1
|
Li YL, Lin J, Huang X, Zeng RH, Zhang G, Xu JN, Lin KJ, Chen XS, He MF, Qiao JD, Cheng X, Zhu D, Xiong ZQ, Chen WJ. Heterozygous Variants in KCNJ10 Cause Paroxysmal Kinesigenic Dyskinesia Via Haploinsufficiency. Ann Neurol 2024; 96:758-773. [PMID: 38979912 DOI: 10.1002/ana.27018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 06/07/2024] [Accepted: 06/12/2024] [Indexed: 07/10/2024]
Abstract
OBJECTIVE Most paroxysmal kinesigenic dyskinesia (PKD) cases are hereditary, yet approximately 60% of patients remain genetically undiagnosed. We undertook the present study to uncover the genetic basis for undiagnosed PKD patients. METHODS Whole-exome sequencing was performed for 106 PRRT2-negative PKD probands. The functional impact of the genetic variants was investigated in HEK293T cells and Drosophila. RESULTS Heterozygous variants in KCNJ10 were identified in 11 individuals from 8 unrelated families, which accounted for 7.5% (8/106) of the PRRT2-negative probands. Both co-segregation of the identified variants and the significantly higher frequency of rare KCNJ10 variants in PKD cases supported impacts from the detected KCNJ10 heterozygous variants on PKD pathogenesis. Moreover, a KCNJ10 mutation-carrying father from a typical EAST/SeSAME family was identified as a PKD patient. All patients manifested dystonia attacks triggered by sudden movement with a short episodic duration. Patch-clamp recordings in HEK293T cells revealed apparent reductions in K+ currents of the patient-derived variants, indicating a loss-of-function. In Drosophila, milder hyperexcitability phenotypes were observed in heterozygous Irk2 knock-in flies compared to homozygotes, supporting haploinsufficiency as the mechanism for the detected heterozygous variants. Electrophysiological recordings showed that excitatory neurons in Irk2 haploinsufficiency flies exhibited increased excitability, and glia-specific complementation with human Kir4.1 rescued the Irk2 mutant phenotypes. INTERPRETATION Our study established haploinsufficiency resulting from heterozygous variants in KCNJ10 can be understood as a previously unrecognized genetic cause for PKD and provided evidence of glial involvement in the pathophysiology of PKD. ANN NEUROL 2024;96:758-773.
Collapse
Affiliation(s)
- Yun-Lu Li
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| | - Jingjing Lin
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| | - Xuejing Huang
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| | - Rui-Huang Zeng
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| | - Guangyu Zhang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jie-Ni Xu
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| | - Kai-Jun Lin
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| | - Xin-Shuo Chen
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| | - Ming-Feng He
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jing-Da Qiao
- Department of Neurology, Institute of Neuroscience, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Xuewen Cheng
- Lingang Laboratory, Shanghai Center for Brain Science and Brain-Inspired Intelligence Technology, Shanghai, China
| | - Dengna Zhu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhi-Qi Xiong
- Center for Excellence in Brain Science and Intelligence Technology, Institute of Neuroscience and State Key Laboratory of Neuroscience, Chinese Academy of Sciences, Shanghai, China
| | - Wan-Jin Chen
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
2
|
Han J, Wang Y, Wei P, Lu D, Shan Y. Unveiling the hidden connection: the blood-brain barrier's role in epilepsy. Front Neurol 2024; 15:1413023. [PMID: 39206290 PMCID: PMC11349696 DOI: 10.3389/fneur.2024.1413023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 07/18/2024] [Indexed: 09/04/2024] Open
Abstract
Epilepsy is characterized by abnormal synchronous electrical activity of neurons in the brain. The blood-brain barrier, which is mainly composed of endothelial cells, pericytes, astrocytes and other cell types and is formed by connections between a variety of cells, is the key physiological structure connecting the blood and brain tissue and is critical for maintaining the microenvironment in the brain. Physiologically, the blood-brain barrier controls the microenvironment in the brain mainly by regulating the passage of various substances. Disruption of the blood-brain barrier and increased leakage of specific substances, which ultimately leading to weakened cell junctions and abnormal regulation of ion concentrations, have been observed during the development and progression of epilepsy in both clinical studies and animal models. In addition, disruption of the blood-brain barrier increases drug resistance through interference with drug trafficking mechanisms. The changes in the blood-brain barrier in epilepsy mainly affect molecular pathways associated with angiogenesis, inflammation, and oxidative stress. Further research on biomarkers is a promising direction for the development of new therapeutic strategies.
Collapse
Affiliation(s)
| | | | | | | | - Yongzhi Shan
- Department of Neurosurgery, Xuanwu Hospital Capital Medical University, Beijing, China
| |
Collapse
|
3
|
Zhao D, Hu M, Liu S. Glial cells in the mammalian olfactory bulb. Front Cell Neurosci 2024; 18:1426094. [PMID: 39081666 PMCID: PMC11286597 DOI: 10.3389/fncel.2024.1426094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
The mammalian olfactory bulb (OB), an essential part of the olfactory system, plays a critical role in odor detection and neural processing. Historically, research has predominantly focused on the neuronal components of the OB, often overlooking the vital contributions of glial cells. Recent advancements, however, underscore the significant roles that glial cells play within this intricate neural structure. This review discus the diverse functions and dynamics of glial cells in the mammalian OB, mainly focused on astrocytes, microglia, oligodendrocytes, olfactory ensheathing cells, and radial glia cells. Each type of glial contributes uniquely to the OB's functionality, influencing everything from synaptic modulation and neuronal survival to immune defense and axonal guidance. The review features their roles in maintaining neural health, their involvement in neurodegenerative diseases, and their potential in therapeutic applications for neuroregeneration. By providing a comprehensive overview of glial cell types, their mechanisms, and interactions within the OB, this article aims to enhance our understanding of the olfactory system's complexity and the pivotal roles glial cells play in both health and disease.
Collapse
Affiliation(s)
| | | | - Shaolin Liu
- Isakson Center for Neurological Disease Research, Department of Physiology and Pharmacology, Department of Biomedical Sciences, University of Georgia College of Veterinary Medicine, Athens, GA, United States
| |
Collapse
|
4
|
Bataveljic D, Pivonkova H, de Concini V, Hébert B, Ezan P, Briault S, Bemelmans AP, Pichon J, Menuet A, Rouach N. Astroglial Kir4.1 potassium channel deficit drives neuronal hyperexcitability and behavioral defects in Fragile X syndrome mouse model. Nat Commun 2024; 15:3583. [PMID: 38678030 PMCID: PMC11055954 DOI: 10.1038/s41467-024-47681-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 04/03/2024] [Indexed: 04/29/2024] Open
Abstract
Fragile X syndrome (FXS) is an inherited form of intellectual disability caused by the loss of the mRNA-binding fragile X mental retardation protein (FMRP). FXS is characterized by neuronal hyperexcitability and behavioral defects, however the mechanisms underlying these critical dysfunctions remain unclear. Here, using male Fmr1 knockout mouse model of FXS, we identify abnormal extracellular potassium homeostasis, along with impaired potassium channel Kir4.1 expression and function in astrocytes. Further, we reveal that Kir4.1 mRNA is a binding target of FMRP. Finally, we show that the deficit in astroglial Kir4.1 underlies neuronal hyperexcitability and several behavioral defects in Fmr1 knockout mice. Viral delivery of Kir4.1 channels specifically to hippocampal astrocytes from Fmr1 knockout mice indeed rescues normal astrocyte potassium uptake, neuronal excitability, and cognitive and social performance. Our findings uncover an important role for astrocyte dysfunction in the pathophysiology of FXS, and identify Kir4.1 channel as a potential therapeutic target for FXS.
Collapse
Affiliation(s)
- Danijela Bataveljic
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
- Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Helena Pivonkova
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
- Department of Physiology, 2nd Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Vidian de Concini
- Experimental and Molecular Immunology and Neurogenetics, CNRS UMR7355 and Orléans University, Orléans, France
| | - Betty Hébert
- Experimental and Molecular Immunology and Neurogenetics, CNRS UMR7355 and Orléans University, Orléans, France
| | - Pascal Ezan
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Sylvain Briault
- Experimental and Molecular Immunology and Neurogenetics, CNRS UMR7355 and Orléans University, Orléans, France
- Department of Genetics, Regional Hospital, Orléans, France
| | - Alexis-Pierre Bemelmans
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Département de la Recherche Fondamentale, Institut de biologie François Jacob, MIRCen, and CNRS UMR 9199, Université Paris-Sud, Neurodegenerative Diseases Laboratory, Fontenay-aux-Roses, 92260, France
| | - Jacques Pichon
- Experimental and Molecular Immunology and Neurogenetics, CNRS UMR7355 and Orléans University, Orléans, France
| | - Arnaud Menuet
- Experimental and Molecular Immunology and Neurogenetics, CNRS UMR7355 and Orléans University, Orléans, France
| | - Nathalie Rouach
- Neuroglial Interactions in Cerebral Physiology and Pathologies, Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France.
| |
Collapse
|
5
|
Passchier EMJ, Bisseling Q, Helman G, van Spaendonk RML, Simons C, Olsthoorn RCL, van der Veen H, Abbink TEM, van der Knaap MS, Min R. Megalencephalic leukoencephalopathy with subcortical cysts: a variant update and review of the literature. Front Genet 2024; 15:1352947. [PMID: 38487253 PMCID: PMC10938252 DOI: 10.3389/fgene.2024.1352947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 01/29/2024] [Indexed: 03/17/2024] Open
Abstract
The leukodystrophy megalencephalic leukoencephalopathy with subcortical cysts (MLC) is characterized by infantile-onset macrocephaly and chronic edema of the brain white matter. With delayed onset, patients typically experience motor problems, epilepsy and slow cognitive decline. No treatment is available. Classic MLC is caused by bi-allelic recessive pathogenic variants in MLC1 or GLIALCAM (also called HEPACAM). Heterozygous dominant pathogenic variants in GLIALCAM lead to remitting MLC, where patients show a similar phenotype in early life, followed by normalization of white matter edema and no clinical regression. Rare patients with heterozygous dominant variants in GPRC5B and classic MLC were recently described. In addition, two siblings with bi-allelic recessive variants in AQP4 and remitting MLC have been identified. The last systematic overview of variants linked to MLC dates back to 2006. We provide an updated overview of published and novel variants. We report on genetic variants from 508 patients with MLC as confirmed by MRI diagnosis (258 from our database and 250 extracted from 64 published reports). We describe 151 unique MLC1 variants, 29 GLIALCAM variants, 2 GPRC5B variants and 1 AQP4 variant observed in these MLC patients. We include experiments confirming pathogenicity for some variants, discuss particularly notable variants, and provide an overview of recent scientific and clinical insight in the pathophysiology of MLC.
Collapse
Affiliation(s)
- Emma M. J. Passchier
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Quinty Bisseling
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Guy Helman
- Translational Bioinformatics, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Parkville, VIC, Australia
| | | | - Cas Simons
- Translational Bioinformatics, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Parkville, VIC, Australia
- Centre for Population Genomics, Garvan Institute of Medical Research, Sydney, NSW, Australia
| | | | - Hieke van der Veen
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
- Department of Complex Trait Genetics, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Truus E. M. Abbink
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Marjo S. van der Knaap
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| | - Rogier Min
- Department of Child Neurology, Amsterdam Leukodystrophy Center, Emma Children’s Hospital, Amsterdam University Medical Center, Amsterdam Neuroscience, Amsterdam, Netherlands
- Department of Integrative Neurophysiology, Center for Neurogenomics and Cognitive Research, Vrije Universiteit Amsterdam, Amsterdam Neuroscience, Amsterdam, Netherlands
| |
Collapse
|
6
|
Maruyama H, Gomi M, Lwin TT, Yoneyama A, Sasaki T. [ 18F]-FDG uptake in brain slices prepared from an aged mouse model of Alzheimer's disease using a dynamic autoradiography technique. Ann Nucl Med 2024; 38:120-130. [PMID: 37921921 DOI: 10.1007/s12149-023-01879-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 09/29/2023] [Indexed: 11/05/2023]
Abstract
OBJECTIVE 2-[18F]fluoro-2-deoxy-D-glucose positron emission tomography ([18F]-FDG-PET) is a imaging modality that has been used to measure of glucose metabolism in the brain in Alzheimer's disease (AD). Clinically, decreased glucose uptake has been reported in the brain of AD, although the precise underlying mechanisms have not yet been elucidated. To elucidate the mechanisms of decreased [18F]-FDG uptake in the AD by PET, [18F]-FDG uptake in the brain of aged model mouse of AD was investigated using a dynamic autoradiography technique "bioradiography". A X-ray phase-contrast imaging (X-PCI) and a histopathological evaluation were also investigated to elucidate the mechanisms underlying the relationships between decreased [18F]-FDG uptake and the pathological changes in the brain of AD mouse. METHODS In this study, AD model mouse (5XFAD, APP+/PS1+) were used. [18F]-FDG-bioradiography was conducted in fresh slices of brain tissue under the condition of resting (slices immersed in 5 mM K+ solution) and metabolically active (in 50 mM K+ solution). Amyloid β42 (Aβ42) deposition in the brain of AD mouse was confirmed by X-PCI. In addition, the positive cells of phosphated tau protein (P-tau) and deposition of Aβ42 were also examined by immunohistochemical staining. RESULTS No significant differences were observed between the two groups in the resting condition. In the activate condition of the brain, [18F]-FDG uptake was significantly decreased in AD mice compared to WT mice. In X-PCI showed Aβ deposition in the AD mouse, but not in the WT. The AD mouse also showed increased P-tau, accumulation of Aβ42, increase in neuronal apoptosis, and decrease in the number of neurons than that of the WT mouse. CONCLUSION Neuronal damage, and induction of neuronal apoptosis, decreased [18F]-FDG uptake, increased Aβ accumulation and P-tau induced neurofibrillary degeneration are observed in AD mouse. In clinical diagnosis, reduction of [18F]-FDG uptake by PET is one of the means of diagnosing the onset of AD. Our results suggest that decreased uptake of [18F]-FDG in the brains of AD may be associated with neuronal dysfunction and cell death in the brain.
Collapse
Affiliation(s)
- Hiroko Maruyama
- Cytopathology, Graduate School of Medical Sciences, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara, 252-0373, Japan.
| | - Misaki Gomi
- Cytopathology, Graduate School of Medical Sciences, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara, 252-0373, Japan
| | - Thet-Thet Lwin
- Molecular Imaging, Graduate School of Medical Sciences, School of Allied Health Sciences, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara, 252-0373, Japan
| | - Akio Yoneyama
- SAGA Light Source, Synchrotron Light Research Center, 8-7 Yayoigaoka, Tosu, Saga, 841-0005, Japan
| | - Toru Sasaki
- Radiation Safety Management, Graduate School of Medical Sciences, Kitasato University, 1-15-1 Kitasato, Minami-ku, Sagamihara, 252-0373, Japan
| |
Collapse
|
7
|
Altas B, Rhee HJ, Ju A, Solís HC, Karaca S, Winchenbach J, Kaplan-Arabaci O, Schwark M, Ambrozkiewicz MC, Lee C, Spieth L, Wieser GL, Chaugule VK, Majoul I, Hassan MA, Goel R, Wojcik SM, Koganezawa N, Hanamura K, Rotin D, Pichler A, Mitkovski M, de Hoz L, Poulopoulos A, Urlaub H, Jahn O, Saher G, Brose N, Rhee J, Kawabe H. Nedd4-2-dependent regulation of astrocytic Kir4.1 and Connexin43 controls neuronal network activity. J Cell Biol 2024; 223:e201902050. [PMID: 38032389 PMCID: PMC10689203 DOI: 10.1083/jcb.201902050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 10/21/2021] [Accepted: 11/02/2023] [Indexed: 12/01/2023] Open
Abstract
Nedd4-2 is an E3 ubiquitin ligase in which missense mutation is related to familial epilepsy, indicating its critical role in regulating neuronal network activity. However, Nedd4-2 substrates involved in neuronal network function have yet to be identified. Using mouse lines lacking Nedd4-1 and Nedd4-2, we identified astrocytic channel proteins inwardly rectifying K+ channel 4.1 (Kir4.1) and Connexin43 as Nedd4-2 substrates. We found that the expression of Kir4.1 and Connexin43 is increased upon conditional deletion of Nedd4-2 in astrocytes, leading to an elevation of astrocytic membrane ion permeability and gap junction activity, with a consequent reduction of γ-oscillatory neuronal network activity. Interestingly, our biochemical data demonstrate that missense mutations found in familial epileptic patients produce gain-of-function of the Nedd4-2 gene product. Our data reveal a process of coordinated astrocytic ion channel proteostasis that controls astrocyte function and astrocyte-dependent neuronal network activity and elucidate a potential mechanism by which aberrant Nedd4-2 function leads to epilepsy.
Collapse
Affiliation(s)
- Bekir Altas
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- International Max Planck Research School and the Göttingen Graduate School for Neurosciences, Biophysics and Molecular Biosciences, Göttingen, Germany
- The Göttingen Graduate School for Neurosciences, Biophysics, and Molecular Biosciences, PhD Program Systems Neuroscience, University of Göttingen, Göttingen, Germany
- Department of Pharmacology and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hong-Jun Rhee
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Anes Ju
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- The Göttingen Graduate School for Neurosciences, Biophysics, and Molecular Biosciences, PhD Program Systems Neuroscience, University of Göttingen, Göttingen, Germany
| | - Hugo Cruces Solís
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- International Max Planck Research School and the Göttingen Graduate School for Neurosciences, Biophysics and Molecular Biosciences, Göttingen, Germany
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Samir Karaca
- International Max Planck Research School and the Göttingen Graduate School for Neurosciences, Biophysics and Molecular Biosciences, Göttingen, Germany
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Jan Winchenbach
- The Göttingen Graduate School for Neurosciences, Biophysics, and Molecular Biosciences, PhD Program Systems Neuroscience, University of Göttingen, Göttingen, Germany
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Oykum Kaplan-Arabaci
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- The Göttingen Graduate School for Neurosciences, Biophysics, and Molecular Biosciences, PhD Program Molecular Physiology of the Brain, University of Göttingen, Göttingen, Germany
| | - Manuela Schwark
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Mateusz C. Ambrozkiewicz
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- International Max Planck Research School and the Göttingen Graduate School for Neurosciences, Biophysics and Molecular Biosciences, Göttingen, Germany
- Institute of Cell Biology and Neurobiology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - ChungKu Lee
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Lena Spieth
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Georg L. Wieser
- City Campus Light Microscopy Facility, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Viduth K. Chaugule
- Department of Epigenetics, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Irina Majoul
- Institute of Biology, Center for Structural and Cell Biology in Medicine, University of Lübeck, Lübeck, Germany
| | - Mohamed A. Hassan
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Protein Research Department, Genetic Engineering and Biotechnology Research Institute (GEBRI), City of Scientific Research and Technological Applications (SRTA-City), New Borg El-Arab City, Egypt
| | - Rashi Goel
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Sonja M. Wojcik
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Noriko Koganezawa
- Department of Pharmacology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Kenji Hanamura
- Department of Pharmacology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Daniela Rotin
- The Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - Andrea Pichler
- Department of Epigenetics, Max Planck Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Miso Mitkovski
- City Campus Light Microscopy Facility, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Livia de Hoz
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Alexandros Poulopoulos
- Department of Pharmacology and Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Henning Urlaub
- Bioanalytical Mass Spectrometry Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Bioanalytics, Institute for Clinical Chemistry, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence “Multiscale Bioimaging: From Molecular Machines to Networks of Excitable Cells” (MBExC), University of Göttingen, Göttingen, Germany
| | - Olaf Jahn
- Department of Molecular Neurobiology, Neuroproteomics Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Psychiatry and Psychotherapy, Translational Neuroproteomics Group, University Medical Center Göttingen, Göttingen, Germany
| | - Gesine Saher
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Nils Brose
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - JeongSeop Rhee
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Hiroshi Kawabe
- Department of Molecular Neurobiology, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Pharmacology, Gunma University Graduate School of Medicine, Maebashi, Japan
- Division of Pathogenetic Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
- Department of Gerontology, Laboratory of Molecular Life Science, Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan
| |
Collapse
|
8
|
Zhang YM, Qi YB, Gao YN, Chen WG, Zhou T, Zang Y, Li J. Astrocyte metabolism and signaling pathways in the CNS. Front Neurosci 2023; 17:1217451. [PMID: 37732313 PMCID: PMC10507181 DOI: 10.3389/fnins.2023.1217451] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/18/2023] [Indexed: 09/22/2023] Open
Abstract
Astrocytes comprise half of the cells in the central nervous system and play a critical role in maintaining metabolic homeostasis. Metabolic dysfunction in astrocytes has been indicated as the primary cause of neurological diseases, such as depression, Alzheimer's disease, and epilepsy. Although the metabolic functionalities of astrocytes are well known, their relationship to neurological disorders is poorly understood. The ways in which astrocytes regulate the metabolism of glucose, amino acids, and lipids have all been implicated in neurological diseases. Metabolism in astrocytes has also exhibited a significant influence on neuron functionality and the brain's neuro-network. In this review, we focused on metabolic processes present in astrocytes, most notably the glucose metabolic pathway, the fatty acid metabolic pathway, and the amino-acid metabolic pathway. For glucose metabolism, we focused on the glycolysis pathway, pentose-phosphate pathway, and oxidative phosphorylation pathway. In fatty acid metabolism, we followed fatty acid oxidation, ketone body metabolism, and sphingolipid metabolism. For amino acid metabolism, we summarized neurotransmitter metabolism and the serine and kynurenine metabolic pathways. This review will provide an overview of functional changes in astrocyte metabolism and provide an overall perspective of current treatment and therapy for neurological disorders.
Collapse
Affiliation(s)
- Yong-mei Zhang
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ying-bei Qi
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Ya-nan Gao
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Wen-gang Chen
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ting Zhou
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yi Zang
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jia Li
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- National Center for Drug Screening, State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
- Institute of Pharmaceutical Sciences, China Pharmaceutical University, Nanjing, Jiangsu, China
| |
Collapse
|
9
|
Gebicke-Haerter PJ. The computational power of the human brain. Front Cell Neurosci 2023; 17:1220030. [PMID: 37608987 PMCID: PMC10441807 DOI: 10.3389/fncel.2023.1220030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/05/2023] [Indexed: 08/24/2023] Open
Abstract
At the end of the 20th century, analog systems in computer science have been widely replaced by digital systems due to their higher computing power. Nevertheless, the question keeps being intriguing until now: is the brain analog or digital? Initially, the latter has been favored, considering it as a Turing machine that works like a digital computer. However, more recently, digital and analog processes have been combined to implant human behavior in robots, endowing them with artificial intelligence (AI). Therefore, we think it is timely to compare mathematical models with the biology of computation in the brain. To this end, digital and analog processes clearly identified in cellular and molecular interactions in the Central Nervous System are highlighted. But above that, we try to pinpoint reasons distinguishing in silico computation from salient features of biological computation. First, genuinely analog information processing has been observed in electrical synapses and through gap junctions, the latter both in neurons and astrocytes. Apparently opposed to that, neuronal action potentials (APs) or spikes represent clearly digital events, like the yes/no or 1/0 of a Turing machine. However, spikes are rarely uniform, but can vary in amplitude and widths, which has significant, differential effects on transmitter release at the presynaptic terminal, where notwithstanding the quantal (vesicular) release itself is digital. Conversely, at the dendritic site of the postsynaptic neuron, there are numerous analog events of computation. Moreover, synaptic transmission of information is not only neuronal, but heavily influenced by astrocytes tightly ensheathing the majority of synapses in brain (tripartite synapse). At least at this point, LTP and LTD modifying synaptic plasticity and believed to induce short and long-term memory processes including consolidation (equivalent to RAM and ROM in electronic devices) have to be discussed. The present knowledge of how the brain stores and retrieves memories includes a variety of options (e.g., neuronal network oscillations, engram cells, astrocytic syncytium). Also epigenetic features play crucial roles in memory formation and its consolidation, which necessarily guides to molecular events like gene transcription and translation. In conclusion, brain computation is not only digital or analog, or a combination of both, but encompasses features in parallel, and of higher orders of complexity.
Collapse
Affiliation(s)
- Peter J. Gebicke-Haerter
- Institute of Psychopharmacology, Central Institute of Mental Health, Faculty of Medicine, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
10
|
Smith LA, Chen C, Lax NZ, Taylor RW, Erskine D, McFarland R. Astrocytic pathology in Alpers' syndrome. Acta Neuropathol Commun 2023; 11:86. [PMID: 37259148 DOI: 10.1186/s40478-023-01579-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/11/2023] [Indexed: 06/02/2023] Open
Abstract
Refractory epilepsy is the main neurological manifestation of Alpers' syndrome, a severe childhood-onset mitochondrial disease caused by bi-allelic pathogenic variants in the mitochondrial DNA (mtDNA) polymerase gamma gene (POLG). The pathophysiological mechanisms underpinning neuronal hyperexcitabilty leading to seizures in Alpers' syndrome remain unknown. However, pathological changes to reactive astrocytes are hypothesised to exacerbate neural dysfunction and seizure-associated cortical activity in POLG-related disease. Therefore, we sought to phenotypically characterise astrocytic pathology in Alpers' syndrome. We performed a detailed quantitative investigation of reactive astrocytes in post-mortem neocortical tissues from thirteen patients with Alpers' syndrome, eight neurologically normal controls and five sudden unexpected death in epilepsy (SUDEP) patients, to control for generalised epilepsy-associated astrocytic pathology. Immunohistochemistry to identify glial fibrillary acidic protein (GFAP)-reactive astrocytes revealed striking reactive astrogliosis localised to the primary visual cortex of Alpers' syndrome tissues, characterised by abnormal-appearing hypertrophic astrocytes. Phenotypic characterisation of individual GFAP-reactive astrocytes demonstrated decreased abundance of mitochondrial oxidative phosphorylation (OXPHOS) proteins and altered expression of key astrocytic proteins including Kir4.1 (subunit of the inwardly rectifying K+ ion channel), AQP4 (astrocytic water channel) and glutamine synthetase (enzyme that metabolises glutamate). These phenotypic astrocytic changes were typically different from the pathology observed in SUDEP tissues, suggesting alternative mechanisms of astrocytic dysfunction between these epilepsies. Crucially, our findings provide further evidence of occipital lobe involvement in Alpers' syndrome and support the involvement of reactive astrocytes in the pathogenesis of POLG-related disease.
Collapse
Affiliation(s)
- Laura A Smith
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| | - Chun Chen
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Nichola Z Lax
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Robert W Taylor
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders of Adults and Children, Newcastle University, Newcastle Upon Tyne, Newcastle, NE2 4HH, UK
| | - Daniel Erskine
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Robert McFarland
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
- NHS Highly Specialised Service for Rare Mitochondrial Disorders of Adults and Children, Newcastle University, Newcastle Upon Tyne, Newcastle, NE2 4HH, UK.
| |
Collapse
|
11
|
Božić M, Pirnat S, Fink K, Potokar M, Kreft M, Zorec R, Stenovec M. Ketamine Reduces the Surface Density of the Astroglial Kir4.1 Channel and Inhibits Voltage-Activated Currents in a Manner Similar to the Action of Ba 2+ on K + Currents. Cells 2023; 12:1360. [PMID: 37408194 DOI: 10.3390/cells12101360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/04/2023] [Accepted: 05/05/2023] [Indexed: 07/07/2023] Open
Abstract
A single sub-anesthetic dose of ketamine evokes rapid and long-lasting beneficial effects in patients with a major depressive disorder. However, the mechanisms underlying this effect are unknown. It has been proposed that astrocyte dysregulation of extracellular K+ concentration ([K+]o) alters neuronal excitability, thus contributing to depression. We examined how ketamine affects inwardly rectifying K+ channel Kir4.1, the principal regulator of K+ buffering and neuronal excitability in the brain. Cultured rat cortical astrocytes were transfected with plasmid-encoding fluorescently tagged Kir4.1 (Kir4.1-EGFP) to monitor the mobility of Kir4.1-EGFP vesicles at rest and after ketamine treatment (2.5 or 25 µM). Short-term (30 min) ketamine treatment reduced the mobility of Kir4.1-EGFP vesicles compared with the vehicle-treated controls (p < 0.05). Astrocyte treatment (24 h) with dbcAMP (dibutyryl cyclic adenosine 5'-monophosphate, 1 mM) or [K+]o (15 mM), which increases intracellular cAMP, mimicked the ketamine-evoked reduction of mobility. Live cell immunolabelling and patch-clamp measurements in cultured mouse astrocytes revealed that short-term ketamine treatment reduced the surface density of Kir4.1 and inhibited voltage-activated currents similar to Ba2+ (300 µM), a Kir4.1 blocker. Thus, ketamine attenuates Kir4.1 vesicle mobility, likely via a cAMP-dependent mechanism, reduces Kir4.1 surface density, and inhibits voltage-activated currents similar to Ba2+, known to block Kir4.1 channels.
Collapse
Affiliation(s)
- Mićo Božić
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
- Department of Medical Oncology, Institute of Oncology Ljubljana, Zaloška 2, 1000 Ljubljana, Slovenia
| | - Samo Pirnat
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
- Celica Biomedical, Tehnološki Park 24, 1000 Ljubljana, Slovenia
| | - Katja Fink
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
| | - Maja Potokar
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
- Celica Biomedical, Tehnološki Park 24, 1000 Ljubljana, Slovenia
| | - Marko Kreft
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
- Celica Biomedical, Tehnološki Park 24, 1000 Ljubljana, Slovenia
- Department of Biology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
- Celica Biomedical, Tehnološki Park 24, 1000 Ljubljana, Slovenia
| | - Matjaž Stenovec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Institute of Pathophysiology, Faculty of Medicine, University of Ljubljana, Zaloška 4, 1000 Ljubljana, Slovenia
- Celica Biomedical, Tehnološki Park 24, 1000 Ljubljana, Slovenia
| |
Collapse
|
12
|
Li X, Helleringer R, Martucci LL, Dallérac G, Cancela JM, Galante M. Low Temperature Delays the Effects of Ischemia in Bergmann Glia and in Cerebellar Tissue Swelling. Biomedicines 2023; 11:biomedicines11051363. [PMID: 37239034 DOI: 10.3390/biomedicines11051363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 04/23/2023] [Accepted: 04/29/2023] [Indexed: 05/28/2023] Open
Abstract
Cerebral ischemia results in oxygen and glucose deprivation that most commonly occurs after a reduction or interruption in the blood supply to the brain. The consequences of cerebral ischemia are complex and involve the loss of metabolic ATP, excessive K+ and glutamate accumulation in the extracellular space, electrolyte imbalance, and brain edema formation. So far, several treatments have been proposed to alleviate ischemic damage, yet few are effective. Here, we focused on the neuroprotective role of lowering the temperature in ischemia mimicked by an episode of oxygen and glucose deprivation (OGD) in mouse cerebellar slices. Our results suggest that lowering the temperature of the extracellular 'milieu' delays both the increases in [K+]e and tissue swelling, two dreaded consequences of cerebellar ischemia. Moreover, radial glial cells (Bergmann glia) display morphological changes and membrane depolarizations that are markedly impeded by lowering the temperature. Overall, in this model of cerebellar ischemia, hypothermia reduces the deleterious homeostatic changes regulated by Bergmann glia.
Collapse
Affiliation(s)
- Xia Li
- Institut des Neurosciences Paris-Saclay, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Romain Helleringer
- Institut des Neurosciences Paris-Saclay, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Lora L Martucci
- Institut des Neurosciences Paris-Saclay, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Glenn Dallérac
- Institut des Neurosciences Paris-Saclay, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - José-Manuel Cancela
- Institut des Neurosciences Paris-Saclay, CNRS, Université Paris-Saclay, 91400 Saclay, France
| | - Micaela Galante
- Institut des Neurosciences Paris-Saclay, CNRS, Université Paris-Saclay, 91400 Saclay, France
| |
Collapse
|
13
|
Barbay T, Pecchi E, Ducrocq M, Rouach N, Brocard F, Bos R. Astrocytic Kir4.1 channels regulate locomotion by orchestrating neuronal rhythmicity in the spinal network. Glia 2023; 71:1259-1277. [PMID: 36645018 DOI: 10.1002/glia.24337] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 12/22/2022] [Accepted: 01/02/2023] [Indexed: 01/17/2023]
Abstract
Neuronal rhythmogenesis in the spinal cord is correlated with variations in extracellular K+ levels ([K+ ]e ). Astrocytes play important role in [K+ ]e homeostasis and compute neuronal information. Yet it is unclear how neuronal oscillations are regulated by astrocytic K+ homeostasis. Here we identify the astrocytic inward-rectifying K+ channel Kir4.1 (a.k.a. Kcnj10) as a key molecular player for neuronal rhythmicity in the spinal central pattern generator (CPG). By combining two-photon calcium imaging with electrophysiology, immunohistochemistry and genetic tools, we report that astrocytes display Ca2+ transients before and during oscillations of neighboring neurons. Inhibition of astrocytic Ca2+ transients with BAPTA decreases the barium-sensitive Kir4.1 current responsible of K+ clearance. Finally, we show in mice that Kir4.1 knockdown in astrocytes progressively prevents neuronal oscillations and alters the locomotor pattern resulting in lower motor performances in challenging tasks. These data identify astroglial Kir4.1 channels as key regulators of neuronal rhythmogenesis in the CPG driving locomotion.
Collapse
Affiliation(s)
- Tony Barbay
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), UMR 7289, Marseille, France
| | - Emilie Pecchi
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), UMR 7289, Marseille, France
| | - Myriam Ducrocq
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), UMR 7289, Marseille, France
| | - Nathalie Rouach
- Center for Interdisciplinary Research in Biology, Collège de France, CNRS, INSERM, Labex Memolife, Université PSL, Paris, France
| | - Frédéric Brocard
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), UMR 7289, Marseille, France
| | - Rémi Bos
- Aix Marseille Univ, CNRS, Institut de Neurosciences de la Timone (INT), UMR 7289, Marseille, France
| |
Collapse
|
14
|
Fomitcheva IV, Sword J, Shi Y, Kirov SA. Plasticity of perisynaptic astroglia during ischemia-induced spreading depolarization. Cereb Cortex 2023; 33:5469-5483. [PMID: 36368909 PMCID: PMC10152098 DOI: 10.1093/cercor/bhac434] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/06/2022] [Accepted: 10/08/2022] [Indexed: 11/13/2022] Open
Abstract
High astroglial capacity for glutamate and potassium clearance aids in recovering spreading depolarization (SD)-evoked disturbance of ion homeostasis during stroke. Since perisynaptic astroglia cannot be imaged with diffraction-limited light microscopy, nothing is known about the impact of SD on the ultrastructure of a tripartite synapse. We used serial section electron microscopy to assess astroglial synaptic coverage in the sensorimotor cortex of urethane-anesthetized male and female mice during and after SD evoked by transient bilateral common carotid artery occlusion. At the subcellular level, astroglial mitochondria were remarkably resilient to SD compared to dendritic mitochondria that were fragmented by SD. Overall, 482 synapses in `Sham' during `SD' and `Recovery' groups were randomly selected and analyzed in 3D. Perisynaptic astroglia was present at the axon-spine interface (ASI) during SD and after recovery. Astrocytic processes were more likely found at large synapses on mushroom spines after recovery, while the length of the ASI perimeter surrounded by astroglia has also significantly increased at large synapses. These findings suggest that as larger synapses have a bigger capacity for neurotransmitter release during SD, they attract astroglial processes to their perimeter during recovery, limiting extrasynaptic glutamate escape and further enhancing the astrocytic ability to protect synapses in stroke.
Collapse
Affiliation(s)
- Ioulia V Fomitcheva
- Department of Neurosurgery, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA 30912, United States
| | - Jeremy Sword
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA 30912, United States
| | - Yang Shi
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA 30912, United States
- Division of Biostatistics and Data Science, Department of Population Health Sciences, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA 30912, United States
| | - Sergei A Kirov
- Department of Neurosurgery, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA 30912, United States
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, 1120 15th Street, Augusta, GA 30912, United States
| |
Collapse
|
15
|
Abrams CK. Mechanisms of Diseases Associated with Mutation in GJC2/Connexin 47. Biomolecules 2023; 13:biom13040712. [PMID: 37189458 DOI: 10.3390/biom13040712] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 05/17/2023] Open
Abstract
Connexins are members of a family of integral membrane proteins that provide a pathway for both electrical and metabolic coupling between cells. Astroglia express connexin 30 (Cx30)-GJB6 and Cx43-GJA1, while oligodendroglia express Cx29/Cx31.3-GJC3, Cx32-GJB1, and Cx47-GJC2. Connexins organize into hexameric hemichannels (homomeric if all subunits are identical or heteromeric if one or more differs). Hemichannels from one cell then form cell-cell channels with a hemichannel from an apposed cell. (These are termed homotypic if the hemichannels are identical and heterotypic if the hemichannels differ). Oligodendrocytes couple to each other through Cx32/Cx32 or Cx47/Cx47 homotypic channels and they couple to astrocytes via Cx32/Cx30 or Cx47/Cx43 heterotypic channels. Astrocytes couple via Cx30/Cx30 and Cx43/Cx43 homotypic channels. Though Cx32 and Cx47 may be expressed in the same cells, all available data suggest that Cx32 and Cx47 cannot interact heteromerically. Animal models wherein one or in some cases two different CNS glial connexins have been deleted have helped to clarify the role of these molecules in CNS function. Mutations in a number of different CNS glial connexin genes cause human disease. Mutations in GJC2 lead to three distinct phenotypes, Pelizaeus Merzbacher like disease, hereditary spastic paraparesis (SPG44) and subclinical leukodystrophy.
Collapse
Affiliation(s)
- Charles K Abrams
- Department of Neurology and Rehabilitation, University of Illinois at Chicago College of Medicine, Chicago, IL 60612, USA
| |
Collapse
|
16
|
Procacci NM, Hastings RL, Aziz AA, Christiansen NM, Zhao J, DeAngeli C, LeBlanc N, Notterpek L, Valdez G, Gould TW. Kir4.1 is specifically expressed and active in non-myelinating Schwann cells. Glia 2023; 71:926-944. [PMID: 36479906 PMCID: PMC9931657 DOI: 10.1002/glia.24315] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022]
Abstract
Non-myelinating Schwann cells (NMSC) play important roles in peripheral nervous system formation and function. However, the molecular identity of these cells remains poorly defined. We provide evidence that Kir4.1, an inward-rectifying K+ channel encoded by the KCNJ10 gene, is specifically expressed and active in NMSC. Immunostaining revealed that Kir4.1 is present in terminal/perisynaptic SCs (TPSC), synaptic glia at neuromuscular junctions (NMJ), but not in myelinating SCs (MSC) of adult mice. To further examine the expression pattern of Kir4.1, we generated BAC transgenic Kir4.1-CreERT2 mice and crossed them to the tdTomato reporter line. Activation of CreERT2 with tamoxifen after the completion of myelination onset led to robust expression of tdTomato in NMSC, including Remak Schwann cells (RSC) along peripheral nerves and TPSC, but not in MSC. In contrast, activating CreERT2 before and during the onset of myelination led to tdTomato expression in NMSC and MSC. These observations suggest that immature SC express Kir4.1, and its expression is then downregulated selectively in myelin-forming SC. In support, we found that while activating CreERT2 induces tdTomato expression in immature SC, it fails to induce tdTomato in MSC associated with sensory axons in culture. NMSC derived from neonatal sciatic nerve were shown to express Kir4.1 and exhibit barium-sensitive inwardly rectifying macroscopic K+ currents. Thus, this study identified Kir4.1 as a potential modulator of immature SC and NMSC function. Additionally, it established a novel transgenic mouse line to introduce or delete genes in NMSC.
Collapse
Affiliation(s)
- Nicole M Procacci
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Robert Louis Hastings
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Aamir A Aziz
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Nina M Christiansen
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Jie Zhao
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Claire DeAngeli
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Normand LeBlanc
- Department of Pharmacology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Lucia Notterpek
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| | - Gregorio Valdez
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, Rhode Island, USA
| | - Thomas W Gould
- Department of Physiology and Cell Biology, University of Nevada School of Medicine, Reno, Nevada, USA
| |
Collapse
|
17
|
Smith BC, Tinkey RA, Shaw BC, Williams JL. Targetability of the neurovascular unit in inflammatory diseases of the central nervous system. Immunol Rev 2022; 311:39-49. [PMID: 35909222 PMCID: PMC9489669 DOI: 10.1111/imr.13121] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The blood-brain barrier (BBB) is a selectively permeable barrier separating the periphery from the central nervous system (CNS). The BBB restricts the flow of most material into and out of the CNS, including many drugs that could be used as potent therapies. BBB permeability is modulated by several cells that are collectively called the neurovascular unit (NVU). The NVU consists of specialized CNS endothelial cells (ECs), pericytes, astrocytes, microglia, and neurons. CNS ECs maintain a complex "seal" via tight junctions, forming the BBB; breakdown of these tight junctions leads to BBB disruption. Pericytes control the vascular flow within capillaries and help maintain the basal lamina. Astrocytes control much of the flow of material that has moved beyond the CNS EC layer and can form a secondary barrier under inflammatory conditions. Microglia survey the border of the NVU for noxious material. Neuronal activity also plays a role in the maintenance of the BBB. Since astrocytes, pericytes, microglia, and neurons are all able to modulate the permeability of the BBB, understating the complex contributions of each member of the NVU will potentially uncover novel and effective methods for delivery of neurotherapies to the CNS.
Collapse
Affiliation(s)
- Brandon C. Smith
- Department of NeurosciencesLerner Research Institute, Cleveland ClinicClevelandOhioUSA,Department of Biological, Geological, and Environmental SciencesCleveland State UniversityClevelandOhioUSA
| | - Rachel A. Tinkey
- Department of NeurosciencesLerner Research Institute, Cleveland ClinicClevelandOhioUSA,School of Biomedical SciencesKent State UniversityKentOhioUSA
| | - Benjamin C. Shaw
- Department of NeurosciencesLerner Research Institute, Cleveland ClinicClevelandOhioUSA
| | - Jessica L. Williams
- Department of NeurosciencesLerner Research Institute, Cleveland ClinicClevelandOhioUSA,Brain Health Research Institute, Kent State UniversityKentOhioUSA
| |
Collapse
|
18
|
Ochoa-de la Paz LD, Gulias-Cañizo R. Glia as a key factor in cell volume regulation processes of the central nervous system. Front Cell Neurosci 2022; 16:967496. [PMID: 36090789 PMCID: PMC9453262 DOI: 10.3389/fncel.2022.967496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 08/03/2022] [Indexed: 11/23/2022] Open
Abstract
Brain edema is a pathological condition with potentially fatal consequences, related to cerebral injuries such as ischemia, chronic renal failure, uremia, and diabetes, among others. Under these pathological states, the cell volume control processes are fully compromised, because brain cells are unable to regulate the movement of water, mainly regulated by osmotic gradients. The processes involved in cell volume regulation are homeostatic mechanisms that depend on the mobilization of osmolytes (ions, organic molecules, and polyols) in the necessary direction to counteract changes in osmolyte concentration in response to water movement. The expression and coordinated function of proteins related to the cell volume regulation process, such as water channels, ion channels, and other cotransport systems in the glial cells, and considering the glial cell proportion compared to neuronal cells, leads to consider the astroglial network the main regulatory unit for water homeostasis in the central nervous system (CNS). In the last decade, several studies highlighted the pivotal role of glia in the cell volume regulation process and water homeostasis in the brain, including the retina; any malfunction of this astroglial network generates a lack of the ability to regulate the osmotic changes and water movements and consequently exacerbates the pathological condition.
Collapse
Affiliation(s)
- Lenin David Ochoa-de la Paz
- Departamento de Bioquímica, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico
- Asociación para Evitar la Ceguera en México (APEC), Unidad de Investigación APEC-UNAM, Mexico
- *Correspondence: Lenin David Ochoa-de la Paz
| | | |
Collapse
|
19
|
Armbruster M, Naskar S, Garcia JP, Sommer M, Kim E, Adam Y, Haydon PG, Boyden ES, Cohen AE, Dulla CG. Neuronal activity drives pathway-specific depolarization of peripheral astrocyte processes. Nat Neurosci 2022; 25:607-616. [PMID: 35484406 PMCID: PMC9988390 DOI: 10.1038/s41593-022-01049-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 03/14/2022] [Indexed: 12/16/2022]
Abstract
Astrocytes are glial cells that interact with neuronal synapses via their distal processes, where they remove glutamate and potassium (K+) from the extracellular space following neuronal activity. Astrocyte clearance of both glutamate and K+ is voltage dependent, but astrocyte membrane potential (Vm) is thought to be largely invariant. As a result, these voltage dependencies have not been considered relevant to astrocyte function. Using genetically encoded voltage indicators to enable the measurement of Vm at peripheral astrocyte processes (PAPs) in mice, we report large, rapid, focal and pathway-specific depolarizations in PAPs during neuronal activity. These activity-dependent astrocyte depolarizations are driven by action potential-mediated presynaptic K+ efflux and electrogenic glutamate transporters. We find that PAP depolarization inhibits astrocyte glutamate clearance during neuronal activity, enhancing neuronal activation by glutamate. This represents a novel class of subcellular astrocyte membrane dynamics and a new form of astrocyte-neuron interaction.
Collapse
Affiliation(s)
- Moritz Armbruster
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA.
| | - Saptarnab Naskar
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Jacqueline P Garcia
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA.,Cell, Molecular, and Developmental Biology Program, Tufts Graduate School of Biomedical Sciences, Boston, MA, USA
| | - Mary Sommer
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Elliot Kim
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Yoav Adam
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.,Edmond and Lily Safra Center for Brain Sciences, the Hebrew University of Jerusalem, Jerusalem, Israel
| | - Philip G Haydon
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA
| | - Edward S Boyden
- Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.,Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.,McGovern Institute, Massachusetts Institute of Technology, Cambridge, MA, USA.,Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, MA, USA.,Koch Institute, Massachusetts Institute of Technology, Cambridge, MA, USA.,Center for Neurobiological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Adam E Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.,Department of Physics, Harvard University, Cambridge, MA, USA
| | - Chris G Dulla
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, USA.
| |
Collapse
|
20
|
Lo J, Forst AL, Warth R, Zdebik AA. EAST/SeSAME Syndrome and Beyond: The Spectrum of Kir4.1- and Kir5.1-Associated Channelopathies. Front Physiol 2022; 13:852674. [PMID: 35370765 PMCID: PMC8965613 DOI: 10.3389/fphys.2022.852674] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/08/2022] [Indexed: 12/13/2022] Open
Abstract
In 2009, two groups independently linked human mutations in the inwardly rectifying K+ channel Kir4.1 (gene name KCNJ10) to a syndrome affecting the central nervous system (CNS), hearing, and renal tubular salt reabsorption. The autosomal recessive syndrome has been named EAST (epilepsy, ataxia, sensorineural deafness, and renal tubulopathy) or SeSAME syndrome (seizures, sensorineural deafness, ataxia, intellectual disability, and electrolyte imbalance), accordingly. Renal dysfunction in EAST/SeSAME patients results in loss of Na+, K+, and Mg2+ with urine, activation of the renin-angiotensin-aldosterone system, and hypokalemic metabolic alkalosis. Kir4.1 is highly expressed in affected organs: the CNS, inner ear, and kidney. In the kidney, it mostly forms heteromeric channels with Kir5.1 (KCNJ16). Biallelic loss-of-function mutations of Kir5.1 can also have disease significance, but the clinical symptoms differ substantially from those of EAST/SeSAME syndrome: although sensorineural hearing loss and hypokalemia are replicated, there is no alkalosis, but rather acidosis of variable severity; in contrast to EAST/SeSAME syndrome, the CNS is unaffected. This review provides a framework for understanding some of these differences and will guide the reader through the growing literature on Kir4.1 and Kir5.1, discussing the complex disease mechanisms and the variable expression of disease symptoms from a molecular and systems physiology perspective. Knowledge of the pathophysiology of these diseases and their multifaceted clinical spectrum is an important prerequisite for making the correct diagnosis and forms the basis for personalized therapies.
Collapse
Affiliation(s)
- Jacky Lo
- Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Anna-Lena Forst
- Medical Cell Biology, Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Richard Warth
- Medical Cell Biology, Institute of Physiology, University of Regensburg, Regensburg, Germany
| | - Anselm A. Zdebik
- Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
- Centre for Nephrology, University College London, London, United Kingdom
| |
Collapse
|
21
|
Huang WY, Lai YL, Liu KH, Lin S, Chen HY, Liang CH, Wu HM, Hsu KS. TNFα-mediated necroptosis in brain endothelial cells as a potential mechanism of increased seizure susceptibility in mice following systemic inflammation. J Neuroinflammation 2022; 19:29. [PMID: 35109859 PMCID: PMC8809013 DOI: 10.1186/s12974-022-02406-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 01/27/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Systemic inflammation is a potent contributor to increased seizure susceptibility. However, information regarding the effects of systemic inflammation on cerebral vascular integrity that influence neuron excitability is scarce. Necroptosis is closely associated with inflammation in various neurological diseases. In this study, necroptosis was hypothesized to be involved in the mechanism underlying sepsis-associated neuronal excitability in the cerebrovascular components (e.g., endothelia cells). METHODS Lipopolysaccharide (LPS) was used to induce systemic inflammation. Kainic acid intraperitoneal injection was used to measure the susceptibility of the mice to seizure. The pharmacological inhibitors C87 and GSK872 were used to block the signaling of TNFα receptors and necroptosis. In order to determine the features of the sepsis-associated response in the cerebral vasculature and CNS, brain tissues of mice were obtained for assays of the necroptosis-related protein expression, and for immunofluorescence staining to identify morphological changes in the endothelia and glia. In addition, microdialysis assay was used to assess the changes in extracellular potassium and glutamate levels in the brain. RESULTS Some noteworthy findings, such as increased seizure susceptibility and brain endothelial necroptosis, Kir4.1 dysfunction, and microglia activation were observed in mice following LPS injection. C87 treatment, a TNFα receptor inhibitor, showed considerable attenuation of increased kainic acid-induced seizure susceptibility, endothelial cell necroptosis, microglia activation and restoration of Kir4.1 protein expression in LPS-treated mice. Treatment with GSK872, a RIP3 inhibitor, such as C87, showed similar effects on these changes following LPS injection. CONCLUSIONS The findings of this study showed that TNFα-mediated necroptosis induced cerebrovascular endothelial damage, neuroinflammation and astrocyte Kir4.1 dysregulation, which may coalesce to contribute to the increased seizure susceptibility in LPS-treated mice. Pharmacologic inhibition targeting this necroptosis pathway may provide a promising therapeutic approach to the reduction of sepsis-associated brain endothelia cell injury, astrocyte ion channel dysfunction, and subsequent neuronal excitability.
Collapse
Affiliation(s)
- Wan-Yu Huang
- Institute of Basic Medical Sciences Basic Medicine, College of Medicine, National Cheng-Kung University, Tainan, Taiwan.,Pediatrics of Kung-Ten General Hospital, Taichung City, Taiwan
| | - Yen-Ling Lai
- Inflammation Research and Drug Development Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Ko-Hung Liu
- Inflammation Research and Drug Development Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Shankung Lin
- Inflammation Research and Drug Development Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Hsuan-Ying Chen
- Inflammation Research and Drug Development Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Chih-Hung Liang
- Department of Food Science, Tunghai University, Taichung City, Taiwan
| | - Hung-Ming Wu
- Inflammation Research and Drug Development Center, Changhua Christian Hospital, Changhua, Taiwan. .,Department of Neurology, Changhua Christian Hospital, Changhua City, Taiwan. .,Institute of Acupuncture, School of Chinese Medicine, China Medical University, Taichung City, Taiwan.
| | - Kuei-Sen Hsu
- Institute of Basic Medical Sciences Basic Medicine, College of Medicine, National Cheng-Kung University, Tainan, Taiwan.
| |
Collapse
|
22
|
Huffels CFM, Osborn LM, Hulshof LA, Kooijman L, Henning L, Steinhäuser C, Hol EM. Amyloid-β plaques affect astrocyte Kir4.1 protein expression but not function in the dentate gyrus of APP/PS1 mice. Glia 2022; 70:748-767. [PMID: 34981861 PMCID: PMC9306581 DOI: 10.1002/glia.24137] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 12/16/2021] [Accepted: 12/17/2021] [Indexed: 01/09/2023]
Abstract
Alzheimer pathology is accompanied by astrogliosis. Reactive astrocytes surrounding amyloid plaques may directly affect neuronal communication, and one of the mechanisms by which astrocytes impact neuronal function is by affecting K+ homeostasis. Here we studied, using hippocampal slices from 9‐month‐old Alzheimer mice (APP/PS1) and wild‐type littermates, whether astrocyte function is changed by analyzing Kir4.1 expression and function and astrocyte coupling in astrocytes surrounding amyloid‐β plaques. Immunohistochemical analysis of Kir4.1 protein in the dentate gyrus revealed localized increases in astrocytes surrounding amyloid‐β plaque deposits. We subsequently focused on changes in astrocyte function by using patch‐clamp slice electrophysiology on both plaque‐ and non‐plaque associated astrocytes to characterize general membrane properties. We found that Ba2+‐sensitive Kir4.1 conductance in astrocytes surrounding plaques was not affected by changes in Kir4.1 protein expression. Additional analysis of astrocyte gap junction coupling efficiency in the dentate gyrus revealed no apparent changes. Quantification of basic features of glutamatergic transmission to granule cells did not indicate disturbed neuronal communication in the dentate gyrus of APP/PS1 mice. Together, these results suggest that astrocytes in the dentate gyrus of APP/PS1 mice maintain their ability to buffer extracellular K+ and attempt to rectify imbalances in K+ concentration to maintain normal neuronal and synaptic function, possibly by localized increases in Kir4.1 protein expression. Our earlier transcriptomic data indicated that chronically activated astrocytes lose their neuronal support function. Here we show that, despite localized increased Kir4.1 protein expression, astrocyte Kir4.1 channel dysfunction is likely not involved in the pathogenesis of Alzheimer's disease.
Collapse
Affiliation(s)
- Christiaan F. M. Huffels
- Department of Translational Neuroscience, University Medical Center Utrecht Brain CenterUtrecht UniversityUtrechtThe Netherlands
| | - Lana M. Osborn
- Swammerdam Institute for Life Sciences, Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Lianne A. Hulshof
- Department of Translational Neuroscience, University Medical Center Utrecht Brain CenterUtrecht UniversityUtrechtThe Netherlands
| | - Lieneke Kooijman
- Swammerdam Institute for Life Sciences, Center for NeuroscienceUniversity of AmsterdamAmsterdamThe Netherlands
| | - Lukas Henning
- Institute of Cellular Neurosciences, Medical FacultyUniversity of BonnBonnGermany
| | | | - Elly M. Hol
- Department of Translational Neuroscience, University Medical Center Utrecht Brain CenterUtrecht UniversityUtrechtThe Netherlands
| |
Collapse
|
23
|
Roman C, Vivi E, Di Benedetto B. Morphological Features of Astrocytes in Health and Neuropsychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2021; 26:75-92. [PMID: 34888831 DOI: 10.1007/978-3-030-77375-5_4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Astroglial cells actively partner with several cell types to regulate the arrangement of neuronal circuits both in the developing and adult brain. Morphological features of astroglial cells strongly impact their functional interactions, thereby supporting the hypothesis that aberrancies in glial morphology may trigger the onset of neuropsychiatric disorders. Thus, understanding the factors which modulate astroglial shapes and the development of tools to examine them may help to gain valuable insights about the role of astroglia in physiological and pathological brain states.Here, we present a collection of representative review and original articles describing the major morphological features which define different subtypes of glial cells and emphasize a high degree of heterogeneity typical of these cell types, besides neurons. Furthermore, we offer an overview about first in vitro and in vivo evidences, which highlight an altered morphology of glial cells in brains of psychiatric patients and animal models of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Celia Roman
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Eugenia Vivi
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany
| | - Barbara Di Benedetto
- Department of Psychiatry and Psychotherapy, University of Regensburg, Regensburg, Germany. .,Regensburg Center of Neuroscience, University of Regensburg, Regensburg, Germany.
| |
Collapse
|
24
|
Dynamic expression of homeostatic ion channels in differentiated cortical astrocytes in vitro. Pflugers Arch 2021; 474:243-260. [PMID: 34734327 PMCID: PMC8766406 DOI: 10.1007/s00424-021-02627-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/02/2021] [Accepted: 09/24/2021] [Indexed: 11/16/2022]
Abstract
The capacity of astrocytes to adapt their biochemical and functional features upon physiological and pathological stimuli is a fundamental property at the basis of their ability to regulate the homeostasis of the central nervous system (CNS). It is well known that in primary cultured astrocytes, the expression of plasma membrane ion channels and transporters involved in homeostatic tasks does not closely reflect the pattern observed in vivo. The individuation of culture conditions that promote the expression of the ion channel array found in vivo is crucial when aiming at investigating the mechanisms underlying their dynamics upon various physiological and pathological stimuli. A chemically defined medium containing growth factors and hormones (G5) was previously shown to induce the growth, differentiation, and maturation of primary cultured astrocytes. Here we report that under these culture conditions, rat cortical astrocytes undergo robust morphological changes acquiring a multi-branched phenotype, which develops gradually during the 2-week period of culturing. The shape changes were paralleled by variations in passive membrane properties and background conductance owing to the differential temporal development of inwardly rectifying chloride (Cl−) and potassium (K+) currents. Confocal and immunoblot analyses showed that morphologically differentiated astrocytes displayed a large increase in the expression of the inward rectifier Cl− and K+ channels ClC-2 and Kir4.1, respectively, which are relevant ion channels in vivo. Finally, they exhibited a large diminution of the intermediate filaments glial fibrillary acidic protein (GFAP) and vimentin which are upregulated in reactive astrocytes in vivo. Taken together the data indicate that long-term culturing of cortical astrocytes in this chemical-defined medium promotes a quiescent functional phenotype. This culture model could aid to address the regulation of ion channel expression involved in CNS homeostasis in response to physiological and pathological challenges.
Collapse
|
25
|
Muñoz Y, Cuevas-Pacheco F, Quesseveur G, Murai KK. Light microscopic and heterogeneity analysis of astrocytes in the common marmoset brain. J Neurosci Res 2021; 99:3121-3147. [PMID: 34716617 PMCID: PMC9541330 DOI: 10.1002/jnr.24967] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 08/19/2021] [Accepted: 08/30/2021] [Indexed: 12/12/2022]
Abstract
Astrocytes are abundant cells of the central nervous system (CNS) and are involved in processes including synapse formation/function, ion homeostasis, neurotransmitter uptake, and neurovascular coupling. Recent evidence indicates that astrocytes show diverse molecular, structural, and physiological properties within the CNS. This heterogeneity is reflected in differences in astrocyte structure, gene expression, functional properties, and responsiveness to injury/pathological conditions. Deeper investigation of astrocytic heterogeneity is needed to understand how astrocytes are configured to enable diverse roles in the CNS. While much has been learned about astrocytic heterogeneity in rodents, much less is known about astrocytic heterogeneity in the primate brain where astrocytes have greater size and complexity. The common marmoset (Callithrix jacchus) is a promising non‐human primate model because of similarities between marmosets and humans with respect to genetics, brain anatomy, and cognition/behavior. Here, we investigated the molecular and structural heterogeneity of marmoset astrocytes using an array of astrocytic markers, multi‐label confocal microscopy, and quantitative analysis. We used male and female marmosets and found that marmoset astrocytes show differences in expression of astrocytic markers in cortex, hippocampus, and cerebellum. These differences were accompanied by intra‐regional variation in expression of markers for glutamate/GABA transporters, and potassium and water channels. Differences in astrocyte structure were also found, along with complex interactions with blood vessels, microglia, and neurons. This study contributes to our knowledge of the cellular and molecular features of marmoset astrocytes and is useful for understanding the complex properties of astrocytes in the primate CNS.
Collapse
Affiliation(s)
- Yorka Muñoz
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
| | - Francisco Cuevas-Pacheco
- Department of Mathematics, Universidad Técnica Federico Santa Maria, Valparaiso, Chile.,Advanced Center for Electrical and Electronic Engineering, Universidad Técnica Federico Santa Maria, Valparaiso, Chile
| | - Gaël Quesseveur
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada
| | - Keith K Murai
- Centre for Research in Neuroscience, Department of Neurology & Neurosurgery, Brain Repair and Integrative Neuroscience Program, Research Institute of the McGill University Health Centre, Montreal General Hospital, Montreal, QC, Canada.,Quantitative Life Sciences Graduate Program, McGill University, Montreal, QC, Canada
| |
Collapse
|
26
|
Twible C, Abdo R, Zhang Q. Astrocyte Role in Temporal Lobe Epilepsy and Development of Mossy Fiber Sprouting. Front Cell Neurosci 2021; 15:725693. [PMID: 34658792 PMCID: PMC8514632 DOI: 10.3389/fncel.2021.725693] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 09/02/2021] [Indexed: 11/13/2022] Open
Abstract
Epilepsy affects approximately 50 million people worldwide, with 60% of adult epilepsies presenting an onset of focal origin. The most common focal epilepsy is temporal lobe epilepsy (TLE). The role of astrocytes in the presentation and development of TLE has been increasingly studied and discussed within the literature. The most common histopathological diagnosis of TLE is hippocampal sclerosis. Hippocampal sclerosis is characterized by neuronal cell loss within the Cornu ammonis and reactive astrogliosis. In some cases, mossy fiber sprouting may be observed. Mossy fiber sprouting has been controversial in its contribution to epileptogenesis in TLE patients, and the mechanisms surrounding the phenomenon have yet to be elucidated. Several studies have reported that mossy fiber sprouting has an almost certain co-existence with reactive astrogliosis within the hippocampus under epileptic conditions. Astrocytes are known to play an important role in the survival and axonal outgrowth of central and peripheral nervous system neurons, pointing to a potential role of astrocytes in TLE and associated cellular alterations. Herein, we review the recent developments surrounding the role of astrocytes in the pathogenic process of TLE and mossy fiber sprouting, with a focus on proposed signaling pathways and cellular mechanisms, histological observations, and clinical correlations in human patients.
Collapse
Affiliation(s)
- Carolyn Twible
- Department of Pathology and Lab Medicine, Western University, London, ON, Canada
| | - Rober Abdo
- Department of Pathology and Lab Medicine, Western University, London, ON, Canada.,Department of Anatomy and Cell Biology, Western University, London, ON, Canada
| | - Qi Zhang
- Department of Pathology and Lab Medicine, Western University, London, ON, Canada.,Department of Pathology and Lab Medicine, London Health Sciences Centre, University Hospital, London, ON, Canada
| |
Collapse
|
27
|
Zhou M, Du Y, Aten S, Terman D. On the electrical passivity of astrocyte potassium conductance. J Neurophysiol 2021; 126:1403-1419. [PMID: 34525325 DOI: 10.1152/jn.00330.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Predominant expression of leak-type K+ channels provides astrocytes a high membrane permeability to K+ ions and a hyperpolarized membrane potential that are crucial for astrocyte function in brain homeostasis. In functionally mature astrocytes, the expression of leak K+ channels creates a unique membrane K+ conductance that lacks voltage-dependent rectification. Accordingly, the conductance is named ohmic or passive K+ conductance. Several inwardly rectifying and two-pore domain K+ channels have been investigated for their contributions to passive conductance. Meanwhile, gap junctional coupling has been postulated to underlie the passive behavior of membrane conductance. It is now clear that the intrinsic properties of K+ channels and gap junctional coupling can each act alone or together to bring about a passive behavior of astrocyte conductance. Additionally, while the passive conductance can generally be viewed as a K+ conductance, the actual representation of this conductance is a combined expression of multiple known and unknown K+ channels, which has been further modified by the intricate morphology of individual astrocytes and syncytial gap junctional coupling. The expression of the inwardly rectifying K+ channels explains the inward-going component of passive conductance disobeying Goldman-Hodgkin-Katz constant field outward rectification. However, the K+ channels encoding the outward-going passive currents remain to be determined in the future. Here, we review our current understanding of ion channels and biophysical mechanisms engaged in the passive astrocyte K+ conductance, propose new studies to resolve this long-standing puzzle in astrocyte physiology, and discuss the functional implication(s) of passive behavior of K+ conductance on astrocyte physiology.
Collapse
Affiliation(s)
- Min Zhou
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Yixing Du
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Sydney Aten
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, Ohio
| | - David Terman
- Department of Mathematics, Ohio State University, Columbus, Ohio
| |
Collapse
|
28
|
Wu YC, Sonninen TM, Peltonen S, Koistinaho J, Lehtonen Š. Blood-Brain Barrier and Neurodegenerative Diseases-Modeling with iPSC-Derived Brain Cells. Int J Mol Sci 2021; 22:7710. [PMID: 34299328 PMCID: PMC8307585 DOI: 10.3390/ijms22147710] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/11/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022] Open
Abstract
The blood-brain barrier (BBB) regulates the delivery of oxygen and important nutrients to the brain through active and passive transport and prevents neurotoxins from entering the brain. It also has a clearance function and removes carbon dioxide and toxic metabolites from the central nervous system (CNS). Several drugs are unable to cross the BBB and enter the CNS, adding complexity to drug screens targeting brain disorders. A well-functioning BBB is essential for maintaining healthy brain tissue, and a malfunction of the BBB, linked to its permeability, results in toxins and immune cells entering the CNS. This impairment is associated with a variety of neurological diseases, including Alzheimer's disease and Parkinson's disease. Here, we summarize current knowledge about the BBB in neurodegenerative diseases. Furthermore, we focus on recent progress of using human-induced pluripotent stem cell (iPSC)-derived models to study the BBB. We review the potential of novel stem cell-based platforms in modeling the BBB and address advances and key challenges of using stem cell technology in modeling the human BBB. Finally, we highlight future directions in this area.
Collapse
Affiliation(s)
- Ying-Chieh Wu
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
| | - Tuuli-Maria Sonninen
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
| | - Sanni Peltonen
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
| | - Jari Koistinaho
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| | - Šárka Lehtonen
- Neuroscience Center, University of Helsinki, 00014 Helsinki, Finland; (Y.-C.W.); (T.-M.S.); (S.P.); (J.K.)
- A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70211 Kuopio, Finland
| |
Collapse
|
29
|
McNeill J, Rudyk C, Hildebrand ME, Salmaso N. Ion Channels and Electrophysiological Properties of Astrocytes: Implications for Emergent Stimulation Technologies. Front Cell Neurosci 2021; 15:644126. [PMID: 34093129 PMCID: PMC8173131 DOI: 10.3389/fncel.2021.644126] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 04/26/2021] [Indexed: 12/12/2022] Open
Abstract
Astrocytes comprise a heterogeneous cell population characterized by distinct morphologies, protein expression and function. Unlike neurons, astrocytes do not generate action potentials, however, they are electrically dynamic cells with extensive electrophysiological heterogeneity and diversity. Astrocytes are hyperpolarized cells with low membrane resistance. They are heavily involved in the modulation of K+ and express an array of different voltage-dependent and voltage-independent channels to help with this ion regulation. In addition to these K+ channels, astrocytes also express several different types of Na+ channels; intracellular Na+ signaling in astrocytes has been linked to some of their functional properties. The physiological hallmark of astrocytes is their extensive intracellular Ca2+ signaling cascades, which vary at the regional, subregional, and cellular levels. In this review article, we highlight the physiological properties of astrocytes and the implications for their function and influence of network and synaptic activity. Furthermore, we discuss the implications of these differences in the context of optogenetic and DREADD experiments and consider whether these tools represent physiologically relevant techniques for the interrogation of astrocyte function.
Collapse
Affiliation(s)
- Jessica McNeill
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| | | | | | - Natalina Salmaso
- Department of Neuroscience, Carleton University, Ottawa, ON, Canada
| |
Collapse
|
30
|
Ethanol inhibition of lateral orbitofrontal cortex neuron excitability is mediated via dopamine D1/D5 receptor-induced release of astrocytic glycine. Neuropharmacology 2021; 192:108600. [PMID: 33965399 DOI: 10.1016/j.neuropharm.2021.108600] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 04/16/2021] [Accepted: 04/30/2021] [Indexed: 01/25/2023]
Abstract
Recent findings from this laboratory demonstrate that ethanol reduces the intrinsic excitability of orbitofrontal cortex (OFC) neurons via activation of strychnine-sensitive glycine receptors. Although the mechanism linking ethanol to the release of glycine is currently unknown, astrocytes are a source of neurotransmitters including glycine and activation of dopamine D1-like receptors has been reported to enhance extracellular levels of glycine via a functional reversal of the astrocytic glycine transporter GlyT1. We recently reported that like ethanol, dopamine or a D1/D5 receptor agonist increases a tonic current in lateral OFC (lOFC) neurons. Therefore, in this study, we used whole-cell patch-clamp electrophysiology to examine whether ethanol inhibition of OFC spiking involves the release of glycine from astrocytes and whether this release is dopamine receptor dependent. Ethanol, applied acutely, decreased spiking of lOFC neurons and this effect was blocked by antagonists of GlyT1, the norepinephrine transporter or D1-like but not D2-like receptors. Ethanol enhanced the tonic current of OFC neurons and occluded the effect of dopamine suggesting that ethanol and dopamine may share a common pathway. Altering astrocyte function by suppressing intracellular astrocytic calcium signaling or blocking the astrocyte-specific Kir4.1 potassium channels reduced but did not completely abolish ethanol inhibition of OFC neuron firing. However, when both astrocytic calcium signaling and Kir4.1 channels were inhibited, ethanol had no effect on firing. Ethanol inhibition was also prevented by inhibitors of phospholipase C and conventional isoforms of protein kinase C (cPKC) previously shown to block D1R-induced GlyT1 reversal and PKC inhibition of Kir4.1 channels. Finally, the membrane potential of OFC astrocytes was depolarized by bath application of a Kir4.1 blocker, a D1 agonist or ethanol and ethanol effect was blocked by a D1 antagonist. Together, these findings suggest that acute ethanol inhibits OFC neuron excitability via a D1 receptor-mediated dysregulation of astrocytic glycine transport.
Collapse
|
31
|
Lv J, Fu X, Li Y, Hong G, Li P, Lin J, Xun Y, Fang L, Weng W, Yue R, Li GL, Guan B, Li H, Huang Y, Chai R. Deletion of Kcnj16 in Mice Does Not Alter Auditory Function. Front Cell Dev Biol 2021; 9:630361. [PMID: 33693002 PMCID: PMC7937937 DOI: 10.3389/fcell.2021.630361] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 01/26/2021] [Indexed: 01/13/2023] Open
Abstract
Endolymphatic potential (EP) is the main driving force behind the sensory transduction of hearing, and K+ is the main charge carrier. Kir5.1 is a K+ transporter that plays a significant role in maintaining EP homeostasis, but the expression pattern and role of Kir5.1 (which is encoded by the Kcnj16 gene) in the mouse auditory system has remained unclear. In this study, we found that Kir5.1 was expressed in the mouse cochlea. We checked the inner ear morphology and measured auditory function in Kcnj16–/– mice and found that loss of Kcnj16 did not appear to affect the development of hair cells. There was no significant difference in auditory function between Kcnj16–/– mice and wild-type littermates, although the expression of Kcnma1, Kcnq4, and Kcne1 were significantly decreased in the Kcnj16–/– mice. Additionally, no significant differences were found in the number or distribution of ribbon synapses between the Kcnj16–/– and wild-type mice. In summary, our results suggest that the Kcnj16 gene is not essential for auditory function in mice.
Collapse
Affiliation(s)
- Jun Lv
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xiaolong Fu
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
| | - Yige Li
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
| | - Guodong Hong
- State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China
| | - Peipei Li
- School of Life Sciences and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Jing Lin
- Waksman Institute, Rutgers, The State University of New Jersey, Piscataway, NJ, United States
| | - Youfang Xun
- Department of Otolaryngology, Head and Neck Surgery, Xiangya School of Medicine, Central South University, Changsha, China.,Department of Otolaryngology, Head and Neck Surgery, Clinical Medical College of Yangzhou University, Yangzhou, China
| | - Lucheng Fang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Weibin Weng
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rongyu Yue
- Department of Otolaryngology-Head and Neck Surgery, Provincial Hospital Affiliated to Shandong University, Jinan, China
| | - Geng-Lin Li
- Department of Otorhinolaryngology and ENT Institute, Eye and ENT Hospital, Fudan University, Shanghai, China
| | - Bing Guan
- Department of Otolaryngology, Head and Neck Surgery, Clinical Medical College of Yangzhou University, Yangzhou, China
| | - He Li
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yideng Huang
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,Department of Otolaryngology-Head and Neck Surgery, Hwa Mei Hospital, University of Chinese Academy of Sciences, Ningbo, China
| | - Renjie Chai
- Department of Otolaryngology-Head and Neck Surgery, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.,State Key Laboratory of Bioelectronics, School of Life Sciences and Technology, Jiangsu Province High-Tech Key Laboratory for Bio-Medical Research, Southeast University, Nanjing, China.,Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
32
|
Romeo R, Glotzbach K, Scheller A, Faissner A. Deletion of LRP1 From Astrocytes Modifies Neuronal Network Activity in an in vitro Model of the Tripartite Synapse. Front Cell Neurosci 2021; 14:567253. [PMID: 33519378 PMCID: PMC7842215 DOI: 10.3389/fncel.2020.567253] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 11/30/2020] [Indexed: 12/30/2022] Open
Abstract
The low-density lipoprotein receptor-related protein 1 (LRP1) is a transmembrane receptor that binds over 40 potential ligands and is involved in processes such as cell differentiation, proliferation, and survival. LRP1 is ubiquitously expressed in the organism and enriched among others in blood vessels, liver, and the central nervous system (CNS). There, it is strongly expressed by neurons, microglia, immature oligodendrocytes, and astrocytes. The constitutive LRP1 knockout leads to embryonic lethality. Therefore, previous studies focused on conditional LRP1-knockout strategies and revealed that the deletion of LRP1 causes an increased differentiation of neural stem and precursor cells into astrocytes. Furthermore, astrocytic LRP1 is necessary for the degradation of Aβ and the reduced accumulation of amyloid plaques in Alzheimer’s disease. Although the role of LRP1 in neurons has intensely been investigated, the function of LRP1 with regard to the differentiation and maturation of astrocytes and their functionality is still unknown. To address this question, we generated an inducible conditional transgenic mouse model, where LRP1 is specifically deleted from GLAST-positive astrocyte precursor cells. The recombination with resulting knockout events was visualized by the simultaneous expression of the fluorescent reporter tdTomato. We observed a significantly increased number of GLT-1 expressing astrocytes in LRP1-depleted astrocytic cultures in comparison to control astrocytes. Furthermore, we investigated the influence of astrocytic LRP1 on neuronal activity and synaptogenesis using the co-culture of hippocampal neurons with control or LRP1-depleted astrocytes. These analyses revealed that the LRP1-deficient astrocytes caused a decreased number of single action potentials as well as a negatively influenced neuronal network activity. Moreover, the proportion of pre- and postsynaptic structures was significantly altered in neurons co-cultured with LPR1-depleted astrocytes. However, the number of structural synapses was not affected. Additionally, the supernatant of hippocampal neurons co-cultured with LRP1-deficient astrocytes showed an altered set of cytokines in comparison to the control condition, which potentially contributed to the altered neuronal transmission and synaptogenesis. Our results suggest astrocytic LRP1 as a modulator of synaptic transmission and synaptogenesis by altering the expression of the glutamate transporter on the cell surface on astrocytes and the release of cytokines in vitro.
Collapse
Affiliation(s)
- Ramona Romeo
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Kristin Glotzbach
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| | - Anja Scheller
- Department of Molecular Physiology, Center for Integrative Physiology and Molecular Medicine (CIPMM), University of Saarland, Homburg, Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology, Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
33
|
Rurak GM, Woodside B, Aguilar-Valles A, Salmaso N. Astroglial cells as neuroendocrine targets in forebrain development: Implications for sex differences in psychiatric disease. Front Neuroendocrinol 2021; 60:100897. [PMID: 33359797 DOI: 10.1016/j.yfrne.2020.100897] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/05/2020] [Accepted: 12/15/2020] [Indexed: 12/23/2022]
Abstract
Astroglial cells are the most abundant cell type in the mammalian brain. They are implicated in almost every aspect of brain physiology, including maintaining homeostasis, building and maintaining the blood brain barrier, and the development and maturation of neuronal networks. Critically, astroglia also express receptors for gonadal sex hormones, respond rapidly to gonadal hormones, and are able to synthesize hormones. Thus, they are positioned to guide and mediate sexual differentiation of the brain, particularly neuronal networks in typical and pathological conditions. In this review, we describe astroglial involvement in the organization and development of the brain, and consider known sex differences in astroglial responses to understand how astroglial cell-mediated organization may play a role in forebrain sexual dimorphisms in human populations. Finally, we consider how sexually dimorphic astroglial responses and functions in development may lead to sex differences in vulnerability for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Gareth M Rurak
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Barbara Woodside
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada; Concordia University, Montreal, Quebec, Canada
| | | | - Natalina Salmaso
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada.
| |
Collapse
|
34
|
Smedlund KB, Hill JW. The role of non-neuronal cells in hypogonadotropic hypogonadism. Mol Cell Endocrinol 2020; 518:110996. [PMID: 32860862 DOI: 10.1016/j.mce.2020.110996] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/01/2020] [Accepted: 08/16/2020] [Indexed: 12/18/2022]
Abstract
The hypothalamic-pituitary-gonadal axis is controlled by gonadotropin-releasing hormone (GnRH) released by the hypothalamus. Disruption of this system leads to impaired reproductive maturation and function, a condition known as hypogonadotropic hypogonadism (HH). Most studies to date have focused on genetic causes of HH that impact neuronal development and function. However, variants may also impact the functioning of non-neuronal cells known as glia. Glial cells make up 50% of brain cells of humans, primates, and rodents. They include radial glial cells, microglia, astrocytes, tanycytes, oligodendrocytes, and oligodendrocyte precursor cells. Many of these cells influence the hypothalamic neuroendocrine system controlling fertility. Indeed, glia regulate GnRH neuronal activity and secretion, acting both at their cell bodies and their nerve endings. Recent work has also made clear that these interactions are an essential aspect of how the HPG axis integrates endocrine, metabolic, and environmental signals to control fertility. Recognition of the clinical importance of interactions between glia and the GnRH network may pave the way for the development of new treatment strategies for dysfunctions of puberty and adult fertility.
Collapse
Affiliation(s)
- Kathryn B Smedlund
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA; Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Jennifer W Hill
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA; Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA.
| |
Collapse
|
35
|
das Neves SP, Sousa JC, Sousa N, Cerqueira JJ, Marques F. Altered astrocytic function in experimental neuroinflammation and multiple sclerosis. Glia 2020; 69:1341-1368. [PMID: 33247866 DOI: 10.1002/glia.23940] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 11/14/2020] [Accepted: 11/17/2020] [Indexed: 12/11/2022]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) that affects about 2.5 million people worldwide. In MS, the patients' immune system starts to attack the myelin sheath, leading to demyelination, neurodegeneration, and, ultimately, loss of vital neurological functions such as walking. There is currently no cure for MS and the available treatments only slow the initial phases of the disease. The later-disease mechanisms are poorly understood and do not directly correlate with the activity of immune system cells, the main target of the available treatments. Instead, evidence suggests that disease progression and disability are better correlated with the maintenance of a persistent low-grade inflammation inside the CNS, driven by local glial cells, like astrocytes and microglia. Depending on the context, astrocytes can (a) exacerbate inflammation or (b) promote immunosuppression and tissue repair. In this review, we will address the present knowledge that exists regarding the role of astrocytes in MS and experimental animal models of the disease.
Collapse
Affiliation(s)
- Sofia Pereira das Neves
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal
| | - João Carlos Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal.,Clinical Academic Center, Braga, Portugal
| | - João José Cerqueira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal.,Clinical Academic Center, Braga, Portugal
| | - Fernanda Marques
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.,ICVS/3B's - PT Government Associate Laboratory, Braga, Portugal
| |
Collapse
|
36
|
Smith KE, Murphy P, Jagger DJ. Divergent membrane properties of mouse cochlear glial cells around hearing onset. J Neurosci Res 2020; 99:679-698. [PMID: 33099767 DOI: 10.1002/jnr.24744] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/21/2020] [Accepted: 10/05/2020] [Indexed: 11/11/2022]
Abstract
Spiral ganglion neurons (SGNs) are the primary afferent neurons of the auditory system, and together with their attendant glia, form the auditory nerve. Within the cochlea, satellite glial cells (SGCs) encapsulate the cell body of SGNs, whereas Schwann cells (SCs) wrap their peripherally- and centrally-directed neurites. Despite their likely importance in auditory nerve function and homeostasis, the physiological properties of auditory glial cells have evaded description. Here, we characterized the voltage-activated membrane currents of glial cells from the mouse cochlea. We identified a prominent weak inwardly rectifying current in SGCs within cochlear slice preparations (postnatal day P5-P6), which was also present in presumptive SGCs within dissociated cultures prepared from the cochleae of hearing mice (P14-P15). Pharmacological block by Ba2+ and desipramine suggested that channels belonging to the Kir4 family mediated the weak inwardly rectifying current, and post hoc immunofluorescence implicated the involvement of Kir4.1 subunits. Additional electrophysiological profiles were identified for glial cells within dissociated cultures, suggesting that glial subtypes may have specific membrane properties to support distinct physiological roles. Immunofluorescence using fixed cochlear sections revealed that although Kir4.1 is restricted to SGCs after the onset of hearing, these channels are more widely distributed within the glial population earlier in postnatal development (i.e., within both SGCs and SCs). The decrease in Kir4.1 immunofluorescence during SC maturation was coincident with a reduction of Sox2 expression and advancing neurite myelination. The data suggest a diversification of glial properties occurs in preparation for sound-driven activity in the auditory nerve.
Collapse
Affiliation(s)
- Katie E Smith
- UCL Ear Institute, University College London, London, UK
| | - Phoebe Murphy
- UCL Ear Institute, University College London, London, UK
| | | |
Collapse
|
37
|
Cohen-Salmon M, Slaoui L, Mazaré N, Gilbert A, Oudart M, Alvear-Perez R, Elorza-Vidal X, Chever O, Boulay AC. Astrocytes in the regulation of cerebrovascular functions. Glia 2020; 69:817-841. [PMID: 33058289 DOI: 10.1002/glia.23924] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022]
Abstract
Astrocytes are the most numerous type of neuroglia in the brain and have a predominant influence on the cerebrovascular system; they control perivascular homeostasis, the integrity of the blood-brain barrier, the dialogue with the peripheral immune system, the transfer of metabolites from the blood, and blood vessel contractility in response to neuronal activity. These regulatory processes occur in a specialized interface composed of perivascular astrocyte extensions that almost completely cover the cerebral blood vessels. Scientists have only recently started to study how this interface is formed and how it influences cerebrovascular functions. Here, we review the literature on the astrocytes' role in the regulation of the cerebrovascular system. We cover the anatomy and development of the gliovascular interface, the known gliovascular functions, and molecular factors, the latter's implication in certain pathophysiological situations, and recent cutting-edge experimental tools developed to examine the astrocytes' role at the vascular interface. Finally, we highlight some open questions in this field of research.
Collapse
Affiliation(s)
- Martine Cohen-Salmon
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Leila Slaoui
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Noémie Mazaré
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Alice Gilbert
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Marc Oudart
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Rodrigo Alvear-Perez
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Xabier Elorza-Vidal
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Oana Chever
- Normandie University, UNIROUEN, INSERM, DC2N, IRIB, Rouen, France
| | - Anne-Cécile Boulay
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| |
Collapse
|
38
|
Kessi M, Chen B, Peng J, Tang Y, Olatoutou E, He F, Yang L, Yin F. Intellectual Disability and Potassium Channelopathies: A Systematic Review. Front Genet 2020; 11:614. [PMID: 32655623 PMCID: PMC7324798 DOI: 10.3389/fgene.2020.00614] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Accepted: 05/20/2020] [Indexed: 01/15/2023] Open
Abstract
Intellectual disability (ID) manifests prior to adulthood as severe limitations to intellectual function and adaptive behavior. The role of potassium channelopathies in ID is poorly understood. Therefore, we aimed to evaluate the relationship between ID and potassium channelopathies. We hypothesized that potassium channelopathies are strongly associated with ID initiation, and that both gain- and loss-of-function mutations lead to ID. This systematic review explores the burden of potassium channelopathies, possible mechanisms, advancements using animal models, therapies, and existing gaps. The literature search encompassed both PubMed and Embase up to October 2019. A total of 75 articles describing 338 cases were included in this review. Nineteen channelopathies were identified, affecting the following genes: KCNMA1, KCNN3, KCNT1, KCNT2, KCNJ10, KCNJ6, KCNJ11, KCNA2, KCNA4, KCND3, KCNH1, KCNQ2, KCNAB1, KCNQ3, KCNQ5, KCNC1, KCNB1, KCNC3, and KCTD3. Twelve of these genes presented both gain- and loss-of-function properties, three displayed gain-of-function only, three exhibited loss-of-function only, and one had unknown function. How gain- and loss-of-function mutations can both lead to ID remains largely unknown. We identified only a few animal studies that focused on the mechanisms of ID in relation to potassium channelopathies and some of the few available therapeutic options (channel openers or blockers) appear to offer limited efficacy. In conclusion, potassium channelopathies contribute to the initiation of ID in several instances and this review provides a comprehensive overview of which molecular players are involved in some of the most prominent disease phenotypes.
Collapse
Affiliation(s)
- Miriam Kessi
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China.,Kilimanjaro Christian Medical University College, Moshi, Tanzania.,Mawenzi Regional Referral Hospital, Moshi, Tanzania
| | - Baiyu Chen
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Jing Peng
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Yulin Tang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Eleonore Olatoutou
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Fang He
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Lifen Yang
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| | - Fei Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China.,Hunan Intellectual and Developmental Disabilities Research Center, Changsha, China
| |
Collapse
|
39
|
Johnson CK, Fernandez-Abascal J, Wang Y, Wang L, Bianchi L. The Na +-K +-ATPase is needed in glia of touch receptors for responses to touch in C. elegans. J Neurophysiol 2020; 123:2064-2074. [PMID: 32292107 PMCID: PMC7444924 DOI: 10.1152/jn.00636.2019] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 03/15/2020] [Accepted: 04/10/2020] [Indexed: 01/04/2023] Open
Abstract
Four of the five types of mammalian mechanosensors are composed of nerve endings and accessory cells. In Caenorhabditis elegans we showed that glia support the function of nose touch neurons via the activity of glial Na+ and K+ channels. We show here that a third regulator of Na+ and K+, the Na+-K+-ATPase, is needed in glia of nose touch neurons for touch. Importantly, we show that two Na+-K+-ATPase genes are needed for the function rather than structural integrity and that their ion transport activity is crucial for touch. Finally, when glial Na+-K+-ATPase genes are knocked out, touch can be restored by activation of a third Na+-K+-ATPase. Taken together, these data show the requirement in glia of touch neurons of the function of the Na+-K+-ATPase. These data underscore the importance of the homeostasis of Na+ and K+, most likely in the space surrounding touch neurons, in touch sensation, a function that might be conserved across species.NEW & NOTEWORTHY Increasing evidence supports that accessory cells in mechanosensors regulate neuronal output; however, the glial molecular mechanisms that control this regulation are not fully understood. We show here in Caenorhabditis elegans that specific glial Na+-K+-ATPase genes are needed for nose touch-avoidance behavior. Our data support the requirement of these Na+-K+-ATPases for homeostasis of Na+ and K+ in nose touch receptors. Our data add to our understanding of glial regulation of mechanosensors.
Collapse
Affiliation(s)
- Christina K Johnson
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida
| | - Jesus Fernandez-Abascal
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida
| | - Ying Wang
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida
| | - Lei Wang
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida
| | - Laura Bianchi
- Department Physiology and Biophysics, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
40
|
Abstract
Astrocytes are the most abundant cell type in the central nervous system and have diverse functions in blood–brain barrier maintenance, neural circuitry formation and function, and metabolic regulation. To better understand the diverse roles of astrocytes, we will summarize what is known about astrocyte development and the challenges limiting our understanding of this process. We will also discuss new approaches and technologies advancing the field.
Collapse
Affiliation(s)
- Ekin Su Akdemir
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, 77030, USA.,Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Anna Yu-Szu Huang
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, 77030, USA.,Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Benjamin Deneen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, 77030, USA.,Program in Developmental Biology, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.,Department of Neurosurgery, Baylor College of Medicine, Houston, TX, 77030, USA
| |
Collapse
|
41
|
Zupanc GKH. Development of a sexual dimorphism in a central pattern generator driving a rhythmic behavior: The role of glia-mediated potassium buffering in the pacemaker nucleus of the weakly electric fish Apteronotus leptorhynchus. Dev Neurobiol 2020; 80:6-15. [PMID: 32090501 DOI: 10.1002/dneu.22736] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Revised: 01/24/2020] [Accepted: 02/20/2020] [Indexed: 01/09/2023]
Abstract
Central pattern generators play a critical role in the neural control of rhythmic behaviors. One of their characteristic features is the ability to modulate the oscillatory output. An important yet little-studied type of modulation involves the generation of oscillations that are sexually dimorphic in frequency. In the weakly electric fish Apteronotus leptorhynchus, the pacemaker nucleus serves as a central pattern generator that drives the electric organ discharge of the fish in a one-to-one fashion. Males discharge at higher frequencies than females-a sexual dimorphism that develops under the influence of steroid hormones. The two principal neurons that constitute the oscillatory network of the pacemaker nucleus are the pacemaker and relay cells. Whereas the number and size of the pacemaker and relay cells are sexually monomorphic, pronounced sex-dependent differences exist in the morphology, and subcellular properties of astrocytes, which form a syncytium closely associated with these neurons. In females, compared to males, the astrocytic syncytium covers a larger area surrounding the pacemaker and relay cells and exhibits higher levels of expression of connexin-43 expression. The latter indicates a strong gap-junction coupling of the individual cells within the syncytium. It is hypothesized that these sex-specific differences result in an increased capacity for buffering of extracellular potassium ions, thereby lowering the potassium equilibrium potential, which, in turn, leads to a decrease in the oscillation frequency. This hypothesis has received strong support from simulations based on computational models of individual neurons and the whole neural network of the pacemaker nucleus.
Collapse
Affiliation(s)
- Günther K H Zupanc
- Laboratory of Neurobiology, Department of Biology, Northeastern University, Boston, Massachusetts, USA
| |
Collapse
|
42
|
Smith KE, Whitcroft K, Law S, Andrews P, Choi D, Jagger DJ. Olfactory ensheathing cells from the nasal mucosa and olfactory bulb have distinct membrane properties. J Neurosci Res 2019; 98:888-901. [PMID: 31797433 DOI: 10.1002/jnr.24566] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 11/14/2019] [Accepted: 11/14/2019] [Indexed: 12/15/2022]
Abstract
Transplantation of olfactory ensheathing cells (OECs) is a potential therapy for the regeneration of damaged neurons. While they maintain tissue homeostasis in the olfactory mucosa (OM) and olfactory bulb (OB), their regenerative properties also support the normal sense of smell by enabling continual turnover and axonal regrowth of olfactory sensory neurons (OSNs). However, the molecular physiology of OECs is not fully understood, especially that of OECs from the mucosa. Here, we carried out whole-cell patch-clamp recordings from individual OECs cultured from the OM and OB of the adult rat, and from the human OM. A subset of OECs from the rat OM cultured 1-3 days in vitro had large weakly rectifying K+ currents, which were sensitive to Ba2+ and desipramine, blockers of Kir4-family channels. Kir4.1 immunofluorescence was detectable in cultured OM cells colabeled for the OEC marker S100, and in S100-labeled cells found adjacent to OSN axons in mucosal sections. OECs cultured from rat OB had distinct properties though, displaying strongly rectifying inward currents at hyperpolarized membrane potentials and strongly rectifying outward currents at depolarized potentials. Kir4.1 immunofluorescence was not evident in OECs adjacent to axons of OSNs in the OB. A subset of human OECs cultured from the OM of adults had membrane properties comparable to those of the rat OM that is dominated by Ba2+ -sensitive weak inwardly rectifying currents. The membrane properties of peripheral OECs are different to those of central OECs, suggesting they may play distinct roles during olfaction.
Collapse
Affiliation(s)
- Katie E Smith
- UCL Ear Institute, University College London, London, UK
| | - Katherine Whitcroft
- UCL Ear Institute, University College London, London, UK.,Royal National Throat Nose & Ear Hospital, London, UK
| | - Stuart Law
- Institute of Neurology, University College London, London, UK
| | - Peter Andrews
- UCL Ear Institute, University College London, London, UK.,Royal National Throat Nose & Ear Hospital, London, UK
| | - David Choi
- Institute of Neurology, University College London, London, UK
| | | |
Collapse
|
43
|
Xu S, Sun Q, Fan J, Jiang Y, Yang W, Cui Y, Yu Z, Jiang H, Li B. Role of Astrocytes in Post-traumatic Epilepsy. Front Neurol 2019; 10:1149. [PMID: 31798512 PMCID: PMC6863807 DOI: 10.3389/fneur.2019.01149] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 10/14/2019] [Indexed: 12/25/2022] Open
Abstract
Traumatic brain injury, a common cause of acquired epilepsy, is typical to find necrotic cell death within the injury core. The dynamic changes in astrocytes surrounding the injury core contribute to epileptic seizures associated with intense neuronal firing. However, little is known about the molecular mechanisms that activate astrocytes during traumatic brain injury or the effect of functional changes of astrocytes on seizures. In this comprehensive review, we present our cumulated understanding of the complex neurological affection in astrocytes after traumatic brain injury. We approached the problem through describing the changes of cell morphology, neurotransmitters, biochemistry, and cytokines in astrocytes during post-traumatic epilepsy. In addition, we also discussed the relationship between dynamic changes in astrocytes and seizures and the current pharmacologic agents used for treatment. Hopefully, this review will provide a more detailed knowledge from which better therapeutic strategies can be developed to treat post-traumatic epilepsy.
Collapse
Affiliation(s)
- Songbai Xu
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, China
| | - Qihan Sun
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Jie Fan
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Yuanyuan Jiang
- School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Yifeng Cui
- Department of Pediatrics, Yanbian Maternal and Child Health Hospital, Yanji, China
| | - Zhenxiang Yu
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, China
| | - Huiyi Jiang
- Department of Neurosurgery, the First Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
44
|
Verkhratsky A, Parpura V, Vardjan N, Zorec R. Physiology of Astroglia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1175:45-91. [PMID: 31583584 DOI: 10.1007/978-981-13-9913-8_3] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Astrocytes are principal cells responsible for maintaining the brain homeostasis. Additionally, these glial cells are also involved in homocellular (astrocyte-astrocyte) and heterocellular (astrocyte-other cell types) signalling and metabolism. These astroglial functions require an expression of the assortment of molecules, be that transporters or pumps, to maintain ion concentration gradients across the plasmalemma and the membrane of the endoplasmic reticulum. Astrocytes sense and balance their neurochemical environment via variety of transmitter receptors and transporters. As they are electrically non-excitable, astrocytes display intracellular calcium and sodium fluctuations, which are not only used for operative signalling but can also affect metabolism. In this chapter we discuss the molecules that achieve ionic gradients and underlie astrocyte signalling.
Collapse
Affiliation(s)
- Alexei Verkhratsky
- Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK. .,Faculty of Health and Medical Sciences, Center for Basic and Translational Neuroscience, University of Copenhagen, 2200, Copenhagen, Denmark. .,Achucarro Center for Neuroscience, IKERBASQUE, Basque Foundation for Science, 48011, Bilbao, Spain.
| | - Vladimir Parpura
- Department of Neurobiology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nina Vardjan
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia.,Celica Biomedical, Ljubljana, Slovenia
| | - Robert Zorec
- Laboratory of Neuroendocrinology-Molecular Cell Physiology, Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Ljubljana, Slovenia.,Celica Biomedical, Ljubljana, Slovenia
| |
Collapse
|
45
|
Lyman KA, Chetkovich DM. Oligodendrocyte K ir4.1 Channels Clear Out Congested K .. Epilepsy Curr 2019; 19:339-340. [PMID: 31409147 PMCID: PMC6864574 DOI: 10.1177/1535759719868185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Oligodendrocytes Control Potassium Accumulation in White Matter and Seizure Susceptibility. Larson VA, Mironova Y, Vanderpool KG, Waisman A, Rash JE, Agarwal A, Bergles DE. Elife. 2018 Mar 29;7. pii: e34829. doi:10.7554/eLife.34829. The inwardly rectifying K+ channel Kir4.1 is broadly expressed by central nervous system glia and deficits in Kir4.1 lead to seizures and myelin vacuolization. However, the role of oligodendrocyte Kir4.1 channels in controlling myelination and K+ clearance in white matter has not been defined. Here, we show that selective deletion of Kir4.1 from oligodendrocyte progenitors or mature oligodendrocytes did not impair their development or disrupt the structure of myelin. However, mice lacking oligodendrocyte Kir4.1 channels exhibited profound functional impairments, including slower clearance of extracellular K+ and delayed recovery of axons from repetitive stimulation in white matter, as well as spontaneous seizures, a lower seizure threshold, and activity-dependent motor deficits. These results indicate that Kir4.1 channels in oligodendrocytes play an important role in extracellular K+ homeostasis in white matter and that selective loss of this channel from oligodendrocytes is sufficient to impair K+ clearance and promote seizures.
Collapse
|
46
|
Garcia VJ, Rushton DJ, Tom CM, Allen ND, Kemp PJ, Svendsen CN, Mattis VB. Huntington's Disease Patient-Derived Astrocytes Display Electrophysiological Impairments and Reduced Neuronal Support. Front Neurosci 2019; 13:669. [PMID: 31316341 PMCID: PMC6610155 DOI: 10.3389/fnins.2019.00669] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 06/11/2019] [Indexed: 12/03/2022] Open
Abstract
In Huntington’s disease (HD), while the ubiquitously expressed mutant Huntingtin (mtHTT) protein primarily compromises striatal and cortical neurons, glia also undergo disease-contributing alterations. Existing HD models using human induced pluripotent stem cells (iPSCs) have not extensively characterized the role of mtHTT in patient-derived astrocytes. Here physiologically mature astrocytes are generated from HD patient iPSCs. These human astrocytes exhibit hallmark HD phenotypes that occur in mouse models, including impaired inward rectifying K+ currents, lengthened spontaneous Ca2+ waves and reduced cell membrane capacitance. HD astrocytes in co-culture provided reduced support for the maturation of iPSC-derived neurons. In addition, neurons exposed to chronic glutamate stimulation are not protected by HD astrocytes. This iPSC-based HD model demonstrates the critical effects of mtHTT on human astrocytes, which not only broadens the understanding of disease susceptibility beyond cortical and striatal neurons but also increases potential drug targets.
Collapse
Affiliation(s)
- Veronica J Garcia
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - David J Rushton
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States.,Divisions of Biomedicine and Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Colton M Tom
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Nicholas D Allen
- Divisions of Biomedicine and Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Paul J Kemp
- Divisions of Biomedicine and Neuroscience, School of Biosciences, Cardiff University, Cardiff, United Kingdom
| | - Clive N Svendsen
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Virginia B Mattis
- Board of Governors Regenerative Medicine Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
47
|
Hill SA, Blaeser AS, Coley AA, Xie Y, Shepard KA, Harwell CC, Gao WJ, Garcia ADR. Sonic hedgehog signaling in astrocytes mediates cell type-specific synaptic organization. eLife 2019; 8:45545. [PMID: 31194676 PMCID: PMC6629371 DOI: 10.7554/elife.45545] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 06/13/2019] [Indexed: 01/08/2023] Open
Abstract
Astrocytes have emerged as integral partners with neurons in regulating synapse formation and function, but the mechanisms that mediate these interactions are not well understood. Here, we show that Sonic hedgehog (Shh) signaling in mature astrocytes is required for establishing structural organization and remodeling of cortical synapses in a cell type-specific manner. In the postnatal cortex, Shh signaling is active in a subpopulation of mature astrocytes localized primarily in deep cortical layers. Selective disruption of Shh signaling in astrocytes produces a dramatic increase in synapse number specifically on layer V apical dendrites that emerges during adolescence and persists into adulthood. Dynamic turnover of dendritic spines is impaired in mutant mice and is accompanied by an increase in neuronal excitability and a reduction of the glial-specific, inward-rectifying K+ channel Kir4.1. These data identify a critical role for Shh signaling in astrocyte-mediated modulation of neuronal activity required for sculpting synapses. A central system of neurons in the spinal cord and brain coordinate most of our body’s actions, ranging from regulating our heart rate to controlling our movement and thoughts. As the brain develops, neurons form specialized contacts with one another known as synapses. If the number of synapses is not properly regulated this can disrupt communication between the neurons, leading to diseases like schizophrenia and autism. As the brain develops, it first forms an excess of synapses and later eliminates unnecessary or weak connections. Various factors, such gene expression or a neuron’s level of activity, regulate this turnover process. However, neurons cannot do this alone, and rely on other types of cells to help regulate their behavior. In the central nervous system, for example, a cell called an astrocyte is known to support the formation and activity of synapses. Now, Hill and Blaeser et al. show that astrocytes also exert influence over synaptic turnover during development, leading to long lasting changes in the number of synapses. Hill, Blaeser et al. revealed that disrupting activity of the signaling pathway known as Sonic hedgehog, or Shh for short, in the astrocytes of mice led to disordered synaptic connections. Notably, neurons produce Shh, suggesting that neurons use this signaling pathway to communicate to specific astrocyte partners. Further experiments showed that reducing astrocyte’s ability to respond to Shh impaired synaptic turnover as the brain developed, leading to an overabundance of synapses. Importantly, these effects were only found to influence neuron populations associated with astrocytes that actively use Shh signaling. This suggests that distinct populations of neurons and astrocytes interact in specialized ways to build the connections within the nervous system. To address how astrocytes use Shh signaling to regulate synaptic turnover, Hill, Blaeser et al. examined gene expression changes in astrocytes that lack Shh signaling. Astrocytes with a reduced capacity to respond to Shh were found to have lower levels of a protein responsible for transporting potassium ions into and out of the cell. This impairs astrocyte’s ability to regulate neuronal activity, which may lead to a failure in eliminating unnecessary synapses. Understanding how synapses are controlled and organized by astrocytes could help identify new ways to treat diseases of the developing nervous system. However, further studies would be needed to improve our understanding of how this process works.
Collapse
Affiliation(s)
- Steven A Hill
- Department of Biology, Drexel University, Philadelphia, United States
| | - Andrew S Blaeser
- Department of Biology, Drexel University, Philadelphia, United States
| | - Austin A Coley
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, United States
| | - Yajun Xie
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | | | - Corey C Harwell
- Department of Neurobiology, Harvard Medical School, Boston, United States
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, United States
| | - A Denise R Garcia
- Department of Biology, Drexel University, Philadelphia, United States.,Department of Neurobiology and Anatomy, Drexel University College of Medicine, Philadelphia, United States
| |
Collapse
|
48
|
Verma V, Paul A, Amrapali Vishwanath A, Vaidya B, Clement JP. Understanding intellectual disability and autism spectrum disorders from common mouse models: synapses to behaviour. Open Biol 2019; 9:180265. [PMID: 31185809 PMCID: PMC6597757 DOI: 10.1098/rsob.180265] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Normal brain development is highly dependent on the timely coordinated actions of genetic and environmental processes, and an aberration can lead to neurodevelopmental disorders (NDDs). Intellectual disability (ID) and autism spectrum disorders (ASDs) are a group of co-occurring NDDs that affect between 3% and 5% of the world population, thus presenting a great challenge to society. This problem calls for the need to understand the pathobiology of these disorders and to design new therapeutic strategies. One approach towards this has been the development of multiple analogous mouse models. This review discusses studies conducted in the mouse models of five major monogenic causes of ID and ASDs: Fmr1, Syngap1, Mecp2, Shank2/3 and Neuroligins/Neurnexins. These studies reveal that, despite having a diverse molecular origin, the effects of these mutations converge onto similar or related aetiological pathways, consequently giving rise to the typical phenotype of cognitive, social and emotional deficits that are characteristic of ID and ASDs. This convergence, therefore, highlights common pathological nodes that can be targeted for therapy. Other than conventional therapeutic strategies such as non-pharmacological corrective methods and symptomatic alleviation, multiple studies in mouse models have successfully proved the possibility of pharmacological and genetic therapy enabling functional recovery.
Collapse
Affiliation(s)
- Vijaya Verma
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research , Jakkur, Bengaluru 560 064, Karnataka, India
| | - Abhik Paul
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research , Jakkur, Bengaluru 560 064, Karnataka, India
| | - Anjali Amrapali Vishwanath
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research , Jakkur, Bengaluru 560 064, Karnataka, India
| | - Bhupesh Vaidya
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research , Jakkur, Bengaluru 560 064, Karnataka, India
| | - James P Clement
- Neuroscience Unit, Jawaharlal Nehru Centre for Advanced Scientific Research , Jakkur, Bengaluru 560 064, Karnataka, India
| |
Collapse
|
49
|
The contribution of astrocytes to the neuroinflammatory response in multiple sclerosis and experimental autoimmune encephalomyelitis. Acta Neuropathol 2019; 137:757-783. [PMID: 30847559 DOI: 10.1007/s00401-019-01980-7] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 02/21/2019] [Accepted: 02/23/2019] [Indexed: 02/06/2023]
Abstract
Neuroinflammation is the coordinated response of the central nervous system (CNS) to threats to its integrity posed by a variety of conditions, including autoimmunity, pathogens and trauma. Activated astrocytes, in concert with other cellular elements of the CNS and immune system, are important players in the modulation of the neuroinflammatory response. During neurological disease, they produce and respond to cellular signals that often lead to dichotomous processes, which can promote further damage or contribute to repair. This occurs also in multiple sclerosis (MS), where astrocytes are now recognized as key components of its immunopathology. Evidence supporting this role has emerged not only from studies in MS patients, but also from animal models, among which the experimental autoimmune encephalomyelitis (EAE) model has proved especially instrumental. Based on this premise, the purpose of the present review is to summarize the current knowledge of astrocyte behavior in MS and EAE. Following a brief description of the pathological characteristics of the two diseases and the main functional roles of astrocytes in CNS physiology, we will delve into the specific responses of this cell population, analyzing MS and EAE in parallel. We will define the temporal and anatomical profile of astroglial activation, then focus on key processes they participate in. These include: (1) production and response to soluble mediators (e.g., cytokines and chemokines), (2) regulation of oxidative stress, and (3) maintenance of BBB integrity and function. Finally, we will review the state of the art on the available methods to measure astroglial activation in vivo in MS patients, and how this could be exploited to optimize diagnosis, prognosis and treatment decisions. Ultimately, we believe that integrating the knowledge obtained from studies in MS and EAE may help not only better understand the pathophysiology of MS, but also uncover new signals to be targeted for therapeutic intervention.
Collapse
|
50
|
Diseases of connexins expressed in myelinating glia. Neurosci Lett 2019; 695:91-99. [DOI: 10.1016/j.neulet.2017.05.037] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 05/15/2017] [Accepted: 05/19/2017] [Indexed: 11/23/2022]
|