1
|
Ding X, Wu Y, Vainshtein A, Rodriguez V, Ricco E, Okoh JT, Liu Y, Kraushaar DC, Peles E, Rasband MN. Age-dependent regulation of axoglial interactions and behavior by oligodendrocyte AnkyrinG. Nat Commun 2024; 15:10865. [PMID: 39738113 DOI: 10.1038/s41467-024-55209-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 12/05/2024] [Indexed: 01/01/2025] Open
Abstract
The bipolar disorder (BD) risk gene ANK3 encodes the scaffolding protein AnkyrinG (AnkG). In neurons, AnkG regulates polarity and ion channel clustering at axon initial segments and nodes of Ranvier. Disruption of neuronal AnkG causes BD-like phenotypes in mice. During development, AnkG is also expressed at comparable levels in oligodendrocytes and facilitates the efficient assembly of paranodal junctions. However, the physiological roles of glial AnkG in the mature nervous system, and its contributions to BD-like phenotypes, remain unexplored. Here, we show that oligodendroglia-specific AnkG conditional knockout results in destabilization of axoglial interactions in aged but not young adult mice. In addition, these mice exhibit significant histological, electrophysiological, and behavioral pathophysiologies. Unbiased translatomic profiling reveals potential compensatory machineries. These results highlight the functions of glial AnkG in maintaining proper axoglial interactions throughout aging and suggest a contribution of glial AnkG to neuropsychiatric disorders.
Collapse
Affiliation(s)
- Xiaoyun Ding
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yu Wu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Anna Vainshtein
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Victoria Rodriguez
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Emily Ricco
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, TX, 77030, USA
| | - James T Okoh
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yanhong Liu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Daniel C Kraushaar
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Elior Peles
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, 77030, USA.
| |
Collapse
|
2
|
Marshall-Phelps KL, Almeida R. Axonal neurotransmitter release in the regulation of myelination. Biosci Rep 2024; 44:BSR20231616. [PMID: 39230890 PMCID: PMC11427734 DOI: 10.1042/bsr20231616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/05/2024] Open
Abstract
Myelination of axons is a key determinant of fast action potential propagation, axonal health and circuit function. Previously considered a static structure, it is now clear that myelin is dynamically regulated in response to neuronal activity in the central nervous system (CNS). However, how activity-dependent signals are conveyed to oligodendrocytes remains unclear. Here, we review the potential mechanisms by which neurons could communicate changing activity levels to myelin, with a focus on the accumulating body of evidence to support activity-dependent vesicular signalling directly onto myelin sheaths. We discuss recent in vivo findings of activity-dependent fusion of neurotransmitter vesicles from non-synaptic axonal sites, and how modulation of this vesicular fusion regulates the stability and growth of myelin sheaths. We also consider the potential mechanisms by which myelin could sense and respond to axon-derived signals to initiate remodelling, and the relevance of these adaptations for circuit function. We propose that axonal vesicular signalling represents an important and underappreciated mode of communication by which neurons can transmit activity-regulated signals to myelinating oligodendrocytes and, potentially, more broadly to other cell types in the CNS.
Collapse
Affiliation(s)
- Katy L.H. Marshall-Phelps
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, U.K
- MS Society Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, U.K
| | - Rafael G. Almeida
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, U.K
- MS Society Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, U.K
| |
Collapse
|
3
|
Eltokhi A, Lundstrom BN, Li J, Zweifel LS, Catterall WA, Gamal El-Din TM. Pathogenic gating pore current conducted by autism-related mutations in the Na V1.2 brain sodium channel. Proc Natl Acad Sci U S A 2024; 121:e2317769121. [PMID: 38564633 PMCID: PMC11009634 DOI: 10.1073/pnas.2317769121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 02/26/2024] [Indexed: 04/04/2024] Open
Abstract
Autism spectrum disorder (ASD) is a complex neurodevelopmental condition characterized by social and communication deficits and repetitive behaviors. The genetic heterogeneity of ASD presents a challenge to the development of an effective treatment targeting the underlying molecular defects. ASD gating charge mutations in the KCNQ/KV7 potassium channel cause gating pore currents (Igp) and impair action potential (AP) firing of dopaminergic neurons in brain slices. Here, we investigated ASD gating charge mutations of the voltage-gated SCN2A/NaV1.2 brain sodium channel, which ranked high among the ion channel genes with mutations in individuals with ASD. Our results show that ASD mutations in the gating charges R2 in Domain-II (R853Q), and R1 (R1626Q) and R2 (R1629H) in Domain-IV of NaV1.2 caused Igp in the resting state of ~0.1% of the amplitude of central pore current. The R1626Q mutant also caused significant changes in the voltage dependence of fast inactivation, and the R1629H mutant conducted proton-selective Igp. These potentially pathogenic Igp were exacerbated by the absence of the extracellular Mg2+ and Ca2+. In silico simulation of the effects of these mutations in a conductance-based single-compartment cortical neuron model suggests that the inward Igp reduces the time to peak for the first AP in a train, increases AP rates during a train of stimuli, and reduces the interstimulus interval between consecutive APs, consistent with increased neural excitability and altered input/output relationships. Understanding this common pathophysiological mechanism among different voltage-gated ion channels at the circuit level will give insights into the underlying mechanisms of ASD.
Collapse
Affiliation(s)
- Ahmed Eltokhi
- Department of Pharmacology, University of Washington, Seattle, WA98195
| | - Brian Nils Lundstrom
- Department of Neurology in the Division of Epilepsy, Mayo Clinic, Rochester, MN55905
| | - Jin Li
- Department of Pharmacology, University of Washington, Seattle, WA98195
| | - Larry S. Zweifel
- Department of Pharmacology, University of Washington, Seattle, WA98195
- Department of Psychiatry & Behavioral Sciences, University of Washington, Seattle, WA98195
| | | | | |
Collapse
|
4
|
Ding X, Wu Y, Rodriguez V, Ricco E, Okoh JT, Liu Y, Kraushaar DC, Rasband MN. Age-dependent regulation of axoglial interactions and behavior by oligodendrocyte AnkyrinG. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.01.587609. [PMID: 38617359 PMCID: PMC11014615 DOI: 10.1101/2024.04.01.587609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
The bipolar disorder (BD) risk gene ANK3 encodes the scaffolding protein AnkyrinG (AnkG). In neurons, AnkG regulates polarity and ion channel clustering at axon initial segments and nodes of Ranvier. Disruption of neuronal AnkG causes BD-like phenotypes in mice. During development, AnkG is also expressed at comparable levels in oligodendrocytes and facilitates the efficient assembly of paranodal junctions. However, the physiological roles of glial AnkG in the mature nervous system, and its contributions to BD-like phenotypes, remain unexplored. Here, we generated oligodendroglia-specific AnkG conditional knockout mice and observed the destabilization of axoglial interactions in aged but not young adult mice. In addition, these mice exhibited profound histological, electrophysiological, and behavioral pathophysiologies. Unbiased translatomic profiling revealed potential compensatory machineries. These results highlight the critical functions of glial AnkG in maintaining proper axoglial interactions throughout aging and suggests a previously unrecognized contribution of oligodendroglial AnkG to neuropsychiatric disorders.
Collapse
Affiliation(s)
- Xiaoyun Ding
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Yu Wu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Victoria Rodriguez
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, TX 77030
| | - Emily Ricco
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, TX 77030
| | - James T. Okoh
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Yanhong Liu
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | - Daniel C. Kraushaar
- Genomic and RNA Profiling Core, Baylor College of Medicine, Houston, TX 77030
| | - Matthew N. Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
5
|
Mouihate A, Kalakh S. Breastfeeding promotes oligodendrocyte precursor cells division and myelination in the demyelinated corpus callosum. Brain Res 2023; 1821:148584. [PMID: 37717888 DOI: 10.1016/j.brainres.2023.148584] [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: 05/10/2023] [Revised: 09/12/2023] [Accepted: 09/14/2023] [Indexed: 09/19/2023]
Abstract
Demyelination alters the conduction of neuronal signals and hampers sensory-motor functions. Experimental and clinical evidence suggest that breastfeeding exerts a promyelinating impact on the maternal brain. The mechanism underlying this neuroprotective effect is not well-understood. In the present paper, we assessed the impact of rat lactation on lysolecithin-induced demyelination injury within the corpus callosum of lactating and non-lactating postpartum rats. We show that lactation enhanced the cell density of oligodendrocyte precursor cells (OPCs), but not that of activated microglia and astrocytes, within the demyelination lesion. Lactation also increased the expression of myelin markers involved in the initial stage of myelin recovery (Myelin-associated glycoprotein and 2',3'-cyclic nucleotide 3'-phosphodiesterase) and reduced the demyelination injury. Altogether, these data suggest that lactation creates a conducive promyelinating environment through increased OPCs cell division, enhanced expression of select myelin proteins, and reduced number of non-myelinated axons.
Collapse
Affiliation(s)
- Abdeslam Mouihate
- Department of Physiology, College of Medicine, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait.
| | - Samah Kalakh
- Department of Physiology, College of Medicine, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait
| |
Collapse
|
6
|
Dolma S, Joshi A. The Node of Ranvier as an Interface for Axo-Glial Interactions: Perturbation of Axo-Glial Interactions in Various Neurological Disorders. J Neuroimmune Pharmacol 2023; 18:215-234. [PMID: 37285016 DOI: 10.1007/s11481-023-10072-z] [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: 09/08/2022] [Accepted: 05/19/2023] [Indexed: 06/08/2023]
Abstract
The action potential conduction along the axon is highly dependent on the healthy interactions between the axon and myelin-producing glial cells. Myelin, which facilitates action potential, is the protective insulation around the axon formed by Schwann cells and oligodendrocytes in the peripheral (PNS) and central nervous system (CNS), respectively. Myelin is a continuous structure with intermittent gaps called nodes of Ranvier, which are the sites enriched with ion channels, transmembrane, scaffolding, and cytoskeletal proteins. Decades-long extensive research has identified a comprehensive proteome with strictly regularized localization at the node of Ranvier. Concurrently, axon-glia interactions at the node of Ranvier have gathered significant attention as the pathophysiological targets for various neurodegenerative disorders. Numerous studies have shown the alterations in the axon-glia interactions culminating in neurological diseases. In this review, we have provided an update on the molecular composition of the node of Ranvier. Further, we have discussed in detail the consequences of disruption of axon-glia interactions during the pathogenesis of various CNS and PNS disorders.
Collapse
Affiliation(s)
- Sonam Dolma
- Department of Pharmacy, Birla Institute of Technology and Sciences- Pilani, Hyderabad campus, Telangana state, India
| | - Abhijeet Joshi
- Department of Pharmacy, Birla Institute of Technology and Sciences- Pilani, Hyderabad campus, Telangana state, India.
| |
Collapse
|
7
|
Jonusaite S, Oulhen N, Izumi Y, Furuse M, Yamamoto T, Sakamoto N, Wessel G, Heyland A. Identification of the genes encoding candidate septate junction components expressed during early development of the sea urchin, Strongylocentrotus purpuratus, and evidence of a role for Mesh in the formation of the gut barrier. Dev Biol 2023; 495:21-34. [PMID: 36587799 DOI: 10.1016/j.ydbio.2022.12.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/30/2022]
Abstract
Septate junctions (SJs) evolved as cell-cell junctions that regulate the paracellular barrier and integrity of epithelia in invertebrates. Multiple morphological variants of SJs exist specific to different epithelia and/or phyla but the biological significance of varied SJ morphology is unclear because the knowledge of the SJ associated proteins and their functions in non-insect invertebrates remains largely unknown. Here we report cell-specific expression of nine candidate SJ genes in the early life stages of the sea urchin Strongylocentrotus purpuratus. By use of in situ RNA hybridization and single cell RNA-seq we found that the expression of selected genes encoding putatively SJ associated transmembrane and cytoplasmic scaffold molecules was dynamically regulated during epithelial development in the embryos and larvae with different epithelia expressing different cohorts of SJ genes. We focused a functional analysis on SpMesh, a homolog of the Drosophila smooth SJ component Mesh, which was highly enriched in the endodermal epithelium of the mid- and hindgut. Functional perturbation of SpMesh by both CRISPR/Cas9 mutagenesis and vivo morpholino-mediated knockdown shows that loss of SpMesh does not disrupt the formation of the gut epithelium during gastrulation. However, loss of SpMesh resulted in a severely reduced gut-paracellular barrier as quantitated by increased permeability to 3-5 kDa FITC-dextran. Together, these studies provide a first look at the molecular SJ physiology during the development of a marine organism and suggest a shared role for Mesh-homologous proteins in forming an intestinal barrier in invertebrates. Results have implications for consideration of the traits underlying species-specific sensitivity of marine larvae to climate driven ocean change.
Collapse
Affiliation(s)
- Sima Jonusaite
- Department of Integrative Biology, University of Guelph, Guelph, ON N1G 2W1, Canada.
| | - Nathalie Oulhen
- Department of Molecular and Cell Biology and Biochemistry, Brown University, Providence, RI, 02912, United States
| | - Yasushi Izumi
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, 444-8787, Japan
| | - Mikio Furuse
- Division of Cell Structure, National Institute for Physiological Sciences, Okazaki, 444-8787, Japan; Nagoya University Graduate School of Medicine, Aichi, 464-8601, Japan
| | - Takashi Yamamoto
- Division of Integrated Sciences for Life, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8526, Japan
| | - Naoaki Sakamoto
- Division of Integrated Sciences for Life, Graduate School of Integrated Sciences for Life, Hiroshima University, Hiroshima, 739-8526, Japan
| | - Gary Wessel
- Department of Molecular and Cell Biology and Biochemistry, Brown University, Providence, RI, 02912, United States
| | - Andreas Heyland
- Department of Integrative Biology, University of Guelph, Guelph, ON N1G 2W1, Canada
| |
Collapse
|
8
|
Bizzoca A, Jirillo E, Flace P, Gennarini G. Overall Role of Contactins Expression in Neurodevelopmental Events and Contribution to Neurological Disorders. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2023; 22:1176-1193. [PMID: 36515028 DOI: 10.2174/1871527322666221212160048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 10/21/2022] [Accepted: 10/28/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND Neurodegenerative disorders may depend upon a misregulation of the pathways which sustain neurodevelopmental control. In this context, this review article focuses on Friedreich ataxia (FA), a neurodegenerative disorder resulting from mutations within the gene encoding the Frataxin protein, which is involved in the control of mitochondrial function and oxidative metabolism. OBJECTIVE The specific aim of the present study concerns the FA molecular and cellular substrates, for which available transgenic mice models are proposed, including mutants undergoing misexpression of adhesive/morphoregulatory proteins, in particular belonging to the Contactin subset of the immunoglobulin supergene family. METHODS In both mutant and control mice, neurogenesis was explored by morphological/morphometric analysis through the expression of cell type-specific markers, including b-tubulin, the Contactin-1 axonal adhesive glycoprotein, as well as the Glial Fibrillary Acidic Protein (GFAP). RESULTS Specific consequences were found to arise from the chosen misexpression approach, consisting of a neuronal developmental delay associated with glial upregulation. Protective effects against the arising phenotype resulted from antioxidants (essentially epigallocatechin gallate (EGCG)) administration, which was demonstrated through the profiles of neuronal (b-tubulin and Contactin 1) as well as glial (GFAP) markers, in turn indicating the concomitant activation of neurodegeneration and neuro repair processes. The latter also implied activation of the Notch-1 signaling. CONCLUSION Overall, this study supports the significance of changes in morphoregulatory proteins expression in the FA pathogenesis and of antioxidant administration in counteracting it, which, in turn, allows to devise potential therapeutic approaches.
Collapse
Affiliation(s)
- Antonella Bizzoca
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Medical School, University of Bari, Piazza Giulio Cesare, 11. Bari I-70124, Italy
| | - Emilio Jirillo
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Medical School, University of Bari, Piazza Giulio Cesare, 11. Bari I-70124, Italy
| | - Paolo Flace
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Medical School, University of Bari, Piazza Giulio Cesare, 11. Bari I-70124, Italy
| | - Gianfranco Gennarini
- Department of Basic Medical Sciences, Neurosciences, and Sensory Organs, Medical School, University of Bari, Piazza Giulio Cesare, 11. Bari I-70124, Italy
| |
Collapse
|
9
|
Abstract
The ankyrin proteins (Ankyrin-R, Ankyrin-B, and Ankyrin-G) are a family of scaffolding, or membrane adaptor proteins necessary for the regulation and targeting of several types of ion channels and membrane transporters throughout the body. These include voltage-gated sodium, potassium, and calcium channels in the nervous system, heart, lungs, and muscle. At these sites, ankyrins recruit ion channels, and other membrane proteins, to specific subcellular domains, which are then stabilized through ankyrin's interaction with the submembranous spectrin-based cytoskeleton. Several recent studies have expanded our understanding of both ankyrin expression and their ion channel binding partners. This review provides an updated overview of ankyrin proteins and their known channel and transporter interactions. We further discuss several potential avenues of future research that would expand our understanding of these important organizational proteins.
Collapse
Affiliation(s)
- Sharon R. Stevens
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA
| | - Matthew N. Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, USA,CONTACT Matthew N. Rasband Department of Neuroscience, Baylor College of Medicine, One Baylor Plaza, Houston, TX77030, USA
| |
Collapse
|
10
|
Song H, McEwan PP, Ameen-Ali KE, Tomasevich A, Kennedy-Dietrich C, Palma A, Arroyo EJ, Dolle JP, Johnson VE, Stewart W, Smith DH. Concussion leads to widespread axonal sodium channel loss and disruption of the node of Ranvier. Acta Neuropathol 2022; 144:967-985. [PMID: 36107227 PMCID: PMC9547928 DOI: 10.1007/s00401-022-02498-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 01/26/2023]
Abstract
Despite being a major health concern, little is known about the pathophysiological changes that underly concussion. Nonetheless, emerging evidence suggests that selective damage to white matter axons, or diffuse axonal injury (DAI), disrupts brain network connectivity and function. While voltage-gated sodium channels (NaChs) and their anchoring proteins at the nodes of Ranvier (NOR) on axons are key elements of the brain's network signaling machinery, changes in their integrity have not been studied in context with DAI. Here, we utilized a clinically relevant swine model of concussion that induces evolving axonal pathology, demonstrated by accumulation of amyloid precursor protein (APP) across the white matter. Over a two-week follow-up post-concussion with this model, we found widespread loss of NaCh isoform 1.6 (Nav1.6), progressive increases in NOR length, the appearance of void and heminodes and loss of βIV-spectrin, ankyrin G, and neurofascin 186 or their collective diffusion into the paranode. Notably, these changes were in close proximity, yet distinct from APP-immunoreactive swollen axonal profiles, potentially representing a unique, newfound phenotype of axonal pathology in DAI. Since concussion in humans is non-fatal, the clinical relevance of these findings was determined through examination of post-mortem brain tissue from humans with higher levels of acute traumatic brain injury. Here, a similar loss of Nav1.6 and changes in NOR structures in brain white matter were observed as found in the swine model of concussion. Collectively, this widespread and progressive disruption of NaChs and NOR appears to be a form of sodium channelopathy, which may represent an important substrate underlying brain network dysfunction after concussion.
Collapse
Affiliation(s)
- Hailong Song
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - Przemyslaw P McEwan
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - Kamar E Ameen-Ali
- School of Neuroscience and Psychology, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Alexandra Tomasevich
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | | | - Alexander Palma
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - Edgardo J Arroyo
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - Jean-Pierre Dolle
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - Victoria E Johnson
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA
| | - William Stewart
- School of Neuroscience and Psychology, University of Glasgow, Glasgow, G12 8QQ, UK
- Department of Neuropathology, Queen Elizabeth University Hospital, Glasgow, G51 4TF, UK
| | - Douglas H Smith
- Department of Neurosurgery, Center for Brain Injury and Repair, University of Pennsylvania, 3320 Smith Walk, 105 Hayden Hall, Philadelphia, PA, 19104, USA.
| |
Collapse
|
11
|
Balraj A, Clarkson-Paredes C, Pajoohesh-Ganji A, Kay MW, Mendelowitz D, Miller RH. Refinement of axonal conduction and myelination in the mouse optic nerve indicate an extended period of postnatal developmental plasticity. Dev Neurobiol 2022; 82:308-325. [PMID: 35403346 PMCID: PMC9128412 DOI: 10.1002/dneu.22875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 02/18/2022] [Accepted: 03/17/2022] [Indexed: 11/07/2022]
Abstract
Retinal ganglion cells generate a pattern of action potentials to communicate visual information from the retina to cortical areas. Myelin, an insulating sheath, wraps axonal segments to facilitate signal propagation and when deficient, can impair visual function. Optic nerve development and initial myelination has largely been considered complete by the fifth postnatal week. However, the relationship between the extent of myelination and axonal signaling in the maturing optic nerve is not well characterized. Here, we examine the relationship between axon conduction and elements of myelination using extracellular nerve recordings, immunohistochemistry, western blot analysis, scanning electron microscopy, and simulations of nerve responses. Comparing compound action potentials from mice aged 4-12 weeks revealed five functional distinct axonal populations, an increase in the number of functional axons, and shifts toward fast-conducting axon populations at 5 and 8 weeks postnatal. At these ages, our analysis revealed increased myelin thickness, lower g-ratios and changes in the 14 kDa MBP isoform, while the density of axons and nodes of Ranvier remained constant. At 5 postnatal weeks, axon diameter increased, while at 8 weeks, increased expression of a mature sodium ion channel subtype, Nav 1.6, was observed at nodes of Ranvier. A simulation model of nerve conduction suggests that ion channel subtype, axon diameter, and myelin thickness are more likely to be key regulators of nerve function than g-ratio. Such refinement of axonal function and myelin rearrangement identified an extended period of maturation in the normal optic nerve that may facilitate the development of visual signaling patterns. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Annika Balraj
- Department of Anatomy, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Cheryl Clarkson-Paredes
- Nanofabrication and Imaging Center, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Ahdeah Pajoohesh-Ganji
- Department of Anatomy, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Matthew W. Kay
- Department of Biomedical Engineering, The George Washington University, Washington, District of Columbia, USA
| | - David Mendelowitz
- Department of Pharmacology and Physiology, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| | - Robert H. Miller
- Department of Anatomy, The George Washington University School of Medicine and Health Sciences, Washington, District of Columbia, USA
| |
Collapse
|
12
|
Panganiban CH, Barth JL, Tan J, Noble KV, McClaskey CM, Howard BA, Jafri SH, Dias JW, Harris KC, Lang H. Two distinct types of nodes of Ranvier support auditory nerve function in the mouse cochlea. Glia 2021; 70:768-791. [DOI: 10.1002/glia.24138] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Revised: 11/12/2021] [Accepted: 12/17/2021] [Indexed: 11/09/2022]
Affiliation(s)
- Clarisse H. Panganiban
- Department of Pathology and Laboratory Medicine Medical University of South Carolina Charleston South Carolina USA
- Wolfson Centre for Age‐Related Diseases King's College London London UK
| | - Jeremy L. Barth
- Department of Regenerative Medicine and Cell Biology Medical University of South Carolina Charleston South Carolina USA
| | - Junying Tan
- Department of Pathology and Laboratory Medicine Medical University of South Carolina Charleston South Carolina USA
| | - Kenyaria V. Noble
- Department of Pathology and Laboratory Medicine Medical University of South Carolina Charleston South Carolina USA
| | - Carolyn M. McClaskey
- Department of Otolaryngology & Head and Neck Surgery Medical University of South Carolina Charleston South Carolina USA
| | - Blake A. Howard
- Department of Pathology and Laboratory Medicine Medical University of South Carolina Charleston South Carolina USA
| | - Shabih H. Jafri
- Department of Pathology and Laboratory Medicine Medical University of South Carolina Charleston South Carolina USA
| | - James W. Dias
- Department of Otolaryngology & Head and Neck Surgery Medical University of South Carolina Charleston South Carolina USA
| | - Kelly C. Harris
- Department of Otolaryngology & Head and Neck Surgery Medical University of South Carolina Charleston South Carolina USA
| | - Hainan Lang
- Department of Pathology and Laboratory Medicine Medical University of South Carolina Charleston South Carolina USA
| |
Collapse
|
13
|
Pathophysiology of the Different Clinical Phenotypes of Chronic Inflammatory Demyelinating Polyradiculoneuropathy (CIDP). Int J Mol Sci 2021; 23:ijms23010179. [PMID: 35008604 PMCID: PMC8745770 DOI: 10.3390/ijms23010179] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 12/22/2022] Open
Abstract
Chronic inflammatory demyelinating polyneuropathy (CIDP) is the most common form of autoimmune polyneuropathy. It is a chronic disease and may be monophasic, progressive or recurrent with exacerbations and incomplete remissions, causing accumulating disability. In recent years, there has been rapid progress in understanding the background of CIDP, which allowed us to distinguish specific phenotypes of this disease. This in turn allowed us to better understand the mechanism of response or non-response to various forms of therapy. On the basis of a review of the relevant literature, the authors present the current state of knowledge concerning the pathophysiology of the different clinical phenotypes of CIDP as well as ongoing research in this field, with reference to key points of immune-mediated processes involved in the background of CIDP.
Collapse
|
14
|
Duncan GJ, Simkins TJ, Emery B. Neuron-Oligodendrocyte Interactions in the Structure and Integrity of Axons. Front Cell Dev Biol 2021; 9:653101. [PMID: 33763430 PMCID: PMC7982542 DOI: 10.3389/fcell.2021.653101] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 02/15/2021] [Indexed: 12/12/2022] Open
Abstract
The myelination of axons by oligodendrocytes is a highly complex cell-to-cell interaction. Oligodendrocytes and axons have a reciprocal signaling relationship in which oligodendrocytes receive cues from axons that direct their myelination, and oligodendrocytes subsequently shape axonal structure and conduction. Oligodendrocytes are necessary for the maturation of excitatory domains on the axon including nodes of Ranvier, help buffer potassium, and support neuronal energy metabolism. Disruption of the oligodendrocyte-axon unit in traumatic injuries, Alzheimer's disease and demyelinating diseases such as multiple sclerosis results in axonal dysfunction and can culminate in neurodegeneration. In this review, we discuss the mechanisms by which demyelination and loss of oligodendrocytes compromise axons. We highlight the intra-axonal cascades initiated by demyelination that can result in irreversible axonal damage. Both the restoration of oligodendrocyte myelination or neuroprotective therapies targeting these intra-axonal cascades are likely to have therapeutic potential in disorders in which oligodendrocyte support of axons is disrupted.
Collapse
Affiliation(s)
- Greg J. Duncan
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Tyrell J. Simkins
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- Vollum Institute, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, VA Portland Health Care System, Portland, OR, United States
| | - Ben Emery
- Jungers Center for Neurosciences Research, Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
15
|
Kunisawa K, Hatanaka N, Shimizu T, Kobayashi K, Osanai Y, Mouri A, Shi Q, Bhat MA, Nambu A, Ikenaka K. Focal loss of the paranodal domain protein Neurofascin155 in the internal capsule impairs cortically induced muscle activity in vivo. Mol Brain 2020; 13:159. [PMID: 33228720 PMCID: PMC7685608 DOI: 10.1186/s13041-020-00698-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 11/13/2020] [Indexed: 11/12/2022] Open
Abstract
Paranodal axoglial junctions are essential for rapid nerve conduction and the organization of axonal domains in myelinated axons. Neurofascin155 (Nfasc155) is a glial cell adhesion molecule that is also required for the assembly of these domains. Previous studies have demonstrated that general ablation of Nfasc155 disorganizes these domains, reduces conduction velocity, and disrupts motor behaviors. Multiple sclerosis (MS), a typical disorder of demyelination in the central nervous system, is reported to have autoantibody to Nfasc. However, the impact of focal loss of Nfasc155, which may occur in MS patients, remains unclear. Here, we examined whether restricted focal loss of Nfasc155 affects the electrophysiological properties of the motor system in vivo. Adeno-associated virus type5 (AAV5) harboring EGFP-2A-Cre was injected into the glial-enriched internal capsule of floxed-Neurofascin (NfascFlox/Flox) mice to focally disrupt paranodal junctions in the cortico-fugal fibers from the motor cortex to the spinal cord. Electromyograms (EMGs) of the triceps brachii muscles in response to electrical stimulation of the motor cortex were successively examined in these awake mice. EMG analysis showed significant delay in the onset and peak latencies after AAV injection compared to control (Nfasc+/+) mice. Moreover, EMG half-widths were increased, and EMG amplitudes were gradually decreased by 13 weeks. Similar EMG changes have been reported in MS patients. These findings provide physiological evidence that motor outputs are obstructed by focal ablation of paranodal junctions in myelinated axons. Our findings may open a new path toward development of a novel biomarker for an early phase of human MS, as Nfasc155 detects microstructural changes in the paranodal junction.
Collapse
Affiliation(s)
- Kazuo Kunisawa
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, 444-8787, Japan
- Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8787, Japan
- Department of Regulatory Science for Evaluation and Development of Pharmaceuticals and Devices, Fujita Health University Graduate School of Health Sciences, Toyoake, 470-1192, Japan
| | - Nobuhiko Hatanaka
- Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8787, Japan.
- Division of System Neurophysiology, National Institute for Physiological Sciences, 38 Nishigonaka, Myodaiji, Okazaki, 444-8585, Japan.
| | - Takeshi Shimizu
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, 444-8787, Japan
- Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8787, Japan
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Kenta Kobayashi
- Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8787, Japan
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan
| | - Yasuyuki Osanai
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, 444-8787, Japan
- Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8787, Japan
| | - Akihiro Mouri
- Department of Regulatory Science for Evaluation and Development of Pharmaceuticals and Devices, Fujita Health University Graduate School of Health Sciences, Toyoake, 470-1192, Japan
| | - Qian Shi
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center, San Antonio, 78229-3900, USA
| | - Manzoor A Bhat
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center, San Antonio, 78229-3900, USA
| | - Atsushi Nambu
- Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8787, Japan
- Division of System Neurophysiology, National Institute for Physiological Sciences, 38 Nishigonaka, Myodaiji, Okazaki, 444-8585, Japan
| | - Kazuhiro Ikenaka
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, 444-8787, Japan
- Department of Physiological Sciences, SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8787, Japan
| |
Collapse
|
16
|
Lubetzki C, Sol-Foulon N, Desmazières A. Nodes of Ranvier during development and repair in the CNS. Nat Rev Neurol 2020; 16:426-439. [DOI: 10.1038/s41582-020-0375-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2020] [Indexed: 01/01/2023]
|
17
|
Abstract
Axons functionally link the somato-dendritic compartment to synaptic terminals. Structurally and functionally diverse, they accomplish a central role in determining the delays and reliability with which neuronal ensembles communicate. By combining their active and passive biophysical properties, they ensure a plethora of physiological computations. In this review, we revisit the biophysics of generation and propagation of electrical signals in the axon and their dynamics. We further place the computational abilities of axons in the context of intracellular and intercellular coupling. We discuss how, by means of sophisticated biophysical mechanisms, axons expand the repertoire of axonal computation, and thereby, of neural computation.
Collapse
Affiliation(s)
- Pepe Alcami
- Division of Neurobiology, Department of Biology II, Ludwig-Maximilians-Universitaet Muenchen, Martinsried, Germany
- Department of Behavioural Neurobiology, Max Planck Institute for Ornithology, Seewiesen, Germany
| | - Ahmed El Hady
- Princeton Neuroscience Institute, Princeton University, Princeton, NJ, United States
- Howard Hughes Medical Institute, Princeton University, Princeton, NJ, United States
| |
Collapse
|
18
|
Dubessy AL, Mazuir E, Rappeneau Q, Ou S, Abi Ghanem C, Piquand K, Aigrot MS, Thétiot M, Desmazières A, Chan E, Fitzgibbon M, Fleming M, Krauss R, Zalc B, Ranscht B, Lubetzki C, Sol-Foulon N. Role of a Contactin multi-molecular complex secreted by oligodendrocytes in nodal protein clustering in the CNS. Glia 2019; 67:2248-2263. [PMID: 31328333 PMCID: PMC6851800 DOI: 10.1002/glia.23681] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/12/2019] [Accepted: 06/14/2019] [Indexed: 01/06/2023]
Abstract
The fast and reliable propagation of action potentials along myelinated fibers relies on the clustering of voltage‐gated sodium channels at nodes of Ranvier. Axo‐glial communication is required for assembly of nodal proteins in the central nervous system, yet the underlying mechanisms remain poorly understood. Oligodendrocytes are known to support node of Ranvier assembly through paranodal junction formation. In addition, the formation of early nodal protein clusters (or prenodes) along axons prior to myelination has been reported, and can be induced by oligodendrocyte conditioned medium (OCM). Our recent work on cultured hippocampal neurons showed that OCM‐induced prenodes are associated with an increased conduction velocity (Freeman et al., 2015). We here unravel the nature of the oligodendroglial secreted factors. Mass spectrometry analysis of OCM identified several candidate proteins (i.e., Contactin‐1, ChL1, NrCAM, Noelin2, RPTP/Phosphacan, and Tenascin‐R). We show that Contactin‐1 combined with RPTP/Phosphacan or Tenascin‐R induces clusters of nodal proteins along hippocampal GABAergic axons. Furthermore, Contactin‐1‐immunodepleted OCM or OCM from Cntn1‐null mice display significantly reduced clustering activity, that is restored by addition of soluble Contactin‐1. Altogether, our results identify Contactin‐1 secreted by oligodendrocytes as a novel factor that may influence early steps of nodal sodium channel cluster formation along specific axon populations.
Collapse
Affiliation(s)
- Anne-Laure Dubessy
- Sorbonne Université, Inserm, CNRS, UMR7225, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Assistance Publique-Hôpitaux de Paris, GH Pitié-Salpêtrière, Paris, France
| | - Elisa Mazuir
- Sorbonne Université, Inserm, CNRS, UMR7225, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Quentin Rappeneau
- Sorbonne Université, Inserm, CNRS, UMR7225, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Sokounthie Ou
- Sorbonne Université, Inserm, CNRS, UMR7225, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Charly Abi Ghanem
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Kevin Piquand
- Sorbonne Université, Inserm, CNRS, UMR7225, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Marie-Stéphane Aigrot
- Sorbonne Université, Inserm, CNRS, UMR7225, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Melina Thétiot
- Sorbonne Université, Inserm, CNRS, UMR7225, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Anne Desmazières
- Sorbonne Université, Inserm, CNRS, UMR7225, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Eric Chan
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts
| | | | - Mark Fleming
- Vertex Pharmaceuticals Incorporated, Boston, Massachusetts
| | - Raul Krauss
- Disarm Therapeutics, Cambridge, Massachusetts
| | - Bernard Zalc
- Sorbonne Université, Inserm, CNRS, UMR7225, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Barbara Ranscht
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California
| | - Catherine Lubetzki
- Sorbonne Université, Inserm, CNRS, UMR7225, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,Assistance Publique-Hôpitaux de Paris, GH Pitié-Salpêtrière, Paris, France
| | - Nathalie Sol-Foulon
- Sorbonne Université, Inserm, CNRS, UMR7225, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| |
Collapse
|
19
|
Kunisawa K, Shimizu T, Kushima I, Aleksic B, Mori D, Osanai Y, Kobayashi K, Taylor AM, Bhat MA, Hayashi A, Baba H, Ozaki N, Ikenaka K. Dysregulation of schizophrenia-related aquaporin 3 through disruption of paranode influences neuronal viability. J Neurochem 2018; 147:395-408. [PMID: 30025158 PMCID: PMC6205917 DOI: 10.1111/jnc.14553] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Revised: 06/28/2018] [Accepted: 07/16/2018] [Indexed: 12/23/2022]
Abstract
Myelinated axons segregate the axonal membrane into four defined regions: the node of Ranvier, paranode, juxtaparanode, and internode. The paranodal junction consists of specific component proteins, such as neurofascin155 (NF155) on the glial side, and Caspr and Contactin on the axonal side. Although paranodal junctions are thought to play crucial roles in rapid saltatory conduction and nodal assembly, the role of their interaction with neurons is not fully understood. In a previous study, conditional NF155 knockout in oligodendrocytes led to disorganization of the paranodal junctions. To examine if disruption of paranodal junctions affects neuronal gene expression, we prepared total RNA from the retina of NF155 conditional knockout, and performed expression analysis. We found that the expression level of 433 genes changed in response to paranodal junction ablation. Interestingly, expression of aquaporin 3 (AQP3) was significantly reduced in NF155 conditional knockout mice, but not in cerebroside sulfotransferase knockout (CST-KO) mice, whose paranodes are not originally formed during development. Copy number variations have an important role in the etiology of schizophrenia (SCZ). We observed rare duplications of AQP3 in SCZ patients, suggesting a correlation between abnormal AQP3 expression and SCZ. To determine if AQP3 over-expression in NF155 conditional knockout mice influences neuronal function, we performed adeno-associated virus (AAV)-mediated over-expression of AQP3 in the motor cortex of mice and found a significant increase in caspase 3-dependent neuronal apoptosis in AQP3-transduced cells. This study may provide new insights into therapeutic approaches for SCZ by regulating AQP3 expression, which is associated with paranodal disruption.
Collapse
Affiliation(s)
- Kazuo Kunisawa
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki 444-8787, Japan
- SOKENDAI (The Graduate University for Advanced Studies), Okazaki 444-8787, Japan
| | - Takeshi Shimizu
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki 444-8787, Japan
- SOKENDAI (The Graduate University for Advanced Studies), Okazaki 444-8787, Japan
| | - Itaru Kushima
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Branko Aleksic
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Daisuke Mori
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
- Brain and Mind Research Center, Nagoya University, Nagoya 466-8550, Japan
| | - Yasuyuki Osanai
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki 444-8787, Japan
- SOKENDAI (The Graduate University for Advanced Studies), Okazaki 444-8787, Japan
| | - Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological Sciences, Okazaki 444-8585, Japan
- SOKENDAI (The Graduate University for Advanced Studies), Okazaki 444-8787, Japan
| | - Anna M. Taylor
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center, San Antonio 78229-3900, USA
| | - Manzoor A. Bhat
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center, San Antonio 78229-3900, USA
| | - Akiko Hayashi
- Department of Molecular Neurobiology, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Japan
| | - Hiroko Baba
- Department of Molecular Neurobiology, Tokyo University of Pharmacy and Life Sciences, Hachioji 192-0392, Japan
| | - Norio Ozaki
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Kazuhiro Ikenaka
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki 444-8787, Japan
- SOKENDAI (The Graduate University for Advanced Studies), Okazaki 444-8787, Japan
| |
Collapse
|
20
|
Abstract
The speed of impulse transmission is critical for optimal neural circuit function, but it is unclear how the appropriate conduction velocity is established in individual axons. The velocity of impulse transmission is influenced by the thickness of the myelin sheath and the morphology of electrogenic nodes of Ranvier along axons. Here we show that myelin thickness and nodal gap length are reversibly altered by astrocytes, glial cells that contact nodes of Ranvier. Thrombin-dependent proteolysis of a cell adhesion molecule that attaches myelin to the axon (neurofascin 155) is inhibited by vesicular release of thrombin protease inhibitors from perinodal astrocytes. Transgenic mice expressing a dominant-negative fragment of VAMP2 in astrocytes, to reduce exocytosis by 50%, exhibited detachment of adjacent paranodal loops of myelin from the axon, increased nodal gap length, and thinning of the myelin sheath in the optic nerve. These morphological changes alter the passive cable properties of axons to reduce conduction velocity and spike-time arrival in the CNS in parallel with a decrease in visual acuity. All effects were reversed by the thrombin inhibitor Fondaparinux. Similar results were obtained by viral transfection of tetanus toxin into astrocytes of rat corpus callosum. Previously, it was unknown how the myelin sheath could be thinned and the functions of perinodal astrocytes were not well understood. These findings describe a form of nervous system plasticity in which myelin structure and conduction velocity are adjusted by astrocytes. The thrombin-dependent cleavage of neurofascin 155 may also have relevance to myelin disruption and repair.
Collapse
|
21
|
Lee JY, Kim MJ, Li L, Velumian AA, Aui PM, Fehlings MG, Petratos S. Nogo receptor 1 regulates Caspr distribution at axo-glial units in the central nervous system. Sci Rep 2017; 7:8958. [PMID: 28827698 PMCID: PMC5567129 DOI: 10.1038/s41598-017-09405-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 07/24/2017] [Indexed: 11/09/2022] Open
Abstract
Axo-glial units are highly organised microstructures propagating saltatory conduction and are disrupted during multiple sclerosis (MS). Nogo receptor 1 (NgR1) has been suggested to govern axonal damage during the progression of disease in the MS-like mouse model, experimental autoimmune encephalomyelitis (EAE). Here we have identified that adult ngr1 -/- mice, previously used in EAE and spinal cord injury experiments, display elongated paranodes, and nodes of Ranvier. Unstructured paranodal regions in ngr1 -/- mice are matched with more distributed expression pattern of Caspr. Compound action potentials of optic nerves and spinal cords from naïve ngr1 -/- mice are delayed and reduced. Molecular interaction studies revealed enhanced Caspr cleavage. Our data suggest that NgR1 may regulate axo-myelin ultrastructure through Caspr-mediated adhesion, regulating the electrophysiological signature of myelinated axons of central nervous system (CNS).
Collapse
Affiliation(s)
- Jae Young Lee
- Department of Medicine, Central Clinical School, Monash University, Prahran, Victoria, 3004, Australia
- ToolGen, Inc., #1204, Byucksan Digital Valley 6-cha, Seoul, South Korea
| | - Min Joung Kim
- Department of Medicine, Central Clinical School, Monash University, Prahran, Victoria, 3004, Australia
| | - Lijun Li
- Krembil Research Institute, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Alexander A Velumian
- Krembil Research Institute, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Krembil Neuroscience Centre, University Health Network, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Pei Mun Aui
- Department of Medicine, Central Clinical School, Monash University, Prahran, Victoria, 3004, Australia
| | - Michael G Fehlings
- Krembil Research Institute, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Krembil Neuroscience Centre, University Health Network, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Surgery, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Steven Petratos
- Department of Medicine, Central Clinical School, Monash University, Prahran, Victoria, 3004, Australia.
| |
Collapse
|
22
|
Liang W, Zhang W, Zhao S, Liang H, Zhang J, Wang L. Alterations of Caspr2 and Nav1.6 on myelinated axon damage in a rat model of chronic cerebral hypoperfusion. Exp Ther Med 2017; 13:2468-2472. [PMID: 28565865 PMCID: PMC5443296 DOI: 10.3892/etm.2017.4228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 01/11/2017] [Indexed: 11/19/2022] Open
Abstract
Myelinated axons require the correct localization of key proteins that are essential for nerve conduction and cognitive function. Little is known regarding the altered expression of contactin-associated protein 2 (Caspr2) at the juxtaparanodal regions and Nav1.6 at the node of Ranvier in response to chronic cerebral hypoperfusion (CCH). The aim of the present study was to examine the alterations in the key protein of myelinated axons and the potential mechanisms that may follow CCH. We established a rat model of CCH by controllable partial narrowing of bilateral common carotid arteries. Then, we detected cerebral blood flow (CBF) after surgery. We also evaluated motor-evoked potentials (MEPs), assessed the Morris water maze test, analyzed Caspr2 expression through immunohistochemistry and Nav1.6 protein expression through western blot analysis at 2, 4 and 12 weeks. The results revealed that the mean CBF value was significantly decreased to 33.90±5.48%. The MEP latencies and the escaping latencies were significantly prolonged. There was also an elongation of the first time passing of the hidden platform with a reduction of crossing platform times in spatial probing. Furthermore, the Caspr2 immunoreactivity demonstrated that the Caspr2 level was significantly downregulated with abnormal locations in the corpus callosum. The western blot analysis of Nav1.6 protein revealed that the level was reduced significantly over time. The results demonstrate that CCH leads to central conductive function loss, cognitive function damage and alterations in the key protein of myelinated axons, which may provide a molecular basis and key link for white matter damage.
Collapse
Affiliation(s)
- Weihua Liang
- No. 263 Clinic of PLA Army General Hospital, Beijing 101149, P.R. China.,Department of Neurology, Xinqiao Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Weiwei Zhang
- PLA Army General Hospital, Beijing 100700, P.R. China
| | - Shifu Zhao
- Department of Neurology, Xinqiao Hospital, The Third Military Medical University, Chongqing 400038, P.R. China
| | - Hua Liang
- The 66083rd of PLA, Beijing 102488, P.R. China
| | - Jinli Zhang
- No. 263 Clinic of PLA Army General Hospital, Beijing 101149, P.R. China
| | - Luyan Wang
- No. 263 Clinic of PLA Army General Hospital, Beijing 101149, P.R. China
| |
Collapse
|
23
|
Duménieu M, Oulé M, Kreutz MR, Lopez-Rojas J. The Segregated Expression of Voltage-Gated Potassium and Sodium Channels in Neuronal Membranes: Functional Implications and Regulatory Mechanisms. Front Cell Neurosci 2017; 11:115. [PMID: 28484374 PMCID: PMC5403416 DOI: 10.3389/fncel.2017.00115] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Accepted: 04/05/2017] [Indexed: 01/25/2023] Open
Abstract
Neurons are highly polarized cells with apparent functional and morphological differences between dendrites and axon. A critical determinant for the molecular and functional identity of axonal and dendritic segments is the restricted expression of voltage-gated ion channels (VGCs). Several studies show an uneven distribution of ion channels and their differential regulation within dendrites and axons, which is a prerequisite for an appropriate integration of synaptic inputs and the generation of adequate action potential (AP) firing patterns. This review article will focus on the signaling pathways leading to segmented expression of voltage-gated potassium and sodium ion channels at the neuronal plasma membrane and the regulatory mechanisms ensuring segregated functions. We will also discuss the relevance of proper ion channel targeting for neuronal physiology and how alterations in polarized distribution contribute to neuronal pathology.
Collapse
Affiliation(s)
- Maël Duménieu
- Research Group Neuroplasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany
| | - Marie Oulé
- Research Group Neuroplasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany
| | - Michael R Kreutz
- Research Group Neuroplasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany.,Leibniz Group "Dendritic Organelles and Synaptic Function", University Medical Center Hamburg-Eppendorf, Center for Molecular Neurobiology (ZMNH)Hamburg, Germany
| | - Jeffrey Lopez-Rojas
- Research Group Neuroplasticity, Leibniz Institute for NeurobiologyMagdeburg, Germany
| |
Collapse
|
24
|
Arancibia-Cárcamo IL, Ford MC, Cossell L, Ishida K, Tohyama K, Attwell D. Node of Ranvier length as a potential regulator of myelinated axon conduction speed. eLife 2017; 6. [PMID: 28130923 PMCID: PMC5313058 DOI: 10.7554/elife.23329] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Accepted: 01/24/2017] [Indexed: 12/30/2022] Open
Abstract
Myelination speeds conduction of the nerve impulse, enhancing cognitive power. Changes of white matter structure contribute to learning, and are often assumed to reflect an altered number of myelin wraps. We now show that, in rat optic nerve and cerebral cortical axons, the node of Ranvier length varies over a 4.4-fold and 8.7-fold range respectively and that variation of the node length is much less along axons than between axons. Modelling predicts that these node length differences will alter conduction speed by ~20%, similar to the changes produced by altering the number of myelin wraps or the internode length. For a given change of conduction speed, the membrane area change needed at the node is >270-fold less than that needed in the myelin sheath. Thus, axon-specific adjustment of node of Ranvier length is potentially an energy-efficient and rapid mechanism for tuning the arrival time of information in the CNS. DOI:http://dx.doi.org/10.7554/eLife.23329.001 Information is transmitted around the nervous system as electrical signals passing along nerve cells. A fatty substance called myelin, which is wrapped around the nerve cells, increases the speed with which the signals travel along the nerve cells. This allows us to think and move faster than we would otherwise be able to do. The electrical signals start at small “nodes” between areas of myelin wrapping. Originally it was thought that we learn things mainly as a result of changes in the strength of connections between nerve cells, but recently it has been proposed that changes in myelin wrapping could also contribute to learning. Arancibia-Cárcamo, Ford, Cossell et al. investigated how much node structure varies in rat nerve cells, and whether differences in the length of nodes can fine-tune the activity of the nervous system. The experiments show that rat nerve cells do indeed have nodes with a range of different lengths. Calculations show that this could result in electrical signals moving at different speeds through different nerve cells. These findings raise the possibility that nerve cells actively alter the length of their nodes in order to alter their signal speed. The next step is to try to show experimentally that this happens during learning in animals. DOI:http://dx.doi.org/10.7554/eLife.23329.002
Collapse
Affiliation(s)
- I Lorena Arancibia-Cárcamo
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Marc C Ford
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Lee Cossell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| | - Kinji Ishida
- The Center for Electron Microscopy and Bio-Imaging Research, Iwate Medical University, Morioka, Japan
| | - Koujiro Tohyama
- The Center for Electron Microscopy and Bio-Imaging Research, Iwate Medical University, Morioka, Japan.,Department of Physiology, School of Dentistry, Iwate Medical University, Morioka, Japan
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London, United Kingdom
| |
Collapse
|
25
|
Zou Y, Zhang WF, Liu HY, Li X, Zhang X, Ma XF, Sun Y, Jiang SY, Ma QH, Xu DE. Structure and function of the contactin-associated protein family in myelinated axons and their relationship with nerve diseases. Neural Regen Res 2017; 12:1551-1558. [PMID: 29090003 PMCID: PMC5649478 DOI: 10.4103/1673-5374.215268] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The contactin-associated protein (Caspr) family participates in nerve excitation and conduction, and neurotransmitter release in myelinated axons. We analyzed the structures and functions of the Caspr family–CNTNAP1 (Caspr1), CNTNAP2 (Caspr2), CNTNAP3 (Caspr3), CNTNAP4 (Caspr4) and CNTNAP5 (Caspr5), Caspr1–5 is not only involved in the formation of myelinated axons, but also participates in maintaining the stability of adjacent connections. Caspr1 participates in the formation, differentiation, and proliferation of neurons and astrocytes, and in motor control and cognitive function. We also analyzed the relationship between the Caspr family and neurodegenerative diseases, multiple sclerosis, and autoimmune encephalitis. However, the effects of Caspr on disease course and prognosis remain poorly understood. The effects of Caspr on disease diagnosis and treatment need further investigation.
Collapse
Affiliation(s)
- Yan Zou
- Department of Neurology, The Second People's Hospital of Wuxi, Wuxi, Jiangsu Province, China
| | - Wei-Feng Zhang
- Department of Neurology, The Second People's Hospital of Wuxi, Wuxi, Jiangsu Province, China
| | - Hai-Ying Liu
- Department of Neurology, The Second People's Hospital of Wuxi, Wuxi, Jiangsu Province, China
| | - Xia Li
- Department of Neurology, The Second People's Hospital of Wuxi, Wuxi, Jiangsu Province, China
| | - Xing Zhang
- Department of Neurology, The Second People's Hospital of Wuxi, Wuxi, Jiangsu Province, China
| | - Xiao-Fang Ma
- Department of Neurology, The Second People's Hospital of Wuxi, Wuxi, Jiangsu Province, China
| | - Yang Sun
- Department of Neurology, The Second People's Hospital of Wuxi, Wuxi, Jiangsu Province, China
| | - Shi-Yi Jiang
- Department of Neurology, The Second People's Hospital of Wuxi, Wuxi, Jiangsu Province, China
| | - Quan-Hong Ma
- Department of Neurology, The Second People's Hospital of Wuxi, Wuxi, Jiangsu Province, China
| | - De-En Xu
- Department of Neurology, The Second People's Hospital of Wuxi, Wuxi, Jiangsu Province, China
| |
Collapse
|
26
|
Gennarini G, Bizzoca A, Picocci S, Puzzo D, Corsi P, Furley AJW. The role of Gpi-anchored axonal glycoproteins in neural development and neurological disorders. Mol Cell Neurosci 2016; 81:49-63. [PMID: 27871938 DOI: 10.1016/j.mcn.2016.11.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 11/10/2016] [Accepted: 11/14/2016] [Indexed: 01/06/2023] Open
Abstract
This review article focuses on the Contactin (CNTN) subset of the Immunoglobulin supergene family (IgC2/FNIII molecules), whose components share structural properties (the association of Immunoglobulin type C2 with Fibronectin type III domains), as well as a general role in cell contact formation and axonal growth control. IgC2/FNIII molecules include 6 highly related components (CNTN 1-6), associated with the cell membrane via a Glycosyl Phosphatidyl Inositol (GPI)-containing lipid tail. Contactin 1 and Contactin 2 share ~50 (49.38)% identity at the aminoacid level. They are components of the cell surface, from which they may be released in soluble forms. They bind heterophilically to multiple partners in cis and in trans, including members of the related L1CAM family and of the Neurexin family Contactin-associated proteins (CNTNAPs or Casprs). Such interactions are important for organising the neuronal membrane, as well as for modulating the growth and pathfinding of axon tracts. In addition, they also mediate the functional maturation of axons by promoting their interactions with myelinating cells at the nodal, paranodal and juxtaparanodal regions. Such interactions also mediate differential ionic channels (both Na+ and K+) distribution, which is of critical relevance in the generation of the peak-shaped action potential. Indeed, thanks to their interactions with Ankyrin G, Na+ channels map within the nodal regions, where they drive axonal depolarization. However, no ionic channels are found in the flanking Contactin1-containing paranodal regions, where CNTN1 interactions with Caspr1 and with the Ig superfamily component Neurofascin 155 in cis and in trans, respectively, build a molecular barrier between the node and the juxtaparanode. In this region K+ channels are clustered, depending upon molecular interactions with Contactin 2 and with Caspr2. In addition to these functions, the Contactins appear to have also a role in degenerative and inflammatory disorders: indeed Contactin 2 is involved in neurodegenerative disorders with a special reference to the Alzheimer disease, given its ability to work as a ligand of the Alzheimer Precursor Protein (APP), which results in increased Alzheimer Intracellular Domain (AICD) release in a γ-secretase-dependent manner. On the other hand Contactin 1 drives Notch signalling activation via the Hes pathway, which could be consistent with its ability to modulate neuroinflammation events, and with the possibility that Contactin 1-dependent interactions may participate to the pathogenesis of the Multiple Sclerosis and of other inflammatory disorders.
Collapse
Affiliation(s)
- Gianfranco Gennarini
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy.
| | - Antonella Bizzoca
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy
| | - Sabrina Picocci
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy
| | - Daniela Puzzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Italy
| | - Patrizia Corsi
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy
| | - Andrew J W Furley
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield S10 2NT, UK
| |
Collapse
|
27
|
Abstract
Myelinated axons are divided into polarized subdomains including axon initial segments and nodes of Ranvier. These domains initiate and propagate action potentials and regulate the trafficking and localization of somatodendritic and axonal proteins. Formation of axon initial segments and nodes of Ranvier depends on intrinsic (neuronal) and extrinsic (glial) interactions. Several levels of redundancy in both mechanisms and molecules also exist to ensure efficient node formation. Furthermore, the establishment of polarized domains at and near nodes of Ranvier reflects the intrinsic polarity of the myelinating glia responsible for node assembly. Here, we discuss the various polarized domains of myelinated axons, how they are established by both intrinsic and extrinsic interactions, and the polarity of myelinating glia.
Collapse
Affiliation(s)
- Daniel R Zollinger
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030;
| | - Kelli L Baalman
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030;
| | - Matthew N Rasband
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030;
| |
Collapse
|
28
|
Rash JE, Vanderpool KG, Yasumura T, Hickman J, Beatty JT, Nagy JI. KV1 channels identified in rodent myelinated axons, linked to Cx29 in innermost myelin: support for electrically active myelin in mammalian saltatory conduction. J Neurophysiol 2016; 115:1836-59. [PMID: 26763782 PMCID: PMC4869480 DOI: 10.1152/jn.01077.2015] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 01/04/2016] [Indexed: 11/22/2022] Open
Abstract
Saltatory conduction in mammalian myelinated axons was thought to be well understood before recent discoveries revealed unexpected subcellular distributions and molecular identities of the K(+)-conductance pathways that provide for rapid axonal repolarization. In this study, we visualize, identify, localize, quantify, and ultrastructurally characterize axonal KV1.1/KV1.2 channels in sciatic nerves of rodents. With the use of light microscopic immunocytochemistry and freeze-fracture replica immunogold labeling electron microscopy, KV1.1/KV1.2 channels are localized to three anatomically and compositionally distinct domains in the internodal axolemmas of large myelinated axons, where they form densely packed "rosettes" of 9-nm intramembrane particles. These axolemmal KV1.1/KV1.2 rosettes are precisely aligned with and ultrastructurally coupled to connexin29 (Cx29) channels, also in matching rosettes, in the surrounding juxtaparanodal myelin collars and along the inner mesaxon. As >98% of transmembrane proteins large enough to represent ion channels in these specialized domains, ∼500,000 KV1.1/KV1.2 channels define the paired juxtaparanodal regions as exclusive membrane domains for the voltage-gated K(+)conductance that underlies rapid axonal repolarization in mammals. The 1:1 molecular linkage of KV1 channels to Cx29 channels in the apposed juxtaparanodal collars, plus their linkage to an additional 250,000-400,000 Cx29 channels along each inner mesaxon in every large-diameter myelinated axon examined, supports previously proposed K(+)conductance directly from juxtaparanodal axoplasm into juxtaparanodal myeloplasm in mammalian axons. With neither Cx29 protein nor myelin rosettes detectable in frog myelinated axons, these data showing axon-to-myelin linkage by abundant KV1/Cx29 channels in rodent axons support renewed consideration of an electrically active role for myelin in increasing both saltatory conduction velocity and maximum propagation frequency in mammalian myelinated axons.
Collapse
Affiliation(s)
- John E Rash
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado; Program in Molecular, Cellular and Integrative Neurosciences, Colorado State University, Fort Collins, Colorado; and
| | - Kimberly G Vanderpool
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Thomas Yasumura
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Jordan Hickman
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Jonathan T Beatty
- Department of Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - James I Nagy
- Department of Physiology, Faculty of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
29
|
Axons provide the secretory machinery for trafficking of voltage-gated sodium channels in peripheral nerve. Proc Natl Acad Sci U S A 2016; 113:1823-8. [PMID: 26839409 DOI: 10.1073/pnas.1514943113] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
The regulation of the axonal proteome is key to generate and maintain neural function. Fast and slow axoplasmic waves have been known for decades, but alternative mechanisms to control the abundance of axonal proteins based on local synthesis have also been identified. The presence of the endoplasmic reticulum has been documented in peripheral axons, but it is still unknown whether this localized organelle participates in the delivery of axonal membrane proteins. Voltage-gated sodium channels are responsible for action potentials and are mostly concentrated in the axon initial segment and nodes of Ranvier. Despite their fundamental role, little is known about the intracellular trafficking mechanisms that govern their availability in mature axons. Here we describe the secretory machinery in axons and its contribution to plasma membrane delivery of sodium channels. The distribution of axonal secretory components was evaluated in axons of the sciatic nerve and in spinal nerve axons after in vivo electroporation. Intracellular protein trafficking was pharmacologically blocked in vivo and in vitro. Axonal voltage-gated sodium channel mRNA and local trafficking were examined by RT-PCR and a retention-release methodology. We demonstrate that mature axons contain components of the endoplasmic reticulum and other biosynthetic organelles. Axonal organelles and sodium channel localization are sensitive to local blockade of the endoplasmic reticulum to Golgi transport. More importantly, secretory organelles are capable of delivering sodium channels to the plasma membrane in isolated axons, demonstrating an intrinsic capacity of the axonal biosynthetic route in regulating the axonal proteome in mammalian axons.
Collapse
|
30
|
Freeman SA, Desmazières A, Fricker D, Lubetzki C, Sol-Foulon N. Mechanisms of sodium channel clustering and its influence on axonal impulse conduction. Cell Mol Life Sci 2016; 73:723-35. [PMID: 26514731 PMCID: PMC4735253 DOI: 10.1007/s00018-015-2081-1] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 10/21/2015] [Accepted: 10/22/2015] [Indexed: 12/16/2022]
Abstract
The efficient propagation of action potentials along nervous fibers is necessary for animals to interact with the environment with timeliness and precision. Myelination of axons is an essential step to ensure fast action potential propagation by saltatory conduction, a process that requires highly concentrated voltage-gated sodium channels at the nodes of Ranvier. Recent studies suggest that the clustering of sodium channels can influence axonal impulse conduction in both myelinated and unmyelinated fibers, which could have major implications in disease, particularly demyelinating pathology. This comprehensive review summarizes the mechanisms governing the clustering of sodium channels at the peripheral and central nervous system nodes and the specific roles of their clustering in influencing action potential conduction. We further highlight the classical biophysical parameters implicated in conduction timing, followed by a detailed discussion on how sodium channel clustering along unmyelinated axons can impact axonal impulse conduction in both physiological and pathological contexts.
Collapse
Affiliation(s)
- Sean A Freeman
- ICM-GHU Pitié-Salpêtrière, Sorbonne Universités UPMC Univ Paris 06, UMR_S 1127, 75013, Paris, France.
- Inserm U1127, 75013, Paris, France.
- CNRS UMR7225, 75013, Paris, France.
| | - Anne Desmazières
- ICM-GHU Pitié-Salpêtrière, Sorbonne Universités UPMC Univ Paris 06, UMR_S 1127, 75013, Paris, France.
- Inserm U1127, 75013, Paris, France.
- CNRS UMR7225, 75013, Paris, France.
| | - Desdemona Fricker
- ICM-GHU Pitié-Salpêtrière, Sorbonne Universités UPMC Univ Paris 06, UMR_S 1127, 75013, Paris, France.
- Inserm U1127, 75013, Paris, France.
- CNRS UMR7225, 75013, Paris, France.
| | - Catherine Lubetzki
- ICM-GHU Pitié-Salpêtrière, Sorbonne Universités UPMC Univ Paris 06, UMR_S 1127, 75013, Paris, France.
- Inserm U1127, 75013, Paris, France.
- CNRS UMR7225, 75013, Paris, France.
- Assistance Publique-Hôpitaux de Paris, Hôpital Pitié-Salpêtrière, Paris, France.
| | - Nathalie Sol-Foulon
- ICM-GHU Pitié-Salpêtrière, Sorbonne Universités UPMC Univ Paris 06, UMR_S 1127, 75013, Paris, France.
- Inserm U1127, 75013, Paris, France.
- CNRS UMR7225, 75013, Paris, France.
| |
Collapse
|
31
|
Rosenzweig S, Carmichael ST. The axon-glia unit in white matter stroke: mechanisms of damage and recovery. Brain Res 2015; 1623:123-34. [PMID: 25704204 PMCID: PMC4545468 DOI: 10.1016/j.brainres.2015.02.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Accepted: 02/10/2015] [Indexed: 01/07/2023]
Abstract
Approximately one quarter of all strokes in humans occur in white matter, and the progressive nature of white matter lesions often results in severe physical and mental disability. Unlike cortical grey matter stroke, the pathology of white matter stroke revolves around disrupted connectivity and injured axons and glial cells, rather than neuronal cell bodies. Consequently, the mechanisms behind ischemic damage to white matter elements, the regenerative responses of glial cells and their signaling pathways, all differ significantly from those in grey matter. Development of effective therapies for white matter stroke would require an enhanced understanding of the complex cellular and molecular interactions within the white matter, leading to the identification of new therapeutic targets. This review will address the unique properties of the axon-glia unit during white matter stroke, describe the challenging process of promoting effective white matter repair, and discuss recently-identified signaling pathways which may hold potential targets for repair in this disease. This article is part of a Special Issue entitled SI: Cell Interactions In Stroke.
Collapse
Affiliation(s)
- Shira Rosenzweig
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| | - S Thomas Carmichael
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA.
| |
Collapse
|
32
|
Chang KJ, Zollinger DR, Susuki K, Sherman DL, Makara MA, Brophy PJ, Cooper EC, Bennett V, Mohler PJ, Rasband MN. Glial ankyrins facilitate paranodal axoglial junction assembly. Nat Neurosci 2014; 17:1673-81. [PMID: 25362471 PMCID: PMC4260775 DOI: 10.1038/nn.3858] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 10/08/2014] [Indexed: 02/07/2023]
Abstract
Neuron-glia interactions establish functional membrane domains along myelinated axons. These include nodes of Ranvier, paranodal axoglial junctions and juxtaparanodes. Paranodal junctions are the largest vertebrate junctional adhesion complex, and they are essential for rapid saltatory conduction and contribute to assembly and maintenance of nodes. However, the molecular mechanisms underlying paranodal junction assembly are poorly understood. Ankyrins are cytoskeletal scaffolds traditionally associated with Na(+) channel clustering in neurons and are important for membrane domain establishment and maintenance in many cell types. Here we show that ankyrin-B, expressed by Schwann cells, and ankyrin-G, expressed by oligodendrocytes, are highly enriched at the glial side of paranodal junctions where they interact with the essential glial junctional component neurofascin 155. Conditional knockout of ankyrins in oligodendrocytes disrupts paranodal junction assembly and delays nerve conduction during early development in mice. Thus, glial ankyrins function as major scaffolds that facilitate early and efficient paranodal junction assembly in the developing CNS.
Collapse
Affiliation(s)
- Kae-Jiun Chang
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Daniel R. Zollinger
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Keiichiro Susuki
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | - Diane L. Sherman
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, United Kingdom EH16 4SB
| | - Michael A. Makara
- Departments of Physiology and Cell Biology, Medicine, and the Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Peter J. Brophy
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh, United Kingdom EH16 4SB
| | - Edward C. Cooper
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neurology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Vann Bennett
- Department of Cell Biology, Duke University, Durham, NC 27710, USA
| | - Peter J. Mohler
- Departments of Physiology and Cell Biology, Medicine, and the Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Matthew N. Rasband
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
33
|
Devaux JJ. [New insights on the organization of the nodes of Ranvier]. Rev Neurol (Paris) 2014; 170:819-24. [PMID: 25459119 DOI: 10.1016/j.neurol.2014.03.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 03/05/2014] [Indexed: 01/06/2023]
Abstract
Myelin plays a crucial role in the rapid and saltatory conduction of the nerve impulse along myelinated axons. In addition, myelin closely regulates the organization of the axonal compartments. This organization involves several complex mechanisms including axo-glial contact, diffusion barriers, the cytoskeletal network, and the extracellular matrix. In peripheral nerves, the axo-glial contact dictates the formation of the nodes and the clustering of the voltage-gated sodium channels (Nav). The axo-glial contact at nodes implicates adhesion molecules expressed by the Schwann cell (gliomedin and NrCAM), which binds a partner, neurofascin-186, on the axonal side. This complex is essential for the recruitment of ankyrin-G, a cytoskeletal scaffolding protein, which binds and concentrates Nav channels at nodes. The paranodal junctions flanking the nodes also play a complementary function in node formation. These junctions are formed by the association of contactin-1/caspr-1/neurofascin-155 and create a diffusion barrier, which traps proteins at the nodes and dampens their diffusion along the internode. In the central nervous system, the mechanisms of node formation are different and the formation of the paranodal junctions precedes the aggregation of Nav channels at nodes. However, node formation can still happen in absence of paranodal junctions in the CNS. One explanation is that NF186 interacts with components of the extracellular matrix around the node and thereby stabilizes the aggregation of nodal proteins. It is likely that many other proteins are also implicated in the signaling pathways that regulate the differentiation of the axonal compartments. The nature and function of these proteins are yet to be identified.
Collapse
Affiliation(s)
- J J Devaux
- Centre de recherche en neurobiologie et neurophysiologie de Marseille, faculté de médecine secteur Nord, Aix-Marseille université, CNRS-UMR7286, 51, boulevard Pierre-Dramard, CS80011, 13344 Marseille cedex 15, France.
| |
Collapse
|
34
|
Luo S, Jaegle M, Li R, Ehring GR, Meijer D, Levinson SR. The sodium channel isoform transition at developing nodes of ranvier in the peripheral nervous system: Dependence on a Genetic program and myelination-induced cluster formation. J Comp Neurol 2014; 522:4057-73. [DOI: 10.1002/cne.23656] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2014] [Revised: 07/18/2014] [Accepted: 07/18/2014] [Indexed: 11/10/2022]
Affiliation(s)
- Songjiang Luo
- Department of Comprehensive Dentistry; University of Texas Health Science Center; San Antonio Texas 78229
| | - Martine Jaegle
- Department of Cell Biology and Genetics; Erasmus University Rotterdam; 3000DR Rotterdam The Netherlands
| | - Roy Li
- Allergan; Irvine California 92612
| | | | - Dies Meijer
- Department of Cell Biology and Genetics; Erasmus University Rotterdam; 3000DR Rotterdam The Netherlands
- Centre for Neuroregeneration; University of Edinburgh; Edinburgh EH16 4SB United Kingdom
| | - Simon R. Levinson
- Department of Physiology and Biophysics; School of Medicine, University of Colorado Anschutz Medical Campus; Aurora Colorado 80045
| |
Collapse
|
35
|
The node of Ranvier in CNS pathology. Acta Neuropathol 2014; 128:161-75. [PMID: 24913350 PMCID: PMC4102831 DOI: 10.1007/s00401-014-1305-z] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2014] [Revised: 05/27/2014] [Accepted: 05/27/2014] [Indexed: 12/11/2022]
Abstract
Healthy nodes of Ranvier are crucial for action potential propagation along myelinated axons, both in the central and in the peripheral nervous system. Surprisingly, the node of Ranvier has often been neglected when describing CNS disorders, with most pathologies classified simply as being due to neuronal defects in the grey matter or due to oligodendrocyte damage in the white matter. However, recent studies have highlighted changes that occur in pathological conditions at the node of Ranvier, and at the associated paranodal and juxtaparanodal regions where neurons and myelinating glial cells interact. Lengthening of the node of Ranvier, failure of the electrically resistive seal between the myelin and the axon at the paranode, and retraction of myelin to expose voltage-gated K+ channels in the juxtaparanode, may contribute to altering the function of myelinated axons in a wide range of diseases, including stroke, spinal cord injury and multiple sclerosis. Here, we review the principles by which the node of Ranvier operates and its molecular structure, and thus explain how defects at the node and paranode contribute to neurological disorders.
Collapse
|
36
|
Abstract
A membrane barrier important for assembly of the nodes of Ranvier is found at the paranodal junction. This junction is comprised of axonal and glial adhesion molecules linked to the axonal actin–spectrin membrane cytoskeleton through specific adaptors. In this issue, Zhang et al. (2013. J. Cell Biol.http://dx.doi.org/10.1083/jcb.201308116) show that axonal βII spectrin maintains the diffusion barrier at the paranodal junction. Thus, βII spectrin serves to compartmentalize the membrane of myelinated axons at specific locations that are determined either intrinsically (i.e., at the axonal initial segment), or by axoglial contacts (i.e., at the paranodal junction).
Collapse
Affiliation(s)
- Yael Eshed-Eisenbach
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | | |
Collapse
|
37
|
Zhang C, Susuki K, Zollinger DR, Dupree JL, Rasband MN. Membrane domain organization of myelinated axons requires βII spectrin. ACTA ACUST UNITED AC 2014; 203:437-43. [PMID: 24217619 PMCID: PMC3824014 DOI: 10.1083/jcb.201308116] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The spectrin-based cytoskeleton functions as a barrier to restrict axonal proteins, such as juxtaparanodal K+ channels, to specific membrane domains. The precise and remarkable subdivision of myelinated axons into molecularly and functionally distinct membrane domains depends on axoglial junctions that function as barriers. However, the molecular basis of these barriers remains poorly understood. Here, we report that genetic ablation and loss of axonal βII spectrin eradicated the paranodal barrier that normally separates juxtaparanodal K+ channel protein complexes located beneath the myelin sheath from Na+ channels located at nodes of Ranvier. Surprisingly, the K+ channels and their associated proteins redistributed into paranodes where they colocalized with intact Caspr-labeled axoglial junctions. Furthermore, electron microscopic analysis of the junctions showed intact paranodal septate-like junctions. Thus, the paranodal spectrin-based submembranous cytoskeleton comprises the paranodal barriers required for myelinated axon domain organization.
Collapse
Affiliation(s)
- Chuansheng Zhang
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| | | | | | | | | |
Collapse
|
38
|
Contactin-1 regulates myelination and nodal/paranodal domain organization in the central nervous system. Proc Natl Acad Sci U S A 2014; 111:E394-403. [PMID: 24385581 DOI: 10.1073/pnas.1313769110] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Myelin, a multilayered membrane sheath formed by oligodendrocytes around axons in the CNS, enables rapid nerve impulse conduction and sustains neuronal health. The signals exchanged between axons and oligodendrocytes in myelin remain to be fully elucidated. Here we provide genetic evidence for multiple and critical functions of Contactin-1 in central myelin. We document dynamic Contactin-1 expression on oligodendrocytes in vivo, and progressive accumulation at nodes of Ranvier and paranodes during postnatal mouse development. Nodal and paranodal expression stabilized in mature myelin, but overall membranous expression diminished. Contactin-1-deficiency disrupted paranodal junction formation as evidenced by loss of Caspr, mislocalized potassium Kv1.2 channels, and abnormal myelin terminal loops. Reduced numbers and impaired maturation of sodium channel clusters accompanied this phenotype. Histological, electron microscopic, and biochemical analyses uncovered significant hypomyelination in Contactin-1-deficient central nerves, with up to 60% myelin loss. Oligodendrocytes were present in normal numbers, albeit a minor population of neuronal/glial antigen 2-positive (NG2(+)) progenitors lagged in maturation by postnatal day 18, when the mouse null mutation was lethal. Major contributing factors to hypomyelination were defects in the generation and organization of myelin membranes, as judged by electron microscopy and quantitative analysis of oligodendrocyte processes labeled by GFP transgenically expressed from the proteolipid protein promoter. These data reveal that Contactin-1 regulates both myelin formation and organization of nodal and paranodal domains in the CNS. These multiple roles distinguish central Contactin-1 functions from its specific role at paranodes in the periphery, and emphasize mechanistic differences in central and peripheral myelination.
Collapse
|
39
|
Wang S, Young KM. White matter plasticity in adulthood. Neuroscience 2013; 276:148-60. [PMID: 24161723 DOI: 10.1016/j.neuroscience.2013.10.018] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 10/09/2013] [Accepted: 10/10/2013] [Indexed: 01/24/2023]
Abstract
CNS white matter is subject to a novel form of neural plasticity which has been termed "myelin plasticity". It is well established that oligodendrocyte generation and the addition of new myelin internodes continue throughout normal adulthood. These new myelin internodes maybe required for the de novo myelination of previously unmyelinated axons, myelin sheath replacement, or even myelin remodeling. Each process could alter axonal conduction velocity, but to what end? We review the changes that occur within the white matter over the lifetime, the known regulators and mediators of white matter plasticity in the mature CNS, and the physiological role this plasticity may play in CNS function.
Collapse
Affiliation(s)
- S Wang
- Menzies Research Institute Tasmania, University of Tasmania, Hobart 7000, Australia
| | - K M Young
- Menzies Research Institute Tasmania, University of Tasmania, Hobart 7000, Australia.
| |
Collapse
|
40
|
Susuki K, Chang KJ, Zollinger DR, Liu Y, Ogawa Y, Eshed-Eisenbach Y, Dours-Zimmermann MT, Oses-Prieto JA, Burlingame AL, Seidenbecher CI, Zimmermann DR, Oohashi T, Peles E, Rasband MN. Three mechanisms assemble central nervous system nodes of Ranvier. Neuron 2013; 78:469-82. [PMID: 23664614 DOI: 10.1016/j.neuron.2013.03.005] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/04/2013] [Indexed: 11/16/2022]
Abstract
Rapid action potential propagation in myelinated axons requires Na⁺ channel clustering at nodes of Ranvier. However, the mechanism of clustering at CNS nodes remains poorly understood. Here, we show that the assembly of nodes of Ranvier in the CNS involves three mechanisms: a glia-derived extracellular matrix (ECM) complex containing proteoglycans and adhesion molecules that cluster NF186, paranodal axoglial junctions that function as barriers to restrict the position of nodal proteins, and axonal cytoskeletal scaffolds (CSs) that stabilize nodal Na⁺ channels. We show that while mice with a single disrupted mechanism had mostly normal nodes, disruptions of the ECM and paranodal barrier, the ECM and CS, or the paranodal barrier and CS all lead to juvenile lethality, profound motor dysfunction, and significantly reduced Na⁺ channel clustering. Our results demonstrate that ECM, paranodal, and axonal cytoskeletal mechanisms ensure robust CNS nodal Na⁺ channel clustering.
Collapse
Affiliation(s)
- Keiichiro Susuki
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Zoupi L, Markoullis K, Kleopa KA, Karagogeos D. Alterations of juxtaparanodal domains in two rodent models of CNS demyelination. Glia 2013; 61:1236-49. [DOI: 10.1002/glia.22511] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 03/20/2013] [Indexed: 01/15/2023]
Affiliation(s)
| | - Kyriaki Markoullis
- Neuroscience Laboratory and Neurology Clinics; The Cyprus Institute of Neurology and Genetics (CING); P.O. Box 23462, 1683 Nicosia; Cyprus
| | - Kleopas A. Kleopa
- Neuroscience Laboratory and Neurology Clinics; The Cyprus Institute of Neurology and Genetics (CING); P.O. Box 23462, 1683 Nicosia; Cyprus
| | | |
Collapse
|
42
|
Buttermore ED, Thaxton CL, Bhat MA. Organization and maintenance of molecular domains in myelinated axons. J Neurosci Res 2013; 91:603-22. [PMID: 23404451 DOI: 10.1002/jnr.23197] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Revised: 09/19/2012] [Accepted: 11/28/2012] [Indexed: 01/17/2023]
Abstract
Over a century ago, Ramon y Cajal first proposed the idea of a directionality involved in nerve conduction and neuronal communication. Decades later, it was discovered that myelin, produced by glial cells, insulated axons with periodic breaks where nodes of Ranvier (nodes) form to allow for saltatory conduction. In the peripheral nervous system (PNS), Schwann cells are the glia that can either individually myelinate the axon from one neuron or ensheath axons of many neurons. In the central nervous system (CNS), oligodendrocytes are the glia that myelinate axons from different neurons. Review of more recent studies revealed that this myelination created polarized domains adjacent to the nodes. However, the molecular mechanisms responsible for the organization of axonal domains are only now beginning to be elucidated. The molecular domains in myelinated axons include the axon initial segment (AIS), where various ion channels are clustered and action potentials are initiated; the node, where sodium channels are clustered and action potentials are propagated; the paranode, where myelin loops contact with the axolemma; the juxtaparanode (JXP), where delayed-rectifier potassium channels are clustered; and the internode, where myelin is compactly wrapped. Each domain contains a unique subset of proteins critical for the domain's function. However, the roles of these proteins in axonal domain organization are not fully understood. In this review, we highlight recent advances on the molecular nature and functions of some of the components of each axonal domain and their roles in axonal domain organization and maintenance for proper neuronal communication.
Collapse
Affiliation(s)
- Elizabeth D Buttermore
- Curriculum in Neurobiology, University of North Carolina School of Medicine, Chapel Hill, North Carolina, USA
| | | | | |
Collapse
|
43
|
Hayashi A, Kaneko N, Tomihira C, Baba H. Sulfatide decrease in myelin influences formation of the paranodal axo-glial junction and conduction velocity in the sciatic nerve. Glia 2013; 61:466-74. [PMID: 23322453 DOI: 10.1002/glia.22447] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 11/02/2012] [Indexed: 11/11/2022]
Abstract
Cerebroside sulfotransferase (CST) catalyzes the production of sulfatide, which is one of the major glycolipids in myelin. Homozygous CST knockout mice were shown to be completely deficient in sulfatide. They were born healthy but began to display progressive neurological deficits from 6 weeks of age. Severe abnormalities of paranodal regions and changes in axonal ion channel distribution were prominent in both the central and peripheral nervous systems. But whether partial decreases in myelin sulfatide levels influence paranodal formation, as well as nerve conduction velocity (NCV), is largely unknown. To determine the functional significance of sulfatide content in myelin, we performed electrophysiological, morphological, and biochemical analyses using heterozygote, homozygote, and wild-type mouse peripheral nerves and compared the results with individual sulfatide content. NCVs were significantly reduced in homozygote animals compared with wild-type mice. In contrast, these values were markedly varied in individual heterozygote mice. On the basis of NCV values, we divided heterozygous mice into two groups: mice with mild but significant reduction of NCV and those with normal NCV. Teased nerve fibers obtained from individual mouse sciatic nerves were immunostained, and Na(+) channel and Caspr cluster lengths were measured to determine abnormal levels of junctional formation at the paranode. Furthermore, sulfatide content in each sciatic nerve was examined by thin layer chromatography. The results demonstrated significant correlations among sulfatide level, severity of paranodal abnormality, and reduction of NCV. Thus, the fine regulation of myelin sulfatide content by CST is important for normal function of myelinated axons.
Collapse
Affiliation(s)
- Akiko Hayashi
- Department of Molecular Neurobiology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | | | | | | |
Collapse
|
44
|
Chang KJ, Rasband MN. Excitable domains of myelinated nerves: axon initial segments and nodes of Ranvier. CURRENT TOPICS IN MEMBRANES 2013; 72:159-92. [PMID: 24210430 DOI: 10.1016/b978-0-12-417027-8.00005-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Neurons are highly polarized cells. They can be subdivided into at least two structurally and functionally distinct domains: somatodendritic and axonal domains. The somatodendritic domain receives and integrates upstream input signals, and the axonal domain generates and relays outputs in the form of action potentials to the downstream target. Demand for quick response to the harsh surroundings prompted evolution to equip vertebrates' neurons with a remarkable glia-derived structure called myelin. Not only Insulating the axon, myelinating glia also rearrange the axonal components and elaborate functional subdomains along the axon. Proper functioning of all theses domains and subdomains is vital for a normal, efficient nervous system.
Collapse
Affiliation(s)
- Kae-Jiun Chang
- Program in Developmental Biology, Baylor College of Medicine, Houston, Texas, USA
| | | |
Collapse
|
45
|
Rosenbluth J, Bobrowski-Khoury N. Structural bases for central nervous system malfunction in the quaking mouse: dysmyelination in a potential model of schizophrenia. J Neurosci Res 2012; 91:374-81. [PMID: 23224912 DOI: 10.1002/jnr.23167] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 09/20/2012] [Accepted: 10/08/2012] [Indexed: 11/07/2022]
Abstract
The dysmyelinating mouse mutant quaking (qk) is thought to be a model of schizophrenia based on diminution of CNS myelin (Andreone et al., 2007) and downregulation of the Qk gene (Haroutunian et al., 2006) in the brains of schizophrenic patients. The purpose of this study was to identify specific structural defects in the qk mouse CNS that could compromise physiologic function and that in humans might account for some of the cognitive defects characteristic of schizophrenia. Ultrastructural analysis of qk mouse CNS myelinated fibers shows abnormalities in nodal, internodal, and paranodal regions, including marked variation in myelin thickness among neighboring fibers, spotty disruption of paranodal junctions, abnormal distribution of nodal and paranodal ion channel complexes, generalized thinning and incompactness of myelin, and on many axonal profiles complete absence of myelin. These structural defects are likely to cause abnormalities in conduction velocity, synchrony of activation, temporal ordering of signals, and other physiological parameters. We conclude that the structural abnormalities described are likely to be responsible for significant functional impairment both in the qk mouse CNS and in the human CNS with comparable myelin pathology.
Collapse
Affiliation(s)
- J Rosenbluth
- Department of Physiology and Neuroscience, New York University School of Medicine, New York, New York 10016, USA.
| | | |
Collapse
|
46
|
Elliott C, Lindner M, Arthur A, Brennan K, Jarius S, Hussey J, Chan A, Stroet A, Olsson T, Willison H, Barnett SC, Meinl E, Linington C. Functional identification of pathogenic autoantibody responses in patients with multiple sclerosis. ACTA ACUST UNITED AC 2012; 135:1819-33. [PMID: 22561643 PMCID: PMC3359756 DOI: 10.1093/brain/aws105] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pathological and clinical studies implicate antibody-dependent mechanisms in the immunopathogenesis of multiple sclerosis. We tested this hypothesis directly by investigating the ability of patient-derived immunoglobulins to mediate demyelination and axonal injury in vitro. Using a myelinating culture system, we developed a sensitive and reproducible bioassay to detect and quantify these effects and applied this to investigate the pathogenic potential of immunoglobulin G preparations obtained from patients with multiple sclerosis (n = 37), other neurological diseases (n = 10) and healthy control donors (n = 13). This identified complement-dependent demyelinating immunoglobulin G responses in approximately 30% of patients with multiple sclerosis, which in two cases was accompanied by significant complement-dependent antibody mediated axonal loss. No pathogenic immunoglobulin G responses were detected in patients with other neurological disease or healthy controls, indicating that the presence of these demyelinating/axopathic autoantibodies is specific for a subset of patients with multiple sclerosis. Immunofluorescence microscopy revealed immunoglobulin G preparations with demyelinating activity contained antibodies that specifically decorated the surface of myelinating oligodendrocytes and their contiguous myelin sheaths. No other binding was observed indicating that the response is restricted to autoantigens expressed by terminally differentiated myelinating oligodendrocytes. In conclusion, our study identifies axopathic and/or demyelinating autoantibody responses in a subset of patients with multiple sclerosis. This observation underlines the mechanistic heterogeneity of multiple sclerosis and provides a rational explanation why some patients benefit from antibody depleting treatments.
Collapse
Affiliation(s)
- Christina Elliott
- Institute of Immunology, Immunity and Infection, University of Glasgow, Glasgow, G12 8TA, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Rodrigues F, Thuma L, Klämbt C. The regulation of glial-specific splicing of Neurexin IV requires HOW and Cdk12 activity. Development 2012; 139:1765-76. [PMID: 22461565 DOI: 10.1242/dev.074070] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The differentiation of the blood-brain barrier (BBB) is an essential process in the development of a complex nervous system and depends on alternative splicing. In the fly BBB, glial cells establish intensive septate junctions that require the cell-adhesion molecule Neurexin IV. Alternative splicing generates two different Neurexin IV isoforms: Neurexin IV(exon3), which is found in cells that form septate junctions, and Neurexin IV(exon4), which is found in neurons that form no septate junctions. Here, we show that the formation of the BBB depends on the RNA-binding protein HOW (Held out wings), which triggers glial specific splicing of Neurexin IV(exon3). Using a set of splice reporters, we show that one HOW-binding site is needed to include one of the two mutually exclusive exons 3 and 4, whereas binding at the three further motifs is needed to exclude exon 4. The differential splicing is controlled by nuclear access of HOW and can be induced in neurons following expression of nuclear HOW. Using a novel in vivo two-color splicing detector, we then screened for genes required for full HOW activity. This approach identified Cyclin-dependent kinase 12 (Cdk12) and the splicesosomal component Prp40 as major determinants in regulating HOW-dependent splicing of Neurexin IV. Thus, in addition to the control of nuclear localization of HOW, the phosphorylation of the C-terminal domain of the RNA polymerase II by Cdk12 provides an elegant mechanism in regulating timed splicing of newly synthesized mRNA molecules.
Collapse
Affiliation(s)
- Floriano Rodrigues
- Institut für Neurobiologie, Universität Münster, Badestrasse 9, 48149 Münster, Germany
| | | | | |
Collapse
|
48
|
Zhang Y, Bekku Y, Dzhashiashvili Y, Armenti S, Meng X, Sasaki Y, Milbrandt J, Salzer JL. Assembly and maintenance of nodes of ranvier rely on distinct sources of proteins and targeting mechanisms. Neuron 2012; 73:92-107. [PMID: 22243749 DOI: 10.1016/j.neuron.2011.10.016] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2011] [Indexed: 01/29/2023]
Abstract
VIDEO ABSTRACT We have investigated the source(s) and targeting of components to PNS nodes of Ranvier. We show adhesion molecules are freely diffusible within the axon membrane and accumulate at forming nodes from local sources, whereas ion channels and cytoskeletal components are largely immobile and require transport to the node. We further characterize targeting of NF186, an adhesion molecule that pioneers node formation. NF186 redistributes to nascent nodes from a mobile, surface pool. Its initial accumulation and clearance from the internode require extracellular interactions, whereas targeting to mature nodes, i.e., those flanked by paranodal junctions, requires intracellular interactions. After incorporation into the node, NF186 is immobile, stable, and promotes node integrity. Thus, nodes assemble from two sources: adhesion molecules, which initiate assembly, accumulate by diffusion trapping via interactions with Schwann cells, whereas ion channels and cytoskeletal components accumulate via subsequent transport. In mature nodes, components turnover slowly and are replenished via transport.
Collapse
Affiliation(s)
- Yanqing Zhang
- Smilow Neuroscience Program, New York University Langone Medical Center, New York, NY 10016, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Pollard JD, Armati PJ. CIDP - the relevance of recent advances in Schwann cell/axonal neurobiology. J Peripher Nerv Syst 2011; 16:15-23. [PMID: 21504498 DOI: 10.1111/j.1529-8027.2011.00323.x] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Early pathological studies in patients with acute and chronic inflammatory demyelinating neuropathies, and the animal model experimental autoimmune neuritis (EAN) showed similarities in the process of demyelination. These studies focused on compact myelin proteins and peptides as targets of immune attack in Guillain-Barré syndrome (GBS), chronic inflammatory demyelinating polyradiculoneuropathy (CIDP), and EAN. However, serological studies in patients with subsets of GBS highlighted the importance of gangliosides - glycolipids enriched in non-compact Schwann cell regions and the node, paranodal, and internodal axolemma. In the acute motor axonal neuropathy (AMAN) rabbit model, antibodies to the ganglioside GM1 bind in the nodal region, impair Na channel clustering and disturb Schwann cell/axon organisation. Schwann cell neurobiological studies now highlight the importance of adhesion molecules, including neurofascins, gliomedin, contactins, and NrCAM to Schwann cell/axon integrity. Changes to nodal fine structure by immune responses against such molecules may provide a mechanism for reversible conduction failure or block. Recovery of patients with CIDP or multifocal motor neuropathy (MMN) following treatment may sometimes be better explained by reversal of conduction failure than remyelination or regeneration. This review considers the importance of the intricate molecular arrangements at the nodal and paranodal regions in inflammatory neuropathies such as CIDP. Early images of compact myelin stripping and phagocytosis, may have diverted the research focus away from these vital non-compact myelin Schwann cell areas.
Collapse
Affiliation(s)
- John D Pollard
- Brain and Mind Research Institute, Nerve Research Foundation, The University of Sydney, Camperdown, NSW, Australia.
| | | |
Collapse
|
50
|
Thaxton C, Pillai AM, Pribisko AL, Dupree JL, Bhat MA. Nodes of Ranvier act as barriers to restrict invasion of flanking paranodal domains in myelinated axons. Neuron 2011; 69:244-57. [PMID: 21262464 DOI: 10.1016/j.neuron.2010.12.016] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/12/2010] [Indexed: 11/16/2022]
Abstract
Accumulation of voltage-gated sodium (Na(v)) channels at nodes of Ranvier is paramount for action potential propagation along myelinated fibers, yet the mechanisms governing nodal development, organization, and stabilization remain unresolved. Here, we report that genetic ablation of the neuron-specific isoform of Neurofascin (Nfasc(NF¹⁸⁶)) in vivo results in nodal disorganization, including loss of Na(v) channel and ankyrin-G (AnkG) enrichment at nodes in the peripheral nervous system (PNS) and central nervous system (CNS). Interestingly, the presence of paranodal domains failed to rescue nodal organization in the PNS and the CNS. Most importantly, using ultrastructural analysis, we demonstrate that the paranodal domains invade the nodal space in Nfasc(NF¹⁸⁶) mutant axons and occlude node formation. Our results suggest that Nfasc(NF¹⁸⁶)-dependent assembly of the nodal complex acts as a molecular boundary to restrict the movement of flanking paranodal domains into the nodal area, thereby facilitating the stereotypic axonal domain organization and saltatory conduction along myelinated axons.
Collapse
Affiliation(s)
- Courtney Thaxton
- Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, Chapel Hill, NC 27599-7545, USA
| | | | | | | | | |
Collapse
|