticus. FASEB J 2006; 21:671-81. [PMID: 17167071 DOI: 10.1096/fj.06-6163com] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The unbalanced excitatory/inhibitory neurotransmitter function in the neuronal network afflicted by seizures is the main biochemical and biophysical hallmark of epilepsy. The aim of this work was to identify changes in the signaling mechanisms associated with neuropeptide Y (NPY)-mediated inhibition of glutamate release that may contribute to hyperexcitability. Using isolated rat hippocampal nerve terminals, we showed that the KCl-evoked glutamate release is inhibited by NPY Y2 receptor activation and is potentiated by the stimulation of protein kinase C (PKC). Moreover, we observed that immediately after status epilepticus (6 h postinjection with kainate, 10 mg/kg), the functional inhibition of glutamate release by NPY Y2 receptors was transiently blocked concomitantly with PKC hyperactivation. The pharmacological blockade of seizure-activated PKC revealed again the Y2 receptor-mediated inhibition of glutamate release. The functional activity of PKC immediately after status epilepticus was assessed by evaluating phosphorylation of the AMPA receptor subunit GluR1 (Ser-831), a substrate for PKC. Moreover, NPY-stimulated [35S]GTPgammaS autoradiographic binding studies indicated that the common target for Y2 receptor and PKC on the inhibition/potentiation of glutamate release was located downstream of the Y2 receptor, or its interacting G-protein, and involves voltage-gated calcium channels.
Collapse
Affiliation(s)
- Ana P Silva
- Institute of Pharmacology and Therapeutics, University of Coimbra, 3004-504 Coimbra, Portugal
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Gorter JA, van Vliet EA, Aronica E, Breit T, Rauwerda H, Lopes da Silva FH, Wadman WJ. Potential new antiepileptogenic targets indicated by microarray analysis in a rat model for temporal lobe epilepsy. J Neurosci 2006; 26:11083-110. [PMID: 17065450 PMCID: PMC6674659 DOI: 10.1523/jneurosci.2766-06.2006] [Citation(s) in RCA: 254] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
To get insight into the mechanisms that may lead to progression of temporal lobe epilepsy, we investigated gene expression during epileptogenesis in the rat. RNA was obtained from three different brain regions [CA3, entorhinal cortex (EC), and cerebellum (CB)] at three different time points after electrically induced status epilepticus (SE): acute phase [group D (1 d)], latent period [group W (1 week)], and chronic epileptic period [group M (3-4 months)]. A group that was stimulated but that had not experienced SE and later epilepsy was also included (group nS). Gene expression analysis was performed using the Affymetrix Gene Chip System (RAE230A). We used GENMAPP and Gene Ontology to identify global biological trends in gene expression data. The immune response was the most prominent process changed during all three phases of epileptogenesis. Synaptic transmission was a downregulated process during the acute and latent phases. GABA receptor subunits involved in tonic inhibition were persistently downregulated. These changes were observed mostly in both CA3 and EC but not in CB. Rats that were stimulated but that did not develop spontaneous seizures later on had also some changes in gene expression, but this was not reflected in a significant change of a biological process. These data suggest that the targeting of specific genes that are involved in these biological processes may be a promising strategy to slow down or prevent the progression of epilepsy. Especially genes related to the immune response, such as complement factors, interleukins, and genes related to prostaglandin synthesis and coagulation pathway may be interesting targets.
Collapse
Affiliation(s)
- Jan A Gorter
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, 1098 SM, Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
40
|
Eva C, Serra M, Mele P, Panzica G, Oberto A. Physiology and gene regulation of the brain NPY Y1 receptor. Front Neuroendocrinol 2006; 27:308-39. [PMID: 16989896 DOI: 10.1016/j.yfrne.2006.07.002] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2005] [Revised: 07/18/2006] [Accepted: 07/25/2006] [Indexed: 10/24/2022]
Abstract
Neuropeptide Y (NPY) is one of the most prominent and abundant neuropeptides in the mammalian brain where it interacts with a family of G-protein coupled receptors, including the Y(1) receptor subtype (Y(1)R). NPY-Y(1)R signalling plays a prominent role in the regulation of several behavioural and physiological functions including feeding behaviour and energy balance, sexual hormone secretion, stress response, emotional behaviour, neuronal excitability and ethanol drinking. Y(1)R expression is regulated by neuronal activity and peripheral hormones. The Y(1)R gene has been isolated from rodents and humans and it contains multiple regulatory elements that may participate in the regulation of its expression. Y(1)R expression in the hypothalamus is modulated by changes in energetic balance induced by a wide variety of conditions (fasting, pregnancy, hyperglycaemic challenge, hypophagia, diet induced obesity). Estrogens up-regulate responsiveness to NPY to stimulate preovulatory GnRH and gonadotropin surges by increasing Y(1)R gene expression both in the hypothalamus and the pituitary. Y(1)R expression is modulated by different kinds of brain insults, such as stress and seizure activity, and alteration in its expression may contribute to antidepressant action. Chronic modulation of GABA(A) receptor function by benzodiazepines or neuroactive steroids also affects Y(1)R expression in the amygdala, suggesting that a functional interaction between the GABA(A) receptor and Y(1)R mediated signalling may contribute to the regulation of emotional behaviour. In this paper, we review the state of the art concerning Y(1)R function and gene expression, including our personal contribution to many of the subjects mentioned above.
Collapse
Affiliation(s)
- Carola Eva
- Sezione di Farmacologia, Dipartimento di Anatomia, Farmacologia e Medicina Legale, Università di Torino, Italy; Centro Rita Levi Montalcini, Università di Torino, Italy.
| | | | | | | | | |
Collapse
|
41
|
Eva C, Oberto A, Mele P, Serra M, Biggio G. Role of brain neuroactive steroids in the functional interplay between the GABA(A) and the NPY-Y1 receptor mediated signals in the amygdala. Pharmacol Biochem Behav 2006; 84:568-80. [PMID: 16824587 DOI: 10.1016/j.pbb.2006.05.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2006] [Accepted: 05/17/2006] [Indexed: 11/18/2022]
Abstract
Various lines of evidence suggest a functional interaction between GABA(A) and Neuropeptide Y (NPY)-Y(1) receptor (Y(1)R) mediated transmissions in various brain regions, which can be important in the regulation of sedation, feeding, anxious behaviour and neuronal excitability. By using a transgenic mouse model carrying the murine Y(1)R gene promoter fused to the lacZ reporter gene (Y(1)R/LacZ mice), we showed that prolonged pharmacologically or physiologically induced changes in the cerebrocortical concentrations of the neuroactive steroids 3alpha-hydroxy-5alpha-pregnan- 20-one (3alpha,5alpha TH PROG) and tetrahydrodeoxycorticosterone (3alpha,5alpha TH DOC) increases Y(1)R/LacZ transgene expression in the central and medial amygdala, an effect similar to that induced by long-term treatment with positive modulators of the GABA(A) receptor complex (diazepam or abecarnil). We also demonstrated that fluctuations in the cerebrocortical concentrations of 3alpha,5alpha-TH PROG and 3alpha,5alpha TH DOC during voluntary ethanol consumption and ethanol withdrawal induces a marked increase in Y(1)R gene expression that becomes apparent 48 h after withdrawal. These data provide evidence that neuroactive steroids may play an important role in the functional interaction between the GABA(A) receptor and NPY-Y(1)R mediated pathways in the amygdala, which might represent an important regulatory mechanism for modulation of several functions, including ethanol withdrawal.
Collapse
Affiliation(s)
- Carola Eva
- Dipartimento di Anatomia, Farmacologia e Medicina Legale, Sezione di Farmacologia, Via Pietro Giuria 13, 10125, Università di Torino, Torino, Italy.
| | | | | | | | | |
Collapse
|
42
|
Lin EJD, Young D, Baer K, Herzog H, During MJ. Differential actions of NPY on seizure modulation via Y1 and Y2 receptors: evidence from receptor knockout mice. Epilepsia 2006; 47:773-80. [PMID: 16650144 DOI: 10.1111/j.1528-1167.2006.00500.x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE Neuropeptide Y (NPY) has been shown to modulate seizure activities. To provide further understanding of the involvement of two of the most abundantly expressed NPY receptors, Y1 and Y2, we assessed the effect of Y1 and Y2 gene deletion on systemic kainic acid-induced seizures. We also examined the effect of rAAV-mediated hippocampal NPY overexpression on seizure susceptibility in these receptor knockout mice. METHODS Recombinant adeno-associated viral vector overexpressing NPY (rAAV-NPY) or an empty vector control (rAAV-Empty) was injected into the hippocampus of adult C57BL/6-129/SvJ wild-type male mice and mice deficient of Y1 or Y2 receptors on the same background. Four weeks after vector injection, mice were subjected to systemic kainic acid-induced seizures, and the seizure behaviors were scored. RESULTS The rAAV-mediated hippocampal overexpression of NPY led to a twofold reduction in seizures induced by systemic kainic acid in wild-type mice and Y1 receptor knockout mice but not in mice deficient of Y2 receptors. A differential action by the receptors was observed in the seizure-induced mortality rate, with increased fatality in Y2-/- mice. In addition, although NPY overexpression did not significantly affect the mortality rate in Y2-/- and wild-type mice, it abolished KA-induced mortality in Y1-/-mice. CONCLUSIONS This study shows for the first time an altered susceptibility to chemically induced seizures in Y1 and Y2 knockout mice and demonstrates a differential seizure modulation mediated by these receptors via a genetic approach.
Collapse
Affiliation(s)
- En-Ju Deborah Lin
- Department of Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand.
| | | | | | | | | |
Collapse
|
43
|
Sapunar D, Modric-Jednacak K, Grkovic I, Michalkiewicz M, Hogan QH. Effect of peripheral axotomy on pain-related behavior and dorsal root ganglion neurons excitability in NPY transgenic rats. Brain Res 2005; 1063:48-58. [PMID: 16259969 DOI: 10.1016/j.brainres.2005.09.019] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2005] [Revised: 09/19/2005] [Accepted: 09/25/2005] [Indexed: 11/30/2022]
Abstract
In order to clarify the physiologic role of NPY in sensory processing, we obtained intracellular recordings of DRG neurons from wild type (WT) and NPY overexpressing transgenic rats (NPY-TG) before and after injury. We investigated medium and large diameter DRG neurons since upregulation of NPY peptide following the nerve injury occurs primarily in those cells. Neurons were classified as Aalpha/beta and Adelta using conduction velocity and action potential duration. Prior to the injury, Aalpha/beta neurons of NPY-TG rats conducted more slowly and had a more brief AHP than similar cells from the WT group. Adelta neurons at baseline conducted faster in TG animals compared to WT. Ligation of the 5th lumbar spinal nerve (SNL) produced certain changes in Aalpha/beta cells that were evident only in the TG group. These include increased refractory period, increased input resistance, AHP prolongation and a depolarizing shift in threshold for AP initiation. The expected injury-induced CV slowing was not seen in NPY-TG Aalpha/beta cells. In the Adelta cell group, injury produced a depolarizing shift in the resting membrane potential, an increase in AP duration and decrease in AHP and refractory period duration only in WT rats, while NPY-TG cells lacked these injury-induced changes. Behavior tests showed diminished sensory response to nerve injury in NPY-TG rats, i.e., shorter duration of enhanced pain-related behavior and attenuation of contralateral effect. In conclusion, our observations suggest that NPY overexpression leads to reduced neuronal activity following nerve injury in a cell-specific manner.
Collapse
Affiliation(s)
- Damir Sapunar
- Department of Anatomy, Histology and Embryology, University of Split Medical School, PAK, KB Split, Spinciceva 1, 21000 Split, Croatia.
| | | | | | | | | |
Collapse
|
44
|
Costantin L, Bozzi Y, Richichi C, Viegi A, Antonucci F, Funicello M, Gobbi M, Mennini T, Rossetto O, Montecucco C, Maffei L, Vezzani A, Caleo M. Antiepileptic effects of botulinum neurotoxin E. J Neurosci 2005; 25:1943-51. [PMID: 15728834 PMCID: PMC6726074 DOI: 10.1523/jneurosci.4402-04.2005] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Experimental studies suggest that the delivery of antiepileptic agents into the seizure focus might be of potential utility for the treatment of focal-onset epilepsies. Botulinum neurotoxin E (BoNT/E) causes a prolonged inhibition of neurotransmitter release after its specific cleavage of the synaptic protein synaptosomal-associated protein of 25 kDa (SNAP-25). Here, we show that BoNT/E injected into the rat hippocampus inhibits glutamate release and blocks spike activity of pyramidal neurons. BoNT/E effects persist for at least 3 weeks, as determined by immunodetection of cleaved SNAP-25 and loss of intact SNAP-25. The delivery of BoNT/E to the rat hippocampus dramatically reduces both focal and generalized kainic acid-induced seizures as documented by behavioral and electrographic analysis. BoNT/E treatment also prevents neuronal loss and long-term cognitive deficits associated with kainic acid seizures. Moreover, BoNT/E-injected rats require 50% more electrical stimulations to reach stage 5 of kindling, thus indicating a delayed epileptogenesis. We conclude that BoNT/E delivery to the hippocampus is both antiictal and antiepileptogenic in experimental models of epilepsy.
Collapse
MESH Headings
- Animals
- Anticonvulsants/administration & dosage
- Anticonvulsants/therapeutic use
- Botulinum Toxins/administration & dosage
- Botulinum Toxins/therapeutic use
- Cell Death/drug effects
- Cognition Disorders/etiology
- Cognition Disorders/prevention & control
- Convulsants/toxicity
- Drug Evaluation, Preclinical
- Electric Stimulation
- Electroencephalography
- Epilepsies, Partial/drug therapy
- Epilepsies, Partial/physiopathology
- Epilepsy, Generalized/chemically induced
- Epilepsy, Generalized/complications
- Epilepsy, Generalized/drug therapy
- Epilepsy, Generalized/physiopathology
- Glutamic Acid/metabolism
- Hippocampus/drug effects
- Hippocampus/physiopathology
- Injections, Intralesional
- Kainic Acid/toxicity
- Kindling, Neurologic/drug effects
- Maze Learning/drug effects
- Membrane Proteins/metabolism
- Nerve Tissue Proteins/metabolism
- Pyramidal Cells/drug effects
- Pyramidal Cells/pathology
- Pyramidal Cells/physiology
- Random Allocation
- Rats
- Rats, Long-Evans
- Stereotaxic Techniques
- Synaptosomal-Associated Protein 25
Collapse
Affiliation(s)
- Laura Costantin
- Scuola Normale Superiore, Consiglio Nazionale delle Ricerche, 56100 Pisa, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
The generation of viral vectors, such as adeno-associated virus (AAV) and lentivirus, which are capable of stable transduction of neurons, offers an attractive strategy for introducing novel genes into the brain, resulting in a long-lasting production of specific proteins. An alternative approach to achieving transgene expression in brain is to graft cells that are genetically engineered to produce neuroactive substances. Neuroactive peptides, adenosine, and gamma-aminobutyric acid, are agents that can be delivered by gene and cell therapy with potential utility in epilepsy therapy.
Collapse
|
46
|
El Bahh B, Balosso S, Hamilton T, Herzog H, Beck-Sickinger AG, Sperk G, Gehlert DR, Vezzani A, Colmers WF. The anti-epileptic actions of neuropeptide Y in the hippocampus are mediated by Y2 and not Y5 receptors. Eur J Neurosci 2005; 22:1417-30. [PMID: 16190896 DOI: 10.1111/j.1460-9568.2005.04338.x] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Neuropeptide Y (NPY) potently inhibits glutamate release and seizure activity in rodent hippocampus in vitro and in vivo, but the nature of the receptor(s) mediating this action is controversial. In hippocampal slices from rats and several wild-type mice, a Y2-preferring agonist mimicked, and the Y2-specific antagonist BIIE0246 blocked, the NPY-mediated inhibition both of glutamatergic transmission and of epileptiform discharges in two different slice models of temporal lobe epilepsy, stimulus train-induced bursting (STIB) and 0-Mg2+ bursting. Whereas Y5 receptor-preferring agonists had small but significant effects in vitro, they were blocked by BIIE0246, and a Y5 receptor-specific antagonist did not affect responses to any agonist tested in any preparation. In slices from mice, NPY was without effect on evoked potentials or in either of the two slice seizure models. In vivo, intrahippocampal injections of Y2- or Y5-preferring agonists inhibited seizures caused by intrahippocampal kainate, but again the Y5 agonist effects were insensitive to a Y5 antagonist. Neither Y2- nor Y5-preferring agonists affected kainate seizures in mice. A Y5-specific antagonist did not displace the binding of two different NPY ligands in WT or mice, whereas all NPY binding was eliminated in the mouse. Thus, we show that Y2 receptors alone mediate all the anti-excitatory actions of NPY seen in the hippocampus, whereas our findings do not support a role for Y5 receptors either in vitro or in vivo. The results suggest that agonists targeting the Y2 receptor may be useful anticonvulsants.
Collapse
Affiliation(s)
- Bouchaïb El Bahh
- Department of Pharmacology, University of Alberta. Edmonton, Alberta, Canada T6G 2H7
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Primeaux SD, Wilson SP, Cusick MC, York DA, Wilson MA. Effects of altered amygdalar neuropeptide Y expression on anxiety-related behaviors. Neuropsychopharmacology 2005; 30:1589-97. [PMID: 15770236 DOI: 10.1038/sj.npp.1300705] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neuropeptide Y (NPY) decreases anxiety-related behaviors in various animal models of anxiety. The purpose of the present study was to examine the role of the amygdalar NPY system in anxiety-related responses in the elevated plus maze. The first experiment determined if herpes virus-mediated alterations in amygdalar NPY levels would alter anxiety-related behaviors in the elevated plus maze. Viral vectors encoding NPY, NPY antisense, or LacZ (control virus) were bilaterally injected into the amygdala, and 4 days postinjection, rats were tested in the elevated plus maze test. NPY-like immunoreactivity (NPY-ir) was measured in the amygdala of these rats. In rats injected with the viral vector encoding NPY, reduced anxiety-related behaviors in the elevated plus maze accompanied by moderate increases in NPY-ir were detected compared to NPY-antisense viral vector-treated subjects. Elevated plus maze behavior did not differ compared to LacZ-treated controls. NPY overexpression at this time point was also suggested by enhanced NPY mRNA expression seen in the amygdala 4 days postinjection using real-time polymerase chain reaction analysis. Experiment 2 was conducted to provide further evidence for a role of amygdalar NPY in regulating anxiety-related behaviors in the elevated plus maze test. The nonpeptide NPY Y1 receptor antagonist, BIBP 3226 (1.5 microg/microl), was bilaterally injected into the amygdala and rats were tested in the elevated plus maze test. Rats receiving BIBP 3226 exhibited increased anxiety-related behaviors in this test. The results of these experiments provide further support for the role of amygdalar NPY in anxiety-related behaviors.
Collapse
Affiliation(s)
- Stefany D Primeaux
- Department of Pharmacology, Physiology and Neuroscience, University of South Carolina School of Medicine, Columbia, SC, USA.
| | | | | | | | | |
Collapse
|
48
|
Wilson DN, Chung H, Elliott RC, Bremer E, George D, Koh S. Microarray analysis of postictal transcriptional regulation of neuropeptides. J Mol Neurosci 2005; 25:285-98. [PMID: 15800381 DOI: 10.1385/jmn:25:3:285] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2004] [Accepted: 11/12/2004] [Indexed: 11/11/2022]
Abstract
Unlike adults, kainic acid (KA)-induced status epilepticus (SE) in immature rats causes neither cell death nor recurrent spontaneous seizures. To elucidate the mechanisms of these distinct responses, transcriptional changes in neuropeptides were examined following KA-induced SE. We aimed to determine whether neuropeptides with anticonvulsant/neuroprotective properties were preferentially increased in immature rats while those with a proconvulsant/neurotoxic role were elevated to a greater extent in mature rats. We used high-density oligonucleotide gene arrays and directly compared transcriptional regulation of seven select neuropeptides at P15 and P30 over five time points. Total RNAs were isolated from hippocampi of 12 animals and pooled to hybridize to triplicate Affymetrix Genechips. Microarray results were validated by real-time quantitative RT-PCR (qRT-PCR). Independent individual RNA samples were purified for triplicate runs of qRT-PCR. Neuropeptides are significantly regulated by seizures in both immature and mature hippocampus. The magnitude of increase is significantly higher at P30 compared with that at P15, not only for neuropeptides with neurotoxic/proconvulsant properties but also for those with neuroprotective/ anticonvulsant properties. Galanin is induced at 24 h only in P30 rats. CST shows high expression in immature hippocampus and is further increased after KA-induced SE only in P15. The expression trends seen in the microarray data are confirmed by qRT-PCR for all six neuropeptides analyzed. CST might play a neuroprotective role in immature rats, and its overexpression might prevent neuronal loss after seizure in adults. Also, suppression of tachykinin and corticotropin-releasing hormone might be effective in alleviating seizure-induced neuronal damage.
Collapse
Affiliation(s)
- Dawn N Wilson
- Division of Neurology, Children's Memorial Hospital, Feinberg School of Medicine, Northwestern University, Chicago, IL 60614, USA
| | | | | | | | | | | |
Collapse
|
49
|
Woldbye DPD, Nanobashvili A, Sørensen AT, Husum H, Bolwig TG, Sørensen G, Ernfors P, Kokaia M. Differential suppression of seizures via Y2 and Y5 neuropeptide Y receptors. Neurobiol Dis 2005; 20:760-72. [PMID: 15979311 DOI: 10.1016/j.nbd.2005.05.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2004] [Revised: 04/14/2005] [Accepted: 05/12/2005] [Indexed: 10/25/2022] Open
Abstract
Neuropeptide Y (NPY) prominently inhibits epileptic seizures in different animal models. The NPY receptors mediating this effect remain controversial partially due to lack of highly selective agonists and antagonists. To circumvent this problem, we used various NPY receptor knockout mice with the same genetic background and explored anti-epileptic action of NPY in vitro and in vivo. In Y2 (Y2-/-) and Y5 (Y5-/-) receptor knockouts, NPY partially inhibited 0 Mg2+-induced epileptiform activity in hippocampal slices. In contrast, in double knockouts (Y2Y5-/-), NPY had no effect, suggesting that in the hippocampus in vitro both receptors mediate anti-epileptiform action of NPY in an additive manner. Systemic kainate induced more severe seizures in Y5-/- and Y2Y5-/-, but not in Y2-/- mice, as compared to wild-type mice. Moreover, kainate seizures were aggravated by administration of the Y5 antagonist L-152,804 in wild-type mice. In Y5-/- mice, hippocampal kindling progressed faster, and afterdischarge durations were longer in amygdala, but not in hippocampus, as compared to wild-type controls. Taken together, these data suggest that, in mice, both Y2 and Y5 receptors regulate hippocampal seizures in vitro, while activation of Y5 receptors in extra-hippocampal regions reduces generalized seizures in vivo.
Collapse
Affiliation(s)
- David P D Woldbye
- Section of Restorative Neurology, Wallenberg Neuroscience Center, BMC A-11, Lund University Hospital, S-221 84 Lund, Sweden
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Silva AP, Xapelli S, Pinheiro PS, Ferreira R, Lourenço J, Cristóvão A, Grouzmann E, Cavadas C, Oliveira CR, Malva JO. Up-regulation of neuropeptide Y levels and modulation of glutamate release through neuropeptide Y receptors in the hippocampus of kainate-induced epileptic rats. J Neurochem 2005; 93:163-70. [PMID: 15773916 DOI: 10.1111/j.1471-4159.2004.03005.x] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Kainate-induced epilepsy has been shown to be associated with increased levels of neuropeptide Y (NPY) in the rat hippocampus. However, there is no information on how increased levels of this peptide might modulate excitation in kainate-induced epilepsy. In this work, we investigated the modulation of glutamate release by NPY receptors in hippocampal synaptosomes isolated from epileptic rats. In the acute phase of epilepsy, a transient decrease in the efficiency of NPY and selective NPY receptor agonists in inhibiting glutamate release was observed. Moreover, in the chronic epileptic hippocampus, a decrease in the efficiency of NPY and the Y(2) receptor agonist, NPY13-36, was also found. Simultaneously, we observed that the epileptic hippocampus expresses higher levels of NPY, which may account for an increased basal inhibition of glutamate release. Consistently, the blockade of Y(2) receptors increased KCl-evoked glutamate release, and there was an increase in Y(2) receptor mRNA levels 30 days after kainic acid injection, suggesting a basal effect of NPY through Y(2) receptors. Taken together, these results indicate that an increased function of the NPY modulatory system in the epileptic hippocampus may contribute to basal inhibition of glutamate release and control hyperexcitability.
Collapse
Affiliation(s)
- Ana P Silva
- Center for Neuroscience and Cell Biology, University of Coimbra, Portugal
| | | | | | | | | | | | | | | | | | | |
Collapse
|