1
|
Alcaide J, Gramuntell Y, Klimczak P, Bueno-Fernandez C, Garcia-Verellen E, Guicciardini C, Sandi C, Castillo-Gómez E, Crespo C, Perez-Rando M, Nacher J. Long term effects of peripubertal stress on the thalamic reticular nucleus of female and male mice. Neurobiol Dis 2024; 200:106642. [PMID: 39173845 DOI: 10.1016/j.nbd.2024.106642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 08/18/2024] [Indexed: 08/24/2024] Open
Abstract
Adverse experiences during infancy and adolescence have an important and enduring effect on the brain and are predisposing factors for mental disorders, particularly major depression. This impact is particularly notable in regions with protracted development, such as the prefrontal cortex. The inhibitory neurons of this cortical region are altered by peripubertal stress (PPS), particularly in female mice. In this study we have explored whether the inhibitory circuits of the thalamus are impacted by PPS in male and female mice. This diencephalic structure, as the prefrontal cortex, also completes its development during postnatal life and is affected by adverse experiences. The long-term changes induced by PPS were exclusively found in adult female mice. We have found that PPS increases depressive-like behavior and induces changes in parvalbumin-expressing (PV+) cells of the thalamic reticular nucleus (TRN). We observed reductions in the volume of the TRN, together with those of parameters related to structures/molecules that regulate the plasticity and connectivity of PV+ cells: perineuronal nets, matricellular structures surrounding PV+ neurons, and the polysialylated form of the neural cell adhesion molecule (PSA-NCAM). The expression of the GluN1, but not of GluN2C, NMDA receptor subunit was augmented in the TRN after PPS. An increase in the fluorescence intensity of PV+ puncta was also observed in the synaptic output of TRN neurons in the lateral posterior thalamic nucleus. These results demonstrate that the inhibitory circuits of the thalamus, as those of the prefrontal cortex, are vulnerable to the effects of aversive experiences during early life, particularly in females. This vulnerability is probably related to the protracted development of the TRN and might contribute to the development of psychiatric disorders.
Collapse
Affiliation(s)
- Julia Alcaide
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain; Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010 Valencia, Spain
| | - Yaiza Gramuntell
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain; Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010 Valencia, Spain
| | - Patrycja Klimczak
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain; Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010 Valencia, Spain
| | - Clara Bueno-Fernandez
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain; Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010 Valencia, Spain
| | - Erica Garcia-Verellen
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain
| | - Chiara Guicciardini
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain
| | - Carmen Sandi
- Laboratory of Behavioral Genetics, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Esther Castillo-Gómez
- Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Department of Medicine, School of Medical Sciences, Universitat Jaume I, Valencia, Spain
| | - Carlos Crespo
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain
| | - Marta Perez-Rando
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain; Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010 Valencia, Spain.
| | - Juan Nacher
- Neurobiology Unit, Institute for Biotechnology and Biomedicine (BIOTECMED), Universitat de València, 46100, Spain; Spanish National Network for Research in Mental Health CIBERSAM, 28029, Spain; Fundación Investigación Hospital Clínico de Valencia, INCLIVA, 46010 Valencia, Spain.
| |
Collapse
|
2
|
Li X, Prudente AS, Prato V, Guo X, Hao H, Jones F, Figoli S, Mullen P, Wang Y, Tonello R, Lee SH, Shah S, Maffei B, Berta T, Du X, Gamper N. Peripheral gating of mechanosensation by glial diazepam binding inhibitor. J Clin Invest 2024; 134:e176227. [PMID: 38888973 PMCID: PMC11324294 DOI: 10.1172/jci176227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 06/13/2024] [Indexed: 06/20/2024] Open
Abstract
We report that diazepam binding inhibitor (DBI) is a glial messenger mediating crosstalk between satellite glial cells (SGCs) and sensory neurons in the dorsal root ganglion (DRG). DBI is highly expressed in SGCs of mice, rats, and humans, but not in sensory neurons or most other DRG-resident cells. Knockdown of DBI results in a robust mechanical hypersensitivity without major effects on other sensory modalities. In vivo overexpression of DBI in SGCs reduces sensitivity to mechanical stimulation and alleviates mechanical allodynia in neuropathic and inflammatory pain models. We further show that DBI acts as an unconventional agonist and positive allosteric modulator at the neuronal GABAA receptors, particularly strongly affecting those with a high-affinity benzodiazepine binding site. Such receptors are selectively expressed by a subpopulation of mechanosensitive DRG neurons, and these are also more enwrapped with DBI-expressing glia, as compared with other DRG neurons, suggesting a mechanism for a specific effect of DBI on mechanosensation. These findings identified a communication mechanism between peripheral neurons and SGCs. This communication modulates pain signaling and can be targeted therapeutically.
Collapse
Affiliation(s)
- Xinmeng Li
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Arthur Silveira Prudente
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Vincenzo Prato
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Xianchuan Guo
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Han Hao
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Frederick Jones
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
- Department of Life Sciences, Manchester Metropolitan University, Manchester, United Kingdom
| | - Sofia Figoli
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Pierce Mullen
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Yujin Wang
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Raquel Tonello
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Sang Hoon Lee
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Shihab Shah
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Benito Maffei
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, Ohio, USA
| | - Xiaona Du
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
| | - Nikita Gamper
- Department of Pharmacology; The Key Laboratory of Neural and Vascular Biology, Ministry of Education; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, Hebei, China
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
3
|
Xia M, Anderson TL, Prantzalos ER, Hawkinson TR, Clarke HA, Keohane SB, Sun RC, Turner JR, Ortinski PI. Voltage-gated potassium channels control extended access cocaine seeking: a role for nucleus accumbens astrocytes. Neuropsychopharmacology 2024; 49:551-560. [PMID: 37660129 PMCID: PMC10789875 DOI: 10.1038/s41386-023-01718-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/03/2023] [Accepted: 08/21/2023] [Indexed: 09/04/2023]
Abstract
Dopaminergic signaling in the nucleus accumbens shell (NAc) regulates neuronal activity relevant to reward-related learning, including cocaine-associated behaviors. Although astrocytes respond to dopamine and cocaine with structural changes, the impact of dopamine and cocaine on astrocyte functional plasticity has not been widely studied. Specifically, behavioral implications of voltage-gated channel activity in the canonically non-excitable astrocytes are not known. We characterized potassium channel function in NAc astrocytes following exposure to exogenous dopamine or cocaine self-administration training under short (2 h/day) and extended (6 h/day) access schedules. Electrophysiological, Ca2+ imaging, mRNA, and mass spectrometry tools were used for molecular characterization. Behavioral effects were examined after NAc-targeted microinjections of channel antagonists and astroglial toxins. Exogenous dopamine increased activity of currents mediated by voltage-gated (Kv7) channels in NAc astrocytes. This was associated with a ~5-fold increase in expression of Kcnq2 transcript level in homogenized NAc micropunches. Matrix-assisted laser desorption/ionization mass spectrometry revealed increased NAc dopamine levels in extended access, relative to short access, rats. Kv7 inhibition selectively increased frequency and amplitude of astrocyte intracellular Ca2+ transients in NAc of extended access rats. Inhibition of Kv7 channels in the NAc attenuated cocaine-seeking in extended access rats only, an effect that was occluded by microinjection of the astrocyte metabolic poison, fluorocitrate. These results suggest that voltage-gated K+ channel signaling in NAc astrocytes is behaviorally relevant, support Kv7-mediated regulation of astrocyte Ca2+ signals, and propose novel mechanisms of neuroglial interactions relevant to drug use.
Collapse
Affiliation(s)
- Mengfan Xia
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY, USA
| | - Tanner L Anderson
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA
| | - Emily R Prantzalos
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA
| | - Tara R Hawkinson
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA
| | - Harrison A Clarke
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA
| | - Shannon B Keohane
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA
| | - Ramon C Sun
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, USA
- Center for Advanced Spatial Biomolecule Research, University of Florida, Gainesville, FL, USA
| | - Jill R Turner
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, KY, USA
| | - Pavel I Ortinski
- Department of Neuroscience, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
4
|
Courtney CD, Sobieski C, Ramakrishnan C, Ingram RJ, Wojnowski NM, DeFazio RA, Deisseroth K, Christian-Hinman CA. Optoα1AR activation in astrocytes modulates basal hippocampal synaptic excitation and inhibition in a stimulation-specific manner. Hippocampus 2023; 33:1277-1291. [PMID: 37767862 PMCID: PMC10842237 DOI: 10.1002/hipo.23580] [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: 06/09/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023]
Abstract
Astrocytes play active roles at synapses and can monitor, respond, and adapt to local synaptic activity. While there is abundant evidence that astrocytes modulate excitatory transmission in the hippocampus, evidence for astrocytic modulation of hippocampal synaptic inhibition remains more limited. Furthermore, to better investigate roles for astrocytes in modulating synaptic transmission, more tools that can selectively activate native G protein signaling pathways in astrocytes with both spatial and temporal precision are needed. Here, we utilized AAV8-GFAP-Optoα1AR-eYFP (Optoα1AR), a viral vector that enables activation of Gq signaling in astrocytes via light-sensitive α1-adrenergic receptors. To determine if stimulating astrocytic Optoα1AR modulates hippocampal synaptic transmission, recordings were made in CA1 pyramidal cells with surrounding astrocytes expressing Optoα1AR, channelrhodopsin (ChR2), or GFP. Both high-frequency (20 Hz, 45-ms light pulses, 5 mW, 5 min) and low-frequency (0.5 Hz, 1-s pulses at increasing 1, 5, and 10 mW intensities, 90 s per intensity) blue light stimulation were tested. 20 Hz Optoα1AR stimulation increased both inhibitory and excitatory postsynaptic current (IPSC and EPSC) frequency, and the effect on miniature IPSCs (mIPSCs) was largely reversible within 20 min. However, low-frequency stimulation of Optoα1AR did not modulate either IPSCs or EPSCs, suggesting that astrocytic Gq -dependent modulation of basal synaptic transmission in the hippocampus is stimulation-dependent. By contrast, low-frequency stimulation of astrocytic ChR2 was effective in increasing both synaptic excitation and inhibition. Together, these data demonstrate that Optoα1AR activation in astrocytes changes basal GABAergic and glutamatergic transmission, but only following high-frequency stimulation, highlighting the importance of temporal dynamics when using optical tools to manipulate astrocyte function.
Collapse
Affiliation(s)
- Connor D. Courtney
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - Courtney Sobieski
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | | | - Robbie J. Ingram
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - Natalia M. Wojnowski
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801
| | - R. Anthony DeFazio
- Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109
| | - Karl Deisseroth
- Department of Bioengineering, Stanford University, Stanford, CA 94305
| | - Catherine A. Christian-Hinman
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL 61801
- Department of Molecular and Integrative Physiology, University of Illinois Urbana-Champaign, Urbana, IL 61801
- Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL 61801
| |
Collapse
|
5
|
Li X, Prudente AS, Prato V, Guo X, Hao H, Jones F, Figoli S, Mullen P, Wang Y, Tonnello R, Lee SH, Shah S, Maffei B, Berta T, Du X, Gamper N. Peripheral gating of pain by glial endozepine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.20.567848. [PMID: 38045227 PMCID: PMC10690183 DOI: 10.1101/2023.11.20.567848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
We report that diazepam binding inhibitor (DBI) is a glial messenger mediating satellite glia-sensory neuron crosstalk in the dorsal root ganglion (DRG). DBI is highly and specifically expressed in satellite glia cells (SGCs) of mice, rat and human, but not in sensory neurons or other DRG-resident cells. Knockdown of DBI results in a robust mechanical hypersensitivity without significant effects on other sensory modalities. In vivo overexpression of DBI in SGCs reduces sensitivity to mechanical stimulation and alleviates mechanical allodynia in neuropathic and inflammatory pain models. We further show that DBI acts as a partial agonist and positive allosteric modulator at the neuronal GABAA receptors, particularly strongly effecting those with a high-affinity benzodiazepine binding site. Such receptors are selectively expressed by a subpopulation of mechanosensitive DRG neurons and these are also more enwrapped with DBI-expressing glia, as compared to other DRG neurons, suggesting a mechanism for specific effect of DBI on mechanosensation. These findings identified a new, peripheral neuron-glia communication mechanism modulating pain signalling, which can be targeted therapeutically.
Collapse
Affiliation(s)
- Xinmeng Li
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Arthur Silveira Prudente
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Vincenzo Prato
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Xianchuan Guo
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Han Hao
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Frederick Jones
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
- Department of Life Sciences, Manchester Metropolitan University, Manchester, UK
| | - Sofia Figoli
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Pierce Mullen
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Yujin Wang
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Raquel Tonnello
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Sang Hoon Lee
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Shihab Shah
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Benito Maffei
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Temugin Berta
- Pain Research Center, Department of Anesthesiology, University of Cincinnati Medical Center, Cincinnati, OH, USA
| | - Xiaona Du
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
| | - Nikita Gamper
- Department of Pharmacology, Hebei Medical University, Shijiazhuang, China
- Faculty of Biological Sciences, School of Biomedical Sciences, University of Leeds, Leeds, UK
| |
Collapse
|
6
|
New LE, Yanagawa Y, McConkey GA, Deuchars J, Deuchars SA. GABAergic regulation of cell proliferation within the adult mouse spinal cord. Neuropharmacology 2023; 223:109326. [PMID: 36336067 DOI: 10.1016/j.neuropharm.2022.109326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 10/17/2022] [Accepted: 10/31/2022] [Indexed: 11/05/2022]
Abstract
Manipulation of neural stem cell proliferation and differentiation in the postnatal CNS is receiving significant attention due to therapeutic potential. In the spinal cord, such manipulations may promote repair in conditions such as multiple sclerosis or spinal cord injury, but may also limit excessive cell proliferation contributing to tumours such as ependymomas. We show that when ambient γ-aminobutyric acid (GABA) is increased in vigabatrin-treated or decreased by GAD67 allele haplodeficiency in glutamic acid decarboxylase67-green fluorescent protein (GAD67-GFP) mice of either sex, the numbers of proliferating cells respectively decreased or increased. Thus, intrinsic spinal cord GABA levels are correlated with the extent of cell proliferation, providing important evidence for manipulating these levels. Diazepam binding inhibitor, an endogenous protein that interacts with GABA receptors and its breakdown product, octadecaneuropeptide, which preferentially activates central benzodiazepine (CBR) sites, were highly expressed in spinal cord, especially in ependymal cells surrounding the central canal. Furthermore, animals with reduced CBR activation via treatment with flumazenil or Ro15-4513, or with a G2F77I mutation in the CBR binding site had greater numbers of Ethynyl-2'-deoxyuridine positive cells compared to control, which maintained their stem cell status since the proportion of newly proliferated cells becoming oligodendrocytes or astrocytes was significantly lower. Altering endogenous GABA levels or modulating GABAergic signalling through specific sites on GABA receptors therefore influences NSC proliferation in the adult spinal cord. These findings provide a basis for further study into how GABAergic signalling could be manipulated to enable spinal cord self-regeneration and recovery or limit pathological proliferative activity.
Collapse
Affiliation(s)
- Lauryn E New
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, UK
| | - Yuchio Yanagawa
- Department of Genetic and Behavioural Neuroscience, Gunma University, Graduate School of Medicine, Maebashi, 371-8511, Japan
| | - Glenn A McConkey
- School of Biology, Faculty of Biological Sciences, University of Leeds, UK
| | - Jim Deuchars
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, UK
| | - Susan A Deuchars
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, UK.
| |
Collapse
|
7
|
Wiera G, Brzdąk P, Lech AM, Lebida K, Jabłońska J, Gmerek P, Mozrzymas JW. Integrins Bidirectionally Regulate the Efficacy of Inhibitory Synaptic Transmission and Control GABAergic Plasticity. J Neurosci 2022; 42:5830-5842. [PMID: 35701161 PMCID: PMC9337602 DOI: 10.1523/jneurosci.1458-21.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 05/17/2022] [Accepted: 05/28/2022] [Indexed: 01/29/2023] Open
Abstract
For many decades, synaptic plasticity was believed to be restricted to excitatory transmission. However, in recent years, this view started to change, and now it is recognized that GABAergic synapses show distinct forms of activity-dependent long-term plasticity, but the underlying mechanisms remain obscure. Herein, we asked whether signaling mediated by β1 or β3 subunit-containing integrins might be involved in regulating the efficacy of GABAergic synapses, including the NMDA receptor-dependent inhibitory long-term potentiation (iLTP) in the hippocampus. We found that activation of β3 integrin with fibrinogen induced a stable depression, whereas inhibition of β1 integrin potentiated GABAergic synapses at CA1 pyramidal neurons in male mice. Additionally, compounds that interfere with the interaction of β1 or β3 integrins with extracellular matrix blocked the induction of NMDA-iLTP. In conclusion, we provide the first evidence that integrins are key players in regulating the endogenous modulatory mechanisms of GABAergic inhibition and plasticity in the hippocampus.SIGNIFICANCE STATEMENT Epilepsy, schizophrenia, and anxiety are just a few medical conditions associated with dysfunctional inhibitory synaptic transmission. GABAergic synapses are known for their extraordinary susceptibility to modulation by endogenous factors and exogenous pharmacological agents. We describe here that integrins, adhesion proteins, play a key role in the modulation of inhibitory synaptic transmission. Specifically, we show that interference with integrin-dependent adhesion results in a variety of effects on the amplitude and frequency of GABAergic mIPSCs. Activation of β3 subunit-containing integrins induces inhibitory long-term depression, whereas the inhibition of β1 subunit-containing integrins induces iLTP. Our results unveil an important mechanism controlling synaptic inhibition, which opens new avenues into the usage of integrin-aimed pharmaceuticals as modulators of GABAergic synapses.
Collapse
Affiliation(s)
- Grzegorz Wiera
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Patrycja Brzdąk
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Anna Maria Lech
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Department of Molecular Physiology and Neurobiology, University of Wroclaw, 50-335 Wroclaw, Poland
| | - Katarzyna Lebida
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Jadwiga Jabłońska
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-368 Wroclaw, Poland
| | - Przemysław Gmerek
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-368 Wroclaw, Poland
- Department of Molecular Physiology and Neurobiology, University of Wroclaw, 50-335 Wroclaw, Poland
| | - Jerzy W Mozrzymas
- Department of Biophysics and Neuroscience, Wroclaw Medical University, 50-368 Wroclaw, Poland
| |
Collapse
|
8
|
Liu J, Feng X, Wang Y, Xia X, Zheng JC. Astrocytes: GABAceptive and GABAergic Cells in the Brain. Front Cell Neurosci 2022; 16:892497. [PMID: 35755777 PMCID: PMC9231434 DOI: 10.3389/fncel.2022.892497] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 05/17/2022] [Indexed: 12/14/2022] Open
Abstract
Astrocytes, the most numerous glial cells in the brain, play an important role in preserving normal neural functions and mediating the pathogenesis of neurological disorders. Recent studies have shown that astrocytes are GABAceptive and GABAergic astrocytes express GABAA receptors, GABAB receptors, and GABA transporter proteins to capture and internalize GABA. GABAceptive astrocytes thus influence both inhibitory and excitatory neurotransmission by controlling the levels of extracellular GABA. Furthermore, astrocytes synthesize and release GABA to directly regulate brain functions. In this review, we highlight recent research progresses that support astrocytes as GABAceptive and GABAergic cells. We also summarize the roles of GABAceptive and GABAergic astrocytes that serve as an inhibitory node in the intercellular communication in the brain. Besides, we discuss future directions for further expanding our knowledge on the GABAceptive and GABAergic astrocyte signaling.
Collapse
Affiliation(s)
- Jianhui Liu
- Department of Anesthesiology, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, China
| | - Xuanran Feng
- Department of Anesthesiology, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, China
| | - Yi Wang
- Translational Research Center, Shanghai Yangzhi Rehabilitation Hospital affiliated to Tongji University School of Medicine, Shanghai, China
| | - Xiaohuan Xia
- Department of Anesthesiology, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, China.,Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, China.,Shanghai Frontiers Science Center of Nanocatalytic Medicine, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, China
| | - Jialin C Zheng
- Department of Anesthesiology, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, China.,Center for Translational Neurodegeneration and Regenerative Therapy, Tongji Hospital affiliated to Tongji University School of Medicine, Shanghai, China.,Shanghai Frontiers Science Center of Nanocatalytic Medicine, Shanghai, China.,Translational Research Institute of Brain and Brain-Like Intelligence, Shanghai Fourth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, China.,Collaborative Innovation Center for Brain Science, Tongji University, Shanghai, China
| |
Collapse
|
9
|
Why won't it stop? The dynamics of benzodiazepine resistance in status epilepticus. Nat Rev Neurol 2022; 18:428-441. [PMID: 35538233 DOI: 10.1038/s41582-022-00664-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/19/2022] [Indexed: 11/08/2022]
Abstract
Status epilepticus is a life-threatening neurological emergency that affects both adults and children. Approximately 36% of episodes of status epilepticus do not respond to the current preferred first-line treatment, benzodiazepines. The proportion of episodes that are refractory to benzodiazepines is higher in low-income and middle-income countries (LMICs) than in high-income countries (HICs). Evidence suggests that longer episodes of status epilepticus alter brain physiology, thereby contributing to the emergence of benzodiazepine resistance. Such changes include alterations in GABAA receptor function and in the transmembrane gradient for chloride, both of which erode the ability of benzodiazepines to enhance inhibitory synaptic signalling. Often, current management guidelines for status epilepticus do not account for these duration-related changes in pathophysiology, which might differentially impact individuals in LMICs, where the average time taken to reach medical attention is longer than in HICs. In this Perspective article, we aim to combine clinical insights and the latest evidence from basic science to inspire a new, context-specific approach to efficiently managing status epilepticus.
Collapse
|
10
|
Alquier T, Christian-Hinman CA, Alfonso J, Færgeman NJ. From benzodiazepines to fatty acids and beyond: revisiting the role of ACBP/DBI. Trends Endocrinol Metab 2021; 32:890-903. [PMID: 34565656 PMCID: PMC8785413 DOI: 10.1016/j.tem.2021.08.009] [Citation(s) in RCA: 4] [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: 07/02/2021] [Revised: 08/29/2021] [Accepted: 08/30/2021] [Indexed: 01/19/2023]
Abstract
Four decades ago Costa and colleagues identified a small, secreted polypeptide in the brain that can displace the benzodiazepine diazepam from the GABAA receptor, and was thus termed diazepam binding inhibitor (DBI). Shortly after, an identical polypeptide was identified in liver by its ability to induce termination of fatty acid synthesis, and was named acyl-CoA binding protein (ACBP). Since then, ACBP/DBI has been studied in parallel without a clear and integrated understanding of its dual roles. The first genetic loss-of-function models have revived the field, allowing targeted approaches to better understand the physiological roles of ACBP/DBI in vivo. We discuss the roles of ACBP/DBI in central and tissue-specific functions in mammals, with an emphasis on metabolism and mechanisms of action.
Collapse
Affiliation(s)
- Thierry Alquier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal Diabetes Research Center, and Departments of Medicine, Pharmacology and Physiology, Biochemistry, and Neurosciences, Université de Montréal, Montreal, QC, Canada.
| | - Catherine A Christian-Hinman
- Department of Molecular and Integrative Physiology, Neuroscience Program, Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL 61801, USA
| | - Julieta Alfonso
- Department of Clinical Neurobiology, University Hospital Heidelberg and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Nils J Færgeman
- Department of Biochemistry and Molecular Biology, Villum Center for Bioanalytical Sciences, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark.
| |
Collapse
|
11
|
Gobbo D, Scheller A, Kirchhoff F. From Physiology to Pathology of Cortico-Thalamo-Cortical Oscillations: Astroglia as a Target for Further Research. Front Neurol 2021; 12:661408. [PMID: 34177766 PMCID: PMC8219957 DOI: 10.3389/fneur.2021.661408] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 05/11/2021] [Indexed: 12/21/2022] Open
Abstract
The electrographic hallmark of childhood absence epilepsy (CAE) and other idiopathic forms of epilepsy are 2.5-4 Hz spike and wave discharges (SWDs) originating from abnormal electrical oscillations of the cortico-thalamo-cortical network. SWDs are generally associated with sudden and brief non-convulsive epileptic events mostly generating impairment of consciousness and correlating with attention and learning as well as cognitive deficits. To date, SWDs are known to arise from locally restricted imbalances of excitation and inhibition in the deep layers of the primary somatosensory cortex. SWDs propagate to the mostly GABAergic nucleus reticularis thalami (NRT) and the somatosensory thalamic nuclei that project back to the cortex, leading to the typical generalized spike and wave oscillations. Given their shared anatomical basis, SWDs have been originally considered the pathological transition of 11-16 Hz bursts of neural oscillatory activity (the so-called sleep spindles) occurring during Non-Rapid Eye Movement (NREM) sleep, but more recent research revealed fundamental functional differences between sleep spindles and SWDs, suggesting the latter could be more closely related to the slow (<1 Hz) oscillations alternating active (Up) and silent (Down) cortical activity and concomitantly occurring during NREM. Indeed, several lines of evidence support the fact that SWDs impair sleep architecture as well as sleep/wake cycles and sleep pressure, which, in turn, affect seizure circadian frequency and distribution. Given the accumulating evidence on the role of astroglia in the field of epilepsy in the modulation of excitation and inhibition in the brain as well as on the development of aberrant synchronous network activity, we aim at pointing at putative contributions of astrocytes to the physiology of slow-wave sleep and to the pathology of SWDs. Particularly, we will address the astroglial functions known to be involved in the control of network excitability and synchronicity and so far mainly addressed in the context of convulsive seizures, namely (i) interstitial fluid homeostasis, (ii) K+ clearance and neurotransmitter uptake from the extracellular space and the synaptic cleft, (iii) gap junction mechanical and functional coupling as well as hemichannel function, (iv) gliotransmission, (v) astroglial Ca2+ signaling and downstream effectors, (vi) reactive astrogliosis and cytokine release.
Collapse
Affiliation(s)
- Davide Gobbo
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Anja Scheller
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| |
Collapse
|
12
|
Yamagata K. Astrocyte-induced synapse formation and ischemic stroke. J Neurosci Res 2021; 99:1401-1413. [PMID: 33604930 DOI: 10.1002/jnr.24807] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 01/26/2021] [Indexed: 12/16/2022]
Abstract
Astrocytes are closely associated with the regulation of synapse formation and function. In addition, astrocytes have been shown to block certain brain impairments, including synaptic damage from stroke and other diseases of the central nervous system (CNS). Although astrocytes do not completely prevent synaptic damage, they appear to be protective and to restore synaptic function following damage. The purpose of this study is to discuss the role of astrocytes in synaptogenesis and synaptic damage in ischemic stroke. I detail the mechanism of action of the multiple factors secreted by astrocytes that are involved in synapse formation. In particular, I describe the characteristics and role in synapse formation of each secreted molecule related to synaptic structure and function. Furthermore, I discuss the effect of astrocytes on synaptogenesis and repair in ischemic stroke and in other CNS diseases. Astrocytes release molecules such as thrombospondin, hevin, secreted protein acidic rich in cysteine, etc., due to activation by ischemia to induce synaptic structure and function, an effect associated with protection of the brain from synaptic damage in ischemic stroke. In conclusion, I show that astrocytes may regulate synaptic transmission while having the potential to block and repair synaptic dysfunction in stroke-associated brain damage.
Collapse
Affiliation(s)
- Kazuo Yamagata
- Department of Food Bioscience & Biotechnology, College of Bioresource Science, Nihon University (UNBS), Fujisawa, Japan
| |
Collapse
|
13
|
Kaczor PT, Wolska AD, Mozrzymas JW. α 1 Subunit Histidine 55 at the Interface between Extracellular and Transmembrane Domains Affects Preactivation and Desensitization of the GABA A Receptor. ACS Chem Neurosci 2021; 12:562-572. [PMID: 33471498 PMCID: PMC7875458 DOI: 10.1021/acschemneuro.0c00781] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
![]()
The
GABAA receptor is a member of the Cys-loop family
and plays a crucial role in the adult mammalian brain inhibition.
Although the static structure of this receptor is emerging, the molecular
mechanisms underlying its conformational transitions remain elusive.
It is known that in the Cys-loop receptors, the interface between
extracellular and transmembrane domains plays a key role in transmitting
the “activation wave” down to the channel gate in the
pore. It has been previously reported that histidine 55 (H55), located
centrally at the interfacial β1−β2 loop of the
α1 subunit, is important in the receptor activation,
but it is unknown which specific gating steps it is affecting. In
the present study, we addressed this issue by taking advantage of
the state-of-the-art macroscopic and single-channel recordings together
with extensive modeling. Considering that H55 is known to affect the
local electrostatic landscape and because it is neighbored by two
negatively charged aspartates, a well conserved feature in the α
subunits, we considered substitution with negative (E) and positive
(K) residues. We found that these mutations markedly affected the
receptor gating, altering primarily preactivation and desensitization
transitions. Importantly, opposite effects were observed for these
two mutations strongly suggesting involvement of electrostatic interactions.
Single-channel recordings suggested also a minor effect on opening/closing
transitions which did not depend on the electric charge of the substituting
amino acid. Altogether, we demonstrate that H55 mutations affect primarily
preactivation and desensitization most likely by influencing local
electrostatic interactions at the receptor interface.
Collapse
Affiliation(s)
- Przemyslaw T. Kaczor
- Department of Biophysics and Neuroscience, Wroclaw Medical University, Chalubinskiego 3a, Wroclaw, Dolnoślaskie 50-368, Poland
| | - Aleksandra D. Wolska
- Department of Biophysics and Neuroscience, Wroclaw Medical University, Chalubinskiego 3a, Wroclaw, Dolnoślaskie 50-368, Poland
| | - Jerzy W. Mozrzymas
- Department of Biophysics and Neuroscience, Wroclaw Medical University, Chalubinskiego 3a, Wroclaw, Dolnoślaskie 50-368, Poland
| |
Collapse
|
14
|
Frost HR. Variance-adjusted Mahalanobis (VAM): a fast and accurate method for cell-specific gene set scoring. Nucleic Acids Res 2020; 48:e94. [PMID: 32633778 PMCID: PMC7498348 DOI: 10.1093/nar/gkaa582] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/10/2020] [Accepted: 06/26/2020] [Indexed: 12/26/2022] Open
Abstract
Statistical analysis of single cell RNA-sequencing (scRNA-seq) data is hindered by high levels of technical noise and inflated zero counts. One promising approach for addressing these challenges is gene set testing, or pathway analysis, which can mitigate sparsity and noise, and improve interpretation and power, by aggregating expression data to the pathway level. Unfortunately, methods optimized for bulk transcriptomics perform poorly on scRNA-seq data and progress on single cell-specific techniques has been limited. Importantly, no existing methods support cell-level gene set inference. To address this challenge, we developed a new gene set testing method, Variance-adjusted Mahalanobis (VAM), that integrates with the Seurat framework and can accommodate the technical noise, sparsity and large sample sizes characteristic of scRNA-seq data. The VAM method computes cell-specific pathway scores to transform a cell-by-gene matrix into a cell-by-pathway matrix that can be used for both data visualization and statistical enrichment analysis. Because the distribution of these scores under the null of uncorrelated technical noise has an accurate gamma approximation, both population and cell-level inference is supported. As demonstrated using simulated and real scRNA-seq data, the VAM method provides superior classification accuracy at a lower computation cost relative to existing single sample gene set testing approaches.
Collapse
Affiliation(s)
- Hildreth Robert Frost
- Department of Biomedical Data Science, Geisel School of Medicine, Dartmouth College, Hanover, NH 03755, USA
| |
Collapse
|
15
|
Tonon MC, Vaudry H, Chuquet J, Guillebaud F, Fan J, Masmoudi-Kouki O, Vaudry D, Lanfray D, Morin F, Prevot V, Papadopoulos V, Troadec JD, Leprince J. Endozepines and their receptors: Structure, functions and pathophysiological significance. Pharmacol Ther 2020; 208:107386. [DOI: 10.1016/j.pharmthera.2019.06.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 06/20/2019] [Indexed: 02/06/2023]
|
16
|
Abstract
Sleep spindles are burstlike signals in the electroencephalogram (EEG) of the sleeping mammalian brain and electrical surface correlates of neuronal oscillations in thalamus. As one of the most inheritable sleep EEG signatures, sleep spindles probably reflect the strength and malleability of thalamocortical circuits that underlie individual cognitive profiles. We review the characteristics, organization, regulation, and origins of sleep spindles and their implication in non-rapid-eye-movement sleep (NREMS) and its functions, focusing on human and rodent. Spatially, sleep spindle-related neuronal activity appears on scales ranging from small thalamic circuits to functional cortical areas, and generates a cortical state favoring intracortical plasticity while limiting cortical output. Temporally, sleep spindles are discrete events, part of a continuous power band, and elements grouped on an infraslow time scale over which NREMS alternates between continuity and fragility. We synthesize diverse and seemingly unlinked functions of sleep spindles for sleep architecture, sensory processing, synaptic plasticity, memory formation, and cognitive abilities into a unifying sleep spindle concept, according to which sleep spindles 1) generate neural conditions of large-scale functional connectivity and plasticity that outlast their appearance as discrete EEG events, 2) appear preferentially in thalamic circuits engaged in learning and attention-based experience during wakefulness, and 3) enable a selective reactivation and routing of wake-instated neuronal traces between brain areas such as hippocampus and cortex. Their fine spatiotemporal organization reflects NREMS as a physiological state coordinated over brain and body and may indicate, if not anticipate and ultimately differentiate, pathologies in sleep and neurodevelopmental, -degenerative, and -psychiatric conditions.
Collapse
Affiliation(s)
- Laura M J Fernandez
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Anita Lüthi
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
17
|
Ujjainwala AL, Courtney CD, Wojnowski NM, Rhodes JS, Christian CA. Differential impacts on multiple forms of spatial and contextual memory in diazepam binding inhibitor knockout mice. J Neurosci Res 2019; 97:683-697. [PMID: 30680776 DOI: 10.1002/jnr.24393] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 01/04/2019] [Accepted: 01/14/2019] [Indexed: 02/03/2023]
Abstract
Learning and memory are fundamental processes that are disrupted in many neurological disorders including Alzheimer's disease and epilepsy. The hippocampus plays an integral role in these functions, and modulation of synaptic transmission mediated by γ-aminobutyric acid (GABA) type-A receptors (GABAA Rs) impacts hippocampus-dependent learning and memory. The protein diazepam binding inhibitor (DBI) differentially modulates GABAA Rs in various brain regions, including hippocampus, and changes in DBI levels may be linked to altered learning and memory. The effects of genetic loss of DBI signaling on these processes, however, have not been determined. In these studies, we examined male and female constitutive DBI knockout mice and wild-type littermates to investigate the role of DBI signaling in modulating multiple forms of hippocampus-dependent spatial learning and memory. DBI knockout mice did not show impaired discrimination of objects in familiar and novel locations in an object location memory test, but did exhibit reduced time spent exploring the objects. Multiple parameters of Barnes maze performance, testing the capability to utilize spatial reference cues, were disrupted in DBI knockout mice. Furthermore, whereas most wild-type mice adopted a direct search strategy upon learning the location of the target hole, knockout mice showed higher rates of using an inefficient random strategy. In addition, DBI knockout mice displayed typical levels of contextual fear conditioning, but lacked a sex difference observed in wild-type mice. Together, these data suggest that DBI selectively influences certain forms of spatial learning and memory, indicating novel roles for DBI signaling in modulating hippocampus-dependent behavior in a task-specific manner.
Collapse
Affiliation(s)
- Ammar L Ujjainwala
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Connor D Courtney
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Natalia M Wojnowski
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Justin S Rhodes
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Department of Psychology, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Catherine A Christian
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
18
|
Courtney CD, Christian CA. Subregion-Specific Impacts of Genetic Loss of Diazepam Binding Inhibitor on Synaptic Inhibition in the Murine Hippocampus. Neuroscience 2018; 388:128-138. [PMID: 30031126 DOI: 10.1016/j.neuroscience.2018.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2018] [Revised: 06/27/2018] [Accepted: 07/09/2018] [Indexed: 11/16/2022]
Abstract
Benzodiazepines are commonly prescribed to treat neurological conditions including epilepsy, insomnia, and anxiety. The discovery of benzodiazepine-specific binding sites on γ-aminobutyric acid type-A receptors (GABAARs) led to the hypothesis that the brain may produce endogenous benzodiazepine-binding site ligands. An endogenous peptide, diazepam binding inhibitor (DBI), which can bind these sites, is thought to be capable of both enhancing and attenuating GABAergic transmission in different brain regions. However, the role that DBI plays in modulating GABAARs in the hippocampus remains unclear. Here, we investigated the role of DBI in modulating synaptic inhibition in the hippocampus using a constitutive DBI knockout mouse. Miniature and evoked inhibitory postsynaptic currents (mIPSCs, eIPSCs) were recorded from CA1 pyramidal cells and dentate gyrus (DG) granule cells. Loss of DBI signaling increased mIPSC frequency and amplitude in CA1 pyramidal cells from DBI knockout mice compared to wild-types. In DG granule cells, conversely, the loss of DBI decreased mIPSC amplitude and increased mIPSC decay time, indicating bidirectional modulation of GABAAR-mediated transmission in specific subregions of the hippocampus. eIPSC paired-pulse ratios were consistent across genotypes, suggesting that alterations in mIPSC frequency were not due to changes in presynaptic release probability. Furthermore, cells from DBI knockout mice did not display altered responsiveness to pharmacological applications of diazepam, a benzodiazepine, nor flumazenil, a benzodiazepine-binding site antagonist. These results provide evidence that genetic loss of DBI alters synaptic inhibition in the adult hippocampus, and that the direction of DBI-mediated modulation can vary discretely between specific subregions of the same brain structure.
Collapse
Affiliation(s)
- Connor D Courtney
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Catherine A Christian
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, IL, United States; Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, IL, United States; Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, United States.
| |
Collapse
|
19
|
Glial cells modulate hippocampal synaptic plasticity in morphine dependent rats. Brain Res Bull 2018; 140:97-106. [DOI: 10.1016/j.brainresbull.2018.04.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/28/2018] [Accepted: 04/11/2018] [Indexed: 12/24/2022]
|
20
|
|
21
|
Mariotti L, Losi G, Lia A, Melone M, Chiavegato A, Gómez-Gonzalo M, Sessolo M, Bovetti S, Forli A, Zonta M, Requie LM, Marcon I, Pugliese A, Viollet C, Bettler B, Fellin T, Conti F, Carmignoto G. Interneuron-specific signaling evokes distinctive somatostatin-mediated responses in adult cortical astrocytes. Nat Commun 2018; 9:82. [PMID: 29311610 PMCID: PMC5758790 DOI: 10.1038/s41467-017-02642-6] [Citation(s) in RCA: 84] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 12/14/2017] [Indexed: 12/25/2022] Open
Abstract
The signaling diversity of GABAergic interneurons to post-synaptic neurons is crucial to generate the functional heterogeneity that characterizes brain circuits. Whether this diversity applies to other brain cells, such as the glial cells astrocytes, remains unexplored. Using optogenetics and two-photon functional imaging in the adult mouse neocortex, we here reveal that parvalbumin- and somatostatin-expressing interneurons, two key interneuron classes in the brain, differentially signal to astrocytes inducing weak and robust GABAB receptor-mediated Ca2+ elevations, respectively. Furthermore, the astrocyte response depresses upon parvalbumin interneuron repetitive stimulations and potentiates upon somatostatin interneuron repetitive stimulations, revealing a distinguished astrocyte plasticity. Remarkably, the potentiated response crucially depends on the neuropeptide somatostatin, released by somatostatin interneurons, which activates somatostatin receptors at astrocytic processes. Our study unveils, in the living brain, a hitherto unidentified signaling specificity between interneuron subtypes and astrocytes opening a new perspective into the role of astrocytes as non-neuronal components of inhibitory circuits.
Collapse
Affiliation(s)
- Letizia Mariotti
- Neuroscience Institute, National Research Council (CNR), 35121, Padova, Italy.,Department of Biomedical Sciences, Università degli Studi di Padova, 35121, Padova, Italy
| | - Gabriele Losi
- Neuroscience Institute, National Research Council (CNR), 35121, Padova, Italy.,Department of Biomedical Sciences, Università degli Studi di Padova, 35121, Padova, Italy
| | - Annamaria Lia
- Neuroscience Institute, National Research Council (CNR), 35121, Padova, Italy.,Department of Biomedical Sciences, Università degli Studi di Padova, 35121, Padova, Italy
| | - Marcello Melone
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy.,Center for Neurobiology of Aging, INRCA IRCCS, 60121, Ancona, Italy
| | - Angela Chiavegato
- Department of Biomedical Sciences, Università degli Studi di Padova, 35121, Padova, Italy
| | - Marta Gómez-Gonzalo
- Neuroscience Institute, National Research Council (CNR), 35121, Padova, Italy.,Department of Biomedical Sciences, Università degli Studi di Padova, 35121, Padova, Italy
| | - Michele Sessolo
- Neuroscience Institute, National Research Council (CNR), 35121, Padova, Italy.,Department of Biomedical Sciences, Università degli Studi di Padova, 35121, Padova, Italy
| | - Serena Bovetti
- Optical Approches to Brain Function Laboratory, Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, 16163, Genova, Italy
| | - Angelo Forli
- Optical Approches to Brain Function Laboratory, Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, 16163, Genova, Italy
| | - Micaela Zonta
- Neuroscience Institute, National Research Council (CNR), 35121, Padova, Italy.,Department of Biomedical Sciences, Università degli Studi di Padova, 35121, Padova, Italy
| | - Linda Maria Requie
- Neuroscience Institute, National Research Council (CNR), 35121, Padova, Italy.,Department of Biomedical Sciences, Università degli Studi di Padova, 35121, Padova, Italy
| | - Iacopo Marcon
- Neuroscience Institute, National Research Council (CNR), 35121, Padova, Italy.,Department of Biomedical Sciences, Università degli Studi di Padova, 35121, Padova, Italy
| | - Arianna Pugliese
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy
| | - Cécile Viollet
- Inserm UMR894, Center for Psychiatry and Neuroscience, Université Paris-Descartes, 75014, Paris, France
| | - Bernhard Bettler
- Departement of Biomedicine, University of Basel, 4031, Basel, Switzerland
| | - Tommaso Fellin
- Optical Approches to Brain Function Laboratory, Department of Neuroscience and Brain Technologies, Istituto Italiano di Tecnologia, 16163, Genova, Italy
| | - Fiorenzo Conti
- Department of Experimental and Clinical Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy.,Center for Neurobiology of Aging, INRCA IRCCS, 60121, Ancona, Italy.,Foundation for Molecular Medicine, Università Politecnica delle Marche, 60126, Ancona, Italy
| | - Giorgio Carmignoto
- Neuroscience Institute, National Research Council (CNR), 35121, Padova, Italy. .,Department of Biomedical Sciences, Università degli Studi di Padova, 35121, Padova, Italy.
| |
Collapse
|
22
|
Abstract
Astrocytes are neural cells of ectodermal, neuroepithelial origin that provide for homeostasis and defense of the central nervous system (CNS). Astrocytes are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters. This complement underlies their remarkable adaptive plasticity that defines the functional maintenance of the CNS in development and aging. Astrocytes are tightly integrated into neural networks and act within the context of neural tissue; astrocytes control homeostasis of the CNS at all levels of organization from molecular to the whole organ.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| | - Maiken Nedergaard
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| |
Collapse
|
23
|
Verkhratsky A, Nedergaard M. Physiology of Astroglia. Physiol Rev 2018; 98:239-389. [PMID: 29351512 PMCID: PMC6050349 DOI: 10.1152/physrev.00042.2016] [Citation(s) in RCA: 1068] [Impact Index Per Article: 152.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 03/22/2017] [Accepted: 04/27/2017] [Indexed: 02/07/2023] Open
Abstract
Astrocytes are neural cells of ectodermal, neuroepithelial origin that provide for homeostasis and defense of the central nervous system (CNS). Astrocytes are highly heterogeneous in morphological appearance; they express a multitude of receptors, channels, and membrane transporters. This complement underlies their remarkable adaptive plasticity that defines the functional maintenance of the CNS in development and aging. Astrocytes are tightly integrated into neural networks and act within the context of neural tissue; astrocytes control homeostasis of the CNS at all levels of organization from molecular to the whole organ.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| | - Maiken Nedergaard
- The University of Manchester , Manchester , United Kingdom ; Achúcarro Basque Center for Neuroscience, IKERBASQUE, Basque Foundation for Science , Bilbao , Spain ; Department of Neuroscience, University of the Basque Country UPV/EHU and CIBERNED, Leioa, Spain ; Center for Basic and Translational Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen , Copenhagen , Denmark ; and Center for Translational Neuromedicine, University of Rochester Medical Center , Rochester, New York
| |
Collapse
|
24
|
Ujjainwala AL, Courtney CD, Rhoads SG, Rhodes JS, Christian CA. Genetic loss of diazepam binding inhibitor in mice impairs social interest. GENES BRAIN AND BEHAVIOR 2017; 17:e12442. [PMID: 29193847 DOI: 10.1111/gbb.12442] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Revised: 11/20/2017] [Accepted: 11/23/2017] [Indexed: 01/21/2023]
Abstract
Neuropsychiatric disorders in which reduced social interest is a common symptom, such as autism, depression, and anxiety, are frequently associated with genetic mutations affecting γ-aminobutyric acid (GABA)ergic transmission. Benzodiazepine treatment, acting via GABA type-A receptors, improves social interaction in male mouse models with autism-like features. The protein diazepam binding inhibitor (DBI) can act as an endogenous benzodiazepine, but a role for DBI in social behavior has not been described. Here, we investigated the role of DBI in the social interest and recognition behavior of mice. The responses of DBI wild-type and knockout male and female mice to ovariectomized female wild-type mice (a neutral social stimulus) were evaluated in a habituation/dishabituation task. Both male and female knockout mice exhibited reduced social interest, and DBI knockout mice lacked the sex difference in social interest levels observed in wild-type mice, in which males showed higher social interest levels than females. The ability to discriminate between familiar and novel stimulus mice (social recognition) was not impaired in DBI-deficient mice of either sex. DBI knockouts could learn a rotarod motor task, and could discriminate between social and nonsocial odors. Both sexes of DBI knockout mice showed increased repetitive grooming behavior, but not in a manner that would account for the decrease in social investigation time. Genetic loss of DBI did not alter seminal vesicle weight, indicating that the social interest phenotype of males lacking DBI is not due to reduced circulating testosterone. Together, these studies show a novel role of DBI in driving social interest and motivation.
Collapse
Affiliation(s)
- A L Ujjainwala
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - C D Courtney
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - S G Rhoads
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - J S Rhodes
- Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Department of Psychology, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - C A Christian
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Neuroscience Program, University of Illinois at Urbana-Champaign, Urbana, Illinois.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
25
|
Um JW. Roles of Glial Cells in Sculpting Inhibitory Synapses and Neural Circuits. Front Mol Neurosci 2017; 10:381. [PMID: 29180953 PMCID: PMC5694142 DOI: 10.3389/fnmol.2017.00381] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 11/01/2017] [Indexed: 01/04/2023] Open
Abstract
Glial cells are essential for every aspect of normal neuronal development, synapse formation, and function in the central nervous system (CNS). Astrocytes secrete a variety of factors that regulate synaptic connectivity and circuit formation. Microglia also modulate synapse development through phagocytic activity. Most of the known actions of CNS glial cells are limited to roles at excitatory synapses. Nevertheless, studies have indicated that both astrocytes and microglia shape inhibitory synaptic connections through various mechanisms, including release of regulatory molecules, direct contact with synaptic terminals, and utilization of mediators in the extracellular matrix. This review summarizes recent investigations into the mechanisms underlying CNS glial cell-mediated inhibitory synapse development.
Collapse
Affiliation(s)
- Ji Won Um
- Department of Brain and Cognitive Sciences, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, South Korea
| |
Collapse
|
26
|
Fogerson PM, Huguenard JR. Tapping the Brakes: Cellular and Synaptic Mechanisms that Regulate Thalamic Oscillations. Neuron 2017; 92:687-704. [PMID: 27883901 DOI: 10.1016/j.neuron.2016.10.024] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 10/03/2016] [Accepted: 10/10/2016] [Indexed: 12/26/2022]
Abstract
Thalamic oscillators contribute to both normal rhythms associated with sleep and anesthesia and abnormal, hypersynchronous oscillations that manifest behaviorally as absence seizures. In this review, we highlight new findings that refine thalamic contributions to cortical rhythms and suggest that thalamic oscillators may be subject to both local and global control. We describe endogenous thalamic mechanisms that limit network synchrony and discuss how these protective brakes might be restored to prevent absence seizures. Finally, we describe how intrinsic and circuit-level specializations among thalamocortical loops may determine their involvement in widespread oscillations and render subsets of thalamic nuclei especially vulnerable to pathological synchrony.
Collapse
Affiliation(s)
- P Michelle Fogerson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - John R Huguenard
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
27
|
Kaczor PT, Mozrzymas JW. Key Metabolic Enzymes Underlying Astrocytic Upregulation of GABAergic Plasticity. Front Cell Neurosci 2017; 11:144. [PMID: 28559800 PMCID: PMC5432623 DOI: 10.3389/fncel.2017.00144] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/02/2017] [Indexed: 11/30/2022] Open
Abstract
GABAergic plasticity is recognized as a key mechanism of shaping the activity of the neuronal networks. However, its description is challenging because of numerous neuron-specific mechanisms. In particular, while essential role of glial cells in the excitatory plasticity is well established, their involvement in GABAergic plasticity only starts to emerge. To address this problem, we used two models: neuronal cell culture (NC) and astrocyte-neuronal co-culture (ANCC), where we chemically induced long-term potentiation at inhibitory synapses (iLTP). iLTP could be induced both in NC and ANCC but in ANCC its extent was larger. Importantly, this functional iLTP manifestation was accompanied by an increase in gephyrin puncta size. Furthermore, blocking astrocyte Krebs cycle with fluoroacetate (FA) in ANCC prevented enhancement of both mIPSC amplitude and gephyrin puncta size but this effect was not observed in NC, indicating a key role in neuron-astrocyte cross-talk. Blockade of monocarboxylate transport with α-Cyano-4-hydroxycinnamic acid (4CIN) abolished iLTP both in NC and ANCC and in the latter model prevented also enlargement of gephyrin puncta. Similarly, blockade of glycogen phosphorylase with BAYU6751 prevented enlargement of gephyrin puncta upon iLTP induction. Finally, block of glutamine synthetase with methionine sulfoxide (MSO) nearly abolished mIPSC increase in both NMDA stimulated cell groups but did not prevent enlargement of gephyrin puncta. In conclusion, we provide further evidence that GABAergic plasticity is strongly regulated by astrocytes and the underlying mechanisms involve key metabolic enzymes. Considering the strategic role of GABAergic interneurons, the plasticity described here indicates possible mechanism whereby metabolism regulates the network activity.
Collapse
Affiliation(s)
- Przemysław T Kaczor
- Department of Molecular Physiology and Neurobiology, Faculty of Biological Sciences, University of WrocławWrocław, Poland
| | - Jerzy W Mozrzymas
- Department of Molecular Physiology and Neurobiology, Faculty of Biological Sciences, University of WrocławWrocław, Poland.,Laboratory of Neuroscience, Department of Biophysics, Wrocław Medical UniversityWrocław, Poland
| |
Collapse
|
28
|
Colonna M, Butovsky O. Microglia Function in the Central Nervous System During Health and Neurodegeneration. Annu Rev Immunol 2017; 35:441-468. [PMID: 28226226 DOI: 10.1146/annurev-immunol-051116-052358] [Citation(s) in RCA: 1672] [Impact Index Per Article: 209.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Microglia are resident cells of the brain that regulate brain development, maintenance of neuronal networks, and injury repair. Microglia serve as brain macrophages but are distinct from other tissue macrophages owing to their unique homeostatic phenotype and tight regulation by the central nervous system (CNS) microenvironment. They are responsible for the elimination of microbes, dead cells, redundant synapses, protein aggregates, and other particulate and soluble antigens that may endanger the CNS. Furthermore, as the primary source of proinflammatory cytokines, microglia are pivotal mediators of neuroinflammation and can induce or modulate a broad spectrum of cellular responses. Alterations in microglia functionality are implicated in brain development and aging, as well as in neurodegeneration. Recent observations about microglia ontogeny combined with extensive gene expression profiling and novel tools to study microglia biology have allowed us to characterize the spectrum of microglial phenotypes during development, homeostasis, and disease. In this article, we review recent advances in our understanding of the biology of microglia, their contribution to homeostasis, and their involvement in neurodegeneration. Moreover, we highlight the complexity of targeting microglia for therapeutic intervention in neurodegenerative diseases.
Collapse
Affiliation(s)
- Marco Colonna
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110;
| | - Oleg Butovsky
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115;
| |
Collapse
|
29
|
Lafourcade C, Ramírez JP, Luarte A, Fernández A, Wyneken U. MiRNAs in Astrocyte-Derived Exosomes as Possible Mediators of Neuronal Plasticity. J Exp Neurosci 2016; 10:1-9. [PMID: 27547038 PMCID: PMC4978198 DOI: 10.4137/jen.s39916] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 07/07/2016] [Accepted: 07/09/2016] [Indexed: 12/21/2022] Open
Abstract
Astrocytes use gliotransmitters to modulate neuronal function and plasticity. However, the role of small extracellular vesicles, called exosomes, in astrocyte-to-neuron signaling is mostly unknown. Exosomes originate in multivesicular bodies of parent cells and are secreted by fusion of the multivesicular body limiting membrane with the plasma membrane. Their molecular cargo, consisting of RNA species, proteins, and lipids, is in part cell type and cell state specific. Among the RNA species transported by exosomes, microRNAs (miRNAs) are able to modify gene expression in recipient cells. Several miRNAs present in astrocytes are regulated under pathological conditions, and this may have far-reaching consequences if they are loaded in exosomes. We propose that astrocyte-derived miRNA-loaded exosomes, such as miR-26a, are dysregulated in several central nervous system diseases; thus potentially controlling neuronal morphology and synaptic transmission through validated and predicted targets. Unraveling the contribution of this new signaling mechanism to the maintenance and plasticity of neuronal networks will impact our understanding on the physiology and pathophysiology of the central nervous system.
Collapse
Affiliation(s)
- Carlos Lafourcade
- Centro de Investigaciones Biomédicas, Facultad de Medicina, Universidad de los Andes, Chile
| | - Juan Pablo Ramírez
- Centro de Investigaciones Biomédicas, Facultad de Medicina, Universidad de los Andes, Chile
| | - Alejandro Luarte
- Centro de Investigaciones Biomédicas, Facultad de Medicina, Universidad de los Andes, Chile
| | - Anllely Fernández
- Centro de Investigaciones Biomédicas, Facultad de Medicina, Universidad de los Andes, Chile
| | - Ursula Wyneken
- Centro de Investigaciones Biomédicas, Facultad de Medicina, Universidad de los Andes, Chile
| |
Collapse
|
30
|
Budry L, Bouyakdan K, Tobin S, Rodaros D, Marcher AB, Mandrup S, Fulton S, Alquier T. DBI/ACBP loss-of-function does not affect anxiety-like behaviour but reduces anxiolytic responses to diazepam in mice. Behav Brain Res 2016; 313:201-207. [PMID: 27363924 DOI: 10.1016/j.bbr.2016.06.052] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 06/13/2016] [Accepted: 06/14/2016] [Indexed: 11/18/2022]
Abstract
Diazepam is well known for its anxiolytic properties, which are mediated via activation of the GABAA receptor. Diazepam Binding Inhibitor (DBI), also called acyl-CoA binding protein (ACBP), is a ubiquitously expressed protein originally identified based on its ability to displace diazepam from its binding site on the GABAA receptor. Central administration of ACBP or its cleaved fragment, commonly referred to as endozepines, induces proconflict and anxiety-like behaviour in rodents. For this reason, ACBP is known as an anxiogenic peptide. However, the role of endogenous ACBP in anxiety-like behaviour and anxiolytic responses to diazepam has not been investigated. To address this question, we assessed anxiety behaviour and anxiolytic responses to diazepam in two complementary loss-of-function mouse models including astrocyte-specific ACBP KO (ACBP(GFAP) KO) and whole-body KO (ACBP KO) mice. Male and female ACBP(GFAP) KO and ACBP KO mice do not show significant changes in anxiety-like behaviour compared to control littermates during elevated plus maze (EPM) and open field (OF) tests. Surprisingly, ACBP(GFAP) KO and ACBP KO mice were unresponsive to the anxiolytic effect of a low dose of diazepam during EPM tests. In conclusion, our experiments using genetic ACBP loss-of-function models suggest that endozepines deficiency does not affect anxiety-like behaviour in mice and impairs the anxiolytic action of diazepam.
Collapse
Affiliation(s)
- Lionel Budry
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Universite de Montreal (CRCHUM), University of Montreal, Montreal, QC, H3T 1J4, Canada; Department of Medicine, University of Montreal, Montreal, QC, H3T 1J4, Canada
| | - Khalil Bouyakdan
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Universite de Montreal (CRCHUM), University of Montreal, Montreal, QC, H3T 1J4, Canada; Department of Biochemistry, University of Montreal, Montreal, QC, H3T 1J4, Canada
| | - Stephanie Tobin
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Universite de Montreal (CRCHUM), University of Montreal, Montreal, QC, H3T 1J4, Canada; Department of Medicine, University of Montreal, Montreal, QC, H3T 1J4, Canada
| | - Demetra Rodaros
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Universite de Montreal (CRCHUM), University of Montreal, Montreal, QC, H3T 1J4, Canada
| | - Ann-Britt Marcher
- Departments of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Susanne Mandrup
- Departments of Biochemistry and Molecular Biology, University of Southern Denmark, DK-5230 Odense M, Denmark
| | - Stephanie Fulton
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Universite de Montreal (CRCHUM), University of Montreal, Montreal, QC, H3T 1J4, Canada; Department of Nutrition, University of Montreal, Montreal, QC, H3T 1J4, Canada
| | - Thierry Alquier
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Universite de Montreal (CRCHUM), University of Montreal, Montreal, QC, H3T 1J4, Canada; Department of Medicine, University of Montreal, Montreal, QC, H3T 1J4, Canada; Department of Biochemistry, University of Montreal, Montreal, QC, H3T 1J4, Canada; Departments of Pathology and Cell Biology, University of Montreal, Montreal, QC, H3T 1J4, Canada.
| |
Collapse
|
31
|
Colavita M, Terral G, Lemercier CE, Drago F, Marsicano G, Massa F. Layer-specific potentiation of network GABAergic inhibition in the CA1 area of the hippocampus. Sci Rep 2016; 6:28454. [PMID: 27345695 PMCID: PMC4921906 DOI: 10.1038/srep28454] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 06/03/2016] [Indexed: 12/04/2022] Open
Abstract
One of the most important functions of GABAergic inhibition in cortical regions is the tight control of spatiotemporal activity of principal neuronal ensembles. However, electrophysiological recordings do not provide sufficient spatial information to determine the spatiotemporal properties of inhibitory plasticity. Using Voltage Sensitive Dye Imaging (VSDI) in mouse hippocampal slices, we demonstrate that GABAA-mediated field inhibitory postsynaptic potentials undergo layer-specific potentiation upon activation of metabotropic glutamate receptors (mGlu). VSDI recordings allowed detection of pharmacologically isolated GABAA-dependent hyperpolarization signals. Bath-application of the selective group-I mGlu receptor agonist, (S)-3,5-Dihydroxyphenylglycine (DHPG), induces an enhancement of the GABAergic VSDI-recorded signal, which is more or less pronounced in different hippocampal layers. This potentiation is mediated by mGlu5 and downstream activation of IP3 receptors. Our results depict network GABAergic activity in the hippocampal CA1 region and its sub-layers, showing also a novel form of inhibitory synaptic plasticity tightly coupled to glutamatergic activity.
Collapse
Affiliation(s)
- Michelangelo Colavita
- INSERM U1215, NeuroCentre Magendie, AVENIR Group "Endocannabinoids and Neuroadaptation", 33077 Bordeaux, France.,Université de Bordeaux, 33077 Bordeaux, France.,University of Catania, Biometec - Department of Biomedical and Biotechnological Sciences, 95125 Catania, Italy
| | - Geoffrey Terral
- INSERM U1215, NeuroCentre Magendie, AVENIR Group "Endocannabinoids and Neuroadaptation", 33077 Bordeaux, France.,Université de Bordeaux, 33077 Bordeaux, France
| | - Clement E Lemercier
- INSERM U1215, NeuroCentre Magendie, AVENIR Group "Endocannabinoids and Neuroadaptation", 33077 Bordeaux, France.,Université de Bordeaux, 33077 Bordeaux, France
| | - Filippo Drago
- University of Catania, Biometec - Department of Biomedical and Biotechnological Sciences, 95125 Catania, Italy
| | - Giovanni Marsicano
- INSERM U1215, NeuroCentre Magendie, AVENIR Group "Endocannabinoids and Neuroadaptation", 33077 Bordeaux, France.,Université de Bordeaux, 33077 Bordeaux, France
| | - Federico Massa
- INSERM U1215, NeuroCentre Magendie, AVENIR Group "Endocannabinoids and Neuroadaptation", 33077 Bordeaux, France.,Université de Bordeaux, 33077 Bordeaux, France
| |
Collapse
|
32
|
Yang H, Wang Y, Zhang Y, Zhang Y, Xu MS, Yuan J, Yu T. Astrocytes contribute to the effects of etomidate on synaptic transmission in rat primary somatosensory cortex. Brain Res 2016; 1642:238-243. [PMID: 27045115 DOI: 10.1016/j.brainres.2016.03.052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 03/26/2016] [Accepted: 03/31/2016] [Indexed: 11/27/2022]
Abstract
Little is known about the mechanisms of unconsciousness induced by general anesthetics. Previous studies have shown that the primary somatosensory cortex (S1) is a sensitive region to a variety of intravenous general anesthetics. Etomidate is a widely used intravenous anesthetic that can influence synaptic transmission. Recently, there are some evidences suggesting that astrocytes, a type of glia cell, also contribute to information transmission in the brain, and modulate synaptic function by releasing neuroactive substances. However, it is unknown whether astrocytes influence the effects of etomidate on information transmission in S1 pyramidal neurons. In the present study, the role of astrocytes in etomidate-induced unconsciousness was investigated by using the whole-cell patch clamp technique. We observed etomidate at clinically relevant concentrations inhibited the spontaneous postsynaptic currents (sPSCs) of rat S1 pyramidal neurons in a concentration-dependent manner, and the EC50 value of etomidate for inhibiting sPSCs from the concentration-effect curve was 6.9μM. Furthermore, in the presence of fluorocitrate, a glia-selective metabolism inhibitor that blocks the aconitase enzyme, both the amplitude and frequency of sPSCs in rat S1 pyramidal neurons were reduced, and the inhibitory effects of etomidate on sPSCs amplitude was strengthened without affecting the effects of etomidate on frequency. From these data, we deduce that etomidate suppresses synaptic activity via presynaptic and postsynaptic components. Furthermore, astrocytes participate in synaptic transmission and influence the effects of etomidate on postsynaptic receptors. This study provides new insight into the role of astrocytes in etomidate-induced unconsciousness.
Collapse
Affiliation(s)
- Hao Yang
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Dalian Road, 563003 Zunyi, Guizhou, China
| | - Yuan Wang
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Dalian Road, 563003 Zunyi, Guizhou, China; Department of Anesthesiology, Affiliated Hospital of Zunyi Medical College, Dalian Road, 563003 Zunyi, Guizhou, China
| | - Yu Zhang
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Dalian Road, 563003 Zunyi, Guizhou, China; Department of Anesthesiology, Affiliated Hospital of Zunyi Medical College, Dalian Road, 563003 Zunyi, Guizhou, China
| | - You Zhang
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Dalian Road, 563003 Zunyi, Guizhou, China
| | - Mao-Sheng Xu
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Dalian Road, 563003 Zunyi, Guizhou, China
| | - Jie Yuan
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Dalian Road, 563003 Zunyi, Guizhou, China
| | - Tian Yu
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical College, Dalian Road, 563003 Zunyi, Guizhou, China; Department of Anesthesiology, Affiliated Hospital of Zunyi Medical College, Dalian Road, 563003 Zunyi, Guizhou, China.
| |
Collapse
|
33
|
Bender CL, Calfa GD, Molina VA. Astrocyte plasticity induced by emotional stress: A new partner in psychiatric physiopathology? Prog Neuropsychopharmacol Biol Psychiatry 2016; 65:68-77. [PMID: 26320029 DOI: 10.1016/j.pnpbp.2015.08.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 08/07/2015] [Accepted: 08/08/2015] [Indexed: 01/18/2023]
Abstract
A growing body of evidence has demonstrated that astrocytes play a pivotal role in the normal functioning of the nervous system. This new conceptual framework has set the groundwork to be able to hypothesize that astrocytes could underlie signs and symptoms of mental diseases. Stress is a major risk factor in the etiology of several psychiatric diseases, such as anxiety disorders and depression. Hence, understanding the effects of stress on astrocytes and how these changes contribute to the development of psychiatric endophenotypes is crucial for both a better comprehension of mental illness and for potential targeted treatment of stress-related mental disorders. Here, we describe the currently used approaches and recent evidence showing astrocyte alterations induced by chronic and acute stress in animals. In addition, the relevance of these changes in stress-induced behavioral sequelae and human data linking astrocytes with neuropsychiatric disorders related to stress are also discussed. All together, the data indicate that astrocytes are also an important target of stress, with both chronic and acute stressors being able to alter the morphology or the expression of several astrocyte specific proteins in brain areas that are known to play a critical role in emotional processing, such as the prefrontal cortex, hippocampus and amygdala. Furthermore, different lines of evidences suggest that these changes may contribute, at less in part, to the behavioral consequences of stress.
Collapse
Affiliation(s)
- Crhistian L Bender
- IFEC-CONICET, Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, (5000) Córdoba, Argentina.
| | - Gaston D Calfa
- IFEC-CONICET, Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, (5000) Córdoba, Argentina
| | - Victor A Molina
- IFEC-CONICET, Departamento de Farmacología, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, (5000) Córdoba, Argentina
| |
Collapse
|
34
|
Peng HZ, Ma LX, Lv MH, Hu T, Liu T. Minocycline enhances inhibitory transmission to substantia gelatinosa neurons of the rat spinal dorsal horn. Neuroscience 2016; 319:183-93. [PMID: 26826332 DOI: 10.1016/j.neuroscience.2016.01.047] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 01/02/2016] [Accepted: 01/21/2016] [Indexed: 12/13/2022]
Abstract
Minocycline, a second-generation tetracycline, is well known for its antibiotic, anti-inflammatory, and antinociceptive effects. Modulation of synaptic transmission is one of the analgesic mechanisms of minocycline. Although it has been reported that minocycline may suppress excitatory glutamatergic synaptic transmission, it remains unclear whether it could affect inhibitory synaptic transmission, which also plays a key role in modulating pain signaling. To examine the effect of minocycline on synaptic transmission in rat spinal substantia gelatinosa (SG) neurons, we recorded spontaneous inhibitory postsynaptic currents (sIPSCs) using whole-cell patch-clamp recording at a holding potential of 0 mV. Bath application of minocycline significantly increased the frequency but not the amplitude of sIPSCs in a reversible and concentration-dependent manner with an EC50 of 85. The enhancement of inhibitory synaptic transmission produced by minocycline was not affected by the glutamate receptor antagonists CNQX and D-APV or by the voltage-gated sodium channel blocker tetrodotoxin (TTX). Moreover, the potency of minocycline for facilitating sIPSC frequency was the same in both glycinergic and GABAergic sIPSCs without changing their decay phases. However, the facilitatory effect of minocycline on sIPSCs was eliminated in a Ca(2+)-free Krebs solution or by co-administration with calcium channel blockers. In summary, our data demonstrate that baseline inhibitory synaptic transmission in SG neurons is markedly enhanced by minocycline. This may function to decrease the excitability of SG neurons, thus leading to a modulation of nociceptive transmission.
Collapse
Affiliation(s)
- H-Z Peng
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - L-X Ma
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - M-H Lv
- Center for Laboratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - T Hu
- Department of Anesthesiology, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China
| | - T Liu
- Department of Pediatrics, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China; Center for Laboratory Medicine, The First Affiliated Hospital of Nanchang University, Nanchang 330006, China.
| |
Collapse
|
35
|
Peña-Ortega F, Rivera-Angulo AJ, Lorea-Hernández JJ. Pharmacological Tools to Study the Role of Astrocytes in Neural Network Functions. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 949:47-66. [DOI: 10.1007/978-3-319-40764-7_3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
36
|
Lorea-Hernández JJ, Morales T, Rivera-Angulo AJ, Alcantara-Gonzalez D, Peña-Ortega F. Microglia modulate respiratory rhythm generation and autoresuscitation. Glia 2015; 64:603-19. [DOI: 10.1002/glia.22951] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Revised: 11/18/2015] [Accepted: 11/18/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Jonathan-Julio Lorea-Hernández
- Departamento de Neurobiología del Desarrollo y Neurofisiología; Instituto De Neurobiología, UNAM Campus Juriquilla; Querétaro México
| | - Teresa Morales
- Departamento de Neurobiología del Desarrollo y Neurofisiología; Instituto De Neurobiología, UNAM Campus Juriquilla; Querétaro México
| | - Ana-Julia Rivera-Angulo
- Departamento de Neurobiología del Desarrollo y Neurofisiología; Instituto De Neurobiología, UNAM Campus Juriquilla; Querétaro México
| | - David Alcantara-Gonzalez
- Departamento de Neurobiología del Desarrollo y Neurofisiología; Instituto De Neurobiología, UNAM Campus Juriquilla; Querétaro México
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología; Instituto De Neurobiología, UNAM Campus Juriquilla; Querétaro México
| |
Collapse
|
37
|
Chen Y, Li G, Huang LYM. p38 MAPK mediates glial P2X7R-neuronal P2Y1R inhibitory control of P2X3R expression in dorsal root ganglion neurons. Mol Pain 2015; 11:68. [PMID: 26542462 PMCID: PMC4635984 DOI: 10.1186/s12990-015-0073-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 10/27/2015] [Indexed: 12/20/2022] Open
Abstract
Background We have previously shown that endogenously active purinergic P2X7 receptors (P2X7Rs) in satellite glial cells of dorsal root ganglia (DRGs) stimulate ATP release. The ATP activates P2Y1Rs located in the enwrapped neuronal somata, resulting in down-regulation of P2X3Rs. This P2X7R-P2Y1-P2X3R inhibitory control significantly reduces P2X3R-mediated nociceptive responses. The underlying mechanism by which the activation of P2Y1Rs inhibits the expression of P2X3Rs remains unexplored. Results Examining the effect of the activation of p38 mitogen-activated protein kinase on the expression of P2X3Rs in DRGs, we found that the p38 activator, anisomycin (Anis), reduced the expression of P2X3Rs. Blocking the activity of SGCs by the glial Krebs cycle inhibitor, fluorocitrate, did not change the effect of Anis. These results suggest that neuronal p38 plays a major role in the inhibition of P2X3R expression. Western blotting analyses showed that inhibiting P2Y1Rs by MRS2179 (MRS) or blocking P2X7Rs by either oxATP or A740003 reduced pp38 and increased P2X3R expression in DRGs. These results are further supported by the immunohistochemical study showing that P2X7R and P2Y1R antagonists reduce the percentage of pp38-positive neurons. These observations suggest that activation of P2X7Rs and P2Y1Rs promotes p38 activity to exert inhibitory control on P2X3R expression. Since activation of p38 by Anis in the presence of either A740003 or MRS could overcome the block of P2X7R-P2Y1R inhibitory control, p38 in DRG neurons is downstream of P2Y1Rs. In addition, inhibition of p38 by SB202190 was found to prevent the P2X7R and P2Y1R block of P2X3R expression and increase P2X3R-mediated nociceptive flinch behaviors. Conclusions p38 in DRG neurons downstream of P2Y1R is necessary and sufficient for the P2X7R-P2Y1R inhibitory control of P2X3R expression.
Collapse
Affiliation(s)
- Yong Chen
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, 77555-1069, USA.
| | - Guangwen Li
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, 77555-1069, USA.
| | - Li-Yen Mae Huang
- Department of Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, 77555-1069, USA.
| |
Collapse
|
38
|
Diversity of astrocyte functions and phenotypes in neural circuits. Nat Neurosci 2015; 18:942-52. [PMID: 26108722 DOI: 10.1038/nn.4043] [Citation(s) in RCA: 815] [Impact Index Per Article: 81.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Accepted: 05/04/2015] [Indexed: 12/11/2022]
Abstract
Astrocytes tile the entire CNS. They are vital for neural circuit function, but have traditionally been viewed as simple, homogenous cells that serve the same essential supportive roles everywhere. Here, we summarize breakthroughs that instead indicate that astrocytes represent a population of complex and functionally diverse cells. Physiological diversity of astrocytes is apparent between different brain circuits and microcircuits, and individual astrocytes display diverse signaling in subcellular compartments. With respect to injury and disease, astrocytes undergo diverse phenotypic changes that may be protective or causative with regard to pathology in a context-dependent manner. These new insights herald the concept that astrocytes represent a diverse population of genetically tractable cells that mediate neural circuit-specific roles in health and disease.
Collapse
|
39
|
Kaczor P, Rakus D, Mozrzymas JW. Neuron-astrocyte interaction enhance GABAergic synaptic transmission in a manner dependent on key metabolic enzymes. Front Cell Neurosci 2015; 9:120. [PMID: 25914620 PMCID: PMC4391237 DOI: 10.3389/fncel.2015.00120] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 03/17/2015] [Indexed: 11/21/2022] Open
Abstract
Gamma aminobutric acid (GABA) is the major inhibitory neurotransmitter in the adult brain and mechanisms of GABAergic inhibition have been intensely investigated in the past decades. Recent studies provided evidence for an important role of astrocytes in shaping GABAergic currents. One of the most obvious, but yet poorly understood, mechanisms of the cross-talk between GABAergic currents and astrocytes is metabolism including neurotransmitter homeostasis. In particular, how modulation of GABAergic currents by astrocytes depends on key enzymes involved in cellular metabolism remains largely unknown. To address this issue, we have considered two simple models of neuronal culture (NC): nominally astrocyte-free NC and neuronal-astrocytic co-cultures (ANCC). Miniature Inhibitory Postsynaptic Currents (mIPSCs) were recorded in control conditions and in the presence of different enzyme blockers. We report that enrichment of NC with astrocytes results in a marked increase in mIPSC frequency. This enhancement of GABAergic activity was accompanied by increased number of GAD65 and vGAT puncta, indicating that at least a part of the frequency enhancement was due to increased number of synaptic contacts. Inhibition of glutamine synthetase (Glns) (with MSO) strongly reduced mIPSC frequency in ANCC but had no effect in NC. Moreover, treatment of ANCC with inhibitor of glycogen phosphorylase (Gys) (BAYU6751) or with selective inhibitor of astrocytic Krebs cycle, fluoroacetate, resulted in a marked reduction of mIPSC frequency in ANCC having no effect in NC. We conclude that GABAergic synaptic transmission strongly depends on neuron-astrocyte interaction in a manner dependent on key metabolic enzymes as well as on the Krebs cycle.
Collapse
Affiliation(s)
- Przemysław Kaczor
- Department of Animal Molecular Physiology, Wroclaw University Wroclaw, Poland
| | - Dariusz Rakus
- Department of Animal Molecular Physiology, Wroclaw University Wroclaw, Poland
| | - Jerzy W Mozrzymas
- Department of Animal Molecular Physiology, Wroclaw University Wroclaw, Poland ; Laboratory of Neuroscience, Department of Biophysics, Wroclaw Medical University Wroclaw, Poland
| |
Collapse
|
40
|
Yang Y, Xu-Friedman MA. Different pools of glutamate receptors mediate sensitivity to ambient glutamate in the cochlear nucleus. J Neurophysiol 2015; 113:3634-45. [PMID: 25855696 DOI: 10.1152/jn.00693.2014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Accepted: 04/08/2015] [Indexed: 01/14/2023] Open
Abstract
Ambient glutamate plays an important role in pathological conditions, such as stroke, but its role during normal activity is not clear. In addition, it is not clear how ambient glutamate acts on glutamate receptors with varying affinities or subcellular localizations. To address this, we studied "endbulb of Held" synapses, which are formed by auditory nerve fibers onto bushy cells (BCs) in the anteroventral cochlear nucleus. When ambient glutamate was increased by applying the glutamate reuptake inhibitor TFB-TBOA, BCs depolarized as a result of activation of N-methyl-D-aspartate receptors (NMDARs) and group I metabotropic glutamate receptors (mGluRs). Application of antagonists against NMDARs (in 0 Mg(2+)) or mGluRs caused hyperpolarization, indicating that these receptors were bound by a tonic source of glutamate. AMPA receptors did not show these effects, consistent with their lower glutamate affinity. We also evaluated the subcellular localization of the receptors activated by ambient glutamate. The mGluRs were not activated by synaptic stimulation and thus appear to be exclusively extrasynaptic. By contrast, NMDARs in both synaptic and extrasynaptic compartments were activated by ambient glutamate, as shown using the use-dependent antagonist MK-801. Levels of ambient glutamate appeared to be regulated in a spike-independent manner, and glia likely play a major role. These low levels of ambient glutamate likely have functional consequences, as even low concentrations of TBOA caused significant increases in BC spiking following synaptic stimulation. These results indicate that normal resting potential appears to be poised in the region of maximal sensitivity to small changes in ambient glutamate.
Collapse
Affiliation(s)
- Yang Yang
- Department of Biological Sciences, University at Buffalo, State University of New York, Buffalo, New York
| | - Matthew A Xu-Friedman
- Department of Biological Sciences, University at Buffalo, State University of New York, Buffalo, New York
| |
Collapse
|
41
|
Astrocytic Regulation of Sleep Processes. CURRENT SLEEP MEDICINE REPORTS 2015. [DOI: 10.1007/s40675-014-0005-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
42
|
Bouyakdan K, Taïb B, Budry L, Zhao S, Rodaros D, Neess D, Mandrup S, Faergeman NJ, Alquier T. A novel role for central ACBP/DBI as a regulator of long-chain fatty acid metabolism in astrocytes. J Neurochem 2015; 133:253-65. [PMID: 25598214 DOI: 10.1111/jnc.13035] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Revised: 12/15/2014] [Accepted: 01/08/2015] [Indexed: 12/29/2022]
Abstract
Acyl-CoA-binding protein (ACBP) is a ubiquitously expressed protein that binds intracellular acyl-CoA esters. Several studies have suggested that ACBP acts as an acyl-CoA pool former and regulates long-chain fatty acids (LCFA) metabolism in peripheral tissues. In the brain, ACBP is known as Diazepam-Binding Inhibitor, a secreted peptide acting as an allosteric modulator of the GABAA receptor. However, its role in central LCFA metabolism remains unknown. In the present study, we investigated ACBP cellular expression, ACBP regulation of LCFA intracellular metabolism, FA profile, and FA metabolism-related gene expression using ACBP-deficient and control mice. ACBP was mainly found in astrocytes with high expression levels in the mediobasal hypothalamus. We demonstrate that ACBP deficiency alters the central LCFA-CoA profile and impairs unsaturated (oleate, linolenate) but not saturated (palmitate, stearate) LCFA metabolic fluxes in hypothalamic slices and astrocyte cultures. In addition, lack of ACBP differently affects the expression of genes involved in FA metabolism in cortical versus hypothalamic astrocytes. Finally, ACBP deficiency increases FA content and impairs their release in response to palmitate in hypothalamic astrocytes. Collectively, these findings reveal for the first time that central ACBP acts as a regulator of LCFA intracellular metabolism in astrocytes. Acyl-CoA-binding protein (ACBP) or diazepam-binding inhibitor is a secreted peptide acting centrally as a GABAA allosteric modulator. Using brain slices, cortical, and hypothalamic astrocyte cultures from ACBP KO mice, we demonstrate that ACBP mainly localizes in astrocytes and regulates unsaturated but not saturated long-chain fatty acids (LCFA) metabolism. In addition, ACBP deficiency alters FA metabolism-related genes and results in intracellular FA accumulation while affecting their release. Our results support a novel role for ACBP in brain lipid metabolism. FA, fatty acids; KO, knockout; PL, phospholipids; TAG, triacylglycerol.
Collapse
Affiliation(s)
- Khalil Bouyakdan
- Montreal Diabetes Research Center, Centre de Recherche du Centre Hospitalier de l'Universite de Montreal (CRCHUM), Montreal, Quebec, Canada; Department of Biochemistry, University of Montreal, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Abstract
Since their introduction in the 1960s, benzodiazepines (BZs) remain one of the most commonly prescribed medications, acting as potent sedatives, hypnotics, anxiolytics, anticonvulsants, and muscle relaxants. The primary neural action of BZs and related compounds is augmentation of inhibitory transmission, which occurs through allosteric modulation of the gamma-aminobutyric acid (GABA)-induced current at the gamma-aminobutyric acid receptor (GABAAR). The discovery of the BZ-binding site on GABAARs encouraged many to speculate that the brain produces its own endogenous ligands to this site (Costa & Guidotti, 1985). The romanticized quest for endozepines, endogenous ligands to the BZ-binding site, has uncovered a variety of ligands that might fulfill this role, including oleamides (Cravatt et al., 1995), nonpeptidic endozepines (Rothstein et al., 1992), and the protein diazepam-binding inhibitor (DBI) (Costa & Guidotti, 1985). Of these ligands, DBI, and affiliated peptide fragments, is the most extensively studied endozepine. The quest for the "brain's Valium" over the decades has been elusive as mainly negative allosteric modulatory effects have been observed (Alfonso, Le Magueresse, Zuccotti, Khodosevich, & Monyer, 2012; Costa & Guidotti, 1985), but recent evidence is accumulating that DBI displays regionally discrete endogenous positive modulation of GABA transmission through activation of the BZ receptor (Christian et al., 2013). Herein, we review the literature on this topic, focusing on identification of the endogenous molecule and its region-specific expression and function.
Collapse
|
44
|
Buckman LB, Ellacott KLJ. The contribution of hypothalamic macroglia to the regulation of energy homeostasis. Front Syst Neurosci 2014; 8:212. [PMID: 25374514 PMCID: PMC4206078 DOI: 10.3389/fnsys.2014.00212] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 10/07/2014] [Indexed: 11/13/2022] Open
Abstract
The hypothalamus is critical for the regulation of energy homeostasis. Genetic and pharmacologic studies have identified a number of key hypothalamic neuronal circuits that integrate signals controlling food intake and energy expenditure. Recently, studies have begun to emerge demonstrating a role for non-neuronal cell types in the regulation of energy homeostasis. In particular the potential importance of different glial cell types is increasingly being recognized. A number of studies have described changes in the activity of hypothalamic macroglia (principally astrocytes and tanycytes) in response to states of positive and negative energy balance, such as obesity and fasting. This article will review these studies and discuss how these findings are changing our understanding of the cellular mechanisms by which energy homeostasis is regulated.
Collapse
Affiliation(s)
- Laura B Buckman
- Department of Molecular Physiology and Biophysics, Vanderbilt University Medical Center Nashville, TN, USA ; Division of Infectious Disease, School of Medicine, University of North Carolina at Chapel Hill Chapel Hill, NC, USA
| | - Kate L J Ellacott
- Biomedical Neuroscience Research Group, University of Exeter Medical School, Hatherly Laboratories Exeter, UK
| |
Collapse
|
45
|
|