1
|
Stephenson KA, Peters P, Rae MG, O'Malley D. Astrocyte proliferation in the hippocampal dentate gyrus is suppressed across the lifespan of dystrophin-deficient mdx mice. Exp Physiol 2025. [PMID: 39792584 DOI: 10.1113/ep092150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025]
Abstract
Absence of the structural protein, dystrophin, results in the neuromuscular disorder Duchenne Muscular Dystrophy (DMD). In addition to progressive skeletal muscle dysfunction, this multisystemic disorder can also result in cognitive deficits and behavioural changes that are likely to be consequences of dystrophin loss from central neurons and astrocytes. Dystrophin-deficient mdx mice exhibit decreases in grey matter volume in the hippocampus, the brain region that encodes and consolidates memories, and this is exacerbated with ageing. To understand changes in cellular composition that might underpin these age-related developments, we have compared neurogenesis and the prevalence of immunofluorescently identified newly born and mature neurons, astrocytes and microglia in the dentate gyrus of mdx and wild-type mice at 2, 4, 8 and 16 months of age. The number of adult-born neurons was suppressed in the dentate gyrus subgranular zone of 2-month-old mdx mice. However, the numbers of granule cells and GABAA receptor, alpha 1-expressing cells were similar in wild-type and mdx mice at all ages. Strikingly, the numbers of astrocytes, particularly in the dentate gyrus molecular layer, were suppressed in mdx mice at all time points. Thus, dystrophin loss was associated with reduced hippocampal neurogenesis in early life but did not impact the prevalence of mature neurons across the lifespan of mdx mice. In contrast, normal age-related dentate gyrus astrocyte proliferation was suppressed in dystrophic mice. Astrocytes are the most abundant cell type in the brain and are crucial in supporting neuronal function, such that loss of these cells is likely to contribute to hippocampal dysfunction reported in mdx mice.
Collapse
Affiliation(s)
| | - Polly Peters
- Department of Physiology, School of Medicine, University College Cork, Cork, Ireland
| | - Mark G Rae
- Department of Physiology, School of Medicine, University College Cork, Cork, Ireland
| | - Dervla O'Malley
- Department of Physiology, School of Medicine, University College Cork, Cork, Ireland
| |
Collapse
|
2
|
Liu J, Fang L, Gong C, Li J, Liu Y, Zeng P, Fan Y, Liu Y, Guo J, Wang L, Li Y. Neurotoxicity study of cenobamate-induced zebrafish early developmental stages. Toxicol Appl Pharmacol 2024; 495:117201. [PMID: 39667564 DOI: 10.1016/j.taap.2024.117201] [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: 09/19/2024] [Revised: 12/04/2024] [Accepted: 12/07/2024] [Indexed: 12/14/2024]
Abstract
Cenobamate (CNB) is a novel anti-seizure medication with significant efficacy in treating epilepsy. However, in clinical trials, the most common adverse reactions observed in patients are central nervous system (CNS) symptoms. In animal studies, administration of CNB during pregnancy or lactation has been associated with adverse effects on neurodevelopment in offspring. To optimize the clinical use of CNB, we investigated the neurotoxicity of different concentrations of CNB (10, 20, 40, 80, and 160 μM) on zebrafish embryos. Following exposure, zebrafish embryos exhibited abnormal phenotypes such as shortened body length, impaired yolk sac absorption, and decreased heart rate. Behavioral experiments showed that CNB caused abnormal movements such as decreased spontaneous tail curling frequency, shortened total movement distance, and reduced average movement speed. We also found that CNB leads to increased acetylcholinesterase (AChE) activity levels in zebrafish embryos, along with differential expression of neurodevelopment-related genes such as nestin, gfap, synapsin IIa, and gap43. In summary, our research findings indicated that CNB may induce developmental and neurotoxic effects in zebrafish embryos by altering neurotransmitter systems and the expression of neurodevelopmental genes, thereby influencing behavior. This study will provide information for the clinical use of CNB, aiming to benefit more epilepsy patients through its appropriate administration.
Collapse
Affiliation(s)
- Jiahao Liu
- Heilongjiang Provincial Key Laboratory of Child Neurorehabilitation, School of Rehabilitation Medicine, Jiamusi University, Jiamusi, Heilongjiang Province 154007, PR China
| | - Liya Fang
- Heilongjiang Provincial Key Laboratory of Child Neurorehabilitation, School of Rehabilitation Medicine, Jiamusi University, Jiamusi, Heilongjiang Province 154007, PR China
| | - Chao Gong
- Heilongjiang Provincial Key Laboratory of Child Neurorehabilitation, School of Rehabilitation Medicine, Jiamusi University, Jiamusi, Heilongjiang Province 154007, PR China
| | - Jiawei Li
- Heilongjiang Provincial Key Laboratory of Child Neurorehabilitation, School of Rehabilitation Medicine, Jiamusi University, Jiamusi, Heilongjiang Province 154007, PR China
| | - Yuanyuan Liu
- Heilongjiang Provincial Key Laboratory of Child Neurorehabilitation, School of Rehabilitation Medicine, Jiamusi University, Jiamusi, Heilongjiang Province 154007, PR China
| | - Pei Zeng
- The Third Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang Province 154007, PR China
| | - Yanping Fan
- The Third Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang Province 154007, PR China
| | - Yao Liu
- School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang Province 154007, PR China
| | - Jin Guo
- The Third Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang Province 154007, PR China.
| | - Luchuan Wang
- The First Affiliated Hospital of Jiamusi University, Jiamusi, Heilongjiang Province 154007, PR China.
| | - Yue Li
- Key laboratory of Microecology-immune Regulatory Network and Related Diseases School of Basic Medicine, Jiamusi University, Jiamusi, Heilongjiang Province 154000, PR China.
| |
Collapse
|
3
|
Alkadhi KA. Giant miniature endplate potentials at vertebrate neuromuscular junctions: A review. Eur J Neurosci 2024; 60:7183-7194. [PMID: 39600045 DOI: 10.1111/ejn.16624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/28/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
An unusually large amplitude spontaneous miniature endplate potentials (gMEPPs) occur naturally at low frequency at the vertebrate neuromuscular junction. Unlike the normal miniature endplate potentials (nMEPPs), these gMEPPs have long duration and long time to peak. More strikingly, gMEPPs seem to be independent of extracellular and intracellular Ca+2. and have a greater temperature sensitivity than nMEPPs. They are potentiated by tetrodotoxin but inhibited by acetylcholine (ACh) receptor blockers indicating ACh is the neurotransmitter responsible for gMEPPs. The frequency of gMEPPs is greatly increased in muscles weakened by various drugs, toxins or disease conditions suggesting that gMEPPs may be a part of possible neurotrophic mechanism to preserve effective neuromuscular transmission when the normal function is compromised.
Collapse
Affiliation(s)
- Karim A Alkadhi
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas, USA
| |
Collapse
|
4
|
Pol E, Côme E, Merlaud Z, Gouhier J, Russeau M, Scotto-Lomassese S, Moutkine I, Marques X, Lévi S. NKCC1 and KCC2 Chloride Transporters Have Different Membrane Dynamics on the Surface of Hippocampal Neurons. Cells 2023; 12:2363. [PMID: 37830575 PMCID: PMC10571912 DOI: 10.3390/cells12192363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/12/2023] [Accepted: 09/20/2023] [Indexed: 10/14/2023] Open
Abstract
Na-K-2Cl cotransporter 1 (NKCC1) regulates chloride influx in neurons and thereby GABAA receptor activity in normal and pathological conditions. Here, we characterized in hippocampal neurons the membrane expression, distribution and dynamics of exogenous NKCC1a and NKCC1b isoforms and compared them to those of the chloride extruder K-Cl cotransporter 2 (KCC2). We found that NKCC1a and NKCC1b behave quite similarly. NKCC1a/1b but not KCC2 are present along the axon initial segment where they are confined. Moreover, NKCC1a/1b are detected in the somato-dendritic compartment at a lower level than KCC2, where they form fewer, smaller and less compact clusters at perisynaptic and extrasynaptic sites. Interestingly, ~60% of dendritic clusters of NKCC1a/1b are colocalized with KCC2. They are larger and brighter than those devoid of KCC2, suggesting a particular NKCC1a/1b-KCC2 relationship. In agreement with the reduced dendritic clustering of NKCC1a/1b compared with that of KCC2, NKCC1a/1b are more mobile on the dendrite than KCC2, suggesting weaker cytoskeletal interaction. NKCC1a/b are confined to endocytic zones, where they spend more time than KCC2. However, they spend less time in these compartments than at the synapses, suggesting that they can rapidly leave endocytic zones to increase the membrane pool, which can happen in pathological conditions. Thus, NKCC1a/b have different membrane dynamics and clustering from KCC2, which helps to explain their low level in the neuronal membrane, while allowing a rapid increase in the membrane pool under pathological conditions.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Sabine Lévi
- Institut du Fer à Moulin, Institut National de la Santé Et de la Recherche Médicale (INSERM) UMR-S 1270, Sorbonne Université, 75005 Paris, France; (E.P.); (E.C.); (Z.M.); (J.G.); (M.R.); (S.S.-L.); (I.M.); (X.M.)
| |
Collapse
|
5
|
Rissardo JP, Fornari Caprara AL. Cenobamate (YKP3089) and Drug-Resistant Epilepsy: A Review of the Literature. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1389. [PMID: 37629678 PMCID: PMC10456719 DOI: 10.3390/medicina59081389] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/08/2023] [Accepted: 07/24/2023] [Indexed: 08/27/2023]
Abstract
Cenobamate (CNB), ([(R)-1-(2-chlorophenyl)-2-(2H-tetrazol-2-yl)ethyl], is a novel tetrazole alkyl carbamate derivative. In November 2019, the Food and Drug Administration approved Xcopri®, marketed by SK Life Science Inc., (Paramus, NJ, USA) for adult focal seizures. The European Medicines Agency approved Ontozry® by Arvelle Therapeutics Netherlands B.V.(Amsterdam, The Neatherlands) in March 2021. Cenobamate is a medication that could potentially change the perspectives regarding the management and prognosis of refractory epilepsy. In this way, this study aims to review the literature on CNB's pharmacological properties, pharmacokinetics, efficacy, and safety. CNB is a highly effective drug in managing focal onset seizures, with more than twenty percent of individuals with drug-resistant epilepsy achieving seizure freedom. This finding is remarkable in the antiseizure medication literature. The mechanism of action of CNB is still poorly understood, but it is associated with transient and persistent sodium currents and GABAergic neurotransmission. In animal studies, CNB showed sustained efficacy and potency in the 6 Hz test regardless of the stimulus intensity. CNB was revealed to be the most cost-effective drug among different third-generation antiseizure medications. Also, CNB could have neuroprotective effects. However, there are still concerns regarding its potential for abuse and suicidality risk, which future studies should clearly assess, after which protocols should be changed. The major drawback of CNB therapy is the slow and complex titration and maintenance phases preventing the wide use of this new agent in clinical practice.
Collapse
Affiliation(s)
- Jamir Pitton Rissardo
- Medicine Department, Federal University of Santa Maria, Santa Maria 97105-900, Brazil;
| | | |
Collapse
|
6
|
Lateral Diffusion of NKCC1 Contributes to Chloride Homeostasis in Neurons and Is Rapidly Regulated by the WNK Signaling Pathway. Cells 2023; 12:cells12030464. [PMID: 36766805 PMCID: PMC9914440 DOI: 10.3390/cells12030464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/27/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
An upregulation of the Na+-K+-2Cl- cotransporter NKCC1, the main chloride importer in mature neurons, can lead to depolarizing/excitatory responses mediated by GABA type A receptors (GABAARs) and, thus, to hyperactivity. Understanding the regulatory mechanisms of NKCC1 would help prevent intra-neuronal chloride accumulation that occurs in pathologies with defective inhibition. The cell mechanisms regulating NKCC1 are poorly understood. Here, we report in mature hippocampal neurons that GABAergic activity controls the membrane diffusion and clustering of NKCC1 via the chloride-sensitive WNK lysine deficient protein kinase 1 (WNK1) and the downstream Ste20 Pro-line Asparagine Rich Kinase (SPAK) kinase that directly phosphorylates NKCC1 on key threonine residues. At rest, this signaling pathway has little effect on intracellular Cl- concentration, but it participates in the elevation of intraneuronal Cl- concentration in hyperactivity conditions associated with an up-regulation of NKCC1. The fact that the main chloride exporter, the K+-Cl- cotransporter KCC2, is also regulated in mature neurons by the WNK1 pathway indicates that this pathway will be a target of choice in the pathology.
Collapse
|
7
|
Marileo AM, Gavilán J, San Martín VP, Lara CO, Sazo A, Muñoz-Montesino C, Castro PA, Burgos CF, Leiva-Salcedo E, Aguayo LG, Moraga-Cid G, Fuentealba J, Yévenes GE. Modulation of GABA A receptors and of GABAergic synapses by the natural alkaloid gelsemine. Front Mol Neurosci 2023; 15:1083189. [PMID: 36733271 PMCID: PMC9887029 DOI: 10.3389/fnmol.2022.1083189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/30/2022] [Indexed: 01/18/2023] Open
Abstract
The Gelsemium elegans plant preparations have shown beneficial activity against common diseases, including chronic pain and anxiety. Nevertheless, their clinical uses are limited by their toxicity. Gelsemine, one of the most abundant alkaloids in the Gelsemium plants, have replicated these therapeutic and toxic actions in experimental behavioral models. However, the molecular targets underlying these biological effects remain unclear. The behavioral activity profile of gelsemine suggests the involvement of GABAA receptors (GABAARs), which are the main biological targets of benzodiazepines (BDZs), a group of drugs with anxiolytic, hypnotic, and analgesic properties. Here, we aim to define the modulation of GABAARs by gelsemine, with a special focus on the subtypes involved in the BDZ actions. The gelsemine actions were determined by electrophysiological recordings of recombinant GABAARs expressed in HEK293 cells, and of native receptors in cortical neurons. Gelsemine inhibited the agonist-evoked currents of recombinant and native receptors. The functional inhibition was not associated with the BDZ binding site. We determined in addition that gelsemine diminished the frequency of GABAergic synaptic events, likely through a presynaptic modulation. Our findings establish gelsemine as a negative modulator of GABAARs and of GABAergic synaptic function. These pharmacological features discard direct anxiolytic or analgesic actions of gelsemine through GABAARs but support a role of GABAARs on the alkaloid induced toxicity. On the other hand, the presynaptic effects of the alkaloid provide an additional mechanism to explain their beneficial effects. Collectively, our results contribute novel information to improve understanding of gelsemine actions in the mammalian nervous system.
Collapse
Affiliation(s)
- Ana M. Marileo
- Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile,Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
| | - Javiera Gavilán
- Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - Victoria P. San Martín
- Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile,Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
| | - Cesar O. Lara
- Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile,Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
| | - Anggelo Sazo
- Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile,Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile
| | - Carola Muñoz-Montesino
- Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - Patricio A. Castro
- Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - Carlos F. Burgos
- Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - Elías Leiva-Salcedo
- Department of Biology, Faculty of Chemistry and Biology, Universidad de Santiago de Chile, Santiago, Chile
| | - Luis G. Aguayo
- Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - Gustavo Moraga-Cid
- Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - Jorge Fuentealba
- Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile
| | - Gonzalo E. Yévenes
- Department of Physiology, Faculty of Biological Sciences, Universidad de Concepción, Concepción, Chile,Millennium Nucleus for the Study of Pain (MiNuSPain), Santiago, Chile,*Correspondence: Gonzalo E. Yévenes, ✉
| |
Collapse
|
8
|
Kurki SN, Uvarov P, Pospelov AS, Trontti K, Hübner AK, Srinivasan R, Watanabe M, Hovatta I, Hübner CA, Kaila K, Virtanen MA. Expression patterns of NKCC1 in neurons and non-neuronal cells during cortico-hippocampal development. Cereb Cortex 2022; 33:5906-5923. [PMID: 36573432 PMCID: PMC10183754 DOI: 10.1093/cercor/bhac470] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/05/2022] [Accepted: 11/06/2022] [Indexed: 12/28/2022] Open
Abstract
Abstract
The Na-K-2Cl cotransporter NKCC1 is widely expressed in cells within and outside the brain. However, our understanding of its roles in brain functions throughout development, as well as in neuropsychiatric and neurological disorders, has been severely hindered by the lack of reliable data on its developmental and (sub)cellular expression patterns. We provide here the first properly controlled analysis of NKCC1 protein expression in various cell types of the mouse brain using custom-made antibodies and an NKCC1 knock-out validated immunohistochemical procedure, with parallel data based on advanced mRNA approaches. NKCC1 protein and mRNA are expressed at remarkably high levels in oligodendrocytes. In immature neurons, NKCC1 protein was located in the somata, whereas in adult neurons, only NKCC1 mRNA could be clearly detected. NKCC1 immunoreactivity is also seen in microglia, astrocytes, developing pericytes, and in progenitor cells of the dentate gyrus. Finally, a differential expression of NKCC1 splice variants was observed, with NKCC1a predominating in non-neuronal cells and NKCC1b in neurons. Taken together, our data provide a cellular basis for understanding NKCC1 functions in the brain and enable the identification of major limitations and promises in the development of neuron-targeting NKCC1-blockers.
Collapse
Affiliation(s)
- Samu N Kurki
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Pavel Uvarov
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Alexey S Pospelov
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Kalevi Trontti
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
- University of Helsinki SleepWell Research Program, Faculty of Medicine, , 00014 Helsinki , Finland
- University of Helsinki Department of Psychology and Logopedics, , 00014 Helsinki , Finland
| | - Antje K Hübner
- Jena University Hospital, Friedrich Schiller Universität Institute of Human Genetics, , 07747 Jena , Germany
| | - Rakenduvadhana Srinivasan
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Masahiko Watanabe
- Hokkaido University Department of Anatomy, Faculty of Medicine, , Sapporo 060–8638 , Japan
| | - Iiris Hovatta
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
- University of Helsinki SleepWell Research Program, Faculty of Medicine, , 00014 Helsinki , Finland
- University of Helsinki Department of Psychology and Logopedics, , 00014 Helsinki , Finland
| | - Christian A Hübner
- Jena University Hospital, Friedrich Schiller Universität Institute of Human Genetics, , 07747 Jena , Germany
| | - Kai Kaila
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| | - Mari A Virtanen
- University of Helsinki Molecular and Integrative Biosciences, , 00014 Helsinki , Finland
- Helsinki Institute of Life Science, University of Helsinki Neuroscience Center, , 00014 Helsinki , Finland
| |
Collapse
|
9
|
Gray SR, Ye L, Ye JY, Paukert M. Noradrenergic terminal short-term potentiation enables modality-selective integration of sensory input and vigilance state. SCIENCE ADVANCES 2021; 7:eabk1378. [PMID: 34919424 PMCID: PMC8682997 DOI: 10.1126/sciadv.abk1378] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Accepted: 10/28/2021] [Indexed: 06/14/2023]
Abstract
Recent years have seen compelling demonstrations of the importance of behavioral state on sensory processing and attention. Arousal plays a dominant role in controlling brain-wide neural activity patterns, particularly through modulation by norepinephrine. Noradrenergic brainstem nuclei, including locus coeruleus, can be activated by stimuli of multiple sensory modalities and broadcast modulatory signals via axonal projections throughout the brain. This organization might suggest proportional brain-wide norepinephrine release during states of heightened vigilance. Here, however, we have found that low-intensity, nonarousing visual stimuli enhanced vigilance-dependent noradrenergic signaling locally in visual cortex, revealed using dual-site fiber photometry to monitor noradrenergic Ca2+ responses of astroglia simultaneously in cerebellum and visual cortex and two-photon microscopy to monitor noradrenergic axonal terminal Ca2+ dynamics. Nitric oxide, following N-methyl-d-aspartate receptor activation in neuronal nitric oxide synthase-positive interneurons, mediated transient acceleration of norepinephrine-dependent astroglia Ca2+ activation. These findings reveal a candidate cortical microcircuit for sensory modality-selective modulation of attention.
Collapse
Affiliation(s)
- Shawn R. Gray
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Joint UTSA/UTHSCSA Graduate Program in Biomedical Engineering, San Antonio, TX, USA
| | - Liang Ye
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jing Yong Ye
- Joint UTSA/UTHSCSA Graduate Program in Biomedical Engineering, San Antonio, TX, USA
- Department of Biomedical Engineering, University of Texas at San Antonio, San Antonio, TX, USA
| | - Martin Paukert
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Joint UTSA/UTHSCSA Graduate Program in Biomedical Engineering, San Antonio, TX, USA
- Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
10
|
Virtanen MA, Uvarov P, Hübner CA, Kaila K. NKCC1, an Elusive Molecular Target in Brain Development: Making Sense of the Existing Data. Cells 2020; 9:cells9122607. [PMID: 33291778 PMCID: PMC7761970 DOI: 10.3390/cells9122607] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/01/2020] [Accepted: 12/03/2020] [Indexed: 12/14/2022] Open
Abstract
Ionotropic GABA transmission is mediated by anion (mainly Cl−)-permeable GABAA receptors (GABAARs). In immature neurons, GABA exerts depolarizing and sometimes functionally excitatory actions, based on active uptake of Cl− by the Na-K-2Cl cotransporter NKCC1. While functional evidence firmly shows NKCC1-mediated ion transport in immature and diseased neurons, molecular detection of NKCC1 in the brain has turned out to be extremely difficult. In this review, we describe the highly inconsistent data that are available on the cell type-specific expression patterns of the NKCC1 mRNA and protein in the CNS. We discuss the major technical caveats, including a lack of knock-out-controlled immunohistochemistry in the forebrain, possible effects of alternative splicing on the binding of antibodies and RNA probes, and the wide expression of NKCC1 in different cell types, which make whole-tissue analyses of NKCC1 useless for studying its neuronal expression. We also review novel single-cell RNAseq data showing that most of the NKCC1 in the adult CNS may, in fact, be expressed in non-neuronal cells, especially in glia. As future directions, we suggest single-cell NKCC1 mRNA and protein analyses and the use of genetically tagged endogenous proteins or systematically designed novel antibodies, together with proper knock-out controls, for the visualization of endogenous NKCC1 in distinct brain cell types and their subcellular compartments.
Collapse
Affiliation(s)
- Mari A. Virtanen
- Molecular and Integrative Biosciences, University of Helsinki, 00014 Helsinki, Finland; (M.A.V.); (P.U.)
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| | - Pavel Uvarov
- Molecular and Integrative Biosciences, University of Helsinki, 00014 Helsinki, Finland; (M.A.V.); (P.U.)
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
| | - Christian A. Hübner
- Institute of Human Genetics, Jena University Hospital, Friedrich Schiller Universität, 07747 Jena, Germany;
| | - Kai Kaila
- Molecular and Integrative Biosciences, University of Helsinki, 00014 Helsinki, Finland; (M.A.V.); (P.U.)
- Neuroscience Center, Helsinki Institute of Life Science, University of Helsinki, 00014 Helsinki, Finland
- Correspondence: ; Tel.: +358-407256759
| |
Collapse
|
11
|
Wiciński M, Puk O, Malinowski B. Cenobamate: Neuroprotective Potential of a New Antiepileptic Drug. Neurochem Res 2020; 46:439-446. [PMID: 33252771 DOI: 10.1007/s11064-020-03188-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 11/17/2020] [Accepted: 11/20/2020] [Indexed: 12/31/2022]
Abstract
Central nervous system (CNS) injuries annually afflict approximately 2.7 million people in United States only, inflicting costs of nearly 100 billion US dollars. The gravity of this problem is a consequence of severe and prolonged disability of patients due to a scarce regeneration of CNS, along with the lack of efficient neuroprotective and neuroregenrative therapies. Therefore, the first and most important task in managing the CNS injury is reduction of the damaged area, and apoptosis of neurons occurs not only during the trauma, but in great extent within the following minutes and hours. This process, called secondary injury phase, is a result of trauma-induced metabolic changes in nervous tissue and neuron apoptosis. Cenobamate is a new antiepileptic drug approved by FDA on November 21, 2019. Regardless of its primary purpose, cenobamate, as a blocker of voltage-gated sodium channels and positive modulator of GABAa receptors, it appears to be a promising neuroprotective agent. Moreover, through activation of PI3K/Akt-CREB-BDNF pathway, it leads to the increase of anti-apoptotic factor levels and the decrease of pro-apoptotic factor levels, which induce inhibition of apoptosis and increase neuron survival. Similarly to riluzole, cenobamate could be an important part of a perioperative procedure in neurosurgery, decreasing the occurrence of neurological deficits. Provided that cenobamate will be effective in aforementioned conditions, it could improve treatment outcomes of millions of patients every year, thereby an extensive investigation of its efficacy as a neuroprotective treatment after central nervous system trauma should follow.
Collapse
Affiliation(s)
- Michał Wiciński
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090, Bydgoszcz, Poland
| | - Oskar Puk
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090, Bydgoszcz, Poland.
| | - Bartosz Malinowski
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, M. Curie 9, 85-090, Bydgoszcz, Poland
| |
Collapse
|
12
|
Hwang KS, Kan H, Kim SS, Chae JS, Yang JY, Shin DS, Ahn SH, Ahn JH, Cho JH, Jang IS, Shin J, Joo J, Kim CH, Bae MA. Efficacy and pharmacokinetics evaluation of 4-(2-chloro-4-fluorobenzyl)-3-(2-thienyl)-1,2,4-oxadiazol-5(4H)-one (GM-90432) as an anti-seizure agent. Neurochem Int 2020; 141:104870. [PMID: 33035603 DOI: 10.1016/j.neuint.2020.104870] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 09/17/2020] [Accepted: 10/02/2020] [Indexed: 11/15/2022]
Abstract
Epilepsy is a common chronic neurological disease characterized by recurrent epileptic seizures. A seizure is an uncontrolled electrical activity in the brain that can cause different levels of behavior, emotion, and consciousness. One-third of patients fail to receive sufficient seizure control, even though more than fifty FDA-approved anti-seizure drugs (ASDs) are available. In this study, we attempted small molecule screening to identify potential therapeutic agents for the treatment of seizures using seizure-induced animal models. Through behavioral phenotype-based screening, 4-(2-chloro-4-fluorobenzyl)-3-(2-thienyl)-1,2,4-oxadiazol-5(4H)-one (GM-90432) was identified as a prototype. GM-90432 treatment effectively decreased seizure-like behaviors in zebrafish and mice with chemically induced seizures. These results were consistent with decreased neuronal activity through immunohistochemistry for pERK in zebrafish larvae. Additionally, electroencephalogram (EEG) analysis revealed that GM-90432 decreases seizure-specific EEG events in adult zebrafish. Moreover, we revealed the preferential binding of GM-90432 to voltage-gated Na+ channels using a whole-cell patch clamp technique. Through pharmacokinetic analysis, GM-90432 effectively penetrated the blood-brain barrier and was distributed into the brain. Taken together, we suggest that GM-90432 has the potential to be developed into a new ASD candidate.
Collapse
Affiliation(s)
- Kyu-Seok Hwang
- Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Hyemin Kan
- Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Seong Soon Kim
- Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Jin Sil Chae
- Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Jung Yoon Yang
- Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Dae-Seop Shin
- Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea
| | - Se Hwan Ahn
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Jin Hee Ahn
- Department of Chemistry, Gwangju Institute of Science and Technology, Gwangju, 61005, Republic of Korea
| | - Jin-Hwa Cho
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu, 41940, Republic of Korea
| | - Il-Sung Jang
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu, 41940, Republic of Korea; Brain Science and Engineering Institute, Kyungpook National University, Daegu, 41940, Republic of Korea
| | | | - Jaeyoung Joo
- Zefit. Inc., Daegu, 42988, Republic of Korea; School of Undergraduate Studies, Daegu Gyeongbuk Institute of Science and Technology, Daegu, 41940, Republic of Korea
| | - Cheol-Hee Kim
- Department of Biology, Chungnam National University, Daejeon, 34114, Republic of Korea
| | - Myung Ae Bae
- Drug Discovery Platform Research Center, Korea Research Institute of Chemical Technology, Daejeon, 34114, Republic of Korea.
| |
Collapse
|
13
|
Sharma R, Nakamura M, Neupane C, Jeon BH, Shin H, Melnick SM, Glenn KJ, Jang IS, Park JB. Positive allosteric modulation of GABAA receptors by a novel antiepileptic drug cenobamate. Eur J Pharmacol 2020; 879:173117. [DOI: 10.1016/j.ejphar.2020.173117] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 04/06/2020] [Accepted: 04/14/2020] [Indexed: 10/24/2022]
|
14
|
Nakamura M, Cho JH, Shin H, Jang IS. Effects of cenobamate (YKP3089), a newly developed anti-epileptic drug, on voltage-gated sodium channels in rat hippocampal CA3 neurons. Eur J Pharmacol 2019; 855:175-182. [PMID: 31063770 DOI: 10.1016/j.ejphar.2019.05.007] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 04/30/2019] [Accepted: 05/03/2019] [Indexed: 11/26/2022]
Abstract
New, more effective pharmacologic treatments for epilepsy are needed, as a substantial portion of patients (>30%) are refractory to currently available anti-epileptic drugs. Cenobamate (YKP3089) is an investigational anti-epileptic drug in clinical development. Two completed adequate and well-controlled studies demonstrated a significant reduction in focal seizures with cenobamate in patients with epilepsy. In this study, we characterized the effects of cenobamate on voltage-gated Na+ channels in acutely isolated rat hippocampal CA3 neurons using a whole-cell patch-clamp technique. While cenobamate had little effect on the peak component of transient Na+ current (INaT) induced by brief depolarizing step pulses, it potently inhibited the non-inactivating persistent component of INa (INaP). In addition, cenobamate potently inhibited the current by slow voltage-ramp stimuli. Cenobamate significantly shifted the steady-state fast inactivation relationship toward a hyperpolarizing range, indicating that cenobamate binds to voltage-gated Na+ channels at the inactivated state with a higher affinity. Cenobamate also accelerated the development of inactivation and retarded recovery from inactivation of voltage-gated Na+ channels. In current clamp experiments, cenobamate hyperpolarized membrane potentials in a concentration-dependent manner, and these effects were mediated by inhibiting the INaP. Cenobamate also increased the threshold for generation of action potentials, and decreased the number of action potentials elicited by depolarizing current injection. Given that the INaP plays a pivotal role in the repetitive and/or burst generation of action potentials, the cenobamate-mediated preferential blockade of INaP might contribute to anti-epileptic activity.
Collapse
Affiliation(s)
- Michiko Nakamura
- Department of Pharmacology, School of Dentistry, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 700-412, Republic of Korea; Brain Science & Engineering Institute, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 700-412, Republic of Korea.
| | - Jin-Hwa Cho
- Department of Pharmacology, School of Dentistry, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 700-412, Republic of Korea.
| | - Hyewon Shin
- Department of Pharmacology, SK Biopharmaceuticals, Co., Ltd., 221 Pangyoyeok-ro, Seongnam, Gyeonggi, 305-712, Republic of Korea.
| | - Il-Sung Jang
- Department of Pharmacology, School of Dentistry, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 700-412, Republic of Korea; Brain Science & Engineering Institute, Kyungpook National University, 2177 Dalgubeol-daero, Jung-gu, Daegu, 700-412, Republic of Korea.
| |
Collapse
|
15
|
Khatri SN, Wu WC, Yang Y, Pugh JR. Direction of action of presynaptic GABA A receptors is highly dependent on the level of receptor activation. J Neurophysiol 2019; 121:1896-1905. [PMID: 30892973 DOI: 10.1152/jn.00779.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Many synapses, including parallel fiber synapses in the cerebellum, express presynaptic GABAA receptors. However, reports of the functional consequences of presynaptic GABAA receptor activation are variable across synapses, from inhibition to enhancement of transmitter release. We find that presynaptic GABAA receptor function is bidirectional at parallel fiber synapses depending on GABA concentration and modulation of GABAA receptors in mice. Activation of GABAA receptors by low GABA concentrations enhances glutamate release, whereas activation of receptors by higher GABA concentrations inhibits release. Furthermore, blocking GABAB receptors reduces GABAA receptor currents and shifts presynaptic responses toward greater enhancement of release across a wide range of GABA concentrations. Conversely, enhancing GABAA receptor currents with ethanol or neurosteroids shifts responses toward greater inhibition of release. The ability of presynaptic GABAA receptors to enhance or inhibit transmitter release at the same synapse depending on activity level provides a new mechanism for fine control of synaptic transmission by GABA and may explain conflicting reports of presynaptic GABAA receptor function across synapses. NEW & NOTEWORTHY GABAA receptors are widely expressed at presynaptic terminals in the central nervous system. However, previous reports have produced conflicting results on the function of these receptors at different synapses. We show that presynaptic GABAA receptor function is strongly dependent on the level of receptor activation. Low levels of receptor activation enhance transmitter release, whereas higher levels of activation inhibit release at the same synapses. This provides a novel mechanism by which presynaptic GABAA receptors fine-tune synaptic transmission.
Collapse
Affiliation(s)
- Shailesh N Khatri
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio , San Antonio, Texas
| | - Wan-Chen Wu
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio , San Antonio, Texas
| | - Ying Yang
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio , San Antonio, Texas.,Xiangya School of Medicine, Central South University , Changsha, Hunan , China
| | - Jason R Pugh
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio , San Antonio, Texas.,Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio , San Antonio, Texas
| |
Collapse
|
16
|
Forster HV. Julius H. Comroe Distinguished Lecture: Interdependence of neuromodulators in the control of breathing. J Appl Physiol (1985) 2018; 125:1511-1525. [PMID: 30138081 DOI: 10.1152/japplphysiol.00477.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
In vitro and in vivo anesthetized studies led to the conclusion that "deficiencies in one neuromodulator are immediately compensated by the action of other neuromodulators," which suggests an interdependence among neuromodulators. This concept was the focus of the 2018 Julius H. Comroe Lecture to the American Physiological Society in which I summarized our published studies testing the hypothesis that if modulatory interdependence was robust, breathing would not decrease during dialysis of antagonists to G protein-coupled excitatory receptors or agonists to inhibitory receptors into the ventral respiratory column (VRC) or the hypoglossal motor nuclei (HMN). We found breathing was not decreased during unilateral VRC dialyses of antagonists to excitatory muscarinic, serotonergic, and neurokinin-1 receptors alone or in combinations nor was breathing decreased with unilateral VRC dialysis of a µ-opioid receptor agonist. Analyses of the effluent dialysate revealed locally increased serotonin (excitatory) during muscarinic receptor blockade and decreased γ-aminobutyric acid (inhibitory) during dialysis of opioid agonists, suggesting an interdependence of neuromodulators through release of compensatory neuromodulators. Bilateral dialysis of receptor antagonists or agonist in the VRC increased breathing, which does not support the concept that unchanged breathing with unilateral dialyses was due to contralateral compensation. In contrast, in the HMN neither unilateral nor bilateral dialysis of the excitatory receptor antagonists altered breathing, but unilateral dialysis of the opioid receptor agonist decreased breathing. We conclude: 1) there is site-dependent interdependence of neuromodulators during physiologic conditions, and 2) attributing physiologic effects to a specific receptor perturbation is complicated by local compensatory mechanisms.
Collapse
Affiliation(s)
- Hubert V Forster
- Department of Physiology, Medical College of Wisconsin, Neuroscience Research Center, Medical College of Wisconsin, Zablocki Veterans Affairs Medical Center , Milwaukee, Wisconsin
| |
Collapse
|
17
|
Messier JE, Chen H, Cai ZL, Xue M. Targeting light-gated chloride channels to neuronal somatodendritic domain reduces their excitatory effect in the axon. eLife 2018; 7:e38506. [PMID: 30091701 PMCID: PMC6130974 DOI: 10.7554/elife.38506] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2018] [Accepted: 08/08/2018] [Indexed: 11/13/2022] Open
Abstract
Light-gated chloride channels are emerging as promising optogenetic tools for inhibition of neural activity. However, their effects depend on the transmembrane chloride electrochemical gradient and may be complex due to the heterogeneity of this gradient in different developmental stages, neuronal types, and subcellular compartments. Here we characterized a light-gated chloride channel, GtACR2, in mouse cortical neurons. We found that GtACR2 activation inhibited the soma, but unexpectedly depolarized the presynaptic terminals resulting in neurotransmitter release. Other light-gated chloride channels had similar effects. Reducing the chloride concentrations in the axon and presynaptic terminals diminished the GtACR2-induced neurotransmitter release, indicating an excitatory effect of chloride channels in these compartments. A novel hybrid somatodendritic targeting motif reduced the GtACR2-induced neurotransmitter release while enhancing the somatic photocurrents. Our results highlight the necessity of precisely determining the effects of light-gated chloride channels under specific experimental conditions and provide a much-improved light-gated chloride channel for optogenetic inhibition.
Collapse
Affiliation(s)
- Jessica E Messier
- Department of NeuroscienceBaylor College of MedicineHouston, TexasUnited States
- The Cain Foundation LaboratoriesJan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHouston, TexasUnited States
| | - Hongmei Chen
- Department of NeuroscienceBaylor College of MedicineHouston, TexasUnited States
- The Cain Foundation LaboratoriesJan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHouston, TexasUnited States
| | - Zhao-Lin Cai
- Department of NeuroscienceBaylor College of MedicineHouston, TexasUnited States
- The Cain Foundation LaboratoriesJan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHouston, TexasUnited States
| | - Mingshan Xue
- Department of NeuroscienceBaylor College of MedicineHouston, TexasUnited States
- The Cain Foundation LaboratoriesJan and Dan Duncan Neurological Research Institute at Texas Children’s HospitalHouston, TexasUnited States
- Department of Molecular and Human GeneticsBaylor College of MedicineHouston, TexasUnited States
| |
Collapse
|
18
|
Langer TM, Neumueller SE, Crumley E, Burgraff NJ, Talwar S, Hodges MR, Pan L, Forster HV. Effects on breathing of agonists to μ-opioid or GABA A receptors dialyzed into the ventral respiratory column of awake and sleeping goats. Respir Physiol Neurobiol 2017; 239:10-25. [PMID: 28137700 PMCID: PMC5996971 DOI: 10.1016/j.resp.2017.01.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 01/09/2017] [Accepted: 01/12/2017] [Indexed: 01/01/2023]
Abstract
Pulmonary ventilation (V̇I) in awake and sleeping goats does not change when antagonists to several excitatory G protein-coupled receptors are dialyzed unilaterally into the ventral respiratory column (VRC). Concomitant changes in excitatory neuromodulators in the effluent mock cerebral spinal fluid (mCSF) suggest neuromodulatory compensation. Herein, we studied neuromodulatory compensation during dialysis of agonists to inhibitory G protein-coupled or ionotropic receptors into the VRC. Microtubules were implanted into the VRC of goats for dialysis of mCSF mixed with agonists to either μ-opioid (DAMGO) or GABAA (muscimol) receptors. We found: (1) V̇I decreased during unilateral but increased during bilateral dialysis of DAMGO, (2) dialyses of DAMGO destabilized breathing, (3) unilateral dialysis of muscimol increased V̇I, and (4) dialysis of DAMGO decreased GABA in the effluent mCSF. We conclude: (1) neuromodulatory compensation can occur during altered inhibitory neuromodulator receptor activity, and (2) the mechanism of compensation differs between G protein-coupled excitatory and inhibitory receptors and between G protein-coupled and inotropic inhibitory receptors.
Collapse
Affiliation(s)
- Thomas M Langer
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Suzanne E Neumueller
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Emma Crumley
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Nicholas J Burgraff
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Sawan Talwar
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Matthew R Hodges
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States; Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States
| | - Lawrence Pan
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States; Department of Physical Therapy, Marquette University, Milwaukee, WI 53226, United States
| | - Hubert V Forster
- Department of Physiology, Medical College of Wisconsin, Milwaukee, WI 53226, United States; Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI 53226, United States; Zablocki Veterans Affairs Medical Center, Milwaukee, WI 53226, United States.
| |
Collapse
|
19
|
Kakizawa K, Watanabe M, Mutoh H, Okawa Y, Yamashita M, Yanagawa Y, Itoi K, Suda T, Oki Y, Fukuda A. A novel GABA-mediated corticotropin-releasing hormone secretory mechanism in the median eminence. SCIENCE ADVANCES 2016; 2:e1501723. [PMID: 27540587 PMCID: PMC4988769 DOI: 10.1126/sciadv.1501723] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Accepted: 07/19/2016] [Indexed: 05/13/2023]
Abstract
Corticotropin-releasing hormone (CRH), which is synthesized in the paraventricular nucleus (PVN) of the hypothalamus, plays an important role in the endocrine stress response. The excitability of CRH neurons is regulated by γ-aminobutyric acid (GABA)-containing neurons projecting to the PVN. We investigated the role of GABA in the regulation of CRH release. The release of CRH was impaired, accumulating in the cell bodies of CRH neurons in heterozygous GAD67-GFP (green fluorescent protein) knock-in mice (GAD67(+/GFP)), which exhibited decreased GABA content. The GABAA receptor (GABAAR) and the Na(+)-K(+)-2Cl(-) cotransporter (NKCC1), but not the K(+)-Cl(-) cotransporter (KCC2), were expressed in the terminals of the CRH neurons at the median eminence (ME). In contrast, CRH neuronal somata were enriched with KCC2 but not with NKCC1. Thus, intracellular Cl(-) concentrations ([Cl(-)]i) may be increased at the terminals of CRH neurons compared with concentrations in the cell body. Moreover, GABAergic terminals projecting from the arcuate nucleus were present in close proximity to CRH-positive nerve terminals. Furthermore, a GABAAR agonist increased the intracellular calcium (Ca(2+)) levels in the CRH neuron terminals but decreased the Ca(2+) levels in their somata. In addition, the increases in Ca(2+) concentrations were prevented by an NKCC1 inhibitor. We propose a novel mechanism by which the excitatory action of GABA maintains a steady-state CRH release from axon terminals in the ME.
Collapse
Affiliation(s)
- Keisuke Kakizawa
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
- Second Division, Department of Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Miho Watanabe
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Hiroki Mutoh
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Yuta Okawa
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
- Second Division, Department of Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Miho Yamashita
- Second Division, Department of Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine, Maebashi, Gunma 371-8511, Japan
| | - Keiichi Itoi
- Laboratory of Information Biology, Graduate School of Information Sciences, Tohoku University, Sendai, Miyagi 980-8579, Japan
| | - Takafumi Suda
- Second Division, Department of Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Yutaka Oki
- Department of Family and Community Medicine, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| | - Atsuo Fukuda
- Department of Neurophysiology, Hamamatsu University School of Medicine, Hamamatsu, Shizuoka 431-3192, Japan
| |
Collapse
|
20
|
Nasehi M, Kamali-Dolatabadi L, Torabi-Nami M, Zarrindast MR. Possible involvement of the CA1 GABAA receptors upon acquisition and expression of the ACPA-induced place preference in mice. Physiol Behav 2016; 161:155-165. [DOI: 10.1016/j.physbeh.2016.04.021] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2015] [Revised: 04/12/2016] [Accepted: 04/12/2016] [Indexed: 12/12/2022]
|
21
|
Abstract
Synapses from neurons of the medial nucleus of the trapezoid body (MNTB) onto neurons of the lateral superior olive (LSO) in the auditory brainstem are glycinergic in maturity, but also GABAergic and glutamatergic in development. The role for this neurotransmitter cotransmission is poorly understood. Here we use electrophysiological recordings in brainstem slices from P3-P21 mice to demonstrate that GABA release evoked from MNTB axons can spill over to neighboring MNTB axons and cause excitation by activating GABAAR. This spillover excitation generates patterns of staggered neurotransmitter release from different MNTB axons resulting in characteristic "doublet" postsynaptic currents in LSO neurons. Postembedding immunogold labeling and electron microscopy provide evidence that GABAARs are localized at MNTB axon terminals. Photolytic uncaging of p-hydroxyphenacyl (pHP) GABA demonstrates backpropagation of GABAAR-mediated depolarizations from MNTB axon terminals to the soma, some hundreds of microns away. These somatic depolarizations enhanced somatic excitability by increasing the probability of action potential generation. GABA spillover excitation between MNTB axon terminals may entrain neighboring MNTB neurons, which may play a role in the developmental refinement of the MNTB-LSO pathway. Axonal spillover excitation persisted beyond the second postnatal week, suggesting that this mechanism may play a role in sound localization, by providing new avenues of communication between MNTB neurons via their distal axonal projections. Significance statement: In this study, a new mechanism of neuronal communication between auditory synapses in the mammalian sound localization pathway is described. Evidence is provided that the inhibitory neurotransmitter GABA can spill over between axon terminals to cause excitation of nearby synapses to further stimulate neurotransmitter release. Excitatory GABA spillover between inhibitory axon terminals may have important implications for the development and refinement of this auditory circuit and may play a role in the ability to precisely localize sound sources.
Collapse
|
22
|
Additive effect of BLA GABAA receptor mechanism and (+)-MK-801 on memory retention deficit, an isobologram analysis. Pharmacol Biochem Behav 2016; 143:57-64. [DOI: 10.1016/j.pbb.2016.02.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Revised: 02/02/2016] [Accepted: 02/03/2016] [Indexed: 11/21/2022]
|
23
|
Zarrabian S, Farahizadeh M, Nasehi M, Zarrindast MR. The role of CA3 GABAA receptors on anxiolytic-like behaviors and avoidance memory deficit induced by NMDA receptor antagonists. J Psychopharmacol 2016; 30:215-23. [PMID: 26755545 DOI: 10.1177/0269881115622239] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cognitive functions are influenced by memory and anxiety states. However, a non-linear relation has been shown between these two domains. The important role of the hippocampus in memory and emotional responses may link the pathogenesis of anxiety to memory-related GABAergic and glutamatergic processes in the hippocampus. To investigate the role of GABAA receptors in relation to blocking N-methyl-D-aspartate (NMDA) receptors in the CA3 region, and balancing the glutamatergic and GABAergic system activities as an approach for the management of related disorders, the elevated plus-maze test-retest paradigm was used to investigate the anxiolytic-like state on the test day and avoidance memory state on the retest day. The data showed that injection of D-AP5, the NMDA receptor antagonist, induced anxiolytic-like behavior and impaired avoidance memory. Injection of GABAA agonist (muscimol), but not the antagonist (bicuculline), induced avoidance memory impairment. Neither muscimol nor bicuculline altered anxiety-like behaviors. Muscimol pretreatment did not change D-AP5-induced anxiolytic-like behaviors but potentiated avoidance memory impairment. Bicuculline pretreatment blocked D-AP5-induced anxiolytic-like behaviors and contradicted its effect on avoidance memory. Our findings indicate that alteration of the CA3 GABAA receptor activity can effectively affect the anxiolytic-like behaviors and avoidance memory deficit induced by D-AP5.
Collapse
Affiliation(s)
| | | | - Mohammad Nasehi
- Cognitive and Neuroscience Research Center (CNRC), Islamic Azad University, Tehran Medical Sciences Branch, Tehran, Iran
| | - Mohammad-Reza Zarrindast
- Institute for Cognitive Science Studies (ICSS), Tehran, Iran Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran Iranian National Center for Addiction Studies; Tehran University of Medical Sciences, Tehran, Iran School of Cognitive Sciences, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran
| |
Collapse
|
24
|
Evidence for the participation of peripheral α5 subunit-containing GABAA receptors in GABAA agonists-induced nociception in rats. Eur J Pharmacol 2014; 734:91-7. [DOI: 10.1016/j.ejphar.2014.03.051] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 03/11/2014] [Accepted: 03/22/2014] [Indexed: 11/17/2022]
|
25
|
Winkelmann A, Maggio N, Eller J, Caliskan G, Semtner M, Häussler U, Jüttner R, Dugladze T, Smolinsky B, Kowalczyk S, Chronowska E, Schwarz G, Rathjen FG, Rechavi G, Haas CA, Kulik A, Gloveli T, Heinemann U, Meier JC. Changes in neural network homeostasis trigger neuropsychiatric symptoms. J Clin Invest 2014; 124:696-711. [PMID: 24430185 PMCID: PMC3904623 DOI: 10.1172/jci71472] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Accepted: 10/31/2013] [Indexed: 12/13/2022] Open
Abstract
The mechanisms that regulate the strength of synaptic transmission and intrinsic neuronal excitability are well characterized; however, the mechanisms that promote disease-causing neural network dysfunction are poorly defined. We generated mice with targeted neuron type-specific expression of a gain-of-function variant of the neurotransmitter receptor for glycine (GlyR) that is found in hippocampectomies from patients with temporal lobe epilepsy. In this mouse model, targeted expression of gain-of-function GlyR in terminals of glutamatergic cells or in parvalbumin-positive interneurons persistently altered neural network excitability. The increased network excitability associated with gain-of-function GlyR expression in glutamatergic neurons resulted in recurrent epileptiform discharge, which provoked cognitive dysfunction and memory deficits without affecting bidirectional synaptic plasticity. In contrast, decreased network excitability due to gain-of-function GlyR expression in parvalbumin-positive interneurons resulted in an anxiety phenotype, but did not affect cognitive performance or discriminative associative memory. Our animal model unveils neuron type-specific effects on cognition, formation of discriminative associative memory, and emotional behavior in vivo. Furthermore, our data identify a presynaptic disease-causing molecular mechanism that impairs homeostatic regulation of neural network excitability and triggers neuropsychiatric symptoms.
Collapse
Affiliation(s)
- Aline Winkelmann
- FU-Berlin, Fachbereich Biologie, Chemie, Pharmazie, Berlin, Germany.
RNA editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Talpiot Medical Leadership Program, Department of Neurology and the J. Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer, Israel.
Cellular and Network Physiology Group, Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Berlin, Germany.
CC2 Zentrum für Physiologie, Freie Universität Berlin, Berlin, Germany.
Experimental Epilepsy Research, Department of Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany.
Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Institute of Biochemistry, University of Cologne and Center for Molecular Medicine, Cologne, Germany.
Department of Physiology II, University of Freiburg, Freiburg, Germany.
Sheba Cancer Research Center, The Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
BrainLinks-BrainTools, Cluster of Excellence and
BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Nicola Maggio
- FU-Berlin, Fachbereich Biologie, Chemie, Pharmazie, Berlin, Germany.
RNA editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Talpiot Medical Leadership Program, Department of Neurology and the J. Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer, Israel.
Cellular and Network Physiology Group, Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Berlin, Germany.
CC2 Zentrum für Physiologie, Freie Universität Berlin, Berlin, Germany.
Experimental Epilepsy Research, Department of Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany.
Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Institute of Biochemistry, University of Cologne and Center for Molecular Medicine, Cologne, Germany.
Department of Physiology II, University of Freiburg, Freiburg, Germany.
Sheba Cancer Research Center, The Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
BrainLinks-BrainTools, Cluster of Excellence and
BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Joanna Eller
- FU-Berlin, Fachbereich Biologie, Chemie, Pharmazie, Berlin, Germany.
RNA editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Talpiot Medical Leadership Program, Department of Neurology and the J. Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer, Israel.
Cellular and Network Physiology Group, Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Berlin, Germany.
CC2 Zentrum für Physiologie, Freie Universität Berlin, Berlin, Germany.
Experimental Epilepsy Research, Department of Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany.
Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Institute of Biochemistry, University of Cologne and Center for Molecular Medicine, Cologne, Germany.
Department of Physiology II, University of Freiburg, Freiburg, Germany.
Sheba Cancer Research Center, The Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
BrainLinks-BrainTools, Cluster of Excellence and
BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Gürsel Caliskan
- FU-Berlin, Fachbereich Biologie, Chemie, Pharmazie, Berlin, Germany.
RNA editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Talpiot Medical Leadership Program, Department of Neurology and the J. Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer, Israel.
Cellular and Network Physiology Group, Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Berlin, Germany.
CC2 Zentrum für Physiologie, Freie Universität Berlin, Berlin, Germany.
Experimental Epilepsy Research, Department of Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany.
Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Institute of Biochemistry, University of Cologne and Center for Molecular Medicine, Cologne, Germany.
Department of Physiology II, University of Freiburg, Freiburg, Germany.
Sheba Cancer Research Center, The Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
BrainLinks-BrainTools, Cluster of Excellence and
BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Marcus Semtner
- FU-Berlin, Fachbereich Biologie, Chemie, Pharmazie, Berlin, Germany.
RNA editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Talpiot Medical Leadership Program, Department of Neurology and the J. Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer, Israel.
Cellular and Network Physiology Group, Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Berlin, Germany.
CC2 Zentrum für Physiologie, Freie Universität Berlin, Berlin, Germany.
Experimental Epilepsy Research, Department of Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany.
Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Institute of Biochemistry, University of Cologne and Center for Molecular Medicine, Cologne, Germany.
Department of Physiology II, University of Freiburg, Freiburg, Germany.
Sheba Cancer Research Center, The Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
BrainLinks-BrainTools, Cluster of Excellence and
BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Ute Häussler
- FU-Berlin, Fachbereich Biologie, Chemie, Pharmazie, Berlin, Germany.
RNA editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Talpiot Medical Leadership Program, Department of Neurology and the J. Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer, Israel.
Cellular and Network Physiology Group, Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Berlin, Germany.
CC2 Zentrum für Physiologie, Freie Universität Berlin, Berlin, Germany.
Experimental Epilepsy Research, Department of Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany.
Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Institute of Biochemistry, University of Cologne and Center for Molecular Medicine, Cologne, Germany.
Department of Physiology II, University of Freiburg, Freiburg, Germany.
Sheba Cancer Research Center, The Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
BrainLinks-BrainTools, Cluster of Excellence and
BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - René Jüttner
- FU-Berlin, Fachbereich Biologie, Chemie, Pharmazie, Berlin, Germany.
RNA editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Talpiot Medical Leadership Program, Department of Neurology and the J. Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer, Israel.
Cellular and Network Physiology Group, Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Berlin, Germany.
CC2 Zentrum für Physiologie, Freie Universität Berlin, Berlin, Germany.
Experimental Epilepsy Research, Department of Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany.
Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Institute of Biochemistry, University of Cologne and Center for Molecular Medicine, Cologne, Germany.
Department of Physiology II, University of Freiburg, Freiburg, Germany.
Sheba Cancer Research Center, The Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
BrainLinks-BrainTools, Cluster of Excellence and
BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Tamar Dugladze
- FU-Berlin, Fachbereich Biologie, Chemie, Pharmazie, Berlin, Germany.
RNA editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Talpiot Medical Leadership Program, Department of Neurology and the J. Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer, Israel.
Cellular and Network Physiology Group, Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Berlin, Germany.
CC2 Zentrum für Physiologie, Freie Universität Berlin, Berlin, Germany.
Experimental Epilepsy Research, Department of Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany.
Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Institute of Biochemistry, University of Cologne and Center for Molecular Medicine, Cologne, Germany.
Department of Physiology II, University of Freiburg, Freiburg, Germany.
Sheba Cancer Research Center, The Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
BrainLinks-BrainTools, Cluster of Excellence and
BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Birthe Smolinsky
- FU-Berlin, Fachbereich Biologie, Chemie, Pharmazie, Berlin, Germany.
RNA editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Talpiot Medical Leadership Program, Department of Neurology and the J. Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer, Israel.
Cellular and Network Physiology Group, Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Berlin, Germany.
CC2 Zentrum für Physiologie, Freie Universität Berlin, Berlin, Germany.
Experimental Epilepsy Research, Department of Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany.
Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Institute of Biochemistry, University of Cologne and Center for Molecular Medicine, Cologne, Germany.
Department of Physiology II, University of Freiburg, Freiburg, Germany.
Sheba Cancer Research Center, The Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
BrainLinks-BrainTools, Cluster of Excellence and
BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Sarah Kowalczyk
- FU-Berlin, Fachbereich Biologie, Chemie, Pharmazie, Berlin, Germany.
RNA editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Talpiot Medical Leadership Program, Department of Neurology and the J. Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer, Israel.
Cellular and Network Physiology Group, Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Berlin, Germany.
CC2 Zentrum für Physiologie, Freie Universität Berlin, Berlin, Germany.
Experimental Epilepsy Research, Department of Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany.
Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Institute of Biochemistry, University of Cologne and Center for Molecular Medicine, Cologne, Germany.
Department of Physiology II, University of Freiburg, Freiburg, Germany.
Sheba Cancer Research Center, The Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
BrainLinks-BrainTools, Cluster of Excellence and
BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Ewa Chronowska
- FU-Berlin, Fachbereich Biologie, Chemie, Pharmazie, Berlin, Germany.
RNA editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Talpiot Medical Leadership Program, Department of Neurology and the J. Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer, Israel.
Cellular and Network Physiology Group, Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Berlin, Germany.
CC2 Zentrum für Physiologie, Freie Universität Berlin, Berlin, Germany.
Experimental Epilepsy Research, Department of Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany.
Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Institute of Biochemistry, University of Cologne and Center for Molecular Medicine, Cologne, Germany.
Department of Physiology II, University of Freiburg, Freiburg, Germany.
Sheba Cancer Research Center, The Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
BrainLinks-BrainTools, Cluster of Excellence and
BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Günter Schwarz
- FU-Berlin, Fachbereich Biologie, Chemie, Pharmazie, Berlin, Germany.
RNA editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Talpiot Medical Leadership Program, Department of Neurology and the J. Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer, Israel.
Cellular and Network Physiology Group, Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Berlin, Germany.
CC2 Zentrum für Physiologie, Freie Universität Berlin, Berlin, Germany.
Experimental Epilepsy Research, Department of Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany.
Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Institute of Biochemistry, University of Cologne and Center for Molecular Medicine, Cologne, Germany.
Department of Physiology II, University of Freiburg, Freiburg, Germany.
Sheba Cancer Research Center, The Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
BrainLinks-BrainTools, Cluster of Excellence and
BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Fritz G. Rathjen
- FU-Berlin, Fachbereich Biologie, Chemie, Pharmazie, Berlin, Germany.
RNA editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Talpiot Medical Leadership Program, Department of Neurology and the J. Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer, Israel.
Cellular and Network Physiology Group, Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Berlin, Germany.
CC2 Zentrum für Physiologie, Freie Universität Berlin, Berlin, Germany.
Experimental Epilepsy Research, Department of Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany.
Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Institute of Biochemistry, University of Cologne and Center for Molecular Medicine, Cologne, Germany.
Department of Physiology II, University of Freiburg, Freiburg, Germany.
Sheba Cancer Research Center, The Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
BrainLinks-BrainTools, Cluster of Excellence and
BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Gideon Rechavi
- FU-Berlin, Fachbereich Biologie, Chemie, Pharmazie, Berlin, Germany.
RNA editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Talpiot Medical Leadership Program, Department of Neurology and the J. Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer, Israel.
Cellular and Network Physiology Group, Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Berlin, Germany.
CC2 Zentrum für Physiologie, Freie Universität Berlin, Berlin, Germany.
Experimental Epilepsy Research, Department of Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany.
Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Institute of Biochemistry, University of Cologne and Center for Molecular Medicine, Cologne, Germany.
Department of Physiology II, University of Freiburg, Freiburg, Germany.
Sheba Cancer Research Center, The Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
BrainLinks-BrainTools, Cluster of Excellence and
BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Carola A. Haas
- FU-Berlin, Fachbereich Biologie, Chemie, Pharmazie, Berlin, Germany.
RNA editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Talpiot Medical Leadership Program, Department of Neurology and the J. Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer, Israel.
Cellular and Network Physiology Group, Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Berlin, Germany.
CC2 Zentrum für Physiologie, Freie Universität Berlin, Berlin, Germany.
Experimental Epilepsy Research, Department of Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany.
Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Institute of Biochemistry, University of Cologne and Center for Molecular Medicine, Cologne, Germany.
Department of Physiology II, University of Freiburg, Freiburg, Germany.
Sheba Cancer Research Center, The Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
BrainLinks-BrainTools, Cluster of Excellence and
BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Akos Kulik
- FU-Berlin, Fachbereich Biologie, Chemie, Pharmazie, Berlin, Germany.
RNA editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Talpiot Medical Leadership Program, Department of Neurology and the J. Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer, Israel.
Cellular and Network Physiology Group, Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Berlin, Germany.
CC2 Zentrum für Physiologie, Freie Universität Berlin, Berlin, Germany.
Experimental Epilepsy Research, Department of Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany.
Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Institute of Biochemistry, University of Cologne and Center for Molecular Medicine, Cologne, Germany.
Department of Physiology II, University of Freiburg, Freiburg, Germany.
Sheba Cancer Research Center, The Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
BrainLinks-BrainTools, Cluster of Excellence and
BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Tengis Gloveli
- FU-Berlin, Fachbereich Biologie, Chemie, Pharmazie, Berlin, Germany.
RNA editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Talpiot Medical Leadership Program, Department of Neurology and the J. Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer, Israel.
Cellular and Network Physiology Group, Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Berlin, Germany.
CC2 Zentrum für Physiologie, Freie Universität Berlin, Berlin, Germany.
Experimental Epilepsy Research, Department of Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany.
Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Institute of Biochemistry, University of Cologne and Center for Molecular Medicine, Cologne, Germany.
Department of Physiology II, University of Freiburg, Freiburg, Germany.
Sheba Cancer Research Center, The Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
BrainLinks-BrainTools, Cluster of Excellence and
BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Uwe Heinemann
- FU-Berlin, Fachbereich Biologie, Chemie, Pharmazie, Berlin, Germany.
RNA editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Talpiot Medical Leadership Program, Department of Neurology and the J. Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer, Israel.
Cellular and Network Physiology Group, Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Berlin, Germany.
CC2 Zentrum für Physiologie, Freie Universität Berlin, Berlin, Germany.
Experimental Epilepsy Research, Department of Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany.
Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Institute of Biochemistry, University of Cologne and Center for Molecular Medicine, Cologne, Germany.
Department of Physiology II, University of Freiburg, Freiburg, Germany.
Sheba Cancer Research Center, The Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
BrainLinks-BrainTools, Cluster of Excellence and
BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Jochen C. Meier
- FU-Berlin, Fachbereich Biologie, Chemie, Pharmazie, Berlin, Germany.
RNA editing and Hyperexcitability Disorders Helmholtz Group, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Talpiot Medical Leadership Program, Department of Neurology and the J. Sagol Neuroscience Center, The Chaim Sheba Medical Center, Tel HaShomer, Israel.
Cellular and Network Physiology Group, Institute of Neurophysiology, Charité Universitätsmedizin Berlin, Berlin, Germany.
CC2 Zentrum für Physiologie, Freie Universität Berlin, Berlin, Germany.
Experimental Epilepsy Research, Department of Neurosurgery, Neurocenter, University of Freiburg, Freiburg, Germany.
Developmental Neurobiology, Max Delbrück Center for Molecular Medicine, Berlin, Germany.
Institute of Biochemistry, University of Cologne and Center for Molecular Medicine, Cologne, Germany.
Department of Physiology II, University of Freiburg, Freiburg, Germany.
Sheba Cancer Research Center, The Chaim Sheba Medical Center and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
BrainLinks-BrainTools, Cluster of Excellence and
BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| |
Collapse
|
26
|
Wakita M, Kotani N, Kogure K, Akaike N. Inhibition of excitatory synaptic transmission in hippocampal neurons by levetiracetam involves Zn²⁺-dependent GABA type A receptor-mediated presynaptic modulation. J Pharmacol Exp Ther 2013; 348:246-59. [PMID: 24259680 DOI: 10.1124/jpet.113.208751] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Levetiracetam (LEV) is an antiepileptic drug with a unique but as yet not fully resolved mechanism of action. Therefore, by use of a simplified rat-isolated nerve-bouton preparation, we have investigated how LEV modulates glutamatergic transmission from mossy fiber terminals to hippocampal CA3 neurons. Action potential-evoked excitatory postsynaptic currents (eEPSCs) were recorded using a conventional whole-cell patch-clamp recording configuration in voltage-clamp mode. The antiepileptic drug phenytoin decreased glutamatergic eEPSCs in a concentration-dependent fashion by inhibiting voltage-dependent Na⁺ and Ca²⁺ channel currents. In contrast, LEV had no effect on eEPSCs or voltage-dependent Na⁺ or Ca²⁺ channel currents. Activation of presynaptic GABA type A (GABA(A)) receptors by muscimol induced presynaptic inhibition of eEPSCs, resulting from depolarization block. Low concentrations of Zn²⁺, which had no effect on eEPSCs, voltage-dependent Na⁺ or Ca²⁺ channel currents, or glutamate receptor-mediated whole cell currents, reduced the muscimol-induced presynaptic inhibition. LEV applied in the continuous presence of 1 µM muscimol and 1 µM Zn²⁺ reversed this Zn²⁺ modulation on eEPSCs. The antagonizing effect of LEV on Zn²⁺-induced presynaptic GABA(A) receptor inhibition was also observed with the Zn²⁺ chelators Ca-EDTA and RhodZin-3. Our results clearly show that LEV removes the Zn²⁺-induced suppression of GABA(A)-mediated presynaptic inhibition, resulting in a presynaptic decrease in glutamate-mediated excitatory transmission. Our results provide a novel mechanism by which LEV may inhibit neuronal activity.
Collapse
Affiliation(s)
- Masahito Wakita
- Research Division for Clinical Pharmacology, Medical Corporation, Jyuryokai, Kumamoto Kinoh Hospital, Kumamoto, Japan (M.W., N.A.); Research Division for Life Science, Kumamoto Health Science University, Kumamoto, Japan (M.W., N.A.); Research Division of Neurophysiology, Kitamoto Hospital, Koshigaya, Japan (N.K., N.A); and Kogure Medical Clinic, Chouseikai Medical Corporation, Fukaya City, Saitama, Japan (K.K.)
| | | | | | | |
Collapse
|
27
|
Wakita M, Kotani N, Nonaka K, Shin MC, Akaike N. Effects of propofol on GABAergic and glutamatergic transmission in isolated hippocampal single nerve-synapse preparations. Eur J Pharmacol 2013; 718:63-73. [DOI: 10.1016/j.ejphar.2013.09.018] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2013] [Revised: 08/26/2013] [Accepted: 09/04/2013] [Indexed: 01/12/2023]
|
28
|
Abstract
The dorsal raphe nucleus (DR) controls forebrain serotonin neurotransmission to influence emotional states. GABA neurotransmission in the DR has been implicated in regulating sleep/wake states and influencing anxiety and aggression. To gain insight into how GABA regulates DR activity, we analyzed the organization of both GABA and glutamate axons in the rat DR using a high-resolution immunofluorescence technique, array tomography, as well as EM. This analysis revealed that a third or more of GABA-containing axons are organized in synaptic triads with a glutamatergic axon and a common postsynaptic target. Electrophysiological recordings showed that GABA has the capacity to presynaptically gate glutamate release in the DR through a combination of GABA-A and GABA-B receptor-mediated effects. Thus, GABA-glutamate synaptic triads are a common feature of the network architecture of the DR with the potential to regulate excitation of the nucleus.
Collapse
|
29
|
Giuliani FA, Escudero C, Casas S, Bazzocchini V, Yunes R, Laconi MR, Cabrera R. Allopregnanolone and puberty: modulatory effect on glutamate and GABA release and expression of 3α-hydroxysteroid oxidoreductase in the hypothalamus of female rats. Neuroscience 2013; 243:64-75. [PMID: 23562943 DOI: 10.1016/j.neuroscience.2013.03.053] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2012] [Revised: 02/26/2013] [Accepted: 03/26/2013] [Indexed: 02/07/2023]
Abstract
The hypothalamic release of glutamate and GABA regulates neurosecretory functions that may control the onset of puberty. This release may be influenced by neurosteroids such as allopregnanolone. Using superfusion experiments we examined the role of allopregnanolone on the K(+)-evoked and basal [(3)H]-glutamate and [(3)H]-GABA release from mediobasal hypothalamus and anterior preoptic area in prepubertal, vaginal opening and pubertal (P) rats and evaluated its modulatory effect on GABAA and NMDA (N-methyl-d-aspartic acid) receptors. Also, we examined the hypothalamic activity and mRNA expression of 3α-hydroxysteroid oxidoreductase (3α-HSOR) - enzyme that synthesizes allopregnanolone - using a spectrophotometric method and RT-PCR, respectively. Allopregnanolone increased both the K(+)-evoked [(3)H]-glutamate and [(3)H]-GABA release in P rats, being the former effect mediated by the modulation of NMDA receptors - as was reverted by Mg(2+) and by the NMDA receptor antagonist AP-7 and the latter by the modulation of NMDA and GABAA receptors - as was reverted by Mg(2+) and the GABAA receptor antagonist bicuculline. The neurosteroid also increased the basal release of [(3)H]-glutamate in VO rats in an effect that was dependent on the modulation of NMDA receptors as was reverted by Mg(2+). On the other hand we show that allopregnanolone reduced the basal release of [(3)H]-GABA in P rats although we cannot elucidate the precise mechanism by which the neurosteroid exerted this latter effect. The enzymatic activity and the mRNA expression of 3α-HSOR were both increased in P rats regarding the other two studied stages of sexual development. These results suggest an important physiological function of allopregnanolone in the hypothalamus of the P rat where it might be involved in the 'fine tuning' of neurosecretory functions related to the biology of reproduction of the female rats.
Collapse
Affiliation(s)
- F A Giuliani
- Instituto de Investigaciones Biomédicas (INBIOMED), Universidad de Mendoza, IMBECU-CONICET, Paseo Dr. Emilio Descotte 720, 5500 Mendoza, Argentina
| | | | | | | | | | | | | |
Collapse
|
30
|
Iwata S, Wakita M, Shin MC, Fukuda A, Akaike N. Modulation of allopregnanolone on excitatory transmitters release from single glutamatergic terminal. Brain Res Bull 2013; 93:39-46. [DOI: 10.1016/j.brainresbull.2012.11.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Revised: 11/02/2012] [Accepted: 11/05/2012] [Indexed: 10/27/2022]
|
31
|
Ruiz AJ, Kullmann DM. Ionotropic receptors at hippocampal mossy fibers: roles in axonal excitability, synaptic transmission, and plasticity. Front Neural Circuits 2013; 6:112. [PMID: 23316138 PMCID: PMC3540408 DOI: 10.3389/fncir.2012.00112] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 12/10/2012] [Indexed: 11/30/2022] Open
Abstract
Dentate granule cells process information from the enthorinal cortex en route to the hippocampus proper. These neurons have a very negative resting membrane potential and are relatively silent in the slice preparation. They are also subject to strong feed-forward inhibition. Their unmyelinated axon or mossy fiber ramifies extensively in the hilus and projects to stratum lucidum where it makes giant en-passant boutons with CA3 pyramidal neurons. There is compelling evidence that mossy fiber boutons express presynaptic GABAA receptors, which are commonly found in granule cell dendrites. There is also suggestive evidence for the presence of other ionotropic receptors, including glycine, NMDA, and kainate receptors, in mossy fiber boutons. These presynaptic receptors have been proposed to lead to mossy fiber membrane depolarization. How this phenomenon alters the excitability of synaptic boutons, the shape of presynaptic action potentials, Ca2+ influx and neurotransmitter release has remained elusive, but high-resolution live imaging of individual varicosities and direct patch-clamp recordings have begun to shed light on these phenomena. Presynaptic GABAA and kainate receptors have also been reported to facilitate the induction of long-term potentiation at mossy fiber—CA3 synapses. Although mossy fibers are highly specialized, some of the principles emerging at this connection may apply elsewhere in the CNS.
Collapse
Affiliation(s)
- Arnaud J Ruiz
- Department of Pharmacology, UCL School of Pharmacy London, UK
| | | |
Collapse
|
32
|
Shin MC, Wakita M, Iwata S, Nonaka K, Kotani N, Akaike N. Comparative effects of pentobarbital on spontaneous and evoked transmitter release from inhibitory and excitatory nerve terminals in rat CA3 neurons. Brain Res Bull 2012; 90:10-8. [PMID: 23026118 DOI: 10.1016/j.brainresbull.2012.09.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 09/14/2012] [Accepted: 09/19/2012] [Indexed: 10/27/2022]
Abstract
Pentobarbital (PB) modulates GABA(A) receptor-mediated postsynaptic responses through various mechanisms, and can directly activate the channel at higher doses. These channels exist both pre- and postsynaptically, and on the soma outside the synapse. PB also inhibits voltage-dependent Na⁺ and Ca²⁺ channels to decrease excitatory synaptic transmission. Just how these different sites of action combine to contribute to the overall effects of PB on inhibitory and excitatory synaptic transmission is less clear. To compare these pre- and postsynaptic actions of PB, we used a 'synaptic bouton' preparation of isolated rat hippocampal CA3 pyramidal neurons where we could measure in single neurons the effects of PB on spontaneous and single bouton evoked GABAergic inhibitory and glutamatergic excitatory postsynaptic currents (sIPSCs, sEPSCs, eIPSCs and eEPSCs), respectively. Low (sedative) concentrations (3-10 μM) of PB increased the frequency and amplitude of sIPSCs and sEPSCs, and also presynaptically increased the amplitude of both eIPSCs and eEPSCs. There was no change in current kinetics at this low concentration. At higher concentrations (30-300 μM), PB decreased the frequency, and increased the amplitude of sIPSCs, and presynaptically decreased the amplitude of eIPSCs. The current decay phase of sIPSCs and eIPSCs was increased. An increase in both frequency and amplitude was seen for sEPSCs, while the eIPSCs was also decreased by a bicuculline-sensitive presynaptic effect. The results confirm the multiple sites of action of PB on inhibitory and excitatory transmission and demonstrate that the most sensitive site of action is on transmitter release, via effects on presynaptic GABA(A) receptors. At low concentrations, however, both glutamate and GABA release is similarly enhanced, making the final effects on neuronal excitability difficult to predict and dependent on the particular systems involved and/or on subtle differences in susceptibility amongst individuals. At higher concentrations, release of both transmitters is decreased, while the postsynaptic effects to increase IPSPs and decrease EPSCs would be expected to both results in reduced neuronal excitability.
Collapse
Affiliation(s)
- Min-Chul Shin
- Research Division for Life Sciences, Kumamoto Health Science University, Kumamoto 861-5598, Japan
| | | | | | | | | | | |
Collapse
|
33
|
Effects of halothane on GABAergic and glutamatergic transmission in isolated hippocampal nerve-synapse preparations. Brain Res 2012; 1473:9-18. [DOI: 10.1016/j.brainres.2012.07.035] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Revised: 07/17/2012] [Accepted: 07/18/2012] [Indexed: 01/31/2023]
|
34
|
The effects of volatile anesthetics on synaptic and extrasynaptic GABA-induced neurotransmission. Brain Res Bull 2012; 93:69-79. [PMID: 22925739 DOI: 10.1016/j.brainresbull.2012.08.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 07/17/2012] [Accepted: 08/01/2012] [Indexed: 02/02/2023]
Abstract
Examination of volatile anesthetic actions at single synapses provides more direct information by reducing interference by surrounding tissue and extrasynaptic modulation. We examined how volatile anesthetics modulate GABA release by measuring spontaneous or miniature GABA-induced inhibitory postsynaptic currents (mIPSCs, sIPSCs) or by measuring action potential-evoked IPSCs (eIPSCs) at individual synapses. Halothane increased both the amplitude and frequency of sIPSCs. Isoflurane and enflurane increased mIPSC frequency while sevoflurane had no effect. These anesthetics did not alter mIPSC amplitudes. Halothane increased the amplitude of eIPSCs, with a decrease in failure rate (Rf) and paired-pulse ratio. In contrast, isoflurane and enflurane decreased the eIPSC amplitude and increased Rf, while sevoflurane decreased the eIPSC amplitude without affecting Rf. Volatile anesthetics did not change kinetics except for sevoflurane, suggesting that presynaptic mechanisms dominate changes in neurotransmission. Each anesthetic showed somewhat different GABA-induced response and these results suggest that GABA-induced synaptic transmission cannot have a uniformly common site of action as suggested for volatile anesthetics. In contrast, all volatile anesthetics concentration-dependently enhanced the GABA-induced extrasynaptic currents. Extrasynaptic receptors containing α4 and α5 subunits are reported to have high sensitivities to volatile anesthetics. Also, inhibition of GABA uptake by volatile anesthetics results in higher extracellular GABA concentration, which may lead to prolonged activation of extrasynaptic GABAA receptors. The extrasynaptic GABA-induced receptors may be major site of volatile anesthetic-induced neurotransmission. This article is part of a Special Issue entitled 'Extrasynaptic ionotropic receptors'.
Collapse
|
35
|
GABAA receptors facilitate spontaneous glutamate release in rat periaqueductal gray neurons. Neuroreport 2012; 22:834-8. [PMID: 21934537 DOI: 10.1097/wnr.0b013e32834bc733] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
The functional role of presynaptic γ-aminobutyric acid (GABA)(A) receptors in excitatory glutamatergic transmission was examined in rat periaqueductal gray neurons recorded using a conventional whole-cell patch clamp technique. Muscimol, a GABA(A) receptor agonist, significantly increased the frequency of spontaneous excitatory postsynaptic currents without affecting their amplitude, and this effect was completely blocked by the selective GABA(A) receptor antagonist. The muscimol-induced facilitation of spontaneous excitatory postsynaptic current frequency disappeared either in the presence of tetrodotoxin or Cd. The results suggest that the activation of presynaptic GABA(A) receptors directly depolarizes glutamatergic terminals resulting in the facilitation of spontaneous glutamate release, and that presynaptic GABA(A) receptors play an important role in the regulation of various physiological functions mediated by the periaqueductal gray.
Collapse
|
36
|
Beltrán JQ, Gutiérrez R. Co-release of glutamate and GABA from single, identified mossy fibre giant boutons. J Physiol 2012; 590:4789-800. [PMID: 22711957 DOI: 10.1113/jphysiol.2012.236372] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Several laboratories have provided immunohistochemical, molecular biological and electrophysiological evidence that the glutamatergic granule cells of the dentate gyrus can transiently express a GABAergic phenotype during development. Electrophysiological recordings on hippocampal slices obtained during this period have shown that stimulation of the mossy fibres (MFs) provokes simultaneous monosynaptic GABA(A) and glutamate receptor-mediated responses in their target cells,which have the pharmacological and physiological characteristics of MF neurotransmission. This evidence, although strongly supporting the hypothesis that MFs co-release glutamate and GABA, is indirect, as the extracellular stimulation used in slice experiments could activate fibres other than MFs. In this study, we show that selective stimulation of single, identified MF boutons (MFBs) attached to the apical dendrites of dissociated pyramidal cells of developing rats produced synaptic currents mediated by either glutamate receptors only or by both glutamate and GABA(A) receptors. By contrast, stimulation of MFBs of adult rats produced exclusively glutamate receptor-mediated responses. All responses evoked by stimulation of MFBs underwent strong frequency-dependent potentiation and were depressed by the activation of presynaptic metabotropic glutamate receptors. On the other hand, synaptic responses evoked by stimulation of interneuronal boutons located on the soma or on the basal dendrites of the same pyramidal cells were exclusively mediated by GABA(A) receptors, underwent frequency-dependent depression and were unaffected by mGluR agonists.We here demonstrate that the simultaneous glutamatergic and GABAergic responses evoked by MF stimulation in pyramidal cells of CA3 during development have a common origin in the giant MFBs.
Collapse
Affiliation(s)
- Jesús Q Beltrán
- Department of Pharmacobiology, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Calzada de los Tenorios No. 235, México
| | | |
Collapse
|
37
|
Beltrán JQ, Reyes S, Pérez-Guzmán JA, Elías-Viñas D, Gutiérrez R. Dissociation of CA3 pyramidal cells with attached, functional, identified mossy fiber and interneuronal boutons for studying glutamatergic and GABAergic synaptic transmission. J Neurosci Methods 2012; 208:155-60. [PMID: 22633895 DOI: 10.1016/j.jneumeth.2012.05.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 05/07/2012] [Accepted: 05/14/2012] [Indexed: 11/29/2022]
Abstract
Pyramidal cells of CA3 area receive glutamatergic signals from the mossy fibers (MFs), perforant path and collaterals of other pyramidal cells, as well as GABAergic inputs from interneurons. In hippocampal slices, an extracellular stimulation electrode is often used to activate the MFs, with the disadvantage of possibly activating fibers other than MFs. We set-up a preparation that allows the analysis of the glutamatergic input from identified, giant MF boutons as well as of GABAergic inputs from boutons of interneurons on single CA3 pyramidal cells. Mossy fiber boutons were labeled by exposing hippocampal slices to a zinc-reactive fluorescent dye, or by injecting a fluorescent dye in the granule cell layer and allowing its transport along the MFs to their terminals in CA3 area. After conducting an enzyme-free, mechanical dissociation of CA3 area, we obtained pyramidal cells containing fluorescent, giant MF boutons attached to their apical dendrites, as well as boutons of interneuronal origin. Whole cell recordings were then performed, whereby synaptic responses could be evoked by selective stimulation of the identified boutons. The synaptic currents evoked by stimulation of MF boutons, unlike those evoked by stimulation of interneuronal boutons, underwent strong frequency potentiation and were depressed by activation of metabotropic glutamate receptors, which are characteristics of transmission of MF origin. Combination of fluorophores can be used to label different tracts/boutons allowing the study of the different characteristics of neurotransmitter release from a variety of sources on single target cells.
Collapse
Affiliation(s)
- Jesús Q Beltrán
- Departments of Physiology, Biophysics and Neurosciences, Centro de Investigación y Estudios Avanzados del Instituto Politécnico Nacional, Mexico DF 07000, Mexico
| | | | | | | | | |
Collapse
|
38
|
Wakita M, Shin MC, Iwata S, Nonaka K, Akaike N. Effects of ethanol on GABA(A) receptors in GABAergic and glutamatergic presynaptic nerve terminals. J Pharmacol Exp Ther 2012; 341:809-19. [PMID: 22434676 DOI: 10.1124/jpet.111.189126] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Ethanol (EtOH) has a number of behavioral effects, including intoxication, amnesia, and/or sedation, that are thought to relate to the activation of GABA(A) receptors. However, GABA(A) receptors at different cellular locations have different sensitivities to EtOH. The present study used the "synaptic bouton" preparation where we could stimulate nerve endings on mechanically dissociated single rat hippocampal CA1 and CA3 pyramidal neurons and investigate the effects of EtOH on presynaptic and postsynaptic GABA(A) receptors. Low concentrations of EtOH (10 mM) had no effect on postsynaptic GABA(A) and glutamate receptors or voltage-dependent Na(+) and Ca(2+) channels. Higher concentrations (≥100 mM) could significantly inhibit these current responses. EtOH at 10 mM had no direct effect on inhibitory postsynaptic currents (IPSCs) and excitatory postsynaptic currents (EPSCs) evoked by focal stimulation of single boutons [evoked IPSCs (eIPSCs) and evoked EPSCs (eEPSCs)]. However, coapplication of 10 mM EtOH with muscimol decreased the amplitude of eIPSCs and eEPSCs and increased their paired-pulse ratio. The effects on eEPSCs were reversed by bicuculline. Coapplication of muscimol and EtOH significantly increased the frequency of spontaneous IPSCs and EPSCs. The EtOH effects on the postsynaptic responses and eEPSCs were similar in neurons from neonatal and mature rats. These results revealed that low concentrations of EtOH can potentiate the activation of presynaptic GABA(A) receptors to inhibit evoked GABA and glutamate release. These results indicate a high sensitivity of presynaptic GABA(A) receptor to EtOH, which needs to be accounted for when considering the cellular mechanisms of EtOH's physiological responses.
Collapse
Affiliation(s)
- Masahito Wakita
- Research Division for Life Sciences, Kumamoto Health Science University, 325 Izumimachi, Kumamoto, 861-5598, Japan
| | | | | | | | | |
Collapse
|
39
|
Kim BG, Cho JH, Choi IS, Lee MG, Jang IS. Modulation of presynaptic GABA(A) receptors by endogenous neurosteroids. Br J Pharmacol 2012; 164:1698-710. [PMID: 21585348 DOI: 10.1111/j.1476-5381.2011.01491.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Although 3α-hydroxy, 5α-reduced pregnane steroids, such as allopregnanolone (AlloP) and tetrahydrodeoxycorticosterone, are endogenous positive modulators of postsynaptic GABA(A) receptors, the functional roles of endogenous neurosteroids in synaptic transmission are still largely unknown. EXPERIMENTAL APPROACH In this study, the effect of AlloP on spontaneous glutamate release was examined in mechanically isolated dentate gyrus hilar neurons by use of the conventional whole-cell patch-clamp technique. KEY RESULTS AlloP increased the frequency of glutamatergic spontaneous excitatory postsynaptic currents (sEPSCs) in a dose-dependent manner. The AlloP-induced increase in sEPSC frequency was completely blocked by a non-competitive GABA(A) receptor blocker, tetrodotoxin or Cd(2+) , suggesting that AlloP acts on presynaptic GABA(A) receptors to depolarize presynaptic nerve terminals to increase the probability of spontaneous glutamate release. On the other hand, γ-cyclodextrin (γ-CD) significantly decreased the basal frequency of sEPSCs. However, γ-CD failed to decrease the basal frequency of sEPSCs in the presence of a non-competitive GABA(A) receptor antagonist or tetrodotoxin. In addition, γ-CD failed to decrease the basal frequency of sEPSCs after blocking the synthesis of endogenous 5α-reduced pregnane steroids. Furthermore, γ-CD decreased the extent of muscimol-induced increase in sEPSC frequency, suggesting that endogenous neurosteroids can directly activate and/or potentiate presynaptic GABA(A) receptors to affect spontaneous glutamate release onto hilar neurons. CONCLUSIONS AND IMPLICATIONS The modulation of presynaptic GABA(A) receptors by endogenous neurosteroids might affect the excitability of the dentate gyrus-hilus-CA3 network, and thus contribute, at least in part, to some pathological conditions, such as catamenial epilepsy and premenstrual dysphoric disorder.
Collapse
Affiliation(s)
- B-G Kim
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu, Korea
| | | | | | | | | |
Collapse
|
40
|
Synergic effect of diazepam and muscimol via presynaptic GABAA receptors on glutamatergic evoked EPSCs. Brain Res 2011; 1416:1-9. [DOI: 10.1016/j.brainres.2011.07.054] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2011] [Revised: 07/25/2011] [Accepted: 07/27/2011] [Indexed: 11/21/2022]
|
41
|
Cid M, Vilcaes A, Rupil L, Salvatierra N, Roth G. Participation of the GABAergic system on the glutamate release of frontal cortex synaptosomes from Wistar rats with experimental autoimmune encephalomyelitis. Neuroscience 2011; 189:337-44. [DOI: 10.1016/j.neuroscience.2011.05.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2010] [Revised: 05/05/2011] [Accepted: 05/05/2011] [Indexed: 11/16/2022]
|
42
|
Luscher B, Fuchs T, Kilpatrick CL. GABAA receptor trafficking-mediated plasticity of inhibitory synapses. Neuron 2011; 70:385-409. [PMID: 21555068 DOI: 10.1016/j.neuron.2011.03.024] [Citation(s) in RCA: 316] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/30/2011] [Indexed: 12/22/2022]
Abstract
Proper developmental, neural cell-type-specific, and activity-dependent regulation of GABAergic transmission is essential for virtually all aspects of CNS function. The number of GABA(A) receptors in the postsynaptic membrane directly controls the efficacy of GABAergic synaptic transmission. Thus, regulated trafficking of GABA(A) receptors is essential for understanding brain function in both health and disease. Here we summarize recent progress in the understanding of mechanisms that allow dynamic adaptation of cell surface expression and postsynaptic accumulation and function of GABA(A) receptors. This includes activity-dependent and cell-type-specific changes in subunit gene expression, assembly of subunits into receptors, as well as exocytosis, endocytic recycling, diffusion dynamics, and degradation of GABA(A) receptors. In particular, we focus on the roles of receptor-interacting proteins, scaffold proteins, synaptic adhesion proteins, and enzymes that regulate the trafficking and function of receptors and associated proteins. In addition, we review neuropeptide signaling pathways that affect neural excitability through changes in GABA(A)R trafficking.
Collapse
Affiliation(s)
- Bernhard Luscher
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA.
| | | | | |
Collapse
|
43
|
Different effects of α-chloralose on spontaneous and evoked GABA release in rat hippocampal CA1 neurons. Brain Res Bull 2011; 85:180-8. [DOI: 10.1016/j.brainresbull.2011.03.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2010] [Revised: 03/18/2011] [Accepted: 03/22/2011] [Indexed: 01/02/2023]
|
44
|
Abstract
Axons are generally considered as reliable transmission cables in which stable propagation occurs once an action potential is generated. Axon dysfunction occupies a central position in many inherited and acquired neurological disorders that affect both peripheral and central neurons. Recent findings suggest that the functional and computational repertoire of the axon is much richer than traditionally thought. Beyond classical axonal propagation, intrinsic voltage-gated ionic currents together with the geometrical properties of the axon determine several complex operations that not only control signal processing in brain circuits but also neuronal timing and synaptic efficacy. Recent evidence for the implication of these forms of axonal computation in the short-term dynamics of neuronal communication is discussed. Finally, we review how neuronal activity regulates both axon morphology and axonal function on a long-term time scale during development and adulthood.
Collapse
Affiliation(s)
- Dominique Debanne
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| | - Emilie Campanac
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| | - Andrzej Bialowas
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| | - Edmond Carlier
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| | - Gisèle Alcaraz
- Institut National de la Santé et de la Recherche Médicale U.641 and Université de la Méditerranée, Faculté de Médecine Secteur Nord, Marseille, France
| |
Collapse
|
45
|
Axonal GABAA receptors increase cerebellar granule cell excitability and synaptic activity. J Neurosci 2011; 31:565-74. [PMID: 21228165 DOI: 10.1523/jneurosci.4506-10.2011] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We report that activation of GABA(A) receptors on cerebellar granule cell axons modulates both transmitter release and the excitability of the axon and soma. Axonal GABA(A) receptors depolarize the axon, increasing its excitability and causing calcium influx at axonal varicosities. GABA-mediated subthreshold depolarizations in the axon spread electrotonically to the soma, promoting orthodromic action potential initiation. When chloride concentrations are unperturbed, GABA iontophoresis elicits spikes and increases excitability of parallel fibers, indicating that GABA(A) receptor-mediated responses are normally depolarizing. GABA release from molecular layer interneurons activates parallel fiber GABA(A) receptors, and this, in turn, increases release probability at synapses between parallel fibers and molecular layer interneurons. These results describe a positive feedback mechanism whereby transmission from granule cells to Purkinje cells and molecular layer interneurons will be strengthened during granule cell spike bursts evoked by sensory stimulation.
Collapse
|
46
|
Vesselkin NP, Natochin YV. Principles of organization and evolution of systems of regulation of functions. J EVOL BIOCHEM PHYS+ 2011. [DOI: 10.1134/s0022093010060083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
47
|
Yamamoto S, Yoshimura M, Shin MC, Wakita M, Nonaka K, Akaike N. GABAA receptor-mediated presynaptic inhibition on glutamatergic transmission. Brain Res Bull 2011; 84:22-30. [DOI: 10.1016/j.brainresbull.2010.10.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 10/20/2010] [Accepted: 10/21/2010] [Indexed: 11/30/2022]
|
48
|
Abstract
We examined the effect of 2'-3'-O-(4-benzoylbenzoyl)-adenosine-5'-triphosphate (Bz-ATP), a P2X7 receptor agonist, on action potential-independent glutamate release from nerve terminals attached to mechanically isolated immature hilar neurons. Bz-ATP increased spontaneous excitatory postsynaptic current (sEPSC) frequency, and this effect was blocked by Brilliant blue G, a P2X7 receptor antagonist, suggesting that P2X7 receptors mediate the facilitatory action of Bz-ATP on sEPSCs. In most of hilar neurons tested, the Bz-ATP-induced increase in sEPSC frequency was blocked by tetrodotoxin or Cd, suggesting that the activation of P2X7 receptors leads to a presynaptic depolarization. The P2X7 receptor-mediated facilitation of glutamate release would modulate the excitability of hilar neurons, and eventually have a broad impact on the pathophysiological functions mediated by the hippocampus.
Collapse
|
49
|
Park HM, Choi IS, Nakamura M, Cho JH, Lee MG, Jang IS. Multiple effects of allopregnanolone on GABAergic responses in single hippocampal CA3 pyramidal neurons. Eur J Pharmacol 2010; 652:46-54. [PMID: 21118679 DOI: 10.1016/j.ejphar.2010.10.097] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2010] [Revised: 10/16/2010] [Accepted: 10/31/2010] [Indexed: 11/27/2022]
Abstract
3α-Hydroxy, 5α-reduced pregnane steroids, such as allopregnanolone, are potent modulators of GABA(A) receptors and have many biological responses including sedative, anxiolytic, anticonvulsant and anesthetic actions. In the present study, we have investigated the effects of allopregnanolone on GABA(A) receptors in acutely isolated single hippocampal CA3 pyramidal neurons using the whole cell patch-clamp technique. Allopregnanolone induced membrane Cl(-) currents in a concentration-dependent manner, and the allopregnanolone-induced currents (I(AlloP)) were blocked by noncompetitive GABA(A) receptor antagonists. The I(AlloP) was not affected by the intracellular loading of γ-cyclodextrin (γ-CD), which efficiently sequesters several kinds of endogenous neurosteroids including allopregnanolone, suggesting that allopregnanolone accesses extracellular but not intracellular sites to activate GABA(A) receptors. Allopregnanolone prolonged the decay time constant of GABAergic spontaneous inhibitory postsynaptic currents (sIPSCs), suggesting that allopregnanolone modulates the desensitization kinetics of postsynaptic GABA(A) receptors. The picrotoxin-sensitive tonic currents (I(tonic)), which were mediated by extrasynaptic GABA(A) receptors, were recorded from CA3 pyramidal neurons. The intracellular loading of γ-CD or allopregnanolone significantly decreased or increased the amplitude of picrotoxin-sensitive I(tonic), respectively, suggesting that endogenous neurosteroids might, at least in part, be involved in the generation of picrotoxin-sensitive I(tonic). Allopregnanolone also increased the frequency of GABAergic sIPSCs, in a manner dependent on the integrity of voltage-dependent Na(+) and Ca(2+) channels, suggesting that allopregnanolone activates presynaptic GABA(A) receptors to depolarize GABAergic nerve terminals. The present results suggest that allopregnanolone exerts its pharmacological and pathophysiological actions via the modulation of multiple types of GABA(A) receptor-mediated responses.
Collapse
Affiliation(s)
- Hye-Mi Park
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu 700-412, Republic of Korea
| | | | | | | | | | | |
Collapse
|
50
|
Lee KH, Cho JH, Choi IS, Park HM, Lee MG, Choi BJ, Jang IS. Pregnenolone sulfate enhances spontaneous glutamate release by inducing presynaptic Ca2+-induced Ca2+ release. Neuroscience 2010; 171:106-16. [PMID: 20816925 DOI: 10.1016/j.neuroscience.2010.07.057] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2010] [Revised: 07/27/2010] [Accepted: 07/27/2010] [Indexed: 01/08/2023]
Abstract
Pregnenolone sulfate (PS) acts as an excitatory neuromodulator and has a variety of neuropharmacological actions, such as memory enhancement and convulsant effects. In the present study, we investigated the effect of PS on glutamatergic spontaneous excitatory postsynaptic currents (sEPSCs) in acutely isolated dentate gyrus (DG) hilar neurons by use of a conventional whole-cell patch-clamp technique. PS significantly increased sEPSC frequency in a concentration-dependent manner without affecting the current amplitude, suggesting that PS acts presynaptically to increase the probability of spontaneous glutamate release. However, known molecular targets of PS, such as α7 nicotinic ACh, NMDA, σ1 receptors and voltage-dependent Ca(2+) channels, were not responsible for the PS-induced increase in sEPSC frequency. In contrast, the PS-induced increase in sEPSC frequency was completely occluded in a Ca(2+)-free external solution, and was significantly reduced by either the depletion of presynaptic Ca(2+) stores or the blockade of ryanodine receptors, suggesting that PS elicits Ca(2+)-induced Ca(2+) release (CICR) within glutamatergic nerve terminals. In addition, the PS-induced increase in sEPSC frequency was completely occluded by transient receptor potential (TRP) channel blockers. These data suggest that PS increases spontaneous glutamate release onto acutely isolated hilar neurons via presynaptic CICR, which was triggered by the influx of Ca(2+) through presynaptic TRP channels. The PS-induced modulation of excitatory transmission onto hilar neurons could have a broad impact on the excitability of hilar neurons and affect the pathophysiological functions mediated by the hippocampus.
Collapse
Affiliation(s)
- K H Lee
- Department of Pharmacology, School of Dentistry, Kyungpook National University, Daegu 700-412, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|