151
|
Thomason EJ, Escalante M, Osterhout DJ, Fuss B. The oligodendrocyte growth cone and its actin cytoskeleton: A fundamental element for progenitor cell migration and CNS myelination. Glia 2019; 68:1329-1346. [PMID: 31696982 DOI: 10.1002/glia.23735] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 09/26/2019] [Accepted: 10/01/2019] [Indexed: 01/06/2023]
Abstract
Cells of the oligodendrocyte (OLG) lineage engage in highly motile behaviors that are crucial for effective central nervous system (CNS) myelination. These behaviors include the guided migration of OLG progenitor cells (OPCs), the surveying of local environments by cellular processes extending from differentiating and pre-myelinating OLGs, and during the process of active myelin wrapping, the forward movement of the leading edge of the myelin sheath's inner tongue along the axon. Almost all of these motile behaviors are driven by actin cytoskeletal dynamics initiated within a lamellipodial structure that is located at the tip of cellular OLG/OPC processes and is structurally as well as functionally similar to the neuronal growth cone. Accordingly, coordinated stoichiometries of actin filament (F-actin) assembly and disassembly at these OLG/OPC growth cones have been implicated in directing process outgrowth and guidance, and the initiation of myelination. Nonetheless, the functional importance of the OLG/OPC growth cone still remains to be fully understood, and, as a unique aspect of actin cytoskeletal dynamics, F-actin depolymerization and disassembly start to predominate at the transition from myelination initiation to myelin wrapping. This review provides an overview of the current knowledge about OLG/OPC growth cones, and it proposes a model in which actin cytoskeletal dynamics in OLG/OPC growth cones are a main driver for morphological transformations and motile behaviors. Remarkably, these activities, at least at the later stages of OLG maturation, may be regulated independently from the transcriptional gene expression changes typically associated with CNS myelination.
Collapse
Affiliation(s)
- Elizabeth J Thomason
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| | - Miguel Escalante
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia.,Departamento de Toxicología, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Donna J Osterhout
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York
| | - Babette Fuss
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Richmond, Virginia
| |
Collapse
|
152
|
Thomas L, Pasquini LA. Galectin-3 Exerts a Pro-differentiating and Pro-myelinating Effect Within a Temporal Window Spanning Precursors and Pre-oligodendrocytes: Insights into the Mechanisms of Action. Mol Neurobiol 2019; 57:976-987. [PMID: 31654317 DOI: 10.1007/s12035-019-01787-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 09/12/2019] [Indexed: 11/29/2022]
Abstract
Oligodendrocytes (OLG) are the cells resident in the CNS responsible for myelination. OLG undergo a succession of morphological and molecular changes along several maturational stages. Galectin-3 (Gal-3) is a 25- to 35-KDa protein belonging to the family of carbohydrate-binding galectins, which bind to glycoconjugates containing β-galactosides. Gal-3 lacks a specific receptor and its binding is thus rather unspecific, as it depends on the cellular environment and the repertoire of glycomolecules at the time when Gal-3 is present. Our previous work revealed that recombinant Gal-3 (rGal-3)-treated OLG showed accelerated differentiation, evidenced by an increase in the number of mature cells to the detriment of immature ones and accelerated actin cytoskeleton dynamics. These changes were a consequence of rGal-3 influence on Akt, Erk 1/2, and β-catenin signaling pathways. Considering this previous evidence, the aim of this study was to identify the temporal window of rGal-3 action on the OLG lineage to induce OLG maturation by using specific single pulses of rGal-3 over the different maturational stages of OLG, and to unravel its main direct targets promoting OLG differentiation by mass spectrometry analysis. Our results reveal a key temporal window spanning between OPC and pre-OLG states in which rGal-3 action promotes OLG differentiation, and identify several targets for rGal-3 binding including proteins related to the cytoskeleton, signaling pathways, metabolism and intracellular trafficking, among others. These results highlight the relevance of Gal-3 in signaling pathways regulating oligodendroglial differentiation and support a potential therapeutic role for rGal-3 in demyelinating diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Laura Thomas
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, Institute of Chemistry and Biological Physicochemistry (IQUIFIB), University of Buenos Aires and National Research Council (CONICET), Buenos Aires, Argentina
| | - Laura Andrea Pasquini
- Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junín 956, C1113, Buenos Aires, Argentina.
| |
Collapse
|
153
|
Djannatian M, Timmler S, Arends M, Luckner M, Weil MT, Alexopoulos I, Snaidero N, Schmid B, Misgeld T, Möbius W, Schifferer M, Peles E, Simons M. Two adhesive systems cooperatively regulate axon ensheathment and myelin growth in the CNS. Nat Commun 2019; 10:4794. [PMID: 31641127 PMCID: PMC6805957 DOI: 10.1038/s41467-019-12789-z] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Accepted: 09/27/2019] [Indexed: 01/06/2023] Open
Abstract
Central nervous system myelin is a multilayered membrane produced by oligodendrocytes to increase neural processing speed and efficiency, but the molecular mechanisms underlying axonal selection and myelin wrapping are unknown. Here, using combined morphological and molecular analyses in mice and zebrafish, we show that adhesion molecules of the paranodal and the internodal segment work synergistically using overlapping functions to regulate axonal interaction and myelin wrapping. In the absence of these adhesive systems, axonal recognition by myelin is impaired with myelin growing on top of previously myelinated fibers, around neuronal cell bodies and above nodes of Ranvier. In addition, myelin wrapping is disturbed with the leading edge moving away from the axon and in between previously formed layers. These data show how two adhesive systems function together to guide axonal ensheathment and myelin wrapping, and provide a mechanistic understanding of how the spatial organization of myelin is achieved.
Collapse
Affiliation(s)
- Minou Djannatian
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Sebastian Timmler
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Martina Arends
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Manja Luckner
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Marie-Theres Weil
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075, Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
| | - Ioannis Alexopoulos
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Nicolas Snaidero
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Bettina Schmid
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Thomas Misgeld
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075, Göttingen, Germany
- Center for Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Göttingen, Germany
- Electron Microscopy Core Unit, Max Planck Institute of Experimental Medicine, 37075, Göttingen, Germany
| | - Martina Schifferer
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany
| | - Elior Peles
- Department of Molecular Cell Biology, The Weizmann Institute of Science, Rehovot, Israel
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
- Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
- Max Planck Institute of Experimental Medicine, Göttingen, Germany.
| |
Collapse
|
154
|
Stadelmann C, Timmler S, Barrantes-Freer A, Simons M. Myelin in the Central Nervous System: Structure, Function, and Pathology. Physiol Rev 2019; 99:1381-1431. [PMID: 31066630 DOI: 10.1152/physrev.00031.2018] [Citation(s) in RCA: 394] [Impact Index Per Article: 65.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Oligodendrocytes generate multiple layers of myelin membrane around axons of the central nervous system to enable fast and efficient nerve conduction. Until recently, saltatory nerve conduction was considered the only purpose of myelin, but it is now clear that myelin has more functions. In fact, myelinating oligodendrocytes are embedded in a vast network of interconnected glial and neuronal cells, and increasing evidence supports an active role of oligodendrocytes within this assembly, for example, by providing metabolic support to neurons, by regulating ion and water homeostasis, and by adapting to activity-dependent neuronal signals. The molecular complexity governing these interactions requires an in-depth molecular understanding of how oligodendrocytes and axons interact and how they generate, maintain, and remodel their myelin sheaths. This review deals with the biology of myelin, the expanded relationship of myelin with its underlying axons and the neighboring cells, and its disturbances in various diseases such as multiple sclerosis, acute disseminated encephalomyelitis, and neuromyelitis optica spectrum disorders. Furthermore, we will highlight how specific interactions between astrocytes, oligodendrocytes, and microglia contribute to demyelination in hereditary white matter pathologies.
Collapse
Affiliation(s)
- Christine Stadelmann
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Sebastian Timmler
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Alonso Barrantes-Freer
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| | - Mikael Simons
- Institute of Neuropathology, University Medical Center Göttingen , Göttingen , Germany ; Institute of Neuronal Cell Biology, Technical University Munich , Munich , Germany ; German Center for Neurodegenerative Diseases (DZNE), Munich , Germany ; Department of Neuropathology, University Medical Center Leipzig , Leipzig , Germany ; Munich Cluster of Systems Neurology (SyNergy), Munich , Germany ; and Max Planck Institute of Experimental Medicine, Göttingen , Germany
| |
Collapse
|
155
|
Stability and flexibility of full-length human oligodendrocytic QKI6. BMC Res Notes 2019; 12:609. [PMID: 31547849 PMCID: PMC6757426 DOI: 10.1186/s13104-019-4629-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/10/2019] [Indexed: 11/15/2022] Open
Abstract
Objective Oligodendrocytes account for myelination in the central nervous system. During myelin compaction, key proteins are translated in the vicinity of the myelin membrane, requiring targeted mRNA transport. Quaking isoform 6 (QKI6) is a STAR domain-containing RNA transport protein, which binds a conserved motif in the 3′-UTR of certain mRNAs, affecting the translation of myelination-involved proteins. RNA binding has been earlier structurally characterized, but information about full-length QKI6 conformation is lacking. Based on known domains and structure predicitons, we expected full-length QKI6 to be flexible and carry disordered regions. Hence, we carried out biophysical and structural characterization of human QKI6. Results We expressed and purified full-length QKI6 and characterized it using mass spectrometry, light scattering, small-angle X-ray scattering, and circular dichroism spectroscopy. QKI6 was monodisperse, folded, and mostly dimeric, being oxidation-sensitive. The C-terminal tail was intrinsically disordered, as predicted. In the absence of RNA, the RNA-binding subdomain is likely to present major flexibility. In thermal stability assays, a double sequential unfolding behaviour was observed in the presence of phosphate, which may interact with the RNA-binding domain. The results confirm the flexibility and partial disorder of QKI6, which may be functionally relevant.
Collapse
|
156
|
Abstract
Microtubules are critical for the extension of oligodendrocyte processes and myelin deposition, yet our knowledge of their microtubule biogenesis is limited. In this issue of Cell, Fu et al. (2019) identify an oligodendrocyte-enriched microtubule regulator that promotes microtubule growth from Golgi outposts and controls myelin sheath elongation, linking microtubule cytoarchitecture and myelination in the CNS.
Collapse
Affiliation(s)
- Antonina Roll-Mecak
- Cell Biology and Biophysics Unit, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA; Biochemistry and Biophysics Center, National Heart, Lung and Blood Institute, NIH, Bethesda, MD, USA.
| |
Collapse
|
157
|
Temporal and partial inhibition of GLI1 in neural stem cells (NSCs) results in the early maturation of NSC derived oligodendrocytes in vitro. Stem Cell Res Ther 2019; 10:272. [PMID: 31455382 PMCID: PMC6712625 DOI: 10.1186/s13287-019-1374-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 11/30/2022] Open
Abstract
Background Oligodendrocytes are a type of glial cells that synthesize the myelin sheath around the axons and are critical for the nerve conduction in the CNS. Oligodendrocyte death and defects are the leading causes of several myelin disorders such as multiple sclerosis, progressive multifocal leukoencephalopathy, periventricular leukomalacia, and several leukodystrophies. Temporal activation of the Sonic Hedgehog (SHH) pathway is critical for the generation of oligodendrocyte progenitors, and their differentiation and maturation in the brain and spinal cord during embryonic development in mammals. Methods Our protocol utilized adherent cultures of human induced pluripotent stem cells (iPSC) and human embryonic stem cells (hESCs) with a green fluorescent protein (GFP) reporter knocked into one allele of the OLIG2 gene locus, dual SMAD inhibition, and transient partial inhibition of glioma-associated oncogene 1 (GLI1) by the small molecule GANT61 during the formation of the SOX2/PAX6-positive neural stem cells (NSCs). The SHH pathway was later restimulated by a Smoothened agonist purmorphamine to induce the generation of OLIG2 glial precursors. One hundred ninety-two individual oligodendrocyte precursor cells (OPCs) from GANT61 and control group were analyzed by single-cell RNA sequencing (RNA-Seq). Results We demonstrate here that transient and partial inhibition of the SHH pathway transcription factor GLI1 in NSCs by a small molecule inhibitor GANT61 was found to generate OPCs that were more migratory and could differentiate earlier toward myelin-producing oligodendrocytes. Single-cell transcriptomic analysis (RNA-Seq) showed that GANT61-NSC-derived oligodendrocyte precursor cells (OPCs) had differential activation of some of the genes in the cytoskeleton rearrangement pathways that are involved in OPC motility and induction of maturation. At the protein level, this was also associated with higher levels of myelin-specific genes in the GANT61 group compared to controls. GANT61-NSC-derived OPCs were functional and could generate compact myelin in vitro and in vivo after transplantation in myelin-deficient shiverer mice. Conclusions This is a small molecule-based in vitro protocol that leads to the faster generation of functional oligodendrocytes. The development of protocols that lead to efficient and faster differentiation of oligodendrocytes from progenitors provides important advances toward the development of autologous neural stem cell-based therapies using human iPSCs. Electronic supplementary material The online version of this article (10.1186/s13287-019-1374-y) contains supplementary material, which is available to authorized users.
Collapse
|
158
|
de Rosa V, Secondo A, Pannaccione A, Ciccone R, Formisano L, Guida N, Crispino R, Fico A, Polishchuk R, D'Aniello A, Annunziato L, Boscia F. D-Aspartate treatment attenuates myelin damage and stimulates myelin repair. EMBO Mol Med 2019; 11:emmm.201809278. [PMID: 30559305 PMCID: PMC6328990 DOI: 10.15252/emmm.201809278] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Glutamate signaling may orchestrate oligodendrocyte precursor cell (OPC) development and myelin regeneration through the activation of glutamate receptors at OPC‐neuron synapses. D‐Aspartate is a D‐amino acid exerting modulatory actions at glutamatergic synapses. Chronic administration of D‐Aspartate has been proposed as therapeutic treatment in diseases related to myelin dysfunction and NMDA receptors hypofunction, including schizophrenia and cognitive deficits. Here, we show, by using an in vivo remyelination model, that administration of D‐Aspartate during remyelination improved motor coordination, accelerated myelin recovery, and significantly increased the number of small‐diameter myelinated axons. Chronically administered during demyelination, D‐Aspartate also attenuated myelin loss and inflammation. Interestingly, D‐Aspartate exposure stimulated OPC maturation and accelerated developmental myelination in organotypic cerebellar slices. D‐Aspartate promoting effects on OPC maturation involved the activation of glutamate transporters, AMPA and NMDA receptors, and the Na+/Ca2+ exchanger NCX3. While blocking NMDA or NCX3 significantly prevented D‐Aspartate‐induced [Ca2+]i oscillations, blocking AMPA and glutamate transporters prevented both the initial and oscillatory [Ca2+]i response as well as D‐Aspartate‐induced inward currents in OPC. Our findings reveal that D‐Aspartate treatment may represent a novel strategy for promoting myelin recovery.
Collapse
Affiliation(s)
- Valeria de Rosa
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Agnese Secondo
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Anna Pannaccione
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Roselia Ciccone
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | - Luigi Formisano
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | | | - Roberta Crispino
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
| | - Annalisa Fico
- Institute of Genetics and Biophysics "A. Buzzati-Traverso", Consiglio Nazionale delle Ricerche, Naples, Italy
| | - Roman Polishchuk
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
| | - Antimo D'Aniello
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| | | | - Francesca Boscia
- Division of Pharmacology, Department of Neuroscience, Reproductive and Dentistry Sciences, School of Medicine, Federico II University of Naples, Naples, Italy
| |
Collapse
|
159
|
Nocita E, Del Giovane A, Tiberi M, Boccuni L, Fiorelli D, Sposato C, Romano E, Basoli F, Trombetta M, Rainer A, Traversa E, Ragnini-Wilson A. EGFR/ErbB Inhibition Promotes OPC Maturation up to Axon Engagement by Co-Regulating PIP2 and MBP. Cells 2019; 8:cells8080844. [PMID: 31390799 PMCID: PMC6721729 DOI: 10.3390/cells8080844] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 08/04/2019] [Accepted: 08/05/2019] [Indexed: 12/11/2022] Open
Abstract
Remyelination in the adult brain relies on the reactivation of the Neuronal Precursor Cell (NPC) niche and differentiation into Oligodendrocyte Precursor Cells (OPCs) as well as on OPC maturation into myelinating oligodendrocytes (OLs). These two distinct phases in OL development are defined by transcriptional and morphological changes. How this differentiation program is controlled remains unclear. We used two drugs that stimulate myelin basic protein (MBP) expression (Clobetasol and Gefitinib) alone or combined with epidermal growth factor receptor (EGFR) or Retinoid X Receptor gamma (RXRγ) gene silencing to decode the receptor signaling required for OPC differentiation in myelinating OLs. Electrospun polystyrene (PS) microfibers were used as synthetic axons to study drug efficacy on fiber engagement. We show that EGFR inhibition per se stimulates MBP expression and increases Clobetasol efficacy in OPC differentiation. Consistent with this, Clobetasol and Gefitinib co-treatment, by co-regulating RXRγ, MBP and phosphatidylinositol 4,5-bisphosphate (PIP2) levels, maximizes synthetic axon engagement. Conversely, RXRγ gene silencing reduces the ability of the drugs to promote MBP expression. This work provides a view of how EGFR/ErbB inhibition controls OPC differentiation and indicates the combination of Clobetasol and Gefitinib as a potent remyelination-enhancing treatment.
Collapse
Affiliation(s)
- Emanuela Nocita
- NeurotechIT Laboratory, Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Alice Del Giovane
- NeurotechIT Laboratory, Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Marta Tiberi
- NeurotechIT Laboratory, Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Laura Boccuni
- NeurotechIT Laboratory, Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Denise Fiorelli
- NeurotechIT Laboratory, Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Carola Sposato
- NeurotechIT Laboratory, Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Elena Romano
- Advanced Microscopy Center, Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy
| | - Francesco Basoli
- Department of Engineering, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Marcella Trombetta
- Department of Engineering, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Alberto Rainer
- Department of Engineering, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Enrico Traversa
- School of Materials and Energy, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Antonella Ragnini-Wilson
- NeurotechIT Laboratory, Department of Biology, University of Rome "Tor Vergata", 00133 Rome, Italy.
| |
Collapse
|
160
|
Cyfip1 haploinsufficient rats show white matter changes, myelin thinning, abnormal oligodendrocytes and behavioural inflexibility. Nat Commun 2019; 10:3455. [PMID: 31371763 PMCID: PMC6671959 DOI: 10.1038/s41467-019-11119-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Accepted: 06/20/2019] [Indexed: 11/30/2022] Open
Abstract
The biological basis of the increased risk for psychiatric disorders seen in 15q11.2 copy number deletion is unknown. Previous work has shown disturbances in white matter tracts in human carriers of the deletion. Here, in a novel rat model, we recapitulated low dosage of the candidate risk gene CYFIP1 present within the 15q11.2 interval. Using diffusion tensor imaging, we first showed extensive white matter changes in Cyfip1 mutant rats, which were most pronounced in the corpus callosum and external capsule. Transmission electron microscopy showed that these changes were associated with thinning of the myelin sheath in the corpus callosum. Myelin thinning was independent of changes in axon number or diameter but was associated with effects on mature oligodendrocytes, including aberrant intracellular distribution of myelin basic protein. Finally, we demonstrated effects on cognitive phenotypes sensitive to both disruptions in myelin and callosal circuitry. People with a genetic deletion of the 15q11.2 locus are at increased risk for psychiatric disorders and white matter disturbances, but the gene(s) responsible are unclear. Here, the authors show that low dosage of CYFIP1, present in the human 15q11.2 region, alters white matter structure and cognition in rats.
Collapse
|
161
|
Puhl DL, D'Amato AR, Gilbert RJ. Challenges of gene delivery to the central nervous system and the growing use of biomaterial vectors. Brain Res Bull 2019; 150:216-230. [PMID: 31173859 PMCID: PMC8284997 DOI: 10.1016/j.brainresbull.2019.05.024] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 05/08/2019] [Accepted: 05/29/2019] [Indexed: 12/18/2022]
Abstract
Gene therapy is a promising form of treatment for those suffering from neurological disorders or central nervous system (CNS) injury, however, obstacles remain that limit its translational potential. The CNS is protected by the blood brain barrier, and this barrier blocks genes from traversing into the CNS if administered outside of the CNS. Viral and non-viral gene delivery vehicles, commonly referred to as vectors, are modified to enhance delivery efficiency to target locations in the CNS. Still, there are few gene therapy approaches approved by the FDA for CNS disease or injury treatment. The lack of viable clinical approaches is due, in part, to the unpredictable nature of many vector systems. In particular, safety concerns exist with the use of viral vectors for CNS gene delivery. To seek some alternatives to viral vectors, development of new non-viral, biomaterial vectors is occurring at a rapid rate. This review discusses the challenges of delivering various forms of genetic material to the CNS, the use and limitations of current viral vector delivery systems, and the use of non-viral, biomaterial vectors for CNS applications.
Collapse
Affiliation(s)
- Devan L Puhl
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, New York, 12180, United States; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th Street, Troy, New York, 12180, United States.
| | - Anthony R D'Amato
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, New York, 12180, United States; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th Street, Troy, New York, 12180, United States.
| | - Ryan J Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, New York, 12180, United States; Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 1623 15th Street, Troy, New York, 12180, United States.
| |
Collapse
|
162
|
Kasuga Y, Fudge AD, Zhang Y, Li H. Characterization of a long noncoding RNA Pcdh17it as a novel marker for immature premyelinating oligodendrocytes. Glia 2019; 67:2166-2177. [PMID: 31328332 DOI: 10.1002/glia.23684] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 07/04/2019] [Accepted: 07/05/2019] [Indexed: 01/12/2023]
Abstract
Oligodendrocyte precursors (OPs) proliferate and differentiate into oligodendrocytes (OLs) during postnatal development and into adulthood in the central nervous system (CNS). Following the initiation of differentiation, OPs give rise to immature, premyelinating OLs, which undergo further differentiation and mature into myelin-forming OLs. We identified an immature OL-specific long noncoding RNA, named Pcdh17it. Through co-localization analysis and morphological characterization of OLs, we found that Pcdh17it is a specific marker for newly born immature OLs in the developing and adult forebrain of mice, and we used this new marker to analyze OL generation over the lifespan of mice. Pcdh17it is an effective tool for monitoring newly born OLs in adult brain, allowing detailed study of the dynamics of OP differentiation into OLs in the normal and pathological CNS.
Collapse
Affiliation(s)
- Yusuke Kasuga
- Wolfson Institute for Biomedical Research, University College London, London, UK
- Department of Life Sciences, Graduate School of Arts and Sciences, University of Tokyo, Tokyo, Japan
| | - Alexander D Fudge
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Yumeng Zhang
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Huiliang Li
- Wolfson Institute for Biomedical Research, University College London, London, UK
| |
Collapse
|
163
|
Villoslada P, Vila G, Colafrancesco V, Moreno B, Fernandez-Diez B, Vazquez R, Pertsovskaya I, Zubizarreta I, Pulido-Valdeolivas I, Messeguer J, Vendrell-Navarro G, Frade JM, López-Sánchez N, Teixido M, Giralt E, Masso M, Dugas JC, Leonoudakis D, Lariosa-Willingham KD, Steinman L, Messeguer A. Axonal and Myelin Neuroprotection by the Peptoid BN201 in Brain Inflammation. Neurotherapeutics 2019; 16:808-827. [PMID: 30815844 PMCID: PMC6694325 DOI: 10.1007/s13311-019-00717-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The development of neuroprotective therapies is a sought-after goal. By screening combinatorial chemical libraries using in vitro assays, we identified the small molecule BN201 that promotes the survival of cultured neural cells when subjected to oxidative stress or when deprived of trophic factors. Moreover, BN201 promotes neuronal differentiation, the differentiation of precursor cells to mature oligodendrocytes in vitro, and the myelination of new axons. BN201 modulates several kinases participating in the insulin growth factor 1 pathway including serum-glucocorticoid kinase and midkine, inducing the phosphorylation of NDRG1 and the translocation of the transcription factor Foxo3 to the cytoplasm. In vivo, BN201 prevents axonal and neuronal loss, and it promotes remyelination in models of multiple sclerosis, chemically induced demyelination, and glaucoma. In summary, we provide a new promising strategy to promote neuroaxonal survival and remyelination, potentially preventing disability in brain diseases.
Collapse
Affiliation(s)
- Pablo Villoslada
- Center for Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Casanova 145, Centre Cellex 3A, 08036, Barcelona, Spain.
| | - Gemma Vila
- Center for Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Casanova 145, Centre Cellex 3A, 08036, Barcelona, Spain
| | - Valeria Colafrancesco
- Center for Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Casanova 145, Centre Cellex 3A, 08036, Barcelona, Spain
| | - Beatriz Moreno
- Center for Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Casanova 145, Centre Cellex 3A, 08036, Barcelona, Spain
| | - Begoña Fernandez-Diez
- Center for Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Casanova 145, Centre Cellex 3A, 08036, Barcelona, Spain
| | - Raquel Vazquez
- Center for Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Casanova 145, Centre Cellex 3A, 08036, Barcelona, Spain
| | - Inna Pertsovskaya
- Center for Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Casanova 145, Centre Cellex 3A, 08036, Barcelona, Spain
| | - Irati Zubizarreta
- Center for Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Casanova 145, Centre Cellex 3A, 08036, Barcelona, Spain
| | - Irene Pulido-Valdeolivas
- Center for Neuroimmunology, Institut d'Investigacions Biomediques August Pi Sunyer, Casanova 145, Centre Cellex 3A, 08036, Barcelona, Spain
| | - Joaquin Messeguer
- Institute for Advanced Chemistry of Catalonia, Consejo Superior de Investigaciones Cientificas, Barcelona, 08034, Spain
| | - Gloria Vendrell-Navarro
- Institute for Advanced Chemistry of Catalonia, Consejo Superior de Investigaciones Cientificas, Barcelona, 08034, Spain
| | - Jose Maria Frade
- Instituto Cajal, Consejo Superior de Investigaciones Cientificas, Madrid, 28002, Spain
| | - Noelia López-Sánchez
- Instituto Cajal, Consejo Superior de Investigaciones Cientificas, Madrid, 28002, Spain
| | | | - Ernest Giralt
- Institute for Research in Biomedicine, Barcelona, 08028, Spain
| | - Mar Masso
- Bionure Farma SL, Barcelona, 94025, Spain
| | | | | | | | | | - Angel Messeguer
- Institute for Advanced Chemistry of Catalonia, Consejo Superior de Investigaciones Cientificas, Barcelona, 08034, Spain
| |
Collapse
|
164
|
Ommer A, Figlia G, Pereira JA, Datwyler AL, Gerber J, DeGeer J, Lalli G, Suter U. Ral GTPases in Schwann cells promote radial axonal sorting in the peripheral nervous system. J Cell Biol 2019; 218:2350-2369. [PMID: 31201267 PMCID: PMC6605813 DOI: 10.1083/jcb.201811150] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/03/2019] [Accepted: 05/15/2019] [Indexed: 12/11/2022] Open
Abstract
Small GTPases of the Rho and Ras families are important regulators of Schwann cell biology. The Ras-like GTPases RalA and RalB act downstream of Ras in malignant peripheral nerve sheath tumors. However, the physiological role of Ral proteins in Schwann cell development is unknown. Using transgenic mice with ablation of one or both Ral genes, we report that Ral GTPases are crucial for axonal radial sorting. While lack of only one Ral GTPase was dispensable for early peripheral nerve development, ablation of both RalA and RalB resulted in persistent radial sorting defects, associated with hallmarks of deficits in Schwann cell process formation and maintenance. In agreement, ex vivo-cultured Ral-deficient Schwann cells were impaired in process extension and the formation of lamellipodia. Our data indicate further that RalA contributes to Schwann cell process extensions through the exocyst complex, a known effector of Ral GTPases, consistent with an exocyst-mediated function of Ral GTPases in Schwann cells.
Collapse
Affiliation(s)
- Andrea Ommer
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Gianluca Figlia
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Jorge A Pereira
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Anna Lena Datwyler
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Joanne Gerber
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Jonathan DeGeer
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Giovanna Lalli
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Ueli Suter
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
165
|
Fricano-Kugler C, Gordon A, Shin G, Gao K, Nguyen J, Berg J, Starks M, Geschwind DH. CYFIP1 overexpression increases fear response in mice but does not affect social or repetitive behavioral phenotypes. Mol Autism 2019; 10:25. [PMID: 31198525 PMCID: PMC6555997 DOI: 10.1186/s13229-019-0278-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 05/22/2019] [Indexed: 12/28/2022] Open
Abstract
Background CYFIP1, a protein that interacts with FMRP and regulates protein synthesis and actin dynamics, is overexpressed in Dup15q syndrome as well as autism spectrum disorder (ASD). While CYFIP1 heterozygosity has been rigorously studied due to its loss in 15q11.2 deletion, Prader-Willi and Angelman syndrome, the effects of CYFIP1 overexpression, as is observed in patients with CYFIP1 duplication, are less well understood. Methods We developed and validated a mouse model of human CYFIP1 overexpression (CYFIP1 OE) using qPCR and western blot analysis. We performed a large battery of behavior testing on these mice, including ultrasonic vocalizations, three-chamber social assay, home-cage behavior, Y-maze, elevated plus maze, open field test, Morris water maze, fear conditioning, prepulse inhibition, and the hot plate assay. We also performed RNA sequencing and analysis on the basolateral amygdala. Results Extensive behavioral testing in CYFIP1 OE mice reveals no changes in the core behaviors related to ASD: social interactions and repetitive behaviors. However, we did observe mild learning deficits and an exaggerated fear response. Using RNA sequencing of the basolateral amygdala, a region associated with fear response, we observed changes in pathways related to cytoskeletal regulation, oligodendrocytes, and myelination. We also identified GABA-A subunit composition changes in basolateral amygdala neurons, which are essential components of the neural fear conditioning circuit. Conclusion Overall, this research identifies the behavioral and molecular consequences of CYFIP1 overexpression and how they contribute to the variable phenotype seen in Dup15q syndrome and in ASD patients with excess CYFIP1.
Collapse
Affiliation(s)
- Catherine Fricano-Kugler
- Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Aaron Gordon
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Grace Shin
- Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Kun Gao
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Jade Nguyen
- Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Jamee Berg
- Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
| | - Mary Starks
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Daniel H. Geschwind
- Program in Neurobehavioral Genetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
- Center for Autism Research and Treatment, Semel Institute, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095 USA
| |
Collapse
|
166
|
Bankston AN, Forston MD, Howard RM, Andres KR, Smith AE, Ohri SS, Bates ML, Bunge MB, Whittemore SR. Autophagy is essential for oligodendrocyte differentiation, survival, and proper myelination. Glia 2019; 67:1745-1759. [PMID: 31162728 DOI: 10.1002/glia.23646] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 05/01/2019] [Accepted: 05/21/2019] [Indexed: 12/12/2022]
Abstract
Deficient myelination, the spiral wrapping of highly specialized membrane around axons, causes severe neurological disorders. Maturation of oligodendrocyte progenitor cells (OPC) to myelinating oligodendrocytes (OL), the sole providers of central nervous system (CNS) myelin, is tightly regulated and involves extensive morphological changes. Here, we present evidence that autophagy, the targeted isolation of cytoplasm and organelles by the double-membrane autophagosome for lysosomal degradation, is essential for OPC/OL differentiation, survival, and proper myelin development. A marked increase in autophagic activity coincides with OL differentiation, with OL processes having the greatest increase in autophagic flux. Multiple lines of evidence indicate that autophagosomes form in developing myelin sheathes before trafficking from myelin to the OL soma. Mice with conditional OPC/OL-specific deletion of the essential autophagy gene Atg5 beginning on postnatal Day 5 develop a rapid tremor and die around postnatal Day 12. Further analysis revealed apoptotic death of OPCs, reduced differentiation, and reduced myelination. Surviving Atg5-/- OLs failed to produce proper myelin structure. In vitro, pharmacological inhibition of autophagy in OPC/dorsal root ganglion (DRG) co-cultures blocked myelination, producing OLs surrounded by many short processes. Conversely, autophagy stimulation enhanced myelination. These results implicate autophagy as a key regulator of OPC survival, maturation, and proper myelination. Autophagy may provide an attractive target to promote both OL survival and subsequent myelin repair after injury.
Collapse
Affiliation(s)
- Andrew N Bankston
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Michael D Forston
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Anatomical Sciences & Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky
| | - Russell M Howard
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Kariena R Andres
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Allison E Smith
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Sujata Saraswat Ohri
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky
| | - Margaret L Bates
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida
| | - Mary B Bunge
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, Florida.,Department of Cell Biology and Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida
| | - Scott R Whittemore
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky.,Department of Anatomical Sciences & Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky
| |
Collapse
|
167
|
Fields RD, Dutta DJ. Treadmilling Model for Plasticity of the Myelin Sheath. Trends Neurosci 2019; 42:443-447. [PMID: 31078309 DOI: 10.1016/j.tins.2019.04.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/09/2019] [Accepted: 04/12/2019] [Indexed: 12/22/2022]
Abstract
We propose a mechanism for myelin plasticity that would complement synaptic plasticity by adjusting conduction velocity for optimal spike-time arrival. In the proposed treadmilling model, myelin sheath thickness is a dynamic balance between the rates of new myelin deposited adjacent to the axon and removal of the outermost layer.
Collapse
Affiliation(s)
- R Douglas Fields
- Section on Nervous System Development and Plasticity, The Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USA.
| | - Dipankar J Dutta
- Section on Nervous System Development and Plasticity, The Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD 20892, USA
| |
Collapse
|
168
|
Zakaria M, Ferent J, Hristovska I, Laouarem Y, Zahaf A, Kassoussi A, Mayeur ME, Pascual O, Charron F, Traiffort E. The Shh receptor Boc is important for myelin formation and repair. Development 2019; 146:146/9/dev172502. [PMID: 31048318 DOI: 10.1242/dev.172502] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 03/28/2019] [Indexed: 12/25/2022]
Abstract
Myelination leads to the formation of myelin sheaths surrounding neuronal axons and is crucial for function, plasticity and repair of the central nervous system (CNS). It relies on the interaction of the axons and the oligodendrocytes: the glial cells producing CNS myelin. Here, we have investigated the role of a crucial component of the Sonic hedgehog (Shh) signalling pathway, the co-receptor Boc, in developmental and repairing myelination. During development, Boc mutant mice display a transient decrease in oligodendroglial cell density together with delayed myelination. Despite recovery of oligodendroglial cells at later stages, adult mutants still exhibit a lower production of myelin basic protein correlated with a significant decrease in the calibre of callosal axons and a reduced amount of the neurofilament NF-M. During myelin repair, the altered OPC differentiation observed in the mutant is reminiscent of the phenotype observed after blockade of Shh signalling. In addition, Boc mutant microglia/macrophages unexpectedly exhibit the apparent inability to transition from a highly to a faintly ramified morphology in vivo Altogether, these results identify Boc as an important component of myelin formation and repair.
Collapse
Affiliation(s)
- Mary Zakaria
- INSERM-University Paris-Sud/Paris-Saclay; Diseases and Hormones of the Nervous System, U1195, 80 rue du Général Leclerc, F-94276, Le Kremlin-Bicêtre, France
| | - Julien Ferent
- IRCM, Molecular Biology of Neural Development, 110 Pine Avenue West, Montreal, Quebec H2W 1R7, Canada; Department of Medicine, University of Montreal, Montreal, Quebec, Canada; McGill University, Montreal, Quebec, Canada
| | - Ines Hristovska
- Institut NeuroMyoGène CNRS UMR 5310-INSERM U1217-Université Claude Bernard Lyon 1, Faculté de Médecine et de Pharmacie 69008 Lyon, France
| | - Yousra Laouarem
- INSERM-University Paris-Sud/Paris-Saclay; Diseases and Hormones of the Nervous System, U1195, 80 rue du Général Leclerc, F-94276, Le Kremlin-Bicêtre, France
| | - Amina Zahaf
- INSERM-University Paris-Sud/Paris-Saclay; Diseases and Hormones of the Nervous System, U1195, 80 rue du Général Leclerc, F-94276, Le Kremlin-Bicêtre, France
| | - Abdelmoumen Kassoussi
- INSERM-University Paris-Sud/Paris-Saclay; Diseases and Hormones of the Nervous System, U1195, 80 rue du Général Leclerc, F-94276, Le Kremlin-Bicêtre, France
| | - Marie-Eve Mayeur
- Institut NeuroMyoGène CNRS UMR 5310-INSERM U1217-Université Claude Bernard Lyon 1, Faculté de Médecine et de Pharmacie 69008 Lyon, France
| | - Olivier Pascual
- Institut NeuroMyoGène CNRS UMR 5310-INSERM U1217-Université Claude Bernard Lyon 1, Faculté de Médecine et de Pharmacie 69008 Lyon, France
| | - Frederic Charron
- IRCM, Molecular Biology of Neural Development, 110 Pine Avenue West, Montreal, Quebec H2W 1R7, Canada; Department of Medicine, University of Montreal, Montreal, Quebec, Canada; McGill University, Montreal, Quebec, Canada
| | - Elisabeth Traiffort
- INSERM-University Paris-Sud/Paris-Saclay; Diseases and Hormones of the Nervous System, U1195, 80 rue du Général Leclerc, F-94276, Le Kremlin-Bicêtre, France
| |
Collapse
|
169
|
Sakers K, Eroglu C. Control of neural development and function by glial neuroligins. Curr Opin Neurobiol 2019; 57:163-170. [PMID: 30991196 DOI: 10.1016/j.conb.2019.03.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 11/16/2022]
Abstract
Neuroligins are a family of cell adhesion molecules, which are best known for their functions as postsynaptic components of the trans-synaptic neurexin-neuroligin complexes. Neuroligins are highly conserved across evolution with important roles in the formation, maturation and function of synaptic structures. Mutations in the genes that encode for neuroligins have been linked to a number of neurodevelopmental disorders such as autism and schizophrenia, which stem from synaptic pathologies. Owing to their essential functions in regulating synaptic connectivity and their link to synaptic dysfunction in disease, previous studies on neuroligins have focused on neurons. Yet a recent work reveals that neuroligins are also expressed in the central nervous system by glial cells, such as astrocytes and oligodendrocytes, and perform important roles in controlling synaptic connectivity in a non-cell autonomous manner. In this review, we will highlight these recent findings demonstrating the important roles of glial neuroligins in regulating the development and connectivity of healthy and diseased brains.
Collapse
Affiliation(s)
- Kristina Sakers
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, United States
| | - Cagla Eroglu
- Department of Cell Biology, Duke University Medical Center, Durham, NC 27710, United States; Department of Neurobiology, Duke University Medical Center, Durham, NC 27710, United States; Duke Institute for Brain Sciences (DIBS), Durham, NC 27710, United States; Regeneration Next Initiative, Duke University, Durham, NC 27710, United States.
| |
Collapse
|
170
|
McCanney GA, McGrath MA, Otto TD, Burchmore R, Yates EA, Bavington CD, Willison HJ, Turnbull JE, Barnett SC. Low sulfated heparins target multiple proteins for central nervous system repair. Glia 2019; 67:668-687. [PMID: 30585359 PMCID: PMC6492281 DOI: 10.1002/glia.23562] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 10/05/2018] [Accepted: 10/17/2018] [Indexed: 01/01/2023]
Abstract
The lack of endogenous repair following spinal cord injury (SCI) accounts for the frequent permanent deficits for which effective treatments are absent. Previously, we demonstrated that low sulfated modified heparin mimetics (LS-mHeps) attenuate astrocytosis, suggesting they may represent a novel therapeutic approach. mHeps are glycomolecules with structural similarities to resident heparan sulfates (HS), which modulate cell signaling by both sequestering ligands, and acting as cofactors in the formation of ligand-receptor complexes. To explore whether mHeps can affect the myelination and neurite outgrowth necessary for repair after SCI, we created lesioned or demyelinated neural cell co-cultures and exposed them with a panel of mHeps with varying degrees and positions of their sulfate moieties. LS-mHep7 enhanced neurite outgrowth and myelination, whereas highly sulfated mHeps (HS-mHeps) had attenuating effects. LS-mHeps had no effects on myelination or neurite extension in developing, uninjured myelinating cultures, suggesting they might exert their proregenerating effects by modulating or sequestering inhibitory factors secreted after injury. To investigate this, we examined conditioned media from cultures using chemokine arrays and conducted an unbiased proteomics approach by applying TMT-LC/MS to mHep7 affinity purified conditioned media from these cultures. Multiple protein factors reported to play a role in damage or repair mechanisms were identified, including amyloid betaA4. Amyloid beta peptide (1-42) was validated as an important candidate by treating myelination cultures and shown to inhibit myelination. Thus, we propose that LS-mHeps exert multiple beneficial effects on mechanisms supporting enhanced repair, and represent novel candidates as therapeutics for CNS damage.
Collapse
Affiliation(s)
- George A. McCanney
- Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgowUK
| | - Michael A. McGrath
- Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgowUK
| | - Thomas D. Otto
- Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgowUK
| | - Richard Burchmore
- Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgowUK
| | - Edwin A. Yates
- Department of BiochemistryInstitute of Integrative Biology, University of LiverpoolLiverpoolUK
| | - Charles D. Bavington
- GlycoMar Limited, European Centre for Marine Biotechnology, Dunstaffnage Marine LaboratoryObanArgyllScotland, UK
| | - Hugh J. Willison
- Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgowUK
| | - Jeremy E. Turnbull
- Department of BiochemistryInstitute of Integrative Biology, University of LiverpoolLiverpoolUK
| | - Susan C. Barnett
- Institute of Infection, Immunity, and Inflammation, College of Medical, Veterinary and Life Sciences, University of GlasgowGlasgowUK
| |
Collapse
|
171
|
Piroli GG, Manuel AM, Patel T, Walla MD, Shi L, Lanci SA, Wang J, Galloway A, Ortinski PI, Smith DS, Frizzell N. Identification of Novel Protein Targets of Dimethyl Fumarate Modification in Neurons and Astrocytes Reveals Actions Independent of Nrf2 Stabilization. Mol Cell Proteomics 2019; 18:504-519. [PMID: 30587509 PMCID: PMC6398201 DOI: 10.1074/mcp.ra118.000922] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 12/01/2018] [Indexed: 12/15/2022] Open
Abstract
The fumarate ester dimethyl fumarate (DMF) has been introduced recently as a treatment for relapsing remitting multiple sclerosis (RRMS), a chronic inflammatory condition that results in neuronal demyelination and axonal loss. DMF is known to act by depleting intracellular glutathione and modifying thiols on Keap1 protein, resulting in the stabilization of the transcription factor Nrf2, which in turn induces the expression of antioxidant response element genes. We have previously shown that DMF reacts with a wide range of protein thiols, suggesting that the complete mechanisms of action of DMF are unknown. Here, we investigated other intracellular thiol residues that may also be irreversibly modified by DMF in neurons and astrocytes. Using mass spectrometry, we identified 24 novel proteins that were modified by DMF in neurons and astrocytes, including cofilin-1, tubulin and collapsin response mediator protein 2 (CRMP2). Using an in vitro functional assay, we demonstrated that DMF-modified cofilin-1 loses its activity and generates less monomeric actin, potentially inhibiting its cytoskeletal remodeling activity, which could be beneficial in the modulation of myelination during RRMS. DMF modification of tubulin did not significantly impact axonal lysosomal trafficking. We found that the oxygen consumption rate of N1E-115 neurons and the levels of proteins related to mitochondrial energy production were only slightly affected by the highest doses of DMF, confirming that DMF treatment does not impair cellular respiratory function. In summary, our work provides new insights into the mechanisms supporting the neuroprotective and remyelination benefits associated with DMF treatment in addition to the antioxidant response by Nrf2.
Collapse
Affiliation(s)
- Gerardo G Piroli
- From the ‡Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, South Carolina 29209
| | - Allison M Manuel
- From the ‡Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, South Carolina 29209
| | - Tulsi Patel
- From the ‡Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, South Carolina 29209
| | - Michael D Walla
- §Mass Spectrometry Center, Department of Chemistry & Biochemistry, University of South Carolina, Columbia, South Carolina 29205
| | - Liang Shi
- ¶Department of Biological Sciences, University of South Carolina, Columbia, South Carolina 29205
| | - Scott A Lanci
- From the ‡Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, South Carolina 29209
| | - Jingtian Wang
- From the ‡Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, South Carolina 29209
| | - Ashley Galloway
- From the ‡Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, South Carolina 29209
| | - Pavel I Ortinski
- From the ‡Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, South Carolina 29209
| | - Deanna S Smith
- ¶Department of Biological Sciences, University of South Carolina, Columbia, South Carolina 29205
| | - Norma Frizzell
- From the ‡Department of Pharmacology, Physiology & Neuroscience, School of Medicine, University of South Carolina, Columbia, South Carolina 29209;
| |
Collapse
|
172
|
Malheiro AR, Correia B, Ferreira da Silva T, Bessa-Neto D, Van Veldhoven PP, Brites P. Leukodystrophy caused by plasmalogen deficiency rescued by glyceryl 1-myristyl ether treatment. Brain Pathol 2019; 29:622-639. [PMID: 30667116 DOI: 10.1111/bpa.12710] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 01/16/2019] [Indexed: 12/29/2022] Open
Abstract
Plasmalogens are the most abundant form of ether phospholipids in myelin and their deficiency causes Rhizomelic Chondrodysplasia Punctata (RCDP), a severe developmental disorder. Using the Gnpat-knockout (KO) mouse as a model of RCDP, we determined the consequences of a plasmalogen deficiency during myelination and myelin homeostasis in the central nervous system (CNS). We unraveled that the lack of plasmalogens causes a generalized hypomyelination in several CNS regions including the optic nerve, corpus callosum and spinal cord. The defect in myelin content evolved to a progressive demyelination concomitant with generalized astrocytosis and white matter-selective microgliosis. Oligodendrocyte precursor cells (OPC) and mature oligodendrocytes were abundant in the CNS of Gnpat KO mice during the active period of demyelination. Axonal loss was minimal in plasmalogen-deficient mice, although axonal damage was observed in spinal cords from aged Gnpat KO mice. Characterization of the plasmalogen-deficient myelin identified myelin basic protein and septin 7 as early markers of dysmyelination, whereas myelin-associated glycoprotein was associated with the active demyelination phase. Using in vitro myelination assays, we unraveled that the intrinsic capacity of oligodendrocytes to ensheath and initiate membrane wrapping requires plasmalogens. The defect in plasmalogens was rescued with glyceryl 1-myristyl ether [1-O-tetradecyl glycerol (1-O-TDG)], a novel alternative precursor in the plasmalogen biosynthesis pathway. 1-O-TDG treatment rescued myelination in plasmalogen-deficient oligodendrocytes and in mutant mice. Our results demonstrate the importance of plasmalogens for oligodendrocyte function and myelin assembly, and identified a novel strategy to promote myelination in nervous tissue.
Collapse
Affiliation(s)
- Ana R Malheiro
- Neurolipid Biology, Instituto de Investigação e Inovação em Saúde - i3S, Instituto de Biologia Molecular e Celular - IBMC e Universidade do Porto, Porto, Portugal.,ICBAS, Instituto Ciências Biomédicas Abel Salazar, Porto, Portugal
| | - Barbara Correia
- Neurolipid Biology, Instituto de Investigação e Inovação em Saúde - i3S, Instituto de Biologia Molecular e Celular - IBMC e Universidade do Porto, Porto, Portugal
| | - Tiago Ferreira da Silva
- Neurolipid Biology, Instituto de Investigação e Inovação em Saúde - i3S, Instituto de Biologia Molecular e Celular - IBMC e Universidade do Porto, Porto, Portugal
| | - Diogo Bessa-Neto
- Neurolipid Biology, Instituto de Investigação e Inovação em Saúde - i3S, Instituto de Biologia Molecular e Celular - IBMC e Universidade do Porto, Porto, Portugal
| | - Paul P Van Veldhoven
- Laboratory of Lipid Biochemistry and Protein Interactions (LIPIT), KU Leuven, Leuven, Belgium
| | - Pedro Brites
- Neurolipid Biology, Instituto de Investigação e Inovação em Saúde - i3S, Instituto de Biologia Molecular e Celular - IBMC e Universidade do Porto, Porto, Portugal
| |
Collapse
|
173
|
Tsai E, Casaccia P. Mechano-modulation of nuclear events regulating oligodendrocyte progenitor gene expression. Glia 2019; 67:1229-1239. [PMID: 30734358 DOI: 10.1002/glia.23595] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 01/03/2019] [Accepted: 01/14/2019] [Indexed: 12/20/2022]
Abstract
Oligodendrocytes differentiate from oligodendrocyte progenitor cells (OPCs) in response to distinct extracellular signals. This process requires changes in gene expression resulting from the interplay between transcription factors and epigenetic modulators. Extracellular signals include chemical and physical stimuli. This review focuses on the signaling mechanisms activated in oligodendrocyte progenitors in response to mechanical forces. Of particular interest is a better understanding on how these forces are transduced into the OPC nuclei and subsequently reshape their epigenetic landscape. Here we will introduce the concept of epigenetic regulation of gene expression, first in general and then focusing on the oligodendrocyte lineage. We will then review the current literature on mechano-transduction in distinct cell types, followed by pathways identified in myelinating oligodendrocytes and their progenitors. Overall, the reader will be provided with a comprehensive review of the signaling pathways which allow oligodendrocyte progenitors to "sense" physical forces and transduce them into patterns of gene expression.
Collapse
Affiliation(s)
- Eric Tsai
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Patrizia Casaccia
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, New York.,Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Biology and Biochemistry, Neuroscience Initiative at the Advanced Science Research Center of the Graduate Center of The City University of New York, New York
| |
Collapse
|
174
|
Erwig MS, Patzig J, Steyer AM, Dibaj P, Heilmann M, Heilmann I, Jung RB, Kusch K, Möbius W, Jahn O, Nave KA, Werner HB. Anillin facilitates septin assembly to prevent pathological outfoldings of central nervous system myelin. eLife 2019; 8:43888. [PMID: 30672734 PMCID: PMC6344079 DOI: 10.7554/elife.43888] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 01/11/2019] [Indexed: 12/15/2022] Open
Abstract
Myelin serves as an axonal insulator that facilitates rapid nerve conduction along axons. By transmission electron microscopy, a healthy myelin sheath comprises compacted membrane layers spiraling around the cross-sectioned axon. Previously we identified the assembly of septin filaments in the innermost non-compacted myelin layer as one of the latest steps of myelin maturation in the central nervous system (CNS) (Patzig et al., 2016). Here we show that loss of the cytoskeletal adaptor protein anillin (ANLN) from oligodendrocytes disrupts myelin septin assembly, thereby causing the emergence of pathological myelin outfoldings. Since myelin outfoldings are a poorly understood hallmark of myelin disease and brain aging we assessed axon/myelin-units in Anln-mutant mice by focused ion beam-scanning electron microscopy (FIB-SEM); myelin outfoldings were three-dimensionally reconstructed as large sheets of multiple compact membrane layers. We suggest that anillin-dependent assembly of septin filaments scaffolds mature myelin sheaths, facilitating rapid nerve conduction in the healthy CNS.
Collapse
Affiliation(s)
- Michelle S Erwig
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Julia Patzig
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Anna M Steyer
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Electron Microscopy Core Unit, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Center for Nanoscale Microscopy and Molecular Physiology of the Brain, Göttingen, Germany
| | - Payam Dibaj
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Mareike Heilmann
- Department of Cellular Biochemistry, Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Ingo Heilmann
- Department of Cellular Biochemistry, Institute of Biochemistry and Biotechnology, Martin-Luther-University Halle-Wittenberg, Halle, Germany
| | - Ramona B Jung
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Kathrin Kusch
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany.,Electron Microscopy Core Unit, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Olaf Jahn
- Proteomics Group, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| |
Collapse
|
175
|
Weil MT, Schulz-Ëberlin G, Mukherjee C, Kuo-Elsner WP, Schäfer I, Müller C, Simons M. Isolation and Culture of Oligodendrocytes. Methods Mol Biol 2019; 1936:79-95. [PMID: 30820894 DOI: 10.1007/978-1-4939-9072-6_5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Primary cultures of brain-derived rodent cells are widely used to study molecular and cellular mechanisms in neurobiology. In this chapter, we describe methods of purifying and culturing oligodendroglial cells from mouse perinatal brains. In addition, we describe methods of coculturing the purified oligodendrocytes with neurons. When prepared and cultured according to these protocols, many essential aspects of the biology of oligodendrocytes, such as their proliferation, differentiation, and myelination, can be studied in culture.
Collapse
Affiliation(s)
- Marie-Theres Weil
- Max Planck Institute of Experimental Medicine, Goettingen, Germany
- Center Nanoscale Microscopy and Molecular Physiology of the Brain (CNMPB), Goettingen, Germany
- AbbVie Germany GmbH & Co. KG, Knollstrasse, Ludwigshafen, Germany
| | | | - Chaitali Mukherjee
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany
- German Center for Neurodegenerative Disease (DZNE), Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Wen Ping Kuo-Elsner
- Department of Biology, Molecular Cell Biology, Johannes Gutenberg University, Mainz, Germany
| | - Isabelle Schäfer
- Institute of Physiology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Christina Müller
- Institute of Physiology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Mikael Simons
- Max Planck Institute of Experimental Medicine, Goettingen, Germany.
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany.
- German Center for Neurodegenerative Disease (DZNE), Munich, Germany.
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
| |
Collapse
|
176
|
Kounakis K, Tavernarakis N. The Cytoskeleton as a Modulator of Aging and Neurodegeneration. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1178:227-245. [PMID: 31493230 DOI: 10.1007/978-3-030-25650-0_12] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The cytoskeleton consists of filamentous protein polymers that form organized structures, contributing to a multitude of cell life aspects. It includes three types of polymers: the actin microfilaments, the microtubules and the intermediate filaments. Decades of research have implicated the cytoskeleton in processes that regulate cellular and organismal aging, as well as neurodegeneration associated with injury or neurodegenerative disease, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic Lateral Sclerosis, or Charcot Marie Tooth disease. Here, we provide a brief overview of cytoskeletal structure and function, and discuss experimental evidence linking cytoskeletal function and dynamics with aging and neurodegeneration.
Collapse
Affiliation(s)
- Konstantinos Kounakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece.,Department of Basic Sciences, Medical School, University of Crete, Heraklion, Greece
| | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece. .,Department of Basic Sciences, Medical School, University of Crete, Heraklion, Greece.
| |
Collapse
|
177
|
Thomas L, Pasquini LA. Galectin-3 prospects as a therapeutic agent for multiple sclerosis. Neural Regen Res 2019; 14:1380-1382. [PMID: 30964061 PMCID: PMC6524501 DOI: 10.4103/1673-5374.253521] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Laura Thomas
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, Institute of Chemistry Biological Physicochemistry (IQUIFIB), University of Buenos Aires and National Research Council (CONICET), Buenos Aires, Argentina
| | - Laura Andrea Pasquini
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, Institute of Chemistry Biological Physicochemistry (IQUIFIB), University of Buenos Aires and National Research Council (CONICET), Buenos Aires, Argentina
| |
Collapse
|
178
|
Vasistha NA, Johnstone M, Barton SK, Mayerl SE, Thangaraj Selvaraj B, Thomson PA, Dando O, Grünewald E, Alloza C, Bastin ME, Livesey MR, Economides K, Magnani D, Makedonopolou P, Burr K, Story DJ, Blackwood DHR, Wyllie DJA, McIntosh AM, Millar JK, ffrench-Constant C, Hardingham GE, Lawrie SM, Chandran S. Familial t(1;11) translocation is associated with disruption of white matter structural integrity and oligodendrocyte-myelin dysfunction. Mol Psychiatry 2019; 24:1641-1654. [PMID: 31481758 PMCID: PMC6814440 DOI: 10.1038/s41380-019-0505-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 05/31/2019] [Accepted: 06/26/2019] [Indexed: 12/12/2022]
Abstract
Although the underlying neurobiology of major mental illness (MMI) remains unknown, emerging evidence implicates a role for oligodendrocyte-myelin abnormalities. Here, we took advantage of a large family carrying a balanced t(1;11) translocation, which substantially increases risk of MMI, to undertake both diffusion tensor imaging and cellular studies to evaluate the consequences of the t(1;11) translocation on white matter structural integrity and oligodendrocyte-myelin biology. This translocation disrupts among others the DISC1 gene which plays a crucial role in brain development. We show that translocation-carrying patients display significant disruption of white matter integrity compared with familial controls. At a cellular level, we observe dysregulation of key pathways controlling oligodendrocyte development and morphogenesis in induced pluripotent stem cell (iPSC) derived case oligodendrocytes. This is associated with reduced proliferation and a stunted morphology in vitro. Further, myelin internodes in a humanized mouse model that recapitulates the human translocation as well as after transplantation of t(1;11) oligodendrocyte progenitors were significantly reduced when compared with controls. Thus we provide evidence that the t(1;11) translocation has biological effects at both the systems and cellular level that together suggest oligodendrocyte-myelin dysfunction.
Collapse
Affiliation(s)
- Navneet A. Vasistha
- 0000 0004 1936 7988grid.4305.2Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK ,0000 0004 1936 7988grid.4305.2MRC Centre for Regenerative Medicine, The University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU UK ,0000 0004 4905 7710grid.475408.aCentre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, GKVK - Post, Bellary Road, Bangalore, 560065 India ,0000 0001 0674 042Xgrid.5254.6Present Address: Biotech Research and Innovation Centre, Ole Maaløes Vej 5, Copenhagen, N 2200 Denmark
| | - Mandy Johnstone
- 0000 0004 1936 7988grid.4305.2Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK ,0000 0004 1936 7988grid.4305.2Division of Psychiatry, The University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, EH10 5HF UK
| | - Samantha K. Barton
- 0000 0004 1936 7988grid.4305.2Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK ,0000 0004 1936 7988grid.4305.2MRC Centre for Regenerative Medicine, The University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU UK ,0000 0004 1936 7988grid.4305.2UK Dementia Research Institute at Edinburgh, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK
| | - Steffen E. Mayerl
- 0000 0004 1936 7988grid.4305.2MRC Centre for Regenerative Medicine, The University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU UK
| | - Bhuvaneish Thangaraj Selvaraj
- 0000 0004 1936 7988grid.4305.2Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK ,0000 0004 1936 7988grid.4305.2MRC Centre for Regenerative Medicine, The University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU UK ,0000 0004 1936 7988grid.4305.2UK Dementia Research Institute at Edinburgh, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK
| | - Pippa A. Thomson
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU UK
| | - Owen Dando
- 0000 0004 1936 7988grid.4305.2UK Dementia Research Institute at Edinburgh, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK ,0000 0004 1936 7988grid.4305.2Centre for Discovery Brain Sciences, The University of Edinburgh, Hugh Robson Building, 15 George Square, Edinburgh, EH8 9XD UK
| | - Ellen Grünewald
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU UK
| | - Clara Alloza
- 0000 0004 1936 7988grid.4305.2Division of Psychiatry, The University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, EH10 5HF UK
| | - Mark E. Bastin
- 0000 0004 1936 7988grid.4305.2Division of Psychiatry, The University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, EH10 5HF UK
| | - Matthew R. Livesey
- 0000 0004 1936 7988grid.4305.2Centre for Discovery Brain Sciences, The University of Edinburgh, Hugh Robson Building, 15 George Square, Edinburgh, EH8 9XD UK
| | | | - Dario Magnani
- 0000 0004 1936 7988grid.4305.2Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK ,0000 0004 1936 7988grid.4305.2MRC Centre for Regenerative Medicine, The University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU UK ,0000 0004 1936 7988grid.4305.2UK Dementia Research Institute at Edinburgh, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK
| | - Paraskevi Makedonopolou
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU UK
| | - Karen Burr
- 0000 0004 1936 7988grid.4305.2Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK ,0000 0004 1936 7988grid.4305.2MRC Centre for Regenerative Medicine, The University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU UK ,0000 0004 1936 7988grid.4305.2UK Dementia Research Institute at Edinburgh, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK
| | - David J. Story
- 0000 0004 1936 7988grid.4305.2Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK ,0000 0004 1936 7988grid.4305.2MRC Centre for Regenerative Medicine, The University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU UK ,0000 0004 1936 7988grid.4305.2UK Dementia Research Institute at Edinburgh, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK
| | - Douglas H. R. Blackwood
- 0000 0004 1936 7988grid.4305.2Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK ,0000 0004 1936 7988grid.4305.2Division of Psychiatry, The University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, EH10 5HF UK
| | - David J. A. Wyllie
- 0000 0004 4905 7710grid.475408.aCentre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, GKVK - Post, Bellary Road, Bangalore, 560065 India ,0000 0004 1936 7988grid.4305.2Centre for Discovery Brain Sciences, The University of Edinburgh, Hugh Robson Building, 15 George Square, Edinburgh, EH8 9XD UK
| | - Andrew M. McIntosh
- 0000 0004 1936 7988grid.4305.2Division of Psychiatry, The University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, EH10 5HF UK
| | - J. Kirsty Millar
- Centre for Genomic and Experimental Medicine, Institute of Genetics and Molecular Medicine, The University of Edinburgh, Western General Hospital, Crewe Road, Edinburgh, EH4 2XU UK
| | - Charles ffrench-Constant
- 0000 0004 1936 7988grid.4305.2MRC Centre for Regenerative Medicine, The University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU UK
| | - Giles E. Hardingham
- 0000 0004 1936 7988grid.4305.2UK Dementia Research Institute at Edinburgh, The University of Edinburgh, Chancellor’s Building, 49 Little France Crescent, Edinburgh, EH16 4SB UK ,0000 0004 1936 7988grid.4305.2Centre for Discovery Brain Sciences, The University of Edinburgh, Hugh Robson Building, 15 George Square, Edinburgh, EH8 9XD UK
| | - Stephen M. Lawrie
- 0000 0004 1936 7988grid.4305.2Division of Psychiatry, The University of Edinburgh, Royal Edinburgh Hospital, Edinburgh, EH10 5HF UK
| | - Siddharthan Chandran
- Centre for Clinical Brain Sciences, The University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK. .,MRC Centre for Regenerative Medicine, The University of Edinburgh, 5 Little France Drive, Edinburgh, EH16 4UU, UK. .,Centre for Brain Development and Repair, Institute for Stem Cell Biology and Regenerative Medicine, GKVK - Post, Bellary Road, Bangalore, 560065, India. .,UK Dementia Research Institute at Edinburgh, The University of Edinburgh, Chancellor's Building, 49 Little France Crescent, Edinburgh, EH16 4SB, UK.
| |
Collapse
|
179
|
Seixas AI, Azevedo MM, Paes de Faria J, Fernandes D, Mendes Pinto I, Relvas JB. Evolvability of the actin cytoskeleton in oligodendrocytes during central nervous system development and aging. Cell Mol Life Sci 2019; 76:1-11. [PMID: 30302529 PMCID: PMC11105620 DOI: 10.1007/s00018-018-2915-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 08/24/2018] [Accepted: 09/04/2018] [Indexed: 01/23/2023]
Abstract
The organization of actin filaments into a wide range of subcellular structures is a defining feature of cell shape and dynamics, important for tissue development and homeostasis. Nervous system function requires morphological and functional plasticity of neurons and glial cells, which is largely determined by the dynamic reorganization of the actin cytoskeleton in response to intrinsic and extracellular signals. Oligodendrocytes are specialized glia that extend multiple actin-based protrusions to form the multilayered myelin membrane that spirally wraps around axons, increasing conduction speed and promoting long-term axonal integrity. Myelination is a remarkable biological paradigm in development, and maintenance of myelin is essential for a healthy adult nervous system. In this review, we discuss how structure and dynamics of the actin cytoskeleton is a defining feature of myelinating oligodendrocytes' biology and function. We also review "old and new" concepts to reflect on the potential role of the cytoskeleton in balancing life and death of myelin membranes and oligodendrocytes in the aging central nervous system.
Collapse
Affiliation(s)
- Ana Isabel Seixas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.
- IBMC - Instituto de Biologia Molecular e Celular, Porto, Portugal.
| | - Maria Manuela Azevedo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Joana Paes de Faria
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Diogo Fernandes
- Instituto Superior Técnico, Universidade de Lisboa, Lisbon, Portugal
- International Iberian Nanotechnology Laboratory - INL, Braga, Portugal
| | - Inês Mendes Pinto
- International Iberian Nanotechnology Laboratory - INL, Braga, Portugal
| | - João Bettencourt Relvas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Porto, Portugal
- The Discoveries Centre for Regeneration and Precision Medicine, Porto Campus, Porto, Portugal
| |
Collapse
|
180
|
Cytoskeletal Regulation of Oligodendrocyte Differentiation and Myelination. J Neurosci 2018; 37:7797-7799. [PMID: 28821599 DOI: 10.1523/jneurosci.1398-17.2017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 07/09/2017] [Accepted: 07/11/2017] [Indexed: 11/21/2022] Open
|
181
|
Sun LO, Mulinyawe SB, Collins HY, Ibrahim A, Li Q, Simon DJ, Tessier-Lavigne M, Barres BA. Spatiotemporal Control of CNS Myelination by Oligodendrocyte Programmed Cell Death through the TFEB-PUMA Axis. Cell 2018; 175:1811-1826.e21. [PMID: 30503207 PMCID: PMC6295215 DOI: 10.1016/j.cell.2018.10.044] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 08/01/2018] [Accepted: 10/19/2018] [Indexed: 12/16/2022]
Abstract
Nervous system function depends on proper myelination for insulation and critical trophic support for axons. Myelination is tightly regulated spatially and temporally, but how it is controlled molecularly remains largely unknown. Here, we identified key molecular mechanisms governing the regional and temporal specificity of CNS myelination. We show that transcription factor EB (TFEB) is highly expressed by differentiating oligodendrocytes and that its loss causes precocious and ectopic myelination in many parts of the murine brain. TFEB functions cell-autonomously through PUMA induction and Bax-Bak activation to promote programmed cell death of a subset of premyelinating oligodendrocytes, allowing selective elimination of oligodendrocytes in normally unmyelinated brain regions. This pathway is conserved across diverse brain areas and is critical for myelination timing. Our findings define an oligodendrocyte-intrinsic mechanism underlying the spatiotemporal specificity of CNS myelination, shedding light on how myelinating glia sculpt the nervous system during development.
Collapse
Affiliation(s)
- Lu O Sun
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Sara B Mulinyawe
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hannah Y Collins
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Adiljan Ibrahim
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Qingyun Li
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - David J Simon
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | | | - Ben A Barres
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
182
|
Daly DT, Ariel M. A novel cerebellar commissure and other myelinated axons in the Purkinje cell layer of a pond turtle (Trachemys scripta elegans). J Comp Neurol 2018; 526:2802-2823. [PMID: 30173417 DOI: 10.1002/cne.24528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 07/23/2018] [Accepted: 07/30/2018] [Indexed: 11/10/2022]
Abstract
Parallel fibers in the molecular layer of the vertebrate cerebellum mediate slow spike conduction in the transverse plane. In contrast, electrophysiological recordings have indicated that rapid spike conduction exists between the lateral regions of the cerebellar cortex of the red-ear pond turtle (Trachemys scripta). The anatomical basis for this commissure is now examined in that species using neuronal tracing techniques. Fluorescently tagged dextrans and lipophilic carbocyanine dyes placed in one lateral edge of this nonfoliated cortex are transported across the midline of living brains in vitro and along the axonal membranes of fixed tissues, respectively. Surprisingly, the labeled commissural axons traversed the cortex within the Purkinje cell layer, and not in the white matter of the molecular layer or the white matter below the granule cell layer. Unlike thin parallel fibers that exhibit characteristic varicosities, this commissure is composed of smooth axons of large diameter that also extend beyond the cerebellar cortex via the cerebellar peduncles. Double labeling with myelin basic protein antibody demonstrated that these commissural axons are ensheathed with myelin. In contrast to this transverse pathway, an orthogonal myelinated tract was observed along the cerebellar midline. The connections of this transverse commissure with the lateral cerebellum, the vestibular nuclear complex, and the cochlear vestibular ganglia indicate that this commissure plays a role in bilateral vestibular connectivity.
Collapse
Affiliation(s)
- Daniel T Daly
- Center for Anatomical Sciences and Education, Department of Surgery, Saint Louis University School of Medicine, St. Louis, Missouri
| | - Michael Ariel
- Center for Anatomical Sciences and Education, Department of Surgery, Saint Louis University School of Medicine, St. Louis, Missouri.,Department of Pharmacology & Physiology, Saint Louis University School of Medicine, St. Louis, Missouri
| |
Collapse
|
183
|
Schell C, Sabass B, Helmstaedter M, Geist F, Abed A, Yasuda-Yamahara M, Sigle A, Maier JI, Grahammer F, Siegerist F, Artelt N, Endlich N, Kerjaschki D, Arnold HH, Dengjel J, Rogg M, Huber TB. ARP3 Controls the Podocyte Architecture at the Kidney Filtration Barrier. Dev Cell 2018; 47:741-757.e8. [PMID: 30503751 PMCID: PMC6302147 DOI: 10.1016/j.devcel.2018.11.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 09/03/2018] [Accepted: 11/01/2018] [Indexed: 12/20/2022]
Abstract
Podocytes, highly specialized epithelial cells, build the outer part of the kidney filtration barrier and withstand high mechanical forces through a complex network of cellular protrusions. Here, we show that Arp2/3-dependent actin polymerization controls actomyosin contractility and focal adhesion maturation of podocyte protrusions and thereby regulates formation, maintenance, and capacity to adapt to mechanical requirements of the filtration barrier. We find that N-WASP-Arp2/3 define the development of complex arborized podocyte protrusions in vitro and in vivo. Loss of dendritic actin networks results in a pronounced activation of the actomyosin cytoskeleton and the generation of over-maturated but less efficient adhesion, leading to detachment of podocytes. Our data provide a model to explain podocyte protrusion morphology and their mechanical stability based on a tripartite relationship between actin polymerization, contractility, and adhesion. ARP3-dependent actin assembly is required for podocyte process formation Arp2/3 thereby links process formation, podocyte adhesion and mechano-adaptation Arp2/3 function is regulated by a reciprocal interplay with actomyosin
Collapse
Affiliation(s)
- Christoph Schell
- Institute of Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany; Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany; Berta-Ottenstein Programme, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Benedikt Sabass
- Institute of Complex Systems-2, Forschungszentrum Jülich, Jülich 52428, Germany
| | - Martin Helmstaedter
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Felix Geist
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Ahmed Abed
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Mako Yasuda-Yamahara
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany; Department of Medicine, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - August Sigle
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Jasmin I Maier
- Institute of Surgical Pathology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany; Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Florian Grahammer
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany; III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany
| | - Florian Siegerist
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald 17487, Germany
| | - Nadine Artelt
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald 17487, Germany
| | - Nicole Endlich
- Department of Anatomy and Cell Biology, University Medicine Greifswald, Greifswald 17487, Germany
| | | | - Hans-Henning Arnold
- Cell and Molecular Biology, Technical University of Braunschweig, Braunschweig 38106, Germany
| | - Jörn Dengjel
- BIOSS Center for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, Freiburg 79106, Germany; Department of Biology, University of Fribourg, Fribourg 1700, Switzerland; Department of Dermatology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany
| | - Manuel Rogg
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany
| | - Tobias B Huber
- Department of Medicine IV, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg 79106, Germany; III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg 20246, Germany; BIOSS Center for Biological Signaling Studies, Albert-Ludwigs-University Freiburg, Freiburg 79106, Germany.
| |
Collapse
|
184
|
Del Giovane A, Ragnini-Wilson A. Targeting Smoothened as a New Frontier in the Functional Recovery of Central Nervous System Demyelinating Pathologies. Int J Mol Sci 2018; 19:E3677. [PMID: 30463396 PMCID: PMC6274747 DOI: 10.3390/ijms19113677] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 11/09/2018] [Accepted: 11/14/2018] [Indexed: 12/20/2022] Open
Abstract
Myelin sheaths on vertebrate axons provide protection, vital support and increase the speed of neuronal signals. Myelin degeneration can be caused by viral, autoimmune or genetic diseases. Remyelination is a natural process that restores the myelin sheath and, consequently, neuronal function after a demyelination event, preventing neurodegeneration and thereby neuron functional loss. Pharmacological approaches to remyelination represent a promising new frontier in the therapy of human demyelination pathologies and might provide novel tools to improve adaptive myelination in aged individuals. Recent phenotypical screens have identified agonists of the atypical G protein-coupled receptor Smoothened and inhibitors of the glioma-associated oncogene 1 as being amongst the most potent stimulators of oligodendrocyte precursor cell (OPC) differentiation in vitro and remyelination in the central nervous system (CNS) of mice. Here, we discuss the current state-of-the-art of studies on the role of Sonic Hedgehog reactivation during remyelination, referring readers to other reviews for the role of Hedgehog signaling in cancer and stem cell maintenance.
Collapse
Affiliation(s)
- Alice Del Giovane
- Department of Biology University of Rome Tor Vergata, Viale Della Ricerca Scientifica, 00133 Rome, Italy.
| | - Antonella Ragnini-Wilson
- Department of Biology University of Rome Tor Vergata, Viale Della Ricerca Scientifica, 00133 Rome, Italy.
| |
Collapse
|
185
|
Ulc A, Zeug A, Bauch J, van Leeuwen S, Kuhlmann T, ffrench-Constant C, Ponimaskin E, Faissner A. The guanine nucleotide exchange factor Vav3 modulates oligodendrocyte precursor differentiation and supports remyelination in white matter lesions. Glia 2018; 67:376-392. [DOI: 10.1002/glia.23548] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 09/03/2018] [Accepted: 09/04/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Annika Ulc
- Department of Cell Morphology and Molecular Neurobiology; Ruhr-University Bochum; Germany
| | - Andre Zeug
- Cellular Neurophysiology, Centre for Physiology; Hannover Medical School; Hannover Germany
| | - Juliane Bauch
- Department of Cell Morphology and Molecular Neurobiology; Ruhr-University Bochum; Germany
| | - Simon van Leeuwen
- Department of Cell Morphology and Molecular Neurobiology; Ruhr-University Bochum; Germany
| | - Tanja Kuhlmann
- Institute of Neuropathology; University Hospital Münster; Germany
| | | | - Evgeni Ponimaskin
- Cellular Neurophysiology, Centre for Physiology; Hannover Medical School; Hannover Germany
| | - Andreas Faissner
- Department of Cell Morphology and Molecular Neurobiology; Ruhr-University Bochum; Germany
| |
Collapse
|
186
|
Pepper RE, Pitman KA, Cullen CL, Young KM. How Do Cells of the Oligodendrocyte Lineage Affect Neuronal Circuits to Influence Motor Function, Memory and Mood? Front Cell Neurosci 2018; 12:399. [PMID: 30524235 PMCID: PMC6262292 DOI: 10.3389/fncel.2018.00399] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Accepted: 10/17/2018] [Indexed: 12/11/2022] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) are immature cells in the central nervous system (CNS) that can rapidly respond to changes within their environment by modulating their proliferation, motility and differentiation. OPCs differentiate into myelinating oligodendrocytes throughout life, and both cell types have been implicated in maintaining and modulating neuronal function to affect motor performance, cognition and emotional state. However, questions remain about the mechanisms employed by OPCs and oligodendrocytes to regulate circuit function, including whether OPCs can only influence circuits through their generation of new oligodendrocytes, or can play other regulatory roles within the CNS. In this review, we detail the molecular and cellular mechanisms that allow OPCs, newborn oligodendrocytes and pre-existing oligodendrocytes to regulate circuit function and ultimately influence behavioral outcomes.
Collapse
Affiliation(s)
- Renee E Pepper
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Kimberley A Pitman
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Carlie L Cullen
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Kaylene M Young
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
187
|
Almeida RG. The Rules of Attraction in Central Nervous System Myelination. Front Cell Neurosci 2018; 12:367. [PMID: 30374292 PMCID: PMC6196289 DOI: 10.3389/fncel.2018.00367] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 09/27/2018] [Indexed: 12/12/2022] Open
Abstract
The wrapping of myelin around axons is crucial for the development and function of the central nervous system (CNS) of vertebrates, greatly regulating the conduction of action potentials. Oligodendrocytes, the myelinating glia of the CNS, have an intrinsic tendency to wrap myelin around any permissive structure in vitro, but in vivo, myelin is targeted with remarkable specificity only to certain axons. Despite the importance of myelination, the mechanisms by which oligodendrocytes navigate a complex milieu that includes many types of cells and their cellular projections and select only certain axons for myelination remains incompletely understood. In this Mini-review, I highlight recent studies that shed light on the molecular and cellular rules governing CNS myelin targeting.
Collapse
Affiliation(s)
- Rafael Góis Almeida
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
188
|
Treichel AJ, Hines JH. Development of an Embryonic Zebrafish Oligodendrocyte-Neuron Mixed Coculture System. Zebrafish 2018; 15:586-596. [PMID: 30300571 DOI: 10.1089/zeb.2018.1625] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
During vertebrate neural development, oligodendrocytes insulate nerve axons with myelin sheaths. Zebrafish (Danio rerio) has emerged as a useful model organism for studying oligodendrocyte development. However, the absence of an in vitro culture system necessitates in vivo manipulations and analyses, which, in some instances, limits the questions that can be addressed. To fill this gap we developed a mixed coculture system for embryonic zebrafish neurons and oligodendrocyte-lineage cells. Cultures harvested from embryos ≥30 hours postfertilization (hpf) yielded oligodendrocyte progenitor cells (OPCs) positive for olig2 and sox10 transgenic reporters. Cultured OPCs exhibited dynamic, exploratory membrane processes, and cell morphologies resembled those established in vivo. Cells harvested from advanced stage embryos possessed more arborized processes than those from early stage embryos. Advanced stage (>60 hpf) embryo culture produced differentiated, mbp+ oligodendrocytes. Genetically tractable neuron subtypes extended neurites when harvested from embryos ≥19 hpf. Coculture produced juxtaposed oligodendrocytes and neurons, demonstrating the practical usefulness of this technique for future studies examining axon-oligodendrocyte interactions under defined conditions. We expect that zebrafish oligodendrocyte culture will complement existing in vivo strengths and may facilitate future studies elucidating the mechanisms of oligodendrocyte specification, proliferation, differentiation, motility, and axon-oligodendrocyte interactions that shape adult myelination patterns.
Collapse
Affiliation(s)
| | - Jacob H Hines
- Department of Biology, Winona State University , Winona, Minnesota
| |
Collapse
|
189
|
Thomas L, Pasquini LA. Galectin-3-Mediated Glial Crosstalk Drives Oligodendrocyte Differentiation and (Re)myelination. Front Cell Neurosci 2018; 12:297. [PMID: 30258354 PMCID: PMC6143789 DOI: 10.3389/fncel.2018.00297] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Accepted: 08/17/2018] [Indexed: 12/17/2022] Open
Abstract
Galectin-3 (Gal-3) is the only chimeric protein in the galectin family. Gal-3 structure comprises unusual tandem repeats of proline and glycine-rich short stretches bound to a carbohydrate-recognition domain (CRD). The present review summarizes Gal-3 functions in the extracellular and intracellular space, its regulation and its internalization and secretion, with a focus on the current knowledge of Gal-3 role in central nervous system (CNS) health and disease, particularly oligodendrocyte (OLG) differentiation, myelination and remyelination in experimental models of multiple sclerosis (MS). During myelination, microglia-expressed Gal-3 promotes OLG differentiation by binding glycoconjugates present only on the cell surface of OLG precursor cells (OPC). During remyelination, microglia-expressed Gal-3 favors an M2 microglial phenotype, hence fostering myelin debris phagocytosis through TREM-2b phagocytic receptor and OLG differentiation. Gal-3 is necessary for myelin integrity and function, as evidenced by myelin ultrastructural and behavioral studies from LGALS3-/- mice. Mechanistically, Gal-3 enhances actin assembly and reduces Erk 1/2 activation, leading to early OLG branching. Gal-3 later induces Akt activation and increases MBP expression, promoting gelsolin release and actin disassembly and thus regulating OLG final differentiation. Altogether, findings indicate that Gal-3 mediates the glial crosstalk driving OLG differentiation and (re)myelination and may be regarded as a target in the design of future therapies for a variety of demyelinating diseases.
Collapse
Affiliation(s)
- Laura Thomas
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Chemistry and Biological Physicochemistry (IQUIFIB), National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Laura Andrea Pasquini
- Department of Biological Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina.,Institute of Chemistry and Biological Physicochemistry (IQUIFIB), National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| |
Collapse
|
190
|
Fauveau M, Wilmet B, Deboux C, Benardais K, Bachelin C, Temporão AC, Kerninon C, Nait Oumesmar B. SOX17 transcription factor negatively regulates oligodendrocyte precursor cell differentiation. Glia 2018; 66:2221-2232. [PMID: 30152028 DOI: 10.1002/glia.23483] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 05/16/2018] [Accepted: 06/08/2018] [Indexed: 11/08/2022]
Abstract
Oligodendrocyte development is a critical process timely and spatially regulated to ensure proper myelination of the central nervous system. HMG-box transcription factors are key regulators of oligodendrocyte lineage progression. Among these factors, Sox17 was previously identified as a positive regulator of oligodendrocyte development. However, the role of Sox17 in oligodendroglial cell lineage progression and differentiation is still poorly understood. To define the functional role of Sox17, we generated new transgenic mouse models with inducible overexpression of Sox17, specifically in oligodendroglial cells. Here, we report that gain of Sox17 function has no effect on oligodendrocyte progenitor cells (OPCs) specification. During early postnatal development, Sox17 overexpression increases the pool of OPCs at the expense of differentiated oligodendrocytes. However, the oligodendroglial cell population, OPC proliferation and apoptosis remained unchanged in Sox17 transgenic mice. RNA sequencing, quantitative RT-PCR and immunohistochemical analysis showed that Sox17 represses the expression of the major myelin genes, resulting in a severe CNS hypomyelination. Overall, our data highlight an unexpected role for Sox17 as a negative regulator of OPC differentiation and myelination, suggesting stage specific functions for this factor during oligodendroglial cell lineage progression.
Collapse
Affiliation(s)
- Melissa Fauveau
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Baptiste Wilmet
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Cyrille Deboux
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Karelle Benardais
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Corinne Bachelin
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Ana C Temporão
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Christophe Kerninon
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| | - Brahim Nait Oumesmar
- Inserm U1127, Institut du Cerveau et de la Moelle Epinière, ICM, Paris, France.,Sorbonne Université UMR-S1127, Paris, France.,CNRS, UMR 7225, Paris, France
| |
Collapse
|
191
|
Pfrieger FW. Learning from Barres. Glia 2018. [DOI: 10.1002/glia.23334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Frank W. Pfrieger
- Institute of Cellular and Integrative Neurosciences (INCI), CNRS UPR 3212, University of Strasbourg; Strasbourg 67084 France
| |
Collapse
|
192
|
Ong W, Lin J, Bechler ME, Wang K, Wang M, ffrench-Constant C, Chew SY. Microfiber drug/gene delivery platform for study of myelination. Acta Biomater 2018; 75:152-160. [PMID: 29885526 DOI: 10.1016/j.actbio.2018.06.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 06/01/2018] [Accepted: 06/05/2018] [Indexed: 01/21/2023]
Abstract
Our ability to rescue functional deficits after demyelinating diseases or spinal cord injuries is limited by our lack of understanding of the complex remyelination process, which is crucial to functional recovery. In this study, we developed an electrospun suspended poly(ε-caprolactone) microfiber platform to enable the screening of therapeutics for remyelination. As a proof of concept, this platform employed scaffold-mediated non-viral delivery of a microRNA (miR) cocktail to promote oligodendrocyte precursor cells (OPCs) differentiation and myelination. We observed enhanced OPCs differentiation when the cells were transfected with miR-219 and miR-338 on the microfiber substrates. Moreover, miRs promoted the formation of MBP+ tubular extensions around the suspended fibers, which was indicative of myelination, instead of flat myelin membranes on 2D substrates. In addition, OPCs that were transfected with the cocktail of miRs formed significantly longer and larger amounts of MBP+ extensions. Taken together, these results demonstrate the efficacy of this functional screening platform for understanding myelination. STATEMENT OF SIGNIFICANCE The lack of understanding of the complex myelination process has hindered the discovery of effective therapeutic treatments for demyelinating diseases. Hence, in vitro models that enable systematic understanding, visualization and quantification of myelination are valuable. Unfortunately, achieving reproducible in vitro myelination by oligodendrocytes (OLs) remains highly challenging. Here, we engineered a suspended microfiber platform that enables sustained non-viral drug/gene delivery to study OL differentiation and myelination. Sustained drug delivery permits the investigation of OL development, which spans several weeks. We show that promyelinogenic microRNAs promoted OL differentiation and myelination on this platform. Our engineered microfiber substrate could serve as a drug/gene screening platform and facilitate future translation into direct implantable devices for in vivo remyelination purposes.
Collapse
|
193
|
Abu-Rub M, Miller RH. Emerging Cellular and Molecular Strategies for Enhancing Central Nervous System (CNS) Remyelination. Brain Sci 2018; 8:brainsci8060111. [PMID: 29914096 PMCID: PMC6024921 DOI: 10.3390/brainsci8060111] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 06/12/2018] [Accepted: 06/13/2018] [Indexed: 02/06/2023] Open
Abstract
Myelination is critical for the normal functioning of the central nervous system (CNS) in vertebrates. Conditions in which the development of myelin is perturbed result in severely compromised individuals often with shorter lifespans, while loss of myelin in the adult results in a variety of functional deficits. Although some form of spontaneous remyelination often takes place, the repair process as a whole often fails. Several lines of evidence suggest it is feasible to develop strategies that enhance the capacity of the CNS to undergo remyelination and potentially reverse functional deficits. Such strategies include cellular therapies using either neural or mesenchymal stem cells as well as molecular regulators of oligodendrocyte development and differentiation. Given the prevalence of demyelinating diseases and their effects on the quality of life for affected individuals it is imperative that effective therapies are developed. Here we discuss some of the new approaches to CNS myelin repair that hold promise for reducing the burden of diseases characterized by myelin loss.
Collapse
Affiliation(s)
- Mohammad Abu-Rub
- Department of Neurology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA.
| | - Robert H Miller
- Department of Anatomy and Regenerative Biology, George Washington University School of Medicine and Health Sciences, Washington, DC 20037, USA.
| |
Collapse
|
194
|
Torvund-Jensen J, Steengaard J, Askebjerg LB, Kjaer-Sorensen K, Laursen LS. The 3'UTRs of Myelin Basic Protein mRNAs Regulate Transport, Local Translation and Sensitivity to Neuronal Activity in Zebrafish. Front Mol Neurosci 2018; 11:185. [PMID: 29946237 PMCID: PMC6006989 DOI: 10.3389/fnmol.2018.00185] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/14/2018] [Indexed: 02/04/2023] Open
Abstract
Formation of functional myelin sheaths within the central nervous system depends on expression of myelin basic protein (MBP). Following process extension and wrapping around axonal segments, this highly basic protein is required for compaction of the multi-layered membrane sheath produced by oligodendrocytes. MBP is hypothesized to be targeted to the membrane sheath by mRNA transport and local translation, which ensures that its expression is temporally and spatially restricted. The mechanistic details of how this might be regulated are still largely unknown, in particular because a model system that allows this process to be studied in vivo is lacking. We here show that the expression of the zebrafish MBP orthologs, mbpa and mbpb, is developmentally regulated, and that expression of specific mbpa isoforms is restricted to the peripheral nervous system. By analysis of transgenic zebrafish, which express a fluorescent reporter protein specifically in myelinating oligodendrocytes, we demonstrate that both mbpa and mbpb include a 3’UTR sequence, by which mRNA transport and translation is regulated in vivo. Further functional analysis suggests that: (1) the 3’UTRs delay the onset of protein expression; and that (2) several regulatory elements contribute to targeting of the mbp mRNA to the myelin sheath. Finally, we show that a pharmacological compound known to enhance neuronal activity stimulates the translation of Mbp in zebrafish in a 3’UTR-dependent manner. A similar effect was obtained following stimulation with a TrkB receptor agonist, and cell-based assays further confirmed that the receptor ligand, BDNF, in combination with other signals reversed the inhibitory effect of the 3’UTR on translation.
Collapse
Affiliation(s)
- Julie Torvund-Jensen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | - Jes Steengaard
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| | | | | | - Lisbeth S Laursen
- Department of Molecular Biology and Genetics, Aarhus University, Aarhus, Denmark
| |
Collapse
|
195
|
Alvarez-Prats A, Bjelobaba I, Aldworth Z, Baba T, Abebe D, Kim YJ, Stojilkovic SS, Stopfer M, Balla T. Schwann-Cell-Specific Deletion of Phosphatidylinositol 4-Kinase Alpha Causes Aberrant Myelination. Cell Rep 2018; 23:2881-2890. [PMID: 29874576 PMCID: PMC7268203 DOI: 10.1016/j.celrep.2018.05.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 04/17/2018] [Accepted: 05/04/2018] [Indexed: 01/01/2023] Open
Abstract
Active membrane remodeling during myelination relies on phospholipid synthesis and membrane polarization, both of which are known to depend on inositol phospholipids. Here, we show that sciatic nerves of mice lacking phosphatidylinositol 4-kinase alpha (PI4KA) in Schwann cells (SCs) show substantially reduced myelin thickness with grave consequences on nerve conductivity and motor functions. Surprisingly, prolonged inhibition of PI4KA in immortalized mouse SCs failed to decrease plasma membrane phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) levels or PI 3-kinase (PI3K) activation, in spite of large reductions in plasma membrane PI4P levels. Instead, it caused rearrangements of the actin cytoskeleton, which was also observed in sciatic nerves of knockout animals. PI4KA inactivation disproportionally reduced phosphatidylserine, phosphatidylethanolamine, and sphingomyelin content in mutant nerves, with similar changes observed in SCs treated with a PI4KA inhibitor. These studies define a role for PI4KA in myelin formation primarily affecting metabolism of key phospholipids and the actin cytoskeleton.
Collapse
Affiliation(s)
- Alejandro Alvarez-Prats
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Ivana Bjelobaba
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Zane Aldworth
- Section on Sensory Coding and Neural Ensembles, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Takashi Baba
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Daniel Abebe
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Yeun Ju Kim
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Stanko S Stojilkovic
- Section on Cellular Signaling, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Mark Stopfer
- Section on Sensory Coding and Neural Ensembles, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA
| | - Tamas Balla
- Section on Molecular Signal Transduction, Program for Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD 20892, USA.
| |
Collapse
|
196
|
Dillenburg A, Ireland G, Holloway RK, Davies CL, Evans FL, Swire M, Bechler ME, Soong D, Yuen TJ, Su GH, Becher JC, Smith C, Williams A, Miron VE. Activin receptors regulate the oligodendrocyte lineage in health and disease. Acta Neuropathol 2018; 135:887-906. [PMID: 29397421 PMCID: PMC5954071 DOI: 10.1007/s00401-018-1813-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 01/16/2018] [Accepted: 01/29/2018] [Indexed: 12/12/2022]
Abstract
The most prevalent neurological disorders of myelin include perinatal brain injury leading to cerebral palsy in infants and multiple sclerosis in adults. Although these disorders have distinct etiologies, they share a common neuropathological feature of failed progenitor differentiation into myelin-producing oligodendrocytes and lack of myelin, for which there is an unmet clinical need. Here, we reveal that a molecular pathology common to both disorders is dysregulation of activin receptors and that activin receptor signaling is required for the majority of myelin generation in development and following injury. Using a constitutive conditional knockout of all activin receptor signaling in oligodendrocyte lineage cells, we discovered this signaling to be required for myelination via regulation of oligodendrocyte differentiation and myelin compaction. These processes were found to be dependent on the activin receptor subtype Acvr2a, which is expressed during oligodendrocyte differentiation and axonal ensheathment in development and following myelin injury. During efficient myelin regeneration, Acvr2a upregulation was seen to coincide with downregulation of Acvr2b, a receptor subtype with relatively higher ligand affinity; Acvr2b was shown to be dispensable for activin receptor-driven oligodendrocyte differentiation and its overexpression was sufficient to impair the abovementioned ligand-driven responses. In actively myelinating or remyelinating areas of human perinatal brain injury and multiple sclerosis tissue, respectively, oligodendrocyte lineage cells expressing Acvr2a outnumbered those expressing Acvr2b, whereas in non-repairing lesions Acvr2b+ cells were increased. Thus, we propose that following human white matter injury, this increase in Acvr2b expression would sequester ligand and consequently impair Acvr2a-driven oligodendrocyte differentiation and myelin formation. Our results demonstrate dysregulated activin receptor signaling in common myelin disorders and reveal Acvr2a as a novel therapeutic target for myelin generation following injury across the lifespan.
Collapse
|
197
|
Eno C, Pelegri F. Modulation of F-actin dynamics by maternal Mid1ip1L controls germ plasm aggregation and furrow recruitment in the zebrafish embryo. Development 2018; 145:dev156596. [PMID: 29724756 PMCID: PMC6001372 DOI: 10.1242/dev.156596] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Accepted: 04/23/2018] [Indexed: 12/11/2022]
Abstract
During the early embryonic cell cycles, zebrafish germ plasm ribonucleoparticles (RNPs) gradually multimerize and become recruited to the forming furrows. RNPs multimerization occurs prior to and during furrow initiation, as forming aggregates move outward through their association with the tips of growing interphase astral microtubules. Germ plasm RNPs are also associated with short cortical F-actin. We show that, in embryos mutant for the cytoskeletal regulator mid1ip1l, germ plasm RNPs fail to become recruited to the furrow, accumulating instead at the periphery of the blastodisc. RNP aggregates are associated with zones of mid1ip1l-dependent cyclical local cortical F-actin network enrichments, as well as contractions at both the cortex and the contractile ring. F-actin inhibition in wild-type embryos mimics the RNP peripheral accumulation defect of mid1ip1l mutants. Our studies suggest that a common mechanism underlies distinct steps of germ plasm RNP segregation. At the cortex, this process attenuates microtubule-dependent outward RNP movement to retain RNPs in the blastodisc cortex and allow their recruitment to the furrows. F-actin network contraction likely also facilitates higher-order germ plasm RNP multimerization.
Collapse
Affiliation(s)
- Celeste Eno
- Laboratory of Genetics, University of Wisconsin, Madison, WI 53706, USA
| | - Francisco Pelegri
- Laboratory of Genetics, University of Wisconsin, Madison, WI 53706, USA
| |
Collapse
|
198
|
Pinezich MR, Russell LN, Murphy NP, Lampe KJ. Encapsulated oligodendrocyte precursor cell fate is dependent on PDGF-AA release kinetics in a 3D microparticle-hydrogel drug delivery system. J Biomed Mater Res A 2018; 106:2402-2411. [PMID: 29660252 DOI: 10.1002/jbm.a.36432] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 02/08/2018] [Accepted: 04/05/2018] [Indexed: 12/14/2022]
Abstract
Biomaterial drug delivery systems (DDS) can be used to regulate growth factor release and combat the limited intrinsic regeneration capabilities of central nervous system (CNS) tissue following injury and disease. Of particular interest are systems that aid in oligodendrocyte regeneration, as oligodendrocytes generate myelin which surrounds neuronal axons and helps transmit signals throughout the CNS. Oligodendrocyte precursor cells (OPCs) are found in small numbers in the adult CNS, but are unable to effectively differentiate following CNS injury. Delivery of signaling molecules can initiate a favorable OPC response, such as proliferation or differentiation. Here, we investigate the delivery of one such molecule, platelet derived growth factor-AA (PDGF-AA), from poly(lactic-co-glycolic) acid microparticles to OPCs in a 3D polyethylene glycol-based hydrogel. The goal of this DDS was to better understand the relationship between PDGF-AA release kinetics and OPC fate. The system approximates native brain tissue stiffness, while incorporating PDGF-AA under seven different delivery scenarios. Within this DDS, supply of PDGF-AA followed by PDGF-AA withdrawal caused OPCs to upregulate gene expression of myelin basic protein (MBP) by factors of 1.6-9.2, whereas continuous supply of PDGF-AA caused OPCs to remain proliferative. At the protein expression level, we observed an upregulation in O1, a marker for mature oligodendrocytes. Together, these results show that burst release followed by withdrawal of PDGF-AA from a hydrogel DDS stimulates survival, proliferation, and differentiation of OPCs in vitro. Our results could inform the development of improved neural regeneration strategies that incorporate delivery of PDGF-AA to the injured CNS. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A:2402-2411, 2018.
Collapse
Affiliation(s)
- Meghan R Pinezich
- Department of Chemical Engineering, University of Virginia, 102 Engineers' Way, Charlottesville, VA, 22904
| | - Lauren N Russell
- Department of Chemical Engineering, University of Virginia, 102 Engineers' Way, Charlottesville, VA, 22904
| | - Nicholas P Murphy
- Department of Chemical Engineering, University of Virginia, 102 Engineers' Way, Charlottesville, VA, 22904
| | - Kyle J Lampe
- Department of Chemical Engineering, University of Virginia, 102 Engineers' Way, Charlottesville, VA, 22904
| |
Collapse
|
199
|
Azevedo MM, Domingues HS, Cordelières FP, Sampaio P, Seixas AI, Relvas JB. Jmy regulates oligodendrocyte differentiation via modulation of actin cytoskeleton dynamics. Glia 2018; 66:1826-1844. [DOI: 10.1002/glia.23342] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 03/26/2018] [Accepted: 04/05/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Maria M. Azevedo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto; Porto Portugal
- IBMC - Instituto de Biologia Molecular e Celular; Porto Portugal
| | - Helena S. Domingues
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto; Porto Portugal
- IBMC - Instituto de Biologia Molecular e Celular; Porto Portugal
| | - Fabrice P. Cordelières
- Bordeaux Imaging Centre, UMS 3420 CNRS, CNRS-INSERM, University of Bordeaux; Bordeaux France
| | - Paula Sampaio
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto; Porto Portugal
- IBMC - Instituto de Biologia Molecular e Celular; Porto Portugal
| | - Ana I. Seixas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto; Porto Portugal
- IBMC - Instituto de Biologia Molecular e Celular; Porto Portugal
| | - João B. Relvas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto; Porto Portugal
- IBMC - Instituto de Biologia Molecular e Celular; Porto Portugal
- The Discoveries Centre for Regeneration and Precision Medicine, Porto campus; Porto Portugal
| |
Collapse
|
200
|
Mørkholt AS, Kastaniegaard K, Trabjerg MS, Gopalasingam G, Niganze W, Larsen A, Stensballe A, Nielsen S, Nieland JD. Identification of brain antigens recognized by autoantibodies in experimental autoimmune encephalomyelitis-induced animals treated with etomoxir or interferon-β. Sci Rep 2018; 8:7092. [PMID: 29728570 PMCID: PMC5935685 DOI: 10.1038/s41598-018-25391-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Accepted: 04/19/2018] [Indexed: 11/08/2022] Open
Abstract
Multiple sclerosis (MS) is a neurodegenerative autoimmune disease, where chronic inflammation plays an essential role in its pathology. A feature of MS is the production of autoantibodies stimulated by an altered-peptide-ligand response and epitope spreading, resulting in loss of tolerance for self-proteins. The involvement of autoantibodies in MS pathogenesis has been suggested to initiate and drive progression of inflammation; however, the etiology of MS remains unknown. The effect of etomoxir and interferon-β (IFN-β) was examined in an experimental-autoimmune-encephalomyelitis (EAE) model of MS. Moreover, the impact of etomoxir and IFN-β on recognition of brain proteins in serum from EAE rats was examined with the purpose of identifying the autoantibody reactivities involved in MS. Animals treated with etomoxir on day 1 exhibited a statistically significantly lower disease score than animals treated with IFN-β (on day 1 or 5) or placebo. Etomoxir treatment on day 5 resulted in a significantly lower disease score than IFN-β treatment on day 1. After disease induction antibodies was induced to a broad pallet of antigens in the brain. Surprisingly, by blocking CPT1 and therewith lipid metabolism several alterations in the antibody response was observed suggesting that autoantibodies play a role in the EAE animal model.
Collapse
Affiliation(s)
| | | | | | - Gopana Gopalasingam
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Wanda Niganze
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Agnete Larsen
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Allan Stensballe
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - Søren Nielsen
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark
| | - John Dirk Nieland
- Department of Health Science and Technology, Aalborg University, Aalborg, Denmark.
| |
Collapse
|