1
|
Ma Z, Zhang W, Wang C, Su Y, Yi C, Niu J. A New Acquaintance of Oligodendrocyte Precursor Cells in the Central Nervous System. Neurosci Bull 2024; 40:1573-1589. [PMID: 39042298 PMCID: PMC11422404 DOI: 10.1007/s12264-024-01261-8] [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: 11/06/2023] [Accepted: 03/21/2024] [Indexed: 07/24/2024] Open
Abstract
Oligodendrocyte precursor cells (OPCs) are a heterogeneous multipotent population in the central nervous system (CNS) that appear during embryogenesis and persist as resident cells in the adult brain parenchyma. OPCs could generate oligodendrocytes to participate in myelination. Recent advances have renewed our knowledge of OPC biology by discovering novel markers of oligodendroglial cells, the myelin-independent roles of OPCs, and the regulatory mechanism of OPC development. In this review, we will explore the updated knowledge on OPC identity, their multifaceted roles in the CNS in health and diseases, as well as the regulatory mechanisms that are involved in their developmental stages, which hopefully would contribute to a further understanding of OPCs and attract attention in the field of OPC biology.
Collapse
Affiliation(s)
- Zexuan Ma
- Department of Histology and Embryology, College of basic medicine, Third Military Medical University, Chongqing, 400038, China
| | - Wei Zhang
- Department of Histology and Embryology, College of basic medicine, Third Military Medical University, Chongqing, 400038, China
| | - Chenmeng Wang
- Department of Histology and Embryology, College of basic medicine, Third Military Medical University, Chongqing, 400038, China
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Yixun Su
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Chenju Yi
- Research Centre, Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Guangzhou, China.
- Shenzhen Key Laboratory of Chinese Medicine Active substance screening and Translational Research, Shenzhen, 518107, China.
| | - Jianqin Niu
- Department of Histology and Embryology, College of basic medicine, Third Military Medical University, Chongqing, 400038, China.
- Chongqing Key Laboratory of Neurobiology, Chongqing, 400038, China.
| |
Collapse
|
2
|
Lu TY, Hanumaihgari P, Hsu ET, Agarwal A, Kawaguchi R, Calabresi PA, Bergles DE. Norepinephrine modulates calcium dynamics in cortical oligodendrocyte precursor cells promoting proliferation during arousal in mice. Nat Neurosci 2023; 26:1739-1750. [PMID: 37697112 PMCID: PMC10630072 DOI: 10.1038/s41593-023-01426-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 08/10/2023] [Indexed: 09/13/2023]
Abstract
Oligodendrocytes, the myelinating cells of the central nervous system (CNS), are generated from oligodendrocyte precursor cells (OPCs) that express neurotransmitter receptors. However, the mechanisms that affect OPC activity in vivo and the physiological roles of neurotransmitter signaling in OPCs are unclear. In this study, we generated a transgenic mouse line that expresses membrane-anchored GCaMP6s in OPCs and used longitudinal two-photon microscopy to monitor OPC calcium (Ca2+) dynamics in the cerebral cortex. OPCs exhibit focal and transient Ca2+ increases within their processes that are enhanced during locomotion-induced increases in arousal. The Ca2+ transients occur independently of excitatory neuron activity, rapidly decline when OPCs differentiate and are inhibited by anesthesia, sedative agents or noradrenergic receptor antagonists. Conditional knockout of α1A adrenergic receptors in OPCs suppresses spontaneous and locomotion-induced Ca2+ increases and reduces OPC proliferation. Our results demonstrate that OPCs are directly modulated by norepinephrine in vivo to enhance Ca2+ dynamics and promote population homeostasis.
Collapse
Affiliation(s)
- Tsai-Yi Lu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Priyanka Hanumaihgari
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Eric T Hsu
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - Amit Agarwal
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
- Chica and Heinz Schaller Research Group, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Riki Kawaguchi
- Department of Psychiatry & Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, CA, USA
| | - Peter A Calabresi
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurology, Division of Neuroimmunology and Neurological Infections, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Dwight E Bergles
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA.
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
3
|
Masson MA, Nait-Oumesmar B. Emerging concepts in oligodendrocyte and myelin formation, inputs from the zebrafish model. Glia 2023; 71:1147-1163. [PMID: 36645033 DOI: 10.1002/glia.24336] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/20/2022] [Accepted: 12/29/2022] [Indexed: 01/17/2023]
Abstract
Oligodendrocytes (OLs) are the myelinating cells of the central nervous system (CNS), which are derived from OL precursor cells. Myelin insulates axons allowing the saltatory conduction of action potentials and also provides trophic and metabolic supports to axons. Interestingly, oligodendroglial cells have the capacity to sense neuronal activity, which regulates myelin sheath formation via the vesicular release of neurotransmitters. Neuronal activity-dependent regulation of myelination is mediated by specialized interaction between axons and oligodendroglia, involving both synaptic and extra-synaptic modes of communications. The zebrafish has provided key advantages for the study of the myelination process in the CNS. External development and transparent larval stages of this vertebrate specie combined with the existence of several transgenic reporter lines provided key advances in oligodendroglial cell biology, axo-glial interactions and CNS myelination. In this publication, we reviewed and discussed the most recent knowledge on OL development and myelin formation, with a focus on mechanisms regulating these fundamental biological processes in the zebrafish. Especially, we highlighted the critical function of axons and oligodendroglia modes of communications and calcium signaling in myelin sheath formation and growth. Finally, we reviewed the relevance of these knowledge's in demyelinating diseases and drug discovery of pharmacological compounds favoring myelin regeneration.
Collapse
Affiliation(s)
- Mary-Amélie Masson
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Brahim Nait-Oumesmar
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute - ICM, Inserm, CNRS, APHP, Hôpital de la Pitié-Salpêtrière, Paris, France
| |
Collapse
|
4
|
Perez-Gianmarco L, Kurt B, Kukley M. Technical approaches and challenges to study AMPA receptors in oligodendrocyte lineage cells: Past, present, and future. Glia 2023; 71:819-847. [PMID: 36453615 DOI: 10.1002/glia.24305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 11/05/2022] [Accepted: 11/10/2022] [Indexed: 12/03/2022]
Abstract
Receptors for α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPARs) are ligand-gated ionotropic receptors for glutamate that is a major excitatory neurotransmitter in the central nervous system. AMPARs are located at postsynaptic sites of neuronal synapses where they mediate fast synaptic signaling and synaptic plasticity. Remarkably, AMPARs are also expressed by glial cells. Their expression by the oligodendrocyte (OL) lineage cells is of special interest because AMPARs mediate fast synaptic communication between neurons and oligodendrocyte progenitor cells (OPCs), modulate proliferation and differentiation of OPCs, and may also be involved in regulation of myelination. On the other hand, during pathological conditions, AMPARs may mediate damage of the OL lineage cells. In the present review, we focus on the technical approaches that have been used to study AMPARs in the OL lineage cells, and discuss future perspectives of AMPAR research in these glial cells.
Collapse
Affiliation(s)
- Lucila Perez-Gianmarco
- Laboratory of Neuronal and Glial Physiology, Achucarro Basque Center for Neuroscience, Leioa, Spain.,Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Begüm Kurt
- Laboratory of Neuronal and Glial Physiology, Achucarro Basque Center for Neuroscience, Leioa, Spain.,Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Maria Kukley
- Laboratory of Neuronal and Glial Physiology, Achucarro Basque Center for Neuroscience, Leioa, Spain.,Ikerbasque - Basque Foundation for Science, Bilbao, Spain
| |
Collapse
|
5
|
Recent Insights into the Functional Role of AMPA Receptors in the Oligodendrocyte Lineage Cells In Vivo. Int J Mol Sci 2023; 24:ijms24044138. [PMID: 36835546 PMCID: PMC9967469 DOI: 10.3390/ijms24044138] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/13/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
This review discusses the experimental findings of several recent studies which investigated the functional role of AMPA receptors (AMPARs) in oligodendrocyte lineage cells in vivo, in mice and in zebrafish. These studies provided valuable information showing that oligodendroglial AMPARs may be involved in the modulation of proliferation, differentiation, and migration of oligodendroglial progenitors, as well as survival of myelinating oligodendrocytes during physiological conditions in vivo. They also suggested that targeting the subunit composition of AMPARs may be an important strategy for treating diseases. However, at the same time, the experimental findings taken together still do not provide a clear picture on the topic. Hence, new ideas and new experimental designs are required for understanding the functional role of AMPARs in the oligodendrocyte lineage cells in vivo. It is also necessary to consider more closely the temporal and spatial aspects of AMPAR-mediated signalling in the oligodendrocyte lineage cells. These two important aspects are routinely discussed by neuronal physiologists studying glutamatergic synaptic transmission, but are rarely debated and thought about by researchers studying glial cells.
Collapse
|
6
|
de Almeida V, Seabra G, Reis-de-Oliveira G, Zuccoli GS, Rumin P, Fioramonte M, Smith BJ, Zuardi AW, Hallak JEC, Campos AC, Crippa JA, Martins-de-Souza D. Cannabinoids modulate proliferation, differentiation, and migration signaling pathways in oligodendrocytes. Eur Arch Psychiatry Clin Neurosci 2022; 272:1311-1323. [PMID: 35622101 DOI: 10.1007/s00406-022-01425-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/02/2022] [Indexed: 11/03/2022]
Abstract
Cannabinoid signaling, mainly via CB1 and CB2 receptors, plays an essential role in oligodendrocyte health and functions. However, the specific molecular signals associated with the activation or blockade of CB1 and CB2 receptors in this glial cell have yet to be elucidated. Mass spectrometry-based shotgun proteomics and in silico biology tools were used to determine which signaling pathways and molecular mechanisms are triggered in a human oligodendrocytic cell line (MO3.13) by several pharmacological stimuli: the phytocannabinoid cannabidiol (CBD); CB1 and CB2 agonists ACEA, HU308, and WIN55, 212-2; CB1 and CB2 antagonists AM251 and AM630; and endocannabinoids anandamide (AEA) and 2-arachidonoylglycerol (2-AG). The modulation of cannabinoid signaling in MO3.13 was found to affect pathways linked to cell proliferation, migration, and differentiation of oligodendrocyte progenitor cells. Additionally, we found that carbohydrate and lipid metabolism, as well as mitochondrial function, were modulated by these compounds. Comparing the proteome changes and upstream regulators among treatments, the highest overlap was between the CB1 and CB2 antagonists, followed by overlaps between AEA and 2-AG. Our study opens new windows of opportunities, suggesting that cannabinoid signaling in oligodendrocytes might be relevant in the context of demyelinating and neurodegenerative diseases. Proteomics data are available at ProteomeXchange (PXD031923).
Collapse
Affiliation(s)
- Valéria de Almeida
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil.
| | - Gabriela Seabra
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil
| | - Guilherme Reis-de-Oliveira
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil
| | - Giuliana S Zuccoli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil
| | - Priscila Rumin
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil
| | - Mariana Fioramonte
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil
| | - Bradley J Smith
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil
| | - Antonio W Zuardi
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,National Institute for Science and Technology, Translational Medicine, São Paulo, Brazil
| | - Jaime E C Hallak
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,National Institute for Science and Technology, Translational Medicine, São Paulo, Brazil
| | - Alline C Campos
- National Institute for Science and Technology, Translational Medicine, São Paulo, Brazil.,Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - José A Crippa
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,National Institute for Science and Technology, Translational Medicine, São Paulo, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil. .,Instituto Nacional de Biomarcadores Em Neuropsiquiatria (INBION) Conselho Nacional de Desenvolvimento Científico E Tecnológico, São Paulo, Brazil. .,Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, SP, Brazil. .,D'Or Institute for Research and Education (IDOR), São Paulo, Brazil.
| |
Collapse
|
7
|
Bayón-Cordero L, Ochoa-Bueno BI, Ruiz A, Ozalla M, Matute C, Sánchez-Gómez MV. GABA Receptor Agonists Protect From Excitotoxic Damage Induced by AMPA in Oligodendrocytes. Front Pharmacol 2022; 13:897056. [PMID: 35959434 PMCID: PMC9360600 DOI: 10.3389/fphar.2022.897056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/06/2022] [Indexed: 11/17/2022] Open
Abstract
Oligodendrocytes are the myelin forming cells of the central nervous system, and their vulnerability to excitotoxicity induced by glutamate contributes to the pathogenesis of neurological disorders including brain ischemia and neurodegenerative diseases, such as multiple sclerosis. In addition to glutamate receptors, oligodendrocytes express GABA receptors (GABAR) that are involved in their survival and differentiation. The interactions between glutamate and GABAergic systems are well documented in neurons, under both physiological and pathological conditions, but this potential crosstalk in oligodendrocytes has not been studied in depth. Here, we evaluated the protective effect of GABAR agonists, baclofen (GABAB) and muscimol (GABAA), against AMPA-induced excitotoxicity in cultured rat oligodendrocytes. First, we observed that both baclofen and muscimol reduced cell death and caspase-3 activation after AMPA insult, proving their oligoprotective potential. Interestingly, analysis of the cell-surface expression of calcium-impermeable GluR2 subunits in oligodendrocytes revealed that GABAergic agonists significantly reverted GluR2 internalization induced by AMPA. We determined that baclofen and muscimol also impaired AMPA-induced intracellular calcium increase and subsequent mitochondrial membrane potential alteration, ROS generation, and calpain activation. However, AMPA-triggered activation of Src, Akt, JNK and CREB was not affected by baclofen or muscimol. Overall, our results suggest that GABAR activation initiates alternative molecular mechanisms that attenuate AMPA-mediated apoptotic excitotoxicity in oligodendrocytes by interfering with expression of GluR subunits in membranes and with calcium-dependent intracellular signaling pathways. Together, these findings provide evidence of GABAR agonists as potential oligodendroglial protectants in central nervous system disorders.
Collapse
Affiliation(s)
- Laura Bayón-Cordero
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Blanca Isabel Ochoa-Bueno
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Asier Ruiz
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Marina Ozalla
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Carlos Matute
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - María Victoria Sánchez-Gómez
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| |
Collapse
|
8
|
Neely SA, Lyons DA. Insights Into Central Nervous System Glial Cell Formation and Function From Zebrafish. Front Cell Dev Biol 2021; 9:754606. [PMID: 34912801 PMCID: PMC8666443 DOI: 10.3389/fcell.2021.754606] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 10/05/2021] [Indexed: 12/23/2022] Open
Abstract
The term glia describes a heterogenous collection of distinct cell types that make up a large proportion of our nervous system. Although once considered the glue of the nervous system, the study of glial cells has evolved significantly in recent years, with a large body of literature now highlighting their complex and diverse roles in development and throughout life. This progress is due, in part, to advances in animal models in which the molecular and cellular mechanisms of glial cell development and function as well as neuron-glial cell interactions can be directly studied in vivo in real time, in intact neural circuits. In this review we highlight the instrumental role that zebrafish have played as a vertebrate model system for the study of glial cells, and discuss how the experimental advantages of the zebrafish lend themselves to investigate glial cell interactions and diversity. We focus in particular on recent studies that have provided insight into the formation and function of the major glial cell types in the central nervous system in zebrafish.
Collapse
Affiliation(s)
- Sarah A. Neely
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - David A. Lyons
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
9
|
Fairless R, Bading H, Diem R. Pathophysiological Ionotropic Glutamate Signalling in Neuroinflammatory Disease as a Therapeutic Target. Front Neurosci 2021; 15:741280. [PMID: 34744612 PMCID: PMC8567076 DOI: 10.3389/fnins.2021.741280] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/30/2021] [Indexed: 01/15/2023] Open
Abstract
Glutamate signalling is an essential aspect of neuronal communication involving many different glutamate receptors, and underlies the processes of memory, learning and synaptic plasticity. Despite neuroinflammatory diseases covering a range of maladies with very different biological causes and pathophysiologies, a central role for dysfunctional glutamate signalling is becoming apparent. This is not just restricted to the well-described role of glutamate in mediating neurodegeneration, but also includes a myriad of other influences that glutamate can exert on the vasculature, as well as immune cell and glial regulation, reflecting the ability of neurons to communicate with these compartments in order to couple their activity with neuronal requirements. Here, we discuss the role of pathophysiological glutamate signalling in neuroinflammatory disease, using both multiple sclerosis and Alzheimer's disease as examples, and how current steps are being made to harness our growing understanding of these processes in the development of neuroprotective strategies. This review focuses in particular on N-methyl-D-aspartate (NMDA) and 2-amino-3-(3-hydroxy-5-methylisooxazol-4-yl) propionate (AMPA) type ionotropic glutamate receptors, although metabotropic, G-protein-coupled glutamate receptors may also contribute to neuroinflammatory processes. Given the indispensable roles of glutamate-gated ion channels in synaptic communication, means of pharmacologically distinguishing between physiological and pathophysiological actions of glutamate will be discussed that allow deleterious signalling to be inhibited whilst minimising the disturbance of essential neuronal function.
Collapse
Affiliation(s)
- Richard Fairless
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Hilmar Bading
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Ricarda Diem
- Department of Neurology, University Clinic Heidelberg, Heidelberg, Germany.,Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
10
|
Hansen KB, Wollmuth LP, Bowie D, Furukawa H, Menniti FS, Sobolevsky AI, Swanson GT, Swanger SA, Greger IH, Nakagawa T, McBain CJ, Jayaraman V, Low CM, Dell'Acqua ML, Diamond JS, Camp CR, Perszyk RE, Yuan H, Traynelis SF. Structure, Function, and Pharmacology of Glutamate Receptor Ion Channels. Pharmacol Rev 2021; 73:298-487. [PMID: 34753794 PMCID: PMC8626789 DOI: 10.1124/pharmrev.120.000131] [Citation(s) in RCA: 267] [Impact Index Per Article: 89.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Many physiologic effects of l-glutamate, the major excitatory neurotransmitter in the mammalian central nervous system, are mediated via signaling by ionotropic glutamate receptors (iGluRs). These ligand-gated ion channels are critical to brain function and are centrally implicated in numerous psychiatric and neurologic disorders. There are different classes of iGluRs with a variety of receptor subtypes in each class that play distinct roles in neuronal functions. The diversity in iGluR subtypes, with their unique functional properties and physiologic roles, has motivated a large number of studies. Our understanding of receptor subtypes has advanced considerably since the first iGluR subunit gene was cloned in 1989, and the research focus has expanded to encompass facets of biology that have been recently discovered and to exploit experimental paradigms made possible by technological advances. Here, we review insights from more than 3 decades of iGluR studies with an emphasis on the progress that has occurred in the past decade. We cover structure, function, pharmacology, roles in neurophysiology, and therapeutic implications for all classes of receptors assembled from the subunits encoded by the 18 ionotropic glutamate receptor genes. SIGNIFICANCE STATEMENT: Glutamate receptors play important roles in virtually all aspects of brain function and are either involved in mediating some clinical features of neurological disease or represent a therapeutic target for treatment. Therefore, understanding the structure, function, and pharmacology of this class of receptors will advance our understanding of many aspects of brain function at molecular, cellular, and system levels and provide new opportunities to treat patients.
Collapse
Affiliation(s)
- Kasper B Hansen
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Lonnie P Wollmuth
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Derek Bowie
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Hiro Furukawa
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Frank S Menniti
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Alexander I Sobolevsky
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Geoffrey T Swanson
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Sharon A Swanger
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Ingo H Greger
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Terunaga Nakagawa
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Chris J McBain
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Vasanthi Jayaraman
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Chian-Ming Low
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Mark L Dell'Acqua
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Jeffrey S Diamond
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Chad R Camp
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Riley E Perszyk
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Hongjie Yuan
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| | - Stephen F Traynelis
- Center for Structural and Functional Neuroscience, Center for Biomolecular Structure and Dynamics, Division of Biological Sciences, University of Montana, Missoula, MT (K.B.H.); Department of Neurobiology and Behavior, Center for Nervous System Disorders, Stony Brook University, Stony Brook, NY (L.P.W.); Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec, Canada (D.B.); WM Keck Structural Biology Laboratory, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY (H.F.); MindImmune Therapeutics, Inc., The George & Anne Ryan Institute for Neuroscience, University of Rhode Island, Kingston, RI (F.S.M.); Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY (A.I.S.); Department of Pharmacology, Northwestern University Feinberg School of Medicine, Chicago, IL (G.T.S.); Fralin Biomedical Research Institute at Virginia Tech Carilion, Virginia Tech, Roanoke, VA and Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Tech, Blacksburg, VA (S.A.S.); Neurobiology Division, MRC Laboratory of Molecular Biology, Cambridge, United Kingdom (I.H.G.); Department of Molecular Physiology and Biophysics, Center for Structural Biology, Vanderbilt Brain Institute, Vanderbilt University, School of Medicine, Nashville, TN (T.N.); Eunice Kennedy Shriver National Institute of Child Health and Human Development (C.J.M.), and Synaptic Physiology Section, NINDS Intramural Research Program, National Institutes of Health, Bethesda, MD (J.S.D.); Department of Biochemistry and Molecular Biology, University of Texas Health Science Center, Houston, TX (V.J.); Department of Pharmacology, Department of Anaesthesia, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore (C.-M.L.); Department of Pharmacology, University of Colorado School of Medicine, Aurora, CO (M.L.D.); and Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA (C.R.C., R.E.P., H.Y., S.F.T.)
| |
Collapse
|
11
|
Piller M, Werkman IL, Brown RI, Latimer AJ, Kucenas S. Glutamate Signaling via the AMPAR Subunit GluR4 Regulates Oligodendrocyte Progenitor Cell Migration in the Developing Spinal Cord. J Neurosci 2021; 41:5353-5371. [PMID: 33975920 PMCID: PMC8221590 DOI: 10.1523/jneurosci.2562-20.2021] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 04/28/2021] [Accepted: 05/03/2021] [Indexed: 11/21/2022] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) are specified from discrete precursor populations during gliogenesis and migrate extensively from their origins, ultimately distributing throughout the brain and spinal cord during early development. Subsequently, a subset of OPCs differentiates into mature oligodendrocytes, which myelinate axons. This process is necessary for efficient neuronal signaling and organism survival. Previous studies have identified several factors that influence OPC development, including excitatory glutamatergic synapses that form between neurons and OPCs during myelination. However, little is known about how glutamate signaling affects OPC migration before myelination. In this study, we use in vivo, time-lapse imaging in zebrafish in conjunction with genetic and pharmacological perturbation to investigate OPC migration and myelination when the GluR4A ionotropic glutamate receptor subunit is disrupted. In our studies, we observed that gria4a mutant embryos and larvae displayed abnormal OPC migration and altered dorsoventral distribution in the spinal cord. Genetic mosaic analysis confirmed that these effects were cell-autonomous, and we identified that voltage-gated calcium channels were downstream of glutamate receptor signaling in OPCs and could rescue the migration and myelination defects we observed when glutamate signaling was perturbed. These results offer new insights into the complex system of neuron-OPC interactions and reveal a cell-autonomous role for glutamatergic signaling in OPCs during neural development.SIGNIFICANCE STATEMENT The migration of oligodendrocyte progenitor cells (OPCs) is an essential process during development that leads to uniform oligodendrocyte distribution and sufficient myelination for central nervous system function. Here, we demonstrate that the AMPA receptor (AMPAR) subunit GluR4A is an important driver of OPC migration and myelination in vivo and that activated voltage-gated calcium channels are downstream of glutamate receptor signaling in mediating this migration.
Collapse
Affiliation(s)
- Melanie Piller
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
| | - Inge L Werkman
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
| | - Robin Isadora Brown
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
| | - Andrew J Latimer
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
| | - Sarah Kucenas
- Department of Biology, University of Virginia, Charlottesville, Virginia 22904
| |
Collapse
|
12
|
Ramos-Vicente D, Grant SG, Bayés À. Metazoan evolution and diversity of glutamate receptors and their auxiliary subunits. Neuropharmacology 2021; 195:108640. [PMID: 34116111 DOI: 10.1016/j.neuropharm.2021.108640] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 05/27/2021] [Accepted: 06/01/2021] [Indexed: 01/18/2023]
Abstract
Glutamate is the major excitatory neurotransmitter in vertebrate and invertebrate nervous systems. Proteins involved in glutamatergic neurotransmission, and chiefly glutamate receptors and their auxiliary subunits, play key roles in nervous system function. Thus, understanding their evolution and uncovering their diversity is essential to comprehend how nervous systems evolved, shaping cognitive function. Comprehensive phylogenetic analysis of these proteins across metazoans have revealed that their evolution is much more complex than what can be anticipated from vertebrate genomes. This is particularly true for ionotropic glutamate receptors (iGluRs), as their current classification into 6 classes (AMPA, Kainate, Delta, NMDA1, NMDA2 and NMDA3) would be largely incomplete. New work proposes a classification of iGluRs into 4 subfamilies that encompass 10 classes. Vertebrate AMPA, Kainate and Delta receptors would belong to one of these subfamilies, named AKDF, the NMDA subunits would constitute another subfamily and non-vertebrate iGluRs would be organised into the previously unreported Epsilon and Lambda subfamilies. Similarly, the animal evolution of metabotropic glutamate receptors has resulted in the formation of four classes of these receptors, instead of the three currently recognised. Here we review our current knowledge on the animal evolution of glutamate receptors and their auxiliary subunits. This article is part of the special issue on 'Glutamate Receptors - Orphan iGluRs'.
Collapse
Affiliation(s)
- David Ramos-Vicente
- Molecular Physiology of the Synapse Laboratory, Biomedical Research Institute Sant Pau, Barcelona, Spain; Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Seth Gn Grant
- Centre for Clinical Brain Sciences, Chancellor's Building, Edinburgh BioQuarter, University of Edinburgh, Edinburgh, EH16 4SB, UK; Simons Initiative for the Developing Brain (SIDB), Centre for Discovery Brain Sciences, University of Edinburgh, Hugh Robson Building, George Square, Edinburgh, EH8 9XD, UK
| | - Àlex Bayés
- Molecular Physiology of the Synapse Laboratory, Biomedical Research Institute Sant Pau, Barcelona, Spain; Universitat Autònoma de Barcelona, Barcelona, Spain.
| |
Collapse
|
13
|
Cull‐Candy SG, Farrant M. Ca 2+ -permeable AMPA receptors and their auxiliary subunits in synaptic plasticity and disease. J Physiol 2021; 599:2655-2671. [PMID: 33533533 PMCID: PMC8436767 DOI: 10.1113/jp279029] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 01/28/2021] [Indexed: 12/14/2022] Open
Abstract
AMPA receptors are tetrameric glutamate-gated ion channels that mediate a majority of fast excitatory neurotransmission in the brain. They exist as calcium-impermeable (CI-) and calcium-permeable (CP-) subtypes, the latter of which lacks the GluA2 subunit. CP-AMPARs display an array of distinctive biophysical and pharmacological properties that allow them to be functionally identified. This has revealed that they play crucial roles in diverse forms of central synaptic plasticity. Here we summarise the functional hallmarks of CP-AMPARs and describe how these are modified by the presence of auxiliary subunits that have emerged as pivotal regulators of AMPARs. A lasting change in the prevalence of GluA2-containing AMPARs, and hence in the fraction of CP-AMPARs, is a feature in many maladaptive forms of synaptic plasticity and neurological disorders. These include modifications of glutamatergic transmission induced by inflammatory pain, fear conditioning, cocaine exposure, and anoxia-induced damage in neurons and glia. Furthermore, defective RNA editing of GluA2 can cause altered expression of CP-AMPARs and is implicated in motor neuron damage (amyotrophic lateral sclerosis) and the proliferation of cells in malignant gliomas. A number of the players involved in CP-AMPAR regulation have been identified, providing useful insight into interventions that may prevent the aberrant CP-AMPAR expression. Furthermore, recent molecular and pharmacological developments, particularly the discovery of TARP subtype-selective drugs, offer the exciting potential to modify some of the harmful effects of increased CP-AMPAR prevalence in a brain region-specific manner.
Collapse
Affiliation(s)
- Stuart G. Cull‐Candy
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonGower StreetLondonWC1E 6BTUK
| | - Mark Farrant
- Department of Neuroscience, Physiology and PharmacologyUniversity College LondonGower StreetLondonWC1E 6BTUK
| |
Collapse
|
14
|
Baglietto-Vargas D, Forner S, Cai L, Martini AC, Trujillo-Estrada L, Swarup V, Nguyen MMT, Do Huynh K, Javonillo DI, Tran KM, Phan J, Jiang S, Kramár EA, Nuñez-Diaz C, Balderrama-Gutierrez G, Garcia F, Childs J, Rodriguez-Ortiz CJ, Garcia-Leon JA, Kitazawa M, Shahnawaz M, Matheos DP, Ma X, Da Cunha C, Walls KC, Ager RR, Soto C, Gutierrez A, Moreno-Gonzalez I, Mortazavi A, Tenner AJ, MacGregor GR, Wood M, Green KN, LaFerla FM. Generation of a humanized Aβ expressing mouse demonstrating aspects of Alzheimer's disease-like pathology. Nat Commun 2021; 12:2421. [PMID: 33893290 PMCID: PMC8065162 DOI: 10.1038/s41467-021-22624-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 03/15/2021] [Indexed: 11/26/2022] Open
Abstract
The majority of Alzheimer's disease (AD) cases are late-onset and occur sporadically, however most mouse models of the disease harbor pathogenic mutations, rendering them better representations of familial autosomal-dominant forms of the disease. Here, we generated knock-in mice that express wildtype human Aβ under control of the mouse App locus. Remarkably, changing 3 amino acids in the mouse Aβ sequence to its wild-type human counterpart leads to age-dependent impairments in cognition and synaptic plasticity, brain volumetric changes, inflammatory alterations, the appearance of Periodic Acid-Schiff (PAS) granules and changes in gene expression. In addition, when exon 14 encoding the Aβ sequence was flanked by loxP sites we show that Cre-mediated excision of exon 14 ablates hAβ expression, rescues cognition and reduces the formation of PAS granules.
Collapse
Affiliation(s)
- David Baglietto-Vargas
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
- Department of Cell Biology, Genetic and Physiology, Faculty of Sciences, Instituto de Investigacion Biomedica de Malaga-IBIMA, Networking Research Center on Neurodegenerative Diseases (CIBERNED), University of Malaga, Malaga, Spain
| | - Stefania Forner
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Lena Cai
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Alessandra C Martini
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Laura Trujillo-Estrada
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
- Department of Cell Biology, Genetic and Physiology, Faculty of Sciences, Instituto de Investigacion Biomedica de Malaga-IBIMA, Networking Research Center on Neurodegenerative Diseases (CIBERNED), University of Malaga, Malaga, Spain
| | - Vivek Swarup
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Marie Minh Thu Nguyen
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Kelly Do Huynh
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Dominic I Javonillo
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Kristine Minh Tran
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Jimmy Phan
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Shan Jiang
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Enikö A Kramár
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Cristina Nuñez-Diaz
- Department of Cell Biology, Genetic and Physiology, Faculty of Sciences, Instituto de Investigacion Biomedica de Malaga-IBIMA, Networking Research Center on Neurodegenerative Diseases (CIBERNED), University of Malaga, Malaga, Spain
| | | | - Franklin Garcia
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Jessica Childs
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Carlos J Rodriguez-Ortiz
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
- Division of Occupational and Environmental Medicine, Department of Medicine. Center for Occupational and Environmental Health (COEH), University of California, Irvine, CA, USA
| | - Juan Antonio Garcia-Leon
- Department of Cell Biology, Genetic and Physiology, Faculty of Sciences, Instituto de Investigacion Biomedica de Malaga-IBIMA, Networking Research Center on Neurodegenerative Diseases (CIBERNED), University of Malaga, Malaga, Spain
| | - Masashi Kitazawa
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
- Division of Occupational and Environmental Medicine, Department of Medicine. Center for Occupational and Environmental Health (COEH), University of California, Irvine, CA, USA
| | - Mohammad Shahnawaz
- The Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Dina P Matheos
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Xinyi Ma
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Celia Da Cunha
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Ken C Walls
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Rahasson R Ager
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
| | - Claudio Soto
- The Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Antonia Gutierrez
- Department of Cell Biology, Genetic and Physiology, Faculty of Sciences, Instituto de Investigacion Biomedica de Malaga-IBIMA, Networking Research Center on Neurodegenerative Diseases (CIBERNED), University of Malaga, Malaga, Spain
| | - Ines Moreno-Gonzalez
- Department of Cell Biology, Genetic and Physiology, Faculty of Sciences, Instituto de Investigacion Biomedica de Malaga-IBIMA, Networking Research Center on Neurodegenerative Diseases (CIBERNED), University of Malaga, Malaga, Spain
- The Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ali Mortazavi
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Andrea J Tenner
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
- Department of Molecular Biology and Biochemistry, University of California, Irvine, CA, USA
| | - Grant R MacGregor
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
| | - Marcelo Wood
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Kim N Green
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA.
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA.
| | - Frank M LaFerla
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, CA, USA.
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA.
| |
Collapse
|
15
|
Kamen Y, Pivonkova H, Evans KA, Káradóttir RT. A Matter of State: Diversity in Oligodendrocyte Lineage Cells. Neuroscientist 2021; 28:144-162. [PMID: 33567971 DOI: 10.1177/1073858420987208] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) give rise to oligodendrocytes which myelinate axons in the central nervous system. Although classically thought to be a homogeneous population, OPCs are reported to have different developmental origins and display regional and temporal diversity in their transcriptome, response to growth factors, and physiological properties. Similarly, evidence is accumulating that myelinating oligodendrocytes display transcriptional heterogeneity. Analyzing this reported heterogeneity suggests that OPCs, and perhaps also myelinating oligodendrocytes, may exist in different functional cell states. Here, we review the evidence indicating that OPCs and oligodendrocytes are diverse, and we discuss the implications of functional OPC states for myelination in the adult brain and for myelin repair.
Collapse
Affiliation(s)
- Yasmine Kamen
- Wellcome-Medical Research Council Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Helena Pivonkova
- Wellcome-Medical Research Council Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Kimberley A Evans
- Wellcome-Medical Research Council Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Ragnhildur T Káradóttir
- Wellcome-Medical Research Council Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, UK.,Department of Physiology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
16
|
Ohashi K, Shibasaki K, Nakazawa H, Kunimasa R, Nagayasu K, Shirakawa H, Kaneko S. Transient Receptor Potential Melastatin 3 Is Functionally Expressed in Oligodendrocyte Precursor Cells and Is Upregulated in Ischemic Demyelinated Lesions. Biol Pharm Bull 2021; 44:181-187. [PMID: 33518671 DOI: 10.1248/bpb.b20-00510] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) are glial cells that differentiate into oligodendrocytes and myelinate axons. The number of OPCs is reportedly increased in brain lesions in some demyelinating diseases and during ischemia; however, these cells also secrete cytokines and elicit both protective and deleterious effects in response to brain injury. The mechanism regulating the behaviors of OPCs in physiological and pathological conditions must be elucidated to control these cells and to treat demyelinating diseases. Here, we focused on transient receptor potential melastatin 3 (TRPM3), a Ca2+-permeable channel that is activated by the neurosteroid pregnenolone sulfate (PS) and body temperature. Trpm3+/Pdgfra+ OPCs were detected in the cerebral cortex (CTX) and corpus callosum (CC) of P4 and adult rats by in situ hybridization. Trpm3 expression was detected in primary cultured rat OPCs and was increased by treatment with tumor necrosis factor α (TNFα). Application of PS (30-100 µM) increased the Ca2+ concentration in OPCs and this effect was inhibited by co-treatment with the TRP channel blocker Gd3+ (100 µM) or the TRPM3 inhibitor isosakuranetin (10 µM). Stimulation of TRPM3 with PS (50 µM) did not affect the differentiation or migration of OPCs. The number of Trpm3+ OPCs was markedly increased in demyelinated lesions in an endothelin-1 (ET-1)-induced ischemic rat model. In conclusion, TRPM3 is functionally expressed in OPCs in vivo and in vitro and is upregulated in inflammatory conditions such as ischemic insults and TNFα treatment, implying that TRPM3 is involved in the regulation of specific behaviors of OPCs in pathological conditions.
Collapse
Affiliation(s)
- Kana Ohashi
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Koji Shibasaki
- Division of Neurochemistry, Graduate School of Human Health Science, University of Nagasaki
| | - Hayaki Nakazawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Ryotaro Kunimasa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Kazuki Nagayasu
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Hisashi Shirakawa
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| | - Shuji Kaneko
- Department of Molecular Pharmacology, Graduate School of Pharmaceutical Sciences, Kyoto University
| |
Collapse
|
17
|
Huang Y, Song YJ, Isaac M, Miretzky S, Patel A, Geoffrey McAuliffe W, Dreyfus CF. Tropomyosin Receptor Kinase B Expressed in Oligodendrocyte Lineage Cells Functions to Promote Myelin Following a Demyelinating Lesion. ASN Neuro 2020; 12:1759091420957464. [PMID: 32927995 PMCID: PMC7495938 DOI: 10.1177/1759091420957464] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The levels of brain-derived neurotrophic factor (BDNF) in the corpus callosum have previously been shown to have a critical impact on oligodendrocyte (OLG) lineage cells during cuprizone-elicited demyelination. In particular, BDNF+/- mice exhibit greater losses in myelin protein levels compared to wild-type mice after cuprizone. To investigate whether OLGs may directly mediate these effects of BDNF during a lesion in vivo, we used the cuprizone model of demyelination with inducible conditional male knockout mice to specifically delete the high-affinity tropomyosin receptor kinase B (TrkB) receptor from proteolipid protein + OLGs during cuprizone-elicited demyelination and subsequent remyelination. The loss of TrkB during cuprizone-elicited demyelination results in an increased sensitivity to demyelination as demonstrated by greater deficits in myelin protein levels, greater decreases in numbers of mature OLGs, increased numbers of demyelinated axons, and decreased myelin thickness. When mice are removed from cuprizone, they exhibit a delayed recovery in myelin proteins and myelin. Our data indicate that following a demyelinating lesion, TrkB in OLGs positively regulates myelin protein expression, myelin itself, and remyelination.
Collapse
Affiliation(s)
- Yangyang Huang
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, United States
| | - Yeri J. Song
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, United States
| | - Maria Isaac
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, United States
| | - Shir Miretzky
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, United States
| | - Ashish Patel
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, United States
| | - W. Geoffrey McAuliffe
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, United States
| | - Cheryl F. Dreyfus
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, New Jersey, United States,Cheryl F. Dreyfus, Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, 683 Hoes Lane West, Piscataway, NJ 08854, United States.
| |
Collapse
|
18
|
Sobel RA, Eaton MJ, Jaju PD, Lowry E, Hinojoza JR. Anti-Myelin Proteolipid Protein Peptide Monoclonal Antibodies Recognize Cell Surface Proteins on Developing Neurons and Inhibit Their Differentiation. J Neuropathol Exp Neurol 2020; 78:819-843. [PMID: 31400116 PMCID: PMC6703999 DOI: 10.1093/jnen/nlz058] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 04/30/2019] [Accepted: 06/18/2019] [Indexed: 12/15/2022] Open
Abstract
Using a panel of monoclonal antibodies (mAbs) to myelin proteolipid protein (PLP) peptides, we found that in addition to CNS myelin, mAbs to external face but not cytoplasmic face epitopes immunostained neurons in immature human CNS tissues and in adult hippocampal dentate gyrus and olfactory bulbs, that is neural stem cell niches (NSCN). To explore the pathobiological significance of these observations, we assessed the mAb effects on neurodifferentiation in vitro. The mAbs to PLP 50-69 (IgG1κ and IgG2aκ), and 178-191 and 200-219 (both IgG1κ) immunostained live cell surfaces and inhibited neurite outgrowth of E18 rat hippocampal precursor cells and of PC12 cells, which do not express PLP. Proteins immunoprecipitated from PC12 cell extracts and captured by mAb-coated magnetic beads were identified by GeLC-MS/MS. Each neurite outgrowth-inhibiting mAb captured a distinct set of neurodifferentiation molecules including sequence-similar M6 proteins and other unrelated membrane and extracellular matrix proteins, for example integrins, Eph receptors, NCAM-1, and protocadherins. These molecules are expressed in adult human NSCN and are implicated in the pathogenesis of many chronic CNS disease processes. Thus, diverse anti-PLP epitope autoantibodies may inhibit neuronal precursor cell differentiation via multispecific recognition of cell surface molecules thereby potentially impeding endogenous neuroregeneration in NSCN and in vivo differentiation of exogenous neural stem cells.
Collapse
Affiliation(s)
- Raymond A Sobel
- Laboratory Service, Veterans Affairs Health Care System, Palo Alto, California.,Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Mary Jane Eaton
- Laboratory Service, Veterans Affairs Health Care System, Palo Alto, California.,Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Prajakta Dilip Jaju
- Laboratory Service, Veterans Affairs Health Care System, Palo Alto, California.,Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Eugene Lowry
- Laboratory Service, Veterans Affairs Health Care System, Palo Alto, California.,Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Julian R Hinojoza
- Laboratory Service, Veterans Affairs Health Care System, Palo Alto, California.,Department of Pathology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
19
|
Li S, Oh BC, Chu C, Arnold A, Jablonska A, Furtmüller GJ, Qin HM, Boltze J, Magnus T, Ludewig P, Janowski M, Brandacher G, Walczak P. Induction of immunological tolerance to myelinogenic glial-restricted progenitor allografts. Brain 2020; 142:3456-3472. [PMID: 31529023 DOI: 10.1093/brain/awz275] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2018] [Revised: 06/22/2019] [Accepted: 07/11/2019] [Indexed: 12/11/2022] Open
Abstract
The immunological barrier currently precludes the clinical utilization of allogeneic stem cells. Although glial-restricted progenitors have become attractive candidates to treat a wide variety of neurological diseases, their survival in immunocompetent recipients is limited. In this study, we adopted a short-term, systemically applicable co-stimulation blockade-based strategy using CTLA4-Ig and anti-CD154 antibodies to modulate T-cell activation in the context of allogeneic glial-restricted progenitor transplantation. We found that co-stimulation blockade successfully prevented rejection of allogeneic glial-restricted progenitors from immunocompetent mouse brains. The long-term engrafted glial-restricted progenitors myelinated dysmyelinated adult mouse brains within one month. Furthermore, we identified a set of plasma miRNAs whose levels specifically correlated to the dynamic changes of immunoreactivity and as such could serve as biomarkers for graft rejection or tolerance. We put forward a successful strategy to induce alloantigen-specific hyporesponsiveness towards stem cells in the CNS, which will foster effective therapeutic application of allogeneic stem cells.
Collapse
Affiliation(s)
- Shen Li
- Neurology Department, Dalian Municipal Central Hospital affiliated to Dalian Medical University, Dalian, China.,Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Byoung Chol Oh
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Chengyan Chu
- Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Antje Arnold
- Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Anna Jablonska
- Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Georg J Furtmüller
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Hua-Min Qin
- Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Johannes Boltze
- School of Life Sciences, University of Warwick, Coventry, UK
| | - Tim Magnus
- Neurology Department, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Peter Ludewig
- Neurology Department, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Mirosław Janowski
- Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Gerald Brandacher
- Department of Plastic and Reconstructive Surgery, Vascularized Composite Allotransplantation (VCA) Laboratory, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Piotr Walczak
- Division of MR Research, Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,Cellular Imaging Section and Vascular Biology Program, Institute for Cell Engineering, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
20
|
Abstract
Cells of the oligodendrocyte lineage express a wide range of Ca2+ channels and receptors that regulate oligodendrocyte progenitor cell (OPC) and oligodendrocyte formation and function. Here we define those key channels and receptors that regulate Ca2+ signaling and OPC development and myelination. We then discuss how the regulation of intracellular Ca2+ in turn affects OPC and oligodendrocyte biology in the healthy nervous system and under pathological conditions. Activation of Ca2+ channels and receptors in OPCs and oligodendrocytes by neurotransmitters converges on regulating intracellular Ca2+, making Ca2+ signaling a central candidate mediator of activity-driven myelination. Indeed, recent evidence indicates that localized changes in Ca2+ in oligodendrocytes can regulate the formation and remodeling of myelin sheaths and perhaps additional functions of oligodendrocytes and OPCs. Thus, decoding how OPCs and myelinating oligodendrocytes integrate and process Ca2+ signals will be important to fully understand central nervous system formation, health, and function.
Collapse
Affiliation(s)
- Pablo M Paez
- Department of Pharmacology and Toxicology and Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, The State University of New York, University at Buffalo, Buffalo, New York 14203, USA;
| | - David A Lyons
- Centre for Discovery Brain Sciences, Centre for Multiple Sclerosis Research, and Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom;
| |
Collapse
|
21
|
Chen TJ, Kula B, Nagy B, Barzan R, Gall A, Ehrlich I, Kukley M. In Vivo Regulation of Oligodendrocyte Precursor Cell Proliferation and Differentiation by the AMPA-Receptor Subunit GluA2. Cell Rep 2019; 25:852-861.e7. [PMID: 30355492 DOI: 10.1016/j.celrep.2018.09.066] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 08/09/2018] [Accepted: 09/19/2018] [Indexed: 01/01/2023] Open
Abstract
The functional role of AMPA receptor (AMPAR)-mediated synaptic signaling between neurons and oligodendrocyte precursor cells (OPCs) remains enigmatic. We modified the properties of AMPARs at axon-OPC synapses in the mouse corpus callosum in vivo during the peak of myelination by targeting the GluA2 subunit. Expression of the unedited (Ca2+ permeable) or the pore-dead GluA2 subunit of AMPARs triggered proliferation of OPCs and reduced their differentiation into oligodendrocytes. Expression of the cytoplasmic C-terminal (GluA2(813-862)) of the GluA2 subunit (C-tail), a modification designed to affect the interaction between GluA2 and AMPAR-binding proteins and to perturb trafficking of GluA2-containing AMPARs, decreased the differentiation of OPCs without affecting their proliferation. These findings suggest that ionotropic and non-ionotropic properties of AMPARs in OPCs, as well as specific aspects of AMPAR-mediated signaling at axon-OPC synapses in the mouse corpus callosum, are important for balancing the response of OPCs to proliferation and differentiation cues.
Collapse
Affiliation(s)
- Ting-Jiun Chen
- Group of Neuron Glia Interaction, Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Graduate Training Centre of Neuroscience, University of Tübingen, 72074 Tübingen, Germany
| | - Bartosz Kula
- Group of Neuron Glia Interaction, Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Graduate Training Centre of Neuroscience, University of Tübingen, 72074 Tübingen, Germany
| | - Bálint Nagy
- Group of Neuron Glia Interaction, Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Graduate Training Centre of Neuroscience, University of Tübingen, 72074 Tübingen, Germany; Institute of Science and Technology (IST) Austria, 3400 Klosterneuburg, Austria
| | - Ruxandra Barzan
- Group of Neuron Glia Interaction, Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Graduate Training Centre of Neuroscience, University of Tübingen, 72074 Tübingen, Germany
| | - Andrea Gall
- Learning and Memory Group, Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Learning and Memory Group, Hertie Institute for Brain Research, University of Tübingen, 72076 Tübingen, Germany; Department of Neurobiology, IBBS, University of Stuttgart, 70569 Stuttgart, Germany
| | - Ingrid Ehrlich
- Learning and Memory Group, Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Learning and Memory Group, Hertie Institute for Brain Research, University of Tübingen, 72076 Tübingen, Germany; Department of Neurobiology, IBBS, University of Stuttgart, 70569 Stuttgart, Germany
| | - Maria Kukley
- Group of Neuron Glia Interaction, Werner Reichardt Centre for Integrative Neuroscience, University of Tübingen, 72076 Tübingen, Germany; Department of Ophthalmology, Research Institute of Ophthalmology, Faculty of Medicine, University Hospital Tübingen, 72076 Tübingen, Germany.
| |
Collapse
|
22
|
Zhang M, Liu Y, Wu S, Zhao X. Ca 2+ Signaling in Oligodendrocyte Development. Cell Mol Neurobiol 2019; 39:1071-1080. [PMID: 31222426 DOI: 10.1007/s10571-019-00705-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 06/11/2019] [Indexed: 12/31/2022]
Abstract
Calcium signaling has essential roles in the development of the nervous system, from neural induction to the proliferation, migration, and differentiation of both neuronal and glia cells. The temporal and spatial dynamics of Ca2+ signals control the highly diverse yet specific transcriptional programs that establish the complex structures of the nervous system. Ca2+-signaling pathways are shaped by interactions among metabotropic signaling cascades, ion channels, intracellular Ca2+ stores, and a multitude of downstream effector proteins that activate specific genetic programs. Progress in the last decade has led to significant advances in our understanding of the functional architecture of Ca2+ signaling networks involved in oligodendrocyte development. In this review, we summarize the molecular and functional organizations of Ca2+-signaling networks during the differentiation of oligodendrocyte, especially its impact on myelin gene expression, proliferation, migration, and myelination. Importantly, the existence of multiple routes of Ca2+ influx opens the possibility that the activity of calcium channels can be manipulated pharmacologically to encourage oligodendrocyte maturation and remyelination after demyelinating episodes in the brain.
Collapse
Affiliation(s)
- Ming Zhang
- Department of Neurobiology, Collaborative Innovation Center for Brain Science and Shaanxi Key Laboratory of Brain Disorders, Fourth Military Medical University, Xi'an, 710032, China
| | - Yuming Liu
- Department of Neurobiology, Collaborative Innovation Center for Brain Science and Shaanxi Key Laboratory of Brain Disorders, Fourth Military Medical University, Xi'an, 710032, China
| | - Shengxi Wu
- Department of Neurobiology, Collaborative Innovation Center for Brain Science and Shaanxi Key Laboratory of Brain Disorders, Fourth Military Medical University, Xi'an, 710032, China.
| | - Xianghui Zhao
- Department of Neurobiology, Collaborative Innovation Center for Brain Science and Shaanxi Key Laboratory of Brain Disorders, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
23
|
López-Valencia D, Medina-Ortega Á, Hoyos-Samboní DF, Saavedra-Torres JS, Salguero C. Epstein-Barr virus infection as a predisposing factor for multiple sclerosis. An update from molecular biology, immunology and epidemiology. REVISTA DE LA FACULTAD DE MEDICINA 2019. [DOI: 10.15446/revfacmed.v67n3.70149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Introduction: Epstein-Barr virus is an infectious agent used to immortalize and induce polyclonal activation of B cells. It has been widely described that this virus produces changes in the cells it infects and in the immune response, and stimulates the development of autoimmune diseases.Objective: To characterize the association between Epstein-Barr virus and multiple sclerosis described in current scientific literature.Materials and methods: A 59-years range literature search was conducted in the PubMed, ScienceDirect, Redalyc and SciELO databases using the following MeSH terms: “Epstein-Barr virus, multiple sclerosis autoimmune diseases, autoimmune diseases of the nervous system”.Results: Many studies describe the association between Epstein-Barr virus and multiple sclerosis. It is believed that acute infection and viral reactivation promote the development of multiple sclerosis.Conclusions: It is necessary to conduct further research on the pathogenesis and morphophysiological and neuroimmunological changes –at the ecological, molecular, cellular, tissue, organic and systemic level– induced by the immune response and that favor the development of multiple sclerosis.
Collapse
|
24
|
Ceprian M, Fulton D. Glial Cell AMPA Receptors in Nervous System Health, Injury and Disease. Int J Mol Sci 2019; 20:E2450. [PMID: 31108947 PMCID: PMC6566241 DOI: 10.3390/ijms20102450] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 04/11/2019] [Accepted: 04/22/2019] [Indexed: 12/16/2022] Open
Abstract
Glia form a central component of the nervous system whose varied activities sustain an environment that is optimised for healthy development and neuronal function. Alpha-amino-3-hydroxy-5-methyl-4-isoxazole (AMPA)-type glutamate receptors (AMPAR) are a central mediator of glutamatergic excitatory synaptic transmission, yet they are also expressed in a wide range of glial cells where they influence a variety of important cellular functions. AMPAR enable glial cells to sense the activity of neighbouring axons and synapses, and as such many aspects of glial cell development and function are influenced by the activity of neural circuits. However, these AMPAR also render glia sensitive to elevations of the extracellular concentration of glutamate, which are associated with a broad range of pathological conditions. Excessive activation of AMPAR under these conditions may induce excitotoxic injury in glial cells, and trigger pathophysiological responses threatening other neural cells and amplifying ongoing disease processes. The aim of this review is to gather information on AMPAR function from across the broad diversity of glial cells, identify their contribution to pathophysiological processes, and highlight new areas of research whose progress may increase our understanding of nervous system dysfunction and disease.
Collapse
Affiliation(s)
- Maria Ceprian
- Instituto de Investigación Sanitaria San Carlos (IdISSC), 28040 Madrid, Spain.
- Departamento de Bioquímica y Biología Molecular, CIBERNED, IRICYS. Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain.
| | - Daniel Fulton
- Neuroscience and Ophthalmology Research Group, Institute of Inflammation and Ageing, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK.
| |
Collapse
|
25
|
Habermacher C, Angulo MC, Benamer N. Glutamate versus GABA in neuron-oligodendroglia communication. Glia 2019; 67:2092-2106. [PMID: 30957306 DOI: 10.1002/glia.23618] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/28/2019] [Accepted: 03/19/2019] [Indexed: 12/20/2022]
Abstract
In the central nervous system (CNS), myelin sheaths around axons are formed by glial cells named oligodendrocytes (OLs). In turn, OLs are generated by oligodendrocyte precursor cells (OPCs) during postnatal development and in adults, according to a process that depends on the proliferation and differentiation of these progenitors. The maturation of OL lineage cells as well as myelination by OLs are complex and highly regulated processes in the CNS. OPCs and OLs express an array of receptors for neurotransmitters, in particular for the two main CNS neurotransmitters glutamate and GABA, and are therefore endowed with the capacity to respond to neuronal activity. Initial studies in cell cultures demonstrated that both glutamate and GABA signaling mechanisms play important roles in OL lineage cell development and function. However, much remains to be learned about the communication of glutamatergic and GABAergic neurons with oligodendroglia in vivo. This review focuses on recent major advances in our understanding of the neuron-oligodendroglia communication mediated by glutamate and GABA in the CNS, and highlights the present controversies in the field. We discuss the expression, activation modes and potential roles of synaptic and extrasynaptic receptors along OL lineage progression. We review the properties of OPC synaptic connectivity with presynaptic glutamatergic and GABAergic neurons in the brain and consider the implication of glutamate and GABA signaling in activity-driven adaptive myelination.
Collapse
Affiliation(s)
- Chloé Habermacher
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France.,Université Paris Descartes, Paris, France
| | - María C Angulo
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France.,Université Paris Descartes, Paris, France
| | - Najate Benamer
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France.,Université Paris Descartes, Paris, France
| |
Collapse
|
26
|
Kula B, Chen T, Kukley M. Glutamatergic signaling between neurons and oligodendrocyte lineage cells: Is it synaptic or non‐synaptic? Glia 2019; 67:2071-2091. [DOI: 10.1002/glia.23617] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/12/2019] [Accepted: 03/18/2019] [Indexed: 12/18/2022]
Affiliation(s)
- Bartosz Kula
- Group of Neuron Glia InteractionUniversity of Tübingen Tübingen Germany
- Graduate Training Centre for NeuroscienceUniversity of Tübingen Tübingen Germany
| | - Ting‐Jiun Chen
- Center for Neuroscience ResearchChildren's Research Institute, Children's National Medical Center Washington District of Columbia
| | - Maria Kukley
- Group of Neuron Glia InteractionUniversity of Tübingen Tübingen Germany
- Research Institute for OphthalmologyUniversity Hospital Tübingen Tübingen Germany
| |
Collapse
|
27
|
Colwell CS, Ghiani CA. Potential Circadian Rhythms in Oligodendrocytes? Working Together Through Time. Neurochem Res 2019; 45:591-605. [PMID: 30906970 DOI: 10.1007/s11064-019-02778-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 03/14/2019] [Indexed: 12/15/2022]
Abstract
Oligodendrocytes (OL) are the only myelinating cells of the central nervous system thus interferences, either environmental or genetic, with their maturation or function have devastating consequences. Albeit so far neglected, one of the less appreciated, nevertheless possible, regulators of OL maturation and function is the circadian cycle. Yet, disruptions in these rhythms are unfortunately becoming a common "disorder" in the today's world. The temporal patterning of behaviour and physiology is controlled by a circadian timing system based in the anterior hypothalamus. At the molecular level, circadian rhythms are generated by a transcriptional/translational feedback system that regulates transcription and has a major impact on cellular function(s). Fundamental cellular properties/functions in most cell types vary with the daily circadian cycle: OL are unlikely an exception! To be clear, the presence of circadian oscillators or the cell-specific function(s) of the circadian clock in OL has yet to be defined. Furthermore, we wish to entertain the idea of links between the "thin" evidence on OL intrinsic circadian rhythms and their interjection(s) at different stages of lineage progression as well as in supporting/regulating OL crucial function: myelination. Individuals with intellectual and developmental syndromes as well as neurodegenerative diseases present with a disrupted sleep/wake cycle; hence, we raise the possibility that these disturbances in timing can contribute to the loss of white matter observed in these disorders. Preclinical and clinical work in this area is needed for a better understanding of how circadian rhythms influence OL maturation and function(s), to aid the development of new therapeutic strategies and standards of care for these patients.
Collapse
Affiliation(s)
- Christopher S Colwell
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.,Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Cristina A Ghiani
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA. .,Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA. .,Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
28
|
Bruce KD, Gorkhali S, Given K, Coates AM, Boyle KE, Macklin WB, Eckel RH. Lipoprotein Lipase Is a Feature of Alternatively-Activated Microglia and May Facilitate Lipid Uptake in the CNS During Demyelination. Front Mol Neurosci 2018; 11:57. [PMID: 29599706 PMCID: PMC5862862 DOI: 10.3389/fnmol.2018.00057] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 02/12/2018] [Indexed: 11/18/2022] Open
Abstract
Severe demyelinating disorders of the central nervous system (CNS) such as multiple sclerosis (MS), can be devastating for many young lives. To date, the factors resulting in poor remyelination and repair are not well understood, and reparative therapies that benefit MS patients have yet to be developed. We have previously shown that the activity and abundance of Lipoprotein Lipase (LPL)—the rate-limiting enzyme in the hydrolysis of triglyceride-rich lipoproteins—is increased in Schwann cells and macrophages following nerve crush injury in the peripheral nervous system (PNS), suggesting that LPL may help scavenge myelin-derived lipids. We hypothesized that LPL may play a similar role in the CNS. To test this, mice were immunized with MOG35–55 peptide to induce experimental allergic encephalomyelitis (EAE). LPL activity was increased (p < 0.05) in the brain at 30 days post-injection, coinciding with partial remission of clinical symptoms. Furthermore, LPL abundance and activity was up-regulated (p < 0.05) at the transition between de- and re-myelination in lysolecithin-treated ex vivo cerebellar slices. Since microglia are the key immune effector cells of the CNS we determined the role of LPL in microglia. Lipid uptake was decreased (p < 0.001) in LPL-deficient BV-2 microglial cells compared to WT. In addition, LPL-deficient cells showed dramatically reduced expression of anti-inflammatory markers, YM1 (−22 fold, p < 0.001), and arginase 1 (Arg1; −265 fold, p < 0.001) and increased expression of pro-inflammatory markers, such as iNOS compared to WT cells (+53 fold, p < 0.001). This suggests that LPL is a feature of reparative microglia, further supported by the metabolic and inflammatory profile of LPL-deficient microglia. Taken together, our data strongly suggest that LPL expression is a novel feature of a microglial phenotype that supports remyelination and repair through the clearance of lipid debris. This mechanism may be exploited to develop future reparative therapies for MS and primary neurodegenerative disorders (Alzheimer’s disease (AD) and Parkinson’s disease).
Collapse
Affiliation(s)
- Kimberley D Bruce
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Sachi Gorkhali
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, United States
| | - Katherine Given
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Alison M Coates
- School of Health Sciences, Sansom Institute for Health Research, University of South Australia, Adelaide, SA, Australia
| | - Kristen E Boyle
- Department of Pediatrics, Section of Nutrition, University of Colorado School of Medicine, Aurora, CO, United States
| | - Wendy B Macklin
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO, United States
| | - Robert H Eckel
- Division of Endocrinology, Metabolism, & Diabetes, Department of Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
29
|
Song XJ, Han W, He R, Li TY, Xie LL, Cheng L, Chen HS, Jiang L. Alterations of Hippocampal Myelin Sheath and Axon Sprouting by Status Convulsion and Regulating Lingo-1 Expression with RNA Interference in Immature and Adult Rats. Neurochem Res 2018; 43:721-735. [PMID: 29383653 DOI: 10.1007/s11064-018-2474-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 12/02/2017] [Accepted: 01/11/2018] [Indexed: 11/24/2022]
Abstract
Seizure-induced brain damage is age-dependent, as evidenced by the different alterations of neural physiopathology in developing and mature brains. However, little is known about the age-dependent characteristics of myelinated fiber injury induced by seizures. Considering the critical functions of oligodendrocyte progenitor cells (OPCs) in myelination and Lingo-1 signaling in regulating OPCs' differentiation, the present study aimed to explore the effects of Lingo-1 on myelin and axon in immature and adult rats after status convulsion (SC) induced by lithium-pilocarpine, and the differences between immature and adult brains. Dynamic variations in electrophysiological activity and spontaneous recurrent seizures were recorded by electroencephalogram monitoring after SC. The impaired microstructures of myelin sheaths and decrease in myelin basic protein caused by SC were observed through transmission electron microscopy and western blot analysis respectively, which became more severe in adult rats, but improved gradually in immature rats. Aberrant axon sprouting occurred in adult rats, which was more prominent than in immature rats, as shown by a Timm stain. This damage was improved or negatively affected after down or upregulating Lingo-1 expression. These results demonstrated that in both immature and adult brains, Lingo-1 signaling plays important roles in seizure-induced damage to myelin sheaths and axon growth. The plasticity of the developing brain may provide a potential window of opportunity to prevent the brain from damage.
Collapse
Affiliation(s)
- Xiao-Jie Song
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Wei Han
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Rong He
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China.,Department of Neurology, Children's Hospital of Chongqing Medical University, No.136 Zhongshan 2nd Road, Chongqing, 400014, China
| | - Tian-Yi Li
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China.,Department of Neurology, Children's Hospital of Chongqing Medical University, No.136 Zhongshan 2nd Road, Chongqing, 400014, China
| | - Ling-Ling Xie
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China.,Department of Neurology, Children's Hospital of Chongqing Medical University, No.136 Zhongshan 2nd Road, Chongqing, 400014, China
| | - Li Cheng
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Heng-Sheng Chen
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China
| | - Li Jiang
- Pediatric Research Institute, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China. .,Department of Neurology, Children's Hospital of Chongqing Medical University, No.136 Zhongshan 2nd Road, Chongqing, 400014, China.
| |
Collapse
|
30
|
Lecca D, Fumagalli M, Ceruti S, Abbracchio MP. Intertwining extracellular nucleotides and their receptors with Ca2+ in determining adult neural stem cell survival, proliferation and final fate. Philos Trans R Soc Lond B Biol Sci 2017; 371:rstb.2015.0433. [PMID: 27377726 DOI: 10.1098/rstb.2015.0433] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2016] [Indexed: 02/07/2023] Open
Abstract
In the central nervous system (CNS), during both brain and spinal cord development, purinergic and pyrimidinergic signalling molecules (ATP, UTP and adenosine) act synergistically with peptidic growth factors in regulating the synchronized proliferation and final specification of multipotent neural stem cells (NSCs) to neurons, astrocytes or oligodendrocytes, the myelin-forming cells. Some NSCs still persist throughout adulthood in both specific 'neurogenic' areas and in brain and spinal cord parenchyma, retaining the potentiality to generate all the three main types of adult CNS cells. Once CNS anatomical structures are defined, purinergic molecules participate in calcium-dependent neuron-to-glia communication and also control the behaviour of adult NSCs. After development, some purinergic mechanisms are silenced, but can be resumed after injury, suggesting a role for purinergic signalling in regeneration and self-repair also via the reactivation of adult NSCs. In this respect, at least three different types of adult NSCs participate in the response of the adult brain and spinal cord to insults: stem-like cells residing in classical neurogenic niches, in particular, in the ventricular-subventricular zone (V-SVZ), parenchymal oligodendrocyte precursor cells (OPCs, also known as NG2-glia) and parenchymal injury-activated astrocytes (reactive astrocytes). Here, we shall review and discuss the purinergic regulation of these three main adult NSCs, with particular focus on how and to what extent modulation of intracellular calcium levels by purinoceptors is mandatory to determine their survival, proliferation and final fate.This article is part of the themed issue 'Evolution brings Ca(2+) and ATP together to control life and death'.
Collapse
Affiliation(s)
- Davide Lecca
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy
| | - Marta Fumagalli
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy
| | - Stefania Ceruti
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy
| | - Maria P Abbracchio
- Laboratory of Molecular and Cellular Pharmacology of Purinergic Transmission, Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, 20133 Milan, Italy
| |
Collapse
|
31
|
Newville J, Jantzie LL, Cunningham LA. Embracing oligodendrocyte diversity in the context of perinatal injury. Neural Regen Res 2017; 12:1575-1585. [PMID: 29171412 PMCID: PMC5696828 DOI: 10.4103/1673-5374.217320] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2017] [Indexed: 12/18/2022] Open
Abstract
Emerging evidence is fueling a new appreciation of oligodendrocyte diversity that is overturning the traditional view that oligodendrocytes are a homogenous cell population. Oligodendrocytes of distinct origins, maturational stages, and regional locations may differ in their functional capacity or susceptibility to injury. One of the most unique qualities of the oligodendrocyte is its ability to produce myelin. Myelin abnormalities have been ascribed to a remarkable array of perinatal brain injuries, with concomitant oligodendrocyte dysregulation. Within this review, we discuss new insights into the diversity of the oligodendrocyte lineage and highlight their relevance in paradigms of perinatal brain injury. Future therapeutic development will be informed by comprehensive knowledge of oligodendrocyte pathophysiology that considers the particular facets of heterogeneity that this lineage exhibits.
Collapse
Affiliation(s)
- Jessie Newville
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Lauren L. Jantzie
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
- Department of Pediatrics, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| | - Lee Anna Cunningham
- Department of Neurosciences, University of New Mexico Health Sciences Center, Albuquerque, NM, USA
| |
Collapse
|
32
|
Lüders KA, Patzig J, Simons M, Nave KA, Werner HB. Genetic dissection of oligodendroglial and neuronalPlp1function in a novel mouse model of spastic paraplegia type 2. Glia 2017; 65:1762-1776. [DOI: 10.1002/glia.23193] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 06/28/2017] [Accepted: 06/29/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Katja A. Lüders
- Department of Neurogenetics; Max Planck Institute of Experimental Medicine; Göttingen 37075 Germany
| | - Julia Patzig
- Department of Neurogenetics; Max Planck Institute of Experimental Medicine; Göttingen 37075 Germany
| | - Mikael Simons
- Cellular Neuroscience; Max Planck Institute of Experimental Medicine; Göttingen 37075 Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics; Max Planck Institute of Experimental Medicine; Göttingen 37075 Germany
| | - Hauke B. Werner
- Department of Neurogenetics; Max Planck Institute of Experimental Medicine; Göttingen 37075 Germany
| |
Collapse
|
33
|
Bu W, Zhao WQ, Li WL, Dong CZ, Zhang Z, Li QJ. Neuropeptide Y suppresses epileptiform discharges by regulating AMPA receptor GluR2 subunit in rat hippocampal neurons. Mol Med Rep 2017; 16:387-395. [DOI: 10.3892/mmr.2017.6567] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 02/09/2017] [Indexed: 11/06/2022] Open
|
34
|
Nevin ZS, Factor DC, Karl RT, Douvaras P, Laukka J, Windrem MS, Goldman SA, Fossati V, Hobson GM, Tesar PJ. Modeling the Mutational and Phenotypic Landscapes of Pelizaeus-Merzbacher Disease with Human iPSC-Derived Oligodendrocytes. Am J Hum Genet 2017; 100:617-634. [PMID: 28366443 DOI: 10.1016/j.ajhg.2017.03.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 03/09/2017] [Indexed: 02/07/2023] Open
Abstract
Pelizaeus-Merzbacher disease (PMD) is a pediatric disease of myelin in the central nervous system and manifests with a wide spectrum of clinical severities. Although PMD is a rare monogenic disease, hundreds of mutations in the X-linked myelin gene proteolipid protein 1 (PLP1) have been identified in humans. Attempts to identify a common pathogenic process underlying PMD have been complicated by an incomplete understanding of PLP1 dysfunction and limited access to primary human oligodendrocytes. To address this, we generated panels of human induced pluripotent stem cells (hiPSCs) and hiPSC-derived oligodendrocytes from 12 individuals with mutations spanning the genetic and clinical diversity of PMD-including point mutations and duplication, triplication, and deletion of PLP1-and developed an in vitro platform for molecular and cellular characterization of all 12 mutations simultaneously. We identified individual and shared defects in PLP1 mRNA expression and splicing, oligodendrocyte progenitor development, and oligodendrocyte morphology and capacity for myelination. These observations enabled classification of PMD subgroups by cell-intrinsic phenotypes and identified a subset of mutations for targeted testing of small-molecule modulators of the endoplasmic reticulum stress response, which improved both morphologic and myelination defects. Collectively, these data provide insights into the pathogeneses of a variety of PLP1 mutations and suggest that disparate etiologies of PMD could require specific treatment approaches for subsets of individuals. More broadly, this study demonstrates the versatility of a hiPSC-based panel spanning the mutational heterogeneity within a single disease and establishes a widely applicable platform for genotype-phenotype correlation and drug screening in any human myelin disorder.
Collapse
Affiliation(s)
- Zachary S Nevin
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Daniel C Factor
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Robert T Karl
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | | | - Jeremy Laukka
- Departments of Neurology and Neuroscience, College of Medicine and Life Science, University of Toledo, Toledo, OH 43614, USA
| | - Martha S Windrem
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, Rochester, NY 14642, USA; Center for Neuroscience, Faculty of Medicine and Health Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Valentina Fossati
- New York Stem Cell Foundation Research Institute, New York, NY 10032, USA
| | - Grace M Hobson
- Nemours Biomedical Research, Alfred I. duPont Hospital for Children, Wilmington, DE 19803, USA; Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA; Department of Pediatrics, Jefferson Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Paul J Tesar
- Department of Genetics and Genome Sciences, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA.
| |
Collapse
|
35
|
A novel PLP1 mutation F240L identified in a patient with connatal type Pelizaeus-Merzbacher disease. Hum Genome Var 2017; 4:16044. [PMID: 28101371 PMCID: PMC5214593 DOI: 10.1038/hgv.2016.44] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/23/2016] [Accepted: 10/24/2016] [Indexed: 11/09/2022] Open
Abstract
Pelizaeus-Merzbacher disease (PMD) is an X-linked recessive hypomyelination disorder caused by mutations in the proteolipid protein 1 gene (PLP1) located on chromosome Xq22. A male patient showed severe developmental delay, pendular nystagmus and laryngeal wheezing. The auditory brain stem response showed only the first wave and brain magnetic resonance imaging showed white matter hypomyelination, suggesting typical PMD. A novel PLP1 mutation, F240L, which was inherited from his mother, was identified.
Collapse
|
36
|
Dulamea AO. Role of Oligodendrocyte Dysfunction in Demyelination, Remyelination and Neurodegeneration in Multiple Sclerosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 958:91-127. [PMID: 28093710 DOI: 10.1007/978-3-319-47861-6_7] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Oligodendrocytes (OLs) are the myelinating cells of the central nervous system (CNS) during development and throughout adulthood. They result from a complex and well controlled process of activation, proliferation, migration and differentiation of oligodendrocyte progenitor cells (OPCs) from the germinative niches of the CNS. In multiple sclerosis (MS), the complex pathological process produces dysfunction and apoptosis of OLs leading to demyelination and neurodegeneration. This review attempts to describe the patterns of demyelination in MS, the steps involved in oligodendrogenesis and myelination in healthy CNS, the different pathways leading to OLs and myelin loss in MS, as well as principles involved in restoration of myelin sheaths. Environmental factors and their impact on OLs and pathological mechanisms of MS are also discussed. Finally, we will present evidence about the potential therapeutic targets in re-myelination processes that can be accessed in order to develop regenerative therapies for MS.
Collapse
Affiliation(s)
- Adriana Octaviana Dulamea
- Neurology Clinic, University of Medicine and Pharmacy "Carol Davila", Fundeni Clinical Institute, Building A, Neurology Clinic, Room 201, 022328, Bucharest, Romania.
| |
Collapse
|
37
|
Liu Y, Harlow DE, Given KS, Owens GP, Macklin WB, Bennett JL. Variable sensitivity to complement-dependent cytotoxicity in murine models of neuromyelitis optica. J Neuroinflammation 2016; 13:301. [PMID: 27905992 PMCID: PMC5134246 DOI: 10.1186/s12974-016-0767-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 11/21/2016] [Indexed: 01/06/2023] Open
Abstract
Background Studies of neuromyelitis optica (NMO), an autoimmune disease of the central nervous system (CNS), have demonstrated that autoantibodies against the water channel aquaporin-4 (AQP4) induce astrocyte damage through complement-dependent cytotoxicity (CDC). In developing experimental models of NMO using cells, tissues or animals from mice, co-administration of AQP4-IgG and normal human serum, which serves as the source of human complement (HC), is required. The sensitivity of mouse CNS cells to HC and CDC in these models is not known. Methods We used HC and recombinant monoclonal antibodies (rAbs) against AQP4 to investigate CDC on mouse neurons, astrocytes, differentiated oligodendrocytes (OLs), and oligodendrocyte progenitors (OPCs) in the context of purified monocultures, neuroglial mixed cultures, and organotypic cerebellar slices. Results We found that murine neurons, OLs, and OPCs were sensitive to HC in monocultures. In mixed murine neuroglial cultures, HC-mediated toxicity to neurons and OLs was reduced; however, astrocyte damage induced by an AQP-specific rAb #53 and HC increased neuronal and oligodendroglial loss. OPCs were resistant to HC toxicity in neuroglial mixed cultures. In mouse cerebellar slices, damage to neurons and OLs following rAb #53-mediated CDC was further reduced, but in contrast to neuroglial mixed cultures, astrocyte damage sensitized OPCs to complement damage. Finally, we established that some injury to neurons, OLs, and OPCs in cell and slice cultures resulted from the activation of HC by anti-tissue antibodies to mouse cells. Conclusions Murine neurons and oligodendroglia demonstrate variable sensitivity to activated complement based on their differentiation and culture conditions. In organotypic cultures, the protection of neurons, OLs, and OPCs against CDC is eliminated by targeted astrocyte destruction. The activation of human complement proteins on mouse CNS cells necessitates caution when interpreting the results of mouse experimental models of NMO using HC. Electronic supplementary material The online version of this article (doi:10.1186/s12974-016-0767-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yiting Liu
- Department of Neurology, University of Colorado, School of Medicine, 12700 E. 19th Ave, Aurora, CO, USA
| | - Danielle E Harlow
- Department of Cell & Developmental Biology, University of Colorado, School of Medicine, 12700 E. 19th Ave, Aurora, CO, USA
| | - Katherine S Given
- Department of Cell & Developmental Biology, University of Colorado, School of Medicine, 12700 E. 19th Ave, Aurora, CO, USA
| | - Gregory P Owens
- Department of Neurology, University of Colorado, School of Medicine, 12700 E. 19th Ave, Aurora, CO, USA
| | - Wendy B Macklin
- Department of Cell & Developmental Biology, University of Colorado, School of Medicine, 12700 E. 19th Ave, Aurora, CO, USA.,Program in Neuroscience, University of Colorado, School of Medicine, 12700 E. 19th Ave, Aurora, CO, USA
| | - Jeffrey L Bennett
- Department of Neurology, University of Colorado, School of Medicine, 12700 E. 19th Ave, Aurora, CO, USA. .,Department of Ophthalmology, University of Colorado, School of Medicine, 12700 E. 19th Ave, Aurora, CO, USA. .,Program in Neuroscience, University of Colorado, School of Medicine, 12700 E. 19th Ave, Aurora, CO, USA.
| |
Collapse
|
38
|
Fernandez-Castaneda A, Gaultier A. Adult oligodendrocyte progenitor cells - Multifaceted regulators of the CNS in health and disease. Brain Behav Immun 2016; 57:1-7. [PMID: 26796621 PMCID: PMC4940337 DOI: 10.1016/j.bbi.2016.01.005] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 12/28/2015] [Accepted: 01/11/2016] [Indexed: 01/17/2023] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) are the often-overlooked fourth glial cell type in the central nervous system (CNS), comprising about 5% of the CNS. For a long time, our vision of OPC function was limited to the generation of mature oligodendrocytes. However, new studies have highlighted the multifaceted nature of OPCs. During homeostatic and pathological conditions, OPCs are the most proliferative cell type in the CNS, a property not consistent with the need to generate new oligodendrocytes. Indeed, OPCs modulate neuronal activity and OPC depletion in the brain can trigger depressive-like behavior. More importantly, OPCs are actively recruited to injury sites, where they orchestrate glial scar formation and contribute to the immune response. The following is a comprehensive analysis of the literature on OPC function beyond myelination, in the context of the healthy and diseased adult CNS.
Collapse
Affiliation(s)
- Anthony Fernandez-Castaneda
- Center for Brain Immunology and Glia, Department of Neuroscience, Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Alban Gaultier
- Center for Brain Immunology and Glia, Department of Neuroscience, Graduate Program in Neuroscience, School of Medicine, University of Virginia, Charlottesville, VA 22908, USA.
| |
Collapse
|
39
|
Wheeler NA, Fuss B. Extracellular cues influencing oligodendrocyte differentiation and (re)myelination. Exp Neurol 2016; 283:512-30. [PMID: 27016069 PMCID: PMC5010977 DOI: 10.1016/j.expneurol.2016.03.019] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/03/2016] [Accepted: 03/18/2016] [Indexed: 02/07/2023]
Abstract
There is an increasing number of neurologic disorders found to be associated with loss and/or dysfunction of the CNS myelin sheath, ranging from the classic demyelinating disease, multiple sclerosis, through CNS injury, to neuropsychiatric diseases. The disabling burden of these diseases has sparked a growing interest in gaining a better understanding of the molecular mechanisms regulating the differentiation of the myelinating cells of the CNS, oligodendrocytes (OLGs), and the process of (re)myelination. In this context, the importance of the extracellular milieu is becoming increasingly recognized. Under pathological conditions, changes in inhibitory as well as permissive/promotional cues are thought to lead to an overall extracellular environment that is obstructive for the regeneration of the myelin sheath. Given the general view that remyelination is, even though limited in human, a natural response to demyelination, targeting pathologically 'dysregulated' extracellular cues and their downstream pathways is regarded as a promising approach toward the enhancement of remyelination by endogenous (or if necessary transplanted) OLG progenitor cells. In this review, we will introduce the extracellular cues that have been implicated in the modulation of (re)myelination. These cues can be soluble, part of the extracellular matrix (ECM) or mediators of cell-cell interactions. Their inhibitory and permissive/promotional roles with regard to remyelination as well as their potential for therapeutic intervention will be discussed.
Collapse
Affiliation(s)
- Natalie A Wheeler
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, United States
| | - Babette Fuss
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, VA 23298, United States.
| |
Collapse
|
40
|
Glutamate signalling: A multifaceted modulator of oligodendrocyte lineage cells in health and disease. Neuropharmacology 2016; 110:574-585. [PMID: 27346208 DOI: 10.1016/j.neuropharm.2016.06.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 05/27/2016] [Accepted: 06/16/2016] [Indexed: 01/10/2023]
Abstract
Myelin is essential for the mammalian brain to function efficiently. Whilst many factors have been associated with regulating the differentiation of oligodendroglia and myelination, glutamate signalling might be particularly important for learning-dependent myelination. The majority of myelinated projection neurons are glutamatergic. Oligodendrocyte precursor cells receive glutamatergic synaptic inputs from unmyelinated axons and oligodendrocyte lineage cells express glutamate receptors which enable them to monitor and respond to changes in neuronal activity. Yet, what role glutamate plays for oligodendroglia is not fully understood. Here, we review glutamate signalling and its effects on oligodendrocyte lineage cells, and myelination in health and disease. Furthermore, we discuss whether glutamate signalling between neurons and oligodendroglia might lay the foundation to activity-dependent white matter plasticity. This article is part of the Special Issue entitled 'Oligodendrocytes in Health and Disease'.
Collapse
|
41
|
Pitman KA, Young KM. Activity-dependent calcium signalling in oligodendrocyte generation. Int J Biochem Cell Biol 2016; 77:30-34. [PMID: 27233230 DOI: 10.1016/j.biocel.2016.05.018] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 05/21/2016] [Accepted: 05/23/2016] [Indexed: 11/26/2022]
Abstract
Throughout postnatal life oligodendrocyte progenitor cells proliferate and differentiate into mature myelinating oligodendrocytes in the central nervous system. Neuronal activity is a major external signal controlling this process. Neurotransmitters, or other signalling molecules released in response to neuronal activity, evoke transient increases in intracellular calcium in oligodendrocyte progenitor cells. As calcium can mediate cellular processes, including the transcription of genes involved in oligodendrocyte progenitor cell division and maturation, a rise in intracellular calcium may be a key signal translating changes in neuronal activity into changes in oligodendrocyte progenitor cell behaviour. Here we review recent advances in our understanding of how neuronal activity can evoke calcium signalling in oligodendrocyte progenitor cells.
Collapse
Affiliation(s)
- Kimberley A Pitman
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania 7000, Australia.
| |
Collapse
|