1
|
Schieferstein N, Del Toro A, Evangelista R, Imbrosci B, Swaminathan A, Schmitz D, Maier N, Kempter R. Propagation of sharp wave-ripple activity in the mouse hippocampal CA3 subfield in vitro. J Physiol 2024; 602:5039-5059. [PMID: 39216085 DOI: 10.1113/jp285671] [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: 09/12/2023] [Accepted: 08/08/2024] [Indexed: 09/04/2024] Open
Abstract
Sharp wave-ripple complexes (SPW-Rs) are spontaneous oscillatory events that characterize hippocampal activity during resting periods and slow-wave sleep. SPW-Rs are related to memory consolidation - the process during which newly acquired memories are transformed into long-lasting memory traces. To test the involvement of SPW-Rs in this process, it is crucial to understand how SPW-Rs originate and propagate throughout the hippocampus. SPW-Rs can originate in CA3, and they typically spread from CA3 to CA1, but little is known about their formation within CA3. To investigate the generation and propagation of SPW-Rs in CA3, we recorded from mouse hippocampal slices using multi-electrode arrays and patch-clamp electrodes. We characterized extracellular and intracellular correlates of SPW-Rs and quantified their propagation along the pyramidal cell layer of CA3. We found that a hippocampal slice can be described by a speed and a direction of propagation of SPW-Rs. The preferred propagation direction was from CA3c (the subfield closer to the dentate gyrus) toward CA3a (the subfield at the boundary to CA2). In patch-clamp recordings from CA3 pyramidal neurons, propagation was estimated separately for excitatory and inhibitory currents associated with SPW-Rs. We found that propagation speed and direction of excitatory and inhibitory currents were correlated. The magnitude of the speed of propagation of SPW-Rs within CA3 was consistent with the speed of propagation of action potentials in axons of CA3 principal cells. KEY POINTS: Hippocampal sharp waves are considered important for memory consolidation; therefore, it is of interest to understand the mechanisms of their generation and propagation. Here, we used two different approaches to study the propagation of sharp waves in mouse CA3 in vitro: multi-electrode arrays and multiple single-cell recordings. We find a preferred direction of propagation of sharp waves from CA3c toward CA3a - both in the local field potential and in sharp wave-associated excitatory and inhibitory synaptic activity. The speed of sharp wave propagation is consistent with the speed of action potential propagation along the axons of CA3 pyramidal neurons. These new insights into the dynamics of sharp waves in the CA3 network will inform future experiments and theoretical models of sharp-wave generation mechanisms.
Collapse
Affiliation(s)
- Natalie Schieferstein
- Department of Biology, Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Ana Del Toro
- Department of Biology, Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Roberta Evangelista
- Department of Biology, Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
| | - Barbara Imbrosci
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Aarti Swaminathan
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Dietmar Schmitz
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- NeuroCure Cluster of Excellence, Berlin, Germany
- Einstein Center for Neurosciences (ECN) Berlin, Berlin, Germany
| | - Nikolaus Maier
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Richard Kempter
- Department of Biology, Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany
- Bernstein Center for Computational Neuroscience Berlin, Berlin, Germany
- Einstein Center for Neurosciences (ECN) Berlin, Berlin, Germany
| |
Collapse
|
2
|
Yamasaki R. Connexins Control Glial Inflammation in Various Neurological Diseases. Int J Mol Sci 2023; 24:16879. [PMID: 38069203 PMCID: PMC10706219 DOI: 10.3390/ijms242316879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/25/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Connexins (Cxs) form gap junctions through homotypic/heterotypic oligomerization. Cxs are initially synthesized in the endoplasmic reticulum, then assembled as hexamers in the Golgi apparatus before being integrated into the cell membrane as hemichannels. These hemichannels remain closed until they combine to create gap junctions, directly connecting neighboring cells. Changes in the intracellular or extracellular environment are believed to trigger the opening of hemichannels, creating a passage between the inside and outside of the cell. The size of the channel pore depends on the Cx isoform and cellular context-specific effects such as posttranslational modifications. Hemichannels allow various bioactive molecules, under ~1 kDa, to move in and out of the host cell in the direction of the electrochemical gradient. In this review, we explore the fundamental roles of Cxs and their clinical implications in various neurological dysfunctions, including hereditary diseases, ischemic brain disorders, degenerative conditions, demyelinating disorders, and psychiatric illnesses. The influence of Cxs on the pathomechanisms of different neurological disorders varies depending on the circumstances. Hemichannels are hypothesized to contribute to proinflammatory effects by releasing ATP, adenosine, glutamate, and other bioactive molecules, leading to neuroglial inflammation. Modulating Cxs' hemichannels has emerged as a promising therapeutic approach.
Collapse
Affiliation(s)
- Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
3
|
Gilloteaux J, De Swert K, Suain V, Brion JP, Nicaise C. Loss of Ephaptic Contacts in the Murine Thalamus during Osmotic Demyelination Syndrome. Ultrastruct Pathol 2023; 47:398-423. [PMID: 37477534 DOI: 10.1080/01913123.2023.2232452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 06/10/2023] [Accepted: 06/29/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND AND AIM A murine model mimicking osmotic demyelination syndrome (ODS) revealed with histology in the relay posterolateral (VPL) and ventral posteromedial (VPM) thalamic nuclei adjoined nerve cell bodies in chronic hyponatremia, amongst the damaged 12 h and 48 h after reinstatement of osmolality. This report aims to verify and complement with ultrastructure other neurophysiology, immunohistochemistry, and molecular biochemistry data to assess the connexin-36 protein, as part of those hinted close contacts.This ODS investigation included four groups of mice: Sham (NN; n = 13), hyponatremic (HN; n = 11), those sacrificed 12 h after a fast restoration of normal natremia (ODS12h; n = 6) and mice sacrificed 48 h afterward, or ODS48 h (n = 9). Out of these, thalamic zones samples included NN (n = 2), HN (n = 2), ODS12h (n = 3) and ODS48h (n = 3). RESULTS Ultrastructure illustrated junctions between nerve cell bodies that were immunolabeled with connexin36 (Cx36) with light microscopy and Western blots. These cell's junctions were reminiscent of low resistance junctions characterized in other regions of the CNS with electrophysiology. Contiguous neurons showed neurolemma contacts in intact and damaged tissues according to their location in the ODS zones, at 12 h and 48 h post correction along with other demyelinating alterations. Neurons and ephaptic contact measurements indicated the highest alterations, including nerve cell necrosis in the ODS epicenter and damages decreased toward the outskirts of the demyelinated zone. CONCLUSION Ephapses contained C × 36between intact or ODS injured neurons in the thalamus appeared to be resilient beyond the core degraded tissue injuries. These could maintain intercellular ionic and metabolite exchanges between these lesser injured regions and, thus, would partake to some brain plasticity repairs.
Collapse
Affiliation(s)
- Jacques Gilloteaux
- Department of Medicine, URPHyM, NARILIS, Université de Namur, Namur, Belgium
- Department of Anatomical Sciences, St George's University School of Medicine, Newcastle Upon Tyne, UK
| | - Kathleen De Swert
- Department of Medicine, URPHyM, NARILIS, Université de Namur, Namur, Belgium
| | - Valérie Suain
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Jean-Pierre Brion
- Laboratory of Histology, Neuroanatomy and Neuropathology, Faculty of Medicine, Université Libre de Bruxelles, Brussels, Belgium
| | - Charles Nicaise
- Department of Medicine, URPHyM, NARILIS, Université de Namur, Namur, Belgium
| |
Collapse
|
4
|
Zlomuzica A, Plank L, Kodzaga I, Dere E. A fatal alliance: Glial connexins, myelin pathology and mental disorders. J Psychiatr Res 2023; 159:97-115. [PMID: 36701970 DOI: 10.1016/j.jpsychires.2023.01.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 01/02/2023] [Accepted: 01/09/2023] [Indexed: 01/12/2023]
Abstract
Mature oligodendrocytes are myelin forming glial cells which are responsible for myelination of neuronal axons in the white matter of the central nervous system. Myelin pathology is a major feature of severe neurological disorders. Oligodendrocyte-specific gene mutations and/or white matter alterations have also been addressed in a variety of mental disorders. Breakdown of myelin integrity and demyelination is associated with severe symptoms, including impairments in motor coordination, breathing, dysarthria, perception (vision and hearing), and cognition. Furthermore, there is evidence indicating that myelin sheath defects and white matter pathology contributes to the affective and cognitive symptoms of patients with mental disorders. Oligodendrocytes express the connexins GJC2; mCx47 [human (GJC2) and mouse (mCx47) connexin gene nomenclature according to Söhl and Willecke (2003)], GJB1; mCx32, and GJD1; mCx29 in both white and gray matter. Preclinical findings indicate that alterations in connexin expression in oligodendrocytes and astrocytes can induce myelin defects. GJC2; mCx47 is expressed at early embryonic stages in oligodendrocyte precursors cells which precedes central nervous system myelination. In adult humans and animals GJC2, respectively mCx47 expression is essential for oligodendrocyte function and ensures adequate myelination as well as myelin maintenance in the central nervous system. In the past decade, evidence has accumulated suggesting that mental disorders can be accompanied by changes in connexin expression, myelin sheath defects and corresponding white matter alterations. This dual pathology could compromise inter-neuronal information transfer, processing and communication and eventually contribute to behavioral, sensory-motor, affective and cognitive symptoms in patients with mental disorders. The induction of myelin repair and remyelination in the central nervous system of patients with mental disorders could help to restore normal neuronal information propagation and ameliorate behavioral and cognitive symptoms in individuals with mental disorders.
Collapse
Affiliation(s)
- Armin Zlomuzica
- Department of Behavioral and Clinical Neuroscience, Ruhr-University Bochum (RUB), Massenbergstraße 9-13, D-44787, Bochum, Germany.
| | - Laurin Plank
- Department of Behavioral and Clinical Neuroscience, Ruhr-University Bochum (RUB), Massenbergstraße 9-13, D-44787, Bochum, Germany
| | - Iris Kodzaga
- Department of Behavioral and Clinical Neuroscience, Ruhr-University Bochum (RUB), Massenbergstraße 9-13, D-44787, Bochum, Germany
| | - Ekrem Dere
- Department of Behavioral and Clinical Neuroscience, Ruhr-University Bochum (RUB), Massenbergstraße 9-13, D-44787, Bochum, Germany; Sorbonne Université, UFR des Sciences de la Vie, 9 quai Saint Bernard, F-75005, Paris, France.
| |
Collapse
|
5
|
Ribarič S. Detecting Early Cognitive Decline in Alzheimer's Disease with Brain Synaptic Structural and Functional Evaluation. Biomedicines 2023; 11:355. [PMID: 36830892 PMCID: PMC9952956 DOI: 10.3390/biomedicines11020355] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Early cognitive decline in patients with Alzheimer's (AD) is associated with quantifiable structural and functional connectivity changes in the brain. AD dysregulation of Aβ and tau metabolism progressively disrupt normal synaptic function, leading to loss of synapses, decreased hippocampal synaptic density and early hippocampal atrophy. Advances in brain imaging techniques in living patients have enabled the transition from clinical signs and symptoms-based AD diagnosis to biomarkers-based diagnosis, with functional brain imaging techniques, quantitative EEG, and body fluids sampling. The hippocampus has a central role in semantic and episodic memory processing. This cognitive function is critically dependent on normal intrahippocampal connections and normal hippocampal functional connectivity with many cortical regions, including the perirhinal and the entorhinal cortex, parahippocampal cortex, association regions in the temporal and parietal lobes, and prefrontal cortex. Therefore, decreased hippocampal synaptic density is reflected in the altered functional connectivity of intrinsic brain networks (aka large-scale networks), including the parietal memory, default mode, and salience networks. This narrative review discusses recent critical issues related to detecting AD-associated early cognitive decline with brain synaptic structural and functional markers in high-risk or neuropsychologically diagnosed patients with subjective cognitive impairment or mild cognitive impairment.
Collapse
Affiliation(s)
- Samo Ribarič
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
6
|
Wang G, Wu X. The potential antiepileptogenic effect of neuronal Cx36 gap junction channel blockage. Transl Neurosci 2021; 12:46-51. [PMID: 33604079 PMCID: PMC7876775 DOI: 10.1515/tnsci-2021-0008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/14/2020] [Accepted: 01/08/2021] [Indexed: 12/21/2022] Open
Abstract
Epilepsy is one of the most prevalent neurological disorders and can result in neuronal injury and degeneration. Consequently, research into new antiepileptic drugs capable of providing protection against neuronal injury and degeneration is extremely important. Neuronal Cx36 gap junction channels have been found to play an important role in epilepsy; thus, pharmacological interference using Cx36 gap junction channel blockers may be a promising strategy for disrupting the synchronization of neurons during seizure activity and protecting neurons. Based on these promising findings, several in vivo and in vitro studies are ongoing and the first encouraging results have been published. The results bring hope that neurons can be protected from injury and degeneration in patients with epilepsy, which is currently impossible.
Collapse
Affiliation(s)
- Guangliang Wang
- Department of Cardiology, Far Eastern Horizon Hospital, Linghai, Liaoning, People's Republic of China
| | - Xuemei Wu
- Department of Pediatric Neurology, First Hospital of Jilin University, 1 Xinmin Street, Changchun 130000, Jilin, People's Republic of China
| |
Collapse
|
7
|
Harnessing the therapeutic potential of antibodies targeting connexin hemichannels. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166047. [PMID: 33418036 DOI: 10.1016/j.bbadis.2020.166047] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/17/2020] [Accepted: 12/03/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Connexin hemichannels have been implicated in pathology-promoting conditions, including inflammation, numerous widespread human diseases, including cancer and diabetes, and several rare diseases linked to pathological point mutations. METHODS We analysed the literature focusing on antibodies capable of modulating hemichannel function, highlighting generation methods, applications to basic biomedical research and translational potential. RESULTS Anti-hemichannel antibodies generated over the past 3 decades targeted mostly connexin 43, with a focus on cancer treatment. A slow transition from relatively unselective polyclonal antibodies to more selective monoclonal antibodies resulted in few products with interesting characteristics that are under evaluation for clinical trials. Selection of antibodies from combinatorial phage-display libraries, has permitted to engineer a monoclonal antibody that binds to and blocks pathological hemichannels formed by connexin 26, 30 and 32. CONCLUSIONS All known antibodies that modulate connexin hemichannels target the two small extracellular loops of the connexin proteins. The extracellular region of different connexins is highly conserved, and few residues of each connexins are exposed. The search for new antibodies may develop an unprecedented potential for therapeutic applications, as it may benefit tremendously from novel whole-cell screening platforms that permit in situ selection of antibodies against membrane proteins in native state. The demonstrated efficacy of mAbs in reaching and modulating hemichannels in vivo, together with their relative specificity for connexins overlapping epitopes, should hopefully stimulate an interest for widening the scope of anti-hemichannel antibodies. There is no shortage of currently incurable diseases for which therapeutic intervention may benefit from anti-hemichannel antibodies capable of modulating hemichannel function selectively and specifically.
Collapse
|
8
|
Generation of Sharp Wave-Ripple Events by Disinhibition. J Neurosci 2020; 40:7811-7836. [PMID: 32913107 PMCID: PMC7548694 DOI: 10.1523/jneurosci.2174-19.2020] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 06/29/2020] [Accepted: 07/17/2020] [Indexed: 11/21/2022] Open
Abstract
Sharp wave-ripple complexes (SWRs) are hippocampal network phenomena involved in memory consolidation. To date, the mechanisms underlying their occurrence remain obscure. Here, we show how the interactions between pyramidal cells, parvalbumin-positive (PV+) basket cells, and an unidentified class of anti-SWR interneurons can contribute to the initiation and termination of SWRs. Using a biophysically constrained model of a network of spiking neurons and a rate-model approximation, we demonstrate that SWRs emerge as a result of the competition between two interneuron populations and the resulting disinhibition of pyramidal cells. Our models explain how the activation of pyramidal cells or PV+ cells can trigger SWRs, as shown in vitro, and suggests that PV+ cell-mediated short-term synaptic depression influences the experimentally reported dynamics of SWR events. Furthermore, we predict that the silencing of anti-SWR interneurons can trigger SWRs. These results broaden our understanding of the microcircuits supporting the generation of memory-related network dynamics. SIGNIFICANCE STATEMENT The hippocampus is a part of the mammalian brain that is crucial for episodic memories. During periods of sleep and inactive waking, the extracellular activity of the hippocampus is dominated by sharp wave-ripple events (SWRs), which have been shown to be important for memory consolidation. The mechanisms regulating the emergence of these events are still unclear. We developed a computational model to study the emergence of SWRs and to explain the roles of different cell types in regulating them. The model accounts for several previously unexplained features of SWRs and thus advances the understanding of memory-related dynamics.
Collapse
|
9
|
Caufriez A, Böck D, Martin C, Ballet S, Vinken M. Peptide-based targeting of connexins and pannexins for therapeutic purposes. Expert Opin Drug Discov 2020; 15:1213-1222. [PMID: 32539572 DOI: 10.1080/17460441.2020.1773787] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Connexin and pannexin (hemi)channels play an important role in paracrine and autocrine signaling pathways. The opening of these cellular pores is linked to a wide range of diseases. Therefore, pharmacological closing of connexin and pannexin (hemi)channels seems a promising therapeutic strategy. However, the currently available inhibitors cope with recurring problems concerning selectivity, specificity, stability and/or solubility. AREAS COVERED A number of peptides that mimic specific regions in the native sequence of connexins and pannexins have the potential to overcome some of these hurdles. In this paper, an overview is provided on these peptide-based inhibitors of connexin and pannexin (hemi)channels for therapeutic purposes. The authors also provide the reader with their expert perspectives on the future of these peptide-based inhibitors. EXPERT OPINION Peptide mimetics can become valuable tools in the treatment of connexin-related and pannexin-related diseases. This can be made possible provided that available peptides are optimized, and new peptide mimetics are designed based on knowledge of the mechanisms underlying the gating control of connexin and pannexin (hemi)channels.
Collapse
Affiliation(s)
- Anne Caufriez
- Department of in Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel , 1090, Brussels, Belgium
| | - Denise Böck
- Department of in Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel , 1090, Brussels, Belgium
| | - Charlotte Martin
- Department of Organic Chemistry, Vrije Universiteit Brussel , 1050, Brussels, Belgium
| | - Steven Ballet
- Department of Organic Chemistry, Vrije Universiteit Brussel , 1050, Brussels, Belgium
| | - Mathieu Vinken
- Department of in Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel , 1090, Brussels, Belgium
| |
Collapse
|
10
|
McKenzie S, Nitzan N, English DF. Mechanisms of neural organization and rhythmogenesis during hippocampal and cortical ripples. Philos Trans R Soc Lond B Biol Sci 2020; 375:20190237. [PMID: 32248777 PMCID: PMC7209923 DOI: 10.1098/rstb.2019.0237] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2019] [Indexed: 12/19/2022] Open
Abstract
Neural activity during ripples has attracted great theoretical and experimental attention over the last three decades. Perhaps one reason for such interest is that ripples occur during quiet waking moments and during sleep, times when we reflect and dream about what has just occurred and what we expect to happen next. The hope is that understanding such 'offline' activity may yield insights into reflection, planning, and the purposes of sleep. This review focuses on the mechanisms by which neurons organize during these high-frequency events. In studying ripples, broader principles have emerged that relate intrinsic neural properties, network topology and synaptic plasticity in controlling neural activity. Ripples, therefore, serve as an excellent model for studying how properties of a neural network relate to neural dynamics. This article is part of the Theo Murphy meeting issue 'Memory reactivation: replaying events past, present and future'.
Collapse
Affiliation(s)
- Sam McKenzie
- NYULMC Neuroscience Institute, New York, NY, USA
| | - Noam Nitzan
- Neuroscience Research Center NWFZ, Berlin, Germany
| | - Daniel F. English
- Virginia Tech School of Neuroscience Blacksburg, Blacksburg, VA, USA
| |
Collapse
|
11
|
Naggar I, Stewart M, Orman R. High Frequency Oscillations in Rat Hippocampal Slices: Origin, Frequency Characteristics, and Spread. Front Neurol 2020; 11:326. [PMID: 32390935 PMCID: PMC7188778 DOI: 10.3389/fneur.2020.00326] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Accepted: 04/03/2020] [Indexed: 11/13/2022] Open
Abstract
Field potential oscillations reflect repetitive firing and synaptic activity by ensembles of neurons in certain areas of the brain. They can be distinguished as slow (e.g., alpha, delta, and theta), fast (e.g., beta and gamma), and high frequency oscillations (HFOs). Neuronal oscillations are involved in a variety of physiological and pathophysiological states such as cognition, consciousness, and seizures. The laminar structure of rat hippocampus affords a way to study these oscillations in hippocampal slices. Rat ventral hippocampal brain slices were cut and maintained in a recording chamber that permitted 64 simultaneous extracellular recordings in the presence of artificial CSF. Brief single stimulus pulses were applied with bipolar electrodes to the CA3 or CA1 regions of hippocampus. Single pulses triggered epileptiform population events that included HFOs in the 150-250 Hz range in the presence of GABAA receptor blockade or kainic acid. HFOs also occurred spontaneously in the presence of kainic acid. The oscillations had the largest amplitude in the CA3c cell layer, regardless of the drug, and were synchronized throughout the cell layer. AMPA receptor blockade stopped these HFOs, whereas NMDA receptor blockade did not. Gap junction activation did not restore HFOs in the presence of AMPA receptor blockade. Our findings suggest that a population of excitatory neurons in CA3c may be a primary focus of seizure-like activity in Ammon's Horn. We suggest that the interconnection of CA3c is different from the rest of CA3.
Collapse
Affiliation(s)
- Isaac Naggar
- EEG Section, NINDS, National Institutes of Health, Bethesda, MD, United States
| | - Mark Stewart
- Department of Neurology, State University of New York Downstate Medical Center, Brooklyn, NY, United States.,Department of Physiology and Pharmacology, State University of New York Downstate Medical Center, Brooklyn, NY, United States
| | - Rena Orman
- Department of Physiology and Pharmacology, State University of New York Downstate Medical Center, Brooklyn, NY, United States
| |
Collapse
|
12
|
Sánchez OF, Rodríguez AV, Velasco-España JM, Murillo LC, Sutachan JJ, Albarracin SL. Role of Connexins 30, 36, and 43 in Brain Tumors, Neurodegenerative Diseases, and Neuroprotection. Cells 2020; 9:E846. [PMID: 32244528 PMCID: PMC7226843 DOI: 10.3390/cells9040846] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 02/15/2020] [Accepted: 02/24/2020] [Indexed: 02/07/2023] Open
Abstract
Gap junction (GJ) channels and their connexins (Cxs) are complex proteins that have essential functions in cell communication processes in the central nervous system (CNS). Neurons, astrocytes, oligodendrocytes, and microglial cells express an extraordinary repertory of Cxs that are important for cell to cell communication and diffusion of metabolites, ions, neurotransmitters, and gliotransmitters. GJs and Cxs not only contribute to the normal function of the CNS but also the pathological progress of several diseases, such as cancer and neurodegenerative diseases. Besides, they have important roles in mediating neuroprotection by internal or external molecules. However, regulation of Cx expression by epigenetic mechanisms has not been fully elucidated. In this review, we provide an overview of the known mechanisms that regulate the expression of the most abundant Cxs in the central nervous system, Cx30, Cx36, and Cx43, and their role in brain cancer, CNS disorders, and neuroprotection. Initially, we focus on describing the Cx gene structure and how this is regulated by epigenetic mechanisms. Then, the posttranslational modifications that mediate the activity and stability of Cxs are reviewed. Finally, the role of GJs and Cxs in glioblastoma, Alzheimer's, Parkinson's, and Huntington's diseases, and neuroprotection are analyzed with the aim of shedding light in the possibility of using Cx regulators as potential therapeutic molecules.
Collapse
Affiliation(s)
- Oscar F. Sánchez
- Department of Nutrition and Biochemistry, Pontificia Universidad Javeriana, 110911 Bogota, Colombia; (A.V.R.); (J.M.V.-E.); (L.C.M.); (J.-J.S.)
| | | | | | | | | | - Sonia-Luz Albarracin
- Department of Nutrition and Biochemistry, Pontificia Universidad Javeriana, 110911 Bogota, Colombia; (A.V.R.); (J.M.V.-E.); (L.C.M.); (J.-J.S.)
| |
Collapse
|
13
|
Lévesque M, Ragsdale D, Avoli M. Evolving Mechanistic Concepts of Epileptiform Synchronization and their Relevance in Curing Focal Epileptic Disorders. Curr Neuropharmacol 2020; 17:830-842. [PMID: 30479217 PMCID: PMC7052840 DOI: 10.2174/1570159x17666181127124803] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 10/26/2018] [Accepted: 11/17/2018] [Indexed: 01/01/2023] Open
Abstract
The synchronized activity of neuronal networks under physiological conditions is mirrored by specific oscillatory patterns of the EEG that are associated with different behavioral states and cognitive functions. Excessive synchronization can, however, lead to focal epileptiform activity characterized by interictal and ictal discharges in epileptic patients and animal models. This review focusses on studies that have addressed epileptiform synchronization in temporal lobe regions by employing in vitro and in vivo recording techniques. First, we consider the role of ionotropic and metabotropic excitatory glutamatergic transmission in seizure generation as well as the paradoxical role of GABAA signaling in initiating and perhaps maintaining focal seizure activity. Second, we address non-synaptic mechanisms (which include voltage-gated ionic currents and gap junctions) in the generation of epileptiform synchronization. For each mechanism, we discuss the actions of antiepileptic drugs that are presumably modulating excitatory or inhibitory signaling and voltage-gated currents to prevent seizures in epileptic patients. These findings provide insights into the mechanisms of seizure initiation and maintenance, thus leading to the development of specific pharmacological treatments for focal epileptic disorders.
Collapse
Affiliation(s)
- Maxime Lévesque
- Montreal Neurological Institute, McGill University, Montreal, H3A 2B4 Quebec, Canada
| | - David Ragsdale
- Montreal Neurological Institute, McGill University, Montreal, H3A 2B4 Quebec, Canada
| | - Massimo Avoli
- Montreal Neurological Institute, McGill University, Montreal, H3A 2B4 Quebec, Canada.,Departments of Neurology & Neurosurgery, and of Physiology, McGill University, Montréal, H3A 2B4 Québec, Canada.,Department of Experimental Medicine, Facoltà di Medicina e Odontoiatria, Sapienza University of Rome, 00185 Roma, Italy
| |
Collapse
|
14
|
Holzbecher A, Kempter R. Interneuronal gap junctions increase synchrony and robustness of hippocampal ripple oscillations. Eur J Neurosci 2019; 48:3446-3465. [PMID: 30414336 DOI: 10.1111/ejn.14267] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 10/12/2018] [Accepted: 10/31/2018] [Indexed: 01/21/2023]
Abstract
Sharp wave-ripples (SWRs) are important for memory consolidation. Their signature in the hippocampal extracellular field potential can be decomposed into a ≈100 ms long sharp wave superimposed by ≈200 Hz ripple oscillations. How ripple oscillations are generated is currently not well understood. A promising model for the genesis of ripple oscillations is based on recurrent interneuronal networks (INT-INT). According to this hypothesis, the INT-INT network in CA1 receives a burst of excitation from CA3 that generates the sharp wave, and recurrent inhibition leads to an ultrafast synchronization of the CA1 network causing the ripple oscillations; fast-spiking parvalbumin-positive basket cells (PV+ BCs) may constitute the ripple-generating interneuronal network. PV+ BCs are also coupled by gap junctions (GJs) but the function of GJs for ripple oscillations has not been quantified. Using simulations of CA1 hippocampal networks of PV+ BCs, we show that GJs promote synchrony beyond a level that could be obtained by only inhibition. GJs also increase the neuronal firing rate of the interneuronal ensemble, while they affect the ripple frequency only mildly. The promoting effect of GJs on ripple oscillations depends on fast GJ transmission ( ≲ 0.5 ms), which requires proximal GJ coupling ( ≲ 100 μm from soma), but is robust to variability in the delay and the amplitude of GJ coupling.
Collapse
Affiliation(s)
- André Holzbecher
- Department of Biology, Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany.,Bernstein Center for Computational Neuroscience, Berlin, Germany
| | - Richard Kempter
- Department of Biology, Institute for Theoretical Biology, Humboldt-Universität zu Berlin, Berlin, Germany.,Bernstein Center for Computational Neuroscience, Berlin, Germany.,Einstein Center for Neurosciences Berlin, Berlin, Germany
| |
Collapse
|
15
|
Shin H, Son Y, Chae U, Kim J, Choi N, Lee HJ, Woo J, Cho Y, Yang SH, Lee CJ, Cho IJ. Multifunctional multi-shank neural probe for investigating and modulating long-range neural circuits in vivo. Nat Commun 2019; 10:3777. [PMID: 31439845 PMCID: PMC6706395 DOI: 10.1038/s41467-019-11628-5] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 07/26/2019] [Indexed: 01/18/2023] Open
Abstract
Investigation and modulation of neural circuits in vivo at the cellular level are very important for studying functional connectivity in a brain. Recently, neural probes with stimulation capabilities have been introduced, and they provided an opportunity for studying neural activities at a specific region in the brain using various stimuli. However, previous methods have a limitation in dissecting long-range neural circuits due to inherent limitations on their designs. Moreover, the large size of the previously reported probes induces more significant tissue damage. Herein, we present a multifunctional multi-shank MEMS neural probe that is monolithically integrated with an optical waveguide for optical stimulation, microfluidic channels for drug delivery, and microelectrode arrays for recording neural signals from different regions at the cellular level. In this work, we successfully demonstrated the functionality of our probe by confirming and modulating the functional connectivity between the hippocampal CA3 and CA1 regions in vivo. Microelectromechanical neural probes can cause tissue damage and often cannot record from distant brain areas. Here the authors combine electrical recording, optical stimulation and microfluidic drug delivery in one multi-shank probe with thinner shanks to reduce damage and a flexible design to target long-range neural circuits.
Collapse
Affiliation(s)
- Hyogeun Shin
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Korea.,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Daejeon, Korea
| | - Yoojin Son
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Korea
| | - Uikyu Chae
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Korea.,School of Electrical Engineering, Korea University, Seoul, Korea
| | | | - Nakwon Choi
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Korea.,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Daejeon, Korea
| | - Hyunjoo J Lee
- School of Electrical Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Korea
| | - Jiwan Woo
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Korea
| | - Yakdol Cho
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Korea
| | - Soo Hyun Yang
- Department of Anatomy, College of Medicine, Korea University, Seoul, Korea
| | - C Justin Lee
- Center for Neuroscience, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Korea
| | - Il-Joo Cho
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Korea. .,Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Daejeon, Korea.
| |
Collapse
|
16
|
Li Q, Li QQ, Jia JN, Liu ZQ, Zhou HH, Mao XY. Targeting gap junction in epilepsy: Perspectives and challenges. Biomed Pharmacother 2018; 109:57-65. [PMID: 30396092 DOI: 10.1016/j.biopha.2018.10.068] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/08/2018] [Accepted: 10/12/2018] [Indexed: 12/21/2022] Open
Abstract
Gap junctions (GJs) are multiple cellular intercellular connections that allow ions to pass directly into the cytoplasm of neighboring cells. Electrical coupling mediated by GJs plays a role in the generation of highly synchronous electrical activity. Accumulative investigations show that GJs in the brain are involved in the generation, synchronization and maintenance of seizure events. At the same time, GJ blockers exert potent curative potential on epilepsy in vivo or in vitro. This review aims to shed light on the role of GJs in epileptogenesis. Targeting GJs is likely to be served as a novel therapeutic approach on epileptic patients.
Collapse
Affiliation(s)
- Qin Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, Hunan, China
| | - Qiu-Qi Li
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, Hunan, China
| | - Ji-Ning Jia
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, Hunan, China
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, Hunan, China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, Hunan, China
| | - Xiao-Yuan Mao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha 410008, China; Institute of Clinical Pharmacology, Hunan Key Laboratory of Pharmacogenetics, Central South University, Changsha 410078, Hunan, China.
| |
Collapse
|
17
|
Traub RD, Whittington MA, Gutiérrez R, Draguhn A. Electrical coupling between hippocampal neurons: contrasting roles of principal cell gap junctions and interneuron gap junctions. Cell Tissue Res 2018; 373:671-691. [PMID: 30112572 DOI: 10.1007/s00441-018-2881-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Accepted: 07/03/2018] [Indexed: 11/28/2022]
Abstract
There is considerable experimental evidence, anatomical and physiological, that gap junctions exist in the hippocampus. Electrical coupling through these gap junctions may be divided into three types: between principal neurons, between interneurons and at mixed chemical (glutamatergic)/electrical synapses. An approach, combining in vitro experimental with modeling techniques, sheds some light on the functional consequences of electrical coupling, for network oscillations and for seizures. Additionally, in vivo experiments, using mouse connexin knockouts, suggest that the presence of electrical coupling is important for optimal performance on selected behavioral tasks; however, the interpretation of such data, in cellular terms, has so far proven difficult. Given that invertebrate central pattern generators so often depend on both chemical and electrical synapses, our hypothesis is that hippocampus-mediated and -influenced behaviors will act likewise. Experiments, likely hard ones, will be required to test this intuition.
Collapse
Affiliation(s)
- Roger D Traub
- Department of Physical Sciences, IBM T.J. Watson Research Center, Yorktown Heights, NY, 10598, USA.
| | | | - Rafael Gutiérrez
- Department of Pharmacobiology, Centro de Investigación y de Estudios Avanzados del IPN, Calzada de los Tenorios 235, 14330, Mexico City, Mexico.,Institut für Physiologie und Pathophysiologie, Universität Heidelberg, Im Neuenheimer Feld 326, 69120, Heidelberg, Germany
| | - Andreas Draguhn
- Institut für Physiologie und Pathophysiologie, Universität Heidelberg, Im Neuenheimer Feld 326, 69120, Heidelberg, Germany
| |
Collapse
|
18
|
Wellbourne-Wood J, Chatton JY. From Cultured Rodent Neurons to Human Brain Tissue: Model Systems for Pharmacological and Translational Neuroscience. ACS Chem Neurosci 2018; 9:1975-1985. [PMID: 29847093 DOI: 10.1021/acschemneuro.8b00098] [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: 12/21/2022] Open
Abstract
To investigate the enormous complexity of the functional and pathological brain there are a number of possible experimental model systems to choose from. Depending on the research question choosing the appropriate model may not be a trivial task, and given the dynamic and intricate nature of an intact living brain several models might be needed to properly address certain questions. In this review, we aim to provide an overview of neural cell and tissue culture, reflecting on historic methodological milestones and providing a brief overview of the state-of-the-art. We additionally present an example of an effective model system pipeline, composed of dissociated mouse cultures, organotypics, acute mouse brain slices, and acute human brain slices, in that order. The sequential use of these four model systems allows a balance and progression from experimental control to human applicability, and provides a meta-model that can help validate basic research findings in a translational setting. We then conclude with a few remarks regarding the necessity of an integrated approach when performing translational and neuropharmacological studies.
Collapse
Affiliation(s)
- Joel Wellbourne-Wood
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland
| | - Jean-Yves Chatton
- Department of Fundamental Neurosciences, University of Lausanne, 1005 Lausanne, Switzerland
| |
Collapse
|
19
|
Affiliation(s)
- Ryo Yamasaki
- Department of Neurology; Neurological Institute; Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| |
Collapse
|
20
|
Wu XL, Ma DM, Zhang W, Zhou JS, Huo YW, Lu M, Tang FR. Cx36 in the mouse hippocampus during and after pilocarpine-induced status epilepticus. Epilepsy Res 2018; 141:64-72. [PMID: 29476948 DOI: 10.1016/j.eplepsyres.2018.02.007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Revised: 01/24/2018] [Accepted: 02/13/2018] [Indexed: 12/24/2022]
Abstract
Gap junctions play an important role in the synchronization activity of coupled cells. Hippocampal inhibitory interneurons are involved in epileptogenesis and seizure activity, and express gap junction protein connexin (Cx) 36. Cx36 is also localized in the axons (mossy fibers) of granule cells in the dentate gyrus. While it has been documented that Cx36 is involved in epileptogenesis, there are still controversies regarding the expression levels of Cx36 at different developmental stages of human and animal models of epileptogenesis. In this study, the expression of Cx36 was investigated in the mouse hippocampus at 1 h, 4 h during pilocarpine-induced status epilepticus (PISE) and 1 week, 2 months after PISE. We found that Cx36 was down-regulated in neurons at different time points during and after PISE, whereas it was increased significantly in the stratum lucidum of CA3 area at 2 months after PISE. Double immunofluorescence indicated that Cx36 was localized in parvalbumin (PV) immunopositive interneuron in CA1 area and in mossy fibers and their terminals in the stratum lucidum of CA3 area. It suggests that decreased expression of Cx36 in interneurons may be related to less effective inhibitory control of excitatory activity of hippocampal principal neurons. However, the increased Cx36 immunopositive product in mossy fibers at the chronic stage after PISE may enhance the contacts between granule cells in the dentate gyrus and pyramidal neurons in CA3 area. The two different changes of Cx36 may be implicated in the epileptogenesis.
Collapse
Affiliation(s)
- X L Wu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an, Shaanxi 710061, China
| | - D M Ma
- Department of Thoracic Surgery, The Ninth Affiliated Hospital of Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710054, China
| | - W Zhang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an, Shaanxi 710061, China
| | - J S Zhou
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an, Shaanxi 710061, China
| | - Y W Huo
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an, Shaanxi 710061, China
| | - M Lu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education of China, Xi'an, Shaanxi 710061, China
| | - F R Tang
- Radiation Physiology Laboratory, Singapore Nuclear Research and Safety Initiative (SNRSI), National University of Singapore, 1 CREATE Way #04-01, CREATE Tower 138602, Singapore.
| |
Collapse
|
21
|
Çalışkan G, Stork O. Hippocampal network oscillations as mediators of behavioural metaplasticity: Insights from emotional learning. Neurobiol Learn Mem 2018; 154:37-53. [PMID: 29476822 DOI: 10.1016/j.nlm.2018.02.022] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2017] [Revised: 02/13/2018] [Accepted: 02/19/2018] [Indexed: 01/15/2023]
Abstract
Behavioural metaplasticity is evident in experience-dependent changes of network activity patterns in neuronal circuits that connect the hippocampus, amygdala and medial prefrontal cortex. These limbic regions are key structures of a brain-wide neural network that translates emotionally salient events into persistent and vivid memories. Communication in this network by-and-large depends on behavioural state-dependent rhythmic network activity patterns that are typically generated and/or relayed via the hippocampus. In fact, specific hippocampal network oscillations have been implicated to the acquisition, consolidation and retrieval, as well as the reconsolidation and extinction of emotional memories. The hippocampal circuits that contribute to these network activities, at the same time, are subject to both Hebbian and non-Hebbian forms of plasticity during memory formation. Further, it has become evident that adaptive changes in the hippocampus-dependent network activity patterns provide an important means of adjusting synaptic plasticity. We here summarise our current knowledge on how these processes in the hippocampus in interaction with amygdala and medial prefrontal cortex mediate the formation and persistence of emotional memories.
Collapse
Affiliation(s)
- Gürsel Çalışkan
- Department of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany.
| | - Oliver Stork
- Department of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg, Leipziger Str. 44, 39120 Magdeburg, Germany; Center for Behavioral Brain Sciences, Universitätsplatz 2, 39106 Magdeburg, Germany
| |
Collapse
|
22
|
Nagy JI, Pereda AE, Rash JE. Electrical synapses in mammalian CNS: Past eras, present focus and future directions. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2018; 1860:102-123. [PMID: 28577972 PMCID: PMC5705454 DOI: 10.1016/j.bbamem.2017.05.019] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/26/2017] [Accepted: 05/27/2017] [Indexed: 12/19/2022]
Abstract
Gap junctions provide the basis for electrical synapses between neurons. Early studies in well-defined circuits in lower vertebrates laid the foundation for understanding various properties conferred by electrical synaptic transmission. Knowledge surrounding electrical synapses in mammalian systems unfolded first with evidence indicating the presence of gap junctions between neurons in various brain regions, but with little appreciation of their functional roles. Beginning at about the turn of this century, new approaches were applied to scrutinize electrical synapses, revealing the prevalence of neuronal gap junctions, the connexin protein composition of many of those junctions, and the myriad diverse neural systems in which they occur in the mammalian CNS. Subsequent progress indicated that electrical synapses constitute key elements in synaptic circuitry, govern the collective activity of ensembles of electrically coupled neurons, and in part orchestrate the synchronized neuronal network activity and rhythmic oscillations that underlie fundamental integrative processes. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- James I Nagy
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - Alberto E Pereda
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, 10461, United States
| | - John E Rash
- Department of Biomedical Sciences, and Program in Molecular, Cellular and Integrative Neurosciences, Colorado State University, Fort Collins, CO 80523, United States
| |
Collapse
|
23
|
Lapato AS, Tiwari-Woodruff SK. Connexins and pannexins: At the junction of neuro-glial homeostasis & disease. J Neurosci Res 2017; 96:31-44. [PMID: 28580666 DOI: 10.1002/jnr.24088] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/08/2017] [Accepted: 05/01/2017] [Indexed: 12/15/2022]
Abstract
In the central nervous system (CNS), connexin (Cx)s and pannexin (Panx)s are an integral component of homeostatic neuronal excitability and synaptic plasticity. Neuronal Cx gap junctions form electrical synapses across biochemically similar GABAergic networks, allowing rapid and extensive inhibition in response to principle neuron excitation. Glial Cx gap junctions link astrocytes and oligodendrocytes in the pan-glial network that is responsible for removing excitotoxic ions and metabolites. In addition, glial gap junctions help constrain excessive excitatory activity in neurons and facilitate astrocyte Ca2+ slow wave propagation. Panxs do not form gap junctions in vivo, but Panx hemichannels participate in autocrine and paracrine gliotransmission, alongside Cx hemichannels. ATP and other gliotransmitters released by Cx and Panx hemichannels maintain physiologic glutamatergic tone by strengthening synapses and mitigating aberrant high frequency bursting. Under pathological depolarizing and inflammatory conditions, gap junctions and hemichannels become dysregulated, resulting in excessive neuronal firing and seizure. In this review, we present known contributions of Cxs and Panxs to physiologic neuronal excitation and explore how the disruption of gap junctions and hemichannels lead to abnormal glutamatergic transmission, purinergic signaling, and seizures.
Collapse
Affiliation(s)
- Andrew S Lapato
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, 92521.,Center for Glial-Neuronal Interactions, University of California Riverside, Riverside, CA, 92521
| | - Seema K Tiwari-Woodruff
- Division of Biomedical Sciences, School of Medicine, University of California Riverside, Riverside, CA, 92521.,Center for Glial-Neuronal Interactions, University of California Riverside, Riverside, CA, 92521.,Neuroscience Graduate Program, University of California Riverside, Riverside, CA, 92521
| |
Collapse
|
24
|
Beheshti S, Zeinali R, Esmaeili A. Rapid upregulation of the hippocampal connexins 36 and 45 mRNA levels during memory consolidation. Behav Brain Res 2017; 320:85-90. [PMID: 27913256 DOI: 10.1016/j.bbr.2016.11.048] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 11/25/2016] [Accepted: 11/28/2016] [Indexed: 10/20/2022]
Abstract
Gap junction channels are implicated in learning and memory process. However, their role on each of the particular stages of memory formation has been studied less. In this study, the time profile of the expression levels of hippocampal connexins 36 and 45 (Cx36 and Cx45) mRNAs was measured during memory consolidation, in a passive avoidance paradigm. Totally 30 adult male rats were distributed into 5 groups of each 6. At different times profiles (30min, 3, 6 and 24h) following training, rats were decapitated and their hippocampi were immediately removed and frozen in liquid nitrogen. Total RNA was extracted and cDNA was synthesized, using oligo-dt primers. A quantitative real-time PCR was used to measure the levels of each of Cx36 and Cx45 mRNAs. Both connexins showed a rapid upregulation (30min) at the transcriptional level, which declined in later times and reached to the control level at 24h. The rapid up-regulation of Cx36 and Cx45 mRNAs might be accompanied with increasing intercellular coupling via gap junction channels and neuronal oscillatory activities required for memory consolidation. The results highlight the role of gap junctional coupling between hippocampal neurons during memory consolidation in the physiological conditions.
Collapse
Affiliation(s)
- Siamak Beheshti
- Division of Animal Sciences, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran.
| | - Reyhaneh Zeinali
- Division of Animal Sciences, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Abolghasem Esmaeili
- Division of Cellular and Molecular Biology, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| |
Collapse
|
25
|
Manjarrez-Marmolejo J, Franco-Pérez J. Gap Junction Blockers: An Overview of their Effects on Induced Seizures in Animal Models. Curr Neuropharmacol 2017; 14:759-71. [PMID: 27262601 PMCID: PMC5050393 DOI: 10.2174/1570159x14666160603115942] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Revised: 02/26/2016] [Accepted: 04/21/2016] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Gap junctions are clusters of intercellular channels allowing the bidirectional pass of ions directly into the cytoplasm of adjacent cells. Electrical coupling mediated by gap junctions plays a role in the generation of highly synchronized electrical activity. The hypersynchronous neuronal activity is a distinctive characteristic of convulsive events. Therefore, it has been postulated that enhanced gap junctional communication is an underlying mechanism involved in the generation and maintenance of seizures. There are some chemical compounds characterized as gap junction blockers because of their ability to disrupt the gap junctional intercellular communication. OBJECTIVE Hence, the aim of this review is to analyze the available data concerning the effects of gap junction blockers specifically in seizure models. RESULTS Carbenoxolone, quinine, mefloquine, quinidine, anandamide, oleamide, heptanol, octanol, meclofenamic acid, niflumic acid, flufenamic acid, glycyrrhetinic acid and retinoic acid have all been evaluated on animal seizure models. In vitro, these compounds share anticonvulsant effects typically characterized by the reduction of both amplitude and frequency of the epileptiform activity induced in brain slices. In vivo, gap junction blockers modify the behavioral parameters related to seizures induced by 4-aminopyridine, pentylenetetrazole, pilocarpine, penicillin and maximal electroshock. CONCLUSION Although more studies are still required, these molecules could be a promising avenue in the search for new pharmaceutical alternatives for the treatment of epilepsy.
Collapse
Affiliation(s)
| | - Javier Franco-Pérez
- Laboratory of Physiology of Reticular Formation, National Institute of Neurology and Neurosurgery, M.V.S. Insurgentes Sur 3877, Col. La Fama, C.P. 14269, Mexico D.F., Mexico
| |
Collapse
|
26
|
Etemadi F, Sayyah M, Pourbadie HG, Babapour V. Facilitation of Hippocampal Kindling and Exacerbation of Kindled Seizures by Intra-CA1 Injection of Quinine: A Possible Role of Cx36 Gap Junctions. IRANIAN BIOMEDICAL JOURNAL 2016; 20:266-72. [PMID: 27108691 PMCID: PMC5075139 DOI: 10.22045/ibj.2016.03] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/14/2015] [Accepted: 11/23/2015] [Indexed: 11/20/2022]
Abstract
BACKGROUND GABAergic interneurons in the hippocampal CA1 area are mutually communicated by gap junctions (GJs) composed of connexin36 (Cx36). We examined the role of Cx36 in CA1 in manifestation of kindled seizures and hippocampal kindling in rats. METHODS Quinine, as the specific blocker of Cx36, was injected into CA1, and kindled seizures severity was examined 10 min afterward. Moreover, quinine was injected into CA1 once daily, and the rate of CA1 kindling was recorded. RESULTS Quinine 0.5 and 1 mM caused 2- and 3.5-fold increase in the duration of total seizure behavior and generalized the seizures. Primary and secondary afterdischarges (AD) were also significantly increased. Quinine 0.1 mM augmented the rate of kindling and the growth of secondary AD. CONCLUSION Cx36 GJs in CA1 are the main components of hippocampal inhibitory circuit. Any interruption in this path by pathologic or physical damages can trigger hippocampal hyperexcitability and facilitate epileptogenesis.
Collapse
Affiliation(s)
- Fatemeh Etemadi
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
- Department of Physiology, Faculty of Veterinary Medicine, Tehran University, Tehran, Iran
| | - Mohammad Sayyah
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Vahab Babapour
- Department of Physiology, Faculty of Veterinary Medicine, Tehran University, Tehran, Iran
| |
Collapse
|
27
|
Ul Haq R, Anderson M, Liotta A, Shafiq M, Sherkheli MA, Heinemann U. Pretreatment with β-adrenergic receptor agonists facilitates induction of LTP and sharp wave ripple complexes in rodent hippocampus. Hippocampus 2016; 26:1486-1492. [PMID: 27699900 DOI: 10.1002/hipo.22665] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 09/14/2016] [Accepted: 09/30/2016] [Indexed: 12/21/2022]
Abstract
Norepinephrine, is involved in the enhancement of learning and memory formation by regulating synaptic mechanisms through its ability to activate pre- and post-synaptic adrenergic receptors. Here we show that β-agonists of norepinephrine facilitate the induction of both associational LTP and sharp wave ripples (SPW-Rs) in acute slices of rat hippocampus in area CA3. Surprisingly, this facilitating effect persists when slices are only pretreated with β-receptor agonists followed by wash out and application of the unspecific β-adrenoreceptor (βAR) antagonist propranolol. During application of βAR agonists repeated stimulation resulted in facilitated induction of SPW-Rs. Since SPW-Rs are thought to be involved in memory replay we studied the effects of βAR-agonists on spontaneous SPW-Rs in murine hippocampus and found that amplitude and incidence of SPW-Rs increased. These effects involve cyclic-AMP and the activation of protein kinase A and suggest a supportive role in memory consolidation. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Rizwan Ul Haq
- Neuroscience Research Center, Charite Universitatsmedizin Berlin, Germany.,Department of Pharmacy, Abbottabad University of Science & Technology, Abbottabad, Pakistan
| | - Marlene Anderson
- Neuroscience Research Center, Charite Universitatsmedizin Berlin, Germany
| | - Agustin Liotta
- Neuroscience Research Center, Charite Universitatsmedizin Berlin, Germany
| | - Maria Shafiq
- Department of Pharmacy, Abbottabad University of Science & Technology, Abbottabad, Pakistan
| | | | - Uwe Heinemann
- Neuroscience Research Center, Charite Universitatsmedizin Berlin, Germany
| |
Collapse
|
28
|
Kay CWP, Ursu D, Sher E, King AE. The role of Cx36 and Cx43 in 4-aminopyridine-induced rhythmic activity in the spinal nociceptive dorsal horn: an electrophysiological study in vitro. Physiol Rep 2016; 4:e12852. [PMID: 27462070 PMCID: PMC4962069 DOI: 10.14814/phy2.12852] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 06/10/2016] [Accepted: 06/13/2016] [Indexed: 01/09/2023] Open
Abstract
Connexin (Cx) proteins and gap junctions support the formation of neuronal and glial syncytia that are linked to different forms of rhythmic firing and oscillatory activity in the CNS. In this study, quantitative reverse transcription polymerase chain reaction (RT-qPCR) was used to profile developmental expression of two specific Cx proteins, namely glial Cx43 and neuronal Cx36, in postnatal lumbar spinal cord aged 4, 7, and 14 days. Extracellular electrophysiology was used to determine the contribution of Cx36 and Cx43 to a previously described form of 4-aminopyridine (4-AP)-induced 4-12 Hz rhythmic activity within substantia gelatinosa (SG) of rat neonatal dorsal horn (DH) in vitro. The involvement of Cx36 and Cx43 was probed pharmacologically using quinine, a specific uncoupler of Cx36 and the mimetic peptide blocker Gap 26 which targets Cx43. After establishment of 4-12 Hz rhythmic activity by 4-AP (25 μmol/L), coapplication of quinine (250 μmol/L) reduced 4-AP-induced 4-12 Hz rhythmic activity (P < 0.05). Preincubation of spinal cord slices with Gap 26 (100 μmol/L), compromised the level of 4-AP-induced 4-12 Hz rhythmic activity in comparison with control slices preincubated in ACSF alone (P < 0.05). Conversely, the nonselective gap junction "opener" trimethylamine (TMA) enhanced 4-12 Hz rhythmic behavior (P < 0.05), further supporting a role for Cx proteins and gap junctions. These data have defined a physiological role for Cx36 and Cx43 in rhythmic firing in SG, a key nociceptive processing area of DH. The significance of these data in the context of pain and Cx proteins as a future analgesic drug target requires further study.
Collapse
Affiliation(s)
- Christopher W P Kay
- School of Biomedical Sciences, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Daniel Ursu
- Eli Lilly & Co., Lilly Research Centre Erl Wood Manor, Windlesham, Surrey, GU20 6PH, United Kingdom
| | - Emanuele Sher
- Eli Lilly & Co., Lilly Research Centre Erl Wood Manor, Windlesham, Surrey, GU20 6PH, United Kingdom
| | - Anne E King
- School of Biomedical Sciences, University of Leeds, Leeds, LS2 9JT, United Kingdom
| |
Collapse
|
29
|
Ramani M, Mylvaganam S, Krawczyk M, Wang L, Zoidl C, Brien J, Reynolds JN, Kapur B, Poulter MO, Zoidl G, Carlen PL. Differential expression of astrocytic connexins in a mouse model of prenatal alcohol exposure. Neurobiol Dis 2016; 91:83-93. [DOI: 10.1016/j.nbd.2016.02.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 01/11/2016] [Accepted: 02/29/2016] [Indexed: 11/24/2022] Open
|
30
|
Buzsáki G. Hippocampal sharp wave-ripple: A cognitive biomarker for episodic memory and planning. Hippocampus 2015; 25:1073-188. [PMID: 26135716 PMCID: PMC4648295 DOI: 10.1002/hipo.22488] [Citation(s) in RCA: 1049] [Impact Index Per Article: 104.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 06/30/2015] [Indexed: 12/23/2022]
Abstract
Sharp wave ripples (SPW-Rs) represent the most synchronous population pattern in the mammalian brain. Their excitatory output affects a wide area of the cortex and several subcortical nuclei. SPW-Rs occur during "off-line" states of the brain, associated with consummatory behaviors and non-REM sleep, and are influenced by numerous neurotransmitters and neuromodulators. They arise from the excitatory recurrent system of the CA3 region and the SPW-induced excitation brings about a fast network oscillation (ripple) in CA1. The spike content of SPW-Rs is temporally and spatially coordinated by a consortium of interneurons to replay fragments of waking neuronal sequences in a compressed format. SPW-Rs assist in transferring this compressed hippocampal representation to distributed circuits to support memory consolidation; selective disruption of SPW-Rs interferes with memory. Recently acquired and pre-existing information are combined during SPW-R replay to influence decisions, plan actions and, potentially, allow for creative thoughts. In addition to the widely studied contribution to memory, SPW-Rs may also affect endocrine function via activation of hypothalamic circuits. Alteration of the physiological mechanisms supporting SPW-Rs leads to their pathological conversion, "p-ripples," which are a marker of epileptogenic tissue and can be observed in rodent models of schizophrenia and Alzheimer's Disease. Mechanisms for SPW-R genesis and function are discussed in this review.
Collapse
Affiliation(s)
- György Buzsáki
- The Neuroscience Institute, School of Medicine and Center for Neural Science, New York University, New York, New York
| |
Collapse
|
31
|
Abstract
The age-dependent progression of tau pathology is a major characteristic of tauopathies, including Alzheimer's disease (AD), and plays an important role in the behavioral phenotypes of AD, including memory deficits. Despite extensive molecular and cellular studies on tau pathology, it remains to be determined how it alters the neural circuit functions underlying learning and memory in vivo. In rTg4510 mice, a Tau-P301L tauopathy model, hippocampal place fields that support spatial memories are abnormal at old age (7-9 months) when tau tangles and neurodegeneration are extensive. However, it is unclear how the abnormality in the hippocampal circuit function arises and progresses with the age-dependent progression of tau pathology. Here we show that in young (2-4 months of age) rTg4510 mice, place fields of hippocampal CA1 cells are largely normal, with only subtle differences from those of age-matched wild-type control mice. Second, high-frequency ripple oscillations of local field potentials in the hippocampal CA1 area are significantly reduced in young rTg4510 mice, and even further deteriorated in old rTg4510 mice. The ripple reduction is associated with less bursty firing and altered synchrony of CA1 cells. Together, the data indicate that deficits in ripples and neuronal synchronization occur before overt deficits in place fields in these mice. The results reveal a tau-pathology-induced progression of hippocampal functional changes in vivo.
Collapse
|
32
|
Decrock E, De Bock M, Wang N, Bultynck G, Giaume C, Naus CC, Green CR, Leybaert L. Connexin and pannexin signaling pathways, an architectural blueprint for CNS physiology and pathology? Cell Mol Life Sci 2015; 72:2823-51. [PMID: 26118660 PMCID: PMC11113968 DOI: 10.1007/s00018-015-1962-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 06/11/2015] [Indexed: 02/06/2023]
Abstract
The central nervous system (CNS) is composed of a highly heterogeneous population of cells. Dynamic interactions between different compartments (neuronal, glial, and vascular systems) drive CNS function and allow to integrate and process information as well as to respond accordingly. Communication within this functional unit, coined the neuro-glio-vascular unit (NGVU), typically relies on two main mechanisms: direct cell-cell coupling via gap junction channels (GJCs) and paracrine communication via the extracellular compartment, two routes to which channels composed of transmembrane connexin (Cx) or pannexin (Panx) proteins can contribute. Multiple isoforms of both protein families are present in the CNS and each CNS cell type is characterized by a unique Cx/Panx portfolio. Over the last two decades, research has uncovered a multilevel platform via which Cxs and Panxs can influence different cellular functions within a tissue: (1) Cx GJCs enable a direct cell-cell communication of small molecules, (2) Cx hemichannels and Panx channels can contribute to autocrine/paracrine signaling pathways, and (3) different structural domains of these proteins allow for channel-independent functions, such as cell-cell adhesion, interactions with the cytoskeleton, and the activation of intracellular signaling pathways. In this paper, we discuss current knowledge on their multifaceted contribution to brain development and to specific processes in the NGVU, including synaptic transmission and plasticity, glial signaling, vasomotor control, and blood-brain barrier integrity in the mature CNS. By highlighting both physiological and pathological conditions, it becomes evident that Cxs and Panxs can play a dual role in the CNS and that an accurate fine-tuning of each signaling mechanism is crucial for normal CNS physiology.
Collapse
Affiliation(s)
- Elke Decrock
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Marijke De Bock
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Nan Wang
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| | - Geert Bultynck
- Laboratory of Molecular and Cellular Signalling, Department of Cellular and Molecular Medicine, KU Leuven, Louvain, Belgium
| | - Christian Giaume
- Collège de France, Center for Interdisciplinary Research in Biology (CIRB)/Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7241/Institut National de la Santé et de la Recherche Médicale U1050, 75231 Paris Cedex 05, France
- University Pierre et Marie
Curie, ED, N°158, 75005 Paris, France
- MEMOLIFE Laboratory of Excellence and Paris Science Lettre Research University, 75005 Paris, France
| | - Christian C. Naus
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Colin R. Green
- Department of Ophthalmology, The University of Auckland, Auckland, New Zealand
| | - Luc Leybaert
- Physiology Group, Department of Basic Medical Sciences, Ghent University, De Pintelaan 185 (Block B, 3rd floor), 9000 Ghent, Belgium
| |
Collapse
|
33
|
Butler JL, Paulsen O. Hippocampal network oscillations - recent insights from in vitro experiments. Curr Opin Neurobiol 2015; 31:40-4. [PMID: 25137641 DOI: 10.1016/j.conb.2014.07.025] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Revised: 07/26/2014] [Accepted: 07/30/2014] [Indexed: 01/07/2023]
Abstract
Network oscillations are present throughout the mammalian brain. They are important for certain cognitive functions, such as learning and memory. The hippocampus exhibits prominent oscillations similar to those seen in other parts of the cortex. Due to its highly organised lamellar structure, ex vivo and in vitro preparations from the hippocampus have provided experimental models within which to study network oscillations. As such, experiments in hippocampal slices continue to progress our understanding about both the mechanisms and functions of cortical network oscillations. Here, advances from the past two years are summarised, and the current state of the field discussed.
Collapse
Affiliation(s)
- James L Butler
- Department of Physiology, Development and Neuroscience, University of Cambridge, Physiological Laboratory, Downing Street, Cambridge CB2 3EG, United Kingdom
| | - Ole Paulsen
- Department of Physiology, Development and Neuroscience, University of Cambridge, Physiological Laboratory, Downing Street, Cambridge CB2 3EG, United Kingdom.
| |
Collapse
|
34
|
Çalışkan G, Albrecht A, Hollnagel JO, Rösler A, Richter-Levin G, Heinemann U, Stork O. Long-term changes in the CA3 associative network of fear-conditioned mice. Stress 2015; 18:188-97. [PMID: 25556979 DOI: 10.3109/10253890.2015.1004628] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The CA3 associative network plays a critical role in the generation of network activity patterns related to emotional state and fear memory. We investigated long-term changes in the corticosterone (CORT)-sensitive function of this network following fear conditioning and fear memory reactivation. In acute slice preparations from mice trained in either condition, the ratio of orthodromic population spike (PS) to antidromic PS was reduced compared to unconditioned animals, indicating a decrease in efficacy of neuronal coupling within the associative CA3 network. However, spontaneous sharp wave-ripples (SW-R), which are thought to arise from this network, remained unaltered. Following CORT application, we observed an increase in orthodromic PS and a normalization to control levels of their ratio to antidromic PS, while SW-R increased in slices of fear conditioned and fear reactivated mice, but not in slices of unconditioned controls. Together with our previous observations of altered hippocampal gamma activity under these learning paradigms, these data suggest that fear conditioning and fear reactivation lastingly alters the CORT-sensitive configuration of different network activity patterns generated by the CA3 associational network. Observed changes in the mRNA expression of receptors for glutamate, GABA and cannabinoids in the stratum pyramidale of area CA3 may provide a molecular mechanism for these adaptive changes.
Collapse
MESH Headings
- Animals
- Anti-Inflammatory Agents/pharmacology
- CA3 Region, Hippocampal/drug effects
- CA3 Region, Hippocampal/metabolism
- CA3 Region, Hippocampal/physiology
- Conditioning, Psychological/physiology
- Corticosterone/pharmacology
- Emotions
- Fear
- Hippocampus/drug effects
- Hippocampus/physiology
- Male
- Memory/physiology
- Mice
- Multiplex Polymerase Chain Reaction
- Nerve Tissue Proteins/genetics
- Neural Pathways/physiology
- Neurons/drug effects
- Neurons/physiology
- Patch-Clamp Techniques
- RNA, Messenger/drug effects
- RNA, Messenger/metabolism
- Receptor, Cannabinoid, CB1/genetics
- Receptors, AMPA/genetics
- Receptors, GABA-A/genetics
- Receptors, N-Methyl-D-Aspartate/genetics
Collapse
Affiliation(s)
- Gürsel Çalışkan
- Department of Genetics & Molecular Neurobiology, Institute of Biology, Otto-von-Guericke-University Magdeburg , Magdeburg , Germany
| | | | | | | | | | | | | |
Collapse
|
35
|
Cepeda C, Chang JW, Owens GC, Huynh MN, Chen JY, Tran C, Vinters HV, Levine MS, Mathern GW. In Rasmussen encephalitis, hemichannels associated with microglial activation are linked to cortical pyramidal neuron coupling: a possible mechanism for cellular hyperexcitability. CNS Neurosci Ther 2014; 21:152-63. [PMID: 25438677 DOI: 10.1111/cns.12352] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Revised: 10/16/2014] [Accepted: 10/20/2014] [Indexed: 12/21/2022] Open
Abstract
AIMS Rasmussen encephalitis (RE) is a rare but devastating condition, mainly in children, characterized by sustained brain inflammation, atrophy of one cerebral hemisphere, epilepsy, and progressive cognitive deterioration. The etiology of RE-induced seizures associated with the inflammatory process remains unknown. METHODS Cortical tissue samples from children undergoing surgical resections for the treatment of RE (n = 16) and non-RE (n = 12) were compared using electrophysiological, morphological, and immunohistochemical techniques to examine neuronal properties and the relationship with microglial activation using the specific microglia/macrophage calcium-binding protein, IBA1 in conjunction with connexins and pannexin expression. RESULTS Compared with non-RE cases, pyramidal neurons from RE cases displayed increased cell capacitance and reduced input resistance. However, neuronal somatic areas were not increased in size. Instead, intracellular injection of biocytin led to increased dye coupling between neurons from RE cases. By Western blot, expression of IBA1 and pannexin was increased while connexin 32 was decreased in RE cases compared with non-RE cases. IBA1 immunostaining overlapped with pannexin and connexin 36 in RE cases. CONCLUSIONS In RE, these results support the notion that a possible mechanism for cellular hyperexcitability may be related to increased intercellular coupling from pannexin linked to increased microglial activation. Such findings suggest that a possible antiseizure treatment for RE may involve the use of gap junction blockers.
Collapse
Affiliation(s)
- Carlos Cepeda
- Intellectual and Developmental Disabilities Research Center, Brain Research Institute, Los Angeles, CA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Reciprocal regulation of epileptiform neuronal oscillations and electrical synapses in the rat hippocampus. PLoS One 2014; 9:e109149. [PMID: 25299405 PMCID: PMC4192321 DOI: 10.1371/journal.pone.0109149] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Accepted: 09/01/2014] [Indexed: 11/19/2022] Open
Abstract
Gap junction (GJ) channels have been recognized as an important mechanism for synchronizing neuronal networks. Herein, we investigated the participation of GJ channels in the pilocarpine-induced status epilepticus (SE) by analyzing electrophysiological activity following the blockade of connexins (Cx)-mediated communication. In addition, we examined the regulation of gene expression, protein levels, phosphorylation profile and distribution of neuronal Cx36, Cx45 and glial Cx43 in the rat hippocampus during the acute and latent periods. Electrophysiological recordings revealed that the GJ blockade anticipates the occurrence of low voltage oscillations and promotes a marked reduction of power in all analyzed frequencies.Cx36 gene expression and protein levels remained stable in acute and latent periods, whereas upregulation of Cx45 gene expression and protein redistribution were detected in the latent period. We also observed upregulation of Cx43 mRNA levels followed by changes in the phosphorylation profile and protein accumulation. Taken together, our results indisputably revealed that GJ communication participates in the epileptiform activity induced by pilocarpine. Moreover, considering that specific Cxs undergo alterations through acute and latent periods, this study indicates that the control of GJ communication may represent a focus in reliable anti-epileptogenic strategies.
Collapse
|
37
|
Decreased fast ripples in the hippocampus of rats with spontaneous recurrent seizures treated with carbenoxolone and quinine. BIOMED RESEARCH INTERNATIONAL 2014; 2014:282490. [PMID: 25276773 PMCID: PMC4168142 DOI: 10.1155/2014/282490] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Accepted: 08/14/2014] [Indexed: 01/14/2023]
Abstract
Background. In models of temporal lobe epilepsy and in patients with this pathology, high frequency oscillations called fast ripples (FRs, 250–600 Hz) can be observed. FRs are considered potential biomarkers for epilepsy and, in the light of many in vitro and in silico studies, we thought that electrical synapses mediated by gap junctions might possibly modulate FRs in vivo. Methods. Animals with spontaneous recurrent seizures induced by pilocarpine administration were implanted with movable microelectrodes in the right anterior and posterior hippocampus to evaluate the effects of gap junction blockers administered in the entorhinal cortex. The effects of carbenoxolone (50 nmoles) and quinine (35 pmoles) on the mean number of spontaneous FR events (occurrence of FRs), as well as on the mean number of oscillation cycles per FR event and their frequency, were assessed using a specific algorithm to analyze FRs in intracranial EEG recordings. Results. We found that these gap junction blockers decreased the mean number of FRs and the mean number of oscillation cycles per FR event in the hippocampus, both during and at different times after carbenoxolone and quinine administration. Conclusion. These data suggest that FRs may be modulated by gap junctions, although additional experiments in vivo will be necessary to determine the precise role of gap junctions in this pathological activity associated with epileptogenesis.
Collapse
|
38
|
Intracellular activities related to in vitro hippocampal sharp waves are altered in CA3 pyramidal neurons of aged mice. Neuroscience 2014; 277:474-85. [PMID: 25088916 DOI: 10.1016/j.neuroscience.2014.07.048] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 07/09/2014] [Accepted: 07/09/2014] [Indexed: 01/11/2023]
Abstract
Pyramidal neurons in the hippocampal CA3 area interconnect intensively via recurrent axonal collaterals, and such CA3-to-CA3 recurrent circuitry plays important roles in the generation of hippocampal network activities. In particular, the CA3 circuitry is able to generate spontaneous sharp waves (SPWs) when examined in vitro. These in vitro SPWs are thought to result from the network activity of GABAergic inhibitory interneurons as SPW-correlating intracellular activities are featured with strong IPSPs in pyramidal neurons and EPSPs or spikes in GABAergic interneurons. In view of accumulating evidence indicating a decrease in subgroups of hippocampal GABAergic interneurons in aged animals, we test the hypothesis that the intracellular activities related to in vitro SPWs are altered in CA3 pyramidal neurons of aged mice. Hippocampal slices were prepared from adult and aged C57 black mice (ages 3-6 and 24-28months respectively). Population and single-cell activities were examined via extracellular and whole-cell patch-clamp recordings. CA3 SPW frequencies were not significantly different between the slices of adult and aged mice but SPW-correlating intracellular activities featured weaker IPSC components in aged CA3 pyramidal neurons compared to adult neurons. It was unlikely that this latter phenomenon was due to general impairments of GABAergic synapses in the aged CA3 circuitry as evoked IPSC responses and pharmacologically isolated IPSCs were observed in aged CA3 pyramidal neurons. In addition, aged CA3 pyramidal neurons displayed more positive resting potentials and had a higher propensity of burst firing than adult neurons. We postulate that alterations of GABAergic network activity may explain the reduced IPCS contributions to in vitro SPWs in aged CA3 pyramidal neurons. Overall, our present observations are supportive of the notion that excitability of hippocampal CA3 circuitry is increased in aged mice.
Collapse
|
39
|
Hollnagel JO, Maslarova A, Haq RU, Heinemann U. GABAB receptor dependent modulation of sharp wave-ripple complexes in the rat hippocampus in vitro. Neurosci Lett 2014; 574:15-20. [PMID: 24796812 DOI: 10.1016/j.neulet.2014.04.045] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 04/08/2014] [Accepted: 04/26/2014] [Indexed: 10/25/2022]
Abstract
Sharp wave-ripple complexes (SPW-R) are observed in vivo during resting immobility, consummatory behavior and during slow wave sleep, and they have been proposed to support memory consolidation. It has been suggested that GABAergic cells play important roles in controlling incidence of sharp waves and of ripple frequency. We report here that the GABAB agonist baclofen reversibly suppresses SPW-R activity in rat hippocampal slices, presumably affecting the strength of neuronal coupling in the associative network of area CA3. The effect is specific as the GABAB receptor antagonist CGP55846 prevents this effect; however, CGP55846 application had no major effect on incidence of SPW-R. Interestingly, repetitive stimulation in the presence of baclofen is able to induce SPW-R activity, which only appears after washout of baclofen. Our findings suggest that GABA levels through activation of GABAB receptors may be involved in the transition from theta-gamma to SPW-R working mode in the hippocampus.
Collapse
Affiliation(s)
- Jan Oliver Hollnagel
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, 14195 Berlin, Germany
| | - Anna Maslarova
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, 14195 Berlin, Germany
| | - Rizwan Ul Haq
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, 14195 Berlin, Germany; Department of Pharmacy, Hazara University, Havelian Campus, Abbottabad 22500, Pakistan
| | - Uwe Heinemann
- Institute of Neurophysiology, Charité-Universitätsmedizin Berlin, 14195 Berlin, Germany; NeuroCure Research Center, Charité-Universitätsmedizin Berlin, 14195 Berlin, Germany.
| |
Collapse
|
40
|
Posłuszny A. The contribution of electrical synapses to field potential oscillations in the hippocampal formation. Front Neural Circuits 2014; 8:32. [PMID: 24772068 PMCID: PMC3982077 DOI: 10.3389/fncir.2014.00032] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 03/18/2014] [Indexed: 11/23/2022] Open
Abstract
Electrical synapses are a type of cellular membrane junction referred to as gap junctions (GJs). They provide a direct way to exchange ions between coupled cells and have been proposed as a structural basis for fast transmission of electrical potentials between neurons in the brain. For this reason GJs have been regarded as an important component within the neuronal networks that underlie synchronous neuronal activity and field potential oscillations. Initially, GJs appeared to play a particularly key role in the generation of high frequency oscillatory patterns in field potentials. In order to assess the scale of neuronal GJs contribution to field potential oscillations in the hippocampal formation, in vivo and in vitro studies are reviewed here. These investigations have shown that blocking the main neuronal GJs, those containing connexin 36 (Cx36-GJs), or knocking out the Cx36 gene affect field potential oscillatory patterns related to awake active behavior (gamma and theta rhythm) but have no effect on high frequency oscillations occurring during silent wake and sleep. Precisely how Cx36-GJs influence population activity of neurons is more complex than previously thought. Analysis of studies on the properties of transmission through GJ channels as well as Cx36-GJs functioning in pairs of coupled neurons provides some explanations of the specific influence of Cx36-GJs on field potential oscillations. It is proposed here that GJ transmission is strongly modulated by the level of neuronal network activity and changing behavioral states. Therefore, contribution of GJs to field potential oscillatory patterns depends on the behavioral state. I propose here a model, based on large body of experimental data gathered in this field by several authors, in which Cx36-GJ transmission especially contributes to oscillations related to active behavior, where it plays a role in filtering and enhancing coherent signals in the network under high-noise conditions. In contrast, oscillations related to silent wake or sleep, especially high frequency oscillations, do not require transmission by neuronal GJs. The reliability of neuronal discharges during those oscillations could be assured by conditions of higher signal-to-noise ratio and some synaptic changes taking place during active behavior.
Collapse
Affiliation(s)
- Anna Posłuszny
- Laboratory of Neuroplasticity, Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences Warsaw, Poland
| |
Collapse
|
41
|
Gigout S, Louvel J, Rinaldi D, Martin B, Pumain R. Thalamocortical relationships and network synchronization in a new genetic model "in mirror" for absence epilepsy. Brain Res 2013; 1525:39-52. [PMID: 23743261 DOI: 10.1016/j.brainres.2013.05.044] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 05/25/2013] [Accepted: 05/27/2013] [Indexed: 11/19/2022]
Abstract
Electroencephalographic generalized spike and wave discharges (SWD), the hallmark of human absence seizures, are generated in thalamocortical networks. However, the potential alterations in these networks in terms of the efficacy of the reciprocal synaptic activities between the cortex and the thalamus are not known in this pathology. Here, the efficacy of these reciprocal connections is assessed in vitro in thalamocortical slices obtained from BS/Orl mice, which is a new genetic model of absence epilepsy. These mice show spontaneous SWD, and their features can be compared to that of BR/Orl mice, which are free of SWD. In addition, since gap junctions may modulate the efficacy of these connections, their implications in pharmacologically-induced epileptiform discharges were studied in the same slices. The thalamus and neocortex were independently stimulated and the electrically-evoked responses in both structures were recorded from the same slice. The synaptic efficacy of thalamocortical and corticothalamic connections were assessed by measuring the dynamic range of synaptic field potential changes in response to increasing stimulation strengths. The connection efficacy was weaker in epileptic mice however, this decrease in efficacy was more pronounced in thalamocortical afferents, thus introducing an imbalance in the reciprocal connections between the cortex and thalamus. However, short-term facilitation of the thalamocortical responses were increased in epileptic mice compared to non-epileptic animals. These features may favor occurrence of rhythmical activities in thalamocortical networks. In addition, carbenoxolone (a gap junction blocker) decreased the cumulative duration of 4-aminopyridine-induced ictal-like activities, with a slower time course in epileptic mice. However, the 4-aminopyridine-induced GABA-dependent negative potentials, which appeared to trigger the ictal-like activities, remained. Our results show that the balance of the reciprocal connections between the thalamus and cortex is altered in favor of the corticothalamic connections in epileptic mice, and suggest that gap junctions mediate a stronger cortical synchronization in this strain.
Collapse
Affiliation(s)
- Sylvain Gigout
- Epilepsie de l'Enfant et Plasticité Cérébrale, INSERM U 663, Paris, France.
| | | | | | | | | |
Collapse
|
42
|
Chang WP, Wu JJS, Shyu BC. Thalamic modulation of cingulate seizure activity via the regulation of gap junctions in mice thalamocingulate slice. PLoS One 2013; 8:e62952. [PMID: 23690968 PMCID: PMC3653920 DOI: 10.1371/journal.pone.0062952] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 03/26/2013] [Indexed: 11/19/2022] Open
Abstract
The thalamus is an important target for deep brain stimulation in the treatment of seizures. However, whether the modulatory effect of thalamic inputs on cortical seizures occurs through the modulation of gap junctions has not been previously studied. Therefore, we tested the effects of different gap junction blockers and couplers in a drug-resistant seizure model and studied the role of gap junctions in the thalamic modulation on cortical seizures. Multielectrode array and calcium imaging were used to record the cortical seizures induced by 4-aminopyridine (250 µM) and bicuculline (5-50 µM) in a novel thalamocingulate slice preparation. Seizure-like activity was significantly attenuated by the pan-gap junction blockers carbenoxolone and octanol and specific neuronal gap junction blocker mefloquine. The gap junction coupler trimethylamine significantly enhanced seizure-like activity. Gap junction blockers did not influence the initial phase of seizure-like activity, but they significantly decreased the amplitude and duration of the maintenance phase. The development of seizures is regulated by extracellular potassium concentration. Carbenoxolone partially restored the amplitude and duration after removing the thalamic inputs. A two-dimensional current source density analysis showed that the sink and source signals shifted to deeper layers after removing the thalamic inputs during the clonic phase. These results indicate that the regulatory mechanism of deep brain stimulation in the thalamus occurs partially though gap junctions.
Collapse
Affiliation(s)
- Wei-Pang Chang
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
| | - José Jiun-Shian Wu
- Institute of Zoology, National Taiwan University, Taipei, Taiwan, Republic of China
| | - Bai-Chuang Shyu
- Graduate Institute of Life Sciences, National Defense Medical Center, Taipei, Taiwan, Republic of China
- Institute of Biomedical Science, Academia Sinica, Taipei, Taiwan, Republic of China
- * E-mail:
| |
Collapse
|
43
|
Belousov AB, Fontes JD. Neuronal gap junctions: making and breaking connections during development and injury. Trends Neurosci 2013; 36:227-36. [PMID: 23237660 PMCID: PMC3609876 DOI: 10.1016/j.tins.2012.11.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 11/02/2012] [Accepted: 11/02/2012] [Indexed: 01/08/2023]
Abstract
In the mammalian central nervous system (CNS), coupling of neurons by gap junctions (i.e., electrical synapses) and the expression of the neuronal gap junction protein, connexin 36 (Cx36), transiently increase during early postnatal development. The levels of both subsequently decline and remain low in the adult, confined to specific subsets of neurons. However, following neuronal injury [such as ischemia, traumatic brain injury (TBI), and epilepsy], the coupling and expression of Cx36 rise. Here we summarize new findings on the mechanisms of regulation of Cx36-containing gap junctions in the developing and mature CNS and following injury. We also review recent studies suggesting various roles for neuronal gap junctions and in particular their role in glutamate-mediated neuronal death.
Collapse
Affiliation(s)
- Andrei B Belousov
- Department of Molecular and Integrative Physiology, University of Kansas Medical Center, Kansas City, KS 66160, USA.
| | | |
Collapse
|
44
|
Simeone TA, Simeone KA, Samson KK, Kim DY, Rho JM. Loss of the Kv1.1 potassium channel promotes pathologic sharp waves and high frequency oscillations in in vitro hippocampal slices. Neurobiol Dis 2013; 54:68-81. [PMID: 23466697 DOI: 10.1016/j.nbd.2013.02.009] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 02/12/2013] [Accepted: 02/22/2013] [Indexed: 01/10/2023] Open
Abstract
In human disease, channelopathies involving functional reduction of the delayed rectifier potassium channel α-subunit Kv1.1 - either by mutation or autoimmune inhibition - result in temporal lobe epilepsy. Kv1.1 is prominently expressed in the axons of the hippocampal tri-synaptic pathway, suggesting its absence will result in widespread effects on normal network oscillatory activity. Here, we performed in vitro extracellular recordings using a multielectrode array to determine the effects of loss of Kv1.1 on spontaneous sharp waves (SPWs) and high frequency oscillations (HFOs). We found that Kcna1-null hippocampi generate SPWs and ripples (80-200Hz bandwidth) with a 50% increased rate of incidence and 50% longer duration, and that epilepsy-associated pathologic HFOs in the fast ripple bandwidth (200-600Hz) are also present. Furthermore, Kcna1-null CA3 has enhanced coupling of excitatory inputs and population spike generation and CA3 principal cells have reduced spike timing reliability. Removing the influence of mossy fiber and perforant path inputs by micro-dissecting the Kcna1-null CA3 region mostly rescued the oscillatory behavior and improved spike timing. We found that Kcna1-null mossy fibers and medial perforant path axons are hyperexcitable and produce greater pre- and post-synaptic responses with reduced paired-pulse ratios suggesting increased neurotransmitter release at these terminals. These findings were recapitulated in wild-type slices exposed to the Kv1.1 inhibitor dendrotoxin-κ. Collectively, these data indicate that loss of Kv1.1 enhances synaptic release in the CA3 region, which reduces spike timing precision of individual neurons leading to disorganization of network oscillatory activity and promotes the emergence of fast ripples.
Collapse
Affiliation(s)
- Timothy A Simeone
- Creighton University, Department of Pharmacology, Omaha, NE 68174, USA.
| | | | | | | | | |
Collapse
|
45
|
Sobayo T, Fine AS, Gunnar E, Kazlauskas C, Nicholls D, Mogul DJ. Synchrony Dynamics Across Brain Structures in Limbic Epilepsy Vary Between Initiation and Termination Phases of Seizures. IEEE Trans Biomed Eng 2013; 60:821-9. [DOI: 10.1109/tbme.2012.2189113] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
46
|
Nagy JI. Evidence for connexin36 localization at hippocampal mossy fiber terminals suggesting mixed chemical/electrical transmission by granule cells. Brain Res 2012; 1487:107-22. [PMID: 22771400 PMCID: PMC3501615 DOI: 10.1016/j.brainres.2012.05.064] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2011] [Revised: 05/14/2012] [Accepted: 05/15/2012] [Indexed: 11/25/2022]
Abstract
Electrical synaptic transmission via gap junctions has become an accepted feature of neuronal communication in the mammalian brain, and occurs often between dendrites of interneurons in major brain structures, including the hippocampus. Electrical and dye-coupling has also been reported to occur between pyramidal cells in the hippocampus, but ultrastructurally-identified gap junctions between these cells have so far eluded detection. Gap junctions can be formed by nerve terminals, where they contribute the electrical component of mixed chemical/electrical synaptic transmission, but mixed synapses have only rarely been described in mammalian CNS. Here, we used immunofluorescence localization of the major gap junction forming protein connexin36 to examine its possible association with hippocampal pyramidal cells. In addition to labeling associated with gap junctions between dendrites of parvalbumin-positive interneurons, a high density of fine, punctate immunolabeling for Cx36, non-overlapping with parvalbumin, was found in subregions of the stratum lucidum in the ventral hippocampus of rat brain. A high percentage of Cx36-positive puncta in the stratum lucidum was localized to mossy fiber terminals, as indicated by co-localization of Cx36-puncta with the mossy terminal marker vesicular glutamate transporter-1, as well as with other proteins that are highly concentrated in, and diagnostic markers of, these terminals. These results suggest that mossy fiber terminals abundantly form mixed chemical/electrical synapses with pyramidal cells, where they may serve as intermediaries for the reported electrical and dye-coupling between ensembles of these principal cells. This article is part of a Special Issue entitled Electrical Synapses.
Collapse
Affiliation(s)
- James I Nagy
- Department of Physiology, Faculty of Medicine, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, Manitoba, Canada R3E 0J9.
| |
Collapse
|
47
|
Eugenin EA, Basilio D, Sáez JC, Orellana JA, Raine CS, Bukauskas F, Bennett MVL, Berman JW. The role of gap junction channels during physiologic and pathologic conditions of the human central nervous system. J Neuroimmune Pharmacol 2012; 7:499-518. [PMID: 22438035 PMCID: PMC3638201 DOI: 10.1007/s11481-012-9352-5] [Citation(s) in RCA: 97] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2012] [Accepted: 02/28/2012] [Indexed: 12/15/2022]
Abstract
Gap junctions (GJs) are expressed in most cell types of the nervous system, including neuronal stem cells, neurons, astrocytes, oligodendrocytes, cells of the blood brain barrier (endothelial cells and astrocytes) and under inflammatory conditions in microglia/macrophages. GJs connect cells by the docking of two hemichannels, one from each cell with each hemichannel being formed by 6 proteins named connexins (Cx). Unapposed hemichannels (uHC) also can be open on the surface of the cells allowing the release of different intracellular factors to the extracellular space. GJs provide a mechanism of cell-to-cell communication between adjacent cells that enables the direct exchange of intracellular messengers, such as calcium, nucleotides, IP(3), and diverse metabolites, as well as electrical signals that ultimately coordinate tissue homeostasis, proliferation, differentiation, metabolism, cell survival and death. Despite their essential functions in physiological conditions, relatively little is known about the role of GJs and uHC in human diseases, especially within the nervous system. The focus of this review is to summarize recent findings related to the role of GJs and uHC in physiologic and pathologic conditions of the central nervous system.
Collapse
Affiliation(s)
- Eliseo A Eugenin
- Department of Pathology, F727, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA.
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
This review summarizes the brain mechanisms controlling sleep and wakefulness. Wakefulness promoting systems cause low-voltage, fast activity in the electroencephalogram (EEG). Multiple interacting neurotransmitter systems in the brain stem, hypothalamus, and basal forebrain converge onto common effector systems in the thalamus and cortex. Sleep results from the inhibition of wake-promoting systems by homeostatic sleep factors such as adenosine and nitric oxide and GABAergic neurons in the preoptic area of the hypothalamus, resulting in large-amplitude, slow EEG oscillations. Local, activity-dependent factors modulate the amplitude and frequency of cortical slow oscillations. Non-rapid-eye-movement (NREM) sleep results in conservation of brain energy and facilitates memory consolidation through the modulation of synaptic weights. Rapid-eye-movement (REM) sleep results from the interaction of brain stem cholinergic, aminergic, and GABAergic neurons which control the activity of glutamatergic reticular formation neurons leading to REM sleep phenomena such as muscle atonia, REMs, dreaming, and cortical activation. Strong activation of limbic regions during REM sleep suggests a role in regulation of emotion. Genetic studies suggest that brain mechanisms controlling waking and NREM sleep are strongly conserved throughout evolution, underscoring their enormous importance for brain function. Sleep disruption interferes with the normal restorative functions of NREM and REM sleep, resulting in disruptions of breathing and cardiovascular function, changes in emotional reactivity, and cognitive impairments in attention, memory, and decision making.
Collapse
Affiliation(s)
- Ritchie E Brown
- Laboratory of Neuroscience, VA Boston Healthcare System and Harvard Medical School, Brockton, Massachusetts 02301, USA
| | | | | | | | | |
Collapse
|
49
|
Taxidis J, Coombes S, Mason R, Owen MR. Modeling sharp wave-ripple complexes through a CA3-CA1 network model with chemical synapses. Hippocampus 2012; 22:995-1017. [PMID: 21452258 DOI: 10.1002/hipo.20930] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/29/2010] [Indexed: 11/08/2022]
Abstract
The hippocampus, and particularly the CA3 and CA1 areas, exhibit a variety of oscillatory rhythms that span frequencies from the slow theta range (4-10 Hz) up to fast ripples (200 Hz). Various computational models of different complexities have been developed in an effort to simulate such population oscillations. Nevertheless the mechanism that underlies the so called Sharp Wave-Ripple complex (SPWR), observed in extracellular recordings in CA1, still remains elusive. We present here, the combination of two simple but realistic models of the rat CA3 and CA1 areas, connected together in a feedforward scheme mimicking Schaffer collaterals. Both network models are computationally simple one-dimensional arrays of excitatory and inhibitory populations interacting only via fast chemical synapses. Connectivity schemes and postsynaptic potentials are based on physiological data, yielding a realistic network topology. The CA3 model exhibits quasi-synchronous population bursts, which give rise to sharp wave-like deep depolarizations in the CA1 dendritic layer accompanied by transient field oscillations at ≈ 150-200 Hz in the somatic layer. The frequency and synchrony of these oscillations is based on interneuronal activity and fast-decaying recurrent inhibition in CA1. Pyramidal cell spikes are sparse and come from a subset of cells receiving stronger than average excitatory input from CA3. The model is shown to accurately reproduce a large number of basic characteristics of SPWRs and yields a new mechanism for the generation of ripples, offering an interpretation to a range of neurophysiological observations, such as the ripple disruption by halothane and the selective firing of pyramidal cells during ripples, which may have implications for memory consolidation during SPWRs.
Collapse
Affiliation(s)
- Jiannis Taxidis
- Division of Applied Mathematics, University of Nottingham, Nottingham, United Kingdom.
| | | | | | | |
Collapse
|
50
|
Munro E, Kopell N. Subthreshold somatic voltage in neocortical pyramidal cells can control whether spikes propagate from the axonal plexus to axon terminals: a model study. J Neurophysiol 2012; 107:2833-52. [PMID: 22378167 DOI: 10.1152/jn.00709.2011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
There is suggestive evidence that pyramidal cell axons in neocortex may be coupled by gap junctions into an "axonal plexus" capable of generating very fast oscillations (VFOs) with frequencies exceeding 80 Hz. It is not obvious, however, how a pyramidal cell in such a network could control its output when action potentials are free to propagate from the axons of other pyramidal cells into its own axon. We address this problem by means of simulations based on three-dimensional reconstructions of pyramidal cells from rat somatosensory cortex. We show that somatic depolarization enables propagation via gap junctions into the initial segment and main axon, while somatic hyperpolarization disables it. We show further that somatic voltage cannot effectively control action potential propagation through gap junctions on minor collaterals; action potentials may therefore propagate freely from such collaterals regardless of somatic voltage. In previous work, VFOs are all but abolished during the hyperpolarization phase of slow oscillations induced by anesthesia in vivo. This finding constrains the density of gap junctions on collaterals in our model and suggests that axonal sprouting due to cortical lesions may result in abnormally high gap junction density on collaterals, leading in turn to excessive VFO activity and hence to epilepsy via kindling.
Collapse
Affiliation(s)
- Erin Munro
- Laboratory for Neural Computation and Adaptation, RIKEN Brain Science Institute, Saitama, Japan.
| | | |
Collapse
|