201
|
Miller AN, Vaisey G, Long SB. Molecular mechanisms of gating in the calcium-activated chloride channel bestrophin. eLife 2019; 8:43231. [PMID: 30628889 PMCID: PMC6342527 DOI: 10.7554/elife.43231] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 01/02/2019] [Indexed: 11/13/2022] Open
Abstract
Bestrophin (BEST1-4) ligand-gated chloride (Cl-) channels are activated by calcium (Ca2+). Mutation of BEST1 causes retinal disease. Partly because bestrophin channels have no sequence or structural similarity to other ion channels, the molecular mechanisms underlying gating are unknown. Here, we present a series of cryo-electron microscopy structures of chicken BEST1, determined at 3.1 Å resolution or better, that represent the channel’s principal gating states. Unlike other channels, opening of the pore is due to the repositioning of tethered pore-lining helices within a surrounding protein shell that dramatically widens a neck of the pore through a concertina of amino acid rearrangements. The neck serves as both the activation and the inactivation gate. Ca2+ binding instigates opening of the neck through allosteric means whereas inactivation peptide binding induces closing. An aperture within the otherwise wide pore controls anion permeability. The studies define a new molecular paradigm for gating among ligand-gated ion channels.
Collapse
Affiliation(s)
- Alexandria N Miller
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, United States
| | - George Vaisey
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, United States.,Louis V. Gerstner Jr. Graduate School of Biomedical Sciences, Memorial Sloan Kettering Cancer Center, New York, United States
| | - Stephen B Long
- Structural Biology Program, Memorial Sloan Kettering Cancer Center, New York, United States
| |
Collapse
|
202
|
|
203
|
Park YM, Chun H, Shin JI, Lee CJ. Astrocyte Specificity and Coverage of hGFAP-CreERT2 [Tg(GFAP-Cre/ERT2)13Kdmc] Mouse Line in Various Brain Regions. Exp Neurobiol 2018; 27:508-525. [PMID: 30636902 PMCID: PMC6318562 DOI: 10.5607/en.2018.27.6.508] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 12/12/2018] [Accepted: 12/12/2018] [Indexed: 11/25/2022] Open
Abstract
Astrocyte is the most abundant cell type in the central nervous system and its importance has been increasingly recognized in the brain pathophysiology. To study in vivo function of astrocyte, astrocyte-specific gene-targeting is regarded as a powerful approach. Especially, hGFAP-CreERT2, which expresses tamoxifen-inducible Cre recombinase under the human GFAP promoter, has been developed and characterized from several research groups. However, one of these mouse lines, [Tg(GFAP-Cre/ERT2)13Kdmc] from Ken McCarthy group has not been quantitatively analyzed, despite its frequent use. Here, we performed comprehensive characterization of this mouse line with quantitative analysis. By crossing this mouse line with Ai14 (RCL-tdTomato), a very sensitive Cre reporter mouse line, we visualized the Cre-expressing cells in various brain regions. For quantitative analysis, we immunostained S100β as an astrocytic marker and NeuN, tyrosine hydroxylase or calbindin as a neuronal marker in different brain regions. We calculated ‘astrocyte specificity’ as the proportion of co-labelled S100β and tdTomato positive cells in the total number of tdTomato positive cells and the ‘astrocyte coverage’ as the proportion of co-labelled S100β and tdTomato positive cells in the total number of S100β positive cells. Interestingly, we found varying degree of astrocyte specificity and coverage in each brain region. In cortex, hypothalamus, substantia nigra pars compacta and cerebellar Purkinje layer, we observed high astrocyte specificity (over 89%) and relatively high astrocyte coverage (over 70%). In striatum, hippocampal CA1 layer, dentate gyrus and cerebellar granule layer, we observed high astrocyte specificity (over 80%), but relative low astrocyte coverage (50–60%). However, thalamus and amygdala showed low astrocyte specificity (about 65%) and significant neuron specificity (over 30%). This hGFAP-CreERT2 mouse line can be useful for genetic modulations of target gene either in gain-of-function or loss-of-function studies in the brain regions with high astrocyte specificity and coverage. However, the use of this mouse line should be restricted to gain-of-function studies in the brain regions with high astrocyte specificity but low coverage. In conclusion, hGFAP-CreERT2 mouse line could be a powerful tool for gene-targeting of astrocytes in cortex, striatum, hippocampus, hypothalamus, substantia nigra pars compacta and cerebellum, but not in thalamus and amygdala.
Collapse
Affiliation(s)
- Yongmin Mason Park
- Division of Bio-Medical Science & Technology, Department of Neuroscience, KIST School, Korea University of Science and Technology, Seoul 02792, Korea.,Center for Glia-Neuron Interaction, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.,Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | - Heejung Chun
- Center for Glia-Neuron Interaction, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.,Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | - Jeong-Im Shin
- Division of Bio-Medical Science & Technology, Department of Neuroscience, KIST School, Korea University of Science and Technology, Seoul 02792, Korea.,Center for Glia-Neuron Interaction, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.,Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| | - C Justin Lee
- Division of Bio-Medical Science & Technology, Department of Neuroscience, KIST School, Korea University of Science and Technology, Seoul 02792, Korea.,Center for Glia-Neuron Interaction, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.,Center for Cognition and Sociality, Institute for Basic Science, Daejeon 34126, Korea
| |
Collapse
|
204
|
Ziegler-Waldkirch S, Meyer-Luehmann M. The Role of Glial Cells and Synapse Loss in Mouse Models of Alzheimer's Disease. Front Cell Neurosci 2018; 12:473. [PMID: 30618627 PMCID: PMC6297249 DOI: 10.3389/fncel.2018.00473] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Accepted: 11/20/2018] [Indexed: 11/13/2022] Open
Abstract
Synapse loss has detrimental effects on cellular communication, leading to network disruptions within the central nervous system (CNS) such as in Alzheimer’s disease (AD). AD is characterized by a progressive decline of memory function, cognition, neuronal and synapse loss. The two main neuropathological hallmarks are amyloid-β (Aβ) plaques and neurofibrillary tangles. In the brain of AD patients and in mouse models of AD several morphological and functional changes, such as microgliosis and astrogliosis around Aβ plaques, as well as dendritic and synaptic alterations, are associated with these lesions. In this review article, we will summarize the current literature on synapse loss in mouse models of AD and discuss current and prospective treatments for AD.
Collapse
Affiliation(s)
- Stephanie Ziegler-Waldkirch
- Department of Neurology, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Melanie Meyer-Luehmann
- Department of Neurology, Medical Center-University of Freiburg, Freiburg, Germany.,Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
205
|
Hsu YT, Chang YG, Chern Y. Insights into GABA Aergic system alteration in Huntington's disease. Open Biol 2018; 8:rsob.180165. [PMID: 30518638 PMCID: PMC6303784 DOI: 10.1098/rsob.180165] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/30/2018] [Indexed: 12/15/2022] Open
Abstract
Huntington's disease (HD) is an autosomal dominant progressive neurodegenerative disease that is characterized by a triad of motor, psychiatric and cognitive impairments. There is still no effective therapy to delay or halt the disease progress. The striatum and cortex are two particularly affected brain regions that exhibit dense reciprocal excitatory glutamate and inhibitory gamma-amino butyric acid (GABA) connections. Imbalance between excitatory and inhibitory signalling is known to greatly affect motor and cognitive processes. Emerging evidence supports the hypothesis that disrupted GABAergic circuits underlie HD pathogenesis. In the present review, we focused on the multiple defects recently found in the GABAergic inhibitory system, including altered GABA level and synthesis, abnormal subunit composition and distribution of GABAA receptors and aberrant GABAA receptor-mediated signalling. In particular, the important role of cation–chloride cotransporters (i.e. NKCC1 and KCC2) is discussed. Recent studies also suggest that neuroinflammation contributes significantly to the abnormal GABAergic inhibition in HD. Thus, GABAA receptors and cation–chloride cotransporters are potential therapeutic targets for HD. Given the limited availability of therapeutic treatments for HD, a better understanding of GABAergic dysfunction in HD could provide novel therapeutic opportunities.
Collapse
Affiliation(s)
- Yi-Ting Hsu
- PhD Program for Translational Medicine, China Medical University and Academia Sinica, Taiwan, Republic of China.,Department of Neurology, China Medical University Hospital, Taichung, Taiwan, Republic of China
| | - Ya-Gin Chang
- Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan, Republic of China.,Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming University and Academia Sinica, Taipei, Taiwan, Republic of China
| | - Yijuang Chern
- PhD Program for Translational Medicine, China Medical University and Academia Sinica, Taiwan, Republic of China .,Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, Republic of China
| |
Collapse
|
206
|
Abudara V, Retamal MA, Del Rio R, Orellana JA. Synaptic Functions of Hemichannels and Pannexons: A Double-Edged Sword. Front Mol Neurosci 2018; 11:435. [PMID: 30564096 PMCID: PMC6288452 DOI: 10.3389/fnmol.2018.00435] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Accepted: 11/08/2018] [Indexed: 01/18/2023] Open
Abstract
The classical view of synapses as the functional contact between presynaptic and postsynaptic neurons has been challenged in recent years by the emerging regulatory role of glial cells. Astrocytes, traditionally considered merely supportive elements are now recognized as active modulators of synaptic transmission and plasticity at the now so-called "tripartite synapse." In addition, an increasing body of evidence indicates that beyond immune functions microglia also participate in various processes aimed to shape synaptic plasticity. Release of neuroactive compounds of glial origin, -process known as gliotransmission-, constitute a widespread mechanism through which glial cells can either potentiate or reduce the synaptic strength. The prevailing vision states that gliotransmission depends on an intracellular Ca2+/exocytotic-mediated release; notwithstanding, growing evidence is pointing at hemichannels (connexons) and pannexin channels (pannexons) as alternative non-vesicular routes for gliotransmitters efflux. In concurrence with this novel concept, both hemichannels and pannexons are known to mediate the transfer of ions and signaling molecules -such as ATP and glutamate- between the cytoplasm and the extracellular milieu. Importantly, recent reports show that glial hemichannels and pannexons are capable to perceive synaptic activity and to respond to it through changes in their functional state. In this article, we will review the current information supporting the "double edge sword" role of hemichannels and pannexons in the function of central and peripheral synapses. At one end, available data support the idea that these channels are chief components of a feedback control mechanism through which gliotransmitters adjust the synaptic gain in either resting or stimulated conditions. At the other end, we will discuss how the excitotoxic release of gliotransmitters and [Ca2+]i overload linked to the opening of hemichannels/pannexons might impact cell function and survival in the nervous system.
Collapse
Affiliation(s)
- Verónica Abudara
- Departamento de Fisiología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Mauricio A Retamal
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo, Santiago, Chile.,Department of Cell Physiology and Molecular Biophysics, Center for Membrane Protein Research, Texas Tech University Health Sciences Center, Lubbock, TX, United States.,Programa de Comunicación Celular en Cáncer, Instituto de Ciencias e Innovación en Medicina, Santiago, Chile
| | - Rodrigo Del Rio
- Laboratory of Cardiorespiratory Control, Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Envejecimiento y Regeneración, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Excelencia en Biomedicina de Magallanes, Universidad de Magallanes, Punta Arenas, Chile
| | - Juan A Orellana
- Departamento de Neurología, Escuela de Medicina and Centro Interdisciplinario de Neurociencias, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile.,Centro de Investigación y Estudio del Consumo de Alcohol en Adolescentes, Santiago, Chile
| |
Collapse
|
207
|
Diverse facets of cortical interneuron migration regulation – Implications of neuronal activity and epigenetics. Brain Res 2018; 1700:160-169. [DOI: 10.1016/j.brainres.2018.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 09/02/2018] [Accepted: 09/03/2018] [Indexed: 01/21/2023]
|
208
|
Mederos S, González-Arias C, Perea G. Astrocyte-Neuron Networks: A Multilane Highway of Signaling for Homeostatic Brain Function. Front Synaptic Neurosci 2018; 10:45. [PMID: 30542276 PMCID: PMC6277918 DOI: 10.3389/fnsyn.2018.00045] [Citation(s) in RCA: 109] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2018] [Accepted: 11/12/2018] [Indexed: 12/22/2022] Open
Abstract
Research on glial cells over the past 30 years has confirmed the critical role of astrocytes in pathophysiological brain states. However, most of our knowledge about astrocyte physiology and of the interactions between astrocytes and neurons is based on the premises that astrocytes constitute a homogeneous cell type, without considering the particular properties of the circuits or brain nuclei in which the astrocytes are located. Therefore, we argue that more-sophisticated experiments are required to elucidate the specific features of astrocytes in different brain regions, and even within different layers of a particular circuit. Thus, in addition to considering the diverse mechanisms used by astrocytes to communicate with neurons and synaptic partners, it is necessary to take into account the cellular heterogeneity that likely contributes to the outcomes of astrocyte-neuron signaling. In this review article, we briefly summarize the current data regarding the anatomical, molecular and functional properties of astrocyte-neuron communication, as well as the heterogeneity within this communication.
Collapse
Affiliation(s)
- Sara Mederos
- Department of Functional and Systems Neurobiology, Instituto Cajal (IC), CSIC, Madrid, Spain
| | - Candela González-Arias
- Department of Functional and Systems Neurobiology, Instituto Cajal (IC), CSIC, Madrid, Spain
| | - Gertrudis Perea
- Department of Functional and Systems Neurobiology, Instituto Cajal (IC), CSIC, Madrid, Spain
| |
Collapse
|
209
|
New Roles for Old Glue: Astrocyte Function in Synaptic Plasticity and Neurological Disorders. Int Neurourol J 2018; 22:S106-114. [PMID: 30396259 PMCID: PMC6234728 DOI: 10.5213/inj.1836214.107] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 10/10/2018] [Indexed: 01/02/2023] Open
Abstract
Previously believed to solely play a supportive role in the central nervous system, astrocytes are now considered active players in normal brain function. Evidence in recent decades extends their contributions beyond the classically held brain glue role; it's now known that astrocytes act as a unique excitable component with functions extending into local network modulation, synaptic plasticity, and memory formation, and postinjury repair. In this review article, we highlight our growing understanding of astrocyte function and physiology, the increasing role of gliotransmitters in neuron-glia communication, and the role of astrocytes in modulating synaptic plasticity and cognitive function. Owing to the duality of both beneficial and deleterious roles attributed to astrocytes, we also discuss the implications of this new knowledge as it applies to neurological disorders including Alzheimer disease, epilepsy, and schizophrenia.
Collapse
|
210
|
A Parkinson’s disease gene, DJ-1, regulates astrogliosis through STAT3. Neurosci Lett 2018; 685:144-149. [DOI: 10.1016/j.neulet.2018.08.025] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 08/10/2018] [Accepted: 08/21/2018] [Indexed: 12/28/2022]
|
211
|
Impaired Autophagy of GABAergic Interneurons in Neuropathic Pain. Pain Res Manag 2018; 2018:9185368. [PMID: 30356379 PMCID: PMC6176324 DOI: 10.1155/2018/9185368] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Accepted: 09/12/2018] [Indexed: 12/30/2022]
Abstract
Neuropathic pain (NP) is caused by lesions of the peripheral fibers and central neurons in the somatosensory nervous system and affects 7-10% of the general population. Although the distinct cause of neuropathic pain has been investigated in primary afferent neurons over the years, pain modulation by central sensitization remains controversial. NP is believed to be driven by cell type-specific spinal synaptic plasticity in the dorsal horn. Upon intense afferent stimulation, spinothalamic tract neurons are potentiated, whereas GABAergic interneurons are inhibited leading to long-term depression. Growing evidences suggest that the inhibition of GABAergic neurons plays pivotal roles in the manifestation of neuropathic and inflammatory pain states. Downregulation of GABA transmission and impairment of GABAergic interneurons in the dorsal horn are critical consequences after spinal cord and peripheral nerve injuries. These impairments in GABAergic interneurons may be associated with dysfunctional autophagy, resulting in neuropathic pain. Here, we review an emerging number of investigations that suggest a pivotal role of impaired autophagy of GABAergic interneurons in NP. We discuss relevant research spurring the development of new targets and therapeutic agents of NP and emphasize the need for a multidisciplinary approach to manage NP in the future.
Collapse
|
212
|
Christensen RK, Delgado-Lezama R, Russo RE, Lind BL, Alcocer EL, Rath MF, Fabbiani G, Schmitt N, Lauritzen M, Petersen AV, Carlsen EM, Perrier JF. Spinal dorsal horn astrocytes release GABA in response to synaptic activation. J Physiol 2018; 596:4983-4994. [PMID: 30079574 DOI: 10.1113/jp276562] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Accepted: 08/01/2018] [Indexed: 02/02/2023] Open
Abstract
KEY POINTS GABA is an essential molecule for sensory information processing. It is usually assumed to be released by neurons. Here we show that in the dorsal horn of the spinal cord, astrocytes respond to glutamate by releasing GABA. Our findings suggest a novel role for astrocytes in somatosensory information processing. ABSTRACT Astrocytes participate in neuronal signalling by releasing gliotransmitters in response to neurotransmitters. We investigated if astrocytes from the dorsal horn of the spinal cord of adult red-eared turtles (Trachemys scripta elegans) release GABA in response to glutamatergic receptor activation. For this, we developed a GABA sensor consisting of HEK cells expressing GABAA receptors. By positioning the sensor recorded in the whole-cell patch-clamp configuration within the dorsal horn of a spinal cord slice, we could detect GABA in the extracellular space. Puff application of glutamate induced GABA release events with time courses that exceeded the duration of inhibitory postsynaptic currents by one order of magnitude. Because the events were neither affected by extracellular addition of nickel, cadmium and tetrodotoxin nor by removal of Ca2+ , we concluded that they originated from non-neuronal cells. Immunohistochemical staining allowed the detection of GABA in a fraction of dorsal horn astrocytes. The selective stimulation of A∂ and C fibres in a dorsal root filament induced a Ca2+ increase in astrocytes loaded with Oregon Green BAPTA. Finally, chelating Ca2+ in a single astrocyte was sufficient to prevent the GABA release evoked by glutamate. Our results indicate that glutamate triggers the release of GABA from dorsal horn astrocytes with a time course compatible with the integration of sensory inputs.
Collapse
Affiliation(s)
- Rasmus Kordt Christensen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Rodolfo Delgado-Lezama
- Departamento de Fisiología, Biofísica y Neurociencias Cinvestav-IPN Avenida IPN 2508, Col. Zacatenco México City, CP, 07300, Mexico
| | - Raúl E Russo
- Neurofisiología Celular y Molecular, Instituto de Investigaciones Biológicas Clemente Estable, 11600, Montevideo, Uruguay
| | - Barbara Lykke Lind
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Emanuel Loeza Alcocer
- Departamento de Fisiología, Biofísica y Neurociencias Cinvestav-IPN Avenida IPN 2508, Col. Zacatenco México City, CP, 07300, Mexico
| | - Martin Fredensborg Rath
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Gabriela Fabbiani
- Neurofisiología Celular y Molecular, Instituto de Investigaciones Biológicas Clemente Estable, 11600, Montevideo, Uruguay
| | - Nicole Schmitt
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Martin Lauritzen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Anders Victor Petersen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Eva Meier Carlsen
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| | - Jean-François Perrier
- Department of Neuroscience, Faculty of Health and Medical Sciences, University of Copenhagen, 2200, Copenhagen, Denmark
| |
Collapse
|
213
|
Zhang Y, Kittredge A, Ward N, Ji C, Chen S, Yang T. ATP activates bestrophin ion channels through direct interaction. Nat Commun 2018; 9:3126. [PMID: 30087350 PMCID: PMC6081419 DOI: 10.1038/s41467-018-05616-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 07/09/2018] [Indexed: 11/13/2022] Open
Abstract
Human Bestrophin1 (hBest1) is a Ca2+-activated Cl- channel in retinal pigment epithelium (RPE) essential for retina physiology, and its mutation results in retinal degenerative diseases that have no available treatments. Here, we discover that hBest1's channel activity in human RPE is significantly enhanced by adenosine triphosphate (ATP) in a dose-dependent manner. We further demonstrate a direct interaction between ATP and bestrophins, and map the ATP-binding motif on hBest1 to an intracellular loop adjacent to the channel activation gate. Importantly, a disease-causing mutation of hBest1 located within the ATP-binding motif, p.I201T, diminishes ATP-dependent activation of the channel in patient-derived RPE, while the corresponding mutants in bestrophin homologs display defective ATP binding and a conformational change in the ATP-binding motif. Taken together, our results identify ATP as a critical activator of bestrophins, and reveal the molecular mechanism of an hBest1 patient-specific mutation.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY, 14642, USA
| | - Alec Kittredge
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY, 14642, USA
| | - Nancy Ward
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY, 14642, USA
| | - Changyi Ji
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY, 14642, USA
| | - Shoudeng Chen
- Molecular Imaging Center, Department of Experimental Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangzhou, 519000, China
| | - Tingting Yang
- Department of Pharmacology and Physiology, University of Rochester, School of Medicine and Dentistry, Rochester, NY, 14642, USA.
| |
Collapse
|
214
|
Shim HG, Jang SS, Kim SH, Hwang EM, Min JO, Kim HY, Kim YS, Ryu C, Chung G, Kim Y, Yoon BE, Kim SJ. TNF-α increases the intrinsic excitability of cerebellar Purkinje cells through elevating glutamate release in Bergmann Glia. Sci Rep 2018; 8:11589. [PMID: 30072733 PMCID: PMC6072779 DOI: 10.1038/s41598-018-29786-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 07/18/2018] [Indexed: 11/17/2022] Open
Abstract
For decades, the glial function has been highlighted not only as the ‘structural glue’, but also as an ‘active participant’ in neural circuits. Here, we suggest that tumor necrosis factor α (TNF-α), a key inflammatory cytokine, alters the neural activity of the cerebellar Purkinje cells (PCs) by facilitating gliotransmission in the juvenile male rat cerebellum. A bath application of TNF-α (100 ng/ml) in acute cerebellar slices elevates spiking activity of PCs with no alterations in the regularity of PC firings. Interestingly, the effect of TNF-α on the intrinsic excitability of PCs was abolished under a condition in which the type1 TNF receptor (TNFR1) in Bergmann glia (BG) was genetically suppressed by viral delivery of an adeno-associated virus (AAV) containing TNFR1-shRNA. In addition, we measured the concentration of glutamate derived from dissociated cerebellar cortical astrocyte cultures treated with TNF-α and observed a progressive increase of glutamate in a time-dependent manner. We hypothesised that TNF-α-induced elevation of glutamate from BGs enveloping the synaptic cleft may directly activate metabotropic glutamate receptor1 (mGluR1). Pharmacological inhibition of mGluR1, indeed, prevented the TNF-α-mediated elevation of the intrinsic excitability in PCs. Taken together, our study reveals that TNF-α triggers glutamate release in BG, thereby increasing the intrinsic excitability of cerebellar PCs in a mGluR1-dependent manner.
Collapse
Affiliation(s)
- Hyun Geun Shim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Korea
| | - Sung-Soo Jang
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.,Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Seung Ha Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Korea
| | - Eun Mi Hwang
- Center for Functional Connectomics, Korea Institute of Science and Technology, Seoul, Korea
| | - Joo Ok Min
- Department of Molecular biology, Dankook University, Chungnam, Korea
| | - Hye Yun Kim
- Department of Pharmacy and Integrated Science and Engineering Division, Yonsei University, Incheon, Korea
| | - Yoo Sung Kim
- Department of Molecular biology, Dankook University, Chungnam, Korea
| | - Changhyeon Ryu
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.,Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Korea.,Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Geehoon Chung
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea.,Department of Brain and Cognitive Science, College of Science, Seoul National University, Seoul, Korea
| | - YoungSoo Kim
- Department of Pharmacy and Integrated Science and Engineering Division, Yonsei University, Incheon, Korea
| | - Bo-Eun Yoon
- Department of Molecular biology, Dankook University, Chungnam, Korea
| | - Sang Jeong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul, Korea. .,Department of Biomedical Science, Seoul National University College of Medicine, Seoul, Korea. .,Neuroscience Research Institute, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
215
|
Chun H, An H, Lim J, Woo J, Lee J, Ryu H, Lee CJ. Astrocytic proBDNF and Tonic GABA Distinguish Active versus Reactive Astrocytes in Hippocampus. Exp Neurobiol 2018; 27:155-170. [PMID: 30022867 PMCID: PMC6050417 DOI: 10.5607/en.2018.27.3.155] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 06/26/2018] [Accepted: 06/30/2018] [Indexed: 01/18/2023] Open
Abstract
Astrocytes are the most abundant cell type in the brain and they make close contacts with neurons and blood vessels. They respond dynamically to various environmental stimuli and change their morphological and functional properties. Both physiological and pathological stimuli can induce versatile changes in astrocytes, as this phenomenon is referred to as ‘astrocytic plasticity’. However, the molecular and cellular mechanisms of astrocytic plasticity in response to various stimuli remain elusive, except for the presence of hypertrophy, a conspicuous structural change which is frequently observed in activated or reactive astrocytes. Here, we investigated differential characteristics of astrocytic plasticity in a stimulus-dependent manner. Strikingly, a stab wound brain injury lead to hypertrophy of astrocytes accompanied by increased GABA expression and tonic GABA release in mouse CA1 hippocampus. In contrast, the mice experiencing enriched environment exhibited astrocytic hypertrophy with enhanced proBDNF immunoreactivity but without GABA signal. Based on the results, we define proBDNF-positive/GABA-negative hypertrophic astrocytes as ‘active’ astrocytes and GABA-positive hypertrophic astrocytes as ‘reactive’ astrocytes, respectively. We propose for the first time that astrocytic proBDNF can be a bona fide molecular marker of the active astrocytes, which are distinct from the reactive astrocytes which show hypertrophy but with aberrant GABA.
Collapse
Affiliation(s)
- Heejung Chun
- Center for Neuroscience and Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.,Center for Glia-Neuron Interaction, Brain Science Institute, KIST, Seoul 02792, Korea
| | - Heeyoung An
- Center for Neuroscience and Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.,Center for Glia-Neuron Interaction, Brain Science Institute, KIST, Seoul 02792, Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Korea
| | - Jiwoon Lim
- Center for Glia-Neuron Interaction, Brain Science Institute, KIST, Seoul 02792, Korea
| | - Junsung Woo
- Center for Neuroscience and Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Jaekwang Lee
- Center for Neuroscience and Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - Hoon Ryu
- Center for Neuromedicine, Brain Science Institute, KIST, Seoul 02792, Korea.,Boston University Alzheimer's Disease Center and Department of Neurology, Boston University School of Medicine, Boston, MA 02118, USA
| | - C Justin Lee
- Center for Neuroscience and Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.,Center for Glia-Neuron Interaction, Brain Science Institute, KIST, Seoul 02792, Korea.,Division of Bio-Medical Science & Technology, KIST School, KIST, Seoul 02792, Korea
| |
Collapse
|
216
|
Plata A, Lebedeva A, Denisov P, Nosova O, Postnikova TY, Pimashkin A, Brazhe A, Zaitsev AV, Rusakov DA, Semyanov A. Astrocytic Atrophy Following Status Epilepticus Parallels Reduced Ca 2+ Activity and Impaired Synaptic Plasticity in the Rat Hippocampus. Front Mol Neurosci 2018; 11:215. [PMID: 29997475 PMCID: PMC6028739 DOI: 10.3389/fnmol.2018.00215] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 05/30/2018] [Indexed: 11/13/2022] Open
Abstract
Epilepsy is a group of neurological disorders commonly associated with the neuronal malfunction leading to generation of seizures. Recent reports point to a possible contribution of astrocytes into this pathology. We used the lithium-pilocarpine model of status epilepticus (SE) in rats to monitor changes in astrocytes. Experiments were performed in acute hippocampal slices 2-4 weeks after SE induction. Nissl staining revealed significant neurodegeneration in the pyramidal cell layers of hippocampal CA1, CA3 areas, and the hilus, but not in the granular cell layer of the dentate gyrus. A significant increase in the density of astrocytes stained with an astrocyte-specific marker, sulforhodamine 101, was observed in CA1 stratum (str.) radiatum. Astrocytes in this area were also whole-cell loaded with a morphological tracer, Alexa Fluor 594, for two-photon excitation imaging. Sholl analyses showed no changes in the size of the astrocytic domain or in the number of primary astrocytic branches, but a significant reduction in the number of distal branches that are resolved with diffraction-limited light microscopy (and are thought to contain Ca2+ stores, such as mitochondria and endoplasmic reticulum). The atrophy of astrocytic branches correlated with the reduced size, but not overall frequency of Ca2+ events. The volume tissue fraction of nanoscopic (beyond the diffraction limit) astrocytic leaflets showed no difference between control and SE animals. The results of spatial entropy-complexity spectrum analysis were also consistent with changes in ratio of astrocytic branches vs. leaflets. In addition, we observed uncoupling of astrocytes through the gap-junctions, which was suggested as a mechanism for reduced K+ buffering. However, no significant difference in time-course of synaptically induced K+ currents in patch-clamped astrocytes argued against possible alterations in K+ clearance by astrocytes. The magnitude of long-term-potentiation (LTP) was reduced after SE. Exogenous D-serine, a co-agonist of NMDA receptors, has rescued the initial phase of LTP. This suggests that the reduced Ca2+-dependent release of D-serine by astrocytes impairs initiation of synaptic plasticity. However, it does not explain the failure of LTP maintenance which may be responsible for cognitive decline associated with epilepsy.
Collapse
Affiliation(s)
- Alex Plata
- UNN Institute of Neuroscience, N. I. Lobachevsky State University of Nizhny Novgorod, University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Albina Lebedeva
- UNN Institute of Neuroscience, N. I. Lobachevsky State University of Nizhny Novgorod, University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Pavel Denisov
- UNN Institute of Neuroscience, N. I. Lobachevsky State University of Nizhny Novgorod, University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Olga Nosova
- UNN Institute of Neuroscience, N. I. Lobachevsky State University of Nizhny Novgorod, University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Tatiana Y. Postnikova
- Laboratory of Molecular Mechanisms of Neural Interactions, Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
- Department of Medical Physics, Peter the Great St. Petersburg Polytechnic University, St. Petersburg, Russia
| | - Alexey Pimashkin
- UNN Institute of Neuroscience, N. I. Lobachevsky State University of Nizhny Novgorod, University of Nizhny Novgorod, Nizhny Novgorod, Russia
| | - Alexey Brazhe
- Department of Biophysics, Faculty of Biology, M. V. Lomonosov Moscow State University, Moscow, Russia
| | - Aleksey V. Zaitsev
- Laboratory of Molecular Mechanisms of Neural Interactions, Sechenov Institute of Evolutionary Physiology and Biochemistry, Russian Academy of Sciences, St. Petersburg, Russia
- Institute of Experimental Medicine, Almazov National Medical Research Centre, St. Petersburg, Russia
| | - Dmitri A. Rusakov
- UNN Institute of Neuroscience, N. I. Lobachevsky State University of Nizhny Novgorod, University of Nizhny Novgorod, Nizhny Novgorod, Russia
- UCL Institute of Neurology, University College London, London, United Kingdom
| | - Alexey Semyanov
- UNN Institute of Neuroscience, N. I. Lobachevsky State University of Nizhny Novgorod, University of Nizhny Novgorod, Nizhny Novgorod, Russia
- Department of Molecular Neurobiology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- All-Russian Research Institute of Medicinal and Aromatic Plants, Moscow, Russia
| |
Collapse
|
217
|
Lia A, Zonta M, Requie LM, Carmignoto G. Dynamic interactions between GABAergic and astrocytic networks. Neurosci Lett 2018; 689:14-20. [PMID: 29908949 DOI: 10.1016/j.neulet.2018.06.026] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 06/12/2018] [Accepted: 06/13/2018] [Indexed: 10/28/2022]
Abstract
Brain network activity derives from the concerted action of different cell populations. Together with interneurons, astrocytes play fundamental roles in shaping the inhibition in brain circuitries and modulating neuronal transmission. In this review, we summarize past and recent findings that reveal in neural networks the importance of the interaction between GABAergic signaling and astrocytes and discuss its physiological and pathological relevance.
Collapse
Affiliation(s)
- Annamaria Lia
- University of Padua, Department of Biomedical Sciences, Padua, Italy; CNR, Neuroscience Institute, Padua, Italy
| | - Micaela Zonta
- University of Padua, Department of Biomedical Sciences, Padua, Italy; CNR, Neuroscience Institute, Padua, Italy.
| | - Linda Maria Requie
- University of Padua, Department of Biomedical Sciences, Padua, Italy; CNR, Neuroscience Institute, Padua, Italy
| | - Giorgio Carmignoto
- University of Padua, Department of Biomedical Sciences, Padua, Italy; CNR, Neuroscience Institute, Padua, Italy
| |
Collapse
|
218
|
Direct neurotransmitter activation of voltage-gated potassium channels. Nat Commun 2018; 9:1847. [PMID: 29748663 PMCID: PMC5945843 DOI: 10.1038/s41467-018-04266-w] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 04/16/2018] [Indexed: 11/25/2022] Open
Abstract
Voltage-gated potassium channels KCNQ2–5 generate the M-current, which controls neuronal excitability. KCNQ2–5 subunits each harbor a high-affinity anticonvulsant drug-binding pocket containing an essential tryptophan (W265 in human KCNQ3) conserved for >500 million years, yet lacking a known physiological function. Here, phylogenetic analysis, electrostatic potential mapping, in silico docking, electrophysiology, and radioligand binding assays reveal that the anticonvulsant binding pocket evolved to accommodate endogenous neurotransmitters including γ-aminobutyric acid (GABA), which directly activates KCNQ5 and KCNQ3 via W265. GABA, and endogenous metabolites β-hydroxybutyric acid (BHB) and γ-amino-β-hydroxybutyric acid (GABOB), competitively and differentially shift the voltage dependence of KCNQ3 activation. Our results uncover a novel paradigm: direct neurotransmitter activation of voltage-gated ion channels, enabling chemosensing of the neurotransmitter/metabolite landscape to regulate channel activity and cellular excitability. M-current is conveyed by voltage-sensitive KCNQ channels, which are enriched in GABAergic neurons and are activated by anticonvulsants such as retigabine. Here the authors show that GABA directly activates KCNQ3, at the residue required for its anticonvulsant activity.
Collapse
|
219
|
Joe EH, Choi DJ, An J, Eun JH, Jou I, Park S. Astrocytes, Microglia, and Parkinson's Disease. Exp Neurobiol 2018; 27:77-87. [PMID: 29731673 PMCID: PMC5934545 DOI: 10.5607/en.2018.27.2.77] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 04/14/2018] [Accepted: 04/16/2018] [Indexed: 12/12/2022] Open
Abstract
Astrocytes and microglia support well-being and well-function of the brain through diverse functions in both intact and injured brain. For example, astrocytes maintain homeostasis of microenvironment of the brain through up-taking ions and neurotransmitters, and provide growth factors and metabolites for neurons, etc. Microglia keep surveying surroundings, and remove abnormal synapses or respond to injury by isolating injury sites and expressing inflammatory cytokines. Therefore, their loss and/or functional alteration may be directly linked to brain diseases. Since Parkinson's disease (PD)-related genes are expressed in astrocytes and microglia, mutations of these genes may alter the functions of these cells, thereby contributing to disease onset and progression. Here, we review the roles of astrocytes and microglia in intact and injured brain, and discuss how PD genes regulate their functions.
Collapse
Affiliation(s)
- Eun-Hye Joe
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16944, Korea.,Department of Biomedical Sciences, Neuroscience Graduate Program, Ajou University School of Medicine, Suwon 16944, Korea.,Department of Brain Science, Ajou University School of Medicine, Suwon 16944, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon 16944, Korea
| | - Dong-Joo Choi
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16944, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon 16944, Korea
| | - Jiawei An
- Department of Biomedical Sciences, Neuroscience Graduate Program, Ajou University School of Medicine, Suwon 16944, Korea
| | - Jin-Hwa Eun
- Department of Biomedical Sciences, Neuroscience Graduate Program, Ajou University School of Medicine, Suwon 16944, Korea
| | - Ilo Jou
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16944, Korea.,Department of Biomedical Sciences, Neuroscience Graduate Program, Ajou University School of Medicine, Suwon 16944, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon 16944, Korea
| | - Sangmyun Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon 16944, Korea.,Department of Biomedical Sciences, Neuroscience Graduate Program, Ajou University School of Medicine, Suwon 16944, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Suwon 16944, Korea
| |
Collapse
|
220
|
Control of motor coordination by astrocytic tonic GABA release through modulation of excitation/inhibition balance in cerebellum. Proc Natl Acad Sci U S A 2018; 115:5004-5009. [PMID: 29691318 DOI: 10.1073/pnas.1721187115] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Tonic inhibition in the brain is mediated through an activation of extrasynaptic GABAA receptors by the tonically released GABA, resulting in a persistent GABAergic inhibitory action. It is one of the key regulators for neuronal excitability, exerting a powerful action on excitation/inhibition balance. We have previously reported that astrocytic GABA, synthesized by monoamine oxidase B (MAOB), mediates tonic inhibition via GABA-permeable bestrophin 1 (Best1) channel in the cerebellum. However, the role of astrocytic GABA in regulating neuronal excitability, synaptic transmission, and cerebellar brain function has remained elusive. Here, we report that a reduction of tonic GABA release by genetic removal or pharmacological inhibition of Best1 or MAOB caused an enhanced neuronal excitability in cerebellar granule cells (GCs), synaptic transmission at the parallel fiber-Purkinje cell (PF-PC) synapses, and motor performance on the rotarod test, whereas an augmentation of tonic GABA release by astrocyte-specific overexpression of MAOB resulted in a reduced neuronal excitability, synaptic transmission, and motor performance. The bidirectional modulation of astrocytic GABA by genetic alteration of Best1 or MAOB was confirmed by immunostaining and in vivo microdialysis. These findings indicate that astrocytes are the key player in motor coordination through tonic GABA release by modulating neuronal excitability and could be a good therapeutic target for various movement and psychiatric disorders, which show a disturbed excitation/inhibition balance.
Collapse
|
221
|
Peripheral GABA A receptor-mediated signaling facilitates persistent inflammatory hypersensitivity. Neuropharmacology 2018; 135:572-580. [PMID: 29634983 DOI: 10.1016/j.neuropharm.2018.04.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 03/30/2018] [Accepted: 04/06/2018] [Indexed: 01/26/2023]
Abstract
Unlike in the central nervous system (CNS), in the adult peripheral nervous system (PNS), activation of GABAA receptors (GABAAR) is excitatory because of the relatively high concentration of intracellular chloride in these neurons. Indeed, exogenous GABA and muscimol, a GABAAR agonist, exacerbate acute inflammatory hypersensitivity in rodents. However, it remains unclear whether peripheral GABAAR and the endogenous GABA play an important role in persistent inflammatory hypersensitivity. In this study, we thus investigated how peripheral GABAAR affects pain hypersensitivity by using the complete Freund's adjuvant (CFA)-induced persistent inflammatory pain mouse model. We found that intraplantar (i.pl.) administration of GABAAR antagonists, picrotoxin, and 1(S),9(R)-(-)-bicuculline methiodide significantly inhibited both spontaneous nociceptive (paw licking and flinching) behavior and mechanical hypersensitivity in CFA-injected mice at day 3 (D3), but not in naïve mice. Interestingly, CFA-induced mechanical hypersensitivity was significantly reversed by anti-GABA antibody (anti-GABA, i.pl.). In addition, RT-qPCR revealed that glutamate decarboxylase Gad1 (GAD 67) and Gad2 (GAD 65) mRNA expression was also upregulated in the ipsilateral hind paw of CFA-injected mice at D3. Finally, 5α-pregnan-3α-ol-20-one (3α,5α-THP), a selective positive allosteric modulator of GABAAR, produced mechanical hypersensitivity in naïve mice in a dose-dependent manner. Taken together, our results indicate that peripheral GABAAR and endogenous GABA, possibly produced by the inflamed tissue, potentiate CFA-induced persistent inflammatory hypersensitivity, suggesting that they can be used as a therapeutic target for alleviating inflammatory pain.
Collapse
|
222
|
Jammal L, Whalley B, Barkai E. Learning-induced modulation of the effect of neuroglial transmission on synaptic plasticity. J Neurophysiol 2018; 119:2373-2379. [PMID: 29561201 DOI: 10.1152/jn.00101.2018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Training rats in a complex olfactory discrimination task results in acquisition of "rule learning" (learning how to learn), a term describing the capability to perform the task superbly. Such rule learning results in strengthening of both excitatory and inhibitory synaptic connections between neurons in the piriform cortex. Moreover, intrinsic excitability is also enhanced throughout the pyramidal neuron population. Surprisingly, the cortical network retains its stability under these long-term modifications. In particular, the susceptibility for long-term potentiation (LTP) induction, while decreased for a short time window, returns to almost its pretraining value, although significant strengthening of AMPA receptor-mediated glutamatergic transmission remains. Such network balance is essential for maintaining the single-cell modifications that underlie long-term memory while preventing hyperexcitability that would result in runaway synaptic activity. However, the mechanisms underlying the long-term maintenance of such balance have yet to be described. In this study, we explored the role of astrocyte-mediated gliotransmission in long-term maintenance of learning-induced modifications in susceptibility for LTP induction and control of the strength of synaptic inhibition. We show that blocking connexin 43 hemichannels, which form gap junctions between astrocytes, decreases significantly the ability to induce LTP by stimulating the excitatory connections between piriform cortex pyramidal neurons after learning only. In parallel, spontaneous miniature inhibitory postsynaptic current amplitude is reduced in neurons from trained rats only, to the level of prelearning. Thus gliotransmission has a key role in maintaining learning-induced cortical stability by a wide-ranged control on synaptic transmission and plasticity. NEW & NOTEWORTHY We explore the role of astrocyte-mediated gliotransmission in maintenance of olfactory discrimination learning-induced modifications. We show that blocking gap junctions between astrocytes decreases significantly the ability to induce long-term potentiation in the piriform cortex after learning only. In parallel, synaptic inhibition is reduced in neurons from trained rats only, to the level of prelearning. Thus gliotransmission has a key role in maintaining learning-induced cortical stability by a wide-ranged control on synaptic transmission and plasticity.
Collapse
Affiliation(s)
- Luna Jammal
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa , Haifa , Israel
| | - Ben Whalley
- School of Chemistry, Food & Nutritional Sciences and Pharmacy, The University of Reading, White Knights, Reading , United Kingdom
| | - Edi Barkai
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa , Haifa , Israel
| |
Collapse
|
223
|
Park A, Uddin O, Li Y, Masri R, Keller A. Pain After Spinal Cord Injury Is Associated With Abnormal Presynaptic Inhibition in the Posterior Nucleus of the Thalamus. THE JOURNAL OF PAIN 2018; 19:727.e1-727.e15. [PMID: 29481977 DOI: 10.1016/j.jpain.2018.02.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 01/29/2018] [Accepted: 02/12/2018] [Indexed: 01/21/2023]
Abstract
Pain after spinal cord injury (SCI-Pain) is one of the most debilitating sequelae of spinal cord injury, characterized as relentless, excruciating pain that is largely refractory to treatments. Although it is generally agreed that SCI-Pain results from maladaptive plasticity in the pain processing pathway that includes the spinothalamic tract and somatosensory thalamus, the specific mechanisms underlying the development and maintenance of such pain are yet unclear. However, accumulating evidence suggests that SCI-Pain may be causally related to abnormal thalamic disinhibition, leading to hyperactivity in the posterior thalamic nucleus (PO), a higher-order nucleus involved in somatosensory and pain processing. We previously described several presynaptic mechanisms by which activity in PO is regulated, including the regulation of GABAergic as well as glutamatergic release by presynaptic metabotropic gamma-aminobutyric acid (GABAB) receptors. Using acute slices from a mouse model of SCI-Pain, we tested whether such mechanisms are affected by SCI-Pain. We reveal 2 abnormal changes in presynaptic signaling in the SCI-Pain condition. The substantial tonic activation of presynaptic GABAB receptors on GABAergic projections to PO-characteristic of normal animals-was absent in mice with SCI-Pain. Also absent in mice with SCI-Pain was the normal presynaptic regulation of glutamatergic projections to the PO by GABAB receptors. The loss of these regulatory presynaptic mechanisms in SCI-Pain may be an element of maladaptive plasticity leading to PO hyperexcitability and behavioral pain, and may suggest targets for development of novel treatments. PERSPECTIVE This report presents synaptic mechanisms that may underlie the development and maintenance of SCI-Pain. Because of the difficulty in treating SCI-Pain, a better understanding of the underlying neurobiological mechanisms is critical, and may allow development of better treatment modalities.
Collapse
Affiliation(s)
- Anthony Park
- Program in Neuroscience and Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Olivia Uddin
- Program in Neuroscience and Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Ying Li
- Program in Neuroscience and Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland
| | - Radi Masri
- Program in Neuroscience and Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland; Department of Endodontics, Periodontics and Prosthodontics, University of Maryland Baltimore, School of Dentistry, Baltimore, Maryland
| | - Asaf Keller
- Program in Neuroscience and Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, Maryland.
| |
Collapse
|
224
|
Tatsumi K, Isonishi A, Yamasaki M, Kawabe Y, Morita-Takemura S, Nakahara K, Terada Y, Shinjo T, Okuda H, Tanaka T, Wanaka A. Olig2-Lineage Astrocytes: A Distinct Subtype of Astrocytes That Differs from GFAP Astrocytes. Front Neuroanat 2018; 12:8. [PMID: 29497365 PMCID: PMC5819569 DOI: 10.3389/fnana.2018.00008] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 01/19/2018] [Indexed: 12/22/2022] Open
Abstract
Astrocytes are the most abundant glia cell type in the central nervous system (CNS), and are known to constitute heterogeneous populations that differ in their morphology, gene expression and function. Although glial fibrillary acidic protein (GFAP) is the cardinal cytological marker of CNS astrocytes, GFAP-negative astrocytes can easily be found in the adult CNS. Astrocytes are also allocated to spatially distinct regional domains during development. This regional heterogeneity suggests that they help to coordinate post-natal neural circuit formation and thereby to regulate eventual neuronal activity. Here, during lineage-tracing studies of cells expressing Olig2 using Olig2CreER; Rosa-CAG-LSL-eNpHR3.0-EYFP transgenic mice, we found Olig2-lineage mature astrocytes in the adult forebrain. Long-term administration of tamoxifen resulted in sufficient recombinant induction, and Olig2-lineage cells were found to be preferentially clustered in some adult brain nuclei. We then made distribution map of Olig2-lineage astrocytes in the adult mouse brain, and further compared the map with the distribution of GFAP-positive astrocytes visualized in GFAPCre; Rosa-CAG-LSL-eNpHR3.0-EYFP mice. Brain regions rich in Olig2-lineage astrocytes (e.g., basal forebrain, thalamic nuclei, and deep cerebellar nuclei) tended to lack GFAP-positive astrocytes, and vice versa. Even within a single brain nucleus, Olig2-lineage astrocytes and GFAP astrocytes frequently occupied mutually exclusive territories. These findings strongly suggest that there is a subpopulation of astrocytes (Olig2-lineage astrocytes) in the adult brain, and that it differs from GFAP-positive astrocytes in its distribution pattern and perhaps also in its function. Interestingly, the brain nuclei rich in Olig2-lineage astrocytes strongly expressed GABA-transporter 3 in astrocytes and vesicular GABA transporter in neurons, suggesting that Olig2-lineage astrocytes are involved in inhibitory neuronal transmission.
Collapse
Affiliation(s)
- Kouko Tatsumi
- Department of Anatomy and Neuroscience, Faculty of Medicine, Nara Medical University, Kashihara, Japan
| | - Ayami Isonishi
- Department of Anatomy and Neuroscience, Faculty of Medicine, Nara Medical University, Kashihara, Japan
| | - Miwako Yamasaki
- Department of Anatomy, Hokkaido University Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshie Kawabe
- Department of Anatomy and Neuroscience, Faculty of Medicine, Nara Medical University, Kashihara, Japan
| | - Shoko Morita-Takemura
- Department of Anatomy and Neuroscience, Faculty of Medicine, Nara Medical University, Kashihara, Japan
| | - Kazuki Nakahara
- Department of Anatomy and Neuroscience, Faculty of Medicine, Nara Medical University, Kashihara, Japan
| | - Yuki Terada
- Department of Anesthesiology, Faculty of Medicine, Nara Medical University, Kashihara, Japan
| | - Takeaki Shinjo
- Department of Anesthesiology, Faculty of Medicine, Nara Medical University, Kashihara, Japan
| | - Hiroaki Okuda
- Department of Functional Anatomy, Graduate School of Medical Science, Kanazawa University, Kanazawa, Japan
| | - Tatsuhide Tanaka
- Department of Anatomy and Neuroscience, Faculty of Medicine, Nara Medical University, Kashihara, Japan
| | - Akio Wanaka
- Department of Anatomy and Neuroscience, Faculty of Medicine, Nara Medical University, Kashihara, Japan
| |
Collapse
|
225
|
Scimemi A. Astrocytes and the Warning Signs of Intracerebral Hemorrhagic Stroke. Neural Plast 2018; 2018:7301623. [PMID: 29531526 PMCID: PMC5817320 DOI: 10.1155/2018/7301623] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 12/13/2017] [Indexed: 12/11/2022] Open
Abstract
Two decades into the two thousands, intracerebral hemorrhagic stroke (ICH) continues to reap lives across the globe. In the US, nearly 12,000 people suffer from ICH every year. Half of them survive, but many are left with permanent physical and cognitive disabilities, the severity of which depends on the location and broadness of the brain region affected by the hemorrhage. The ongoing efforts to identify risk factors for hemorrhagic stroke have been instrumental for the development of new medical practices to prevent, aid the recovery and reduce the risk of recurring ICH. Recent efforts approach the study of ICH from a different angle, providing information on how we can limit brain damage by manipulating astrocyte receptors. These results provide a novel understanding of how astrocytes contribute to brain injury and recovery from small ICH. Here, we discuss current knowledge on the risk factors and molecular pathology of ICH and the functional properties of astrocytes and their role in ICH. Last, we discuss candidate astrocyte receptors that may prove to be valuable therapeutic targets to treat ICH. Together, these findings provide basic and clinical scientists useful information for the future development of strategies to improve the detection of small ICH, limit brain damage, and prevent the onset of more severe episodes of brain hemorrhage.
Collapse
Affiliation(s)
- Annalisa Scimemi
- SUNY Albany, Department of Biology, 1400 Washington Avenue, Albany, NY 12222, USA
| |
Collapse
|
226
|
Schousboe A. Metabolic signaling in the brain and the role of astrocytes in control of glutamate and GABA neurotransmission. Neurosci Lett 2018; 689:11-13. [PMID: 29378296 DOI: 10.1016/j.neulet.2018.01.038] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2017] [Revised: 01/19/2018] [Accepted: 01/20/2018] [Indexed: 12/15/2022]
Abstract
Neurotransmission mediated by the two amino acids glutamate and GABA is based on recycling of the two signaling molecules between the presynaptic nerve endings and the surrounding astrocytes. During the recycling process, a fraction of the transmitter pool is lost since both transmitters undergo oxidative metabolism. This loss must be replenished by de novo synthesis which involves the action of pyruvate carboxylase, aminotransferases, glutamate dehydrogenase and glutamine synthetase. Among these enzymes, pyruvate carboxylase and glutamine synthetase are selectively expressed in astrocytes and thus these cells are obligatory partners in synaptic replenishment of both glutamate and GABA. The cycling processes also involve transporters for glutamate, GABA and glutamine and the operation of these transporters is discussed. Additionally, astrocytes appear to be essential for production of the neuromodulators, citrate, glycine and d-serine, aspects that will be briefly discussed.
Collapse
Affiliation(s)
- Arne Schousboe
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, 2 Universitetsparken, DK-2100, Copenhagen, Denmark.
| |
Collapse
|
227
|
Wilson CS, Mongin AA. The signaling role for chloride in the bidirectional communication between neurons and astrocytes. Neurosci Lett 2018; 689:33-44. [PMID: 29329909 DOI: 10.1016/j.neulet.2018.01.012] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 01/04/2018] [Accepted: 01/05/2018] [Indexed: 01/01/2023]
Abstract
It is well known that the electrical signaling in neuronal networks is modulated by chloride (Cl-) fluxes via the inhibitory GABAA and glycine receptors. Here, we discuss the putative contribution of Cl- fluxes and intracellular Cl- to other forms of information transfer in the CNS, namely the bidirectional communication between neurons and astrocytes. The manuscript (i) summarizes the generic functions of Cl- in cellular physiology, (ii) recaps molecular identities and properties of Cl- transporters and channels in neurons and astrocytes, and (iii) analyzes emerging studies implicating Cl- in the modulation of neuroglial communication. The existing literature suggests that neurons can alter astrocytic Cl- levels in a number of ways; via (a) the release of neurotransmitters and activation of glial transporters that have intrinsic Cl- conductance, (b) the metabotropic receptor-driven changes in activity of the electroneutral cation-Cl- cotransporter NKCC1, and (c) the transient, activity-dependent changes in glial cell volume which open the volume-regulated Cl-/anion channel VRAC. Reciprocally, astrocytes are thought to alter neuronal [Cl-]i through either (a) VRAC-mediated release of the inhibitory gliotransmitters, GABA and taurine, which open neuronal GABAA and glycine receptor/Cl- channels, or (b) the gliotransmitter-driven stimulation of NKCC1. The most important recent developments in this area are the identification of the molecular composition and functional heterogeneity of brain VRAC channels, and the discovery of a new cytosolic [Cl-] sensor - the Wnk family protein kinases. With new work in the field, our understanding of the role of Cl- in information processing within the CNS is expected to be significantly updated.
Collapse
Affiliation(s)
- Corinne S Wilson
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
| | - Alexander A Mongin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States; Department of Biophysics and Functional Diagnostics, Siberian State Medical University, Tomsk, Russian Federation.
| |
Collapse
|
228
|
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
|
229
|
The Cerebellar GABA AR System as a Potential Target for Treating Alcohol Use Disorder. Handb Exp Pharmacol 2018; 248:113-156. [PMID: 29736774 DOI: 10.1007/164_2018_109] [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] [Indexed: 12/14/2022]
Abstract
In the brain, fast inhibitory neurotransmission is mediated primarily by the ionotropic subtype of the gamma-aminobutyric acid (GABA) receptor subtype A (GABAAR). It is well established that the brain's GABAAR system mediates many aspects of neurobehavioral responses to alcohol (ethanol; EtOH). Accordingly, in both preclinical studies and some clinical scenarios, pharmacologically targeting the GABAAR system can alter neurobehavioral responses to acute and chronic EtOH consumption. However, many of the well-established interactions of EtOH and the GABAAR system have been identified at concentrations of EtOH ([EtOH]) that would only occur during abusive consumption of EtOH (≥40 mM), and there are still inadequate treatment options for prevention of or recovery from alcohol use disorder (AUD, including abuse and dependence). Accordingly, there is a general acknowledgement that more research is needed to identify and characterize: (1) neurobehavioral targets of lower [EtOH] and (2) associated brain structures that would involve such targets in a manner that may influence the development and maintenance of AUDs.Nearly 15 years ago it was discovered that the GABAAR system of the cerebellum is highly sensitive to EtOH, responding to concentrations as low as 10 mM (as would occur in the blood of a typical adult human after consuming 1-2 standard units of EtOH). This high sensitivity to EtOH, which likely mediates the well-known motor impairing effects of EtOH, combined with recent advances in our understanding of the role of the cerebellum in non-motor, cognitive/emotive/reward processes has renewed interest in this system in the specific context of AUD. In this chapter we will describe recent advances in our understanding of cerebellar processing, actions of EtOH on the cerebellar GABAAR system, and the potential relationship of such actions to the development of AUD. We will finish with speculation about how cerebellar specific GABAAR ligands might be effective pharmacological agents for treating aspects of AUD.
Collapse
|
230
|
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: 1077] [Impact Index Per Article: 153.9] [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
|
231
|
Effects of Ca2+, Glu and GABA on hBest1 and composite hBest1/POPC surface films. Colloids Surf B Biointerfaces 2018; 161:192-199. [DOI: 10.1016/j.colsurfb.2017.10.051] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 10/11/2017] [Accepted: 10/17/2017] [Indexed: 12/22/2022]
|
232
|
Rorsman P, Ashcroft FM. Pancreatic β-Cell Electrical Activity and Insulin Secretion: Of Mice and Men. Physiol Rev 2018; 98:117-214. [PMID: 29212789 PMCID: PMC5866358 DOI: 10.1152/physrev.00008.2017] [Citation(s) in RCA: 520] [Impact Index Per Article: 74.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 05/30/2017] [Accepted: 06/18/2017] [Indexed: 12/14/2022] Open
Abstract
The pancreatic β-cell plays a key role in glucose homeostasis by secreting insulin, the only hormone capable of lowering the blood glucose concentration. Impaired insulin secretion results in the chronic hyperglycemia that characterizes type 2 diabetes (T2DM), which currently afflicts >450 million people worldwide. The healthy β-cell acts as a glucose sensor matching its output to the circulating glucose concentration. It does so via metabolically induced changes in electrical activity, which culminate in an increase in the cytoplasmic Ca2+ concentration and initiation of Ca2+-dependent exocytosis of insulin-containing secretory granules. Here, we review recent advances in our understanding of the β-cell transcriptome, electrical activity, and insulin exocytosis. We highlight salient differences between mouse and human β-cells, provide models of how the different ion channels contribute to their electrical activity and insulin secretion, and conclude by discussing how these processes become perturbed in T2DM.
Collapse
Affiliation(s)
- Patrik Rorsman
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom; Department of Neuroscience and Physiology, Metabolic Research Unit, Göteborg, Sweden; and Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Frances M Ashcroft
- Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, United Kingdom; Department of Neuroscience and Physiology, Metabolic Research Unit, Göteborg, Sweden; and Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
233
|
Moldavan M, Cravetchi O, Allen CN. GABA transporters regulate tonic and synaptic GABA A receptor-mediated currents in the suprachiasmatic nucleus neurons. J Neurophysiol 2017; 118:3092-3106. [PMID: 28855287 PMCID: PMC5814714 DOI: 10.1152/jn.00194.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 08/23/2017] [Accepted: 08/24/2017] [Indexed: 11/22/2022] Open
Abstract
GABA is a principal neurotransmitter in the hypothalamic suprachiasmatic nucleus (SCN) that contributes to intercellular communication between individual circadian oscillators within the SCN network and the stability and precision of the circadian rhythms. GABA transporters (GAT) regulate the extracellular GABA concentration and modulate GABAA receptor (GABAAR)-mediated currents. GABA transport inhibitors were applied to study how GABAAR-mediated currents depend on the expression and function of GAT. Nipecotic acid inhibits GABA transport and induced an inward tonic current in concentration-dependent manner during whole cell patch-clamp recordings from SCN neurons. Application of either the selective GABA transporter 1 (GAT1) inhibitors NNC-711 or SKF-89976A, or the GABA transporter 3 (GAT3) inhibitor SNAP-5114, produced only small changes of the baseline current. Coapplication of GAT1 and GAT3 inhibitors induced a significant GABAAR-mediated tonic current that was blocked by gabazine. GAT inhibitors decreased the amplitude and decay time constant and increased the rise time of spontaneous GABAAR-mediated postsynaptic currents. However, inhibition of GAT did not alter the expression of either GAT1 or GAT3 in the hypothalamus. Thus GAT1 and GAT3 functionally complement each other to regulate the extracellular GABA concentration and GABAAR-mediated synaptic and tonic currents in the SCN. Coapplication of SKF-89976A and SNAP-5114 (50 µM each) significantly reduced the circadian period of Per1 expression in the SCN by 1.4 h. Our studies demonstrate that GAT are important regulators of GABAAR-mediated currents and the circadian clock in the SCN.NEW & NOTEWORTHY In the suprachiasmatic nucleus (SCN), the GABA transporters GAT1 and GAT3 are expressed in astrocytes. Inhibition of these GABA transporters increased a tonic GABA current and reduced the circadian period of Per1 expression in SCN neurons. GAT1 and GAT3 showed functional cooperativity: inhibition of one GAT increased the activity but not the expression of the other. Our data demonstrate that GABA transporters are important regulators of GABAA receptor-mediated currents and the circadian clock.
Collapse
Affiliation(s)
- Michael Moldavan
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon; and
| | - Olga Cravetchi
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon; and
| | - Charles N Allen
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, Oregon; and
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
234
|
Barros-Barbosa AR, Oliveira Â, Lobo MG, Cordeiro JM, Correia-de-Sá P. Under stressful conditions activation of the ionotropic P2X7 receptor differentially regulates GABA and glutamate release from nerve terminals of the rat cerebral cortex. Neurochem Int 2017; 112:81-95. [PMID: 29154812 DOI: 10.1016/j.neuint.2017.11.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 11/08/2017] [Accepted: 11/10/2017] [Indexed: 01/08/2023]
Abstract
γ-Aminobutyric acid (GABA) and glutamate (Glu) are the main inhibitory and excitatory neurotransmitters in the central nervous system (CNS), respectively. Fine tuning regulation of extracellular levels of these amino acids is essential for normal brain activity. Recently, we showed that neocortical nerve terminals from patients with epilepsy express higher amounts of the non-desensitizing ionotropic P2X7 receptor. Once activated by ATP released from neuronal cells, the P2X7 receptor unbalances GABAergic vs. glutamatergic neurotransmission by differentially interfering with GABA and Glu uptake. Here, we investigated if activation of the P2X7 receptor also affects [3H]GABA and [14C]Glu release measured synchronously from isolated nerve terminals (synaptosomes) of the rat cerebral cortex. Data show that activation of the P2X7 receptor consistently increases [14C]Glu over [3H]GABA release from cortical nerve terminals, but the GABA/Glu ratio depends on extracellular Ca2+ concentrations. While the P2X7-induced [3H]GABA release is operated by a Ca2+-dependent pathway when external Ca2+ is available, this mechanism shifts towards the reversal of the GAT1 transporter in low Ca2+ conditions. A different scenario is verified regarding [14C]Glu outflow triggered by the P2X7 receptor, since the amino acid seems to be consistently released through the recruitment of connexin-containing hemichannels upon P2X7 activation, both in the absence and in the presence of external Ca2+. Data from this study add valuable information suggesting that ATP, via P2X7 activation, not only interferes with the high-affinity uptake of GABA and Glu but actually favors the release of these amino acids through distinct molecular mechanisms amenable to differential therapeutic control.
Collapse
Affiliation(s)
- Aurora R Barros-Barbosa
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
| | - Ângela Oliveira
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
| | - M Graça Lobo
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
| | - J Miguel Cordeiro
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal
| | - Paulo Correia-de-Sá
- Laboratório de Farmacologia e Neurobiologia, Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal; Center for Drug Discovery and Innovative Medicines (MedInUP), Instituto de Ciências Biomédicas de Abel Salazar (ICBAS), Universidade do Porto (UP), Porto, Portugal.
| |
Collapse
|
235
|
Direct interaction with 14-3-3γ promotes surface expression of Best1 channel in astrocyte. Mol Brain 2017; 10:51. [PMID: 29121962 PMCID: PMC5679146 DOI: 10.1186/s13041-017-0331-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 10/28/2017] [Indexed: 01/01/2023] Open
Abstract
Background Bestrophin-1 (Best1) is a calcium-activated anion channel (CAAC) that is expressed broadly in mammalian tissues including the brain. We have previously reported that Best1 is expressed in hippocampal astrocytes at the distal peri-synaptic regions, called microdomains, right next to synaptic junctions, and that it disappears from the microdomains in Alzheimer’s disease mouse model. Although Best1 appears to be dynamically regulated, the mechanism of its regulation and modulation is poorly understood. It has been reported that a regulatory protein, 14-3-3 affects the surface expression of numerous membrane proteins in mammalian cells. Methods The protein-protein interaction between Best1 and 14-3-3γ was confirmed by yeast-two hybrid assay and BiFC method. The effect of 14-3-3γ on Best1-mediated current was measured by whole-cell patch clamp technique. Results We identified 14-3-3γ as novel binding partner of Best1 in astrocytes: among 7 isoforms of 14-3-3 protein, only 14-3-3γ was found to bind specifically. We determined a binding domain on the C-terminus of Best1 which is critical for an interaction with 14-3-3γ. We also revealed that interaction between Best1 and 14-3-3γ was mediated by phosphorylation of S358 in the C-terminus of Best1. We confirmed that surface expression of Best1 and Best1-mediated whole-cell current were significantly decreased after a gene-silencingof 14-3-3γ without a significant change in total Best1 expression in cultured astrocytes. Furthermore, we discovered that 14-3-3γ-shRNA reduced Best1-mediated glutamate release from hippocampal astrocyte by recording a PAR1 receptor-induced NMDA receptor-mediated current from CA1 pyramidal neurons in hippocampal slices injected with adenovirus carrying 14-3-3γ-shRNA. Finally, through a structural modeling, we found critical amino acid residues containing S358 of Best1 exhibiting binding affinities to 14-3-3γ. Conclusions 14-3-3γ promotes surface expression of Best1 channel in astrocytes through direct interaction.
Collapse
|
236
|
Brown DA. Norman Bowery's discoveries about extrasynaptic and asynaptic GABA systems and their significance. Neuropharmacology 2017; 136:3-9. [PMID: 29128306 DOI: 10.1016/j.neuropharm.2017.11.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 11/04/2017] [Indexed: 11/26/2022]
Abstract
Before discovering the GABA-B receptor, Norman Bowery completed a series of studies on an extrasynaptic or asynaptic "GABA system" in the rat superior cervical sympathetic ganglion. First, he discovered an uptake system for GABA in neuroglial cells in the ganglia and in peripheral nerves, with a different substrate specificity than that in neurons. Second, he showed that accumulated GABA in sympathetic glial cells was metabolized to succinate by a transaminase enzyme. Third, he provided detailed structure-activity information about compounds activating an extrasynaptic GABA-A receptor on neurons in the rat sympathetic ganglion. Fourth, he showed that some amino acid substrates for the neuroglial transporter could indirectly stimulate neurons by releasing GABA from adjacent glial cells, and that GABA could also be released from neuroglial cells by membrane depolarization. In this review, these discoveries are briefly described and updated and some of their implications assessed. This article is part of the "Special Issue Dedicated to Norman G. Bowery".
Collapse
Affiliation(s)
- David A Brown
- Department of Neuroscience, Physiology & Pharmacology, University College London, London WC1E 6BT, UK.
| |
Collapse
|
237
|
Choi DJ, Eun JH, Kim BG, Jou I, Park SM, Joe EH. A Parkinson's disease gene, DJ-1, repairs brain injury through Sox9 stabilization and astrogliosis. Glia 2017; 66:445-458. [PMID: 29105838 DOI: 10.1002/glia.23258] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 10/23/2017] [Accepted: 10/24/2017] [Indexed: 12/13/2022]
Abstract
Defects in repair of damaged brain accumulate injury and contribute to slow-developing neurodegeneration. Here, we report that a deficiency of DJ-1, a Parkinson's disease (PD) gene, delays repair of brain injury due to destabilization of Sox9, a positive regulator of astrogliosis. Stereotaxic injection of ATP into the brain striatum produces similar size of acute injury in wild-type and DJ-1-knockout (KO) mice. However, recovery of the injury is delayed in KO mice, which is confirmed by 9.4T magnetic resonance imaging and tyrosine hydroxylase immunostaining. DJ-1 regulates neurite outgrowth from damaged neurons in a non-cell autonomous manner. In DJ-1 KO brains and astrocytes, Sox9 protein levels are decreased due to enhanced ubiquitination, resulting in defects in astrogliosis and glial cell-derived neurotrophic factor/ brain-derived neurotrophic factor expression in injured brain and astrocytes. These results indicate that DJ-1 deficiency causes defects in astrocyte-mediated repair of brain damage, which may contribute to the development of PD.
Collapse
Affiliation(s)
- Dong-Joo Choi
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| | - Jin-Hwa Eun
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| | - Byung Gon Kim
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Brain Science, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Neurology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| | - Ilo Jou
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| | - Sang Myun Park
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| | - Eun-Hye Joe
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Pharmacology, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Chronic Inflammatory Disease Research Center, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea.,Department of Brain Science, Ajou University School of Medicine, Worldcup-ro 164, Suwon, Kyunggi-do, 16499, Korea
| |
Collapse
|
238
|
Li Y, Zhang Y, Xu Y, Kittredge A, Ward N, Chen S, Tsang SH, Yang T. Patient-specific mutations impair BESTROPHIN1's essential role in mediating Ca 2+-dependent Cl - currents in human RPE. eLife 2017; 6. [PMID: 29063836 PMCID: PMC5655127 DOI: 10.7554/elife.29914] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 09/13/2017] [Indexed: 01/06/2023] Open
Abstract
Mutations in the human BEST1 gene lead to retinal degenerative diseases displaying progressive vision loss and even blindness. BESTROPHIN1, encoded by BEST1, is predominantly expressed in retinal pigment epithelium (RPE), but its physiological role has been a mystery for the last two decades. Using a patient-specific iPSC-based disease model and interdisciplinary approaches, we comprehensively analyzed two distinct BEST1 patient mutations, and discovered mechanistic correlations between patient clinical phenotypes, electrophysiology in their RPEs, and the structure and function of BESTROPHIN1 mutant channels. Our results revealed that the disease-causing mechanism of BEST1 mutations is centered on the indispensable role of BESTROPHIN1 in mediating the long speculated Ca2+-dependent Cl- current in RPE, and demonstrate that the pathological potential of BEST1 mutations can be evaluated and predicted with our iPSC-based 'disease-in-a-dish' approach. Moreover, we demonstrated that patient RPE is rescuable with viral gene supplementation, providing a proof-of-concept for curing BEST1-associated diseases.
Collapse
Affiliation(s)
- Yao Li
- Jonas Children's Vision Care, and Bernard and Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology and Pathology & Cell Biology, Edward S. Harkness Eye Institute, New York Presbyterian Hospital/Columbia University, New York, United States
| | - Yu Zhang
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, Rochester, United States
| | - Yu Xu
- Jonas Children's Vision Care, and Bernard and Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology and Pathology & Cell Biology, Edward S. Harkness Eye Institute, New York Presbyterian Hospital/Columbia University, New York, United States.,Department of Ophthalmology, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Alec Kittredge
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, Rochester, United States
| | - Nancy Ward
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, Rochester, United States
| | - Shoudeng Chen
- Molecular Imaging Center, Department of Experimental Medicine, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Stephen H Tsang
- Jonas Children's Vision Care, and Bernard and Shirlee Brown Glaucoma Laboratory, Department of Ophthalmology and Pathology & Cell Biology, Edward S. Harkness Eye Institute, New York Presbyterian Hospital/Columbia University, New York, United States
| | - Tingting Yang
- Department of Pharmacology and Physiology, School of Medicine and Dentistry, University of Rochester, Rochester, United States
| |
Collapse
|
239
|
Brawek B, Chesters R, Klement D, Müller J, Lerdkrai C, Hermes M, Garaschuk O. A bell-shaped dependence between amyloidosis and GABA accumulation in astrocytes in a mouse model of Alzheimer's disease. Neurobiol Aging 2017; 61:187-197. [PMID: 29107186 DOI: 10.1016/j.neurobiolaging.2017.09.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/25/2017] [Accepted: 09/27/2017] [Indexed: 10/18/2022]
Abstract
Functioning at the interface between the nervous and immune systems, in the amyloid-depositing brain, astrocytes become hypertrophic and accumulate around senile plaques. Moreover, hippocampal astrocytes upregulate their γ-aminobutyric acid (GABA) content and enhance tonic inhibition, likely causing local circuit imbalance. It remains, however, unclear whether this effect is hippocampus specific and how it is regulated during disease progression. Here, we studied changes in astrocytic morphology and GABA content in the frontal cortex and dentate gyrus of control and amyloid-depositing mice. Healthy aging was accompanied by a transient increase in astrocytic GABA content at middle age and region-specific alterations of soma size. In contrast, amyloid deposition caused a gradual cortex-accentuated increase in soma size. Importantly, our data uncovered a bell-shaped relationship between the mouse age and astrocytic GABA content in both brain regions. Moreover, in mice carrying an Alzheimer's disease-related mutation in presenilin 1, astrocytes accumulated GABA even in the absence of amyloidosis. These data question the proposed inhibition of astrocytic GABA synthesis as a universal strategy for treating network dysfunction in Alzheimer's disease.
Collapse
Affiliation(s)
- Bianca Brawek
- Department of Neurophysiology, Institute of Physiology, University of Tübingen, Tübingen, Germany
| | - Robert Chesters
- Department of Neurophysiology, Institute of Physiology, University of Tübingen, Tübingen, Germany
| | - Daniel Klement
- Department of Neurophysiology, Institute of Physiology, University of Tübingen, Tübingen, Germany
| | - Julia Müller
- Department of Neurophysiology, Institute of Physiology, University of Tübingen, Tübingen, Germany
| | - Chommanad Lerdkrai
- Department of Neurophysiology, Institute of Physiology, University of Tübingen, Tübingen, Germany
| | - Marina Hermes
- Department of Neurophysiology, Institute of Physiology, University of Tübingen, Tübingen, Germany
| | - Olga Garaschuk
- Department of Neurophysiology, Institute of Physiology, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
240
|
Eto K, Kim SK, Takeda I, Nabekura J. The roles of cortical astrocytes in chronic pain and other brain pathologies. Neurosci Res 2017; 126:3-8. [PMID: 28870605 DOI: 10.1016/j.neures.2017.08.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 08/01/2017] [Accepted: 08/18/2017] [Indexed: 01/21/2023]
Abstract
Astrocytes are the most abundant cell type in the brain. Several decades ago, they were considered to be only support cells in the central nervous system. Recent studies using advanced technologies have clarified that astrocytes play more active roles in regulating neuronal function and remodeling synaptic structures by releasing molecules called gliotransmitters. In addition to various physiological functions, astrocytes are activated under disease conditions, such as chronic pain, releasing molecules that in turn cause reorganization of the central nervous system microstructure and disrupt behavior in pathological conditions. In the present review, we summarize cortical astrocyte function in chronic pain and other neurological disorders and discuss the role of astrocytes in brain pathologies.
Collapse
Affiliation(s)
- Kei Eto
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences, The Graduate School for Advanced Studies, Hayama, Kanagawa 240-0193, Japan
| | - Sun Kwang Kim
- Department of Physiology, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ikuko Takeda
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan
| | - Junichi Nabekura
- Division of Homeostatic Development, National Institute for Physiological Sciences, Okazaki, Aichi 444-8585, Japan; Department of Physiological Sciences, The Graduate School for Advanced Studies, Hayama, Kanagawa 240-0193, Japan; Core Research for Evolutional Science and Technology, Japan Science and Technology Agency, Tokyo 102-0076, Japan.
| |
Collapse
|
241
|
Kim YS, Woo J, Lee CJ, Yoon BE. Decreased Glial GABA and Tonic Inhibition in Cerebellum of Mouse Model for Attention-Deficit/Hyperactivity Disorder (ADHD). Exp Neurobiol 2017; 26:206-212. [PMID: 28912643 PMCID: PMC5597551 DOI: 10.5607/en.2017.26.4.206] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 08/23/2017] [Accepted: 08/23/2017] [Indexed: 01/19/2023] Open
Abstract
About 5~12% of school-aged children suffer from the Attention-Deficit/Hyperactivity Disorder (ADHD). However, the core mechanism of ADHD remains unclear. G protein-coupled receptor kinase-interacting protein-1 (GIT1) has recently been reported to be associated with ADHD in human and the genetic deletion of GIT1 result in ADHD-like behaviors in mice. Mice lacking GIT1 shows a shift in neuronal excitation/inhibition (E/I) balance. However, the pricise mechanism for E/I imbalance and the role of neuron-glia interaction in GIT1 knockout (KO) mice have not been studied. Especially, a possible contribution of glial GABA and tonic inhibition mediated by astrocytic GABA release in the mouse model for ADHD remains unexplored. Therefore, we investigated the changes in the amount of GABA and degree of tonic inhibition in GIT1 KO mice. We observed a decreased glial GABA intensity in GIT1 KO mice compared to wild type (WT) mice and an attenuation of tonic current from cerebellar granule cells in GIT1 KO mice. Our study identifies the previously unknown mechanism of reduced astrocytic GABA and tonic inhibition in GIT1 lacking mice as a potential cause of hyperactivity disorder.
Collapse
Affiliation(s)
- Yoo Sung Kim
- Department of Molecular biology, Dankook University, Cheonan, Chungnam 31116, Korea
| | - Junsung Woo
- Center for Neuroscience and Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.,Neuroscience Program, University of Science and Technology (UST), Daejeon 34113, Korea
| | - C Justin Lee
- Center for Neuroscience and Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.,Neuroscience Program, University of Science and Technology (UST), Daejeon 34113, Korea.,KU-KIST Graduate School of Converging Sciences and Technologies, Korea University, Seoul 02841, Korea
| | - Bo-Eun Yoon
- Department of Molecular biology, Dankook University, Cheonan, Chungnam 31116, Korea
| |
Collapse
|
242
|
Chen X, Keramidas A, Lynch JW. Physiological and pharmacological properties of inhibitory postsynaptic currents mediated by α5β1γ2, α5β2γ2 and α5β3γ2 GABA A receptors. Neuropharmacology 2017; 125:243-253. [PMID: 28757051 DOI: 10.1016/j.neuropharm.2017.07.027] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 07/18/2017] [Accepted: 07/26/2017] [Indexed: 02/07/2023]
Abstract
α5-containing GABAARs are potential therapeutic targets for clinical conditions including age-related dementia, stroke, schizophrenia, Down syndrome, anaesthetic-induced amnesia, anxiety and pain. α5-containing GABAARs are expressed in layer 5 cortical neurons and hippocampal pyramidal neurons where they mediate both tonic currents and slow inhibitory postsynaptic currents (IPSCs). A range of drugs has been developed to specifically modulate these receptors. The main α5-containing GABAARs that are likely to exist in vivo are the α5β1γ2, α5β2γ2 and α5β3γ2 isoforms. We currently have few clues as to how these isoforms are distributed between synaptic and extrasynaptic compartments or their relative roles in controlling neuronal excitability. Accordingly, the aim of this study was to define the basic biophysical and pharmacological properties of IPSCs mediated by the three isoforms in a hippocampal neuron-HEK293 cell co-culture assay. The IPSC decay time constants were slow (α5β1γ2L: 45 ms; α5β1γ2L: 80 ms; α5β3γ2L: 184 ms) and were largely dominated by the intrinsic channel deactivation rates. By comparing IPSC rise times, we inferred that α5β1γ2L GABAARs are located postsynaptically whereas the other two are predominantly perisynaptic. α5β3γ2L GABAARs alone mediated tonic currents. We quantified the effects of four α5-specific inverse agonists (TB-21007, MRK-016, α5IA and L-655708) on IPSCs mediated by the three isoforms. All compounds selectively inhibited IPSC amplitudes and accelerated IPSC decay rates, albeit with distinct isoform specificities. MRK-016 also significantly accelerated IPSC rise times. These results provide a reference for future studies seeking to identify and characterize the properties of IPSCs mediated by α5-containing GABAAR isoforms in neurons.
Collapse
Affiliation(s)
- Xiumin Chen
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Angelo Keramidas
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Joseph W Lynch
- Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia.
| |
Collapse
|
243
|
Szabó Z, Héja L, Szalay G, Kékesi O, Füredi A, Szebényi K, Dobolyi Á, Orbán TI, Kolacsek O, Tompa T, Miskolczy Z, Biczók L, Rózsa B, Sarkadi B, Kardos J. Extensive astrocyte synchronization advances neuronal coupling in slow wave activity in vivo. Sci Rep 2017; 7:6018. [PMID: 28729692 PMCID: PMC5519671 DOI: 10.1038/s41598-017-06073-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 05/26/2017] [Indexed: 01/19/2023] Open
Abstract
Slow wave activity (SWA) is a characteristic brain oscillation in sleep and quiet wakefulness. Although the cell types contributing to SWA genesis are not yet identified, the principal role of neurons in the emergence of this essential cognitive mechanism has not been questioned. To address the possibility of astrocytic involvement in SWA, we used a transgenic rat line expressing a calcium sensitive fluorescent protein in both astrocytes and interneurons and simultaneously imaged astrocytic and neuronal activity in vivo. Here we demonstrate, for the first time, that the astrocyte network display synchronized recurrent activity in vivo coupled to UP states measured by field recording and neuronal calcium imaging. Furthermore, we present evidence that extensive synchronization of the astrocytic network precedes the spatial build-up of neuronal synchronization. The earlier extensive recruitment of astrocytes in the synchronized activity is reinforced by the observation that neurons surrounded by active astrocytes are more likely to join SWA, suggesting causality. Further supporting this notion, we demonstrate that blockade of astrocytic gap junctional communication or inhibition of astrocytic Ca2+ transients reduces the ratio of both astrocytes and neurons involved in SWA. These in vivo findings conclusively suggest a causal role of the astrocytic syncytium in SWA generation.
Collapse
Affiliation(s)
- Zsolt Szabó
- Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117, Budapest, Hungary
| | - László Héja
- Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117, Budapest, Hungary.
| | - Gergely Szalay
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony 43, 1083, Budapest, Hungary
| | - Orsolya Kékesi
- Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117, Budapest, Hungary
| | - András Füredi
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117, Budapest, Hungary.,Institute of Cancer Research, Medical University Wien, Borschkegasse 8a, 1090, Wien, Austria
| | - Kornélia Szebényi
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117, Budapest, Hungary.,Institute of Cancer Research, Medical University Wien, Borschkegasse 8a, 1090, Wien, Austria
| | - Árpád Dobolyi
- MTA-ELTE Laboratory of Molecular and Systems Neurobiology, Department of Physiology and Neurobiology, Eötvös Loránd University, Pázmány Péter sétány 1C, 1117, Budapest, Hungary
| | - Tamás I Orbán
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117, Budapest, Hungary
| | - Orsolya Kolacsek
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117, Budapest, Hungary
| | - Tamás Tompa
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony 43, 1083, Budapest, Hungary
| | - Zsombor Miskolczy
- Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117, Budapest, Hungary
| | - László Biczók
- Institute of Materials and Environmental Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117, Budapest, Hungary
| | - Balázs Rózsa
- Institute of Experimental Medicine, Hungarian Academy of Sciences, Szigony 43, 1083, Budapest, Hungary
| | - Balázs Sarkadi
- Institute of Enzymology, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117, Budapest, Hungary
| | - Julianna Kardos
- Institute of Organic Chemistry, Research Centre for Natural Sciences, Hungarian Academy of Sciences, Magyar tudósok körútja 2, 1117, Budapest, Hungary
| |
Collapse
|
244
|
Optogenetic Visualization of Presynaptic Tonic Inhibition of Cerebellar Parallel Fibers. J Neurosci 2017; 36:5709-23. [PMID: 27225762 DOI: 10.1523/jneurosci.4366-15.2016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 03/31/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Tonic inhibition was imaged in cerebellar granule cells of transgenic mice expressing the optogenetic chloride indicator, Clomeleon. Blockade of GABAA receptors substantially reduced chloride concentration in granule cells due to block of tonic inhibition. This indicates that tonic inhibition is a significant contributor to the resting chloride concentration of these cells. Tonic inhibition was observed not only in granule cell bodies, but also in their axons, the parallel fibers (PFs). This presynaptic tonic inhibition could be observed in slices both at room and physiological temperatures, as well as in vivo, and has many of the same properties as tonic inhibition measured in granule cell bodies. GABA application revealed that PFs possess at least two types of GABAA receptor: one high-affinity receptor that is activated by ambient GABA and causes a chloride influx that mediates tonic inhibition, and a second with a low affinity for GABA that causes a chloride efflux that excites PFs. Presynaptic tonic inhibition regulates glutamate release from PFs because GABAA receptor blockade enhanced both the frequency of spontaneous EPSCs and the amplitude of evoked EPSCs at the PF-Purkinje cell synapse. We conclude that tonic inhibition of PFs could play an important role in regulating information flow though cerebellar synaptic circuits. Such cross talk between phasic and tonic signaling could be a general mechanism for fine tuning of synaptic circuits. SIGNIFICANCE STATEMENT This paper demonstrates that an unconventional form of signaling, known as tonic inhibition, is found in presynaptic terminals and affects conventional synaptic communication. Our results establish the basic characteristics and mechanisms of presynaptic tonic inhibition and show that it occurs in vivo as well as in isolated brain tissue.
Collapse
|
245
|
Oh SJ, Lee CJ. Distribution and Function of the Bestrophin-1 (Best1) Channel in the Brain. Exp Neurobiol 2017; 26:113-121. [PMID: 28680296 PMCID: PMC5491579 DOI: 10.5607/en.2017.26.3.113] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Revised: 06/08/2017] [Accepted: 06/08/2017] [Indexed: 12/29/2022] Open
Abstract
Bestrophin-1 (Best1) is a calcium-activated anion channel identified from retinal pigment epithelium where human mutations are associated with Best's macular degeneration. Best1 is known to be expressed in a variety of tissues including the brain, and is thought to be involved in many physiological processes. This review focuses on the current state of knowledge on aspects of expression and function of Best1 in the brain. Best1 protein is observed in cortical and hippocampal astrocytes, in cerebellar Bergmann glia and lamellar astrocytes, in thalamic reticular neurons, in meninges and in the epithelial cells of the choroid plexus. The most prominent feature of Best1 is its significant permeability to glutamate and GABA in addition to chloride ions because glutamate and GABA are important transmitters in the brain. Under physiological conditions, both Best1-mediated glutamate release and tonic GABA release from astrocytes modulate neuronal excitability, synaptic transmission and synaptic plasticity. Under pathological conditions such as neuroinflammation and neurodegeneration, reactive astrocytes phenotypically switch from GABA-negative to GABA-producing and redistribute Best1 from the perisynaptic microdomains to the soma and processes to tonically release GABA via Best1. This implicates that tonic GABA release from reactive astrocyte via redistributed Best1 is a common phenomenon that occur in various pathological conditions with astrogliosis such as traumatic brain injury, neuroinflammation, neurodegeneration, and hypoxic and ischemic insults. These properties of Best1, including the permeation and release of glutamate and GABA and its redistribution in reactive astrocytes, promise us exciting discoveries of novel brain functions to be uncovered in the future.
Collapse
Affiliation(s)
- Soo-Jin Oh
- Center for Neuroscience, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.,Convergence Research Center for Diagnosis, Treatment and Care System of Dementia, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.,Center for Glia-Neuron Interaction, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| | - C Justin Lee
- Center for Neuroscience, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea.,Center for Glia-Neuron Interaction, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea
| |
Collapse
|
246
|
Opening a New Time Window for Treatment of Stroke by Targeting HDAC2. J Neurosci 2017; 37:6712-6728. [PMID: 28592694 DOI: 10.1523/jneurosci.0341-17.2017] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 05/10/2017] [Accepted: 06/01/2017] [Indexed: 11/21/2022] Open
Abstract
Narrow therapeutic window limits treatments with thrombolysis and neuroprotection for most stroke patients. Widening therapeutic window remains a critical challenge. Understanding the key mechanisms underlying the pathophysiological events in the peri-infarct area where secondary injury coexists with neuroplasticity over days to weeks may offer an opportunity for expanding the therapeutic window. Here we show that ischemia-induced histone deacetylase 2 (HDAC2) upregulation from 5 to 7 d after stroke plays a crucial role. In this window phase, suppressing HDAC2 in the peri-infarct cortex of rodents by HDAC inhibitors, knockdown or knock-out of Hdac2 promoted recovery of motor function from stroke via epigenetically enhancing cells survival and neuroplasticity of surviving neurons as well as reducing neuroinflammation, whereas overexpressing HDAC2 worsened stroke-induced functional impairment of both WT and Hdac2 conditional knock-out mice. More importantly, inhibiting other isoforms of HDACs had no effect. Thus, the intervention by precisely targeting HDAC2 in this window phase is a novel strategy for the functional recovery of stroke survivors.SIGNIFICANCE STATEMENT Narrow time window phase impedes current therapies for stroke patients. Understanding the key mechanisms underlying secondary injury may open a new window for pharmacological interventions to promote recovery from stroke. Our study indicates that ischemia-induced histone deacetylase 2 upregulation from 5 to 7 d after stroke mediates the secondary functional loss by reducing survival and neuroplasticity of peri-infarct neurons as well as augmenting neuroinflammation. Thus, precisely targeting histone deacetylase 2 in the window phase provides a novel therapeutic strategy for stroke recovery.
Collapse
|
247
|
Min JO, Yoon BE. Glia and gliotransmitters on carbon nanotubes. NANO REVIEWS & EXPERIMENTS 2017; 8:1323853. [PMID: 30410703 PMCID: PMC6167025 DOI: 10.1080/20022727.2017.1323853] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Revised: 11/20/2016] [Accepted: 05/11/2017] [Indexed: 11/11/2022]
Abstract
Introduction: Functionalised carbon nanotubes (CNTs) have been shown to be promising biomaterials in neural systems, such as CNT -based nerve scaffolds to drive nerve regeneration. CNTs have been shown to modulate neuronal growth and improve electrical conductivity of neurons. Methods: Cultured astrocytes on the functionalized CNTs (PEG, caroboxyl group) were assessed for distribution of GABA, glutamate uptake assay using isotope and change of conductance of CNTs by ATP. Immunostaining of GABA using anti-GABA (red), anti-GFAP (green) antibody in primary cortical astrocytes on MW-CNT and PDL coverslips. Results: The functionalization of CNTs has improved their solubility and biocompatibility and alters their cellular interaction pathways. Recently, CNTs have been shown to modulate morphofunctional characteristics of glia as well as neurons. Among the various types of glia, astrocytes express diverse receptors for corresponding neurotransmitters and release gliotransmitters, including glutamate, adenosine triphosphate, and γ-amino butyric acid. Gliotransmitters are primarily released from astrocytes and play important roles in glia–neuron crosstalk. Conclusion: This review focuses on the effects of CNTs on glial cells and discusses how functionalized CNTs can modulate morphology and gliotransmitters of glial cells. Based on exciting new findings, they look to be a promising material for use in brain disease therapy or neuroprosthetics.
Collapse
Affiliation(s)
- Joo-Ok Min
- Department of Nanobiomedical Science, Dankook University, Cheonan-si, Chungnam, Republic of Korea
| | - Bo-Eun Yoon
- Department of Nanobiomedical Science, Dankook University, Cheonan-si, Chungnam, Republic of Korea.,Department of Molecular Biology, Dankook University, Cheonan-si, Chungnam, Republic of Korea
| |
Collapse
|
248
|
Acosta C, Anderson HD, Anderson CM. Astrocyte dysfunction in Alzheimer disease. J Neurosci Res 2017; 95:2430-2447. [PMID: 28467650 DOI: 10.1002/jnr.24075] [Citation(s) in RCA: 177] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 04/07/2017] [Accepted: 04/10/2017] [Indexed: 12/11/2022]
Abstract
Astrocytes are glial cells that are distributed throughout the central nervous system in an arrangement optimal for chemical and physical interaction with neuronal synapses and brain blood supply vessels. Neurotransmission modulates astrocytic excitability by activating an array of cell surface receptors and transporter proteins, resulting in dynamic changes in intracellular Ca2+ or Na+ . Ionic and electrogenic astrocytic changes, in turn, drive vital cell nonautonomous effects supporting brain function, including regulation of synaptic activity, neuronal metabolism, and regional blood supply. Alzheimer disease (AD) is associated with aberrant oligomeric amyloid β generation, which leads to extensive proliferation of astrocytes with a reactive phenotype and abnormal regulation of these processes. Astrocytic morphology, Ca2+ responses, extracellular K+ removal, glutamate transport, amyloid clearance, and energy metabolism are all affected in AD, resulting in a deleterious set of effects that includes glutamate excitotoxicity, impaired synaptic plasticity, reduced carbon delivery to neurons for oxidative phosphorylation, and dysregulated linkages between neuronal energy demand and regional blood supply. This review summarizes how astrocytes are affected in AD and describes how these changes are likely to influence brain function. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Crystal Acosta
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.,Canadian Centre for Agri-food Research in Health and Medicine, St. Boniface Hospital Research, Winnipeg, Manitoba, Canada
| | - Hope D Anderson
- Canadian Centre for Agri-food Research in Health and Medicine, St. Boniface Hospital Research, Winnipeg, Manitoba, Canada.,College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Christopher M Anderson
- Department of Pharmacology and Therapeutics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada.,Neuroscience Research Program, Kleysen Institute for Advanced Medicine, Health Sciences Centre, Winnipeg, Manitoba, Canada
| |
Collapse
|
249
|
Du X, Hao H, Yang Y, Huang S, Wang C, Gigout S, Ramli R, Li X, Jaworska E, Edwards I, Deuchars J, Yanagawa Y, Qi J, Guan B, Jaffe DB, Zhang H, Gamper N. Local GABAergic signaling within sensory ganglia controls peripheral nociceptive transmission. J Clin Invest 2017; 127:1741-1756. [PMID: 28375159 PMCID: PMC5409786 DOI: 10.1172/jci86812] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 02/02/2017] [Indexed: 01/05/2023] Open
Abstract
The integration of somatosensory information is generally assumed to be a function of the central nervous system (CNS). Here we describe fully functional GABAergic communication within rodent peripheral sensory ganglia and show that it can modulate transmission of pain-related signals from the peripheral sensory nerves to the CNS. We found that sensory neurons express major proteins necessary for GABA synthesis and release and that sensory neurons released GABA in response to depolarization. In vivo focal infusion of GABA or GABA reuptake inhibitor to sensory ganglia dramatically reduced acute peripherally induced nociception and alleviated neuropathic and inflammatory pain. In addition, focal application of GABA receptor antagonists to sensory ganglia triggered or exacerbated peripherally induced nociception. We also demonstrated that chemogenetic or optogenetic depolarization of GABAergic dorsal root ganglion neurons in vivo reduced acute and chronic peripherally induced nociception. Mechanistically, GABA depolarized the majority of sensory neuron somata, yet produced a net inhibitory effect on the nociceptive transmission due to the filtering effect at nociceptive fiber T-junctions. Our findings indicate that peripheral somatosensory ganglia represent a hitherto underappreciated site of somatosensory signal integration and offer a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Xiaona Du
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Han Hao
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Yuehui Yang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Sha Huang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Caixue Wang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Sylvain Gigout
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Rosmaliza Ramli
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
- School of Dental Sciences, Universiti Sains Malaysia, Kelantan, Malaysia
| | - Xinmeng Li
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Ewa Jaworska
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Ian Edwards
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Jim Deuchars
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| | - Yuchio Yanagawa
- Department of Genetic and Behavioral Neuroscience, Gunma University Graduate School of Medicine and Japan Science and Technology Agency, CREST, Maebashi, Japan
| | - Jinlong Qi
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Bingcai Guan
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - David B. Jaffe
- Department of Biology, UTSA Neurosciences Institute, University of Texas at San Antonio, San Antonio, Texas, USA
| | - Hailin Zhang
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
| | - Nikita Gamper
- Department of Pharmacology, Hebei Medical University, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Province; Shijiazhuang, China
- Faculty of Biological Sciences, University of Leeds, Leeds, United Kingdom
| |
Collapse
|
250
|
Ransom CB, Ye Z, Spain WJ, Richerson GB. Modulation of Tonic GABA Currents by Anion Channel and Connexin Hemichannel Antagonists. Neurochem Res 2017; 42:2551-2559. [PMID: 28401401 DOI: 10.1007/s11064-017-2246-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2016] [Revised: 03/17/2017] [Accepted: 03/22/2017] [Indexed: 12/19/2022]
Abstract
Anion channels and connexin hemichannels are permeable to amino acid neurotransmitters. It is hypothesized that these conductive pathways release GABA, thereby influencing ambient GABA levels and tonic GABAergic inhibition. To investigate this, we measured the effects of anion channel/hemichannel antagonists on tonic GABA currents of rat hippocampal neurons. In contrast to predictions, blockade of anion channels and hemichannels with NPPB potentiated tonic GABA currents of neurons in culture and acute hippocampal slices. In contrast, the anion channel/hemichannel antagonist carbenoxolone (CBX) inhibited tonic currents. These findings could result from alterations of ambient GABA concentration or direct effects on GABAA receptors. To test for effects on GABAA receptors, we measured currents evoked by exogenous GABA. Coapplication of NPPB with GABA potentiated GABA-evoked currents. CBX dose-dependently inhibited GABA-evoked currents. These results are consistent with direct effects of NPPB and CBX on GABAA receptors. GABA release from hippocampal cell cultures was directly measured using HPLC. Inhibition of anion channels with NPPB or CBX did not affect GABA release from cultured hippocampal neurons. NPPB reduced GABA release from pure astrocytic cultures by 21%, but the total GABA release from astrocytes was small compared to that of mixed cultures. These data indicate that drugs commonly used to antagonize anion channels and connexin hemichannels may affect tonic currents via direct effects on GABAA receptors and have negligible effects on ambient GABA concentrations. Interpretation of experiments using NPPB or CBX should include consideration of their effects on tonic GABA currents.
Collapse
Affiliation(s)
- Christopher B Ransom
- Epilepsy Center of Excellence, VA Puget Sound HealthCare System, Seattle, WA, 98108, USA. .,Department of Neurology, University of Washington, Seattle, WA, 98105, USA. .,Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA.
| | - Zucheng Ye
- Department of Neurology, University of Washington, Seattle, WA, 98105, USA
| | - William J Spain
- Epilepsy Center of Excellence, VA Puget Sound HealthCare System, Seattle, WA, 98108, USA.,Department of Neurology, University of Washington, Seattle, WA, 98105, USA.,Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - George B Richerson
- Department of Neurology, University of Iowa, Iowa City, IA, 52242, USA.,Department of Molecular Physiology and Biophysics, University of Iowa, Iowa City, IA, USA
| |
Collapse
|