1
|
Beitchman JA, Krishna G, Bromberg CE, Thomas TC. Effects of isoflurane and urethane anesthetics on glutamate neurotransmission in rat brain using in vivo amperometry. BMC Neurosci 2023; 24:52. [PMID: 37817064 PMCID: PMC10563344 DOI: 10.1186/s12868-023-00822-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 09/19/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND Aspects of glutamate neurotransmission implicated in normal and pathological conditions are predominantly evaluated using in vivo recording paradigms in rats anesthetized with isoflurane or urethane. Urethane and isoflurane anesthesia influence glutamate neurotransmission through different mechanisms; however, real-time outcome measures of potassium chloride (KCl)-evoked glutamate overflow and glutamate clearance kinetics have not been compared within and between regions of the brain. In order to maintain rigor and reproducibility within the literature between the two most common methods of anesthetized in vivo recording of glutamate, we compared glutamate signaling as a function of anesthesia and brain region in the rat strain most used in neuroscience. METHODS In the following experiments, in vivo amperometric recordings of KCl-evoked glutamate overflow and glutamate clearance kinetics (uptake rate and T80) in the cortex, hippocampus, and thalamus were performed using glutamate-selective microelectrode arrays (MEAs) in young adult male, Sprague-Dawley rats anesthetized with either isoflurane or urethane. RESULTS Potassium chloride (KCl)-evoked glutamate overflow was similar under urethane and isoflurane anesthesia in all brain regions studied. Analysis of glutamate clearance determined that the uptake rate was significantly faster (53.2%, p < 0.05) within the thalamus under urethane compared to isoflurane, but no differences were measured in the cortex or hippocampus. Under urethane, glutamate clearance parameters were region-dependent, with significantly faster glutamate clearance in the thalamus compared to the cortex but not the hippocampus (p < 0.05). No region-dependent differences were measured for glutamate overflow using isoflurane. CONCLUSIONS These data support that amperometric recordings of KCl-evoked glutamate under isoflurane and urethane anesthesia result in similar and comparable data. However, certain parameters of glutamate clearance can vary based on choice of anesthesia and brain region. In these circumstances, special considerations are needed when comparing previous literature and planning future experiments.
Collapse
Affiliation(s)
- Joshua A Beitchman
- Department of Child Health, University of Arizona College of Medicine - Phoenix, 425 N. 5th St. | 322 ABC-1 Building, Phoenix, AZ, 85004-2127, USA
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix Children's Hospital, Phoenix, AZ, USA
- College of Graduate Studies, Midwestern University, Glendale, AZ, USA
| | - Gokul Krishna
- Department of Child Health, University of Arizona College of Medicine - Phoenix, 425 N. 5th St. | 322 ABC-1 Building, Phoenix, AZ, 85004-2127, USA
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix Children's Hospital, Phoenix, AZ, USA
| | - Caitlin E Bromberg
- Department of Child Health, University of Arizona College of Medicine - Phoenix, 425 N. 5th St. | 322 ABC-1 Building, Phoenix, AZ, 85004-2127, USA
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix Children's Hospital, Phoenix, AZ, USA
| | - Theresa Currier Thomas
- Department of Child Health, University of Arizona College of Medicine - Phoenix, 425 N. 5th St. | 322 ABC-1 Building, Phoenix, AZ, 85004-2127, USA.
- Barrow Neurological Institute at Phoenix Children's Hospital, Phoenix Children's Hospital, Phoenix, AZ, USA.
- Phoenix VA Healthcare System, Phoenix, AZ, USA.
| |
Collapse
|
2
|
Huffels CFM, Middeldorp J, Hol EM. Aß Pathology and Neuron-Glia Interactions: A Synaptocentric View. Neurochem Res 2023; 48:1026-1046. [PMID: 35976488 PMCID: PMC10030451 DOI: 10.1007/s11064-022-03699-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 06/30/2022] [Accepted: 07/15/2022] [Indexed: 10/15/2022]
Abstract
Alzheimer's disease (AD) causes the majority of dementia cases worldwide. Early pathological hallmarks include the accumulation of amyloid-ß (Aß) and activation of both astrocytes and microglia. Neurons form the building blocks of the central nervous system, and astrocytes and microglia provide essential input for its healthy functioning. Their function integrates at the level of the synapse, which is therefore sometimes referred to as the "quad-partite synapse". Increasing evidence puts AD forward as a disease of the synapse, where pre- and postsynaptic processes, as well as astrocyte and microglia functioning progressively deteriorate. Here, we aim to review the current knowledge on how Aß accumulation functionally affects the individual components of the quad-partite synapse. We highlight a selection of processes that are essential to the healthy functioning of the neuronal synapse, including presynaptic neurotransmitter release and postsynaptic receptor functioning. We further discuss how Aß affects the astrocyte's capacity to recycle neurotransmitters, release gliotransmitters, and maintain ion homeostasis. We additionally review literature on how Aß changes the immunoprotective function of microglia during AD progression and conclude by summarizing our main findings and highlighting the challenges in current studies, as well as the need for further research.
Collapse
Affiliation(s)
- Christiaan F M Huffels
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Jinte Middeldorp
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands
- Department of Neurobiology & Aging, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
3
|
Astrocyte reactivity in the glia limitans superficialis of the rat medial prefrontal cortex following sciatic nerve injury. Histochem Cell Biol 2023; 159:185-198. [PMID: 36326875 DOI: 10.1007/s00418-022-02161-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/14/2022] [Indexed: 11/06/2022]
Abstract
The glia limitans superficialis (GLS) on the rodent cortical surface consists of astrocyte bodies intermingled with their cytoplasmic processes. Many studies have observed astrocyte reactivity in the medial prefrontal cortex (mPFC) parenchyma induced by a peripheral nerve injury, while the response of GLS astrocytes is still not fully understood. The aim of our study was to identify the reactivity of rat GLS astrocytes in response to sciatic nerve compression (SNC) over different time periods. The alteration of GLS astrocyte reactivity was monitored using immunofluorescence (IF) intensities of glial fibrillary acidic protein (GFAP), glutamine synthetase (GS), and NFκBp65. Our results demonstrated that SNC induced GLS astrocyte reactivity seen as increased intensities of GFAP-IF, and longer extensions of cytoplasmic processes into lamina I. First significant increase of GFAP-IF was observed on post-operation day 7 (POD7) after SNC with further increases on POD14 and POD21. In contrast, dynamic alteration of the extension of cytoplasmic processes into lamina I was detected as early as POD1 and continued throughout the monitored survival periods of both sham and SNC operations. The reactivity of GLS astrocytes was not associated with their proliferation. In addition, GLS astrocytes also displayed a significant decrease in GS immunofluorescence (GS-IF) and NFκB immunofluorescence (NFκB-IF) in response to sham and SNC operation compared with naïve control rats. These results suggest that damaged peripheral tissues (following sham operation as well as peripheral nerve lesions) may induce significant changes in GLS astrocyte reactivity. The signaling mechanism from injured peripheral tissue and nerve remains to be elucidated.
Collapse
|
4
|
Rahiminejad E, Azad F, Parvizi-Fard A, Amiri M, Linares-Barranco B. A Neuromorphic CMOS Circuit With Self-Repairing Capability. IEEE TRANSACTIONS ON NEURAL NETWORKS AND LEARNING SYSTEMS 2022; 33:2246-2258. [PMID: 33417568 DOI: 10.1109/tnnls.2020.3045019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Neurophysiological observations confirm that the brain not only is able to detect the impaired synapses (in brain damage) but also it is relatively capable of repairing faulty synapses. It has been shown that retrograde signaling by astrocytes leads to the modulation of synaptic transmission and thus bidirectional collaboration of astrocyte with nearby neurons is an important aspect of self-repairing mechanism. Specifically, the retrograde signaling via astrocyte can increase the transmission probability of the healthy synapses linked to the neuron. Motivated by these findings, in the present research, a CMOS neuromorphic circuit with self-repairing capabilities is proposed based on astrocyte signaling. In this way, the computational model of self-repairing process is hired as a basis for designing a novel analog integrated circuit in the 180-nm CMOS technology. It is illustrated that the proposed analog circuit is able to successfully recompense the damaged synapses by appropriately modifying the voltage signals of the remaining healthy synapses in the wide range of frequency. The proposed circuit occupies 7500- [Formula: see text] silicon area and its power consumption is about [Formula: see text]. This neuromorphic fault-tolerant circuit can be considered as a key candidate for future silicon neuronal systems and implementation of neurorobotic and neuro-inspired circuits.
Collapse
|
5
|
Fabris F, Šoštarić P, Matak I, Binz T, Toffan A, Simonato M, Montecucco C, Pirazzini M, Rossetto O. Detection of VAMP Proteolysis by Tetanus and Botulinum Neurotoxin Type B In Vivo with a Cleavage-Specific Antibody. Int J Mol Sci 2022; 23:ijms23084355. [PMID: 35457172 PMCID: PMC9024618 DOI: 10.3390/ijms23084355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/08/2022] [Accepted: 04/12/2022] [Indexed: 11/17/2022] Open
Abstract
Tetanus and Botulinum type B neurotoxins are bacterial metalloproteases that specifically cleave the vesicle-associated membrane protein VAMP at an identical peptide bond, resulting in inhibition of neuroexocytosis. The minute amounts of these neurotoxins commonly used in experimental animals are not detectable, nor is detection of their VAMP substrate sensitive enough. The immune detection of the cleaved substrate is much more sensitive, as we have previously shown for botulinum neurotoxin type A. Here, we describe the production in rabbit of a polyclonal antibody raised versus a peptide encompassing the 13 residues C-terminal with respect to the neurotoxin cleavage site. The antibody was affinity purified and found to recognize, with high specificity and selectivity, the novel N-terminus of VAMP that becomes exposed after cleavage by tetanus toxin and botulinum toxin type B. This antibody recognizes the neoepitope not only in native and denatured VAMP but also in cultured neurons and in neurons in vivo in neurotoxin-treated mice or rats, suggesting the great potential of this novel tool to elucidate tetanus and botulinum B toxin activity in vivo.
Collapse
Affiliation(s)
- Federico Fabris
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (F.F.); (C.M.)
| | - Petra Šoštarić
- Department of Pharmacology, School of Medicine, University of Zagreb, Šalata 11, 10000 Zagreb, Croatia; (P.Š.); (I.M.)
| | - Ivica Matak
- Department of Pharmacology, School of Medicine, University of Zagreb, Šalata 11, 10000 Zagreb, Croatia; (P.Š.); (I.M.)
| | - Thomas Binz
- Institute of Cellular Biochemistry, Hannover Medical School, Carl-Neuberg-Straße 1, 30625 Hannover, Germany;
| | - Anna Toffan
- Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell’Università 10, 35020 Legnaro, Italy;
| | - Morena Simonato
- Institute of Neuroscience, Italian Research Council, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy;
| | - Cesare Montecucco
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (F.F.); (C.M.)
- Institute of Neuroscience, Italian Research Council, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy;
| | - Marco Pirazzini
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (F.F.); (C.M.)
- Interdepartmental Research Center of Myology CIR-Myo, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy
- Correspondence: (M.P.); (O.R.)
| | - Ornella Rossetto
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (F.F.); (C.M.)
- Institute of Neuroscience, Italian Research Council, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy;
- Interdepartmental Research Center of Myology CIR-Myo, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy
- Correspondence: (M.P.); (O.R.)
| |
Collapse
|
6
|
Ozgur M, Özyurt MG, Arkan S, Cavdar S. The Effects of Optogenetic Activation of Astrocytes on Spike-and-Wave Discharges in Genetic Absence Epileptic Rats. Ann Neurosci 2022; 29:53-61. [PMID: 35875425 PMCID: PMC9305907 DOI: 10.1177/09727531211072423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 11/29/2021] [Indexed: 11/16/2022] Open
Abstract
Background Absence seizures (petit mal seizures) are characterized by a brief loss of consciousness without loss of postural tone. The disease is diagnosed by an electroencephalogram (EEG) showing spike-wave discharges (SWD) caused by hypersynchronous thalamocortical (TC) oscillations. There has been an explosion of research highlighting the role of astrocytes in supporting and modulating neuronal activity. Despite established in vitro evidence, astrocytes' influence on the TC network remains to be elucidated in vivo in the absence epilepsy (AE). Purpose In this study, we investigated the role of astrocytes in the generation and modulation of SWDs. We hypothesize that disturbances in astrocytes' function may affect the pathomechanism of AE. Methods To direct the expression of channelrhodopsin-2 (ChR2) rAAV8-GFAP-ChR2(H134R)-EYFP or to control the effect of surgical intervention, AAV-CaMKIIa-EYFP was injected into the ventrobasal nucleus (VB) of the thalamus of 18 animals. After four weeks following the injection, rats were stimulated using blue light (~473 nm) and, simultaneously, the electrophysiological activity of the frontal cortical neurons was recorded for three consecutive days. The animals were then perfused, and the brain tissue was analyzed by confocal microscopy. Results A significant increase in the duration of SWD without affecting the number of SWD in genetic absence epileptic rats from Strasbourg (GAERS) compared to control injections was observed. The duration of the SWD was increased from 12.50 ± 4.41 s to 17.44 ± 6.07 following optogenetic stimulation in GAERS. The excitation of the astrocytes in Wistar Albino Glaxo Rijswijk (WAG-Rij) did not change the duration of SWD; however, stimulation resulted in a significant increase in the number of SWD from 18.52 ± 11.46 bursts/30 min to 30.17 ± 18.43 bursts/30 min. Whereas in control injection, the duration and the number of SWDs were similar at pre- and poststimulus. Both the background and poststimulus average firing rates of the SWD in WAG-Rij were significantly higher than the firing recorded in GAERS. Conclusion These findings suggest that VB astrocytes play a role in modulating the SWD generation in both rat models with distinct mechanisms and can present an essential target for the possible therapeutic approach for AE.
Collapse
Affiliation(s)
- Merve Ozgur
- Graduate School of Health Sciences, Division of Neuroscience, Koc University, Istanbul Turkey
- Department of Anatomy, Faculty of Medicine, Izmir University of Economics, Izmir, Turkey
- Department of Anatomy, Koç University School of Medicine, Istanbul, Turkey
| | - Mustafa Görkem Özyurt
- Graduate School of Sciences and Engineering, Koç University, Istanbul, Turkey
- Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Sertan Arkan
- Department of Experimental Medical Science, Molecular Neurobiology Unit, Lund University, Lund, Sweden
| | - Safiye Cavdar
- Department of Anatomy, Koç University School of Medicine, Istanbul, Turkey
| |
Collapse
|
7
|
Manninen T, Saudargiene A, Linne ML. Astrocyte-mediated spike-timing-dependent long-term depression modulates synaptic properties in the developing cortex. PLoS Comput Biol 2020; 16:e1008360. [PMID: 33170856 PMCID: PMC7654831 DOI: 10.1371/journal.pcbi.1008360] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 09/22/2020] [Indexed: 12/26/2022] Open
Abstract
Astrocytes have been shown to modulate synaptic transmission and plasticity in specific cortical synapses, but our understanding of the underlying molecular and cellular mechanisms remains limited. Here we present a new biophysicochemical model of a somatosensory cortical layer 4 to layer 2/3 synapse to study the role of astrocytes in spike-timing-dependent long-term depression (t-LTD) in vivo. By applying the synapse model and electrophysiological data recorded from rodent somatosensory cortex, we show that a signal from a postsynaptic neuron, orchestrated by endocannabinoids, astrocytic calcium signaling, and presynaptic N-methyl-D-aspartate receptors coupled with calcineurin signaling, induces t-LTD which is sensitive to the temporal difference between post- and presynaptic firing. We predict for the first time the dynamics of astrocyte-mediated molecular mechanisms underlying t-LTD and link complex biochemical networks at presynaptic, postsynaptic, and astrocytic sites to the time window of t-LTD induction. During t-LTD a single astrocyte acts as a delay factor for fast neuronal activity and integrates fast neuronal sensory processing with slow non-neuronal processing to modulate synaptic properties in the brain. Our results suggest that astrocytes play a critical role in synaptic computation during postnatal development and are of paramount importance in guiding the development of brain circuit functions, learning and memory.
Collapse
Affiliation(s)
- Tiina Manninen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Neurobiology, Stanford University, Stanford, CA, USA
| | - Ausra Saudargiene
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania
- Department of Informatics, Vytautas Magnus University, Kaunas, Lithuania
| | - Marja-Leena Linne
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
8
|
Lenk K, Satuvuori E, Lallouette J, Ladrón-de-Guevara A, Berry H, Hyttinen JAK. A Computational Model of Interactions Between Neuronal and Astrocytic Networks: The Role of Astrocytes in the Stability of the Neuronal Firing Rate. Front Comput Neurosci 2020; 13:92. [PMID: 32038210 PMCID: PMC6987305 DOI: 10.3389/fncom.2019.00092] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022] Open
Abstract
Recent research in neuroscience indicates the importance of tripartite synapses and gliotransmission mediated by astrocytes in neuronal system modulation. Although the astrocyte and neuronal network functions are interrelated, they are fundamentally different in their signaling patterns and, possibly, the time scales at which they operate. However, the exact nature of gliotransmission and the effect of the tripartite synapse function at the network level are currently elusive. In this paper, we propose a computational model of interactions between an astrocyte network and a neuron network, starting from tripartite synapses and spanning to a joint network level. Our model focuses on a two-dimensional setup emulating a mixed in vitro neuron-astrocyte cell culture. The model depicts astrocyte-released gliotransmitters exerting opposing effects on the neurons: increasing the release probability of the presynaptic neuron while hyperpolarizing the post-synaptic one at a longer time scale. We simulated the joint networks with various levels of astrocyte contributions and neuronal activity levels. Our results indicate that astrocytes prolong the burst duration of neurons, while restricting hyperactivity. Thus, in our model, the effect of astrocytes is homeostatic; the firing rate of the network stabilizes to an intermediate level independently of neuronal base activity. Our computational model highlights the plausible roles of astrocytes in interconnected astrocytic and neuronal networks. Our simulations support recent findings in neurons and astrocytes in vivo and in vitro suggesting that astrocytic networks provide a modulatory role in the bursting of the neuronal network.
Collapse
Affiliation(s)
- Kerstin Lenk
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Eero Satuvuori
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Institute for Complex Systems (ISC), National Research Council (CNR), Sesto Fiorentino, Italy.,Department of Physics and Astronomy, University of Florence, Sesto Fiorentino, Italy.,Department of Human Movement Sciences, MOVE Research Institute Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Jules Lallouette
- INRIA, Villeurbanne, France.,LIRIS UMR5205, University of Lyon, Villeurbanne, France
| | | | - Hugues Berry
- INRIA, Villeurbanne, France.,LIRIS UMR5205, University of Lyon, Villeurbanne, France
| | - Jari A K Hyttinen
- BioMediTech, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| |
Collapse
|
9
|
Mao Z, He S, Mesnard C, Synowicki P, Zhang Y, Chung L, Wiesman AI, Wilson TW, Monaghan DT. NMDA receptors containing GluN2C and GluN2D subunits have opposing roles in modulating neuronal oscillations; potential mechanism for bidirectional feedback. Brain Res 2019; 1727:146571. [PMID: 31786200 DOI: 10.1016/j.brainres.2019.146571] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 11/21/2019] [Accepted: 11/25/2019] [Indexed: 12/22/2022]
Abstract
NMDA receptor (NMDAR) antagonists such as ketamine, can reproduce many of the symptoms of schizophrenia. A reliable indicator of NMDAR channel blocker action in vivo is the augmentation of neuronal oscillation power. Since the coordinated and rhythmic activation of neuronal assemblies (oscillations) is necessary for perception, cognition and working memory, their disruption (inappropriate augmentation or inhibition of oscillatory power or inter-regional coherence) both in psychiatric conditions and with NMDAR antagonists may reflect the underlying defects causing schizophrenia symptoms. NMDAR antagonists and knockout (KO) mice were used to evaluate the role of GluN2C and GluN2D NMDAR subunits in generating NMDAR antagonist-induced oscillations. We find that basal oscillatory power was elevated in GluN2C-KO mice, especially in the low gamma frequencies while there was no statistically significant difference in basal oscillations between WT and GluN2D-KO mice. Compared to wildtype (WT) mice, NMDAR channel blockers caused a greater increase in oscillatory power in GluN2C-KO mice and were relatively ineffective in inducing oscillations in GluN2D-KO mice. In contrast, preferential blockade of GluN2A- and GluN2B-containing receptors induced oscillations that did not appear to be changed in either KO animal. We propose a model wherein NMDARs containing GluN2C in astrocytes and GluN2D in interneurons serve to detect local cortical excitatory synaptic activity and provide excitatory and inhibitory feedback, respectively, to local populations of postsynaptic excitatory neurons and thereby bidirectionally modulate oscillatory power.
Collapse
Affiliation(s)
- Zhihao Mao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Shengxi He
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Christopher Mesnard
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Paul Synowicki
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Yuning Zhang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Lucy Chung
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA
| | - Alex I Wiesman
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Tony W Wilson
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Daniel T Monaghan
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5800, USA.
| |
Collapse
|
10
|
Vardjan N, Parpura V, Verkhratsky A, Zorec R. Gliocrine System: Astroglia as Secretory Cells of the CNS. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1175:93-115. [PMID: 31583585 DOI: 10.1007/978-981-13-9913-8_4] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Astrocytes are secretory cells, actively participating in cell-to-cell communication in the central nervous system (CNS). They sense signaling molecules in the extracellular space, around the nearby synapses and also those released at much farther locations in the CNS, by their cell surface receptors, get excited to then release their own signaling molecules. This contributes to the brain information processing, based on diffusion within the extracellular space around the synapses and on convection when locales relatively far away from the release sites are involved. These functions resemble secretion from endocrine cells, therefore astrocytes were termed to be a part of the gliocrine system in 2015. An important mechanism, by which astrocytes release signaling molecules is the merger of the vesicle membrane with the plasmalemma, i.e., exocytosis. Signaling molecules stored in astroglial secretory vesicles can be discharged into the extracellular space after the vesicle membrane fuses with the plasma membrane. This leads to a fusion pore formation, a channel that must widen to allow the exit of the Vesiclal cargo. Upon complete vesicle membrane fusion, this process also integrates other proteins, such as receptors, transporters and channels into the plasma membrane, determining astroglial surface signaling landscape. Vesiclal cargo, together with the whole vesicle can also exit astrocytes by the fusion of multivesicular bodies with the plasma membrane (exosomes) or by budding of vesicles (ectosomes) from the plasma membrane into the extracellular space. These astroglia-derived extracellular vesicles can later interact with various target cells. Here, the characteristics of four types of astroglial secretory vesicles: synaptic-like microvesicles, dense-core vesicles, secretory lysosomes, and extracellular vesicles, are discussed. Then machinery for vesicle-based exocytosis, second messenger regulation and the kinetics of exocytotic vesicle content discharge or release of extracellular vesicles are considered. In comparison to rapidly responsive, electrically excitable neurons, the receptor-mediated cytosolic excitability-mediated astroglial exocytotic vesicle-based transmitter release is a relatively slow process.
Collapse
Affiliation(s)
- Nina Vardjan
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, 1000, Ljubljana, Slovenia. .,Celica Biomedical, 1000, Ljubljana, Slovenia.
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK.,Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark.,Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, 1000, Ljubljana, Slovenia. .,Celica Biomedical, 1000, Ljubljana, Slovenia.
| |
Collapse
|
11
|
Mahmoud S, Gharagozloo M, Simard C, Amrani A, Gris D. NLRX1 Enhances Glutamate Uptake and Inhibits Glutamate Release by Astrocytes. Cells 2019; 8:cells8050400. [PMID: 31052241 PMCID: PMC6562695 DOI: 10.3390/cells8050400] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 04/25/2019] [Accepted: 04/28/2019] [Indexed: 01/24/2023] Open
Abstract
Uptake of glutamate from the extracellular space and glutamate release to neurons are two major processes conducted by astrocytes in the central nervous system (CNS) that protect against glutamate excitotoxicity and strengthen neuronal firing, respectively. During inflammatory conditions in the CNS, astrocytes may lose one or both of these functions, resulting in accumulation of the extracellular glutamate, which eventually leads to excitotoxic neuronal death, which in turn worsens the CNS inflammation. NLRX1 is an innate immune NOD-like receptor that inhibits the major inflammatory pathways. It is localized in the mitochondria and was shown to inhibit cell death, enhance ATP production, and dampen oxidative stress. In the current work, using primary murine astrocyte cultures from WT and Nlrx1-/- mice, we demonstrate that NLRX1 potentiates astrocytic glutamate uptake by enhancing mitochondrial functions and the functional activity of glutamate transporters. Also, we report that NLRX1 inhibits glutamate release from astrocytes by repressing Ca2+-mediated glutamate exocytosis. Our study, for the first time, identified NLRX1 as a potential regulator of glutamate homeostasis in the CNS.
Collapse
Affiliation(s)
- Shaimaa Mahmoud
- Program of Immunology, Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Marjan Gharagozloo
- Program of Immunology, Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Camille Simard
- Program of Immunology, Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Abdelaziz Amrani
- Program of Immunology, Department of Pediatrics, CR-CHUS, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Denis Gris
- Program of Immunology, Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| |
Collapse
|
12
|
Mahmoud S, Gharagozloo M, Simard C, Gris D. Astrocytes Maintain Glutamate Homeostasis in the CNS by Controlling the Balance between Glutamate Uptake and Release. Cells 2019; 8:E184. [PMID: 30791579 PMCID: PMC6406900 DOI: 10.3390/cells8020184] [Citation(s) in RCA: 378] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 01/26/2023] Open
Abstract
Glutamate is one of the most prevalent neurotransmitters released by excitatory neurons in the central nervous system (CNS); however, residual glutamate in the extracellular space is, potentially, neurotoxic. It is now well-established that one of the fundamental functions of astrocytes is to uptake most of the synaptically-released glutamate, which optimizes neuronal functions and prevents glutamate excitotoxicity. In the CNS, glutamate clearance is mediated by glutamate uptake transporters expressed, principally, by astrocytes. Interestingly, recent studies demonstrate that extracellular glutamate stimulates Ca2+ release from the astrocytes' intracellular stores, which triggers glutamate release from astrocytes to the adjacent neurons, mostly by an exocytotic mechanism. This released glutamate is believed to coordinate neuronal firing and mediate their excitatory or inhibitory activity. Therefore, astrocytes contribute to glutamate homeostasis in the CNS, by maintaining the balance between their opposing functions of glutamate uptake and release. This dual function of astrocytes represents a potential therapeutic target for CNS diseases associated with glutamate excitotoxicity. In this regard, we summarize the molecular mechanisms of glutamate uptake and release, their regulation, and the significance of both processes in the CNS. Also, we review the main features of glutamate metabolism and glutamate excitotoxicity and its implication in CNS diseases.
Collapse
Affiliation(s)
- Shaimaa Mahmoud
- Program of Immunology, Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Marjan Gharagozloo
- Program of Immunology, Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Camille Simard
- Program of Immunology, Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| | - Denis Gris
- Program of Immunology, Department of Pharmacology-Physiology, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, QC J1H 5N4, Canada.
| |
Collapse
|
13
|
New Roles for Old Glue: Astrocyte Function in Synaptic Plasticity and Neurological Disorders. Int Neurourol J 2018; 22:S106-114. [PMID: 30396259 PMCID: PMC6234728 DOI: 10.5213/inj.1836214.107] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 10/10/2018] [Indexed: 01/02/2023] Open
Abstract
Previously believed to solely play a supportive role in the central nervous system, astrocytes are now considered active players in normal brain function. Evidence in recent decades extends their contributions beyond the classically held brain glue role; it's now known that astrocytes act as a unique excitable component with functions extending into local network modulation, synaptic plasticity, and memory formation, and postinjury repair. In this review article, we highlight our growing understanding of astrocyte function and physiology, the increasing role of gliotransmitters in neuron-glia communication, and the role of astrocytes in modulating synaptic plasticity and cognitive function. Owing to the duality of both beneficial and deleterious roles attributed to astrocytes, we also discuss the implications of this new knowledge as it applies to neurological disorders including Alzheimer disease, epilepsy, and schizophrenia.
Collapse
|
14
|
Gómez-Gonzalo M, Zehnder T, Requie LM, Bezzi P, Carmignoto G. Insights into the release mechanism of astrocytic glutamate evoking in neurons NMDA receptor-mediated slow depolarizing inward currents. Glia 2018; 66:2188-2199. [DOI: 10.1002/glia.23473] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/27/2018] [Accepted: 05/25/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Marta Gómez-Gonzalo
- Neuroscience Institute, National Research Council (CNR) and Department of Biomedical Sciences; University of Padova; Padova Italy
| | - Tamara Zehnder
- Department of Fundamental Neurosciences; University of Lausanne; Lausanne Switzerland
| | - Linda Maria Requie
- Neuroscience Institute, National Research Council (CNR) and Department of Biomedical Sciences; University of Padova; Padova Italy
| | - Paola Bezzi
- Department of Fundamental Neurosciences; University of Lausanne; Lausanne Switzerland
| | - Giorgio Carmignoto
- Neuroscience Institute, National Research Council (CNR) and Department of Biomedical Sciences; University of Padova; Padova Italy
| |
Collapse
|
15
|
Pál B. Involvement of extrasynaptic glutamate in physiological and pathophysiological changes of neuronal excitability. Cell Mol Life Sci 2018; 75:2917-2949. [PMID: 29766217 PMCID: PMC11105518 DOI: 10.1007/s00018-018-2837-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 04/27/2018] [Accepted: 05/07/2018] [Indexed: 12/14/2022]
Abstract
Glutamate is the most abundant neurotransmitter of the central nervous system, as the majority of neurons use glutamate as neurotransmitter. It is also well known that this neurotransmitter is not restricted to synaptic clefts, but found in the extrasynaptic regions as ambient glutamate. Extrasynaptic glutamate originates from spillover of synaptic release, as well as from astrocytes and microglia. Its concentration is magnitudes lower than in the synaptic cleft, but receptors responding to it have higher affinity for it. Extrasynaptic glutamate receptors can be found in neuronal somatodendritic location, on astroglia, oligodendrocytes or microglia. Activation of them leads to changes of neuronal excitability with different amplitude and kinetics. Extrasynaptic glutamate is taken up by neurons and astrocytes mostly via EAAT transporters, and astrocytes, in turn metabolize it to glutamine. Extrasynaptic glutamate is involved in several physiological phenomena of the central nervous system. It regulates neuronal excitability and synaptic strength by involving astroglia; contributing to learning and memory formation, neurosecretory and neuromodulatory mechanisms, as well as sleep homeostasis.The extrasynaptic glutamatergic system is affected in several brain pathologies related to excitotoxicity, neurodegeneration or neuroinflammation. Being present in dementias, neurodegenerative and neuropsychiatric diseases or tumor invasion in a seemingly uniform way, the system possibly provides a common component of their pathogenesis. Although parts of the system are extensively discussed by several recent reviews, in this review I attempt to summarize physiological actions of the extrasynaptic glutamate on neuronal excitability and provide a brief insight to its pathology for basic understanding of the topic.
Collapse
Affiliation(s)
- Balázs Pál
- Department of Physiology, Faculty of Medicine, University of Debrecen, Nagyerdei krt 98, Debrecen, 4012, Hungary.
| |
Collapse
|
16
|
Abstract
A recently identified mechanism for oligomeric Aβ-induced glutamate release from astrocytes involves intracellular Ca elevation, potentially by Ca-dependent vesicular release. Evidence suggests that levetiracetam (LEV; Keppra), an antiepileptic drug, can improve cognitive performance in both humans with mild cognitive impairment and animal models of Alzheimer disease. Because LEV acts by modulating neurotransmitter release from neurons by interaction with synaptic vesicles, we tested the effect of LEV on Aβ-induced astrocytic release of glutamate. We used a fluorescence resonance energy transfer-based glutamate sensor (termed SuperGluSnFR), whose structure is based on the ligand-binding site of glutamate receptors, to monitor glutamate release from primary cultures of human astrocytes exposed to oligomeric amyloid-β peptide 1-42 (Aβ42). We found that LEV (10 µM) inhibited oligomeric Aβ-induced astrocytic glutamate release. In addition, we show that this Aβ-induced glutamate release from astrocytes is sensitive to tetanus neurotoxin, an inhibitor of the vesicle release machinery. Taken together, our evidence suggests that LEV inhibits Aβ-induced vesicular glutamate release from astrocytes and thus may underlie, at least in part, the ability of LEV to reduce hyperexcitability in Alzheimer disease.
Collapse
|
17
|
Okuda H. A review of functional heterogeneity among astrocytes and the CS56-specific antibody-mediated detection of a subpopulation of astrocytes in adult brains. Anat Sci Int 2017; 93:161-168. [PMID: 29086253 DOI: 10.1007/s12565-017-0420-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 10/19/2017] [Indexed: 12/13/2022]
Abstract
Astrocytes comprise the largest class of glial cells in the mammalian central nerve system (CNS). Although astrocytes were long considered to be a homogeneous population of neuron-supporting cells, recent decades have seen a shift toward the recognition that astrocytes exhibit morphological and functional heterogeneities and serve as essential modulators of brain functions. However, the mechanism underlying astrocyte diversity remains unclear, and the different subpopulations are difficult to identify due to a lack of specific cell markers. In this review, I discuss current knowledge regarding astrocyte heterogeneity and introduce a subpopulation that can be detected via labeling with a chondroitin sulfate-specific antibody (CS56). These CS56-positive astrocytes were found to selectively express tenascin-R (TNR) in the adult mouse cerebral cortex. Further research demonstrated significantly lower levels of glutamate uptake activity and glutamate aspartate transporter expression in TNR-knockdown astrocytes relative to controls, suggesting that the expression and secretion of Tnr by a subpopulation of astrocytes may contribute to region-specific neuron-astrocyte interactions. In summary, these results suggest that CS56-specific antibody and Tnr could be used as novel markers to detect an astrocyte subpopulation in the adult CNS.
Collapse
Affiliation(s)
- Hiroaki Okuda
- Department of Functional Anatomy, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan.
| |
Collapse
|
18
|
Yoon MN, Kim MJ, Koong HS, Kim DK, Kim SH, Park HS. Ethanol suppresses carbamylcholine-induced intracellular calcium oscillation in mouse pancreatic acinar cells. Alcohol 2017; 63:53-59. [PMID: 28847382 DOI: 10.1016/j.alcohol.2017.03.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 03/23/2017] [Accepted: 03/23/2017] [Indexed: 12/31/2022]
Abstract
Oscillation of intracellular calcium levels is closely linked to initiating secretion of digestive enzymes from pancreatic acinar cells. Excessive alcohol consumption is known to relate to a variety of disorders in the digestive system, including the exocrine pancreas. In this study, we have investigated the role and mechanism of ethanol on carbamylcholine (CCh)-induced intracellular calcium oscillation in murine pancreatic acinar cells. Ethanol at concentrations of 30 and 100 mM reversibly suppressed CCh-induced Ca2+ oscillation in a dose-dependent manner. Pretreatment of ethanol has no effect on the store-operated calcium entry induced by 10 μM of CCh. Ethanol significantly reduced the initial calcium peak induced by low concentrations of CCh and therefore, the CCh-induced dose-response curve of the initial calcium peak was shifted to the right by ethanol pretreatment. Furthermore, ethanol significantly dose-dependently reduced inositol 1,4,5-trisphosphate-induced calcium release from the internal stores in permeabilized acinar cells. These results provide evidence that excessive alcohol intake could impair cytosolic calcium oscillation through inhibiting calcium release from intracellular stores in mouse pancreatic acinar cells.
Collapse
Affiliation(s)
- Mi Na Yoon
- Department of Physiology, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea
| | - Min Jae Kim
- Department of Physiology, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea
| | - Hwa Soo Koong
- Department of Dental Hygiene, College of Medical Science, Konyang University, Daejeon 35365, Republic of Korea
| | - Dong Kwan Kim
- Department of Physiology, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea
| | - Se Hoon Kim
- Department of Physiology, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea
| | - Hyung Seo Park
- Department of Physiology, College of Medicine, Konyang University, Daejeon 35365, Republic of Korea; Myunggok Medical Research Institute, Konyang University, Daejeon 35365, Republic of Korea.
| |
Collapse
|
19
|
Zhu H, Lin W, Zhao Y, Wang Z, Lao W, Kuang P, Zhou H. Transient upregulation of Nav1.6 expression in the genu of corpus callosum following middle cerebral artery occlusion in the rats. Brain Res Bull 2017; 132:20-27. [PMID: 28434994 DOI: 10.1016/j.brainresbull.2017.04.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2016] [Accepted: 04/12/2017] [Indexed: 12/19/2022]
Abstract
Focal ischemic stroke can lead to brain damage and cause human disability and death. Increased excitatory transmission and reduced neuronal inhibition are important pathological alterations in the cerebral ischemia, which can induce abnormal brain excitability. Nav1.6 is a key determinant of neuronal excitability in the nervous system. Here we investigate the expression of Nav1.6 at protein and mRNA levels in the rats subjected to middle cerebral artery occlusion (MCAO). Nav1.6 expression at mRNA levels in the ischemic and contralateral hemispheres of MCAO rats were persistently decreased at 6h, 12h and 24h after reperfusion compared to the sham-operated rats. However, a prominent, dynamic increase of Nav1.6 immunoreactivity in reactive astrocytes was observed in the genu of corpus callosum (GCC) of MCAO rats in the acute phase, reaching the peak at 6h after reperfusion, rapidly dropping at 12h and 24h after reperfusion. Furthermore, the upregulation of Nav1.6 expression was strongly correlated with the severity of reactive astrogliosis. Collectively, these findings suggest that this upregulated astrocytic sodium channel expression in the GCC of MCAO rats may contribute to the functional roles of reactive astrocytes in response to brain ischemia.
Collapse
Affiliation(s)
- Hongyan Zhu
- School of Life Science, Shanghai University, Nanchen Road 333, Shanghai, 200444, China.
| | - Weide Lin
- School of Life Science, Shanghai University, Nanchen Road 333, Shanghai, 200444, China
| | - Yuxiao Zhao
- School of Life Science, Shanghai University, Nanchen Road 333, Shanghai, 200444, China
| | - Ziyi Wang
- School of Life Science, Shanghai University, Nanchen Road 333, Shanghai, 200444, China
| | - Wenwen Lao
- School of Life Science, Shanghai University, Nanchen Road 333, Shanghai, 200444, China
| | - Ping Kuang
- School of Life Science, Shanghai University, Nanchen Road 333, Shanghai, 200444, China
| | - Houguang Zhou
- Department of Geriatrics Neurology, Huashan Hospital, Fudan University, Middle Wulumuqi Road, Shanghai, 200040, China
| |
Collapse
|
20
|
Oschmann F, Berry H, Obermayer K, Lenk K. From in silico astrocyte cell models to neuron-astrocyte network models: A review. Brain Res Bull 2017; 136:76-84. [PMID: 28189516 DOI: 10.1016/j.brainresbull.2017.01.027] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 01/30/2017] [Accepted: 01/31/2017] [Indexed: 01/25/2023]
Abstract
The idea that astrocytes may be active partners in synaptic information processing has recently emerged from abundant experimental reports. Because of their spatial proximity to neurons and their bidirectional communication with them, astrocytes are now considered as an important third element of the synapse. Astrocytes integrate and process synaptic information and by doing so generate cytosolic calcium signals that are believed to reflect neuronal transmitter release. Moreover, they regulate neuronal information transmission by releasing gliotransmitters into the synaptic cleft affecting both pre- and postsynaptic receptors. Concurrent with the first experimental reports of the astrocytic impact on neural network dynamics, computational models describing astrocytic functions have been developed. In this review, we give an overview over the published computational models of astrocytic functions, from single-cell dynamics to the tripartite synapse level and network models of astrocytes and neurons.
Collapse
Affiliation(s)
- Franziska Oschmann
- Technical University Berlin, Neural Information Processing Group, Sekr. MAR 5-6, Marchstrasse 23, 10587 Berlin, Germany; Bernstein Center for Computational Neuroscience, Berlin, Germany.
| | - Hugues Berry
- INRIA, 69603 Villeurbanne, France; LIRIS UMR5205, University of Lyon, 69622 Villeurbanne, France
| | - Klaus Obermayer
- Technical University Berlin, Neural Information Processing Group, Sekr. MAR 5-6, Marchstrasse 23, 10587 Berlin, Germany; Bernstein Center for Computational Neuroscience, Berlin, Germany
| | - Kerstin Lenk
- Tampere University of Technology, BioMediTech, PL100, 33014 Tampere, Finland.
| |
Collapse
|
21
|
Astrocytic Pathological Calcium Homeostasis and Impaired Vesicle Trafficking in Neurodegeneration. Int J Mol Sci 2017; 18:ijms18020358. [PMID: 28208745 PMCID: PMC5343893 DOI: 10.3390/ijms18020358] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 01/30/2017] [Accepted: 01/31/2017] [Indexed: 02/08/2023] Open
Abstract
Although the central nervous system (CNS) consists of highly heterogeneous populations of neurones and glial cells, clustered into diverse anatomical regions with specific functions, there are some conditions, including alertness, awareness and attention that require simultaneous, coordinated and spatially homogeneous activity within a large area of the brain. During such events, the brain, representing only about two percent of body mass, but consuming one fifth of body glucose at rest, needs additional energy to be produced. How simultaneous energy procurement in a relatively extended area of the brain takes place is poorly understood. This mechanism is likely to be impaired in neurodegeneration, for example in Alzheimer’s disease, the hallmark of which is brain hypometabolism. Astrocytes, the main neural cell type producing and storing glycogen, a form of energy in the brain, also hold the key to metabolic and homeostatic support in the central nervous system and are impaired in neurodegeneration, contributing to the slow decline of excitation-energy coupling in the brain. Many mechanisms are affected, including cell-to-cell signalling. An important question is how changes in cellular signalling, a process taking place in a rather short time domain, contribute to the neurodegeneration that develops over decades. In this review we focus initially on the slow dynamics of Alzheimer’s disease, and on the activity of locus coeruleus, a brainstem nucleus involved in arousal. Subsequently, we overview much faster processes of vesicle traffic and cytosolic calcium dynamics, both of which shape the signalling landscape of astrocyte-neurone communication in health and neurodegeneration.
Collapse
|
22
|
Bohmbach K, Schwarz MK, Schoch S, Henneberger C. The structural and functional evidence for vesicular release from astrocytes in situ. Brain Res Bull 2017; 136:65-75. [PMID: 28122264 DOI: 10.1016/j.brainresbull.2017.01.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 01/17/2017] [Accepted: 01/19/2017] [Indexed: 12/31/2022]
Abstract
The concept of the tripartite synapse states that bi-directional signalling between perisynaptic astrocyte processes, presynaptic axonal boutons and postsynaptic neuronal structures defines the properties of synaptic information processing. Ca2+-dependent vesicular release from astrocytes, as one of the mechanisms of astrocyte-neuron communication, has attracted particular attention but has also been the subject of intense debate. In neurons, regulated vesicular release is a strongly coordinated process. It requires a complex release machinery comprised of many individual components ranging from vesicular neurotransmitter transporters and soluble NSF attachment protein receptors (SNARE) proteins to Ca2+-sensors and the proteins that spatially and temporally control exocytosis of synaptic vesicles. If astrocytes employ similar mechanisms to release neurotransmitters is less well understood. The aim of this review is therefore to discuss recent experimental evidence that sheds light on the central structural components responsible for vesicular release from astrocytes in situ.
Collapse
Affiliation(s)
- Kirsten Bohmbach
- Institute of Cellular Neurosciences, University of Bonn Medical School, Bonn, Germany.
| | - Martin K Schwarz
- Department of Epileptology, University of Bonn Medical School, Bonn, Germany
| | - Susanne Schoch
- Institute of Neuropathology, University of Bonn Medical School, Bonn, Germany
| | - Christian Henneberger
- Institute of Cellular Neurosciences, University of Bonn Medical School, Bonn, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; Institute of Neurology, University College London, London, United Kingdom.
| |
Collapse
|
23
|
Pendin D, Greotti E, Lefkimmiatis K, Pozzan T. Exploring cells with targeted biosensors. J Gen Physiol 2016; 149:1-36. [PMID: 28028123 PMCID: PMC5217087 DOI: 10.1085/jgp.201611654] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Revised: 10/26/2016] [Accepted: 12/01/2016] [Indexed: 01/10/2023] Open
Abstract
Cellular signaling networks are composed of multiple pathways, often interconnected, that form complex networks with great potential for cross-talk. Signal decoding depends on the nature of the message as well as its amplitude, temporal pattern, and spatial distribution. In addition, the existence of membrane-bound organelles, which are both targets and generators of messages, add further complexity to the system. The availability of sensors that can localize to specific compartments in live cells and monitor their targets with high spatial and temporal resolution is thus crucial for a better understanding of cell pathophysiology. For this reason, over the last four decades, a variety of strategies have been developed, not only to generate novel and more sensitive probes for ions, metabolites, and enzymatic activity, but also to selectively deliver these sensors to specific intracellular compartments. In this review, we summarize the principles that have been used to target organic or protein sensors to different cellular compartments and their application to cellular signaling.
Collapse
Affiliation(s)
- Diana Pendin
- Neuroscience Institute, National Research Council, Padua Section, 35121 Padua, Italy.,Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
| | - Elisa Greotti
- Neuroscience Institute, National Research Council, Padua Section, 35121 Padua, Italy.,Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
| | - Konstantinos Lefkimmiatis
- Neuroscience Institute, National Research Council, Padua Section, 35121 Padua, Italy.,Venetian Institute of Molecular Medicine, 35129 Padua, Italy
| | - Tullio Pozzan
- Neuroscience Institute, National Research Council, Padua Section, 35121 Padua, Italy.,Venetian Institute of Molecular Medicine, 35129 Padua, Italy.,Department of Biomedical Sciences, University of Padua, 35121 Padua, Italy
| |
Collapse
|
24
|
Kostic M, Zivkovic N, Cvetanovic A, Stojanovic I, Colic M. IL-17 signalling in astrocytes promotes glutamate excitotoxicity: Indications for the link between inflammatory and neurodegenerative events in multiple sclerosis. Mult Scler Relat Disord 2016; 11:12-17. [PMID: 28104249 DOI: 10.1016/j.msard.2016.11.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Revised: 10/24/2016] [Accepted: 11/13/2016] [Indexed: 11/15/2022]
Abstract
OBJECTIVE Th-17 cells have been exclusively referred to inflammatory events in multiple sclerosis (MS), while their importance in the development of glutamate excitotoxicity and the consequent neurodegeneration has been a completely unexplored concept. Accordingly, the objective of our study was to assess IL-17A effect on astrocyte ability to metabolize and release glutamate, considering that astrocytes had the central role in glutamate homeostasis. METHODS By using primary rat astrocyte cultures, astrocyte ability to uptake glutamate was estimated by the alterations of glutamate transporters (GLAST and GLT-1) expression, whereas changes in glutamine synthetase expression were used to estimate the ability to metabolize glutamate. Gene expression was determined by real time polymerase chain reaction (rtPCR). IL-17A effect on astrocyte ability to produce glutamate was investigated directly, by measuring the level of released glutamate using high performance liquid chromatography (HPLC). RESULTS Lower concentrations of IL-17A reduced the expressions of both glutamate transporters and glutamine synthetase; however, this effect was lost when IL-17A was applied in a higher dose. IL-17A did not significantly modify glutamate release from astrocyte in basal conditions, but following Ca2+ stimulation, as well as Ca2+ removal from the culture medium, IL-17A stimulated glutamate release in dose-dependent manner. CONCLUSION Together, these results support that IL-17A could promote glutamate excitotoxicity by decreasing astrocyte ability to uptake and convert glutamate to non-toxic glutamine, but also by stimulating Ca2+ dependent glutamate release. Such interactions between IL-17A and glutamate excitotoxicity implicate the potential link between inflammation and neurodegeneration during MS pathogenesis, and identify astrocytes as a potential target in achieving neuroprotective effects in MS.
Collapse
Affiliation(s)
- Milos Kostic
- Department of Immunology, Medical Faculty, University of Nis, Blvd. dr Zorana Djindjica 81, 18000 Nis, Serbia.
| | - Nikola Zivkovic
- Department of Pathology, Medical Faculty, University of Nis, Blvd. dr Zorana Djindjica 81, 18000 Nis, Serbia
| | - Ana Cvetanovic
- Clinic of Oncology, Clinical Center Nis, Blvd. dr Zorana Djindjica 48, 18000 Nis, Serbia
| | - Ivana Stojanovic
- Department of Biochemistry, Medical Faculty, University of Nis, Blvd. dr Zorana Djindjica 81, 18000 Nis, Serbia
| | - Miodrag Colic
- Department of Immunology, Medical Faculty, University of Nis, Blvd. dr Zorana Djindjica 81, 18000 Nis, Serbia
| |
Collapse
|
25
|
Bang J, Kim HY, Lee H. Optogenetic and Chemogenetic Approaches for Studying Astrocytes and Gliotransmitters. Exp Neurobiol 2016; 25:205-221. [PMID: 27790055 PMCID: PMC5081467 DOI: 10.5607/en.2016.25.5.205] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 08/19/2016] [Accepted: 08/19/2016] [Indexed: 12/20/2022] Open
Abstract
The brain consists of heterogeneous populations of neuronal and non-neuronal cells. The revelation of their connections and interactions is fundamental to understanding normal brain functions as well as abnormal changes in pathological conditions. Optogenetics and chemogenetics have been developed to allow functional manipulations both in vitro and in vivo to examine causal relationships between cellular changes and functional outcomes. These techniques are based on genetically encoded effector molecules that respond exclusively to exogenous stimuli, such as a certain wavelength of light or a synthetic ligand. Activation of effector molecules provokes diverse intracellular changes, such as an influx or efflux of ions, depolarization or hyperpolarization of membranes, and activation of intracellular signaling cascades. Optogenetics and chemogenetics have been applied mainly to the study of neuronal circuits, but their use in studying non-neuronal cells has been gradually increasing. Here we introduce recent studies that have employed optogenetics and chemogenetics to reveal the function of astrocytes and gliotransmitters.
Collapse
Affiliation(s)
- Juwon Bang
- Department of Brain and Cognitive Sciences, DGIST, Daegu 42988, Korea
| | - Hak Yeong Kim
- Department of Brain and Cognitive Sciences, DGIST, Daegu 42988, Korea
| | - Hyosang Lee
- Department of Brain and Cognitive Sciences, DGIST, Daegu 42988, Korea
| |
Collapse
|
26
|
Tzour A, Leibovich H, Barkai O, Biala Y, Lev S, Yaari Y, Binshtok AM. K V 7/M channels as targets for lipopolysaccharide-induced inflammatory neuronal hyperexcitability. J Physiol 2016; 595:713-738. [PMID: 27506492 DOI: 10.1113/jp272547] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 07/27/2016] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS Neuroinflammation associated with CNS insults leads to neuronal hyperexcitability, which may culminate in epileptiform discharges. Application of the endotoxin lipopolysaccharide (LPS) to brain tissue initiates a neuroinflammatory cascade, providing an experimental model to study the mechanisms of neuroinflammatory neuronal hyperexcitability. Here we show that LPS application to hippocampal slices markedly enhances the excitability of CA1 pyramidal cells by inhibiting a specific potassium current, the M-current, generated by KV 7/M channels, which controls the excitability of almost every neuron in the CNS. The LPS-induced M-current inhibition is triggered by sequential activation of microglia, astrocytes and pyramidal cells, mediated by metabotropic purinergic and glutamatergic transmission, leading to blockade of KV 7/M channels by calcium released from intracellular stores. The identification of the downstream molecular target of neuroinflammation, namely the KV 7/M channel, potentially has far reaching implications for the understanding and treatment of many acute and chronic brain disorders. ABSTRACT Acute brain insults and many chronic brain diseases manifest an innate inflammatory response. The hallmark of this response is glia activation, which promotes repair of damaged tissue, but also induces structural and functional changes that may lead to an increase in neuronal excitability. We have investigated the mechanisms involved in the modulation of neuronal activity by acute inflammation. Initiating inflammatory responses in hippocampal tissue rapidly led to neuronal depolarization and repetitive firing even in the absence of active synaptic transmission. This action was mediated by a complex metabotropic purinergic and glutamatergic glia-to-neuron signalling cascade, leading to the blockade of neuronal KV 7/M channels by Ca2+ released from internal stores. These channels generate the low voltage-activating, non-inactivating M-type K+ current (M-current) that controls intrinsic neuronal excitability, and its inhibition was the predominant cause of the inflammation-induced hyperexcitability. Our discovery that the ubiquitous KV 7/M channels are the downstream target of the inflammation-induced cascade, has far reaching implications for the understanding and treatment of many acute and chronic brain disorders.
Collapse
Affiliation(s)
- Arik Tzour
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah School of Medicine, Jerusalem, 91120, Israel.,The Edmond and Lily Safra Center for Brain Sciences, Hebrew University of Jerusalem, Israel
| | - Hodaya Leibovich
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah School of Medicine, Jerusalem, 91120, Israel.,The Edmond and Lily Safra Center for Brain Sciences, Hebrew University of Jerusalem, Israel
| | - Omer Barkai
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah School of Medicine, Jerusalem, 91120, Israel.,The Edmond and Lily Safra Center for Brain Sciences, Hebrew University of Jerusalem, Israel
| | - Yoav Biala
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah School of Medicine, Jerusalem, 91120, Israel
| | - Shaya Lev
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah School of Medicine, Jerusalem, 91120, Israel.,The Edmond and Lily Safra Center for Brain Sciences, Hebrew University of Jerusalem, Israel
| | - Yoel Yaari
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah School of Medicine, Jerusalem, 91120, Israel
| | - Alexander M Binshtok
- Department of Medical Neurobiology, Institute for Medical Research Israel-Canada, Hebrew University-Hadassah School of Medicine, Jerusalem, 91120, Israel.,The Edmond and Lily Safra Center for Brain Sciences, Hebrew University of Jerusalem, Israel
| |
Collapse
|
27
|
Zorec R, Parpura V, Verkhratsky A. Astroglial Vesicular Trafficking in Neurodegenerative Diseases. Neurochem Res 2016; 42:905-917. [DOI: 10.1007/s11064-016-2055-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 08/30/2016] [Accepted: 08/31/2016] [Indexed: 12/20/2022]
|
28
|
Abstract
Neurons and glia are the principal cellular components of the nervous system. Although the glia are 10 times more numerous than neurons, until recently they were thought to be passive cells that monitor and support the active neurons by taking up used neurotransmitters from the synapses. In the past few years, this concept has been challenged by the findings that Ca2+ waves spread from one astrocyte to another via Ca2+-and SNARE (soluble N-ethylmaleimide-sensitive factor attachment protein receptor)-dependent gliotransmitter release in pure cultures of astrocytes, raising the possibility that glia are not so passive as previously thought. This hypothesis was further advanced by two recent reports, which demonstrated that astrocytes release glutamate via vesicular exocytosis in response to stimuli. The kinetics of single vesicle exocytosis is distinct from its neural equivalent, because in response to physiological stimulation, gliotransmitter release is exclusively in the mode of “kiss and run.” These advances were made possible by newly available techniques for single vesicle recordings, which will also be briefly reviewed here.
Collapse
Affiliation(s)
- Xiao-Ke Chen
- Institute of Molecular Medicine, Peking University, 5 Yi-He-Yuan Road, Beijing 100871, China
| | | | | |
Collapse
|
29
|
Hedgehog Signaling Modulates the Release of Gliotransmitters from Cultured Cerebellar Astrocytes. Neurochem Res 2015; 41:278-89. [PMID: 26694649 DOI: 10.1007/s11064-015-1791-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2015] [Revised: 11/24/2015] [Accepted: 11/24/2015] [Indexed: 10/22/2022]
Abstract
Sonic hedgehog (Shh), a member of the Hedgehog (Hh) family, plays essential roles in the development of the central nervous system. Recent studies suggest that the Hh signaling pathway also functions in mature astrocytes under physiological conditions. We first examined the expression of genes encoding Hh signaling molecules in the adult mouse cerebellum by in situ hybridization histochemistry. mRNA for Patched homolog 1 (Ptch1), a receptor for Hh family members, was expressed in S100β-positive astrocytes and Shh mRNA was expressed in HuC/D-positive neurons, implying that the Hh signaling pathway contributes to neuro-glial interactions. To test this hypothesis, we next examined the effects of recombinant SHH N-terminal protein (rSHH-N) on the functions of cultured cerebellar astrocytes. rSHH-N up-regulated Hh signal target genes such as Ptch1 and Gli-1, a key transcription factor of the Hh signaling pathway. Although activation of Hh signaling by rSHH-N or purmorphamine influenced neither glutamate uptake nor gliotransmitters release, inhibition of the Hh signaling pathway by cyclopamine, neutralizing antibody against SHH or intracellular Ca(2+) chelation decreased glutamate and ATP release from cultured cerebellar astrocytes. On the other hand, cyclopamine, neutralizing antibody against SHH or Ca(2+) chelator hardly affected D-serine secretion. Various kinase inhibitors attenuated glutamate and ATP release, while only U0126 reduced D-serine secretion from the astrocytes. These results suggested that the Hh signaling pathway sustains the release of glutamate and ATP and participates in neuro-glial interactions in the adult mouse brain. We also propose that signaling pathways distinct from the Hh pathway govern D-serine secretion from adult cerebellar astrocytes.
Collapse
|
30
|
Sahlender DA, Savtchouk I, Volterra A. What do we know about gliotransmitter release from astrocytes? Philos Trans R Soc Lond B Biol Sci 2015; 369:20130592. [PMID: 25225086 PMCID: PMC4173278 DOI: 10.1098/rstb.2013.0592] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Astrocytes participate in information processing by actively modulating synaptic properties via gliotransmitter release. Various mechanisms of astrocytic release have been reported, including release from storage organelles via exocytosis and release from the cytosol via plasma membrane ion channels and pumps. It is still not fully clear which mechanisms operate under which conditions, but some of them, being Ca2+-regulated, may be physiologically relevant. The properties of Ca2+-dependent transmitter release via exocytosis or via ion channels are different and expected to produce different extracellular transmitter concentrations over time and to have distinct functional consequences. The molecular aspects of these two release pathways are still under active investigation. Here, we discuss the existing morphological and functional evidence in support of either of them. Transgenic mouse models, specific antagonists and localization studies have provided insight into regulated exocytosis, albeit not in a systematic fashion. Even more remains to be uncovered about the details of channel-mediated release. Better functional tools and improved ultrastructural approaches are needed in order fully to define specific modalities and effects of astrocytic gliotransmitter release pathways.
Collapse
Affiliation(s)
- Daniela A Sahlender
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, Lausanne 1005, Switzerland
| | - Iaroslav Savtchouk
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, Lausanne 1005, Switzerland
| | - Andrea Volterra
- Department of Fundamental Neurosciences, University of Lausanne, Rue du Bugnon 9, Lausanne 1005, Switzerland
| |
Collapse
|
31
|
Ropert N, Jalil A, Li D. Expression and cellular function of vSNARE proteins in brain astrocytes. Neuroscience 2015; 323:76-83. [PMID: 26518463 DOI: 10.1016/j.neuroscience.2015.10.036] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Revised: 10/07/2015] [Accepted: 10/21/2015] [Indexed: 12/27/2022]
Abstract
Gray matter protoplasmic astrocytes, a major type of glial cell in the mammalian brain, extend thin processes ensheathing neuronal synaptic terminals. Albeit electrically silent, astrocytes respond to neuronal activity with Ca(2+) signals that trigger the release of gliotransmitters, such as glutamate, d-serine, and ATP, which modulate synaptic transmission. It has been suggested that the astrocytic processes, together with neuronal pre- and post-synaptic elements, constitute a tripartite synapse, and that astrocytes actively regulate information processing. Astrocytic vesicles expressing VAMP2 and VAMP3 vesicular SNARE (vSNARE) proteins have been suggested to be a key feature of the tripartite synapse and mediate gliotransmitter release through Ca(2+)-regulated exocytosis. However, the concept of exocytotic release of gliotransmitters by astrocytes has been challenged. Here we review studies investigating the expression profile of VAMP2 and VAMP3 vSNARE proteins in rodent astrocytes, and the functional implication of VAMP2/VAMP3 vesicles in astrocyte signaling. We also discuss our recent data suggesting that astrocytic VAMP3 vesicles regulate the trafficking of glutamate transporters at the plasma membrane and glutamate uptake. A better understanding of the functional consequences of the astrocytic vSNARE vesicles on glutamate signaling, neuronal excitability and plasticity, will require the development of new strategies to selectively interrogate the astrocytic vesicles trafficking in vivo.
Collapse
Affiliation(s)
- N Ropert
- Brain Physiology Laboratory, CNRS UMR8118, Paris F-75006, France; Fédération de Recherche en Neurosciences, FR 3636, Université Paris Descartes, 45 rue des Saints Pères, Paris F-75006, France; Sorbonne Paris Cité, 190, avenue de France, Paris F-75013, France
| | - A Jalil
- Brain Physiology Laboratory, CNRS UMR8118, Paris F-75006, France; Fédération de Recherche en Neurosciences, FR 3636, Université Paris Descartes, 45 rue des Saints Pères, Paris F-75006, France; Sorbonne Paris Cité, 190, avenue de France, Paris F-75013, France
| | - D Li
- Brain Physiology Laboratory, CNRS UMR8118, Paris F-75006, France; Fédération de Recherche en Neurosciences, FR 3636, Université Paris Descartes, 45 rue des Saints Pères, Paris F-75006, France; Sorbonne Paris Cité, 190, avenue de France, Paris F-75013, France.
| |
Collapse
|
32
|
Sreetama SC, Takano T, Nedergaard M, Simon SM, Jaiswal JK. Injured astrocytes are repaired by Synaptotagmin XI-regulated lysosome exocytosis. Cell Death Differ 2015; 23:596-607. [PMID: 26450452 DOI: 10.1038/cdd.2015.124] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2015] [Revised: 08/03/2015] [Accepted: 08/17/2015] [Indexed: 12/21/2022] Open
Abstract
Astrocytes are known to facilitate repair following brain injury; however, little is known about how injured astrocytes repair themselves. Repair of cell membrane injury requires Ca(2+)-triggered vesicle exocytosis. In astrocytes, lysosomes are the main Ca(2+)-regulated exocytic vesicles. Here we show that astrocyte cell membrane injury results in a large and rapid calcium increase. This triggers robust lysosome exocytosis where the fusing lysosomes release all luminal contents and merge fully with the plasma membrane. In contrast to this, receptor stimulation produces a small sustained calcium increase, which is associated with partial release of the lysosomal luminal content, and the lysosome membrane does not merge into the plasma membrane. In most cells, lysosomes express the synaptotagmin (Syt) isoform Syt VII; however, this isoform is not present on astrocyte lysosomes and exogenous expression of Syt VII on lysosome inhibits their exocytosis. Deletion of one of the most abundant Syt isoform in astrocyte--Syt XI--suppresses astrocyte lysosome exocytosis. This identifies lysosome as Syt XI-regulated exocytic vesicle in astrocytes. Further, inhibition of lysosome exocytosis (by Syt XI depletion or Syt VII expression) prevents repair of injured astrocytes. These results identify the lysosomes and Syt XI as the sub-cellular and molecular regulators, respectively of astrocyte cell membrane repair.
Collapse
Affiliation(s)
- S C Sreetama
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC, USA
| | - T Takano
- Center for Translational Neuromedicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, USA
| | - M Nedergaard
- Center for Translational Neuromedicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY, USA
| | - S M Simon
- Laboratory of Cellular Biophysics, The Rockefeller University, 1230 York Avenue, New York, NY, USA
| | - J K Jaiswal
- Center for Genetic Medicine Research, Children's National Medical Center, 111 Michigan Avenue NW, Washington, DC, USA.,Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, 111 Michigan Avenue NW, Washington, DC, USA
| |
Collapse
|
33
|
Vardjan N, Parpura V, Zorec R. Loose excitation-secretion coupling in astrocytes. Glia 2015; 64:655-67. [PMID: 26358496 DOI: 10.1002/glia.22920] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2015] [Accepted: 08/26/2015] [Indexed: 12/19/2022]
Abstract
Astrocytes play an important housekeeping role in the central nervous system. Additionally, as secretory cells, they actively participate in cell-to-cell communication, which can be mediated by membrane-bound vesicles. The gliosignaling molecules stored in these vesicles are discharged into the extracellular space after the vesicle membrane fuses with the plasma membrane. This process is termed exocytosis, regulated by SNARE proteins, and triggered by elevations in cytosolic calcium levels, which are necessary and sufficient for exocytosis in astrocytes. For astrocytic exocytosis, calcium is sourced from the intracellular endoplasmic reticulum store, although its entry from the extracellular space contributes to cytosolic calcium dynamics in astrocytes. Here, we discuss calcium management in astrocytic exocytosis and the properties of the membrane-bound vesicles that store gliosignaling molecules, including the vesicle fusion machinery and kinetics of vesicle content discharge. In astrocytes, the delay between the increase in cytosolic calcium activity and the discharge of secretions from the vesicular lumen is orders of magnitude longer than that in neurons. This relatively loose excitation-secretion coupling is likely tailored to the participation of astrocytes in modulating neural network processing.
Collapse
Affiliation(s)
- Nina Vardjan
- Celica Biomedical, Ljubljana, Slovenia.,Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Vladimir Parpura
- Department of Neurobiology, Center for Glial Biology in Medicine, Civitan International Research Center, Atomic Force Microscopy & Nanotechnology Laboratories, Evelyn F. McKnight Brain Institute, University of Alabama at Birmingham, Birmingham, Alabama
| | - Robert Zorec
- Celica Biomedical, Ljubljana, Slovenia.,Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
34
|
Abstract
Neuroglia, the "glue" that fills the space between neurons in the central nervous system, takes active part in nerve cell signaling. Neuroglial cells, astroglia, oligodendroglia, and microglia, are together about as numerous as neurons in the brain as a whole, and in the cerebral cortex grey matter, but the proportion varies widely among brain regions. Glial volume, however, is less than one-fifth of the tissue volume in grey matter. When stimulated by neurons or other cells, neuroglial cells release gliotransmitters by exocytosis, similar to neurotransmitter release from nerve endings, or by carrier-mediated transport or channel flux through the plasma membrane. Gliotransmitters include the common neurotransmitters glutamate and GABA, the nonstandard amino acid d-serine, the high-energy phosphate ATP, and l-lactate. The latter molecule is a "buffer" between glycolytic and oxidative metabolism as well as a signaling substance recently shown to act on specific lactate receptors in the brain. Complementing neurotransmission at a synapse, neuroglial transmission often implies diffusion of the transmitter over a longer distance and concurs with the concept of volume transmission. Transmission from glia modulates synaptic neurotransmission based on energetic and other local conditions in a volume of tissue surrounding the individual synapse. Neuroglial transmission appears to contribute significantly to brain functions such as memory, as well as to prevalent neuropathologies.
Collapse
Affiliation(s)
- Vidar Gundersen
- SN-Lab, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, and CMBN/SERTA/Healthy Brain Ageing Centre, University of Oslo, Oslo, Norway; Department of Neurology, Oslo University Hospital-Rikshospitalet, Oslo, Norway; Center for Healthy Aging, Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; and Brain and Muscle Energy Group, Department of Oral Biology and Division of Anatomy, Department of Molecular Medicine, University of Oslo, Oslo, Norway
| | - Jon Storm-Mathisen
- SN-Lab, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, and CMBN/SERTA/Healthy Brain Ageing Centre, University of Oslo, Oslo, Norway; Department of Neurology, Oslo University Hospital-Rikshospitalet, Oslo, Norway; Center for Healthy Aging, Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; and Brain and Muscle Energy Group, Department of Oral Biology and Division of Anatomy, Department of Molecular Medicine, University of Oslo, Oslo, Norway
| | - Linda Hildegard Bergersen
- SN-Lab, Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, and CMBN/SERTA/Healthy Brain Ageing Centre, University of Oslo, Oslo, Norway; Department of Neurology, Oslo University Hospital-Rikshospitalet, Oslo, Norway; Center for Healthy Aging, Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark; and Brain and Muscle Energy Group, Department of Oral Biology and Division of Anatomy, Department of Molecular Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
35
|
Cervetto C, Frattaroli D, Venturini A, Passalacqua M, Nobile M, Alloisio S, Tacchetti C, Maura G, Agnati LF, Marcoli M. Calcium-permeable AMPA receptors trigger vesicular glutamate release from Bergmann gliosomes. Neuropharmacology 2015; 99:396-407. [PMID: 26260232 DOI: 10.1016/j.neuropharm.2015.08.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Revised: 08/04/2015] [Accepted: 08/05/2015] [Indexed: 11/29/2022]
Abstract
The Bergmann glia is equipped with Ca2+-permeable AMPA receptors for glutamate, indispensable for structural and functional relations between the Bergmann glia and parallel/climbing fibers-Purkinje cell synapses. To better understand roles for the Bergmann AMPA receptors, herein we investigate on gliotransmitter release and Ca2+ signals in isolated Bergmann glia processes obtained from adult rat cerebellum. We found that: 1) the rat cerebellar purified astrocyte processes (gliosomes) expressed astrocytic and Bergmann markers and exhibited negligible contamination by nerve terminals, microglia, or oligodendrocytes; 2) activation of Ca2+-permeable AMPA receptors caused Ca2+ signals in the processes, and the release of glutamate from the processes; 3) effectiveness of rose bengal, trypan blue or bafilomycin A1, indicated that activation of the AMPA receptors evoked vesicular glutamate release. Cerebellar purified nerve terminals appeared devoid of glutamate-releasing Ca2+-permeable AMPA receptors, indicating that neuronal contamination may not be the source of the signals detected. Ultrastructural analysis indicated the presence of vesicles in the cytoplasm of the processes; confocal imaging confirmed the presence of vesicular glutamate transporters in Bergmann glia processes. We conclude that: a vesicular mechanism for release of the gliotransmitter glutamate is present in mature Bergmann processes; entry of Ca2+ through the AMPA receptors located on Bergmann processes is coupled with vesicular glutamate release. The findings would add a new role for a well-known Bergmann target for glutamate (the Ca2+-permeable AMPA receptors) and a new actor (the gliotransmitter glutamate) at the cerebellar excitatory synapses onto Purkinje cells.
Collapse
Affiliation(s)
- Chiara Cervetto
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy; Centre of Excellence for Biomedical Research CEBR, University of Genova, Viale Benedetto XV, 5, 16132 Genova, Italy
| | - Daniela Frattaroli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy
| | - Arianna Venturini
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy
| | - Mario Passalacqua
- Department of Experimental Medicine, Section of Biochemistry, Italian Institute of Biostructures and Biosystems, University of Genova, Via L.B. Alberti 2, 16132 Genova, Italy
| | - Mario Nobile
- CNR, Biophysics Institute, Via de Marinis 6, 16146 Genova, Italy
| | - Susanna Alloisio
- CNR, Biophysics Institute, Via de Marinis 6, 16146 Genova, Italy
| | - Carlo Tacchetti
- Department of Experimental Medicine, University of Genova, Via L. B. Alberti 2, 16132 Genova, Italy; Experimental Imaging Center, Scientific Institute San Raffaele, Via Olgettina 60, 20132 Milano, Italy
| | - Guido Maura
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy; Centre of Excellence for Biomedical Research CEBR, University of Genova, Viale Benedetto XV, 5, 16132 Genova, Italy
| | - Luigi Francesco Agnati
- Department of Biomedical, Metabolic Sciences and Neuroscience, University of Modena and Reggio Emilia, Via Campi 287, 41125 Modena, Italy; Department of Neuroscience, Karolinska Institutet, Retzius väg 8, Stockholm, Sweden
| | - Manuela Marcoli
- Department of Pharmacy, Section of Pharmacology and Toxicology, University of Genova, Viale Cembrano 4, 16148 Genova, Italy; Centre of Excellence for Biomedical Research CEBR, University of Genova, Viale Benedetto XV, 5, 16132 Genova, Italy.
| |
Collapse
|
36
|
Komin N, Moein M, Ellisman MH, Skupin A. Multiscale Modeling Indicates That Temperature Dependent [Ca2+]i Spiking in Astrocytes Is Quantitatively Consistent with Modulated SERCA Activity. Neural Plast 2015; 2015:683490. [PMID: 26347125 PMCID: PMC4539483 DOI: 10.1155/2015/683490] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 03/19/2015] [Indexed: 11/17/2022] Open
Abstract
Changes in the cytosolic Ca(2+) concentration ([Ca(2+)]i) are the most predominant active signaling mechanism in astrocytes that can modulate neuronal activity and is assumed to influence neuronal plasticity. Although Ca(2+) signaling in astrocytes has been intensively studied in the past, our understanding of the signaling mechanism and its impact on tissue level is still incomplete. Here we revisit our previously published data on the strong temperature dependence of Ca(2+) signals in both cultured primary astrocytes and astrocytes in acute brain slices of mice. We apply multiscale modeling to test the hypothesis that the temperature dependent [Ca(2+)]i spiking is mainly caused by the increased activity of the sarcoendoplasmic reticulum ATPases (SERCAs) that remove Ca(2+) from the cytosol into the endoplasmic reticulum. Quantitative comparison of experimental data with multiscale simulations supports the SERCA activity hypothesis. Further analysis of multiscale modeling and traditional rate equations indicates that the experimental observations are a spatial phenomenon where increasing pump strength leads to a decoupling of Ca(2+) release sites and subsequently to vanishing [Ca(2+)]i spikes.
Collapse
Affiliation(s)
- Niko Komin
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 7 Avenue des Hauts-Fourneaux, 4362 Esch-sur-Alzette, Luxembourg
- National Centre for Microscopy and Imaging Research, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0608, USA
| | - Mahsa Moein
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 7 Avenue des Hauts-Fourneaux, 4362 Esch-sur-Alzette, Luxembourg
| | - Mark H. Ellisman
- National Centre for Microscopy and Imaging Research, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0608, USA
| | - Alexander Skupin
- Luxembourg Centre for Systems Biomedicine, University of Luxembourg, 7 Avenue des Hauts-Fourneaux, 4362 Esch-sur-Alzette, Luxembourg
- National Centre for Microscopy and Imaging Research, University of California San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0608, USA
| |
Collapse
|
37
|
Astrocytes: Orchestrating synaptic plasticity? Neuroscience 2015; 323:43-61. [PMID: 25862587 DOI: 10.1016/j.neuroscience.2015.04.001] [Citation(s) in RCA: 161] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 03/25/2015] [Accepted: 04/01/2015] [Indexed: 01/09/2023]
Abstract
Synaptic plasticity is the capacity of a preexisting connection between two neurons to change in strength as a function of neural activity. Because synaptic plasticity is the major candidate mechanism for learning and memory, the elucidation of its constituting mechanisms is of crucial importance in many aspects of normal and pathological brain function. In particular, a prominent aspect that remains debated is how the plasticity mechanisms, that encompass a broad spectrum of temporal and spatial scales, come to play together in a concerted fashion. Here we review and discuss evidence that pinpoints to a possible non-neuronal, glial candidate for such orchestration: the regulation of synaptic plasticity by astrocytes.
Collapse
|
38
|
Irizarry-Valle Y, Parker AC. An astrocyte neuromorphic circuit that influences neuronal phase synchrony. IEEE TRANSACTIONS ON BIOMEDICAL CIRCUITS AND SYSTEMS 2015; 9:175-187. [PMID: 25934997 DOI: 10.1109/tbcas.2015.2417580] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Neuromorphic circuits are designed and simulated to emulate the role of astrocytes in phase synchronization of neuronal activity. We emulate, to a first order, the ability of slow inward currents (SICs) evoked by the astrocyte, acting on extrasynaptic N-methyl-D-aspartate receptors (NMDAR) of adjacent neurons, as a mechanism for phase synchronization. We run a simulation test incorporating two small networks of neurons interacting with astrocytic microdomains. These microdomains are designed using a resistive and capacitive ladder network and their interactions occur through pass transistors. Upon enough synaptic activity, the astrocytic microdomains interact with each other, generating SIC events on synapses of adjacent neurons. Since the amplitude of SICs is several orders of magnitude larger compared to synaptic currents, a SIC event drastically enhances the excitatory postsynaptic potential (EPSP) on adjacent neurons simultaneously. This causes neurons to fire synchronously in phase. Phase synchrony holds for a duration of time proportional to the time constant of the SIC decay. Once the SIC decay has completed, the neurons are able to go back to their natural phase difference, inducing desynchronization of their firing of spikes. This paper incorporates some biological aspects observed by recent experiments showing astrocytic influence on neuronal synchronization, and intends to offer a circuit view on the hypothesis of astrocytic role on synchronous activity that could potentially lead to the binding of neuronal information.
Collapse
|
39
|
Vardjan N, Zorec R. Excitable Astrocytes: Ca(2+)- and cAMP-Regulated Exocytosis. Neurochem Res 2015; 40:2414-24. [PMID: 25732760 DOI: 10.1007/s11064-015-1545-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 02/15/2015] [Accepted: 02/17/2015] [Indexed: 01/19/2023]
Abstract
During neural activity, neurotransmitters released at synapses reach neighbouring cells, such as astrocytes. These get excited via numerous mechanisms, including the G protein coupled receptors that regulate the cytosolic concentration of second messengers, such as Ca(2+) and cAMP. The stimulation of these pathways leads to feedback modulation of neuronal activity and the activity of other cells by the release of diverse substances, gliosignals that include classical neurotransmitters such as glutamate, ATP, or neuropeptides. Gliosignal molecules are released from astrocytes through several distinct molecular mechanisms, for example, by diffusion through membrane channels, by translocation via plasmalemmal transporters, or by vesicular exocytosis. Vesicular release regulated by a stimulus-mediated increase in cytosolic second messengers involves a SNARE-dependent merger of the vesicle membrane with the plasmalemma. The coupling between the stimulus and vesicular secretion of gliosignals in astrocytes is not as tight as in neurones. This is considered an adaptation to regulate homeostatic processes in a slow time domain as is the case in the endocrine system (slower than the nervous system), hence glial functions constitute the gliocrine system. This article provides an overview of the mechanisms of excitability, involving Ca(2+) and cAMP, where the former mediates phasic signalling and the latter tonic signalling. The molecular, anatomic, and physiologic properties of the vesicular apparatus mediating the release of gliosignals is presented.
Collapse
Affiliation(s)
- Nina Vardjan
- Celica Biomedical, 1000, Ljubljana, Slovenia.
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, 1000, Ljubljana, Slovenia.
| | - Robert Zorec
- Celica Biomedical, 1000, Ljubljana, Slovenia.
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, 1000, Ljubljana, Slovenia.
| |
Collapse
|
40
|
Zorec R, Verkhratsky A, Rodríguez JJ, Parpura V. Astrocytic vesicles and gliotransmitters: Slowness of vesicular release and synaptobrevin2-laden vesicle nanoarchitecture. Neuroscience 2015; 323:67-75. [PMID: 25727638 DOI: 10.1016/j.neuroscience.2015.02.033] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 02/01/2015] [Accepted: 02/18/2015] [Indexed: 12/30/2022]
Abstract
Neurotransmitters released at synapses activate neighboring astrocytes, which in turn, modulate neuronal activity by the release of diverse neuroactive substances that include classical neurotransmitters such as glutamate, GABA or ATP. Neuroactive substances are released from astrocytes through several distinct molecular mechanisms, for example, by diffusion through membrane channels, by translocation via plasmalemmal transporters or by vesicular exocytosis. Vesicular release regulated by a stimulus-mediated increase in cytosolic calcium involves soluble N-ethyl maleimide-sensitive fusion protein attachment protein receptor (SNARE)-dependent merger of the vesicle membrane with the plasmalemma. Up to 25 molecules of synaptobrevin 2 (Sb2), a SNARE complex protein, reside at a single astroglial vesicle; an individual neuronal, i.e. synaptic, vesicle contains ∼70 Sb2 molecules. It is proposed that this paucity of Sb2 molecules in astrocytic vesicles may determine the slow secretion. In the present essay we shall overview multiple aspects of vesicular architecture and types of vesicles based on their cargo and dynamics in astroglial cells.
Collapse
Affiliation(s)
- R Zorec
- University of Ljubljana, Institute of Pathophysiology, Laboratory of Neuroendocrinology and Molecular Cell Physiology, Zaloska cesta 4, SI-1000 Ljubljana, Slovenia; Celica, BIOMEDICAL, Technology Park 24, 1000 Ljubljana, Slovenia.
| | - A Verkhratsky
- University of Ljubljana, Institute of Pathophysiology, Laboratory of Neuroendocrinology and Molecular Cell Physiology, Zaloska cesta 4, SI-1000 Ljubljana, Slovenia; Celica, BIOMEDICAL, Technology Park 24, 1000 Ljubljana, Slovenia; Faculty of Life Sciences, The University of Manchester, Manchester M13 9PT, UK; Achucarro Center for Neuroscience, IKERBASQUE, 48011 Bilbao, Spain; University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain.
| | - J J Rodríguez
- Achucarro Center for Neuroscience, IKERBASQUE, 48011 Bilbao, Spain; University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain.
| | - V Parpura
- Department of Neurobiology, Civitan International Research Center and Center for Glial Biology in Medicine, Evelyn F. McKnight Brain Institute, Atomic Force Microscopy & Nanotechnology Laboratories, 1719 6th Avenue South, CIRC 429, University of Alabama at Birmingham, Birmingham, AL 35294-0021, USA; Department of Biotechnology, University of Rijeka, Radmile Matejčić 2, 51000 Rijeka, Croatia.
| |
Collapse
|
41
|
Abstract
Glial cells are an integral part of functional communication in the brain. Here we show that astrocytes contribute to the fast dynamics of neural circuits that underlie normal cognitive behaviors. In particular, we found that the selective expression of tetanus neurotoxin (TeNT) in astrocytes significantly reduced the duration of carbachol-induced gamma oscillations in hippocampal slices. These data prompted us to develop a novel transgenic mouse model, specifically with inducible tetanus toxin expression in astrocytes. In this in vivo model, we found evidence of a marked decrease in electroencephalographic (EEG) power in the gamma frequency range in awake-behaving mice, whereas neuronal synaptic activity remained intact. The reduction in cortical gamma oscillations was accompanied by impaired behavioral performance in the novel object recognition test, whereas other forms of memory, including working memory and fear conditioning, remained unchanged. These results support a key role for gamma oscillations in recognition memory. Both EEG alterations and behavioral deficits in novel object recognition were reversed by suppression of tetanus toxin expression. These data reveal an unexpected role for astrocytes as essential contributors to information processing and cognitive behavior.
Collapse
|
42
|
Mehta S, Aye-Han NN, Ganesan A, Oldach L, Gorshkov K, Zhang J. Calmodulin-controlled spatial decoding of oscillatory Ca2+ signals by calcineurin. eLife 2014; 3:e03765. [PMID: 25056880 PMCID: PMC4141273 DOI: 10.7554/elife.03765] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Calcineurin is responsible for mediating a wide variety of cellular processes in response to dynamic calcium (Ca(2+)) signals, yet the precise mechanisms involved in the spatiotemporal control of calcineurin signaling are poorly understood. Here, we use genetically encoded fluorescent biosensors to directly probe the role of cytosolic Ca(2+) oscillations in modulating calcineurin activity dynamics in insulin-secreting MIN6 β-cells. We show that Ca(2+) oscillations induce distinct temporal patterns of calcineurin activity in the cytosol and plasma membrane vs at the ER and mitochondria in these cells. Furthermore, we found that these differential calcineurin activity patterns are determined by variations in the subcellular distribution of calmodulin (CaM), indicating that CaM plays an active role in shaping both the spatial and temporal aspects of calcineurin signaling. Together, our findings provide new insights into the mechanisms by which oscillatory signals are decoded to generate specific functional outputs within different cellular compartments.
Collapse
Affiliation(s)
- Sohum Mehta
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, United States
| | - Nwe-Nwe Aye-Han
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, United States
| | - Ambhighainath Ganesan
- Department of Biomedical Engineering, The Johns Hopkins University, Baltimore, United States
| | - Laurel Oldach
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, United States
| | - Kirill Gorshkov
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, United States
| | - Jin Zhang
- Department of Pharmacology and Molecular Sciences, The Johns Hopkins University School of Medicine, Baltimore, United States The Solomon H. Snyder Department of Neuroscience, The Johns Hopkins University School of Medicine, Baltimore, United States Department of Oncology, The Johns Hopkins University School of Medicine, Baltimore, United States
| |
Collapse
|
43
|
Cao X, Li LP, Qin XH, Li SJ, Zhang M, Wang Q, Hu HH, Fang YY, Gao YB, Li XW, Sun LR, Xiong WC, Gao TM, Zhu XH. Astrocytic adenosine 5'-triphosphate release regulates the proliferation of neural stem cells in the adult hippocampus. Stem Cells 2014; 31:1633-43. [PMID: 23630193 DOI: 10.1002/stem.1408] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Accepted: 03/27/2013] [Indexed: 12/13/2022]
Abstract
Astrocytes are key components of the niche for neural stem cells (NSCs) in the adult hippocampus and play a vital role in regulating NSC proliferation and differentiation. However, the exact molecular mechanisms by which astrocytes modulate NSC proliferation have not been identified. Here, we identified adenosine 5'-triphosphate (ATP) as a proliferative factor required for astrocyte-mediated proliferation of NSCs in the adult hippocampus. Our results indicate that ATP is necessary and sufficient for astrocytes to promote NSC proliferation in vitro. The lack of inositol 1,4,5-trisphosphate receptor type 2 and transgenic blockage of vesicular gliotransmission induced deficient ATP release from astrocytes. This deficiency led to a dysfunction in NSC proliferation that could be rescued via the administration of exogenous ATP. Moreover, P2Y1-mediated purinergic signaling is involved in the astrocyte promotion of NSC proliferation. As adult hippocampal neurogenesis is potentially involved in major mood disorder, our results might offer mechanistic insights into this disease.
Collapse
Affiliation(s)
- Xiong Cao
- Department of Neurobiology, School of Basic Medical Sciences; Key Laboratory of Neuroplasticity of Guangdong Higher Education Institutes, and; School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Araque A, Carmignoto G, Haydon PG, Oliet SHR, Robitaille R, Volterra A. Gliotransmitters travel in time and space. Neuron 2014; 81:728-39. [PMID: 24559669 DOI: 10.1016/j.neuron.2014.02.007] [Citation(s) in RCA: 883] [Impact Index Per Article: 80.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/03/2014] [Indexed: 12/12/2022]
Abstract
The identification of the presence of active signaling between astrocytes and neurons in a process termed gliotransmission has caused a paradigm shift in our thinking about brain function. However, we are still in the early days of the conceptualization of how astrocytes influence synapses, neurons, networks, and ultimately behavior. In this Perspective, our goal is to identify emerging principles governing gliotransmission and consider the specific properties of this process that endow the astrocyte with unique functions in brain signal integration. We develop and present hypotheses aimed at reconciling confounding reports and define open questions to provide a conceptual framework for future studies. We propose that astrocytes mainly signal through high-affinity slowly desensitizing receptors to modulate neurons and perform integration in spatiotemporal domains complementary to those of neurons.
Collapse
Affiliation(s)
- Alfonso Araque
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, 28002 Madrid, Spain; Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| | - Giorgio Carmignoto
- Istituto di Neuroscienze, Consiglio Nazionale delle Ricerche and Dipartimento Scienze Biomediche, Università di Padova, 35121 Padova, Italy.
| | - Philip G Haydon
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Stéphane H R Oliet
- Inserm U862, Neurocentre Magendie, 33077 Bordeaux, France; Université de Bordeaux, 33077 Bordeaux, France
| | - Richard Robitaille
- Département de Neurosciences, Université de Montréal, Montréal, QC H3C 3J7, Canada; Groupe de Recherche sur le Système Nerveux Central, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Andrea Volterra
- Département de Neurosciences Fondamentales (DNF), Faculté de Biologie et de Médecine, Université de Lausanne, 1005 Lausanne, Switzerland
| |
Collapse
|
45
|
|
46
|
Decreased astroglial monocarboxylate transporter 4 expression in temporal lobe epilepsy. Mol Neurobiol 2014; 50:327-38. [PMID: 24464262 DOI: 10.1007/s12035-013-8619-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 12/12/2013] [Indexed: 10/25/2022]
Abstract
Efflux of monocaroxylates like lactate, pyruvate, and ketone bodies from astrocytes through monocarboxylate transporter 4 (MCT4) supplies the local neuron population with metabolic intermediates to meet energy requirements under conditions of increased demand. Disruption of this astroglial-neuron metabolic coupling pathway may contribute to epileptogenesis. We measured MCT4 expression in temporal lobe epileptic foci excised from patients with intractable epilepsy and in rats injected with pilocarpine, an animal model of temporal lobe epilepsy (TLE). Cortical MCT4 expression levels were significantly lower in TLE patients compared with controls, due at least partially to MCT4 promoter methylation. Expression of MCT4 also decreased progressively in pilocarpine-treated rats from 12 h to 14 days post-administration. Underexpression of MCT4 in cultured astrocytes induced by a short hairpin RNA promoted apoptosis. Knockdown of astrocyte MCT4 also suppressed excitatory amino acid transporter 1 (EAAT1) expression. Reduced MCT4 and EAAT1 expression by astrocytes may lead to neuronal hyperexcitability and epileptogenesis in the temporal lobe by reducing the supply of metabolic intermediates and by allowing accumulation of extracellular glutamate.
Collapse
|
47
|
Salamone A, Mura E, Zappettini S, Grilli M, Olivero G, Preda S, Govoni S, Marchi M. Inhibitory effects of beta-amyloid on the nicotinic receptors which stimulate glutamate release in rat hippocampus: the glial contribution. Eur J Pharmacol 2013; 723:314-21. [PMID: 24275353 DOI: 10.1016/j.ejphar.2013.11.011] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2013] [Revised: 10/22/2013] [Accepted: 11/11/2013] [Indexed: 01/20/2023]
Abstract
We investigated on the neuronal nicotinic acetylcholine receptor subtypes involved in the cholinergic control of in vivo hippocampal glutamate (GLU), aspartate (ASP) and inhibitory γ-aminobutyric acid (GABA) overflow. We also investigated on the possible contribution of nicotinic acetylcholine receptors subtypes present on astrocytes in the regulation of the three neurotransmitter amino acids overflow using hippocampal gliosomes and on the effects of beta-amyloid (Aβ) 1-40 on the nicotinic control of amino acid neurotransmitter release. Nicotine was able to enhance the in vivo overflow of the three amino acids being more potent in stimulating GLU overflow. The α7 selective agonist PHA543613 induced an overflow very similar to that of nicotine. The α4β2 selective agonist 5IA85380 was significantly less potent in inducing GLU overflow while the overflow of ASP and GABA were almost inconsistent. Aβ1-40 inhibited the neurotransmitter overflow stimulated by PHA543613 but not the one evoked by 5IA85380. In hippocampal gliosomes nicotine elicited selectively GLU overflow which was also evoked by 5IA85380 and by the α7 selective agonist choline. Nicotine- and choline-induced glutamate overflow in gliosomes was inhibited by Aα1-40. In conclusion nicotine administration in vivo elicits hippocampal GLU release mostly through α7 nicotinic acetylcholine receptors likely present both on neurons and astrocytes. Aβ inhibitory effect on the nicotinic-control of GLU release seems to depend primarily to the inhibition of α7 nicotinic acetylcholine receptors functional responses.
Collapse
Affiliation(s)
- Alessia Salamone
- Section of Pharmacology and Toxicology, Department of Pharmacy, University of Genoa, Genoa, Italy
| | - Elisa Mura
- Department of Drug Sciences, Centre of Excellence in Applied Biology, University of Pavia, Pavia, Italy
| | - Stefania Zappettini
- Section of Pharmacology and Toxicology, Department of Pharmacy, University of Genoa, Genoa, Italy
| | - Massimo Grilli
- Section of Pharmacology and Toxicology, Department of Pharmacy, University of Genoa, Genoa, Italy
| | - Guendalina Olivero
- Section of Pharmacology and Toxicology, Department of Pharmacy, University of Genoa, Genoa, Italy
| | - Stefania Preda
- Department of Drug Sciences, Centre of Excellence in Applied Biology, University of Pavia, Pavia, Italy
| | - Stefano Govoni
- Department of Drug Sciences, Centre of Excellence in Applied Biology, University of Pavia, Pavia, Italy
| | - Mario Marchi
- Section of Pharmacology and Toxicology, Department of Pharmacy, University of Genoa, Genoa, Italy; Center of Excellence for Biomedical Research, University of Genoa, Italy.
| |
Collapse
|
48
|
Castillo C, Norcini M, Martin Hernandez L, Correa G, Blanck T, Recio-Pinto E. Satellite glia cells in dorsal root ganglia express functional NMDA receptors. Neuroscience 2013; 240:135-46. [DOI: 10.1016/j.neuroscience.2013.02.031] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 01/29/2013] [Accepted: 02/13/2013] [Indexed: 01/06/2023]
|
49
|
Lutgen V, Qualmann K, Resch J, Kong L, Choi S, Baker DA. Reduction in phencyclidine induced sensorimotor gating deficits in the rat following increased system xc⁻ activity in the medial prefrontal cortex. Psychopharmacology (Berl) 2013; 226:531-40. [PMID: 23192314 PMCID: PMC3595356 DOI: 10.1007/s00213-012-2926-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 11/09/2012] [Indexed: 12/25/2022]
Abstract
RATIONALE Aspects of schizophrenia, including deficits in sensorimotor gating, have been linked to glutamate dysfunction and/or oxidative stress in the prefrontal cortex. System xc(-), a cystine-glutamate antiporter, is a poorly understood mechanism that contributes to both cellular antioxidant capacity and glutamate homeostasis. OBJECTIVES Our goal was to determine whether increased system xc(-) activity within the prefrontal cortex would normalize a rodent measure of sensorimotor gating. METHODS In situ hybridization was used to map messenger RNA (mRNA) expression of xCT, the active subunit of system xc(-), in the prefrontal cortex. Prepulse inhibition was used to measure sensorimotor gating; deficits in prepulse inhibition were produced using phencyclidine (0.3-3 mg/kg, sc). N-Acetylcysteine (10-100 μM) and the system xc(-) inhibitor (S)-4-carboxyphenylglycine (CPG, 0.5 μM) were used to increase and decrease system xc(-) activity, respectively. The uptake of (14)C-cystine into tissue punches obtained from the prefrontal cortex was used to assay system xc(-) activity. RESULTS The expression of xCT mRNA in the prefrontal cortex was most prominent in a lateral band spanning primarily the prelimbic cortex. Although phencyclidine did not alter the uptake of (14)C-cystine in prefrontal cortical tissue punches, intraprefrontal cortical infusion of N-acetylcysteine (10-100 μM) significantly reduced phencyclidine- (1.5 mg/kg, sc) induced deficits in prepulse inhibition. N-Acetylcysteine was without effect when coinfused with CPG (0.5 μM), indicating an involvement of system xc(-). CONCLUSIONS These results indicate that phencyclidine disrupts sensorimotor gating through system xc(-) independent mechanisms, but that increasing cystine-glutamate exchange in the prefrontal cortex is sufficient to reduce behavioral deficits produced by phencyclidine.
Collapse
Affiliation(s)
- Victoria Lutgen
- Department of Biomedical Sciences, Marquette University, Suite 446, 561 N. 15St, Milwaukee, WI 53233
| | - Krista Qualmann
- Department of Biomedical Sciences, Marquette University, Suite 446, 561 N. 15St, Milwaukee, WI 53233
| | - Jon Resch
- Department of Biomedical Sciences, Marquette University, Suite 446, 561 N. 15St, Milwaukee, WI 53233
| | - Linghai Kong
- Department of Biomedical Sciences, Marquette University, Suite 446, 561 N. 15St, Milwaukee, WI 53233
| | - SuJean Choi
- Department of Biomedical Sciences, Marquette University, Suite 446, 561 N. 15St, Milwaukee, WI 53233
| | - David A. Baker
- Department of Biomedical Sciences, Marquette University, Suite 446, 561 N. 15St, Milwaukee, WI 53233
| |
Collapse
|
50
|
Crunelli V, Carmignoto G. New vistas on astroglia in convulsive and non-convulsive epilepsy highlight novel astrocytic targets for treatment. J Physiol 2012; 591:775-85. [PMID: 23230232 DOI: 10.1113/jphysiol.2012.243378] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Our current knowledge of the role of astrocytes in health and disease states supports the view that many physiological brain functions and neurological diseases are finely tuned, and in certain cases fully determined, by the continuous cross-talk between astrocytes and neurons. This novel way of interpreting brain activity as a dynamic and reciprocal interplay between astrocytic and neuronal networks has also influenced our understanding of epilepsy, not only forcing a reinterpretation of old findings, but also being a catalyst for novel experimentation. In this review, we summarize some of the recent studies that highlight these novel distinct contributions of astrocytes to the expression of convulsive and non-convulsive epileptiform discharges and seizures. The emerging picture suggests a general framework based on bilateral signalling between astrocytes and neurons for a fuller understanding of epileptogenic and epileptic mechanisms in the brain network. Astrocytes potentially represent targets for the development of those novel chemical entities with improved efficacy for the treatment of convulsive and non-convulsive epilepsy that expert groups have recognized as one of the key priorities for the management of epilepsy.
Collapse
Affiliation(s)
- Vincenzo Crunelli
- Neuroscience Division, School of Bioscience, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK.
| | | |
Collapse
|