1
|
Chang Y, Zhu J, Li X, Deng Y, Lai B, Ma Y, Tong J, Liu H, Li J, Yang C, Chen Q, Lu C, Liang Y, Qi S, Wang X, Kong E. Palmitoylation regulates myelination by modulating the ZDHHC3-Cadm4 axis in the central nervous system. Signal Transduct Target Ther 2024; 9:254. [PMID: 39327467 PMCID: PMC11427461 DOI: 10.1038/s41392-024-01971-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 08/10/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024] Open
Abstract
The downregulation of Cadm4 (Cell adhesion molecular 4) is a prominent feature in demyelination diseases, yet, the underlying molecular mechanism remains elusive. Here, we reveal that Cadm4 undergoes specific palmitoylation at cysteine-347 (C347), which is crucial for its stable localization on the plasma membrane (PM). Mutation of C347 to alanine (C347A), blocking palmitoylation, causes Cadm4 internalization from the PM and subsequent degradation. In vivo experiments introducing the C347A mutation (Cadm4-KI) lead to severe myelin abnormalities in the central nervous system (CNS), characterized by loss, demyelination, and hypermyelination. We further identify ZDHHC3 (Zinc finger DHHC-type palmitoyltransferase 3) as the enzyme responsible for catalyzing Cadm4 palmitoylation. Depletion of ZDHHC3 reduces Cadm4 palmitoylation and diminishes its PM localization. Remarkably, genetic deletion of ZDHHC3 results in decreased Cadm4 palmitoylation and defects in CNS myelination, phenocopying the Cadm4-KI mouse model. Consequently, altered Cadm4 palmitoylation impairs neuronal transmission and cognitive behaviors in both Cadm4-KI and ZDHHC3 knockout mice. Importantly, attenuated ZDHHC3-Cadm4 signaling significantly influences neuroinflammation in diverse demyelination diseases. Mechanistically, we demonstrate the predominant expression of Cadm4 in the oligodendrocyte lineage and its potential role in modulating cell differentiation via the WNT-β-Catenin pathway. Together, our findings propose that dysregulated ZDHHC3-Cadm4 signaling contributes to myelin abnormalities, suggesting a common pathological mechanism underlying demyelination diseases associated with neuroinflammation.
Collapse
Affiliation(s)
- Yanli Chang
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
| | - Jiangli Zhu
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Department of Urology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, China
| | - Xiaopeng Li
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Yi Deng
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
| | - Birou Lai
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
| | - Yidan Ma
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jia Tong
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
| | - Huicong Liu
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
| | - Juanjuan Li
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
| | - Chenyu Yang
- Center of Cryo-Electron Microscopy, Zhejiang University, Hangzhou, China
| | - Qiao Chen
- Department of Nutrition, Third Medical Center of PLA General Hospital, Beijing, China
| | - Chengbiao Lu
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
| | - Yinming Liang
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China
| | - Shiqian Qi
- Department of Urology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, China
| | - Xiaoning Wang
- School of Life Sciences, Fudan University, Shanghai, China.
| | - Eryan Kong
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, China.
- Institute of Psychiatry and Neuroscience, Xinxiang Key Laboratory of Protein Palmitoylation and Major Human Diseases, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
2
|
Krauter D, Stausberg D, Hartmann TJ, Volkmann S, Kungl T, Rasche DA, Saher G, Fledrich R, Stassart RM, Nave KA, Goebbels S, Ewers D, Sereda MW. Targeting PI3K/Akt/mTOR signaling in rodent models of PMP22 gene-dosage diseases. EMBO Mol Med 2024; 16:616-640. [PMID: 38383802 PMCID: PMC10940316 DOI: 10.1038/s44321-023-00019-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 12/15/2023] [Accepted: 12/15/2023] [Indexed: 02/23/2024] Open
Abstract
Haplo-insufficiency of the gene encoding the myelin protein PMP22 leads to focal myelin overgrowth in the peripheral nervous system and hereditary neuropathy with liability to pressure palsies (HNPP). Conversely, duplication of PMP22 causes Charcot-Marie-Tooth disease type 1A (CMT1A), characterized by hypomyelination of medium to large caliber axons. The molecular mechanisms of abnormal myelin growth regulation by PMP22 have remained obscure. Here, we show in rodent models of HNPP and CMT1A that the PI3K/Akt/mTOR-pathway inhibiting phosphatase PTEN is correlated in abundance with PMP22 in peripheral nerves, without evidence for direct protein interactions. Indeed, treating DRG neuron/Schwann cell co-cultures from HNPP mice with PI3K/Akt/mTOR pathway inhibitors reduced focal hypermyelination. When we treated HNPP mice in vivo with the mTOR inhibitor Rapamycin, motor functions were improved, compound muscle amplitudes were increased and pathological tomacula in sciatic nerves were reduced. In contrast, we found Schwann cell dedifferentiation in CMT1A uncoupled from PI3K/Akt/mTOR, leaving partial PTEN ablation insufficient for disease amelioration. For HNPP, the development of PI3K/Akt/mTOR pathway inhibitors may be considered as the first treatment option for pressure palsies.
Collapse
Affiliation(s)
- Doris Krauter
- Research Group "Translational Neurogenetics", Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
- Division of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Daniela Stausberg
- Research Group "Translational Neurogenetics", Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Timon J Hartmann
- Research Group "Translational Neurogenetics", Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Stefan Volkmann
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Theresa Kungl
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - David A Rasche
- Research Group "Translational Neurogenetics", Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Gesine Saher
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Robert Fledrich
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Ruth M Stassart
- Institute of Neuropathology, University of Leipzig, Leipzig, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Sandra Goebbels
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
| | - David Ewers
- Research Group "Translational Neurogenetics", Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany.
| | - Michael W Sereda
- Research Group "Translational Neurogenetics", Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany.
| |
Collapse
|
3
|
Iyer M, Kantarci H, Cooper MH, Ambiel N, Novak SW, Andrade LR, Lam M, Jones G, Münch AE, Yu X, Khakh BS, Manor U, Zuchero JB. Oligodendrocyte calcium signaling promotes actin-dependent myelin sheath extension. Nat Commun 2024; 15:265. [PMID: 38177161 PMCID: PMC10767123 DOI: 10.1038/s41467-023-44238-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 12/05/2023] [Indexed: 01/06/2024] Open
Abstract
Myelin is essential for rapid nerve signaling and is increasingly found to play important roles in learning and in diverse diseases of the CNS. Morphological parameters of myelin such as sheath length are thought to precisely tune conduction velocity, but the mechanisms controlling sheath morphology are poorly understood. Local calcium signaling has been observed in nascent myelin sheaths and can be modulated by neuronal activity. However, the role of calcium signaling in sheath formation remains incompletely understood. Here, we use genetic tools to attenuate oligodendrocyte calcium signaling during myelination in the developing mouse CNS. Surprisingly, genetic calcium attenuation does not grossly affect the number of myelinated axons or myelin thickness. Instead, calcium attenuation causes myelination defects resulting in shorter, dysmorphic sheaths. Mechanistically, calcium attenuation reduces actin filaments in oligodendrocytes, and an intact actin cytoskeleton is necessary and sufficient to achieve accurate myelin morphology. Together, our work reveals a cellular mechanism required for accurate CNS myelin formation and may provide mechanistic insight into how oligodendrocytes respond to neuronal activity to sculpt and refine myelin sheaths.
Collapse
Affiliation(s)
- Manasi Iyer
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Husniye Kantarci
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Madeline H Cooper
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Nicholas Ambiel
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Sammy Weiser Novak
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Leonardo R Andrade
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Mable Lam
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Graham Jones
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexandra E Münch
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Xinzhu Yu
- Department of Molecular and Integrative Physiology, Beckman Institute, University of Illinois at Urbana-, Champaign, IL, USA
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Baljit S Khakh
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Uri Manor
- Waitt Advanced Biophotonics Center, Salk Institute for Biological Studies, La Jolla, CA, USA
- Department of Cell and Developmental Biology, University of California, San Diego, San Diego, CA, USA
| | - J Bradley Zuchero
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
4
|
Terada N, Saitoh Y, Saito M, Yamada T, Kamijo A, Yoshizawa T, Sakamoto T. Recent Progress on Genetically Modified Animal Models for Membrane Skeletal Proteins: The 4.1 and MPP Families. Genes (Basel) 2023; 14:1942. [PMID: 37895291 PMCID: PMC10606877 DOI: 10.3390/genes14101942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
The protein 4.1 and membrane palmitoylated protein (MPP) families were originally found as components in the erythrocyte membrane skeletal protein complex, which helps maintain the stability of erythrocyte membranes by linking intramembranous proteins and meshwork structures composed of actin and spectrin under the membranes. Recently, it has been recognized that cells and tissues ubiquitously use this membrane skeletal system. Various intramembranous proteins, including adhesion molecules, ion channels, and receptors, have been shown to interact with the 4.1 and MPP families, regulating cellular and tissue dynamics by binding to intracellular signal transduction proteins. In this review, we focus on our previous studies regarding genetically modified animal models, especially on 4.1G, MPP6, and MPP2, to describe their functional roles in the peripheral nervous system, the central nervous system, the testis, and bone formation. As the membrane skeletal proteins are located at sites that receive signals from outside the cell and transduce signals inside the cell, it is necessary to elucidate their molecular interrelationships, which may broaden the understanding of cell and tissue functions.
Collapse
Affiliation(s)
- Nobuo Terada
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, Matsumoto City, Nagano 390-8621, Japan
| | - Yurika Saitoh
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, Matsumoto City, Nagano 390-8621, Japan
- Center for Medical Education, Teikyo University of Science, Adachi-ku, Tokyo 120-0045, Japan
| | - Masaki Saito
- School of Pharma-Science, Teikyo University, Itabashi-ku, Tokyo 173-8605, Japan;
| | - Tomoki Yamada
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, Matsumoto City, Nagano 390-8621, Japan
| | - Akio Kamijo
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, Matsumoto City, Nagano 390-8621, Japan
- Division of Basic & Clinical Medicine, Nagano College of Nursing, Komagane City, Nagano 399-4117, Japan
| | - Takahiro Yoshizawa
- Division of Animal Research, Research Center for Advanced Science and Technology, Shinshu University, Matsumoto City, Nagano 390-8621, Japan
| | - Takeharu Sakamoto
- Department of Cancer Biology, Institute of Biomedical Science, Kansai Medical University, Hirakata City, Osaka 573-1010, Japan
| |
Collapse
|
5
|
Pauziene N, Ranceviene D, Rysevaite-Kyguoliene K, Inokaitis H, Saburkina I, Plekhanova K, Sabeckiene D, Sabeckis I, Martinaityte R, Pilnikovaite E, Pauza DH. Comparative analysis of intracardiac neural structures in the aged rats with essential hypertension. Anat Rec (Hoboken) 2023; 306:2313-2332. [PMID: 36342958 DOI: 10.1002/ar.25109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 09/16/2022] [Accepted: 10/09/2022] [Indexed: 11/09/2022]
Abstract
Persistent arterial hypertension initiates cardiac autonomic imbalance and alters cardiac tissues. Previous studies have shown that neural component contributes to arterial hypertension etiology, maintenance, and progression and leads to brain damage, peripheral neuropathy, and remodeling of intrinsic cardiac neural plexus. Recently, significant structural changes of the intracardiac neural plexus were demonstrated in young prehypertensive and adult hypertensive spontaneously hypertensive rats (SHR), yet structural alterations of intracardiac neural plexus that occur in the aged SHR remain undetermined. Thus, we analyzed the impact of uncontrolled arterial hypertension in old (48-52 weeks) SHR and the age-matched Wistar-Kyoto rats (WKY). Intrinsic cardiac neural plexus was examined using a combination of immunofluorescence confocal microscopy and transmission electron microscopy in cardiac sections and whole-mount preparations. Our findings demonstrate that structural changes of intrinsic cardiac neural plexus caused by arterial hypertension are heterogeneous and may support recent physiological implications about cardiac denervation occurring together with the hyperinnervation of the SHR heart. We conclude that arterial hypertension leads to (i) the decrease of the neuronal body area, the thickness of atrial nerves, the number of myelinated nerve fibers, unmyelinated axon area and cumulative axon area in the nerve, and the density of myocardial nerve fibers, and (ii) the increase in myelinated nerve fiber area and density of neuronal bodies within epicardiac ganglia. Despite neuropathic alterations of myelinated fibers were exposed within intracardiac nerves of both groups, SHR and WKY, we consider that the determined significant changes in structure of intrinsic cardiac neural plexus were predisposed by arterial hypertension.
Collapse
Affiliation(s)
| | | | | | | | - Inga Saburkina
- Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | | | - Ignas Sabeckis
- Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | | | | |
Collapse
|
6
|
Martens AK, Erwig M, Patzig J, Fledrich R, Füchtbauer EM, Werner HB. Targeted inactivation of the Septin2 and Septin9 genes in myelinating Schwann cells of mice. Cytoskeleton (Hoboken) 2023; 80:290-302. [PMID: 36378242 DOI: 10.1002/cm.21736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/03/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022]
Abstract
The formation of axon-enwrapping myelin sheaths by oligodendrocytes in the central nervous system involves the assembly of a scaffolding septin filament comprised of the subunits SEPTIN2, SEPTIN4, SEPTIN7 and SEPTIN8. Conversely, in the peripheral nervous system (PNS), myelin is synthesized by a different cell type termed Schwann cells, and it remained unknown if septins also assemble as a multimer in PNS myelin. According to prior proteome analysis, PNS myelin comprises the subunits SEPTIN2, SEPTIN7, SEPTIN8, SEPTIN9, and SEPTIN11, which localize to the paranodal and abaxonal myelin subcompartments. Here, we use the Cre/loxP-system to delete the Septin9-gene specifically in Schwann cells, causing a markedly reduced abundance of SEPTIN9 in sciatic nerves, implying that Schwann cells are the main cell type expressing SEPTIN9 in the nerve. However, Septin9-deficiency in Schwann cells did not affect the abundance or localization of other septin subunits. In contrast, when deleting the Septin2-gene in Schwann cells the abundance of all relevant septin subunits was markedly reduced, including SEPTIN9. Notably, we did not find evidence that deleting Septin2 or Septin9 in Schwann cells impairs myelin biogenesis, nerve conduction velocity or motor/sensory capabilities, at least at the assessed timepoints. Our data thus show that SEPTIN2 but not SEPTIN9 is required for the formation or stabilization of a septin multimer in PNS myelin in vivo; however, its functional relevance remains to be established.
Collapse
Affiliation(s)
- Ann-Kristin Martens
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Michelle Erwig
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Julia Patzig
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Robert Fledrich
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | | | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| |
Collapse
|
7
|
Duong P, Ramesh R, Schneider A, Won S, Cooper AJ, Svaren J. Modulation of Schwann cell homeostasis by the BAP1 deubiquitinase. Glia 2023; 71:1466-1480. [PMID: 36790040 PMCID: PMC10073320 DOI: 10.1002/glia.24351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/16/2023]
Abstract
Schwann cell programming during myelination involves transcriptional networks that activate gene expression but also repress genes that are active in neural crest/embryonic differentiation of Schwann cells. We previously found that a Schwann cell-specific deletion of the EED subunit of the Polycomb Repressive Complex (PRC2) led to inappropriate activation of many such genes. Moreover, some of these genes become re-activated in the pro-regenerative response of Schwann cells to nerve injury, and we found premature activation of the nerve injury program in a Schwann cell-specific knockout of Eed. Polycomb-associated histone modifications include H3K27 trimethylation formed by PRC2 and H2AK119 monoubiquitination (H2AK119ub1), deposited by Polycomb repressive complex 1 (PRC1). We recently found dynamic regulation of H2AK119ub1 in Schwann cell genes after injury. Therefore, we hypothesized that H2AK119 deubiquitination modulates the dynamic polycomb repression of genes involved in Schwann cell maturation. To determine the role of H2AK119 deubiquitination, we generated a Schwann cell-specific knockout of the H2AK119 deubiquitinase Bap1 (BRCA1-associated protein). We found that loss of Bap1 causes tomacula formation, decreased axon diameters and eventual loss of myelinated axons. The gene expression changes are accompanied by redistribution of H2AK119ub1 and H3K27me3 modifications to extragenic sites throughout the genome. BAP1 interacts with OGT in the PR-DUB complex, and our data suggest that the PR-DUB complex plays a multifunctional role in repression of the injury program. Overall, our results indicate Bap1 is required to restrict the spread of polycomb-associated histone modifications in Schwann cells and to promote myelin homeostasis in peripheral nerve.
Collapse
Affiliation(s)
- Phu Duong
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Raghu Ramesh
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Andrew Schneider
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Seongsik Won
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Aaron J Cooper
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - John Svaren
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department Of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
8
|
El-Bazzal L, Ghata A, Estève C, Gadacha J, Quintana P, Castro C, Roeckel-Trévisiol N, Lembo F, Lenfant N, Mégarbané A, Borg JP, Lévy N, Bartoli M, Poitelon Y, Roubertoux PL, Delague V, Bernard-Marissal N. Imbalance of NRG1-ERBB2/3 signalling underlies altered myelination in Charcot-Marie-Tooth disease 4H. Brain 2023; 146:1844-1858. [PMID: 36314052 PMCID: PMC10151191 DOI: 10.1093/brain/awac402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 08/30/2022] [Accepted: 10/02/2022] [Indexed: 11/12/2022] Open
Abstract
Charcot-Marie-Tooth (CMT) disease is one of the most common inherited neurological disorders, affecting either axons from the motor and/or sensory neurons or Schwann cells of the peripheral nervous system (PNS) and caused by more than 100 genes. We previously identified mutations in FGD4 as responsible for CMT4H, an autosomal recessive demyelinating form of CMT disease. FGD4 encodes FRABIN, a GDP/GTP nucleotide exchange factor, particularly for the small GTPase Cdc42. Remarkably, nerves from patients with CMT4H display excessive redundant myelin figures called outfoldings that arise from focal hypermyelination, suggesting that FRABIN could play a role in the control of PNS myelination. To gain insights into the role of FGD4/FRABIN in Schwann cell myelination, we generated a knockout mouse model (Fgd4SC-/-), with conditional ablation of Fgd4 in Schwann cells. We show that the specific deletion of FRABIN in Schwann cells leads to aberrant myelination in vitro, in dorsal root ganglia neuron/Schwann cell co-cultures, as well as in vivo, in distal sciatic nerves from Fgd4SC-/- mice. We observed that those myelination defects are related to an upregulation of some interactors of the NRG1 type III/ERBB2/3 signalling pathway, which is known to ensure a proper level of myelination in the PNS. Based on a yeast two-hybrid screen, we identified SNX3 as a new partner of FRABIN, which is involved in the regulation of endocytic trafficking. Interestingly, we showed that the loss of FRABIN impairs endocytic trafficking, which may contribute to the defective NRG1 type III/ERBB2/3 signalling and myelination. Using RNA-Seq, in vitro, we identified new potential effectors of the deregulated pathways, such as ERBIN, RAB11FIP2 and MAF, thereby providing cues to understand how FRABIN contributes to proper ERBB2 trafficking or even myelin membrane addition through cholesterol synthesis. Finally, we showed that the re-establishment of proper levels of the NRG1 type III/ERBB2/3 pathway using niacin treatment reduces myelin outfoldings in nerves of CMT4H mice. Overall, our work reveals a new role of FRABIN in the regulation of NRG1 type III/ERBB2/3 NRG1signalling and myelination and opens future therapeutic strategies based on the modulation of the NRG1 type III/ERBB2/3 pathway to reduce CMT4H pathology and more generally other demyelinating types of CMT disease.
Collapse
Affiliation(s)
- Lara El-Bazzal
- Aix Marseille Univ, INSERM, MMG, U 1251, Marseille, France
| | - Adeline Ghata
- Aix Marseille Univ, INSERM, MMG, U 1251, Marseille, France
| | | | - Jihane Gadacha
- Aix Marseille Univ, INSERM, MMG, U 1251, Marseille, France
| | | | | | | | - Frédérique Lembo
- Aix Marseille Univ, INSERM, CNRS, CRCM, Institut Paoli-Calmettes, Marseille, France
| | | | - André Mégarbané
- Department of Human Genetics, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| | - Jean-Paul Borg
- Aix Marseille Univ, INSERM, CNRS, CRCM, Institut Paoli-Calmettes, Marseille, France
| | - Nicolas Lévy
- Aix Marseille Univ, INSERM, MMG, U 1251, Marseille, France
| | - Marc Bartoli
- Aix Marseille Univ, INSERM, MMG, U 1251, Marseille, France
| | - Yannick Poitelon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | | | | | | |
Collapse
|
9
|
Iyer M, Kantarci H, Ambiel N, Novak SW, Andrade LR, Lam M, Münch AE, Yu X, Khakh BS, Manor U, Zuchero JB. Oligodendrocyte calcium signaling sculpts myelin sheath morphology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.11.536299. [PMID: 37090556 PMCID: PMC10120717 DOI: 10.1101/2023.04.11.536299] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Myelin is essential for rapid nerve signaling and is increasingly found to play important roles in learning and in diverse diseases of the CNS. Morphological parameters of myelin such as sheath length and thickness are regulated by neuronal activity and can precisely tune conduction velocity, but the mechanisms controlling sheath morphology are poorly understood. Local calcium signaling has been observed in nascent myelin sheaths and can be modulated by neuronal activity. However, the role of calcium signaling in sheath formation and remodeling is unknown. Here, we used genetic tools to attenuate oligodendrocyte calcium signaling during active myelination in the developing mouse CNS. Surprisingly, we found that genetic calcium attenuation did not grossly affect the number of myelinated axons or myelin thickness. Instead, calcium attenuation caused striking myelination defects resulting in shorter, dysmorphic sheaths. Mechanistically, calcium attenuation reduced actin filaments in oligodendrocytes, and an intact actin cytoskeleton was necessary and sufficient to achieve accurate myelin morphology. Together, our work reveals a novel cellular mechanism required for accurate CNS myelin formation and provides mechanistic insight into how oligodendrocytes may respond to neuronal activity to sculpt myelin sheaths throughout the nervous system.
Collapse
|
10
|
Steyer AM, Buscham TJ, Lorenz C, Hümmert S, Eichel-Vogel MA, Schadt LC, Edgar JM, Köster S, Möbius W, Nave KA, Werner HB. Focused ion beam-scanning electron microscopy links pathological myelin outfoldings to axonal changes in mice lacking Plp1 or Mag. Glia 2023; 71:509-523. [PMID: 36354016 DOI: 10.1002/glia.24290] [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: 07/11/2022] [Revised: 10/10/2022] [Accepted: 10/17/2022] [Indexed: 11/11/2022]
Abstract
Healthy myelin sheaths consist of multiple compacted membrane layers closely encasing the underlying axon. The ultrastructure of CNS myelin requires specialized structural myelin proteins, including the transmembrane-tetraspan proteolipid protein (PLP) and the Ig-CAM myelin-associated glycoprotein (MAG). To better understand their functional relevance, we asked to what extent the axon/myelin-units display similar morphological changes if PLP or MAG are lacking. We thus used focused ion beam-scanning electron microscopy (FIB-SEM) to re-investigate axon/myelin-units side-by-side in Plp- and Mag-null mutant mice. By three-dimensional reconstruction and morphometric analyses, pathological myelin outfoldings extend up to 10 μm longitudinally along myelinated axons in both models. More than half of all assessed outfoldings emerge from internodal myelin. Unexpectedly, three-dimensional reconstructions demonstrated that both models displayed complex axonal pathology underneath the myelin outfoldings, including axonal sprouting. Axonal anastomosing was additionally observed in Plp-null mutant mice. Importantly, normal-appearing axon/myelin-units displayed significantly increased axonal diameters in both models according to quantitative assessment of electron micrographs. These results imply that healthy CNS myelin sheaths facilitate normal axonal diameters and shape, a function that is impaired when structural myelin proteins PLP or MAG are lacking.
Collapse
Affiliation(s)
- Anna M Steyer
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.,Electron Microscopy-City Campus, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Tobias J Buscham
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Charlotta Lorenz
- Institute for X-Ray Physics, University of Göttingen, Göttingen, Germany
| | - Sophie Hümmert
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Maria A Eichel-Vogel
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Leonie C Schadt
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Julia M Edgar
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.,Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Sarah Köster
- Institute for X-Ray Physics, University of Göttingen, Göttingen, Germany.,Cluster of Excellence 'Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells' (MBExC), University of Göttingen, Göttingen, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.,Electron Microscopy-City Campus, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany.,Cluster of Excellence 'Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells' (MBExC), University of Göttingen, Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| |
Collapse
|
11
|
Abstract
Myelin is a key evolutionary specialization and adaptation of vertebrates formed by the plasma membrane of glial cells, which insulate axons in the nervous system. Myelination not only allows rapid and efficient transmission of electric impulses in the axon by decreasing capacitance and increasing resistance but also influences axonal metabolism and the plasticity of neural circuits. In this review, we will focus on Schwann cells, the glial cells which form myelin in the peripheral nervous system. Here, we will describe the main extrinsic and intrinsic signals inducing Schwann cell differentiation and myelination and how myelin biogenesis is achieved. Finally, we will also discuss how the study of human disorders in which molecules and pathways relevant for myelination are altered has enormously contributed to the current knowledge on myelin biology.
Collapse
Affiliation(s)
- Alessandra Bolino
- Human Inherited Neuropathies Unit, Institute of Experimental Neurology INSPE, Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy.
| |
Collapse
|
12
|
Dysregulation of myelin synthesis and actomyosin function underlies aberrant myelin in CMT4B1 neuropathy. Proc Natl Acad Sci U S A 2021; 118:2009469118. [PMID: 33653949 DOI: 10.1073/pnas.2009469118] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Charcot-Marie-Tooth type 4B1 (CMT4B1) is a severe autosomal recessive demyelinating neuropathy with childhood onset, caused by loss-of-function mutations in the myotubularin-related 2 (MTMR2) gene. MTMR2 is a ubiquitously expressed catalytically active 3-phosphatase, which in vitro dephosphorylates the 3-phosphoinositides PtdIns3P and PtdIns(3,5)P 2, with a preference for PtdIns(3,5)P 2 A hallmark of CMT4B1 neuropathy are redundant loops of myelin in the nerve termed myelin outfoldings, which can be considered the consequence of altered growth of myelinated fibers during postnatal development. How MTMR2 loss and the resulting imbalance of 3'-phosphoinositides cause CMT4B1 is unknown. Here we show that MTMR2 by regulating PtdIns(3,5)P 2 levels coordinates mTORC1-dependent myelin synthesis and RhoA/myosin II-dependent cytoskeletal dynamics to promote myelin membrane expansion and longitudinal myelin growth. Consistent with this, pharmacological inhibition of PtdIns(3,5)P 2 synthesis or mTORC1/RhoA signaling ameliorates CMT4B1 phenotypes. Our data reveal a crucial role for MTMR2-regulated lipid turnover to titrate mTORC1 and RhoA signaling thereby controlling myelin growth.
Collapse
|
13
|
Ishii A, Furusho M, Bansal R. Mek/ERK1/2-MAPK and PI3K/Akt/mTOR signaling plays both independent and cooperative roles in Schwann cell differentiation, myelination and dysmyelination. Glia 2021; 69:2429-2446. [PMID: 34157170 DOI: 10.1002/glia.24049] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 04/29/2021] [Accepted: 06/04/2021] [Indexed: 01/15/2023]
Abstract
Multiple signals are involved in the regulation of developmental myelination by Schwann cells and in the maintenance of a normal myelin homeostasis throughout adult life, preserving the integrity of the axons in the PNS. Recent studies suggest that Mek/ERK1/2-MAPK and PI3K/Akt/mTOR intracellular signaling pathways play important, often overlapping roles in the regulation of myelination in the PNS. In addition, hyperactivation of these signaling pathways in Schwann cells leads to a late onset of various pathological changes in the sciatic nerves. However, it remains poorly understood whether these pathways function independently or sequentially or converge using a common mechanism to facilitate Schwann cell differentiation and myelin growth during development and in causing pathological changes in the adult animals. To address these questions, we analyzed multiple genetically modified mice using simultaneous loss- and constitutive gain-of-function approaches. We found that during development, the Mek/ERK1/2-MAPK pathway plays a primary role in Schwann cell differentiation, distinct from mTOR. However, during active myelination, ERK1/2 is dependent on mTOR signaling to drive the growth of the myelin sheath and regulate its thickness. Finally, our data suggest that peripheral nerve pathology during adulthood caused by hyperactivation of Mek/ERK1/2-MAPK or PI3K is likely to be independent or dependent on mTOR-signaling in different contexts. Thus, this study highlights the complexities in the roles played by two major intracellular signaling pathways in Schwann cells that affect their differentiation, myelination, and later PNS pathology and predicts that potential therapeutic modulation of these pathways in PNS neuropathies could be a complex process.
Collapse
Affiliation(s)
- Akihiro Ishii
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Miki Furusho
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Rashmi Bansal
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| |
Collapse
|
14
|
A Golgi-associated lipid kinase controls peripheral nerve myelination. Proc Natl Acad Sci U S A 2020; 117:30873-30875. [PMID: 33188090 DOI: 10.1073/pnas.2021130117] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
15
|
Skedsmo FS, Espenes A, Tranulis MA, Matiasek K, Gunnes G, Bjerkås I, Moe L, Røed SS, Berendt M, Fredholm M, Rohdin C, Shelton GD, Bruheim P, Stafsnes MH, Bartosova Z, Hermansen LC, Stigen Ø, Jäderlund KH. Impaired NDRG1 functions in Schwann cells cause demyelinating neuropathy in a dog model of Charcot-Marie-Tooth type 4D. Neuromuscul Disord 2020; 31:56-68. [PMID: 33334662 DOI: 10.1016/j.nmd.2020.11.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Revised: 11/13/2020] [Accepted: 11/18/2020] [Indexed: 11/25/2022]
Abstract
Mutations in the N-myc downstream-regulated gene 1 (NDRG1) cause degenerative polyneuropathy in ways that are poorly understood. We have investigated Alaskan Malamute dogs with neuropathy caused by a missense mutation in NDRG1. In affected animals, nerve levels of NDRG1 protein were reduced by more than 70% (p< 0.03). Nerve fibers were thinly myelinated, loss of large myelinated fibers was pronounced and teased fiber preparations showed both demyelination and remyelination. Inclusions of filamentous material containing actin were present in adaxonal Schwann cell cytoplasm and Schmidt-Lanterman clefts. This condition strongly resembles the human Charcot-Marie-Tooth type 4D. However, the focally folded myelin with adaxonal infoldings segregating the axon found in this study are ultrastructural changes not described in the human disease. Furthermore, lipidomic analysis revealed a profound loss of peripheral nerve lipids. Our data suggest that the low levels of mutant NDRG1 is insufficient to support Schwann cells in maintaining myelin homeostasis.
Collapse
Affiliation(s)
- Fredrik S Skedsmo
- Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ullevålsveien 72, 0454 Oslo, Norway.
| | - Arild Espenes
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ullevålsveien 72, 0454 Oslo, Norway
| | - Michael A Tranulis
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ullevålsveien 72, 0454 Oslo, Norway
| | - Kaspar Matiasek
- Section of Clinical & Comparative Neuropathology, Centre for Clinical Veterinary Medicine, Ludwig-Maximilians-Universität, Veterinärstr. 13, D-80539 Munich, Germany
| | - Gjermund Gunnes
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ullevålsveien 72, 0454 Oslo, Norway
| | - Inge Bjerkås
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ullevålsveien 72, 0454 Oslo, Norway
| | - Lars Moe
- Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ullevålsveien 72, 0454 Oslo, Norway
| | - Susan Skogtvedt Røed
- Department of Preclinical Sciences and Pathology, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ullevålsveien 72, 0454 Oslo, Norway
| | - Mette Berendt
- Department of Veterinary Clinical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Dyrlægevej 16, 1870 Frederiksberg C, Denmark
| | - Merete Fredholm
- Department of Veterinary and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Grønnegårdsvej 2, 1870 Frederiksberg C, Denmark
| | - Cecilia Rohdin
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Ultunaalléen 5A, 756 51 Uppsala, Sweden; Anicura Albano Small Animal Hospital, Rinkebyvägen 21, 182 36 Danderyd, Sweden
| | - G Diane Shelton
- Department of Pathology, School of Medicine, University of California San Diego, 9500 Gilman Drive, La Jolla, California 92093-0709, United States of America
| | - Per Bruheim
- Department of Biotechnology and Food Science, Faculty of Natural Sciences, Norwegian University of Science and Technology, Sem Sælands vei 6, 7034 Trondheim, Norway
| | - Marit H Stafsnes
- Department of Biotechnology and Food Science, Faculty of Natural Sciences, Norwegian University of Science and Technology, Sem Sælands vei 6, 7034 Trondheim, Norway
| | - Zdenka Bartosova
- Department of Biotechnology and Food Science, Faculty of Natural Sciences, Norwegian University of Science and Technology, Sem Sælands vei 6, 7034 Trondheim, Norway
| | - Lene C Hermansen
- Department of Plant Sciences, Faculty of Biosciences, Norwegian University of Life Sciences, Universitetstunet 3, 1433 Ås, Norway
| | - Øyvind Stigen
- Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ullevålsveien 72, 0454 Oslo, Norway
| | - Karin H Jäderlund
- Department of Companion Animal Clinical Sciences, Faculty of Veterinary Medicine, Norwegian University of Life Sciences, Ullevålsveien 72, 0454 Oslo, Norway
| |
Collapse
|
16
|
Lüningschrör P, Slotta C, Heimann P, Briese M, Weikert UM, Massih B, Appenzeller S, Sendtner M, Kaltschmidt C, Kaltschmidt B. Absence of Plekhg5 Results in Myelin Infoldings Corresponding to an Impaired Schwann Cell Autophagy, and a Reduced T-Cell Infiltration Into Peripheral Nerves. Front Cell Neurosci 2020; 14:185. [PMID: 32733205 PMCID: PMC7358705 DOI: 10.3389/fncel.2020.00185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 05/28/2020] [Indexed: 12/14/2022] Open
Abstract
Inflammation and dysregulation of the immune system are hallmarks of several neurodegenerative diseases. An activated immune response is considered to be the cause of myelin breakdown in demyelinating disorders. In the peripheral nervous system (PNS), myelin can be degraded in an autophagy-dependent manner directly by Schwann cells or by macrophages, which are modulated by T-lymphocytes. Here, we show that the NF-κB activator Pleckstrin homology containing family member 5 (Plekhg5) is involved in the regulation of both Schwann cell autophagy and recruitment of T-lymphocytes in peripheral nerves during motoneuron disease. Plekhg5-deficient mice show defective axon/Schwann cell units characterized by myelin infoldings in peripheral nerves. Even at late stages, Plekhg5-deficient mice do not show any signs of demyelination and inflammation. Using RNAseq, we identified a transcriptional signature for an impaired immune response in sciatic nerves, which manifested in a reduced number of CD4+ and CD8+ T-cells. These findings identify Plekhg5 as a promising target to impede myelin breakdown in demyelinating PNS disorders.
Collapse
Affiliation(s)
- Patrick Lüningschrör
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Carsten Slotta
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany.,Molecular Neurobiology, University of Bielefeld, Bielefeld, Germany
| | - Peter Heimann
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
| | - Michael Briese
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Ulrich M Weikert
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany
| | - Bita Massih
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Silke Appenzeller
- Core Unit Systems Medicine, University of Wuerzburg, Wuerzburg, Germany.,Comprehensive Cancer Center Mainfranken, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Michael Sendtner
- Institute of Clinical Neurobiology, University Hospital Wuerzburg, Wuerzburg, Germany
| | | | - Barbara Kaltschmidt
- Department of Cell Biology, University of Bielefeld, Bielefeld, Germany.,Molecular Neurobiology, University of Bielefeld, Bielefeld, Germany
| |
Collapse
|
17
|
Sawade L, Grandi F, Mignanelli M, Patiño-López G, Klinkert K, Langa-Vives F, Di Guardo R, Echard A, Bolino A, Haucke V. Rab35-regulated lipid turnover by myotubularins represses mTORC1 activity and controls myelin growth. Nat Commun 2020; 11:2835. [PMID: 32503983 PMCID: PMC7275063 DOI: 10.1038/s41467-020-16696-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 05/18/2020] [Indexed: 01/21/2023] Open
Abstract
Inherited peripheral neuropathies (IPNs) represent a broad group of disorders including Charcot-Marie-Tooth (CMT) neuropathies characterized by defects primarily arising in myelin, axons, or both. The molecular mechanisms by which mutations in nearly 100 identified IPN/CMT genes lead to neuropathies are poorly understood. Here we show that the Ras-related GTPase Rab35 controls myelin growth via complex formation with the myotubularin-related phosphatidylinositol (PI) 3-phosphatases MTMR13 and MTMR2, encoded by genes responsible for CMT-types 4B2 and B1 in humans, and found that it downregulates lipid-mediated mTORC1 activation, a pathway known to crucially regulate myelin biogenesis. Targeted disruption of Rab35 leads to hyperactivation of mTORC1 signaling caused by elevated levels of PI 3-phosphates and to focal hypermyelination in vivo. Pharmacological inhibition of phosphatidylinositol 3,5-bisphosphate synthesis or mTORC1 signaling ameliorates this phenotype. These findings reveal a crucial role for Rab35-regulated lipid turnover by myotubularins to repress mTORC1 activity and to control myelin growth. Charcot-Marie-Tooth (CMT) is an inherited peripheral neuropathy. Here, the authors show that Rab35 forms a complex with genes implicated in CMT, MTMR13 and MTMR2, which regulates myelin growth by controlling mTORC1 signaling through lipid turnover.
Collapse
Affiliation(s)
- Linda Sawade
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125, Berlin, Germany
| | - Federica Grandi
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Marianna Mignanelli
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy.,San Raffaele Vita-Salute University, Via Olgettina 60, 20132, Milan, Italy
| | - Genaro Patiño-López
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México, Federico Gómez. C.P, 06720, Ciudad de México, México
| | - Kerstin Klinkert
- Membrane Traffic and Cell Division Lab, Institut Pasteur, UMR3691, CNRS, 25-28 rue du Dr Roux, F-75015, Paris, France.,Sorbonne Université, Collège doctoral, F-75005, Paris, France
| | - Francina Langa-Vives
- Centre d'Ingénierie Génétique Murine, Institut Pasteur, 25-28 rue du Dr Roux, F-75015, Paris, France
| | - Roberta Di Guardo
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Arnaud Echard
- Membrane Traffic and Cell Division Lab, Institut Pasteur, UMR3691, CNRS, 25-28 rue du Dr Roux, F-75015, Paris, France
| | - Alessandra Bolino
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy.
| | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125, Berlin, Germany. .,Freie Universität Berlin, Faculty of Biology, Chemistry and Pharmacy, 14195, Berlin, Germany. .,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, NeuroCure Cluster of Excellence, 10117, Berlin, Germany.
| |
Collapse
|
18
|
Dahlin LB, Rix KR, Dahl VA, Dahl AB, Jensen JN, Cloetens P, Pacureanu A, Mohseni S, Thomsen NOB, Bech M. Three-dimensional architecture of human diabetic peripheral nerves revealed by X-ray phase contrast holographic nanotomography. Sci Rep 2020; 10:7592. [PMID: 32371896 PMCID: PMC7200696 DOI: 10.1038/s41598-020-64430-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/14/2020] [Indexed: 01/06/2023] Open
Abstract
A deeper knowledge of the architecture of the peripheral nerve with three-dimensional (3D) imaging of the nerve tissue at the sub-cellular scale may contribute to unravel the pathophysiology of neuropathy. Here we demonstrate the feasibility of X-ray phase contrast holographic nanotomography to enable 3D imaging of nerves at high resolution, while covering a relatively large tissue volume. We show various subcomponents of human peripheral nerves in biopsies from patients with type 1 and 2 diabetes and in a healthy subject. Together with well-organized, parallel myelinated nerve fibres we show regenerative clusters with twisted nerve fibres, a sprouted axon from a node of Ranvier and other specific details. A novel 3D construction (with movie created) of a node of Ranvier with end segment of a degenerated axon and sprout of a regenerated one is captured. Many of these architectural elements are not described in the literature. Thus, X-ray phase contrast holographic nanotomography enables identifying specific morphological structures in 3D in peripheral nerve biopsies from a healthy subject and from patients with type 1 and 2 diabetes.
Collapse
Affiliation(s)
- Lars B Dahlin
- Department of Translational Medicine - Hand Surgery, Lund University, Jan Waldenströms gata 5, SE-205 02, Malmö, Sweden
- Department of Hand Surgery, Skåne University Hospital, Jan Waldenströms gata 5, SE-205 02, Malmö, Sweden
| | - Kristian R Rix
- Niels Bohr Institute, Copenhagen University, Blegdamsvej 17, 2100, Copenhagen, Denmark
| | - Vedrana A Dahl
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Richard Petersens Plads Building 324, 2800, Kgs Lyngby, Denmark
| | - Anders B Dahl
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Richard Petersens Plads Building 324, 2800, Kgs Lyngby, Denmark
| | - Janus N Jensen
- Department of Applied Mathematics and Computer Science, Technical University of Denmark, Richard Petersens Plads Building 324, 2800, Kgs Lyngby, Denmark
| | - Peter Cloetens
- ESRF, The European Synchrotron, 71 Avenue des Martyrs, 38000, Grenoble, France
| | - Alexandra Pacureanu
- ESRF, The European Synchrotron, 71 Avenue des Martyrs, 38000, Grenoble, France
| | - Simin Mohseni
- Department of Biomedical and Clinical Sciences, Linköping University, 581 83, Linköping, Sweden
| | - Niels O B Thomsen
- Department of Hand Surgery, Skåne University Hospital, Jan Waldenströms gata 5, SE-205 02, Malmö, Sweden
| | - Martin Bech
- Department of Medical Radiation Physics, Clinical Sciences Lund, Lund University, 221 85, Lund, Sweden.
| |
Collapse
|
19
|
Song GJ, Gupta DP, Rahman MH, Park HT, Al Ghouleh I, Bisello A, Lee MG, Park JY, Park HH, Jun JH, Chung KW, Choi BO, Suk K. Loss-of-function of EBP50 is a new cause of hereditary peripheral neuropathy: EBP50 functions in peripheral nerve system. Glia 2020; 68:1794-1809. [PMID: 32077526 DOI: 10.1002/glia.23805] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 02/04/2020] [Accepted: 02/07/2020] [Indexed: 12/12/2022]
Abstract
Finding causative genetic mutations is important in the diagnosis and treatment of hereditary peripheral neuropathies. This study was conducted to find new genes involved in the pathophysiology of hereditary peripheral neuropathy. We identified a new mutation in the EBP50 gene, which is co-segregated with neuropathic phenotypes, including motor and sensory deficit in a family with Charcot-Marie-Tooth disease. EBP50 is known to be important for the formation of microvilli in epithelial cells, and the discovery of this gene mutation allowed us to study the function of EBP50 in the nervous system. EBP50 was strongly expressed in the nodal and paranodal regions of sciatic nerve fibers, where Schwann cell microvilli contact the axolemma, and at the growth tips of primary Schwann cells. In addition, EBP50 expression was decreased in mouse models of peripheral neuropathy. Knockout mice were used to study EBP50 function in the peripheral nervous system. Interestingly motor function deficit and abnormal histology of nerve fibers were observed in EBP50+/- heterozygous mice at 12 months of age, but not 3 months. in vitro studies using Schwann cells showed that NRG1-induced AKT activation and migration were significantly reduced in cells overexpressing the I325V mutant of EBP50 or cells with knocked-down EBP50 expression. In conclusion, we show for the first time that loss of function due to EBP50 gene deficiency or mutation can cause peripheral neuropathy.
Collapse
Affiliation(s)
- Gyun Jee Song
- Department of Medical Science, Institute for Bio-Medical Convergence, Catholic Kwandong University, International St. Mary's Hospital, Incheon, Republic of Korea
| | - Deepak Prasad Gupta
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Md Habibur Rahman
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Hwan Tae Park
- Department of Molecular Neuroscience, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Imad Al Ghouleh
- Division of Cardiology, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Alessandro Bisello
- Department of Pharmacology and Chemical Biology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Maan-Gee Lee
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| | - Jae-Yong Park
- School of Biosystems and Biomedical Sciences, College of Health Sciences, Korea University, Seoul, Republic of Korea
| | - Hyun Ho Park
- College of Pharmacy, Chung-Ang University, Seoul, Republic of Korea
| | - Jin Hyun Jun
- Department of Senior Healthcare, BK21 Plus Program, Graduate School of Eulji University, Department of Biomedical Laboratory Science, College of Health Science, Eulji University, Seongnam, Republic of Korea
| | - Ki Wha Chung
- Department of Biological Sciences, Kongju National University, Gongju, Republic of Korea
| | - Byung-Ok Choi
- Department of Neurology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Kyoungho Suk
- Department of Pharmacology, Brain Science and Engineering Institute, BK21 Plus KNU Biomedical Convergence Program, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
| |
Collapse
|
20
|
Clarke BA, Majumder S, Zhu H, Lee YT, Kono M, Li C, Khanna C, Blain H, Schwartz R, Huso VL, Byrnes C, Tuymetova G, Dunn TM, Allende ML, Proia RL. The Ormdl genes regulate the sphingolipid synthesis pathway to ensure proper myelination and neurologic function in mice. eLife 2019; 8:51067. [PMID: 31880535 PMCID: PMC6934382 DOI: 10.7554/elife.51067] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Accepted: 12/09/2019] [Indexed: 12/23/2022] Open
Abstract
Sphingolipids are membrane and bioactive lipids that are required for many aspects of normal mammalian development and physiology. However, the importance of the regulatory mechanisms that control sphingolipid levels in these processes is not well understood. The mammalian ORMDL proteins (ORMDL1, 2 and 3) mediate feedback inhibition of the de novo synthesis pathway of sphingolipids by inhibiting serine palmitoyl transferase in response to elevated ceramide levels. To understand the function of ORMDL proteins in vivo, we studied mouse knockouts (KOs) of the Ormdl genes. We found that Ormdl1 and Ormdl3 function redundantly to suppress the levels of bioactive sphingolipid metabolites during myelination of the sciatic nerve. Without proper ORMDL-mediated regulation of sphingolipid synthesis, severe dysmyelination results. Our data indicate that the Ormdls function to restrain sphingolipid metabolism in order to limit levels of dangerous metabolic intermediates that can interfere with essential physiological processes such as myelination.
Collapse
Affiliation(s)
- Benjamin A Clarke
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| | - Saurav Majumder
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| | - Hongling Zhu
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| | - Y Terry Lee
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| | - Mari Kono
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| | - Cuiling Li
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| | - Caroline Khanna
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| | - Hailey Blain
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| | - Ronit Schwartz
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| | - Vienna L Huso
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| | - Colleen Byrnes
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| | - Galina Tuymetova
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| | - Teresa M Dunn
- Department of Biochemistry, Uniformed Services University of the Health Sciences, Bethesda, United States
| | - Maria L Allende
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| | - Richard L Proia
- Genetics of Development and Disease Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, United States
| |
Collapse
|
21
|
Torii T, Miyamoto Y, Yamauchi J. Cellular Signal-Regulated Schwann Cell Myelination and Remyelination. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1190:3-22. [PMID: 31760634 DOI: 10.1007/978-981-32-9636-7_1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Increasing studies have demonstrated multiple signaling molecules responsible for oligodendrocytes and Schwann cells development such as migration, differentiation, myelination, and axo-glial interaction. However, complicated roles in these events are still poorly understood. This chapter focuses on well established intracellular signaling transduction and recent topics that control myelination and are elucidated from accumulating evidences. The underlying molecular mechanisms, which involved in membrane trafficking through small GTPase Arf6 and its activator cytohesins, demonstrate the crosstalk between well established intracellular signaling transduction and a new finding signaling pathway in glial cells links to physiological phenotype and essential role in peripheral nerve system (PNS). Since Arf family proteins affect the expression levels of myelin protein zero (MPZ) and Krox20, which is a transcription factor regulatory factor in early developmental stages of Schwann cells, Arf proteins likely to be key regulator for Schwann cells development. Herein, we discuss how intracellular signaling transductions in Schwann cells associate with myelination in CNS and PNS.
Collapse
Affiliation(s)
- Tomohiro Torii
- Graduate School of Brain Science, Doshisha University, Kyotanabe-shi, Kyoto, Japan
| | - Yuki Miyamoto
- Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya, Tokyo, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Science, Hachioji, Tokyo, Japan.
| |
Collapse
|
22
|
Hackett AR, Strickland A, Milbrandt J. Disrupting insulin signaling in Schwann cells impairs myelination and induces a sensory neuropathy. Glia 2019; 68:963-978. [PMID: 31758725 DOI: 10.1002/glia.23755] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/28/2019] [Accepted: 11/05/2019] [Indexed: 12/14/2022]
Abstract
Although diabetic mice have been studied for decades, little is known about the cell type specific contributions to diabetic neuropathy (DN). Schwann cells (SCs) myelinate and provide trophic support to peripheral nervous system axons. Altered SC metabolism leads to myelin defects, which can be seen both in inherited and DNs. How SC metabolism is altered in DN is not fully understood, but it is clear that insulin resistance underlies impaired lipid metabolism in many cell types throughout the body via the phosphoinositide 3-kinase/protein kinase b (PKB)/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway. Here, we created an insulin resistant SC by deleting both insulin receptor (INSR) and insulin-like growth factor receptor 1 (IGF1R), to determine the role of this signaling pathway in development and response to injury in order to understand SC defects in DN. We found that myelin is thinner throughout development and adulthood in INSR/IGF1R Schwann cell specific knock out mice. The nerves of these mutant mice had reduced expression of key genes that mediate fatty acid and cholesterol synthesis due to reduced mTOR-sterol regulatory element-binding protein signaling. In adulthood, these mice show sensory neuropathy phenotypes reminiscent of diabetic mice. Altogether, these data suggest that SCs may play an important role in DN and targeting their metabolism could lead to new therapies for DN.
Collapse
Affiliation(s)
- Amber R Hackett
- Department of Genetics, Washington University School of Medicine, Saint Louis, Missouri
| | - Amy Strickland
- Department of Genetics, Washington University School of Medicine, Saint Louis, Missouri
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine, Saint Louis, Missouri
| |
Collapse
|
23
|
Fledrich R, Kungl T, Nave KA, Stassart RM. Axo-glial interdependence in peripheral nerve development. Development 2019; 146:146/21/dev151704. [PMID: 31719044 DOI: 10.1242/dev.151704] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
During the development of the peripheral nervous system, axons and myelinating Schwann cells form a unique symbiotic unit, which is realized by a finely tuned network of molecular signals and reciprocal interactions. The importance of this complex interplay becomes evident after injury or in diseases in which aspects of axo-glial interaction are perturbed. This Review focuses on the specific interdependence of axons and Schwann cells in peripheral nerve development that enables axonal outgrowth, Schwann cell lineage progression, radial sorting and, finally, formation and maintenance of the myelin sheath.
Collapse
Affiliation(s)
- Robert Fledrich
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany .,Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Theresa Kungl
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany.,Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Ruth M Stassart
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany .,Department of Neuropathology, University Clinic Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
24
|
Li L, Yao L, Wang F, Zhang Z. Knock-down of JAK2 and PTEN on pain behavior in rat model of trigeminal neuropathic pain. Gene 2019; 719:144080. [PMID: 31454541 DOI: 10.1016/j.gene.2019.144080] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 08/23/2019] [Accepted: 08/23/2019] [Indexed: 10/26/2022]
Abstract
Trigeminal neuropathic pain is seen as a huge clinical challenge. Although numerous drugs have been developed to treat the condition, some patients have shown intolerance to the drugs and thus continue to suffer. In the present study, a rat model of trigeminal neuropathic pain was established using incorrectly positioned dental implants, which had various manifestations that were similar to human trigeminal neuropathic pain. Using this model, we investigated the differential regulation of JAK2 and PTEN. Firstly, we examined the expression of JAK2 and PTEN in the medullary dorsal horn. After inhibiting JAK2/PTEN, we evaluated nociception-related behavioral alterations. The rat models were established by replacing the left lower second molar with a mini dental implant. Immunoblot assay and immunofluorescence experiments indicated high expression of JAK2 and PTEN in medullary dorsal horn after the nerve injury, which attained plateau levels on post-operative day (POD) 5-10 and 10-20. Administration of adenovirus-shRNA-JAK2 on POD 1 reduced mechanical allodynia and downstream STAT activation. Meanwhile, the administration of adenovirus-shRNA-PTEN on POD 1 attenuated mechanical allodynia while upregulating AKT. In addition to postoperative JAK2 and PTEN activation, dexmedetomidine treatment (10 mg/kg) also modulated the downstream sensors of these signaling molecules. These data suggest that JAK2 and PTEN are pivotal to the development of trigeminal neuropathic pain, and that JAK2 and PTEN suppression alleviates the neuropathic pain.
Collapse
Affiliation(s)
- Linan Li
- Department of Pain, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Sciences, Xiangyang, Hubei, China
| | - Lingling Yao
- Department of Pain, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Sciences, Xiangyang, Hubei, China
| | - Fengjuan Wang
- Department of Stomatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Sciences, Xiangyang, Hubei, China.
| | - Zhihong Zhang
- Department of Stomatology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Sciences, Xiangyang, Hubei, China.
| |
Collapse
|
25
|
Zhou Y, Miles JR, Tavori H, Lin M, Khoshbouei H, Borchelt DR, Bazick H, Landreth GE, Lee S, Fazio S, Notterpek L. PMP22 Regulates Cholesterol Trafficking and ABCA1-Mediated Cholesterol Efflux. J Neurosci 2019; 39:5404-5418. [PMID: 31061090 PMCID: PMC6607759 DOI: 10.1523/jneurosci.2942-18.2019] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 04/22/2019] [Accepted: 04/26/2019] [Indexed: 12/21/2022] Open
Abstract
The absence of functional peripheral myelin protein 22 (PMP22) is associated with shortened lifespan in rodents and severe peripheral nerve myelin abnormalities in several species including humans. Schwann cells and nerves from PMP22 knock-out (KO) mice show deranged cholesterol distribution and aberrant lipid raft morphology, supporting an unrecognized role for PMP22 in cellular lipid metabolism. To examine the mechanisms underlying these abnormalities, we studied Schwann cells and nerves from male and female PMP22 KO mice. Whole-cell current-clamp recordings in cultured Schwann cells revealed increased membrane capacitance and decreased membrane resistance in the absence of PMP22, which was consistent with a reduction in membrane cholesterol. Nerves from PMP22-deficient mice contained abnormal lipid droplets, with both mRNA and protein levels of apolipoprotein E (apoE) and ATP-binding cassette transporter A1 (ABCA1) being highly upregulated. Despite the upregulation of ABCA1 and apoE, the absence of PMP22 resulted in reduced localization of the transporter to the cell membrane and diminished secretion of apoE. The absence of PMP22 also impaired ABCA1-mediated cholesterol efflux capacity. In nerves from ABCA1 KO mice, the expression of PMP22 was significantly elevated and the subcellular processing of the overproduced protein was aberrant. In wild-type samples, double immunolabeling identified overlapping distribution of PMP22 and ABCA1 at the Schwann cell plasma membrane and the two proteins were coimmunoprecipitated from Schwann cell and nerve lysates. Together, these results reveal a novel role for PMP22 in regulating lipid metabolism and cholesterol trafficking through functional interaction with the cholesterol efflux regulatory protein ABCA1.SIGNIFICANCE STATEMENT Understanding the subcellular events that underlie abnormal myelin formation in hereditary neuropathies is critical for advancing therapy development. Peripheral myelin protein 22 (PMP22) is an essential peripheral myelin protein because its genetic abnormalities account for ∼80% of hereditary neuropathies. Here, we demonstrate that in the absence of PMP22, the cellular and electrophysiological properties of the Schwann cells' plasma membrane are altered and cholesterol trafficking and lipid homeostasis are perturbed. The molecular mechanisms for these abnormalities involve a functional interplay among PMP22, cholesterol, apolipoprotein E, and the major cholesterol-efflux transporter protein ATP-binding cassette transporter A1 (ABCA1). These findings establish a critical role for PMP22 in the maintenance of cholesterol homeostasis in Schwann cells.
Collapse
Affiliation(s)
| | - Joshua R Miles
- Department of Medicine
- Department of Physiology and Pharmacology, Knight Cardiovascular Institute, Center of Preventive Cardiology, Oregon Health & Science University, Portland, Oregon 27332, and
| | - Hagai Tavori
- Department of Medicine
- Department of Physiology and Pharmacology, Knight Cardiovascular Institute, Center of Preventive Cardiology, Oregon Health & Science University, Portland, Oregon 27332, and
| | | | | | | | | | - Gary E Landreth
- Department of Neurosciences, Indiana University, Indianapolis, Indiana 46202
| | | | - Sergio Fazio
- Department of Medicine
- Department of Physiology and Pharmacology, Knight Cardiovascular Institute, Center of Preventive Cardiology, Oregon Health & Science University, Portland, Oregon 27332, and
| | - Lucia Notterpek
- Department of Neuroscience,
- Department of Neurology, College of Medicine, McKnight Brain Institute, University of Florida, Gainesville, Florida 32610
| |
Collapse
|
26
|
Meng X, Maurel P, Lam I, Heffernan C, Stiffler MA, McBeath G, Salzer JL. Necl-4/Cadm4 recruits Par-3 to the Schwann cell adaxonal membrane. Glia 2019; 67:884-895. [PMID: 30585357 PMCID: PMC7138615 DOI: 10.1002/glia.23578] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 11/18/2018] [Accepted: 11/26/2018] [Indexed: 12/12/2022]
Abstract
Interactions between axons and Schwann cells are essential for the acquisition of Schwann cell radial and longitudinal polarity and myelin sheath assembly. In the internode, the largest of these longitudinal domains, axon-Schwann cell interactions are mediated by the Nectin-like (Necl) cell adhesion proteins, also known as SynCAMs or Cadms. In particular, Necl-1/Cadm3 expressed on the axon surface binds to Necl-4/Cadm4 expressed along the adaxonal membrane of myelinating Schwann cells. Necl-4 promotes myelination in vitro and is required for the timely onset of myelination and the fidelity of the organization of the myelin sheath and the internode in vivo. A key question is the identity of the downstream effectors of Necl-4 that mediate its effects. The cytoplasmic terminal region (CTR) of Necl-4 contains a PDZ-domain binding motif. Accordingly, we used the CTR of Necl-4 in an unbiased proteomic screen of PDZ-domain proteins. We identify Par-3, a multi-PDZ domain containing protein of the Par-aPKC polarity complex previously implicated in myelination, as an interacting protein. Necl-4 and Par-3 are colocalized along the inner Schwann cell membrane and coprecipitate from Schwann cell lysates. The CTR of Necl-4 binds to the first PDZ domain of Par-3 thereby recruiting Par-3 to sites of Necl-4/Necl-1 interaction. Knockdown of Necl-4 perturbs Par-3 localization to the inner membrane of Schwann cells in myelinating co-cultures. These findings implicate interactions of Necl-1/Necl-4 in the recruitment of Par-3 to the Schwann cell adaxonal membrane and the establishment of Schwann cell radial polarity.
Collapse
Affiliation(s)
- Xiaosong Meng
- Departments of Neuroscience and Physiology and Neurology,
the Neuroscience Institute, NYU Langone Medical Center, New York, NY 10016
| | - Patrice Maurel
- Department of Biological Sciences, Rutgers, Newark, NJ
07102
| | - Isabel Lam
- Dana-Faber Cancer Institute, Boston, MA 02215
| | - Corey Heffernan
- Department of Biological Sciences, Rutgers, Newark, NJ
07102
| | | | - Gavin McBeath
- Department of Systems Biology, Harvard Medical School,
Boston, MA 02115
| | - James L. Salzer
- Departments of Neuroscience and Physiology and Neurology,
the Neuroscience Institute, NYU Langone Medical Center, New York, NY 10016
- Departments of Neuroscience and Physiology and Neurology,
the Neuroscience Institute, NYU Langone Medical Center, New York, NY 10016
| |
Collapse
|
27
|
Belin S, Ornaghi F, Shackleford G, Wang J, Scapin C, Lopez-Anido C, Silvestri N, Robertson N, Williamson C, Ishii A, Taveggia C, Svaren J, Bansal R, Schwab MH, Nave K, Fratta P, D’Antonio M, Poitelon Y, Feltri ML, Wrabetz L. Neuregulin 1 type III improves peripheral nerve myelination in a mouse model of congenital hypomyelinating neuropathy. Hum Mol Genet 2019; 28:1260-1273. [PMID: 30535360 PMCID: PMC6452193 DOI: 10.1093/hmg/ddy420] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/06/2018] [Accepted: 12/02/2018] [Indexed: 12/13/2022] Open
Abstract
Myelin sheath thickness is precisely regulated and essential for rapid propagation of action potentials along myelinated axons. In the peripheral nervous system, extrinsic signals from the axonal protein neuregulin 1 (NRG1) type III regulate Schwann cell fate and myelination. Here we ask if modulating NRG1 type III levels in neurons would restore myelination in a model of congenital hypomyelinating neuropathy (CHN). Using a mouse model of CHN, we improved the myelination defects by early overexpression of NRG1 type III. Surprisingly, the improvement was independent from the upregulation of Egr2 or essential myelin genes. Rather, we observed the activation of MAPK/ERK and other myelin genes such as peripheral myelin protein 2 and oligodendrocyte myelin glycoprotein. We also confirmed that the permanent activation of MAPK/ERK in Schwann cells has detrimental effects on myelination. Our findings demonstrate that the modulation of axon-to-glial NRG1 type III signaling has beneficial effects and improves myelination defects during development in a model of CHN.
Collapse
Affiliation(s)
- Sophie Belin
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, NY, USA
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Francesca Ornaghi
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, NY, USA
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
- SR-TIGET, IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | - Ghjuvan’Ghjacumu Shackleford
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, NY, USA
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Jie Wang
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Cristina Scapin
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | | | - Nicholas Silvestri
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Neil Robertson
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Courtney Williamson
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, NY, USA
| | - Akihiro Ishii
- Department of Neuroscience, University of Connecticut Medical School, Farmington, CT, USA
| | - Carla Taveggia
- Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - John Svaren
- Waisman Center, University of Wisconsin–Madison, Madison, WI, USA
| | - Rashmi Bansal
- Department of Neuroscience, University of Connecticut Medical School, Farmington, CT, USA
| | - Markus H Schwab
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Klaus Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Pietro Fratta
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London, UK
| | - Maurizio D’Antonio
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Yannick Poitelon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - M Laura Feltri
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, NY, USA
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Lawrence Wrabetz
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, NY, USA
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
28
|
Davies AJ, Kim HW, Gonzalez-Cano R, Choi J, Back SK, Roh SE, Johnson E, Gabriac M, Kim MS, Lee J, Lee JE, Kim YS, Bae YC, Kim SJ, Lee KM, Na HS, Riva P, Latremoliere A, Rinaldi S, Ugolini S, Costigan M, Oh SB. Natural Killer Cells Degenerate Intact Sensory Afferents following Nerve Injury. Cell 2019; 176:716-728.e18. [PMID: 30712871 PMCID: PMC6418410 DOI: 10.1016/j.cell.2018.12.022] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 11/12/2018] [Accepted: 12/14/2018] [Indexed: 02/06/2023]
Abstract
Sensory axons degenerate following separation from their cell body, but partial injury to peripheral nerves may leave the integrity of damaged axons preserved. We show that an endogenous ligand for the natural killer (NK) cell receptor NKG2D, Retinoic Acid Early 1 (RAE1), is re-expressed in adult dorsal root ganglion neurons following peripheral nerve injury, triggering selective degeneration of injured axons. Infiltration of cytotoxic NK cells into the sciatic nerve by extravasation occurs within 3 days following crush injury. Using a combination of genetic cell ablation and cytokine-antibody complex stimulation, we show that NK cell function correlates with loss of sensation due to degeneration of injured afferents and reduced incidence of post-injury hypersensitivity. This neuro-immune mechanism of selective NK cell-mediated degeneration of damaged but intact sensory axons complements Wallerian degeneration and suggests the therapeutic potential of modulating NK cell function to resolve painful neuropathy through the clearance of partially damaged nerves. Cytotoxic NK cells infiltrate the damaged peripheral nerve within days of injury Injured sensory axons express NKG2D ligand RAE1 to signal degeneration by NK cells Clearance of damaged axons reduces development of chronic pain after nerve injury NK cells complement Wallerian degeneration to aid functional regeneration of PNS
Collapse
Affiliation(s)
- Alexander J Davies
- Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea; Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Hyoung Woo Kim
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Rafael Gonzalez-Cano
- Departments of Anesthesia and Neurobiology, Children's Hospital Boston and Harvard Medical School, Boston, MA 02115, USA
| | - Jahyang Choi
- Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea
| | - Seung Keun Back
- Departments of Physiology, Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Seung Eon Roh
- Department of Physiology, Seoul National University College of Medicine, Seoul 03087, Republic of Korea
| | - Errin Johnson
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, UK
| | - Melanie Gabriac
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Mi-Sun Kim
- Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea
| | - Jaehee Lee
- Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea; Departments of Physiology, Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Jeong Eun Lee
- Departments of Physiology, Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Yun Sook Kim
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 700-412, Korea
| | - Yong Chul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu 700-412, Korea
| | - Sang Jeong Kim
- Department of Physiology, Seoul National University College of Medicine, Seoul 03087, Republic of Korea
| | - Kyung-Mi Lee
- Departments of Physiology, Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Heung Sik Na
- Departments of Physiology, Biochemistry and Molecular Biology, College of Medicine, Korea University, Seoul 02841, Republic of Korea
| | - Priscilla Riva
- Departments of Anesthesia and Neurobiology, Children's Hospital Boston and Harvard Medical School, Boston, MA 02115, USA
| | - Alban Latremoliere
- Neurosurgery Department, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
| | - Simon Rinaldi
- Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Sophie Ugolini
- Aix Marseille Univ, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Marseille, France
| | - Michael Costigan
- Departments of Anesthesia and Neurobiology, Children's Hospital Boston and Harvard Medical School, Boston, MA 02115, USA.
| | - Seog Bae Oh
- Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, Seoul 03080, Republic of Korea; Department of Brain and Cognitive Sciences, College of Natural Sciences, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
29
|
Abstract
The use of electron microscopy allows analysis of the microarchitecture of peripheral nerves both during development and in the processes of nerve regeneration and repair.We describe a novel method for the rapid analysis and quantification of myelin in peripheral nerve using a low vacuum scanning electron microscopy protocol. For this methodology, excised nerves are prepared for traditional transmission electron microscopy (TEM) imaging, but at the stage where semi-thin sections would be taken, the resin block is instead imaged at low vacuum in the scanning electron microscope (SEM) using the backscattered electron signal. Any features in the tissue which have incorporated high concentrations of osmium from the fixation process (e.g., myelin) appear as bright regions in the image.Myelin therefore is easily identifiable in the images, and since there is a high contrast difference between it and the surrounding tissue, automated measurements for myelin thickness (e.g., G ratio) using standard and freely available image analysis software (e.g., ImageJ) are easily achievable, consistent, and repeatable. This method therefore greatly speeds up the analysis of nerve samples and, in many cases, will obviate the need for ultrathin sections and TEM for analyzing nerve morphology.Low vacuum (LV) SEM imaging has benefits compared with light microscopy; magnification is continuous, resolution is higher, and contrast and brightness are controllable. Since the resin block does not need a metal coating for imaging in LV mode, once the images have been collected, the block is still ready to section for both light microscopy (LM) and TEM.
Collapse
|
30
|
Terada N, Saitoh Y, Kamijo A, Yamauchi J, Ohno N, Sakamoto T. Structures and Molecular Composition of Schmidt-Lanterman Incisures. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1190:181-198. [PMID: 31760645 DOI: 10.1007/978-981-32-9636-7_12] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Schmidt-Lanterman incisure (SLI) is a circular-truncated cone shape in the myelin internode that is a specific feature of myelinated nerve fibers formed in Schwann cells in the peripheral nervous system (PNS). The SLI circular-truncated cones elongate like spring at the narrow sites of beaded appearance nerve fibers under the stretched condition. In this chapter, we demonstrate various molecular complexes in SLI, and especially focus on membrane skeleton, protein 4.1G-membrane protein palmitoylated 6 (MPP6)-cell adhesion molecule 4 (CADM4). 4.1G was essential for the molecular targeting of MPP6 and CADM4 in SLI. Motor activity and myelin ultrastructures were abnormal in 4.1G-deficient mice, indicating the 4.1G function as a signal for proper formation of myelin in PNS. Thus, SLI probably has potential roles in the regulation of adhesion and signal transduction as well as in structural stability in Schwann cell myelin formation.
Collapse
Affiliation(s)
- Nobuo Terada
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, Matsumoto City, Nagano, Japan.
| | - Yurika Saitoh
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, Matsumoto City, Nagano, Japan
- Center for Medical Education, Teikyo University of Science, Adachi-ku, Tokyo, Japan
| | - Akio Kamijo
- Health Science Division, Department of Medical Sciences, Shinshu University Graduate School of Medicine, Science and Technology, Matsumoto City, Nagano, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Science, Hachioji, Tokyo, Japan
| | - Nobuhiko Ohno
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Aichi, Japan
- Department of Anatomy, Division of Histology and Cell Biology, Jichi Medical University, School of Medicine, Shimotsuke, Japan
| | - Takeharu Sakamoto
- Division of Cellular and Molecular Biology, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo, Japan
| |
Collapse
|
31
|
The membrane palmitoylated protein, MPP6, is involved in myelin formation in the mouse peripheral nervous system. Histochem Cell Biol 2018; 151:385-394. [PMID: 30357511 DOI: 10.1007/s00418-018-1745-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2018] [Indexed: 01/01/2023]
Abstract
A membrane skeletal molecular complex, protein 4.1G-membrane palmitoylated protein 6 (MPP6)-Lin7-cell adhesion molecule 4 (CADM4), is incorporated in Schwann cells, especially in Schmidt-Lanterman incisures (SLIs), in the mouse peripheral nervous system (PNS). MPP6, Lin7, and CADM4 are transported to SLIs by 4.1G. In this study, we created MPP6-deficient mice and evaluated myelin structure and MPP6 protein complexes. In SLIs in MPP6-deficient nerves, Lin7 was rarely detected by immunohistochemistry and western blotting, but the localization and amount of CADM4 and 4.1G were not altered. Motor activity was not significantly impaired in a tail-suspension test, but the sciatic nerves of MPP6-deficient mice had thicker myelin in internodes by electron microscopy compared to that of wild-type mice. These results indicate that the MPP6-Lin7 complex regulates myelin formation.
Collapse
|
32
|
Shi Q, Saifetiarova J, Taylor AM, Bhat MA. mTORC1 Activation by Loss of Tsc1 in Myelinating Glia Causes Downregulation of Quaking and Neurofascin 155 Leading to Paranodal Domain Disorganization. Front Cell Neurosci 2018; 12:201. [PMID: 30050412 PMCID: PMC6052123 DOI: 10.3389/fncel.2018.00201] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 06/20/2018] [Indexed: 11/29/2022] Open
Abstract
Mutations in human tuberous sclerosis complex (TSC) genes TSC1 and TSC2 are the leading causes of developmental brain abnormalities and large tumors in other tissues. Murine Tsc1/2 have been shown to negatively regulate the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway in most tissues, and this pathway has been shown to be essential for proper oligodendrocytes/Schwann cell differentiation and myelination. Here, we report that ablation of Tsc1 gene specifically in oligodendrocytes/Schwann cells activates mTORC1 signaling resulting in severe motor disabilities, weight loss, and early postnatal death. The mutant mice of either sex showed reduced myelination, disrupted paranodal domains in myelinated axons, and disorganized unmyelinated Remak bundles. mRNA and protein expression analyses revealed strong reduction in the RNA-binding protein Quaking (Qk) and the 155 kDa glial Neurofascin (NfascNF155). Re-introduction of exogenous Qk gene in Tsc1 mutant oligodendrocytes restored NfascNF155 protein levels indicating that Qk is required for the stabilization of NfascNF155 mRNA. Interestingly, injection of Rapamycin, a pharmacological mTORC1 inhibitor, to pregnant mothers increased the lifespan of the mutant offspring, restored myelination as well as the levels of Qk and NfascNF155, and consequently the organization of the paranodal domains. Together our studies show a critical role of mTORC1 signaling in the differentiation of myelinating glial cells and proper organization of axonal domains and provide insights into TSC-associated myelinated axon abnormalities.
Collapse
Affiliation(s)
| | | | | | - Manzoor A. Bhat
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
33
|
Bansagi B, Phan V, Baker MR, O'Sullivan J, Jennings MJ, Whittaker RG, Müller JS, Duff J, Griffin H, Miller JAL, Gorman GS, Lochmüller H, Chinnery PF, Roos A, Swan LE, Horvath R. Multifocal demyelinating motor neuropathy and hamartoma syndrome associated with a de novo PTEN mutation. Neurology 2018; 90:e1842-e1848. [PMID: 29720545 PMCID: PMC5962916 DOI: 10.1212/wnl.0000000000005566] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2017] [Accepted: 03/01/2018] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVE To describe a patient with a multifocal demyelinating motor neuropathy with onset in childhood and a mutation in phosphatase and tensin homolog (PTEN), a tumor suppressor gene associated with inherited tumor susceptibility conditions, macrocephaly, autism, ataxia, tremor, and epilepsy. Functional implications of this protein have been investigated in Parkinson and Alzheimer diseases. METHODS We performed whole-exome sequencing in the patient's genomic DNA validated by Sanger sequencing. Immunoblotting, in vitro enzymatic assay, and label-free shotgun proteomic profiling were performed in the patient's fibroblasts. RESULTS The predominant clinical presentation of the patient was a childhood onset, asymmetric progressive multifocal motor neuropathy. In addition, he presented with macrocephaly, autism spectrum disorder, and skin hamartomas, considered as clinical criteria for PTEN-related hamartoma tumor syndrome. Extensive tumor screening did not detect any malignancies. We detected a novel de novo heterozygous c.269T>C, p.(Phe90Ser) PTEN variant, which was absent in both parents. The pathogenicity of the variant is supported by altered expression of several PTEN-associated proteins involved in tumorigenesis. Moreover, fibroblasts showed a defect in catalytic activity of PTEN against the secondary substrate, phosphatidylinositol 3,4-trisphosphate. In support of our findings, focal hypermyelination leading to peripheral neuropathy has been reported in PTEN-deficient mice. CONCLUSION We describe a novel phenotype, PTEN-associated multifocal demyelinating motor neuropathy with a skin hamartoma syndrome. A similar mechanism may potentially underlie other forms of Charcot-Marie-Tooth disease with involvement of the phosphatidylinositol pathway.
Collapse
Affiliation(s)
- Boglarka Bansagi
- From the Wellcome Centre for Mitochondrial Research (G.S.G.), Institute of Genetic Medicine (B.B., M.J., J.S.M., J.D., H.G., H.L., P.F.C., A.R., R.H.), and Institute of Neuroscience (M.R.B., R.G.W., G.S.G.), Newcastle University, Newcastle upon Tyne, UK; Leibniz-Institute für Analytische Wissenschaften-ISAS-e.V. (V.P., A.R.), Dortmund, Germany; Departments of Neurology (M.R.B., J.A.L.M., G.S.G.) and Clinical Neurophysiology (M.R.B., R.G.W., R.H.), Royal Victoria Infirmary, Newcastle upon Tyne; Department of Cellular and Molecular Physiology (J.O., L.E.S.), Institute of Translational Medicine, University of Liverpool; Department of Clinical Neurosciences (P.F.C.), University of Cambridge, Cambridge Biomedical Campus, UK; Department of Neuropediatrics and Muscle Disorders (H.L.), Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; and Centro Nacional de Análisis Genómico (CNAG-CRG) (H.L.), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Vietxuan Phan
- From the Wellcome Centre for Mitochondrial Research (G.S.G.), Institute of Genetic Medicine (B.B., M.J., J.S.M., J.D., H.G., H.L., P.F.C., A.R., R.H.), and Institute of Neuroscience (M.R.B., R.G.W., G.S.G.), Newcastle University, Newcastle upon Tyne, UK; Leibniz-Institute für Analytische Wissenschaften-ISAS-e.V. (V.P., A.R.), Dortmund, Germany; Departments of Neurology (M.R.B., J.A.L.M., G.S.G.) and Clinical Neurophysiology (M.R.B., R.G.W., R.H.), Royal Victoria Infirmary, Newcastle upon Tyne; Department of Cellular and Molecular Physiology (J.O., L.E.S.), Institute of Translational Medicine, University of Liverpool; Department of Clinical Neurosciences (P.F.C.), University of Cambridge, Cambridge Biomedical Campus, UK; Department of Neuropediatrics and Muscle Disorders (H.L.), Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; and Centro Nacional de Análisis Genómico (CNAG-CRG) (H.L.), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Mark R Baker
- From the Wellcome Centre for Mitochondrial Research (G.S.G.), Institute of Genetic Medicine (B.B., M.J., J.S.M., J.D., H.G., H.L., P.F.C., A.R., R.H.), and Institute of Neuroscience (M.R.B., R.G.W., G.S.G.), Newcastle University, Newcastle upon Tyne, UK; Leibniz-Institute für Analytische Wissenschaften-ISAS-e.V. (V.P., A.R.), Dortmund, Germany; Departments of Neurology (M.R.B., J.A.L.M., G.S.G.) and Clinical Neurophysiology (M.R.B., R.G.W., R.H.), Royal Victoria Infirmary, Newcastle upon Tyne; Department of Cellular and Molecular Physiology (J.O., L.E.S.), Institute of Translational Medicine, University of Liverpool; Department of Clinical Neurosciences (P.F.C.), University of Cambridge, Cambridge Biomedical Campus, UK; Department of Neuropediatrics and Muscle Disorders (H.L.), Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; and Centro Nacional de Análisis Genómico (CNAG-CRG) (H.L.), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Julia O'Sullivan
- From the Wellcome Centre for Mitochondrial Research (G.S.G.), Institute of Genetic Medicine (B.B., M.J., J.S.M., J.D., H.G., H.L., P.F.C., A.R., R.H.), and Institute of Neuroscience (M.R.B., R.G.W., G.S.G.), Newcastle University, Newcastle upon Tyne, UK; Leibniz-Institute für Analytische Wissenschaften-ISAS-e.V. (V.P., A.R.), Dortmund, Germany; Departments of Neurology (M.R.B., J.A.L.M., G.S.G.) and Clinical Neurophysiology (M.R.B., R.G.W., R.H.), Royal Victoria Infirmary, Newcastle upon Tyne; Department of Cellular and Molecular Physiology (J.O., L.E.S.), Institute of Translational Medicine, University of Liverpool; Department of Clinical Neurosciences (P.F.C.), University of Cambridge, Cambridge Biomedical Campus, UK; Department of Neuropediatrics and Muscle Disorders (H.L.), Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; and Centro Nacional de Análisis Genómico (CNAG-CRG) (H.L.), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Matthew J Jennings
- From the Wellcome Centre for Mitochondrial Research (G.S.G.), Institute of Genetic Medicine (B.B., M.J., J.S.M., J.D., H.G., H.L., P.F.C., A.R., R.H.), and Institute of Neuroscience (M.R.B., R.G.W., G.S.G.), Newcastle University, Newcastle upon Tyne, UK; Leibniz-Institute für Analytische Wissenschaften-ISAS-e.V. (V.P., A.R.), Dortmund, Germany; Departments of Neurology (M.R.B., J.A.L.M., G.S.G.) and Clinical Neurophysiology (M.R.B., R.G.W., R.H.), Royal Victoria Infirmary, Newcastle upon Tyne; Department of Cellular and Molecular Physiology (J.O., L.E.S.), Institute of Translational Medicine, University of Liverpool; Department of Clinical Neurosciences (P.F.C.), University of Cambridge, Cambridge Biomedical Campus, UK; Department of Neuropediatrics and Muscle Disorders (H.L.), Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; and Centro Nacional de Análisis Genómico (CNAG-CRG) (H.L.), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Roger G Whittaker
- From the Wellcome Centre for Mitochondrial Research (G.S.G.), Institute of Genetic Medicine (B.B., M.J., J.S.M., J.D., H.G., H.L., P.F.C., A.R., R.H.), and Institute of Neuroscience (M.R.B., R.G.W., G.S.G.), Newcastle University, Newcastle upon Tyne, UK; Leibniz-Institute für Analytische Wissenschaften-ISAS-e.V. (V.P., A.R.), Dortmund, Germany; Departments of Neurology (M.R.B., J.A.L.M., G.S.G.) and Clinical Neurophysiology (M.R.B., R.G.W., R.H.), Royal Victoria Infirmary, Newcastle upon Tyne; Department of Cellular and Molecular Physiology (J.O., L.E.S.), Institute of Translational Medicine, University of Liverpool; Department of Clinical Neurosciences (P.F.C.), University of Cambridge, Cambridge Biomedical Campus, UK; Department of Neuropediatrics and Muscle Disorders (H.L.), Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; and Centro Nacional de Análisis Genómico (CNAG-CRG) (H.L.), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Juliane S Müller
- From the Wellcome Centre for Mitochondrial Research (G.S.G.), Institute of Genetic Medicine (B.B., M.J., J.S.M., J.D., H.G., H.L., P.F.C., A.R., R.H.), and Institute of Neuroscience (M.R.B., R.G.W., G.S.G.), Newcastle University, Newcastle upon Tyne, UK; Leibniz-Institute für Analytische Wissenschaften-ISAS-e.V. (V.P., A.R.), Dortmund, Germany; Departments of Neurology (M.R.B., J.A.L.M., G.S.G.) and Clinical Neurophysiology (M.R.B., R.G.W., R.H.), Royal Victoria Infirmary, Newcastle upon Tyne; Department of Cellular and Molecular Physiology (J.O., L.E.S.), Institute of Translational Medicine, University of Liverpool; Department of Clinical Neurosciences (P.F.C.), University of Cambridge, Cambridge Biomedical Campus, UK; Department of Neuropediatrics and Muscle Disorders (H.L.), Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; and Centro Nacional de Análisis Genómico (CNAG-CRG) (H.L.), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Jennifer Duff
- From the Wellcome Centre for Mitochondrial Research (G.S.G.), Institute of Genetic Medicine (B.B., M.J., J.S.M., J.D., H.G., H.L., P.F.C., A.R., R.H.), and Institute of Neuroscience (M.R.B., R.G.W., G.S.G.), Newcastle University, Newcastle upon Tyne, UK; Leibniz-Institute für Analytische Wissenschaften-ISAS-e.V. (V.P., A.R.), Dortmund, Germany; Departments of Neurology (M.R.B., J.A.L.M., G.S.G.) and Clinical Neurophysiology (M.R.B., R.G.W., R.H.), Royal Victoria Infirmary, Newcastle upon Tyne; Department of Cellular and Molecular Physiology (J.O., L.E.S.), Institute of Translational Medicine, University of Liverpool; Department of Clinical Neurosciences (P.F.C.), University of Cambridge, Cambridge Biomedical Campus, UK; Department of Neuropediatrics and Muscle Disorders (H.L.), Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; and Centro Nacional de Análisis Genómico (CNAG-CRG) (H.L.), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Helen Griffin
- From the Wellcome Centre for Mitochondrial Research (G.S.G.), Institute of Genetic Medicine (B.B., M.J., J.S.M., J.D., H.G., H.L., P.F.C., A.R., R.H.), and Institute of Neuroscience (M.R.B., R.G.W., G.S.G.), Newcastle University, Newcastle upon Tyne, UK; Leibniz-Institute für Analytische Wissenschaften-ISAS-e.V. (V.P., A.R.), Dortmund, Germany; Departments of Neurology (M.R.B., J.A.L.M., G.S.G.) and Clinical Neurophysiology (M.R.B., R.G.W., R.H.), Royal Victoria Infirmary, Newcastle upon Tyne; Department of Cellular and Molecular Physiology (J.O., L.E.S.), Institute of Translational Medicine, University of Liverpool; Department of Clinical Neurosciences (P.F.C.), University of Cambridge, Cambridge Biomedical Campus, UK; Department of Neuropediatrics and Muscle Disorders (H.L.), Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; and Centro Nacional de Análisis Genómico (CNAG-CRG) (H.L.), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - James A L Miller
- From the Wellcome Centre for Mitochondrial Research (G.S.G.), Institute of Genetic Medicine (B.B., M.J., J.S.M., J.D., H.G., H.L., P.F.C., A.R., R.H.), and Institute of Neuroscience (M.R.B., R.G.W., G.S.G.), Newcastle University, Newcastle upon Tyne, UK; Leibniz-Institute für Analytische Wissenschaften-ISAS-e.V. (V.P., A.R.), Dortmund, Germany; Departments of Neurology (M.R.B., J.A.L.M., G.S.G.) and Clinical Neurophysiology (M.R.B., R.G.W., R.H.), Royal Victoria Infirmary, Newcastle upon Tyne; Department of Cellular and Molecular Physiology (J.O., L.E.S.), Institute of Translational Medicine, University of Liverpool; Department of Clinical Neurosciences (P.F.C.), University of Cambridge, Cambridge Biomedical Campus, UK; Department of Neuropediatrics and Muscle Disorders (H.L.), Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; and Centro Nacional de Análisis Genómico (CNAG-CRG) (H.L.), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Grainne S Gorman
- From the Wellcome Centre for Mitochondrial Research (G.S.G.), Institute of Genetic Medicine (B.B., M.J., J.S.M., J.D., H.G., H.L., P.F.C., A.R., R.H.), and Institute of Neuroscience (M.R.B., R.G.W., G.S.G.), Newcastle University, Newcastle upon Tyne, UK; Leibniz-Institute für Analytische Wissenschaften-ISAS-e.V. (V.P., A.R.), Dortmund, Germany; Departments of Neurology (M.R.B., J.A.L.M., G.S.G.) and Clinical Neurophysiology (M.R.B., R.G.W., R.H.), Royal Victoria Infirmary, Newcastle upon Tyne; Department of Cellular and Molecular Physiology (J.O., L.E.S.), Institute of Translational Medicine, University of Liverpool; Department of Clinical Neurosciences (P.F.C.), University of Cambridge, Cambridge Biomedical Campus, UK; Department of Neuropediatrics and Muscle Disorders (H.L.), Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; and Centro Nacional de Análisis Genómico (CNAG-CRG) (H.L.), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Hanns Lochmüller
- From the Wellcome Centre for Mitochondrial Research (G.S.G.), Institute of Genetic Medicine (B.B., M.J., J.S.M., J.D., H.G., H.L., P.F.C., A.R., R.H.), and Institute of Neuroscience (M.R.B., R.G.W., G.S.G.), Newcastle University, Newcastle upon Tyne, UK; Leibniz-Institute für Analytische Wissenschaften-ISAS-e.V. (V.P., A.R.), Dortmund, Germany; Departments of Neurology (M.R.B., J.A.L.M., G.S.G.) and Clinical Neurophysiology (M.R.B., R.G.W., R.H.), Royal Victoria Infirmary, Newcastle upon Tyne; Department of Cellular and Molecular Physiology (J.O., L.E.S.), Institute of Translational Medicine, University of Liverpool; Department of Clinical Neurosciences (P.F.C.), University of Cambridge, Cambridge Biomedical Campus, UK; Department of Neuropediatrics and Muscle Disorders (H.L.), Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; and Centro Nacional de Análisis Genómico (CNAG-CRG) (H.L.), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Patrick F Chinnery
- From the Wellcome Centre for Mitochondrial Research (G.S.G.), Institute of Genetic Medicine (B.B., M.J., J.S.M., J.D., H.G., H.L., P.F.C., A.R., R.H.), and Institute of Neuroscience (M.R.B., R.G.W., G.S.G.), Newcastle University, Newcastle upon Tyne, UK; Leibniz-Institute für Analytische Wissenschaften-ISAS-e.V. (V.P., A.R.), Dortmund, Germany; Departments of Neurology (M.R.B., J.A.L.M., G.S.G.) and Clinical Neurophysiology (M.R.B., R.G.W., R.H.), Royal Victoria Infirmary, Newcastle upon Tyne; Department of Cellular and Molecular Physiology (J.O., L.E.S.), Institute of Translational Medicine, University of Liverpool; Department of Clinical Neurosciences (P.F.C.), University of Cambridge, Cambridge Biomedical Campus, UK; Department of Neuropediatrics and Muscle Disorders (H.L.), Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; and Centro Nacional de Análisis Genómico (CNAG-CRG) (H.L.), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Andreas Roos
- From the Wellcome Centre for Mitochondrial Research (G.S.G.), Institute of Genetic Medicine (B.B., M.J., J.S.M., J.D., H.G., H.L., P.F.C., A.R., R.H.), and Institute of Neuroscience (M.R.B., R.G.W., G.S.G.), Newcastle University, Newcastle upon Tyne, UK; Leibniz-Institute für Analytische Wissenschaften-ISAS-e.V. (V.P., A.R.), Dortmund, Germany; Departments of Neurology (M.R.B., J.A.L.M., G.S.G.) and Clinical Neurophysiology (M.R.B., R.G.W., R.H.), Royal Victoria Infirmary, Newcastle upon Tyne; Department of Cellular and Molecular Physiology (J.O., L.E.S.), Institute of Translational Medicine, University of Liverpool; Department of Clinical Neurosciences (P.F.C.), University of Cambridge, Cambridge Biomedical Campus, UK; Department of Neuropediatrics and Muscle Disorders (H.L.), Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; and Centro Nacional de Análisis Genómico (CNAG-CRG) (H.L.), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Laura E Swan
- From the Wellcome Centre for Mitochondrial Research (G.S.G.), Institute of Genetic Medicine (B.B., M.J., J.S.M., J.D., H.G., H.L., P.F.C., A.R., R.H.), and Institute of Neuroscience (M.R.B., R.G.W., G.S.G.), Newcastle University, Newcastle upon Tyne, UK; Leibniz-Institute für Analytische Wissenschaften-ISAS-e.V. (V.P., A.R.), Dortmund, Germany; Departments of Neurology (M.R.B., J.A.L.M., G.S.G.) and Clinical Neurophysiology (M.R.B., R.G.W., R.H.), Royal Victoria Infirmary, Newcastle upon Tyne; Department of Cellular and Molecular Physiology (J.O., L.E.S.), Institute of Translational Medicine, University of Liverpool; Department of Clinical Neurosciences (P.F.C.), University of Cambridge, Cambridge Biomedical Campus, UK; Department of Neuropediatrics and Muscle Disorders (H.L.), Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; and Centro Nacional de Análisis Genómico (CNAG-CRG) (H.L.), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Rita Horvath
- From the Wellcome Centre for Mitochondrial Research (G.S.G.), Institute of Genetic Medicine (B.B., M.J., J.S.M., J.D., H.G., H.L., P.F.C., A.R., R.H.), and Institute of Neuroscience (M.R.B., R.G.W., G.S.G.), Newcastle University, Newcastle upon Tyne, UK; Leibniz-Institute für Analytische Wissenschaften-ISAS-e.V. (V.P., A.R.), Dortmund, Germany; Departments of Neurology (M.R.B., J.A.L.M., G.S.G.) and Clinical Neurophysiology (M.R.B., R.G.W., R.H.), Royal Victoria Infirmary, Newcastle upon Tyne; Department of Cellular and Molecular Physiology (J.O., L.E.S.), Institute of Translational Medicine, University of Liverpool; Department of Clinical Neurosciences (P.F.C.), University of Cambridge, Cambridge Biomedical Campus, UK; Department of Neuropediatrics and Muscle Disorders (H.L.), Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany; and Centro Nacional de Análisis Genómico (CNAG-CRG) (H.L.), Center for Genomic Regulation, Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
| |
Collapse
|
34
|
Kim M, Wende H, Walcher J, Kühnemund J, Cheret C, Kempa S, McShane E, Selbach M, Lewin GR, Birchmeier C. Maf links Neuregulin1 signaling to cholesterol synthesis in myelinating Schwann cells. Genes Dev 2018; 32:645-657. [PMID: 29748249 PMCID: PMC6004071 DOI: 10.1101/gad.310490.117] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 04/16/2018] [Indexed: 11/25/2022]
Abstract
Kim et al. define a crucial role of the transcription factor Maf in myelination and cholesterol biosynthesis and show that Maf acts downstream from Neuregulin1 in myelinating Schwann cells. Cholesterol is a major constituent of myelin membranes, which insulate axons and allow saltatory conduction. Therefore, Schwann cells, the myelinating glia of the peripheral nervous system, need to produce large amounts of cholesterol. Here, we define a crucial role of the transcription factor Maf in myelination and cholesterol biosynthesis and show that Maf acts downstream from Neuregulin1 (Nrg1). Maf expression is induced when Schwann cells begin myelination. Genetic ablation of Maf resulted in hypomyelination that resembled mice with defective Nrg1 signaling. Importantly, loss of Maf or Nrg1 signaling resulted in a down-regulation of the cholesterol synthesis program, and Maf directly binds to enhancers of cholesterol synthesis genes. Furthermore, we identified the molecular mechanisms by which Nrg1 signaling regulates Maf levels. Transcription of Maf depends on calmodulin-dependent kinases downstream from Nrg1, whereas Nrg1–MAPK signaling stabilizes Maf protein. Our results delineate a novel signaling cascade regulating cholesterol synthesis in myelinating Schwann cells.
Collapse
Affiliation(s)
- Minchul Kim
- Department of Developmental Biology/Signal Transduction, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Hagen Wende
- Department of Developmental Biology/Signal Transduction, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Jan Walcher
- Department of Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Johannes Kühnemund
- Department of Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Cyril Cheret
- Department of Developmental Biology/Signal Transduction, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Stefan Kempa
- Department of Integrative Proteomics and Metabolomics, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Erik McShane
- Department of Proteome Dynamics, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Matthias Selbach
- Department of Proteome Dynamics, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Gary R Lewin
- Department of Molecular Physiology of Somatic Sensation, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| | - Carmen Birchmeier
- Department of Developmental Biology/Signal Transduction, Max Delbrück Center for Molecular Medicine, 13125 Berlin, Germany
| |
Collapse
|
35
|
Bachiller S, Roca-Ceballos MA, García-Domínguez I, Pérez-Villegas EM, Martos-Carmona D, Pérez-Castro MÁ, Real LM, Rosa JL, Tabares L, Venero JL, Armengol JÁ, Carrión ÁM, Ruiz R. HERC1 Ubiquitin Ligase Is Required for Normal Axonal Myelination in the Peripheral Nervous System. Mol Neurobiol 2018; 55:8856-8868. [PMID: 29603094 DOI: 10.1007/s12035-018-1021-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 03/16/2018] [Indexed: 12/14/2022]
Abstract
A missense mutation in HERC1 provokes loss of cerebellar Purkinje cells, tremor, and unstable gait in tambaleante (tbl) mice. Recently, we have shown that before cerebellar degeneration takes place, the tbl mouse suffers from a reduction in the number of vesicles available for release at the neuromuscular junction (NMJ). The aim of the present work was to study to which extent the alteration in HERC1 may affect other cells in the nervous system and how this may influence the motor dysfunction observed in these mice. The functional analysis showed a consistent delay in the propagation of the action potential in mutant mice in comparison with control littermates. Morphological analyses of glial cells in motor axons revealed signs of compact myelin damage as tomacula and local hypermyelination foci. Moreover, we observed an alteration in non-myelinated terminal Schwann cells at the level of the NMJ. Additionally, we found a significant increment of phosphorylated Akt-2 in the sciatic nerve. Based on these findings, we propose a molecular model that could explain how mutated HERC1 in tbl mice affects the myelination process in the peripheral nervous system. Finally, since the myelin abnormalities found in tbl mice are histological hallmarks of neuropathic periphery diseases, tbl mutant mice could be considered as a new mouse model for this type of diseases.
Collapse
Affiliation(s)
- Sara Bachiller
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Seville, Spain
| | - María Angustias Roca-Ceballos
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Calle Profesor García González 2, 41012, Sevilla, Spain
| | - Irene García-Domínguez
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Calle Profesor García González 2, 41012, Sevilla, Spain
| | - Eva María Pérez-Villegas
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Seville, Spain
| | - David Martos-Carmona
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Seville, Spain
| | - Miguel Ángel Pérez-Castro
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Calle Profesor García González 2, 41012, Sevilla, Spain
| | - Luis Miguel Real
- Unit of Infectious Diseases and Microbiology, Valme University Hospital, Seville, Spain
| | - José Luis Rosa
- Departament de Ciències Fisiològiques II, IDIBELL, Campus de Bellvitge, Universitat de Barcelona, L'Hospitalet de Llobregat, E-08907, Barcelona, Spain
| | - Lucía Tabares
- Department of Medical Physiology and Biophysics, School of Medicine, University of Seville, Seville, Spain
| | - José Luis Venero
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Calle Profesor García González 2, 41012, Sevilla, Spain
| | - José Ángel Armengol
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Seville, Spain
| | - Ángel Manuel Carrión
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Seville, Spain
| | - Rocío Ruiz
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Seville, Spain. .,Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Calle Profesor García González 2, 41012, Sevilla, Spain.
| |
Collapse
|
36
|
Poitelon Y, Matafora V, Silvestri N, Zambroni D, McGarry C, Serghany N, Rush T, Vizzuso D, Court FA, Bachi A, Wrabetz L, Feltri ML. A dual role for Integrin α6β4 in modulating hereditary neuropathy with liability to pressure palsies. J Neurochem 2018; 145:245-257. [PMID: 29315582 DOI: 10.1111/jnc.14295] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 12/14/2017] [Accepted: 12/21/2017] [Indexed: 12/23/2022]
Abstract
Peripheral myelin protein 22 (PMP22) is a component of compact myelin in the peripheral nervous system. The amount of PMP22 in myelin is tightly regulated, and PMP22 over or under-expression cause Charcot-Marie-Tooth 1A (CMT1A) and Hereditary Neuropathy with Pressure Palsies (HNPP). Despite the importance of PMP22, its function remains largely unknown. It was reported that PMP22 interacts with the β4 subunit of the laminin receptor α6β4 integrin, suggesting that α6β4 integrin and laminins may contribute to the pathogenesis of CMT1A or HNPP. Here we asked if the lack of α6β4 integrin in Schwann cells influences myelin stability in the HNPP mouse model. Our data indicate that PMP22 and β4 integrin may not interact directly in myelinating Schwann cells, however, ablating β4 integrin delays the formation of tomacula, a characteristic feature of HNPP. In contrast, ablation of integrin β4 worsens nerve conduction velocities and non-compact myelin organization in HNPP animals. This study demonstrates that indirect interactions between an extracellular matrix receptor and a myelin protein influence the stability and function of myelinated fibers.
Collapse
Affiliation(s)
- Yannick Poitelon
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York, USA.,Department of Biochemistry, University at Buffalo, Buffalo, New York, USA
| | - Vittoria Matafora
- Division of Genetics and Cell Biology, San Raffaele Hospital, Milan, Italy
| | | | - Desirée Zambroni
- Division of Genetics and Cell Biology, San Raffaele Hospital, Milan, Italy
| | - Claire McGarry
- Department of Biochemistry, University at Buffalo, Buffalo, New York, USA
| | - Nora Serghany
- Department of Biochemistry, University at Buffalo, Buffalo, New York, USA
| | - Thomas Rush
- Department of Biochemistry, University at Buffalo, Buffalo, New York, USA
| | - Domenica Vizzuso
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York, USA.,Division of Genetics and Cell Biology, San Raffaele Hospital, Milan, Italy
| | - Felipe A Court
- Division of Genetics and Cell Biology, San Raffaele Hospital, Milan, Italy.,Center for Integrative Biology, Universidad Mayor de Chile, Santiago, Chile
| | - Angela Bachi
- Division of Genetics and Cell Biology, San Raffaele Hospital, Milan, Italy
| | - Lawrence Wrabetz
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York, USA.,Department of Biochemistry, University at Buffalo, Buffalo, New York, USA.,Division of Genetics and Cell Biology, San Raffaele Hospital, Milan, Italy.,Department of Neurology, University at Buffalo, Buffalo, New York, USA
| | - Maria Laura Feltri
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York, USA.,Department of Biochemistry, University at Buffalo, Buffalo, New York, USA.,Division of Genetics and Cell Biology, San Raffaele Hospital, Milan, Italy.,Department of Neurology, University at Buffalo, Buffalo, New York, USA
| |
Collapse
|
37
|
Wong KM, Beirowski B. Multiple lines of inhibitory feedback on AKT kinase in Schwann cells lacking TSC1/2 hint at distinct functions of mTORC1 and AKT in nerve development. Commun Integr Biol 2018; 11:e1433441. [PMID: 29497474 PMCID: PMC5824964 DOI: 10.1080/19420889.2018.1433441] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 01/02/2018] [Accepted: 01/19/2018] [Indexed: 11/29/2022] Open
Abstract
During nerve development, Schwann cells (SCs) build multilayered myelin sheaths around axons to accelerate nerve conduction. The mechanistic target of rapamycin complex 1 (mTORC1) downstream of PI3K/AKT signaling lately emerged as a central anabolic regulator of myelination. Using mutant mice with sustained mTORC1 hyperactivity in developing SCs we recently uncovered that mTORC1 impedes developmental myelination by promoting proliferation of immature SCs while antagonizing SC differentiation. In contrast, mTORC1 stimulates myelin production, rather than SC proliferation, in already differentiated SCs. Importantly, these diametrical mTORC1 functions were unmasked under settings of greatly suppressed AKT signaling. Here we demonstrate, inter alia, additional mechanisms of feedback inhibition of AKT by mTORC1, such as strikingly elevated PTEN levels in SCs with disruption of the mTORC1 inhibitory complex, TSC1/2. These data lead us to propose a model wherein mTORC1 and AKT have distinct roles in developing SCs that have to be precisely coordinated for normal myelinogenesis.
Collapse
Affiliation(s)
- Keit Men Wong
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Bogdan Beirowski
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
38
|
Cervellini I, Galino J, Zhu N, Allen S, Birchmeier C, Bennett DL. Sustained MAPK/ERK Activation in Adult Schwann Cells Impairs Nerve Repair. J Neurosci 2018; 38:679-690. [PMID: 29217688 PMCID: PMC5777114 DOI: 10.1523/jneurosci.2255-17.2017] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Revised: 09/29/2017] [Accepted: 10/19/2017] [Indexed: 11/21/2022] Open
Abstract
The MAPK/ERK pathway has a critical role in PNS development. It is required for Schwann cell (SC) differentiation and myelination; sustained embryonic MAPK/ERK activation in SCs enhances myelin growth overcoming signals that normally end myelination. Excess activation of this pathway can be maladaptive as in adulthood acute strong activation of MAPK/ERK has been shown to cause SC dedifferentiation and demyelination. We used a mouse model (including male and female animals) in which the gain-of-function MEK1DD allele produces sustained MAPK/ERK activation in adult SCs, and we determined the impact of such activation on nerve repair. In the uninjured nerve, MAPK/ERK activation neither impaired myelin nor reactivated myelination. However, in the injured nerve it was detrimental and resulted in delayed repair and functional recovery. In the early phase of injury, the rate of myelin clearance was faster. Four weeks following injury, when nerve repair is normally advanced, myelinated axons of MEK1DD mutants demonstrated higher rates of myelin decompaction, a reduced number of Cajal bands. and decreased internodal length. We noted the presence of abnormal Remak bundles with long SCs processes and reduced numbers of C-fibers/Remak bundle. Both the total number of regenerating axons and the intraepidermal nerve fiber density in the skin were reduced. Sustained activation of MAPK/ERK in adult SCs is therefore deleterious to successful nerve repair, emphasizing the differences in the signaling processes coordinating nerve development and repair. Our results also underline the key role of SCs in axon regeneration and successful target reinnervation.SIGNIFICANCE STATEMENT The MAPK/ERK pathway promotes developmental myelination and its sustained activation in SCs induced continuous myelin growth, compensating for the absence of essential myelination signals. However, the strength of activation is fundamental because acute strong induction of MAPK/ERK in adulthood induces demyelination. What has been unknown is the effect of a mild but sustained MAPK/ERK activation in SCs on nerve repair in adulthood. This promoted myelin clearance but led to abnormalities in nonmyelinating and myelinating SCs in the later phases of nerve repair, resulting in slowed axon regeneration, cutaneous reinnervation, and functional recovery. Our results emphasize the distinct role of the MAPK/ERK pathway in developmental myelination versus remyelination and the importance of signaling between SCs and axons for successful axon regeneration.
Collapse
Affiliation(s)
- Ilaria Cervellini
- The Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom, and
| | - Jorge Galino
- The Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom, and
| | - Ning Zhu
- The Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom, and
| | - Shannen Allen
- The Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom, and
| | - Carmen Birchmeier
- Developmental Biology/Signal Transduction Group, Max Delbrück Center for Molecular Medicine (MDC) in the Helmholtz Society, 13125 Berlin, Germany
| | - David L Bennett
- The Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom, and
| |
Collapse
|
39
|
Tricaud N. Myelinating Schwann Cell Polarity and Mechanically-Driven Myelin Sheath Elongation. Front Cell Neurosci 2018; 11:414. [PMID: 29354031 PMCID: PMC5760505 DOI: 10.3389/fncel.2017.00414] [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: 10/03/2017] [Accepted: 12/11/2017] [Indexed: 11/13/2022] Open
Abstract
Myelin sheath geometry, encompassing myelin sheath thickness relative to internodal length, is critical to optimize nerve conduction velocity and these parameters are carefully adjusted by the myelinating cells in mammals. In the central nervous system these adjustments could regulate neuronal activities while in the peripheral nervous system they lead to the optimization and the reliability of the nerve conduction velocity. However, the physiological and cellular mechanisms that underlie myelin sheath geometry regulation are not yet fully elucidated. In peripheral nerves the myelinating Schwann cell uses several molecular mechanisms to reach and maintain the correct myelin sheath geometry, such that myelin sheath thickness and internodal length are regulated independently. One of these mechanisms is the epithelial-like cell polarization process that occurs during the early phases of the myelin biogenesis. Epithelial cell polarization factors are known to control cell size and morphology in invertebrates and mammals making these processes critical in the organogenesis. Correlative data indicate that internodal length is regulated by postnatal body growth that elongates peripheral nerves in mammals. In addition, the mechanical stretching of peripheral nerves in adult animals shows that myelin sheath length can be increased by mechanical cues. Recent results describe the important role of YAP/TAZ co-transcription factors during Schwann cell myelination and their functions have linked to the mechanotransduction through the HIPPO pathway and the epithelial polarity factor Crb3. In this review the molecular mechanisms that govern mechanically-driven myelin sheath elongation and how a Schwann cell can modulate internodal myelin sheath length, independent of internodal thickness, will be discussed regarding these recent data. In addition, the potential relevance of these mechanosensitive mechanisms in peripheral pathologies will be highlighted.
Collapse
Affiliation(s)
- Nicolas Tricaud
- Institut National de la Santé et de la Recherche Médicale, Institut des Neurosciences de Montpellier, Université de Montpellier, Montpellier, France
| |
Collapse
|
40
|
Figlia G, Gerber D, Suter U. Myelination and mTOR. Glia 2017; 66:693-707. [PMID: 29210103 PMCID: PMC5836902 DOI: 10.1002/glia.23273] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/08/2017] [Accepted: 11/17/2017] [Indexed: 02/06/2023]
Abstract
Myelinating cells surround axons to accelerate the propagation of action potentials, to support axonal health, and to refine neural circuits. Myelination is metabolically demanding and, consistent with this notion, mTORC1—a signaling hub coordinating cell metabolism—has been implicated as a key signal for myelination. Here, we will discuss metabolic aspects of myelination, illustrate the main metabolic processes regulated by mTORC1, and review advances on the role of mTORC1 in myelination of the central nervous system and the peripheral nervous system. Recent progress has revealed a complex role of mTORC1 in myelinating cells that includes, besides positive regulation of myelin growth, additional critical functions in the stages preceding active myelination. Based on the available evidence, we will also highlight potential nonoverlapping roles between mTORC1 and its known main upstream pathways PI3K‐Akt, Mek‐Erk1/2, and AMPK in myelinating cells. Finally, we will discuss signals that are already known or hypothesized to be responsible for the regulation of mTORC1 activity in myelinating cells. Myelination is metabolically demanding. The metabolic regulator mTORC1 controls differentiation of myelinating cells and promotes myelin
growth. mTORC1‐independent targets of the PI3K‐Akt and Mek‐Erk1/2 pathways may also be significant in myelination.
Collapse
Affiliation(s)
- Gianluca Figlia
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology, ETH Zürich, Zürich, CH 8093, Switzerland
| | - Daniel Gerber
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology, ETH Zürich, Zürich, CH 8093, Switzerland
| | - Ueli Suter
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology, ETH Zürich, Zürich, CH 8093, Switzerland
| |
Collapse
|
41
|
Figlia G, Norrmén C, Pereira JA, Gerber D, Suter U. Dual function of the PI3K-Akt-mTORC1 axis in myelination of the peripheral nervous system. eLife 2017; 6:e29241. [PMID: 28880149 PMCID: PMC5589416 DOI: 10.7554/elife.29241] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 07/31/2017] [Indexed: 01/24/2023] Open
Abstract
Myelination is a biosynthetically demanding process in which mTORC1, the gatekeeper of anabolism, occupies a privileged regulatory position. We have shown previously that loss of mTORC1 function in Schwann cells (SCs) hampers myelination. Here, we genetically disrupted key inhibitory components upstream of mTORC1, TSC1 or PTEN, in mouse SC development, adult homeostasis, and nerve injury. Surprisingly, the resulting mTORC1 hyperactivity led to markedly delayed onset of both developmental myelination and remyelination after injury. However, if mTORC1 was hyperactivated after myelination onset, radial hypermyelination was observed. At early developmental stages, physiologically high PI3K-Akt-mTORC1 signaling suppresses expression of Krox20 (Egr2), the master regulator of PNS myelination. This effect is mediated by S6K and contributes to control mechanisms that keep SCs in a not-fully differentiated state to ensure proper timing of myelination initiation. An ensuing decline in mTORC1 activity is crucial to allow myelination to start, while remaining mTORC1 activity drives myelin growth.
Collapse
Affiliation(s)
- Gianluca Figlia
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| | - Camilla Norrmén
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| | - Jorge A Pereira
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| | - Daniel Gerber
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| | - Ueli Suter
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| |
Collapse
|
42
|
Schwann Cell O-GlcNAc Glycosylation Is Required for Myelin Maintenance and Axon Integrity. J Neurosci 2017; 36:9633-46. [PMID: 27629714 PMCID: PMC5039245 DOI: 10.1523/jneurosci.1235-16.2016] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 07/25/2016] [Indexed: 12/29/2022] Open
Abstract
UNLABELLED Schwann cells (SCs), ensheathing glia of the peripheral nervous system, support axonal survival and function. Abnormalities in SC metabolism affect their ability to provide this support and maintain axon integrity. To further interrogate this metabolic influence on axon-glial interactions, we generated OGT-SCKO mice with SC-specific deletion of the metabolic/nutrient sensing protein O-GlcNAc transferase that mediates the O-linked addition of N-acetylglucosamine (GlcNAc) moieties to Ser and Thr residues. The OGT-SCKO mice develop tomaculous demyelinating neuropathy characterized by focal thickenings of the myelin sheath (tomacula), progressive demyelination, axonal loss, and motor and sensory nerve dysfunction. Proteomic analysis identified more than 100 O-GlcNAcylated proteins in rat sciatic nerve, including Periaxin (PRX), a myelin protein whose mutation causes inherited neuropathy in humans. PRX lacking O-GlcNAcylation is mislocalized within the myelin sheath of these mutant animals. Furthermore, phenotypes of OGT-SCKO and Prx-deficient mice are very similar, suggesting that metabolic control of PRX O-GlcNAcylation is crucial for myelin maintenance and axonal integrity. SIGNIFICANCE STATEMENT The nutrient sensing protein O-GlcNAc transferase (OGT) mediates post-translational O-linked N-acetylglucosamine (GlcNAc) modification. Here we find that OGT functions in Schwann cells (SCs) to maintain normal myelin and prevent axonal loss. SC-specific deletion of OGT (OGT-SCKO mice) causes a tomaculous demyelinating neuropathy accompanied with progressive axon degeneration and motor and sensory nerve dysfunction. We also found Periaxin (PRX), a myelin protein whose mutation causes inherited neuropathy in humans, is O-GlcNAcylated. Importantly, phenotypes of OGT-SCKO and Prx mutant mice are very similar, implying that compromised PRX function contributes to the neuropathy of OGT-SCKO mice. This study will be useful in understanding how SC metabolism contributes to PNS function and in developing new strategies for treating peripheral neuropathy by targeting SC function.
Collapse
|
43
|
Deficiency of a membrane skeletal protein, 4.1G, results in myelin abnormalities in the peripheral nervous system. Histochem Cell Biol 2017; 148:597-606. [PMID: 28755316 DOI: 10.1007/s00418-017-1600-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2017] [Indexed: 12/24/2022]
Abstract
We previously demonstrated that a membrane skeletal molecular complex, 4.1G-membrane palmitoylated protein 6 (MPP6)-cell adhesion molecule 4, is incorporated in Schwann cells in the peripheral nervous system (PNS). In this study, we evaluated motor activity and myelin ultrastructures in 4.1G-deficient (-/-) mice. When suspended by the tail, aged 4.1G-/- mice displayed spastic leg extension, especially after overwork. Motor-conduction velocity in 4.1G-/- mice was slower than that in wild-type mice. Using electron microscopy, 4.1G-/- mice exhibited myelin abnormalities: myelin was thicker in internodes, and attachment of myelin tips was distorted in some paranodes. In addition, we found a novel function of 4.1G for sorting a scaffold protein, Lin7, due to disappearance of the immunolocalization and reduction of the production of Lin7c and Lin7a in 4.1G-/- sciatic nerves, as well as the interaction of MPP6 and Lin7 with immunoprecipitation. Thus, we herein propose 4.1G functions as a signal for proper formation of myelin in PNS.
Collapse
|
44
|
Strength of ERK1/2 MAPK Activation Determines Its Effect on Myelin and Axonal Integrity in the Adult CNS. J Neurosci 2017; 36:6471-87. [PMID: 27307235 DOI: 10.1523/jneurosci.0299-16.2016] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 05/10/2016] [Indexed: 11/21/2022] Open
Abstract
UNLABELLED Myelin growth is a tightly regulated process driven by multiple signals. ERK1/2-MAPK signaling is an important regulator of myelin thickness. Because, in demyelinating diseases, the myelin formed during remyelination fails to achieve normal thickness, increasing ERK1/2 activity in oligodendrocytes is of obvious therapeutic potential for promoting efficient remyelination. However, other studies have suggested that increased levels of ERK1/2 activity could, in fact, have detrimental effects on myelinating cells. Because the strength, duration, or timing of ERK1/2 activation may alter the biological outcomes of cellular responses markedly, here, we investigated the effect of modulating ERK1/2 activity in myelinating cells using transgenic mouse lines in which ERK1/2 activation was upregulated conditionally in a graded manner. We found enhanced myelin gene expression and myelin growth in the adult CNS at both moderate and hyperactivated levels of ERK1/2 when upregulation commenced during developmental myelination or was induced later during adulthood in quiescent preexisting oligodendrocytes, after active myelination is largely terminated. However, a late onset of demyelination and axonal degeneration occurred at hyperelevated, but not moderately elevated, levels regardless of the timing of the upregulation. Similarly, myelin and axonal pathology occurred with elevated ERK1/2 activity in Schwann cells. We conclude that a fine tuning of ERK1/2 signaling strength is critically important for normal oligodendrocyte and Schwann cell function and that disturbance of this balance has negative consequences for myelin and axonal integrity in the long term. Therefore, therapeutic modulation of ERK1/2 activity in demyelinating disease or peripheral neuropathies must be approached with caution. SIGNIFICANCE STATEMENT ERK1/2-MAPK activation in oligodendrocytes and Schwann cells is an important signal for promoting myelin growth during developmental myelination. Here, we show that, when ERK1/2 are activated in mature quiescent oligodendrocytes during adulthood, new myelin growth is reinitiated even after active myelination is terminated, which has implications for understanding the mechanism underlying plasticity of myelin in adult life. Paradoxically, simply increasing the "strength" of ERK1/2 activation changed the biological outcome from beneficial to detrimental, adversely affecting myelin and axonal integrity in both the CNS and PNS. Therefore, this study highlights the complexity of ERK1/2-MAPK signaling in the context of oligodendrocyte and Schwann cell function in the adult animal and emphasizes the need to approach potential therapeutic modulation of ERK1/2 activity with caution.
Collapse
|
45
|
mTORC1 promotes proliferation of immature Schwann cells and myelin growth of differentiated Schwann cells. Proc Natl Acad Sci U S A 2017; 114:E4261-E4270. [PMID: 28484008 PMCID: PMC5448230 DOI: 10.1073/pnas.1620761114] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The myelination of axons in peripheral nerves requires precisely coordinated proliferation and differentiation of Schwann cells (SCs). We found that the activity of the mechanistic target of rapamycin complex 1 (mTORC1), a key signaling hub for the regulation of cellular growth and proliferation, is progressively extinguished as SCs differentiate during nerve development. To study the effects of different levels of sustained mTORC1 hyperactivity in the SC lineage, we disrupted negative regulators of mTORC1, including TSC2 or TSC1, in developing SCs of mutant mice. Surprisingly, the phenotypes ranged from arrested myelination in nerve development to focal hypermyelination in adulthood, depending on the level and timing of mTORC1 hyperactivity. For example, mice lacking TSC2 in developing SCs displayed hyperproliferation of undifferentiated SCs incompatible with normal myelination. However, these defects and myelination could be rescued by pharmacological mTORC1 inhibition. The subsequent reconstitution of SC mTORC1 hyperactivity in adult animals resulted in focal hypermyelination. Together our data suggest a model in which high mTORC1 activity promotes proliferation of immature SCs and antagonizes SC differentiation during nerve development. Down-regulation of mTORC1 activity is required for terminal SC differentiation and subsequent initiation of myelination. In distinction to this developmental role, excessive SC mTORC1 activity stimulates myelin growth, even overgrowth, in adulthood. Thus, our work delineates two distinct functions of mTORC1 in the SC lineage essential for proper nerve development and myelination. Moreover, our studies show that SCs retain their plasticity to myelinate and remodel myelin via mTORC1 throughout life.
Collapse
|
46
|
Bansagi B, Griffin H, Whittaker RG, Antoniadi T, Evangelista T, Miller J, Greenslade M, Forester N, Duff J, Bradshaw A, Kleinle S, Boczonadi V, Steele H, Ramesh V, Franko E, Pyle A, Lochmüller H, Chinnery PF, Horvath R. Genetic heterogeneity of motor neuropathies. Neurology 2017; 88:1226-1234. [PMID: 28251916 PMCID: PMC5373778 DOI: 10.1212/wnl.0000000000003772] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 01/06/2017] [Indexed: 11/22/2022] Open
Abstract
OBJECTIVE To study the prevalence, molecular cause, and clinical presentation of hereditary motor neuropathies in a large cohort of patients from the North of England. METHODS Detailed neurologic and electrophysiologic assessments and next-generation panel testing or whole exome sequencing were performed in 105 patients with clinical symptoms of distal hereditary motor neuropathy (dHMN, 64 patients), axonal motor neuropathy (motor Charcot-Marie-Tooth disease [CMT2], 16 patients), or complex neurologic disease predominantly affecting the motor nerves (hereditary motor neuropathy plus, 25 patients). RESULTS The prevalence of dHMN is 2.14 affected individuals per 100,000 inhabitants (95% confidence interval 1.62-2.66) in the North of England. Causative mutations were identified in 26 out of 73 index patients (35.6%). The diagnostic rate in the dHMN subgroup was 32.5%, which is higher than previously reported (20%). We detected a significant defect of neuromuscular transmission in 7 cases and identified potentially causative mutations in 4 patients with multifocal demyelinating motor neuropathy. CONCLUSIONS Many of the genes were shared between dHMN and motor CMT2, indicating identical disease mechanisms; therefore, we suggest changing the classification and including dHMN also as a subcategory of Charcot-Marie-Tooth disease. Abnormal neuromuscular transmission in some genetic forms provides a treatable target to develop therapies.
Collapse
Affiliation(s)
- Boglarka Bansagi
- From the MRC Centre for Neuromuscular Diseases and John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine (B.B., H.G., T.E., J.D., A.B., V.B., H.S., E.F., A.P., H.L., P.F.C., R.H.), and Institute of Neuroscience (R.G.W., J.M.), Newcastle University, Newcastle upon Tyne; Bristol Genetics Laboratory (T.A., M.G., N.F.), Pathology Sciences, North Bristol NHS Trust, Southmead Hospital; Medical Genetic Center (S.K.), Munich, Germany; Department of Paediatric Neurology (V.R.), Royal Victoria Infirmary, Newcastle upon Tyne Foundation Hospitals NHS Trust; Nuffield Department of Clinical Neurosciences (E.F.), University of Oxford; and Department of Clinical Neurosciences (P.F.C.), Cambridge Biomedical Campus, University of Cambridge, UK
| | - Helen Griffin
- From the MRC Centre for Neuromuscular Diseases and John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine (B.B., H.G., T.E., J.D., A.B., V.B., H.S., E.F., A.P., H.L., P.F.C., R.H.), and Institute of Neuroscience (R.G.W., J.M.), Newcastle University, Newcastle upon Tyne; Bristol Genetics Laboratory (T.A., M.G., N.F.), Pathology Sciences, North Bristol NHS Trust, Southmead Hospital; Medical Genetic Center (S.K.), Munich, Germany; Department of Paediatric Neurology (V.R.), Royal Victoria Infirmary, Newcastle upon Tyne Foundation Hospitals NHS Trust; Nuffield Department of Clinical Neurosciences (E.F.), University of Oxford; and Department of Clinical Neurosciences (P.F.C.), Cambridge Biomedical Campus, University of Cambridge, UK
| | - Roger G Whittaker
- From the MRC Centre for Neuromuscular Diseases and John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine (B.B., H.G., T.E., J.D., A.B., V.B., H.S., E.F., A.P., H.L., P.F.C., R.H.), and Institute of Neuroscience (R.G.W., J.M.), Newcastle University, Newcastle upon Tyne; Bristol Genetics Laboratory (T.A., M.G., N.F.), Pathology Sciences, North Bristol NHS Trust, Southmead Hospital; Medical Genetic Center (S.K.), Munich, Germany; Department of Paediatric Neurology (V.R.), Royal Victoria Infirmary, Newcastle upon Tyne Foundation Hospitals NHS Trust; Nuffield Department of Clinical Neurosciences (E.F.), University of Oxford; and Department of Clinical Neurosciences (P.F.C.), Cambridge Biomedical Campus, University of Cambridge, UK
| | - Thalia Antoniadi
- From the MRC Centre for Neuromuscular Diseases and John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine (B.B., H.G., T.E., J.D., A.B., V.B., H.S., E.F., A.P., H.L., P.F.C., R.H.), and Institute of Neuroscience (R.G.W., J.M.), Newcastle University, Newcastle upon Tyne; Bristol Genetics Laboratory (T.A., M.G., N.F.), Pathology Sciences, North Bristol NHS Trust, Southmead Hospital; Medical Genetic Center (S.K.), Munich, Germany; Department of Paediatric Neurology (V.R.), Royal Victoria Infirmary, Newcastle upon Tyne Foundation Hospitals NHS Trust; Nuffield Department of Clinical Neurosciences (E.F.), University of Oxford; and Department of Clinical Neurosciences (P.F.C.), Cambridge Biomedical Campus, University of Cambridge, UK
| | - Teresinha Evangelista
- From the MRC Centre for Neuromuscular Diseases and John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine (B.B., H.G., T.E., J.D., A.B., V.B., H.S., E.F., A.P., H.L., P.F.C., R.H.), and Institute of Neuroscience (R.G.W., J.M.), Newcastle University, Newcastle upon Tyne; Bristol Genetics Laboratory (T.A., M.G., N.F.), Pathology Sciences, North Bristol NHS Trust, Southmead Hospital; Medical Genetic Center (S.K.), Munich, Germany; Department of Paediatric Neurology (V.R.), Royal Victoria Infirmary, Newcastle upon Tyne Foundation Hospitals NHS Trust; Nuffield Department of Clinical Neurosciences (E.F.), University of Oxford; and Department of Clinical Neurosciences (P.F.C.), Cambridge Biomedical Campus, University of Cambridge, UK
| | - James Miller
- From the MRC Centre for Neuromuscular Diseases and John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine (B.B., H.G., T.E., J.D., A.B., V.B., H.S., E.F., A.P., H.L., P.F.C., R.H.), and Institute of Neuroscience (R.G.W., J.M.), Newcastle University, Newcastle upon Tyne; Bristol Genetics Laboratory (T.A., M.G., N.F.), Pathology Sciences, North Bristol NHS Trust, Southmead Hospital; Medical Genetic Center (S.K.), Munich, Germany; Department of Paediatric Neurology (V.R.), Royal Victoria Infirmary, Newcastle upon Tyne Foundation Hospitals NHS Trust; Nuffield Department of Clinical Neurosciences (E.F.), University of Oxford; and Department of Clinical Neurosciences (P.F.C.), Cambridge Biomedical Campus, University of Cambridge, UK
| | - Mark Greenslade
- From the MRC Centre for Neuromuscular Diseases and John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine (B.B., H.G., T.E., J.D., A.B., V.B., H.S., E.F., A.P., H.L., P.F.C., R.H.), and Institute of Neuroscience (R.G.W., J.M.), Newcastle University, Newcastle upon Tyne; Bristol Genetics Laboratory (T.A., M.G., N.F.), Pathology Sciences, North Bristol NHS Trust, Southmead Hospital; Medical Genetic Center (S.K.), Munich, Germany; Department of Paediatric Neurology (V.R.), Royal Victoria Infirmary, Newcastle upon Tyne Foundation Hospitals NHS Trust; Nuffield Department of Clinical Neurosciences (E.F.), University of Oxford; and Department of Clinical Neurosciences (P.F.C.), Cambridge Biomedical Campus, University of Cambridge, UK
| | - Natalie Forester
- From the MRC Centre for Neuromuscular Diseases and John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine (B.B., H.G., T.E., J.D., A.B., V.B., H.S., E.F., A.P., H.L., P.F.C., R.H.), and Institute of Neuroscience (R.G.W., J.M.), Newcastle University, Newcastle upon Tyne; Bristol Genetics Laboratory (T.A., M.G., N.F.), Pathology Sciences, North Bristol NHS Trust, Southmead Hospital; Medical Genetic Center (S.K.), Munich, Germany; Department of Paediatric Neurology (V.R.), Royal Victoria Infirmary, Newcastle upon Tyne Foundation Hospitals NHS Trust; Nuffield Department of Clinical Neurosciences (E.F.), University of Oxford; and Department of Clinical Neurosciences (P.F.C.), Cambridge Biomedical Campus, University of Cambridge, UK
| | - Jennifer Duff
- From the MRC Centre for Neuromuscular Diseases and John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine (B.B., H.G., T.E., J.D., A.B., V.B., H.S., E.F., A.P., H.L., P.F.C., R.H.), and Institute of Neuroscience (R.G.W., J.M.), Newcastle University, Newcastle upon Tyne; Bristol Genetics Laboratory (T.A., M.G., N.F.), Pathology Sciences, North Bristol NHS Trust, Southmead Hospital; Medical Genetic Center (S.K.), Munich, Germany; Department of Paediatric Neurology (V.R.), Royal Victoria Infirmary, Newcastle upon Tyne Foundation Hospitals NHS Trust; Nuffield Department of Clinical Neurosciences (E.F.), University of Oxford; and Department of Clinical Neurosciences (P.F.C.), Cambridge Biomedical Campus, University of Cambridge, UK
| | - Anna Bradshaw
- From the MRC Centre for Neuromuscular Diseases and John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine (B.B., H.G., T.E., J.D., A.B., V.B., H.S., E.F., A.P., H.L., P.F.C., R.H.), and Institute of Neuroscience (R.G.W., J.M.), Newcastle University, Newcastle upon Tyne; Bristol Genetics Laboratory (T.A., M.G., N.F.), Pathology Sciences, North Bristol NHS Trust, Southmead Hospital; Medical Genetic Center (S.K.), Munich, Germany; Department of Paediatric Neurology (V.R.), Royal Victoria Infirmary, Newcastle upon Tyne Foundation Hospitals NHS Trust; Nuffield Department of Clinical Neurosciences (E.F.), University of Oxford; and Department of Clinical Neurosciences (P.F.C.), Cambridge Biomedical Campus, University of Cambridge, UK
| | - Stephanie Kleinle
- From the MRC Centre for Neuromuscular Diseases and John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine (B.B., H.G., T.E., J.D., A.B., V.B., H.S., E.F., A.P., H.L., P.F.C., R.H.), and Institute of Neuroscience (R.G.W., J.M.), Newcastle University, Newcastle upon Tyne; Bristol Genetics Laboratory (T.A., M.G., N.F.), Pathology Sciences, North Bristol NHS Trust, Southmead Hospital; Medical Genetic Center (S.K.), Munich, Germany; Department of Paediatric Neurology (V.R.), Royal Victoria Infirmary, Newcastle upon Tyne Foundation Hospitals NHS Trust; Nuffield Department of Clinical Neurosciences (E.F.), University of Oxford; and Department of Clinical Neurosciences (P.F.C.), Cambridge Biomedical Campus, University of Cambridge, UK
| | - Veronika Boczonadi
- From the MRC Centre for Neuromuscular Diseases and John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine (B.B., H.G., T.E., J.D., A.B., V.B., H.S., E.F., A.P., H.L., P.F.C., R.H.), and Institute of Neuroscience (R.G.W., J.M.), Newcastle University, Newcastle upon Tyne; Bristol Genetics Laboratory (T.A., M.G., N.F.), Pathology Sciences, North Bristol NHS Trust, Southmead Hospital; Medical Genetic Center (S.K.), Munich, Germany; Department of Paediatric Neurology (V.R.), Royal Victoria Infirmary, Newcastle upon Tyne Foundation Hospitals NHS Trust; Nuffield Department of Clinical Neurosciences (E.F.), University of Oxford; and Department of Clinical Neurosciences (P.F.C.), Cambridge Biomedical Campus, University of Cambridge, UK
| | - Hannah Steele
- From the MRC Centre for Neuromuscular Diseases and John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine (B.B., H.G., T.E., J.D., A.B., V.B., H.S., E.F., A.P., H.L., P.F.C., R.H.), and Institute of Neuroscience (R.G.W., J.M.), Newcastle University, Newcastle upon Tyne; Bristol Genetics Laboratory (T.A., M.G., N.F.), Pathology Sciences, North Bristol NHS Trust, Southmead Hospital; Medical Genetic Center (S.K.), Munich, Germany; Department of Paediatric Neurology (V.R.), Royal Victoria Infirmary, Newcastle upon Tyne Foundation Hospitals NHS Trust; Nuffield Department of Clinical Neurosciences (E.F.), University of Oxford; and Department of Clinical Neurosciences (P.F.C.), Cambridge Biomedical Campus, University of Cambridge, UK
| | - Venkateswaran Ramesh
- From the MRC Centre for Neuromuscular Diseases and John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine (B.B., H.G., T.E., J.D., A.B., V.B., H.S., E.F., A.P., H.L., P.F.C., R.H.), and Institute of Neuroscience (R.G.W., J.M.), Newcastle University, Newcastle upon Tyne; Bristol Genetics Laboratory (T.A., M.G., N.F.), Pathology Sciences, North Bristol NHS Trust, Southmead Hospital; Medical Genetic Center (S.K.), Munich, Germany; Department of Paediatric Neurology (V.R.), Royal Victoria Infirmary, Newcastle upon Tyne Foundation Hospitals NHS Trust; Nuffield Department of Clinical Neurosciences (E.F.), University of Oxford; and Department of Clinical Neurosciences (P.F.C.), Cambridge Biomedical Campus, University of Cambridge, UK
| | - Edit Franko
- From the MRC Centre for Neuromuscular Diseases and John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine (B.B., H.G., T.E., J.D., A.B., V.B., H.S., E.F., A.P., H.L., P.F.C., R.H.), and Institute of Neuroscience (R.G.W., J.M.), Newcastle University, Newcastle upon Tyne; Bristol Genetics Laboratory (T.A., M.G., N.F.), Pathology Sciences, North Bristol NHS Trust, Southmead Hospital; Medical Genetic Center (S.K.), Munich, Germany; Department of Paediatric Neurology (V.R.), Royal Victoria Infirmary, Newcastle upon Tyne Foundation Hospitals NHS Trust; Nuffield Department of Clinical Neurosciences (E.F.), University of Oxford; and Department of Clinical Neurosciences (P.F.C.), Cambridge Biomedical Campus, University of Cambridge, UK
| | - Angela Pyle
- From the MRC Centre for Neuromuscular Diseases and John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine (B.B., H.G., T.E., J.D., A.B., V.B., H.S., E.F., A.P., H.L., P.F.C., R.H.), and Institute of Neuroscience (R.G.W., J.M.), Newcastle University, Newcastle upon Tyne; Bristol Genetics Laboratory (T.A., M.G., N.F.), Pathology Sciences, North Bristol NHS Trust, Southmead Hospital; Medical Genetic Center (S.K.), Munich, Germany; Department of Paediatric Neurology (V.R.), Royal Victoria Infirmary, Newcastle upon Tyne Foundation Hospitals NHS Trust; Nuffield Department of Clinical Neurosciences (E.F.), University of Oxford; and Department of Clinical Neurosciences (P.F.C.), Cambridge Biomedical Campus, University of Cambridge, UK
| | - Hanns Lochmüller
- From the MRC Centre for Neuromuscular Diseases and John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine (B.B., H.G., T.E., J.D., A.B., V.B., H.S., E.F., A.P., H.L., P.F.C., R.H.), and Institute of Neuroscience (R.G.W., J.M.), Newcastle University, Newcastle upon Tyne; Bristol Genetics Laboratory (T.A., M.G., N.F.), Pathology Sciences, North Bristol NHS Trust, Southmead Hospital; Medical Genetic Center (S.K.), Munich, Germany; Department of Paediatric Neurology (V.R.), Royal Victoria Infirmary, Newcastle upon Tyne Foundation Hospitals NHS Trust; Nuffield Department of Clinical Neurosciences (E.F.), University of Oxford; and Department of Clinical Neurosciences (P.F.C.), Cambridge Biomedical Campus, University of Cambridge, UK
| | - Patrick F Chinnery
- From the MRC Centre for Neuromuscular Diseases and John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine (B.B., H.G., T.E., J.D., A.B., V.B., H.S., E.F., A.P., H.L., P.F.C., R.H.), and Institute of Neuroscience (R.G.W., J.M.), Newcastle University, Newcastle upon Tyne; Bristol Genetics Laboratory (T.A., M.G., N.F.), Pathology Sciences, North Bristol NHS Trust, Southmead Hospital; Medical Genetic Center (S.K.), Munich, Germany; Department of Paediatric Neurology (V.R.), Royal Victoria Infirmary, Newcastle upon Tyne Foundation Hospitals NHS Trust; Nuffield Department of Clinical Neurosciences (E.F.), University of Oxford; and Department of Clinical Neurosciences (P.F.C.), Cambridge Biomedical Campus, University of Cambridge, UK
| | - Rita Horvath
- From the MRC Centre for Neuromuscular Diseases and John Walton Muscular Dystrophy Research Centre, Institute of Genetic Medicine (B.B., H.G., T.E., J.D., A.B., V.B., H.S., E.F., A.P., H.L., P.F.C., R.H.), and Institute of Neuroscience (R.G.W., J.M.), Newcastle University, Newcastle upon Tyne; Bristol Genetics Laboratory (T.A., M.G., N.F.), Pathology Sciences, North Bristol NHS Trust, Southmead Hospital; Medical Genetic Center (S.K.), Munich, Germany; Department of Paediatric Neurology (V.R.), Royal Victoria Infirmary, Newcastle upon Tyne Foundation Hospitals NHS Trust; Nuffield Department of Clinical Neurosciences (E.F.), University of Oxford; and Department of Clinical Neurosciences (P.F.C.), Cambridge Biomedical Campus, University of Cambridge, UK.
| |
Collapse
|
47
|
Boerboom A, Dion V, Chariot A, Franzen R. Molecular Mechanisms Involved in Schwann Cell Plasticity. Front Mol Neurosci 2017; 10:38. [PMID: 28261057 PMCID: PMC5314106 DOI: 10.3389/fnmol.2017.00038] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 01/31/2017] [Indexed: 01/09/2023] Open
Abstract
Schwann cell incredible plasticity is a hallmark of the utmost importance following nerve damage or in demyelinating neuropathies. After injury, Schwann cells undergo dedifferentiation before redifferentiating to promote nerve regeneration and complete functional recovery. This review updates and discusses the molecular mechanisms involved in the negative regulation of myelination as well as in the reprogramming of Schwann cells taking place early following nerve lesion to support repair. Significant advance has been made on signaling pathways and molecular components that regulate SC regenerative properties. These include for instance transcriptional regulators such as c-Jun or Notch, the MAPK and the Nrg1/ErbB2/3 pathways. This comprehensive overview ends with some therapeutical applications targeting factors that control Schwann cell plasticity and highlights the need to carefully modulate and balance this capacity to drive nerve repair.
Collapse
Affiliation(s)
| | - Valérie Dion
- GIGA-Neurosciences, University of Liège Liège, Belgium
| | - Alain Chariot
- GIGA-Molecular Biology of Diseases, University of LiègeLiège, Belgium; Walloon Excellence in Lifesciences and Biotechnology (WELBIO)Wavre, Belgium
| | | |
Collapse
|
48
|
Mindos T, Dun XP, North K, Doddrell RDS, Schulz A, Edwards P, Russell J, Gray B, Roberts SL, Shivane A, Mortimer G, Pirie M, Zhang N, Pan D, Morrison H, Parkinson DB. Merlin controls the repair capacity of Schwann cells after injury by regulating Hippo/YAP activity. J Cell Biol 2017; 216:495-510. [PMID: 28137778 PMCID: PMC5294779 DOI: 10.1083/jcb.201606052] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 10/23/2016] [Accepted: 12/27/2016] [Indexed: 02/06/2023] Open
Abstract
Loss of the Merlin tumor suppressor and activation of the Hippo signaling pathway play major roles in the control of cell proliferation and tumorigenesis. We have identified completely novel roles for Merlin and the Hippo pathway effector Yes-associated protein (YAP) in the control of Schwann cell (SC) plasticity and peripheral nerve repair after injury. Injury to the peripheral nervous system (PNS) causes a dramatic shift in SC molecular phenotype and the generation of repair-competent SCs, which direct functional repair. We find that loss of Merlin in these cells causes a catastrophic failure of axonal regeneration and remyelination in the PNS. This effect is mediated by activation of YAP expression in Merlin-null SCs, and loss of YAP restores axonal regrowth and functional repair. This work identifies new mechanisms that control the regenerative potential of SCs and gives new insight into understanding the correct control of functional nerve repair in the PNS.
Collapse
Affiliation(s)
- Thomas Mindos
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
| | - Xin-Peng Dun
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
| | - Katherine North
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
- University of Bath, Bath BA2 7AY, England, UK
| | - Robin D S Doddrell
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
| | - Alexander Schulz
- Leibniz Institute for Age Research - Fritz Lipmann Institute Jena, D-07745 Jena, Germany
| | - Philip Edwards
- Department of Cellular and Anatomical Pathology, Derriford Hospital, Plymouth PL6 8DH, England, UK
| | - James Russell
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
| | - Bethany Gray
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
- University of Bath, Bath BA2 7AY, England, UK
| | - Sheridan L Roberts
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
| | - Aditya Shivane
- Department of Cellular and Anatomical Pathology, Derriford Hospital, Plymouth PL6 8DH, England, UK
| | - Georgina Mortimer
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
| | - Melissa Pirie
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
| | - Nailing Zhang
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Duojia Pan
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Helen Morrison
- Leibniz Institute for Age Research - Fritz Lipmann Institute Jena, D-07745 Jena, Germany
| | - David B Parkinson
- Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth PL6 8BU, England, UK
| |
Collapse
|
49
|
Heffernan C, Jain MR, Liu T, Kim H, Barretto K, Li H, Maurel P. Nectin-like 4 Complexes with Choline Transporter-like Protein-1 and Regulates Schwann Cell Choline Homeostasis and Lipid Biogenesis in Vitro. J Biol Chem 2017; 292:4484-4498. [PMID: 28119456 DOI: 10.1074/jbc.m116.747816] [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: 07/12/2016] [Revised: 01/13/2017] [Indexed: 11/06/2022] Open
Abstract
Nectin-like 4 (NECL4, CADM4) is a Schwann cell-specific cell adhesion molecule that promotes axo-glial interactions. In vitro and in vivo studies have shown that NECL4 is necessary for proper peripheral nerve myelination. However, the molecular mechanisms that are regulated by NECL4 and affect peripheral myelination currently remain unclear. We used an in vitro approach to begin identifying some of the mechanisms that could explain NECL4 function. Using mass spectrometry and Western blotting techniques, we have identified choline transporter-like 1 (CTL1) as a putative complexing partner with NECL4. We show that intracellular choline levels are significantly elevated in NECL4-deficient Schwann cells. The analysis of extracellular d9-choline uptake revealed a deficit in the amount of d9-choline found inside NECL4-deficient Schwann cells, suggestive of either reduced transport capabilities or increased metabolization of transported choline. An extensive lipidomic screen of choline derivatives showed that total phosphatidylcholine and phosphatidylinositol (but not diacylglycerol or sphingomyelin) are significantly elevated in NECL4-deficient Schwann cells, particularly specific subspecies of phosphatidylcholine carrying very long polyunsaturated fatty acid chains. Finally, CTL1-deficient Schwann cells are significantly impaired in their ability to myelinate neurites in vitro To our knowledge, this is the first demonstration of a bona fide cell adhesion molecule, NECL4, regulating choline homeostasis and lipid biogenesis. Phosphatidylcholines are major myelin phospholipids, and several phosphorylated phosphatidylinositol species are known to regulate key aspects of peripheral myelination. Furthermore, the biophysical properties imparted to plasma membranes are regulated by fatty acid chain profiles. Therefore, it will be important to translate these in vitro observations to in vivo studies of NECL4 and CTL1-deficient mice.
Collapse
Affiliation(s)
- Corey Heffernan
- From the Department of Biological Sciences, Rutgers, the State University of New Jersey, Newark, New Jersey 07102-1814 and
| | - Mohit R Jain
- the Center for Advanced Proteomics Research, New Jersey Medical School, Newark, New Jersey 07103
| | - Tong Liu
- the Center for Advanced Proteomics Research, New Jersey Medical School, Newark, New Jersey 07103
| | - Hyosung Kim
- From the Department of Biological Sciences, Rutgers, the State University of New Jersey, Newark, New Jersey 07102-1814 and
| | - Kevin Barretto
- From the Department of Biological Sciences, Rutgers, the State University of New Jersey, Newark, New Jersey 07102-1814 and
| | - Hong Li
- the Center for Advanced Proteomics Research, New Jersey Medical School, Newark, New Jersey 07103
| | - Patrice Maurel
- From the Department of Biological Sciences, Rutgers, the State University of New Jersey, Newark, New Jersey 07102-1814 and
| |
Collapse
|
50
|
Castelnovo LF, Bonalume V, Melfi S, Ballabio M, Colleoni D, Magnaghi V. Schwann cell development, maturation and regeneration: a focus on classic and emerging intracellular signaling pathways. Neural Regen Res 2017; 12:1013-1023. [PMID: 28852375 PMCID: PMC5558472 DOI: 10.4103/1673-5374.211172] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The development, maturation and regeneration of Schwann cells (SCs), the main glial cells of the peripheral nervous system, require the coordinate and complementary interaction among several factors, signals and intracellular pathways. These regulatory molecules consist of integrins, neuregulins, growth factors, hormones, neurotransmitters, as well as entire intracellular pathways including protein-kinase A, C, Akt, Erk/MAPK, Hippo, mTOR, etc. For instance, Hippo pathway is overall involved in proliferation, apoptosis, regeneration and organ size control, being crucial in cancer proliferation process. In SCs, Hippo is linked to merlin and YAP/TAZ signaling and it seems to respond to mechanic/physical challenges. Recently, among factors regulating SCs, also the signaling intermediates Src tyrosine kinase and focal adhesion kinase (FAK) proved relevant for SC fate, participating in the regulation of adhesion, motility, migration and in vitro myelination. In SCs, the factors Src and FAK are regulated by the neuroactive steroid allopregnanolone, thus corroborating the importance of this steroid in the control of SC maturation. In this review, we illustrate some old and novel signaling pathways modulating SC biology and functions during the different developmental, mature and regenerative states.
Collapse
Affiliation(s)
- Luca Franco Castelnovo
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Veronica Bonalume
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Simona Melfi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Marinella Ballabio
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Deborah Colleoni
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Valerio Magnaghi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|