1
|
Salzer J, Feltri ML, Jacob C. Schwann Cell Development and Myelination. Cold Spring Harb Perspect Biol 2024; 16:a041360. [PMID: 38503507 PMCID: PMC11368196 DOI: 10.1101/cshperspect.a041360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Glial cells in the peripheral nervous system (PNS), which arise from the neural crest, include axon-associated Schwann cells (SCs) in nerves, synapse-associated SCs at the neuromuscular junction, enteric glia, perikaryon-associated satellite cells in ganglia, and boundary cap cells at the border between the central nervous system (CNS) and the PNS. Here, we focus on axon-associated SCs. These SCs progress through a series of formative stages, which culminate in the generation of myelinating SCs that wrap large-caliber axons and of nonmyelinating (Remak) SCs that enclose multiple, small-caliber axons. In this work, we describe SC development, extrinsic signals from the axon and extracellular matrix (ECM) and the intracellular signaling pathways they activate that regulate SC development, and the morphogenesis and organization of myelinating SCs and the myelin sheath. We review the impact of SCs on the biology and integrity of axons and their emerging role in regulating peripheral nerve architecture. Finally, we explain how transcription and epigenetic factors control and fine-tune SC development and myelination.
Collapse
Affiliation(s)
- James Salzer
- Neuroscience Institute, New York University Grossman School of Medicine, New York, New York 10016, USA
| | - M Laura Feltri
- Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York 14203, USA
- IRCCS Neurological Institute Carlo Besta, Milano 20133, Italy
- Department of Biotechnology and Translational Sciences, Universita' Degli Studi di Milano, Milano 20133, Italy
| | - Claire Jacob
- Faculty of Biology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz 55128, Germany
| |
Collapse
|
2
|
Miyazaki Y, Otsuka T, Yamagata Y, Endo T, Sanbo M, Sano H, Kobayashi K, Inahashi H, Kornau HC, Schmitz D, Prüss H, Meijer D, Hirabayashi M, Fukata Y, Fukata M. Oligodendrocyte-derived LGI3 and its receptor ADAM23 organize juxtaparanodal Kv1 channel clustering for short-term synaptic plasticity. Cell Rep 2024; 43:113634. [PMID: 38194969 PMCID: PMC10828548 DOI: 10.1016/j.celrep.2023.113634] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/31/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024] Open
Abstract
Neurodevelopmental disorders, such as intellectual disability (ID), epilepsy, and autism, involve altered synaptic transmission and plasticity. Functional characterization of their associated genes is vital for understanding physio-pathological brain functions. LGI3 is a recently recognized ID-associated gene encoding a secretory protein related to an epilepsy-gene product, LGI1. Here, we find that LGI3 is uniquely secreted from oligodendrocytes in the brain and enriched at juxtaparanodes of myelinated axons, forming nanoscale subclusters. Proteomic analysis using epitope-tagged Lgi3 knockin mice shows that LGI3 uses ADAM23 as a receptor and selectively co-assembles with Kv1 channels. A lack of Lgi3 in mice disrupts juxtaparanodal clustering of ADAM23 and Kv1 channels and suppresses Kv1-channel-mediated short-term synaptic plasticity. Collectively, this study identifies an extracellular organizer of juxtaparanodal Kv1 channel clustering for finely tuned synaptic transmission. Given the defective secretion of the LGI3 missense variant, we propose a molecular pathway, the juxtaparanodal LGI3-ADAM23-Kv1 channel, for understanding neurodevelopmental disorders.
Collapse
Affiliation(s)
- Yuri Miyazaki
- Division of Neuropharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Takeshi Otsuka
- Section of Cellular Electrophysiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan; Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Aichi 444-8585, Japan
| | - Yoko Yamagata
- Section of Multilayer Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan
| | | | - Makoto Sanbo
- Section of Mammalian Transgenesis, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Hiromi Sano
- Division of Behavioral Neuropharmacology, International Center for Brain Science, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Kenta Kobayashi
- Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Aichi 444-8585, Japan; Section of Viral Vector Development, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8585, Japan
| | - Hiroki Inahashi
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Hans-Christian Kornau
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany; Neuroscience Research Center (NWFZ), Cluster NeuroCure, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dietmar Schmitz
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany; Neuroscience Research Center (NWFZ), Cluster NeuroCure, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Harald Prüss
- German Center for Neurodegenerative Diseases (DZNE) Berlin, Berlin, Germany; Helmholtz Innovation Lab BaoBab (Brain Antibody-omics and B-cell Lab), Berlin, Germany; Department of Neurology and Experimental Neurology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Dies Meijer
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK; Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK
| | - Masumi Hirabayashi
- Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Aichi 444-8585, Japan; Section of Mammalian Transgenesis, Center for Genetic Analysis of Behavior, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan
| | - Yuko Fukata
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan; Division of Molecular and Cellular Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan.
| | - Masaki Fukata
- Division of Neuropharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan; Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki, Aichi 444-8787, Japan; Graduate Institute for Advanced Studies, SOKENDAI, Okazaki, Aichi 444-8585, Japan.
| |
Collapse
|
3
|
Nosková L, Fukata Y, Stránecký V, Šaligová J, Bodnárová O, Giertlová M, Fukata M, Kmoch S. ADAM22 ethnic-specific variant reducing binding of membrane-associated guanylate kinases causes focal epilepsy and behavioural disorder. Brain Commun 2023; 5:fcad295. [PMID: 37953841 PMCID: PMC10636567 DOI: 10.1093/braincomms/fcad295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 07/19/2023] [Accepted: 10/25/2023] [Indexed: 11/14/2023] Open
Abstract
Pathogenic variants of ADAM22 affecting either its biosynthesis and/or its interactions with either LGI1 and/or PSD-95 have been recently identified in individuals with developmental and epileptic encephalopathy. Here, we describe a girl with seizures, delayed psychomotor development, and behavioural disorder, carrying a homozygous variant in ADAM22 (NM_021723.5:c.2714C > T). The variant has a surprisingly high frequency in the Roma population of the Czech and Slovak Republic, with 11 of 213 (∼5.2%) healthy Roma individuals identified as heterozygous carriers. Structural in silico characterization revealed that the genetic variant encodes the missense variant p.S905F, which localizes to the PDZ-binding motif of ADAM22. Studies in transiently transfected mammalian cells revealed that the variant has no effect on biosynthesis and stability of ADAM22. Rather, protein-protein interaction studies showed that the p.S905F variant specifically impairs ADAM22 binding to PSD-95 and other proteins from a family of membrane-associated guanylate kinases, while it has only minor effect on ADAM22-LGI1 interaction. Our study indicates that a significant proportion of epilepsy in patients of Roma ancestry may be caused by homozygous c.2714C > T variants in ADAM22. The study of this ADAM22 variant highlights a novel pathogenic mechanism of ADAM22 dysfunction and reconfirms an essential role of interaction of ADAM22 with membrane-associated guanylate kinases in seizure protection in humans.
Collapse
Affiliation(s)
- Lenka Nosková
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, 1st Faculty of Medicine, Charles University in Prague, 128 08 Prague 2, Czech Republic
| | - Yuko Fukata
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki 444-8585, Japan
- Division of Molecular and Cellular Pharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Viktor Stránecký
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, 1st Faculty of Medicine, Charles University in Prague, 128 08 Prague 2, Czech Republic
| | - Jana Šaligová
- Children's Faculty Hospital, Košice 040 11, Slovakia
| | | | - Mária Giertlová
- Medical Genetics Outpatient Service, Unilabs Slovakia Ltd, Košice 040 01, Slovakia
- Department of Paediatric and Adolescent Medicine, Faculty of Medicine, P.J. Šafárik University,Košice 040 01, Slovak Republic
| | - Masaki Fukata
- Division of Membrane Physiology, Department of Molecular and Cellular Physiology, National Institute for Physiological Sciences, National Institutes of Natural Sciences, Okazaki 444-8787, Japan
- Department of Physiological Sciences, School of Life Science, SOKENDAI (The Graduate University for Advanced Studies), Okazaki 444-8585, Japan
- Division of Neuropharmacology, Nagoya University Graduate School of Medicine, Nagoya 466-8550, Japan
| | - Stanislav Kmoch
- Research Unit for Rare Diseases, Department of Pediatrics and Inherited Metabolic Disorders, 1st Faculty of Medicine, Charles University in Prague, 128 08 Prague 2, Czech Republic
| |
Collapse
|
4
|
Kozar-Gillan N, Velichkova A, Kanatouris G, Eshed-Eisenbach Y, Steel G, Jaegle M, Aunin E, Peles E, Torsney C, Meijer DN. LGI3/2-ADAM23 interactions cluster Kv1 channels in myelinated axons to regulate refractory period. J Cell Biol 2023; 222:e202211031. [PMID: 36828548 PMCID: PMC9997507 DOI: 10.1083/jcb.202211031] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/18/2022] [Accepted: 01/17/2023] [Indexed: 02/26/2023] Open
Abstract
Along myelinated axons, Shaker-type potassium channels (Kv1) accumulate at high density in the juxtaparanodal region, directly adjacent to the paranodal axon-glia junctions that flank the nodes of Ranvier. However, the mechanisms that control the clustering of Kv1 channels, as well as their function at this site, are still poorly understood. Here we demonstrate that axonal ADAM23 is essential for both the accumulation and stability of juxtaparanodal Kv1 complexes. The function of ADAM23 is critically dependent on its interaction with its extracellular ligands LGI2 and LGI3. Furthermore, we demonstrate that juxtaparanodal Kv1 complexes affect the refractory period, thus enabling high-frequency burst firing of action potentials. Our findings not only reveal a previously unknown molecular pathway that regulates Kv1 channel clustering, but they also demonstrate that the juxtaparanodal Kv1 channels that are concealed below the myelin sheath, play a significant role in modifying axonal physiology.
Collapse
Affiliation(s)
- Nina Kozar-Gillan
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh. UK
| | | | - George Kanatouris
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh. UK
| | - Yael Eshed-Eisenbach
- Department of Molecular Cell Biology and Molecular Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Gavin Steel
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh. UK
| | | | - Eerik Aunin
- Biomedical Sciences, ErasmusMC, Rotterdam, Netherlands
| | - Elior Peles
- Department of Molecular Cell Biology and Molecular Neurobiology, Weizmann Institute of Science, Rehovot, Israel
| | - Carole Torsney
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh. UK
- Simons Initiative for the Developing Brain, University of Edinburgh, Edinburgh. UK
| | - Dies N. Meijer
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh. UK
- Muir Maxwell Epilepsy Centre, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
5
|
Zhang X, Kira JI, Ogata H, Imamura T, Mitsuishi M, Fujii T, Kobayashi M, Kitagawa K, Namihira Y, Ohya Y, Maimaitijiang G, Yamasaki R, Fukata Y, Fukata M, Isobe N, Nakamura Y. Anti-LGI4 Antibody Is a Novel Juxtaparanodal Autoantibody for Chronic Inflammatory Demyelinating Polyneuropathy. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2023; 10:10/2/e200081. [PMID: 36631269 PMCID: PMC9833819 DOI: 10.1212/nxi.0000000000200081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 11/10/2022] [Indexed: 01/13/2023]
Abstract
BACKGROUND AND OBJECTIVES The objective of this study was to discover novel nodal autoantibodies in chronic inflammatory demyelinating polyneuropathy (CIDP). METHODS We screened for autoantibodies that bind to mouse sciatic nerves and dorsal root ganglia (DRG) using indirect immunofluorescence (IFA) assays with sera from 113 patients with CIDP seronegative for anti-neurofascin 155 and anticontactin-1 antibodies and 127 controls. Western blotting, IFA assays using HEK293T cells transfected with relevant antigen expression plasmids, and cell-based RNA interference assays were used to identify target antigens. Krox20 and Periaxin expression, both of which independently control peripheral nerve myelination, was assessed by quantitative real-time PCR after application of patient and control sera to Schwann cells. RESULTS Sera from 4 patients with CIDP, but not control sera, selectively bound to the nodal regions of sciatic nerves and DRG satellite glia (p = 0.048). The main immunoglobulin G (IgG) subtype was IgG4. IgG from these 4 patients stained a 60-kDa band on Western blots of mouse DRG and sciatic nerve lysates. These features indicated leucine-rich repeat LGI family member 4 (LGI4) as a candidate antigen. A commercial anti-LGI4 antibody and IgG from all 4 seropositive patients with CIDP showed the same immunostaining patterns of DRG and cultured rat Schwann cells and bound to the 60-kDa protein in Western blots of LGI4 overexpression lysates. IgG from 3 seropositive patients, but none from controls, bound to cells cotransfected with plasmids containing LGI4 and a disintegrin and metalloprotease domain-containing protein 22 (ADAM22), an LGI4 receptor. In cultured rat Schwann and human melanoma cells constitutively expressing LGI4, LGI4 siRNA effectively downregulated LGI4 and reduced patients' IgG binding compared with scrambled siRNA. Application of serum from a positive patient to Schwann cells expressing ADAM22 significantly reduced the expression of Krox20, but not Periaxin. Anti-LGI4 antibody-positive patients had a relatively old age at onset (mean age 58 years), motor weakness, deep and superficial sensory impairment with Romberg sign, and extremely high levels of CSF protein. Three patients showed subacute CIDP onset resembling Guillain-Barré syndrome. DISCUSSION IgG4 anti-LGI4 antibodies are found in some elderly patients with CIDP who present subacute sensory impairment and motor weakness and are worth measuring, particularly in patients with symptoms resembling Guillain-Barré syndrome.
Collapse
Affiliation(s)
| | - Jun-Ichi Kira
- From the Translational Neuroscience Center (X.Z., J.K., T.I., M.M., G.M., Y. Nakamura), Graduate School of Medicine, International University of Health and Welfare, Okawa; School of Pharmacy at Fukuoka (J.K., T.I., Y. Nakamura), International University of Health and Welfare, Okawa; Department of Neurology (J.K., Y. Nakamura), Brain and Nerve Center, Fukuoka Central Hospital, International University of Health and Welfare, Fukuoka; Department of Neurology (H.O., T.F., R.Y., N.I.), Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka; Department of Neurology (M.K., K.K.), Tokyo Women's Medical University Hospital, Tokyo; Department of Cardiovascular Medicine (Y. Namihira, Y.O.), Nephrology, and Neurology, Graduate School of Medicine, University of Ryukyus, Okinawa; and Division of Membrane Physiology (Y.F., M.F.), National Institute for Physiological Sciences, Okazaki, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Kokan N, Witt S, Sandhu S, Hutter H. lron-11 guides axons in the ventral nerve cord of Caenorhabditis elegans. PLoS One 2022; 17:e0278258. [PMID: 36449480 PMCID: PMC9710760 DOI: 10.1371/journal.pone.0278258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/13/2022] [Indexed: 12/02/2022] Open
Abstract
For the nervous system to develop properly, neurons must connect in a precise way to form functional networks. This requires that outgrowing neuronal processes (axons) navigate to their target areas, where they establish proper synaptic connections. The molecular basis of this navigation process is not firmly understood. A candidate family containing putative receptors acting in various aspects of neuronal development including axon navigation are transmembrane proteins of the extracellular Leucine-Rich Repeat family (eLRRs). We systematically tested members of this family in C. elegans for a role in axon navigation in the ventral nerve cord (VNC). We found that lron-11 mutants showed VNC navigation defects in several classes of neurons, including a pioneer neuron and various classes of interneurons and motoneurons. This suggests that while most members of the lron-family do not seem to have a role in axon navigation in the VNC, lron-11 is likely to be a receptor required for correct navigation of axons in the VNC of C. elegans.
Collapse
Affiliation(s)
- Nikolas Kokan
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Skyla Witt
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Saru Sandhu
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
| | - Harald Hutter
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC, Canada
- * E-mail:
| |
Collapse
|
7
|
Marafi D, Kozar N, Duan R, Bradley S, Yokochi K, Al Mutairi F, Saadi NW, Whalen S, Brunet T, Kotzaeridou U, Choukair D, Keren B, Nava C, Kato M, Arai H, Froukh T, Faqeih EA, AlAsmari AM, Saleh MM, Pinto e Vairo F, Pichurin PN, Klee EW, Schmitz CT, Grochowski CM, Mitani T, Herman I, Calame DG, Fatih JM, Du H, Coban-Akdemir Z, Pehlivan D, Jhangiani SN, Gibbs RA, Miyatake S, Matsumoto N, Wagstaff LJ, Posey JE, Lupski JR, Meijer D, Wagner M. A reverse genetics and genomics approach to gene paralog function and disease: Myokymia and the juxtaparanode. Am J Hum Genet 2022; 109:1713-1723. [PMID: 35948005 PMCID: PMC9502070 DOI: 10.1016/j.ajhg.2022.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 07/01/2022] [Indexed: 11/16/2022] Open
Abstract
The leucine-rich glioma-inactivated (LGI) family consists of four highly conserved paralogous genes, LGI1-4, that are highly expressed in mammalian central and/or peripheral nervous systems. LGI1 antibodies are detected in subjects with autoimmune limbic encephalitis and peripheral nerve hyperexcitability syndromes (PNHSs) such as Isaacs and Morvan syndromes. Pathogenic variations of LGI1 and LGI4 are associated with neurological disorders as disease traits including familial temporal lobe epilepsy and neurogenic arthrogryposis multiplex congenita 1 with myelin defects, respectively. No human disease has been reported associated with either LGI2 or LGI3. We implemented exome sequencing and family-based genomics to identify individuals with deleterious variants in LGI3 and utilized GeneMatcher to connect practitioners and researchers worldwide to investigate the clinical and electrophysiological phenotype in affected subjects. We also generated Lgi3-null mice and performed peripheral nerve dissection and immunohistochemistry to examine the juxtaparanode LGI3 microarchitecture. As a result, we identified 16 individuals from eight unrelated families with loss-of-function (LoF) bi-allelic variants in LGI3. Deep phenotypic characterization showed LGI3 LoF causes a potentially clinically recognizable PNHS trait characterized by global developmental delay, intellectual disability, distal deformities with diminished reflexes, visible facial myokymia, and distinctive electromyographic features suggestive of motor nerve instability. Lgi3-null mice showed reduced and mis-localized Kv1 channel complexes in myelinated peripheral axons. Our data demonstrate bi-allelic LoF variants in LGI3 cause a clinically distinguishable disease trait of PNHS, most likely caused by disturbed Kv1 channel distribution in the absence of LGI3.
Collapse
Affiliation(s)
- Dana Marafi
- Department of Pediatrics, Faculty of Medicine, Kuwait University, P.O. Box 24923, Safat 13110, Kuwait,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Nina Kozar
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Ruizhi Duan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Stephen Bradley
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Kenji Yokochi
- Department of Pediatrics, Toyohashi Municipal Hospital, Toyohashi, Aichi 441-8570, Japan,Department of Pediatrics, Seirei Mikatahara General Hospital, Shizuoka 433-8558, Japan
| | - Fuad Al Mutairi
- Genetics and Precision Medicine Department, King Abdullah Specialized Children’s Hospital, King Abdulaziz Medical City, Ministry of National Guard Health Affairs, P.O. Box 22490, Riyadh 11426, Kingdom of Saudi Arabia,King Abdullah International Research Center, King Saud Bin Abdulaziz University for Health Sciences, Ministry of National Guard Health Affairs, Riyadh, Kingdom of Saudi Arabia
| | - Nebal Waill Saadi
- College of Medicine, University of Baghdad, Baghdad 10001, Iraq,Children Welfare Teaching Hospital, Medical City Complex, Baghdad 10001, Iraq
| | - Sandra Whalen
- UF de Génétique Clinique et Centre de Reference Anomalies du Développement et Syndromes Malformatifs, APHP, Sorbonne Université, Hôpital Trousseau, 75005 Paris, France
| | - Theresa Brunet
- Institute of Human Genetics, Faculty of Medicine, Technical University Munich, Munich, Germany,Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Urania Kotzaeridou
- Division of Child Neurology and Inherited Metabolic Diseases, Centre for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniela Choukair
- Division of Pediatric Endocrinology, Centre for Pediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Boris Keren
- Département de Génétique, Hôpital Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris, Paris 75013, France
| | - Caroline Nava
- Département de Génétique, Hôpital Pitié-Salpêtrière, Assistance Publique - Hôpitaux de Paris, Paris 75013, France
| | - Mitsuhiro Kato
- Department of Pediatrics, Showa University School of Medicine, Tokyo 142-8666, Japan
| | - Hiroshi Arai
- Department of Pediatric Neurology, Bobath Memorial Hospital, Osaka 536-0023, Japan
| | - Tawfiq Froukh
- Department of Biotechnology and Genetic Engineering, Philadelphia University, Amman, Jordan
| | - Eissa Ali Faqeih
- Section of Medical Genetics, King Fahad Medical City, Children’s Specialist Hospital, Riyadh, Saudi Arabia
| | - Ali M. AlAsmari
- Section of Medical Genetics, King Fahad Medical City, Children’s Specialist Hospital, Riyadh, Saudi Arabia
| | - Mohammed M. Saleh
- Section of Medical Genetics, King Fahad Medical City, Children’s Specialist Hospital, Riyadh, Saudi Arabia
| | - Filippo Pinto e Vairo
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA,Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA
| | | | - Eric W. Klee
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA,Department of Clinical Genomics, Mayo Clinic, Rochester, MN, USA,Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, USA
| | | | | | - Tadahiro Mitani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Isabella Herman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA,Section of Pediatric Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA,Texas Children’s Hospital, Houston, TX 77030, USA
| | - Daniel G. Calame
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA,Section of Pediatric Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA,Texas Children’s Hospital, Houston, TX 77030, USA
| | - Jawid M. Fatih
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Haowei Du
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Zeynep Coban-Akdemir
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA,Human Genetics Center, Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Davut Pehlivan
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA,Section of Pediatric Neurology and Developmental Neuroscience, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA,Texas Children’s Hospital, Houston, TX 77030, USA
| | - Shalini N. Jhangiani
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Richard A. Gibbs
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA,Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA
| | - Satoko Miyatake
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan,Clinical Genetics Department, Yokohama City University Hospital, Yokohama, Kanagawa 236-0004, Japan
| | - Naomichi Matsumoto
- Department of Human Genetics, Yokohama City University Graduate School of Medicine, Yokohama, Kanagawa 236-0004, Japan
| | - Laura J. Wagstaff
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Jennifer E. Posey
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
| | - James R. Lupski
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA,Texas Children’s Hospital, Houston, TX 77030, USA,Human Genome Sequencing Center, Baylor College of Medicine, Houston, TX 77030, USA,Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA,Corresponding author
| | - Dies Meijer
- Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, UK.
| | - Matias Wagner
- Institute for Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany,Institute of Human Genetics, Technical University Munich, Munich, Germany
| |
Collapse
|
8
|
Abstract
Schwann cells in the peripheral nervous system (PNS) are essential for the support and myelination of axons, ensuring fast and accurate communication between the central nervous system and the periphery. Schwann cells and related glia accompany innervating axons in virtually all tissues in the body, where they exhibit remarkable plasticity and the ability to modulate pathology in extraordinary, and sometimes surprising, ways. Here, we provide a brief overview of the various glial cell types in the PNS and describe the cornerstone cellular and molecular processes that enable Schwann cells to perform their canonical functions. We then dive into discussing exciting noncanonical functions of Schwann cells and related PNS glia, which include their role in organizing the PNS, in regulating synaptic activity and pain, in modulating immunity, in providing a pool of stem cells for different organs, and, finally, in influencing cancer.
Collapse
Affiliation(s)
- Carla Taveggia
- Axo-Glial Interaction Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy;
| | - M. Laura Feltri
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| |
Collapse
|
9
|
Previtali SC. Peripheral Nerve Development and the Pathogenesis of Peripheral Neuropathy: the Sorting Point. Neurotherapeutics 2021; 18:2156-2168. [PMID: 34244926 PMCID: PMC8804061 DOI: 10.1007/s13311-021-01080-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2021] [Indexed: 12/12/2022] Open
Abstract
Nerve development requires a coordinated sequence of events and steps to be accomplished for the generation of functional peripheral nerves to convey sensory and motor signals. Any abnormality during development may result in pathological structure and function of the nerve, which evolves in peripheral neuropathy. In this review, we will briefly describe different steps of nerve development while we will mostly focus on the molecular mechanisms involved in radial sorting of axons, one of these nerve developmental steps. We will summarize current knowledge of molecular pathways so far reported in radial sorting and their possible interactions. Finally, we will describe how disruption of these pathways may result in human neuropathies.
Collapse
Affiliation(s)
- Stefano C Previtali
- Neuromuscular Repair Unit, InSpe (Institute of Experimental Neurology) and Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
10
|
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
|
11
|
The Efficacy of Schwann-Like Differentiated Muscle-Derived Stem Cells in Treating Rodent Upper Extremity Peripheral Nerve Injury. Plast Reconstr Surg 2021; 148:787-798. [PMID: 34550935 DOI: 10.1097/prs.0000000000008383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND There is a pressing need to identify alternative mesenchymal stem cell sources for Schwann cell cellular replacement therapy, to improve peripheral nerve regeneration. This study assessed the efficacy of Schwann cell-like cells (induced muscle-derived stem cells) differentiated from muscle-derived stem cells (MDSCs) in augmenting nerve regeneration and improving muscle function after nerve trauma. METHODS The Schwann cell-like nature of induced MDSCs was characterized in vitro using immunofluorescence, flow cytometry, microarray, and reverse-transcription polymerase chain reaction. In vivo, four groups (n = 5 per group) of rats with median nerve injuries were examined: group 1 animals were treated with intraneural phosphate-buffered saline after cold and crush axonotmesis (negative control); group 2 animals were no-injury controls; group 3 animals were treated with intraneural green fluorescent protein-positive MDSCs; and group 4 animals were treated with green fluorescent protein-positive induced MDSCs. All animals underwent weekly upper extremity functional testing. Rats were euthanized 5 weeks after treatment. The median nerve and extrinsic finger flexors were harvested for nerve histomorphometry, myelination, muscle weight, and atrophy analyses. RESULTS In vitro, induced MDSCs recapitulated native Schwann cell gene expression patterns and up-regulated pathways involved in neuronal growth/signaling. In vivo, green fluorescent protein-positive induced MDSCs remained stably transformed 5 weeks after injection. Induced MDSC therapy decreased muscle atrophy after median nerve injury (p = 0.0143). Induced MDSC- and MDSC-treated animals demonstrated greater functional muscle recovery when compared to untreated controls (hand grip after induced MDSC treatment: group 1, 0.91 N; group 4, 3.38 N); p < 0.0001) at 5 weeks after treatment. This may demonstrate the potential beneficial effects of MDSC therapy, regardless of differentiation stage. CONCLUSION Both MDSCs and induced MDSCs decrease denervation muscle atrophy and improve subsequent functional outcomes after upper extremity nerve trauma in rodents.
Collapse
|
12
|
Drulis-Fajdasz D, Gostomska-Pampuch K, Duda P, Wiśniewski JR, Rakus D. Quantitative Proteomics Reveals Significant Differences between Mouse Brain Formations in Expression of Proteins Involved in Neuronal Plasticity during Aging. Cells 2021; 10:2021. [PMID: 34440790 PMCID: PMC8393337 DOI: 10.3390/cells10082021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/30/2021] [Accepted: 08/05/2021] [Indexed: 12/22/2022] Open
Abstract
Aging is associated with a general decline in cognitive functions, which appears to be due to alterations in the amounts of proteins involved in the regulation of synaptic plasticity. Here, we present a quantitative analysis of proteins involved in neurotransmission in three brain regions, namely, the hippocampus, the cerebral cortex and the cerebellum, in mice aged 1 and 22 months, using the total protein approach technique. We demonstrate that although the titer of some proteins involved in neurotransmission and synaptic plasticity is affected by aging in a similar manner in all the studied brain formations, in fact, each of the formations represents its own mode of aging. Generally, the hippocampal and cortical proteomes are much more unstable during the lifetime than the cerebellar proteome. The data presented here provide a general picture of the effect of physiological aging on synaptic plasticity and might suggest potential drug targets for anti-aging therapies.
Collapse
Affiliation(s)
- Dominika Drulis-Fajdasz
- Department of Molecular Physiology and Neurobiology, University of Wrocław, Sienkiewicza 21, 50-335 Wrocław, Poland; (D.D.-F.); (P.D.)
| | - Kinga Gostomska-Pampuch
- Biochemical Proteomics Group, Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; (K.G.-P.); (J.R.W.)
- Department of Biochemistry and Immunochemistry, Wrocław Medical University, Chałubińskiego 10, 50-368 Wrocław, Poland
| | - Przemysław Duda
- Department of Molecular Physiology and Neurobiology, University of Wrocław, Sienkiewicza 21, 50-335 Wrocław, Poland; (D.D.-F.); (P.D.)
| | - Jacek Roman Wiśniewski
- Biochemical Proteomics Group, Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, 82152 Martinsried, Germany; (K.G.-P.); (J.R.W.)
| | - Dariusz Rakus
- Department of Molecular Physiology and Neurobiology, University of Wrocław, Sienkiewicza 21, 50-335 Wrocław, Poland; (D.D.-F.); (P.D.)
| |
Collapse
|
13
|
Booth DG, Kozar N, Bradley S, Meijer D. Characterizing the molecular etiology of arthrogryposis multiplex congenita in patients with LGI4 mutations. Glia 2021; 69:2605-2617. [PMID: 34288120 DOI: 10.1002/glia.24061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/07/2021] [Accepted: 07/09/2021] [Indexed: 11/05/2022]
Abstract
Disruption of axon-glia interactions in the peripheral nervous system has emerged as a major cause of arthrogryposis multiplex congenita (AMC), a condition characterized by multiple congenital postural abnormalities involving the major joints. Several genes crucially important to the biology of Schwann cells have now been implicated with AMC. One such gene is LGI4 which encodes a secreted glycoprotein. LGI4 is expressed and secreted by Schwann cells and binds its receptor ADAM22 on the axonal membrane to drive myelination. Homozygous mutations in LGI4 or ADAM22 results in severe congenital hypomyelination and joint contractures in mice. Recently bi-allelic LGI4 loss of function mutations has been described in three unrelated families with severe AMC. Two individuals in a fourth, non-consanguineous family were found to be compound heterozygous for two LGI4 missense mutations. It is not known how these missense mutations affect the biology of LGI4. Here we investigated whether these missense mutations affected the secretion of the protein, its ADAM22 binding capacity, or its myelination-promoting function. We demonstrate that the mutations largely affect the progression of the mutant protein through the endomembrane system resulting in severely reduced expression. Importantly, binding to ADAM22 and myelination-promoting activity appear largely unaffected, suggesting that treatment with chemical chaperones to improve secretion of the mutant proteins might prove beneficial.
Collapse
Affiliation(s)
- Daniel G Booth
- Centre for Discovery Brain Sciences and MS Society Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Nina Kozar
- Centre for Discovery Brain Sciences and MS Society Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Stephen Bradley
- Centre for Discovery Brain Sciences and MS Society Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Dies Meijer
- Centre for Discovery Brain Sciences and MS Society Edinburgh Centre for MS Research, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
14
|
Perveen N, Ashraf W, Alqahtani F, Fawad Rasool M, Samad N, Imran I. Temporal Lobe Epilepsy: What do we understand about protein alterations? Chem Biol Drug Des 2021; 98:377-394. [PMID: 34132061 DOI: 10.1111/cbdd.13858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/22/2021] [Accepted: 04/18/2021] [Indexed: 01/19/2023]
Abstract
During neuronal diseases, neuronal proteins get disturbed due to changes in the connections of neurons. As a result, neuronal proteins get disturbed and cause epilepsy. At the genetic level, many mutations may take place in proteins like axon guidance proteins, leucine-rich glioma inactivated 1 protein, microtubular protein, pore-forming, chromatin remodeling, and chemokine proteins which may lead toward temporal lobe epilepsy. These proteins can be targeted in the future for the treatment purpose of epilepsy. Novel avenues can be developed for therapeutic interventions by these new insights.
Collapse
Affiliation(s)
- Nadia Perveen
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Waseem Ashraf
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Muhammad Fawad Rasool
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Noreen Samad
- Department of Biochemistry, Faculty of Science, Bahauddin Zakariya University, Multan, Pakistan
| | - Imran Imran
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| |
Collapse
|
15
|
Catignas KK, Frick LR, Pellegatta M, Hurley E, Kolb Z, Addabbo K, McCarty JH, Hynes RO, van der Flier A, Poitelon Y, Wrabetz L, Feltri ML. α V integrins in Schwann cells promote attachment to axons, but are dispensable in vivo. Glia 2021; 69:91-108. [PMID: 32744761 PMCID: PMC8491627 DOI: 10.1002/glia.23886] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 12/22/2022]
Abstract
In the developing peripheral nervous system, Schwann cells (SCs) extend their processes to contact, sort, and myelinate axons. The mechanisms that contribute to the interaction between SCs and axons are just beginning to be elucidated. Using a SC-neuron coculture system, we demonstrate that Arg-Gly-Asp (RGD) peptides that inhibit αV -containing integrins delay the extension of SCs elongating on axons. αV integrins in SC localize to sites of contact with axons and are expressed early in development during radial sorting and myelination. Short interfering RNA-mediated knockdown of the αV integrin subunit also delays SC extension along axons in vitro, suggesting that αV -containing integrins participate in axo-glial interactions. However, mice lacking the αV subunit in SCs, alone or in combination with the potentially compensating α5 subunit, or the αV partners β3 or β8 , myelinate normally during development and remyelinate normally after nerve crush, indicating that overlapping or compensatory mechanisms may hide the in vivo role of RGD-binding integrins.
Collapse
Affiliation(s)
- Kathleen K. Catignas
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York
- Department of Biochemistry, University at Buffalo, Buffalo, New York
| | - Luciana R. Frick
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - Marta Pellegatta
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York
- IRCCS San Raffaele Scientific Institute and Vita Salute San Raffaele University, Milan, Italy
| | - Edward Hurley
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - Zachary Kolb
- Department of Biochemistry, University at Buffalo, Buffalo, New York
| | - Kathryn Addabbo
- Department of Biochemistry, University at Buffalo, Buffalo, New York
| | - Joseph H. McCarty
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Richard O. Hynes
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Boston, Massachusetts
| | - Arjan van der Flier
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Boston, Massachusetts
- Sanofi, Boston, Massachusetts
| | - Yannick Poitelon
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York
- Department of Biochemistry, University at Buffalo, Buffalo, New York
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York
| | - Lawrence Wrabetz
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York
- Department of Biochemistry, University at Buffalo, Buffalo, New York
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - Maria Laura Feltri
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York
- Department of Biochemistry, University at Buffalo, Buffalo, New York
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| |
Collapse
|
16
|
Assembly and Function of the Juxtaparanodal Kv1 Complex in Health and Disease. Life (Basel) 2020; 11:life11010008. [PMID: 33374190 PMCID: PMC7824554 DOI: 10.3390/life11010008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/21/2020] [Accepted: 12/23/2020] [Indexed: 02/07/2023] Open
Abstract
The precise axonal distribution of specific potassium channels is known to secure the shape and frequency of action potentials in myelinated fibers. The low-threshold voltage-gated Kv1 channels located at the axon initial segment have a significant influence on spike initiation and waveform. Their role remains partially understood at the juxtaparanodes where they are trapped under the compact myelin bordering the nodes of Ranvier in physiological conditions. However, the exposure of Kv1 channels in de- or dys-myelinating neuropathy results in alteration of saltatory conduction. Moreover, cell adhesion molecules associated with the Kv1 complex, including Caspr2, Contactin2, and LGI1, are target antigens in autoimmune diseases associated with hyperexcitability such as encephalitis, neuromyotonia, or neuropathic pain. The clustering of Kv1.1/Kv1.2 channels at the axon initial segment and juxtaparanodes is based on interactions with cell adhesion molecules and cytoskeletal linkers. This review will focus on the trafficking and assembly of the axonal Kv1 complex in the peripheral and central nervous system (PNS and CNS), during development, and in health and disease.
Collapse
|
17
|
Muppirala AN, Limbach LE, Bradford EF, Petersen SC. Schwann cell development: From neural crest to myelin sheath. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2020; 10:e398. [PMID: 33145925 DOI: 10.1002/wdev.398] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/16/2022]
Abstract
Vertebrate nervous system function requires glial cells, including myelinating glia that insulate axons and provide trophic support that allows for efficient signal propagation by neurons. In vertebrate peripheral nervous systems, neural crest-derived glial cells known as Schwann cells (SCs) generate myelin by encompassing and iteratively wrapping membrane around single axon segments. SC gliogenesis and neurogenesis are intimately linked and governed by a complex molecular environment that shapes their developmental trajectory. Changes in this external milieu drive developing SCs through a series of distinct morphological and transcriptional stages from the neural crest to a variety of glial derivatives, including the myelinating sublineage. Cues originate from the extracellular matrix, adjacent axons, and the developing SC basal lamina to trigger intracellular signaling cascades and gene expression changes that specify stages and transitions in SC development. Here, we integrate the findings from in vitro neuron-glia co-culture experiments with in vivo studies investigating SC development, particularly in zebrafish and mouse, to highlight critical factors that specify SC fate. Ultimately, we connect classic biochemical and mutant studies with modern genetic and visualization tools that have elucidated the dynamics of SC development. This article is categorized under: Signaling Pathways > Cell Fate Signaling Nervous System Development > Vertebrates: Regional Development.
Collapse
Affiliation(s)
- Anoohya N Muppirala
- Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neuroscience, Kenyon College, Gambier, Ohio, USA
| | | | | | - Sarah C Petersen
- Department of Neuroscience, Kenyon College, Gambier, Ohio, USA.,Department of Biology, Kenyon College, Gambier, Ohio, USA
| |
Collapse
|
18
|
Wilson ER, Della-Flora Nunes G, Weaver MR, Frick LR, Feltri ML. Schwann cell interactions during the development of the peripheral nervous system. Dev Neurobiol 2020; 81:464-489. [PMID: 32281247 DOI: 10.1002/dneu.22744] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/14/2020] [Accepted: 04/06/2020] [Indexed: 12/21/2022]
Abstract
Schwann cells play a critical role in the development of the peripheral nervous system (PNS), establishing important relationships both with the extracellular milieu and other cell types, particularly neurons. In this review, we discuss various Schwann cell interactions integral to the proper establishment, spatial arrangement, and function of the PNS. We include signals that cascade onto Schwann cells from axons and from the extracellular matrix, bidirectional signals that help to establish the axo-glial relationship and how Schwann cells in turn support the axon. Further, we speculate on how Schwann cell interactions with other components of the developing PNS ultimately promote the complete construction of the peripheral nerve.
Collapse
Affiliation(s)
- Emma R Wilson
- Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, NY, USA.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Gustavo Della-Flora Nunes
- Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, NY, USA.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Michael R Weaver
- Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, NY, USA.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Luciana R Frick
- Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, NY, USA.,Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - M Laura Feltri
- Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, NY, USA.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA.,Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
19
|
Mishra S, Rai A, Srivastava P, Phadke SR. A mild phenotype of LGI4-Related arthrogryposis multiplex congenita with intrafamilial variability. Eur J Med Genet 2020; 63:103756. [DOI: 10.1016/j.ejmg.2019.103756] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 07/10/2019] [Accepted: 09/08/2019] [Indexed: 01/22/2023]
|
20
|
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
|
21
|
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
|
22
|
Xie YJ, Zhou L, Wang Y, Jiang NW, Cao S, Shao CY, Wang XT, Li XY, Shen Y, Zhou L. Leucine-Rich Glioma Inactivated 1 Promotes Oligodendrocyte Differentiation and Myelination via TSC-mTOR Signaling. Front Mol Neurosci 2018; 11:231. [PMID: 30034322 PMCID: PMC6043672 DOI: 10.3389/fnmol.2018.00231] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 06/12/2018] [Indexed: 12/26/2022] Open
Abstract
Leucine-rich glioma inactivated 1 (Lgi1), a putative tumor suppressor, is tightly associated with autosomal dominant lateral temporal lobe epilepsy (ADLTE). It has been shown that Lgi1 regulates the myelination of Schwann cells in the peripheral nervous system (PNS). However, the function and underlying mechanisms for Lgi1 regulation of oligodendrocyte differentiation and myelination in the central nervous system (CNS) remain elusive. In addition, whether Lgi1 is required for myelin maintenance is unknown. Here, we show that Lgi1 is necessary and sufficient for the differentiation of oligodendrocyte precursor cells and is also required for the maintenance of myelinated fibers. The hypomyelination in Lgi1-/- mice attributes to the inhibition of the biosynthesis of lipids and proteins in oligodendrocytes (OLs). Moreover, we found that Lgi1 deficiency leads to a decrease in expression of tuberous sclerosis complex 1 (TSC1) and activates mammalian target of rapamycin signaling. Together, the present work establishes that Lgi1 is a regulator of oligodendrocyte development and myelination in CNS.
Collapse
Affiliation(s)
- Ya-Jun Xie
- Key Laboratory of Medical Neurobiology of Ministry of Health, Department of Neurobiology, Zhejiang University School of MedicineHangzhou, China
| | - Lin Zhou
- Key Laboratory of Medical Neurobiology of Ministry of Health, Department of Neurobiology, Zhejiang University School of MedicineHangzhou, China
| | - Yin Wang
- Laboratory of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical UniversityYinchuan, China
| | - Nan-Wei Jiang
- Ningbo Key Laboratory of Behavioral Neuroscience, Department of Physiology and Pharmacology, Ningbo University School of MedicineNingbo, China
| | - Shenglong Cao
- Department of Neurosurgery, Second Affiliated Hospital of Zhejiang University School of MedicineHangzhou, China
| | - Chong-Yu Shao
- Key Laboratory of Medical Neurobiology of Ministry of Health, Department of Neurobiology, Zhejiang University School of MedicineHangzhou, China
| | - Xin-Tai Wang
- Key Laboratory of Medical Neurobiology of Ministry of Health, Department of Neurobiology, Zhejiang University School of MedicineHangzhou, China
| | - Xiang-Yao Li
- Key Laboratory of Medical Neurobiology of Ministry of Health, Department of Neurobiology, Zhejiang University School of MedicineHangzhou, China
| | - Ying Shen
- Key Laboratory of Medical Neurobiology of Ministry of Health, Department of Neurobiology, Zhejiang University School of MedicineHangzhou, China
| | - Liang Zhou
- Key Laboratory of Medical Neurobiology of Ministry of Health, Department of Neurobiology, Zhejiang University School of MedicineHangzhou, China
| |
Collapse
|
23
|
Structural basis of epilepsy-related ligand-receptor complex LGI1-ADAM22. Nat Commun 2018; 9:1546. [PMID: 29670100 PMCID: PMC5906670 DOI: 10.1038/s41467-018-03947-w] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 03/23/2018] [Indexed: 01/17/2023] Open
Abstract
Epilepsy is a common brain disorder throughout history. Epilepsy-related ligand–receptor complex, LGI1–ADAM22, regulates synaptic transmission and has emerged as a determinant of brain excitability, as their mutations and acquired LGI1 autoantibodies cause epileptic disorders in human. Here, we report the crystal structure of human LGI1–ADAM22 complex, revealing a 2:2 heterotetrameric assembly. The hydrophobic pocket of the C-terminal epitempin-repeat (EPTP) domain of LGI1 binds to the metalloprotease-like domain of ADAM22. The N-terminal leucine-rich repeat and EPTP domains of LGI1 mediate the intermolecular LGI1–LGI1 interaction. A pathogenic R474Q mutation of LGI1, which does not exceptionally affect either the secretion or the ADAM22 binding, is located in the LGI1–LGI1 interface and disrupts the higher-order assembly of the LGI1–ADAM22 complex in vitro and in a mouse model for familial epilepsy. These studies support the notion that the LGI1–ADAM22 complex functions as the trans-synaptic machinery for precise synaptic transmission. LGI1 is an epilepsy-related gene that encodes a secreted neuronal protein. Here the authors present the crystal structure of LGI1 bound to its receptor ADAM22, which provides structural insights into epilepsy-causing LGI1 mutations and might facilitate the development of novel anti-epilepsy drugs.
Collapse
|
24
|
Loss-of-Function Mutations in LGI4, a Secreted Ligand Involved in Schwann Cell Myelination, Are Responsible for Arthrogryposis Multiplex Congenita. Am J Hum Genet 2017; 100:659-665. [PMID: 28318499 DOI: 10.1016/j.ajhg.2017.02.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 02/16/2017] [Indexed: 01/26/2023] Open
Abstract
Arthrogryposis multiplex congenita (AMC) is a developmental condition characterized by multiple joint contractures resulting from reduced or absent fetal movements. Through genetic mapping of disease loci and whole-exome sequencing in four unrelated multiplex families presenting with severe AMC, we identified biallelic loss-of-function mutations in LGI4 (leucine-rich glioma-inactivated 4). LGI4 is a ligand secreted by Schwann cells that regulates peripheral nerve myelination via its cognate receptor ADAM22 expressed by neurons. Immunolabeling experiments and transmission electron microscopy of the sciatic nerve from one of the affected individuals revealed a lack of myelin. Functional tests using affected individual-derived iPSCs showed that these germline mutations caused aberrant splicing of the endogenous LGI4 transcript and in a cell-based assay impaired the secretion of truncated LGI4 protein. This is consistent with previous studies reporting arthrogryposis in Lgi4-deficient mice due to peripheral hypomyelination. This study adds to the recent reports implicating defective axoglial function as a key cause of AMC.
Collapse
|
25
|
Fukata Y, Yokoi N, Miyazaki Y, Fukata M. The LGI1–ADAM22 protein complex in synaptic transmission and synaptic disorders. Neurosci Res 2017; 116:39-45. [DOI: 10.1016/j.neures.2016.09.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 09/18/2016] [Accepted: 09/22/2016] [Indexed: 12/21/2022]
|
26
|
Schwann cells–axon interaction in myelination. Curr Opin Neurobiol 2016; 39:24-9. [DOI: 10.1016/j.conb.2016.03.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/15/2016] [Accepted: 03/16/2016] [Indexed: 01/01/2023]
|
27
|
Birchmeier C, Bennett DLH. Neuregulin/ErbB Signaling in Developmental Myelin Formation and Nerve Repair. Curr Top Dev Biol 2016; 116:45-64. [PMID: 26970613 DOI: 10.1016/bs.ctdb.2015.11.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Myelin is essential for rapid and accurate conduction of electrical impulses by axons in the central and peripheral nervous system (PNS). Myelin is formed in the early postnatal period, and developmental myelination in the PNS depends on axonal signals provided by Nrg1/ErbB receptors. In addition, Nrg1 is required for effective nerve repair and remyelination in adulthood. We discuss here similarities and differences in Nrg1/ErbB functions in developmental myelination and remyelination after nerve injury.
Collapse
Affiliation(s)
- Carmen Birchmeier
- Developmental Biology, Max-Delbrück-Center for Molecular Medicine, Berlin, Germany.
| | - David L H Bennett
- The Nuffield Department of Clinical Neurosciences, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom.
| |
Collapse
|
28
|
Abstract
Myelin is essential for rapid and efficient action potential propagation in vertebrates. However, the molecular mechanisms regulating myelination remain incompletely characterized. For example, even before myelination begins in the PNS, Schwann cells must radially sort axons to form 1:1 associations. Schwann cells then ensheathe and wrap axons, and establish polarized, subcellular domains, including apical and basolateral domains, paranodes, and Schmidt-Lanterman incisures. Intriguingly, polarity proteins, such as Pals1/Mpp5, are highly enriched in some of these domains, suggesting that they may regulate the polarity of Schwann cells and myelination. To test this, we generated mice with Schwann cells and oligodendrocytes that lack Pals1. During early development of the PNS, Pals1-deficient mice had impaired radial sorting of axons, delayed myelination, and reduced nerve conduction velocities. Although myelination and conduction velocities eventually recovered, polyaxonal myelination remained a prominent feature of adult Pals1-deficient nerves. Despite the enrichment of Pals1 at paranodes and incisures of control mice, nodes of Ranvier and paranodes were unaffected in Pals1-deficient mice, although we measured a significant increase in the number of incisures. As in other polarized cells, we found that Pals1 interacts with Par3 and loss of Pals1 reduced levels of Par3 in Schwann cells. In the CNS, loss of Pals1 affected neither myelination nor the establishment of polarized membrane domains. These results demonstrate that Schwann cells and oligodendrocytes use distinct mechanisms to control their polarity, and that radial sorting in the PNS is a key polarization event that requires Pals1. Significance statement: This paper reveals the role of the canonical polarity protein Pals1 in radial sorting of axons by Schwann cells. Radial sorting is essential for efficient and proper myelination and is disrupted in some types of congenital muscular dystrophy.
Collapse
|
29
|
Poitelon Y, Bogni S, Matafora V, Della-Flora Nunes G, Hurley E, Ghidinelli M, Katzenellenbogen BS, Taveggia C, Silvestri N, Bachi A, Sannino A, Wrabetz L, Feltri ML. Spatial mapping of juxtacrine axo-glial interactions identifies novel molecules in peripheral myelination. Nat Commun 2015; 6:8303. [PMID: 26383514 PMCID: PMC4576721 DOI: 10.1038/ncomms9303] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 08/07/2015] [Indexed: 12/13/2022] Open
Abstract
Cell–cell interactions promote juxtacrine signals in specific subcellular domains, which are difficult to capture in the complexity of the nervous system. For example, contact between axons and Schwann cells triggers signals required for radial sorting and myelination. Failure in this interaction causes dysmyelination and axonal degeneration. Despite its importance, few molecules at the axo-glial surface are known. To identify novel molecules in axo-glial interactions, we modified the ‘pseudopodia' sub-fractionation system and isolated the projections that glia extend when they receive juxtacrine signals from axons. By proteomics we identified the signalling networks present at the glial-leading edge, and novel proteins, including members of the Prohibitin family. Glial-specific deletion of Prohibitin-2 in mice impairs axo-glial interactions and myelination. We thus validate a novel method to model morphogenesis and juxtacrine signalling, provide insights into the molecular organization of the axo-glial contact, and identify a novel class of molecules in myelination. Neuron–glia interactions are critical in the nervous system, where they result in the extension of glial pseudopodia. Poitelon et al. isolate these protrusions using an in vitro assay, and, by characterising their proteomes, identify Prohibitin-2 as a regulator of myelination.
Collapse
Affiliation(s)
- Y Poitelon
- Hunter James Kelly Research Institute, Department Biochemistry, University at Buffalo, Buffalo, New York 14203, USA.,Division of Genetics and Cell Biology, San Raffaele Hospital, Milano 20132, Italy
| | - S Bogni
- Division of Genetics and Cell Biology, San Raffaele Hospital, Milano 20132, Italy
| | - V Matafora
- Division of Genetics and Cell Biology, San Raffaele Hospital, Milano 20132, Italy
| | - G Della-Flora Nunes
- Hunter James Kelly Research Institute, Department Biochemistry, University at Buffalo, Buffalo, New York 14203, USA
| | - E Hurley
- Hunter James Kelly Research Institute, Department Biochemistry, University at Buffalo, Buffalo, New York 14203, USA
| | - M Ghidinelli
- Hunter James Kelly Research Institute, Department Biochemistry, University at Buffalo, Buffalo, New York 14203, USA.,Division of Genetics and Cell Biology, San Raffaele Hospital, Milano 20132, Italy
| | - B S Katzenellenbogen
- Department of Molecular and Integrative Physiology, University of Illinois and College of Medicine, Urbana Illinois 61801, USA
| | - C Taveggia
- Division of Neuroscience, San Raffaele Hospital, Milano 20132, Italy
| | - N Silvestri
- Department of Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - A Bachi
- Division of Genetics and Cell Biology, San Raffaele Hospital, Milano 20132, Italy
| | - A Sannino
- Department of Engineering for Innovation, University of Salento, Lecce 73100, Italy
| | - L Wrabetz
- Hunter James Kelly Research Institute, Department Biochemistry, University at Buffalo, Buffalo, New York 14203, USA.,Division of Genetics and Cell Biology, San Raffaele Hospital, Milano 20132, Italy.,Department of Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| | - M L Feltri
- Hunter James Kelly Research Institute, Department Biochemistry, University at Buffalo, Buffalo, New York 14203, USA.,Division of Genetics and Cell Biology, San Raffaele Hospital, Milano 20132, Italy.,Department of Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14203, USA
| |
Collapse
|
30
|
Monk KR, Feltri ML, Taveggia C. New insights on Schwann cell development. Glia 2015; 63:1376-93. [PMID: 25921593 PMCID: PMC4470834 DOI: 10.1002/glia.22852] [Citation(s) in RCA: 179] [Impact Index Per Article: 19.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 04/13/2015] [Indexed: 12/11/2022]
Abstract
In the peripheral nervous system, Schwann cells are glial cells that are in intimate contact with axons throughout development. Schwann cells generate the insulating myelin sheath and provide vital trophic support to the neurons that they ensheathe. Schwann cell precursors arise from neural crest progenitor cells, and a highly ordered developmental sequence controls the progression of these cells to become mature myelinating or nonmyelinating Schwann cells. Here, we discuss both seminal discoveries and recent advances in our understanding of the molecular mechanisms that drive Schwann cell development and myelination with a focus on cell-cell and cell-matrix signaling events.
Collapse
Affiliation(s)
- Kelly R Monk
- Department of Developmental Biology, Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri
| | - M Laura Feltri
- Department of Biochemistry and Neurology, Hunter James Kelly Research Institute, University at Buffalo, State University of New York, Buffalo, New York
| | - Carla Taveggia
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
31
|
Abstract
Myelinated nerve fibers are essential for the rapid propagation of action potentials by saltatory conduction. They form as the result of reciprocal interactions between axons and Schwann cells. Extrinsic signals from the axon, and the extracellular matrix, drive Schwann cells to adopt a myelinating fate, whereas myelination reorganizes the axon for its role in conduction and is essential for its integrity. Here, we review our current understanding of the development, molecular organization, and function of myelinating Schwann cells. Recent findings into the extrinsic signals that drive Schwann cell myelination, their cognate receptors, and the downstream intracellular signaling pathways they activate will be described. Together, these studies provide important new insights into how these pathways converge to activate the transcriptional cascade of myelination and remodel the actin cytoskeleton that is critical for morphogenesis of the myelin sheath.
Collapse
Affiliation(s)
- James L Salzer
- Department of Neuroscience and Physiology, New York University Neuroscience Institute, New York University School of Medicine, New York, New York 10016
| |
Collapse
|
32
|
Pakozdy A, Patzl M, Zimmermann L, Jokinen TS, Glantschnigg U, Kelemen A, Hasegawa D. LGI Proteins and Epilepsy in Human and Animals. J Vet Intern Med 2015; 29:997-1005. [PMID: 26032921 PMCID: PMC4895363 DOI: 10.1111/jvim.12610] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Revised: 03/23/2015] [Accepted: 04/11/2015] [Indexed: 12/16/2022] Open
Abstract
Leucine‐rich glioma‐inactivated (LGI) protein was first thought to have a suppressor effect in the formation of some cancers. Developments in physiology and medicine made it possible to characterize the function of the LGI protein family and its crucial role in different conditions more precisely. These proteins play an important role in synaptic transmission, and dysfunction may cause hyperexcitability. Genetic mutation of LGI1was confirmed to be the cause of autosomal dominant lateral temporal lobe epilepsy in humans. The LGI2 mutation was identified in benign familial juvenile epilepsy in Lagotto Romagnolo (LR) dogs. Cats with familial spontaneous temporal lobe epilepsy have been reported, and the etiology might be associated with LGI protein family dysfunction. In addition, an autoimmune reaction against LGI1 was detected in humans and cats with limbic encephalitis. These advances prompted a review of LGI protein function and its role in different seizure disorders.
Collapse
Affiliation(s)
- A Pakozdy
- University Clinic of Small Animals, University of Veterinary Medicine, Vienna, Austria
| | - M Patzl
- Institute of Immunology, University of Veterinary Medicine, Vienna, Austria
| | - L Zimmermann
- Unit of Physiology and Biophysics, University of Veterinary Medicine, Vienna, Austria
| | - T S Jokinen
- Department of Equine and Small Animal Medicine, University of Helsinki, Helsinki, Finland
| | - U Glantschnigg
- University Clinic of Small Animals, University of Veterinary Medicine, Vienna, Austria
| | - A Kelemen
- Epilepsy Center, National Institute of Clinical Neurosciences, Budapest, Hungary
| | - D Hasegawa
- Department of Clinical Veterinary Medicine, Nippon Veterinary and Life Science University, Musashinoshi, Tokyo, Japan
| |
Collapse
|
33
|
Grigoryan T, Birchmeier W. Molecular signaling mechanisms of axon-glia communication in the peripheral nervous system. Bioessays 2015; 37:502-13. [PMID: 25707700 DOI: 10.1002/bies.201400172] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this article we discuss the molecular signaling mechanisms that coordinate interactions between Schwann cells and the neurons of the peripheral nervous system. Such interactions take place perpetually during development and in adulthood, and are critical for the homeostasis of the peripheral nervous system (PNS). Neurons provide essential signals to control Schwann cell functions, whereas Schwann cells promote neuronal survival and allow efficient transduction of action potentials. Deregulation of neuron-Schwann cell interactions often results in developmental abnormalities and diseases. Recent investigations have shown that during development, neuronally provided signals, such as Neuregulin, Jagged, and Wnt interact to fine-tune the Schwann cell lineage progression. In adult, the signal exchange between neurons and Schwann cells ensures proper nerve function and regeneration. Identification of the mechanisms of neuron-Schwann cell interactions is therefore essential for our understanding of the development, function and pathology of the peripheral nervous system as a whole.
Collapse
Affiliation(s)
- Tamara Grigoryan
- Max-Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | | |
Collapse
|
34
|
Abstract
Peripheral nerves contain large myelinated and small unmyelinated (Remak) fibers that perform different functions. The choice to myelinate or not is dictated to Schwann cells by the axon itself, based on the amount of neuregulin I-type III exposed on its membrane. Peripheral axons are more important in determining the final myelination fate than central axons, and the implications for this difference in Schwann cells and oligodendrocytes are discussed. Interestingly, this choice is reversible during pathology, accounting for the remarkable plasticity of Schwann cells, and contributing to the regenerative potential of the peripheral nervous system. Radial sorting is the process by which Schwann cells choose larger axons to myelinate during development. This crucial morphogenetic step is a prerequisite for myelination and for differentiation of Remak fibers, and is arrested in human diseases due to mutations in genes coding for extracellular matrix and linkage molecules. In this review we will summarize progresses made in the last years by a flurry of reverse genetic experiments in mice and fish. This work revealed novel molecules that control radial sorting, and contributed unexpected ideas to our understanding of the cellular and molecular mechanisms that control radial sorting of axons.
Collapse
Affiliation(s)
- M Laura Feltri
- Hunter James Kelly Research Institute, Departments of Biochemistry & Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Yannick Poitelon
- Hunter James Kelly Research Institute, Departments of Biochemistry & Neurology, School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Stefano Carlo Previtali
- Institute of Experimental Neurology (INSPE), Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
35
|
Helmbrecht MS, Soellner H, Truckenbrodt AML, Sundermeier J, Cohrs C, Hans W, de Angelis MH, Feuchtinger A, Aichler M, Fouad K, Huber AB. Loss of Npn1 from motor neurons causes postnatal deficits independent from Sema3A signaling. Dev Biol 2014; 399:2-14. [PMID: 25512301 DOI: 10.1016/j.ydbio.2014.11.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Revised: 11/24/2014] [Accepted: 11/25/2014] [Indexed: 12/20/2022]
Abstract
The correct wiring of neuronal circuits is of crucial importance for the function of the vertebrate nervous system. Guidance cues like the neuropilin receptors (Npn) and their ligands, the semaphorins (Sema) provide a tight spatiotemporal control of sensory and motor axon growth and guidance. Among this family of guidance partners the Sema3A-Npn1 interaction has been shown to be of great importance, since defective signaling leads to wiring deficits and defasciculation. For the embryonic stage these defects have been well described, however, also after birth the organism can adapt to new challenges by compensational mechanisms. Therefore, we used the mouse lines Olig2-Cre;Npn1(cond) and Npn1(Sema-) to investigate how postnatal organisms cope with the loss of Npn1 selectively from motor neurons or a systemic dysfunctional Sema3A-Npn1 signaling in the entire organism, respectively. While in Olig2-Cre(+);Npn1(cond-/-) mice clear anatomical deficits in paw posturing, bone structure, as well as muscle and nerve composition became evident, Npn1(Sema-) mutants appeared anatomically normal. Furthermore, Olig2-Cre(+);Npn1(cond) mutants revealed a dysfunctional extensor muscle innervation after single-train stimulation of the N.radial. Interestingly, these mice did not show obvious deficits in voluntary locomotion, however, skilled motor function was affected. In contrast, Npn1(Sema-) mutants were less affected in all behavioral tests and able to improve their performance over time. Our data suggest that loss of Sema3A-Npn1 signaling is not the only cause for the observed deficits in Olig2-Cre(+);Npn1(cond-/-) mice and that additional, yet unknown binding partners for Npn1 may be involved that allow Npn1(Sema-) mutants to compensate for their developmental deficits.
Collapse
Affiliation(s)
- Michaela S Helmbrecht
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany.
| | - Heidi Soellner
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Anna M L Truckenbrodt
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Julia Sundermeier
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Christian Cohrs
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany; DFG-Research Center for Regenerative Therapies Dresden, Technische Universität and Paul Langerhans Institute Dresden, German Center for Diabetes Research (DZD), Germany
| | - Wolfgang Hans
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Martin Hrabě de Angelis
- German Mouse Clinic, Institute of Experimental Genetics, Helmholtz Zentrum Muenchen, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany; Chair of Experimental Genetics, Center of Life and Food Sciences Weihenstephan, Technische Universität München, 85354 Freising-Weihenstephan, Germany; German Center for Diabetes Research (DZD), Ingostädter Landstr. 1, 85764 Neuherberg, Germany
| | - Annette Feuchtinger
- Research Unit Analytical Pathology, Insititute of Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Michaela Aichler
- Research Unit Analytical Pathology, Insititute of Pathology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany
| | - Karim Fouad
- Faculty of Rehabilitation Medicine and Centre for Neuroscience, University of Alberta, Canada
| | - Andrea B Huber
- Institute of Developmental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Ingolstaedter Landstr. 1, 85764 Neuherberg, Germany.
| |
Collapse
|
36
|
Peripheral nerve proteins as potential autoantigens in acute and chronic inflammatory demyelinating polyneuropathies. Autoimmun Rev 2014; 13:1070-8. [DOI: 10.1016/j.autrev.2014.08.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2014] [Accepted: 06/27/2014] [Indexed: 01/06/2023]
|
37
|
Kegel L, Jaegle M, Driegen S, Aunin E, Leslie K, Fukata Y, Watanabe M, Fukata M, Meijer D. Functional phylogenetic analysis of LGI proteins identifies an interaction motif crucial for myelination. Development 2014; 141:1749-56. [PMID: 24715463 DOI: 10.1242/dev.107995] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The cellular interactions that drive the formation and maintenance of the insulating myelin sheath around axons are only partially understood. Leucine-rich glioma-inactivated (LGI) proteins play important roles in nervous system development and mutations in their genes have been associated with epilepsy and amyelination. Their function involves interactions with ADAM22 and ADAM23 cell surface receptors, possibly in apposing membranes, thus attenuating cellular interactions. LGI4-ADAM22 interactions are required for axonal sorting and myelination in the developing peripheral nervous system (PNS). Functional analysis revealed that, despite their high homology and affinity for ADAM22, LGI proteins are functionally distinct. To dissect the key residues in LGI proteins required for coordinating axonal sorting and myelination in the developing PNS, we adopted a phylogenetic and computational approach and demonstrate that the mechanism of action of LGI4 depends on a cluster of three amino acids on the outer surface of the LGI4 protein, thus providing a structural basis for the mechanistic differences in LGI protein function in nervous system development and evolution.
Collapse
Affiliation(s)
- Linde Kegel
- Erasmus University Medical Center, Biomedical Sciences, Departments of Genetics and Cell Biology, Rotterdam 3015GE, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Costa ET, Barnabé GF, Li M, Dias AAM, Machado TR, Asprino PF, Cavalher FP, Ferreira EN, Del Mar Inda M, Nagai MH, Malnic B, Duarte ML, Leite KRM, de Barros ACSD, Carraro DM, Chammas R, Armelin HA, Cavenee W, Furnari F, Camargo AA. Intratumoral heterogeneity of ADAM23 promotes tumor growth and metastasis through LGI4 and nitric oxide signals. Oncogene 2014; 34:1270-9. [PMID: 24662834 DOI: 10.1038/onc.2014.70] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 01/06/2014] [Accepted: 01/14/2014] [Indexed: 12/22/2022]
Abstract
Intratumoral heterogeneity (ITH) represents an obstacle for cancer diagnosis and treatment, but little is known about its functional role in cancer progression. The A Desintegrin And Metalloproteinase 23 (ADAM23) gene is epigenetically silenced in different types of tumors, and silencing is often associated with advanced disease and metastasis. Here, we show that invasive breast tumors exhibit significant ADAM23-ITH and that this heterogeneity is critical for tumor growth and metastasis. We demonstrate that while loss of ADAM23 expression enhances invasion, it causes a severe proliferative deficiency and is not itself sufficient to trigger metastasis. Rather, we observed that, in ADAM23-heterotypic environments, ADAM23-negative cells promote tumor growth and metastasis by enhancing the proliferation and invasion of adjacent A23-positive cells through the production of LGI4 (Leucine-rich Glioma Inactivated 4) and nitric oxide (NO). Ablation of LGI4 and NO in A23-negative cells significantly attenuates A23-positive cell proliferation and invasion. Our work denotes a driving role of ADAM23-ITH during disease progression, shifting the malignant phenotype from the cellular to the tissue level. Our findings also provide insights for therapeutic intervention, enforcing the need to ascertain ITH to improve cancer diagnosis and therapy.
Collapse
Affiliation(s)
- E T Costa
- 1] Centro de Oncologia Molecular, Hospital Sírio Libanês, São Paulo, Brazil [2] Ludwig Institute for Cancer Research (LICR), São Paulo, Brazil
| | - G F Barnabé
- 1] Centro de Oncologia Molecular, Hospital Sírio Libanês, São Paulo, Brazil [2] Ludwig Institute for Cancer Research (LICR), São Paulo, Brazil
| | - M Li
- Ludwig Institute for Cancer Research (LICR), University of California, San Diego, CA, USA
| | - A A M Dias
- Departamento de Biologia Geral (ICB), Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - T R Machado
- Ludwig Institute for Cancer Research (LICR), São Paulo, Brazil
| | - P F Asprino
- 1] Centro de Oncologia Molecular, Hospital Sírio Libanês, São Paulo, Brazil [2] Ludwig Institute for Cancer Research (LICR), São Paulo, Brazil
| | - F P Cavalher
- Ludwig Institute for Cancer Research (LICR), São Paulo, Brazil
| | - E N Ferreira
- Centro Internacional de Pesquisa, Hospital AC Camargo, São Paulo, Brazil
| | - M Del Mar Inda
- Ludwig Institute for Cancer Research (LICR), University of California, San Diego, CA, USA
| | - M H Nagai
- Departamento de Bioquímica (IQ), Universidade de São Paulo, São Paulo, Brazil
| | - B Malnic
- Departamento de Bioquímica (IQ), Universidade de São Paulo, São Paulo, Brazil
| | - M L Duarte
- 1] Centro de Oncologia Molecular, Hospital Sírio Libanês, São Paulo, Brazil [2] Ludwig Institute for Cancer Research (LICR), São Paulo, Brazil
| | - K R M Leite
- Departamento de Urologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - A C S D de Barros
- Departamento de Mastologia, Hospital Sírio Libanês, São Paulo, Brazil
| | - D M Carraro
- Centro Internacional de Pesquisa, Hospital AC Camargo, São Paulo, Brazil
| | - R Chammas
- Departamento de Urologia, Faculdade de Medicina, Universidade de São Paulo, São Paulo, Brazil
| | - H A Armelin
- 1] Departamento de Bioquímica (IQ), Universidade de São Paulo, São Paulo, Brazil [2] Instituto Butantan, São Paulo, Brazil
| | - W Cavenee
- Ludwig Institute for Cancer Research (LICR), University of California, San Diego, CA, USA
| | - F Furnari
- Ludwig Institute for Cancer Research (LICR), University of California, San Diego, CA, USA
| | - A A Camargo
- 1] Centro de Oncologia Molecular, Hospital Sírio Libanês, São Paulo, Brazil [2] Ludwig Institute for Cancer Research (LICR), São Paulo, Brazil
| |
Collapse
|
39
|
Abstract
Mutations in the LGI1 gene predispose to autosomal dominant lateral temporal lobe epilepsy, a rare hereditary form with incomplete penetrance and associated with acoustic auras. LGI1 is not a structural component of an ion channel like most epilepsy-related genes, but is a secreted protein. Mutant null mice exhibit early-onset seizures, and electrophysiological analysis shows abnormal synaptic transmission. LGI1 binds to ADAM23 on the presynaptic membrane and ADAM22 on the postsynaptic membrane, further implicating it in regulating the strength of synaptic transmission. Patients with limbic encephalitis show autoantibodies against LGI1 and develop seizures, supporting a role for LGI1 in synapse transmission in the post developmental brain. LGI1, however, also seems to be involved in aspects of neurite development and dendritic pruning, suggesting an additional role in corticogenesis. LGI1 is also involved in cell movement and suppression of dendritic outgrowth in in vitro systems, possibly involving actin cytoskeleton dynamics. Expression patterns in embryonic development correspond to areas of neuronal migration. Loss of LGI1 expression also impacts on myelination of the central and peripheral nervous systems. In zebrafish embryos, knockdown of lgi1a leads to a seizure-like behavior and abnormal brain development, providing a system to study its role in early embryogenesis. Despite being implicated in a role in both synapse transmission and neuronal development, how LGI1 predisposes to epilepsy is still largely unknown. It appears, however, that LGI1 may function differently in a cell context-specific manner, implying a complex involvement in brain development and function that remains to be defined.
Collapse
Affiliation(s)
- John K Cowell
- Georgia Regents University Cancer Center, Augusta, GA, USA.
| |
Collapse
|
40
|
Abstract
Axons in the vertebrate peripheral nervous system are intimately associated with Schwann cells. Axons regulate the Schwann cell phenotype, determining whether they myelinate individual axons or ensheathe multiple, small axons in Remak bundles. Our current understanding of the axonal signals that drive Schwann cells towards these distinct morphological and phenotypic fates is briefly reviewed here. Elucidation of these signals, and the intracellular pathways they regulate, may lead to new, rational therapies for the treatment of inherited and acquired neuropathies.
Collapse
Affiliation(s)
- James L Salzer
- Department of Cell Biology, New York University School of Medicine, 550 First Avenue, New York, NY 10016, USA.
| |
Collapse
|
41
|
Abstract
The development and function of the vertebrate nervous system depend on specific interactions between different cell types. Two examples of such interactions are synaptic transmission and myelination. LGI1-4 (leucine-rich glioma inactivated proteins) play important roles in these processes. They are secreted proteins consisting of an LRR (leucine-rich repeat) domain and a so-called epilepsy-associated or EPTP (epitempin) domain. Both domains are thought to function in protein–protein interactions. The first LGI gene to be identified, LGI1, was found at a chromosomal translocation breakpoint in a glioma cell line. It was subsequently found mutated in ADLTE (autosomal dominant lateral temporal (lobe) epilepsy) also referred to as ADPEAF (autosomal dominant partial epilepsy with auditory features). LGI1 protein appears to act at synapses and antibodies against LGI1 may cause the autoimmune disorder limbic encephalitis. A similar function in synaptic remodelling has been suggested for LGI2, which is mutated in canine Benign Familial Juvenile Epilepsy. LGI4 is required for proliferation of glia in the peripheral nervous system and binds to a neuronal receptor, ADAM22, to foster ensheathment and myelination of axons by Schwann cells. Thus, LGI proteins play crucial roles in nervous system development and function and their study is highly important, both to understand their biological functions and for their therapeutic potential. Here, we review our current knowledge about this important family of proteins, and the progress made towards understanding their functions.
Collapse
|
42
|
Srinivasan R, Sun G, Keles S, Jones EA, Jang SW, Krueger C, Moran JJ, Svaren J. Genome-wide analysis of EGR2/SOX10 binding in myelinating peripheral nerve. Nucleic Acids Res 2012; 40:6449-60. [PMID: 22492709 PMCID: PMC3413122 DOI: 10.1093/nar/gks313] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 03/25/2012] [Accepted: 03/26/2012] [Indexed: 11/17/2022] Open
Abstract
Myelin is essential for the rapidity of saltatory nerve conduction, and also provides trophic support for axons to prevent axonal degeneration. Two critical determinants of myelination are SOX10 and EGR2/KROX20. SOX10 is required for specification of Schwann cells from neural crest, and is required at every stage of Schwann cell development. Egr2/Krox20 expression is activated by axonal signals in myelinating Schwann cells, and is required for cell cycle arrest and myelin formation. To elucidate the integrated function of these two transcription factors during peripheral nerve myelination, we performed in vivo ChIP-Seq analysis of myelinating peripheral nerve. Integration of these binding data with loss-of-function array data identified a range of genes regulated by these factors. In addition, although SOX10 itself regulates Egr2/Krox20 expression, leading to coordinate activation of several major myelin genes by the two factors, there is a large subset of genes that are activated independent of EGR2. Finally, the results identify a set of SOX10-dependent genes that are expressed in early Schwann cell development, but become subsequently repressed by EGR2/KROX20.
Collapse
Affiliation(s)
- Rajini Srinivasan
- Waisman Center, Department of Statistics, Department of Biostatistics and Medical Informatics, Program in Cellular and Molecular Biology and, Department of Comparative Biosciences, University of Wisconsin, Madison, WI, USA
| | - Guannan Sun
- Waisman Center, Department of Statistics, Department of Biostatistics and Medical Informatics, Program in Cellular and Molecular Biology and, Department of Comparative Biosciences, University of Wisconsin, Madison, WI, USA
| | - Sunduz Keles
- Waisman Center, Department of Statistics, Department of Biostatistics and Medical Informatics, Program in Cellular and Molecular Biology and, Department of Comparative Biosciences, University of Wisconsin, Madison, WI, USA
| | - Erin A. Jones
- Waisman Center, Department of Statistics, Department of Biostatistics and Medical Informatics, Program in Cellular and Molecular Biology and, Department of Comparative Biosciences, University of Wisconsin, Madison, WI, USA
| | - Sung-Wook Jang
- Waisman Center, Department of Statistics, Department of Biostatistics and Medical Informatics, Program in Cellular and Molecular Biology and, Department of Comparative Biosciences, University of Wisconsin, Madison, WI, USA
| | - Courtney Krueger
- Waisman Center, Department of Statistics, Department of Biostatistics and Medical Informatics, Program in Cellular and Molecular Biology and, Department of Comparative Biosciences, University of Wisconsin, Madison, WI, USA
| | - John J. Moran
- Waisman Center, Department of Statistics, Department of Biostatistics and Medical Informatics, Program in Cellular and Molecular Biology and, Department of Comparative Biosciences, University of Wisconsin, Madison, WI, USA
| | - John Svaren
- Waisman Center, Department of Statistics, Department of Biostatistics and Medical Informatics, Program in Cellular and Molecular Biology and, Department of Comparative Biosciences, University of Wisconsin, Madison, WI, USA
| |
Collapse
|
43
|
Heinen A, Tzekova N, Graffmann N, Torres KJ, Uhrberg M, Hartung HP, Küry P. Histone methyltransferase enhancer of zeste homolog 2 regulates Schwann cell differentiation. Glia 2012; 60:1696-708. [DOI: 10.1002/glia.22388] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2011] [Revised: 05/31/2012] [Accepted: 06/19/2012] [Indexed: 01/05/2023]
|
44
|
Parakalan R, Jiang B, Nimmi B, Janani M, Jayapal M, Lu J, Tay SSW, Ling EA, Dheen ST. Transcriptome analysis of amoeboid and ramified microglia isolated from the corpus callosum of rat brain. BMC Neurosci 2012; 13:64. [PMID: 22697290 PMCID: PMC3441342 DOI: 10.1186/1471-2202-13-64] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2011] [Accepted: 05/16/2012] [Indexed: 02/03/2023] Open
Abstract
Background Microglia, the resident immune cells of the central nervous system (CNS), have two distinct phenotypes in the developing brain: amoeboid form, known to be amoeboid microglial cells (AMC) and ramified form, known to be ramified microglial cells (RMC). The AMC are characterized by being proliferative, phagocytic and migratory whereas the RMC are quiescent and exhibit a slow turnover rate. The AMC transform into RMC with advancing age, and this transformation is indicative of the gradual shift in the microglial functions. Both AMC and RMC respond to CNS inflammation, and they become hypertrophic when activated by trauma, infection or neurodegenerative stimuli. The molecular mechanisms and functional significance of morphological transformation of microglia during normal development and in disease conditions is not clear. It is hypothesized that AMC and RMC are functionally regulated by a specific set of genes encoding various signaling molecules and transcription factors. Results To address this, we carried out cDNA microarray analysis using lectin-labeled AMC and RMC isolated from frozen tissue sections of the corpus callosum of 5-day and 4-week old rat brain respectively, by laser capture microdissection. The global gene expression profiles of both microglial phenotypes were compared and the differentially expressed genes in AMC and RMC were clustered based on their functional annotations. This genome wide comparative analysis identified genes that are specific to AMC and RMC. Conclusions The novel and specific molecules identified from the trancriptome explains the quiescent state functioning of microglia in its two distinct morphological states.
Collapse
Affiliation(s)
- Rangarajan Parakalan
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Blk MD10, 4 Medical Drive, Singapore, 117597, Singapore
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Delmaghani S, Aghaie A, Michalski N, Bonnet C, Weil D, Petit C. Defect in the gene encoding the EAR/EPTP domain-containing protein TSPEAR causes DFNB98 profound deafness. Hum Mol Genet 2012; 21:3835-44. [PMID: 22678063 DOI: 10.1093/hmg/dds212] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
We report a consanguineous Iranian family affected by congenital profound sensorineural deafness segregating in an autosomal recessive mode. Auditory tests implicated at least a cochlear defect in these patients. We mapped the deafness, autosomal recessive (DFNB) locus involved by linkage analysis to a 4.8 Mb region at chromosome 21q22.3-qter. Exclusion of the DFNB8/10 gene TMPRSS3, located in this chromosomal interval, led us to identify a new deafness locus, DFNB98. Whole exome sequencing allowed us to identify a homozygous frame-shifting mutation (c.1726G>T+c.1728delC) in the gene TSPEAR (thrombospondin-type laminin G domain and EAR repeats). This truncating mutation (p.V576LfsX37) impeded the secretion of the encoded protein by cells transfected with the mutated gene. Alternative splicing of TSPEAR transcripts predict two protein isoforms, 522 and 669 amino acids in length, both of which would be affected by the mutation. These isoforms are composed of a thrombospondin-type laminin G (TSP) domain followed by seven tandemly organized epilepsy-associated repeats (EARs), probably forming a β-propeller domain. Tspear is expressed in a variety of murine tissues. Only the larger Tspear transcript was found in the cochlea, and the protein was detected by immunofluorescence at the surface of the hair bundles of sensory cells. The mammalian EAR protein family includes six known members. Defects in four of them, i.e. Lgi1, Lgi2, Vlgr1 and, we show here, TSPEAR, cause disorders with auditory features: epilepsy, which can include auditory features in humans; audiogenic seizures in animals; and/or hearing impairments in humans and mice. These observations demonstrate that EAR-containing proteins are essential for the development and function of the auditory system.
Collapse
Affiliation(s)
- Sedigheh Delmaghani
- Institut Pasteur, Unité de Génétique et Physiologie de l’Audition, Paris, France
| | | | | | | | | | | |
Collapse
|
46
|
Pereira JA, Lebrun-Julien F, Suter U. Molecular mechanisms regulating myelination in the peripheral nervous system. Trends Neurosci 2011; 35:123-34. [PMID: 22192173 DOI: 10.1016/j.tins.2011.11.006] [Citation(s) in RCA: 181] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2011] [Revised: 11/17/2011] [Accepted: 11/18/2011] [Indexed: 12/21/2022]
Abstract
Glial cells and neurons are engaged in a continuous and highly regulated bidirectional dialog. A remarkable example is the control of myelination. Oligodendrocytes in the central nervous system (CNS) and Schwann cells (SCs) in the peripheral nervous system (PNS) wrap their plasma membranes around axons to organize myelinated nerve fibers that allow rapid saltatory conduction. The functionality of this system is critical, as revealed by numerous neurological diseases that result from deregulation of the system, including multiple sclerosis and peripheral neuropathies. In this review we focus on PNS myelination and present a conceptual framework that integrates crucial signaling mechanisms with basic SC biology. We will highlight signaling hubs and overarching molecular mechanisms, including genetic, epigenetic, and post-translational controls, which together regulate the interplay between SCs and axons, extracellular signals, and the transcriptional network.
Collapse
Affiliation(s)
- Jorge A Pereira
- Institute of Cell Biology, Department of Biology, Swiss Federal Institute of Technology, ETH Zürich, Zürich, Switzerland
| | | | | |
Collapse
|
47
|
Seppälä EH, Jokinen TS, Fukata M, Fukata Y, Webster MT, Karlsson EK, Kilpinen SK, Steffen F, Dietschi E, Leeb T, Eklund R, Zhao X, Rilstone JJ, Lindblad-Toh K, Minassian BA, Lohi H. LGI2 truncation causes a remitting focal epilepsy in dogs. PLoS Genet 2011; 7:e1002194. [PMID: 21829378 PMCID: PMC3145619 DOI: 10.1371/journal.pgen.1002194] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2010] [Accepted: 06/05/2011] [Indexed: 12/17/2022] Open
Abstract
One quadrillion synapses are laid in the first two years of postnatal construction of the human brain, which are then pruned until age 10 to 500 trillion synapses composing the final network. Genetic epilepsies are the most common neurological diseases with onset during pruning, affecting 0.5% of 2–10-year-old children, and these epilepsies are often characterized by spontaneous remission. We previously described a remitting epilepsy in the Lagotto romagnolo canine breed. Here, we identify the gene defect and affected neurochemical pathway. We reconstructed a large Lagotto pedigree of around 34 affected animals. Using genome-wide association in 11 discordant sib-pairs from this pedigree, we mapped the disease locus to a 1.7 Mb region of homozygosity in chromosome 3 where we identified a protein-truncating mutation in the Lgi2 gene, a homologue of the human epilepsy gene LGI1. We show that LGI2, like LGI1, is neuronally secreted and acts on metalloproteinase-lacking members of the ADAM family of neuronal receptors, which function in synapse remodeling, and that LGI2 truncation, like LGI1 truncations, prevents secretion and ADAM interaction. The resulting epilepsy onsets at around seven weeks (equivalent to human two years), and remits by four months (human eight years), versus onset after age eight in the majority of human patients with LGI1 mutations. Finally, we show that Lgi2 is expressed highly in the immediate post-natal period until halfway through pruning, unlike Lgi1, which is expressed in the latter part of pruning and beyond. LGI2 acts at least in part through the same ADAM receptors as LGI1, but earlier, ensuring electrical stability (absence of epilepsy) during pruning years, preceding this same function performed by LGI1 in later years. LGI2 should be considered a candidate gene for common remitting childhood epilepsies, and LGI2-to-LGI1 transition for mechanisms of childhood epilepsy remission. Major remodeling of the neuronal synaptic network occurs during childhood. The quadrillion synapses formed till the end of age two are trimmed to 500 trillion by age 10 through a selective process of strengthening of ideal connections, removal of redundant ones, and formation of new contacts. Very little is known about the basic mechanisms that direct this massive reorganization that leads to the adult brain. The most common epilepsies of humans occur in childhood and are characterized by remission prior to adulthood. Not much is known about their genetics and basic remission mechanisms. We describe here a canine equivalent disease and identify the defective gene, Lgi2. We show that the gene product is a secreted protein and interacts with neuronal ADAM receptors known to be involved in the regulation of synaptic remodeling in the developing brain. Our work sheds important light on the basic mechanisms of the most common neurological disease of children and discloses processes of epilepsy remission. The identification of the first focal epilepsy gene in dogs has also enabled the development of a genetic test to identify carriers for breeding purposes.
Collapse
Affiliation(s)
- Eija H. Seppälä
- Department of Veterinary Biosciences, Department of Medical Genetics, Research Programs Unit, Molecular Medicine, University of Helsinki, Helsinki, Finland
- Department of Molecular Genetics, Folkhälsan Research Center, Helsinki, Finland
| | - Tarja S. Jokinen
- Department of Clinical Veterinary Sciences, University of Helsinki, Helsinki, Finland
| | - Masaki Fukata
- Division of Membrane Physiology, Department of Cell Physiology, National Institute for Physiological Sciences and National Institutes of Natural Sciences, Okazaki, Japan
| | - Yuko Fukata
- Division of Membrane Physiology, Department of Cell Physiology, National Institute for Physiological Sciences and National Institutes of Natural Sciences, Okazaki, Japan
- PRESTO, Japan Science and Technology Agency, Tokyo, Japan
| | - Matthew T. Webster
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Elinor K. Karlsson
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States of America
- FAS Center for Systems Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Sami K. Kilpinen
- Institute for Molecular Medicine Finland (FIMM), University of Helsinki, Helsinki, Finland
| | - Frank Steffen
- Department for Small Animals, Neurology Services, University of Zurich, Zurich, Switzerland
| | - Elisabeth Dietschi
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bern, Switzerland
- Department of Clinical Veterinary Medicine, Division of Small Animal Orthopedics and Surgery, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Tosso Leeb
- Institute of Genetics, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Ranja Eklund
- Department of Veterinary Biosciences, Department of Medical Genetics, Research Programs Unit, Molecular Medicine, University of Helsinki, Helsinki, Finland
- Department of Molecular Genetics, Folkhälsan Research Center, Helsinki, Finland
| | - Xiaochu Zhao
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Jennifer J. Rilstone
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Kerstin Lindblad-Toh
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts, United States of America
| | - Berge A. Minassian
- Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- Division of Neurology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Canada
| | - Hannes Lohi
- Department of Veterinary Biosciences, Department of Medical Genetics, Research Programs Unit, Molecular Medicine, University of Helsinki, Helsinki, Finland
- Department of Molecular Genetics, Folkhälsan Research Center, Helsinki, Finland
- * E-mail:
| |
Collapse
|
48
|
de Wit J, Hong W, Luo L, Ghosh A. Role of leucine-rich repeat proteins in the development and function of neural circuits. Annu Rev Cell Dev Biol 2011; 27:697-729. [PMID: 21740233 DOI: 10.1146/annurev-cellbio-092910-154111] [Citation(s) in RCA: 107] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The nervous system consists of an ensemble of billions of neurons interconnected in a highly specific pattern that allows proper propagation and integration of neural activities. The organization of these specific connections emerges from sequential developmental events including axon guidance, target selection, and synapse formation. These events critically rely on cell-cell recognition and communication mediated by cell-surface ligands and receptors. Recent studies have uncovered central roles for leucine-rich repeat (LRR) domain-containing proteins, not only in organizing neural connectivity from axon guidance to target selection to synapse formation, but also in various nervous system disorders. Their versatile LRR domains, in particular, serve as key sites for interactions with a wide diversity of binding partners. Here, we focus on a few exquisite examples of secreted or membrane-associated LRR proteins in Drosophila and mammals and review the mechanisms by which they regulate diverse aspects of nervous system development and function.
Collapse
Affiliation(s)
- Joris de Wit
- Neurobiology Section, Division of Biology, University of California, San Diego, La Jolla, California 92093-0366, USA
| | | | | | | |
Collapse
|
49
|
Monk KR, Oshima K, Jörs S, Heller S, Talbot WS. Gpr126 is essential for peripheral nerve development and myelination in mammals. Development 2011; 138:2673-80. [PMID: 21613327 DOI: 10.1242/dev.062224] [Citation(s) in RCA: 160] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In peripheral nerves, Schwann cells form the myelin sheath that insulates axons and allows rapid propagation of action potentials. Although a number of regulators of Schwann cell development are known, the signaling pathways that control myelination are incompletely understood. In this study, we show that Gpr126 is essential for myelination and other aspects of peripheral nerve development in mammals. A mutation in Gpr126 causes a severe congenital hypomyelinating peripheral neuropathy in mice, and expression of differentiated Schwann cell markers, including Pou3f1, Egr2, myelin protein zero and myelin basic protein, is reduced. Ultrastructural studies of Gpr126-/- mice showed that axonal sorting by Schwann cells is delayed, Remak bundles (non-myelinating Schwann cells associated with small caliber axons) are not observed, and Schwann cells are ultimately arrested at the promyelinating stage. Additionally, ectopic perineurial fibroblasts form aberrant fascicles throughout the endoneurium of the mutant sciatic nerve. This analysis shows that Gpr126 is required for Schwann cell myelination in mammals, and defines new roles for Gpr126 in axonal sorting, formation of mature non-myelinating Schwann cells and organization of the perineurium.
Collapse
Affiliation(s)
- Kelly R Monk
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|
50
|
Li H, Yang H, Liu Y, Huan W, Zhang S, Wu G, Lu Q, Wang Q, Wang Y. The cyclin-dependent kinase inhibitor p27(Kip1) is a positive regulator of Schwann cell differentiation in vitro. J Mol Neurosci 2011; 45:277-83. [PMID: 21484444 DOI: 10.1007/s12031-011-9518-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Accepted: 03/24/2011] [Indexed: 10/18/2022]
Abstract
Schwann cell precursors differentiating into a myelinating phenotype are critical for peripheral nerve development and regeneration. However, little is known about the underlying molecular mechanisms of Schwann cell differentiation. In the present study, we performed a cyclic adenosine monophosphate-induced Schwann cell differentiation model in vitro. Western blot analysis showed that p27(Kip1) expression was upregulated during the differentiation of Schwann cell, while the inhibition of p27(Kip1) expression by short hairpin RNA-mediated knockdown significantly abolished the expression of promyelinating markers and the alteration of cellular morphology. In addition, immunofluorescence revealed a decrease of p27(Kip1) nuclear staining and a concomitant increase of cytoplasmic staining in differentiated Schwann cells. In summary, our data indicated that p27(Kip1) was a positive regulator of Schwann cell differentiation in vitro.
Collapse
Affiliation(s)
- Honghui Li
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong 22600, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|