1
|
Matsumoto M, Matsushita K, Hane M, Wen C, Kurematsu C, Ota H, Bang Nguyen H, Quynh Thai T, Herranz-Pérez V, Sawada M, Fujimoto K, García-Verdugo JM, Kimura KD, Seki T, Sato C, Ohno N, Sawamoto K. Neuraminidase inhibition promotes the collective migration of neurons and recovery of brain function. EMBO Mol Med 2024; 16:1228-1253. [PMID: 38789599 PMCID: PMC11178813 DOI: 10.1038/s44321-024-00073-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/26/2024] Open
Abstract
In the injured brain, new neurons produced from endogenous neural stem cells form chains and migrate to injured areas and contribute to the regeneration of lost neurons. However, this endogenous regenerative capacity of the brain has not yet been leveraged for the treatment of brain injury. Here, we show that in healthy brain chains of migrating new neurons maintain unexpectedly large non-adherent areas between neighboring cells, allowing for efficient migration. In instances of brain injury, neuraminidase reduces polysialic acid levels, which negatively regulates adhesion, leading to increased cell-cell adhesion and reduced migration efficiency. The administration of zanamivir, a neuraminidase inhibitor used for influenza treatment, promotes neuronal migration toward damaged regions, fosters neuronal regeneration, and facilitates functional recovery. Together, these findings shed light on a new mechanism governing efficient neuronal migration in the adult brain under physiological conditions, pinpoint the disruption of this mechanism during brain injury, and propose a promising therapeutic avenue for brain injury through drug repositioning.
Collapse
Affiliation(s)
- Mami Matsumoto
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
- Division of Neural Development and Regeneration, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan
| | - Katsuyoshi Matsushita
- Department of Mathematical and Life Sciences, Hiroshima University, Higashi-Hiroshima, 739-8526, Japan
| | - Masaya Hane
- Bioscience and Biotechnology Center, Graduate School of Bioagricultural Sciences, and Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan
| | - Chentao Wen
- Graduate School of Science, Nagoya City University, Nagoya, 467-8501, Japan
- Laboratory for Developmental Dynamics, RIKEN Center for Biosystems Dynamics Research, Kobe, 650-0047, Japan
| | - Chihiro Kurematsu
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
| | - Haruko Ota
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
- Department of Anesthesiology and Intensive Care Medicine, Graduate School of Medical Sciences, Nagoya City University, Nagoya, 467-8601, Japan
| | - Huy Bang Nguyen
- Section of Electron Microscopy, Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki, 444-8787, Japan
- Department of Anatomy, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City (UMP), Ho Chi Minh City, 70000, Vietnam
| | - Truc Quynh Thai
- Section of Electron Microscopy, Supportive Center for Brain Research, National Institute for Physiological Sciences, Okazaki, 444-8787, Japan
- Department of Histology-Embryology-Genetics, Faculty of Basic Medical Sciences, Pham Ngoc Thach University of Medicine, Ho Chi Minh City, 70000, Vietnam
| | - Vicente Herranz-Pérez
- Laboratory of Comparative Neurobiology, Cavanilles Institute, University of Valencia, CIBERNED-ISCIII, Valencia, 46980, Spain
- Department of Cell Biology, Functional Biology and Physical Anthropology, University of Valencia, Burjassot, 46100, Spain
| | - Masato Sawada
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan
- Division of Neural Development and Regeneration, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan
| | - Koichi Fujimoto
- Department of Mathematical and Life Sciences, Hiroshima University, Higashi-Hiroshima, 739-8526, Japan
| | - José Manuel García-Verdugo
- Laboratory of Comparative Neurobiology, Cavanilles Institute, University of Valencia, CIBERNED-ISCIII, Valencia, 46980, Spain
| | - Koutarou D Kimura
- Graduate School of Science, Nagoya City University, Nagoya, 467-8501, Japan
| | - Tatsunori Seki
- Department of Histology and Neuroanatomy, Tokyo Medical University, Tokyo, 160-8402, Japan
- Department of Anatomy and Life Structure, Juntendo University Graduate School of Medicine, Tokyo, 160-8402, Japan
| | - Chihiro Sato
- Bioscience and Biotechnology Center, Graduate School of Bioagricultural Sciences, and Institute for Glyco-core Research (iGCORE), Nagoya University, Nagoya, 464-8601, Japan
| | - Nobuhiko Ohno
- Department of Anatomy, Division of Histology and Cell Biology, Jichi Medical University, Shimotsuke, 329-0498, Japan
- Division of Ultrastructural Research, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan
| | - Kazunobu Sawamoto
- Department of Developmental and Regenerative Neurobiology, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, 467-8601, Japan.
- Division of Neural Development and Regeneration, National Institute for Physiological Sciences, Okazaki, 444-8585, Japan.
| |
Collapse
|
2
|
Khullar S, Huang X, Ramesh R, Svaren J, Wang D. NetREm: Network Regression Embeddings reveal cell-type transcription factor coordination for gene regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.25.563769. [PMID: 37961577 PMCID: PMC10634989 DOI: 10.1101/2023.10.25.563769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Transcription factor (TF) coordination plays a key role in target gene (TG) regulation via protein-protein interactions (PPIs) and DNA co-binding to regulatory elements. Single-cell technologies facilitate gene expression measurement for individual cells and cell-type identification, yet the connection between TF coordination and TG regulation of various cell types remains unclear. To address this, we have developed a novel computational approach, Network Regression Embeddings (NetREm), to reveal cell-type TF-TF coordination activities for TG regulation. NetREm leverages network-constrained regularization using prior knowledge of direct and/or indirect PPIs among TFs to analyze single-cell gene expression data. We test NetREm by simulation data and benchmark its performance in 4 real-world applications that have gold standard TF-TG networks available: mouse (mESCs) and simulated human (hESCs) embryonic stem (ESCs), human hematopoietic stem (HSCs), and mouse dendritic (mDCs) cells. Further, we use NetREm to prioritize valid novel TF-TF coordination links in human Peripheral Blood Mononuclear cell (PBMC) sub-types. We apply NetREm to analyze various cell types in both central (CNS) and peripheral (PNS) nerve system (NS) (e.g. neuronal, glial, Schwann cells (SCs)) as well as in Alzheimers disease (AD). Our findings uncover cell-type coordinating TFs and identify new TF-TG candidate links. We validate our top predictions using Cut&Run and knockout loss-of-function expression data in rat/mouse models and compare results with additional functional genomic data, including expression quantitative trait loci (eQTL) and Genome-Wide Association Studies (GWAS) to link genetic variants (single nucleotide polymorphisms (SNPs)) to TF coordination.
Collapse
|
3
|
Klein D, Groh J, Yuan X, Berve K, Stassart R, Fledrich R, Martini R. Early targeting of endoneurial macrophages alleviates the neuropathy and affects abnormal Schwann cell differentiation in a mouse model of Charcot-Marie-Tooth 1A. Glia 2022; 70:1100-1116. [PMID: 35188681 DOI: 10.1002/glia.24158] [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: 12/10/2021] [Revised: 01/26/2022] [Accepted: 01/31/2022] [Indexed: 12/11/2022]
Abstract
We have previously shown that targeting endoneurial macrophages with the orally applied CSF-1 receptor specific kinase (c-FMS) inhibitor PLX5622 from the age of 3 months onwards led to a substantial alleviation of the neuropathy in mouse models of Charcot-Marie-Tooth (CMT) 1X and 1B disease, which are genetically-mediated nerve disorders not treatable in humans. The same approach failed in a model of CMT1A (PMP22-overexpressing mice, line C61), representing the most frequent form of CMT. This was unexpected since previous studies identified macrophages contributing to disease severity in the same CMT1A model. Here we re-approached the possibility of alleviating the neuropathy in a model of CMT1A by targeting macrophages at earlier time points. As a proof-of-principle experiment, we genetically inactivated colony-stimulating factor-1 (CSF-1) in CMT1A mice, which resulted in lower endoneurial macrophage numbers and alleviated the neuropathy. Based on these observations, we pharmacologically ablated macrophages in newborn CMT1A mice by feeding their lactating mothers with chow containing PLX5622, followed by treatment of the respective progenies after weaning until the age of 6 months. We found that peripheral neuropathy was substantially alleviated after early postnatal treatment, leading to preserved motor function in CMT1A mice. Moreover, macrophage depletion affected the altered Schwann cell differentiation phenotype. These findings underscore the targetable role of macrophage-mediated inflammation in peripheral nerves of inherited neuropathies, but also emphasize the need for an early treatment start confined to a narrow therapeutic time window in CMT1A models and potentially in respective patients.
Collapse
Affiliation(s)
- Dennis Klein
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Janos Groh
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Xidi Yuan
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Kristina Berve
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Ruth Stassart
- Paul-Flechsig-Institute of Neuropathology, University Clinic Leipzig, Leipzig, Germany
| | - Robert Fledrich
- Institute of Anatomy, University of Leipzig, Leipzig, Germany
| | - Rudolf Martini
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
4
|
Castillo G, Kleene R, Schachner M, Loers G, Torda AE. Proteins Binding to the Carbohydrate HNK-1: Common Origins? Int J Mol Sci 2021; 22:ijms22158116. [PMID: 34360882 PMCID: PMC8347730 DOI: 10.3390/ijms22158116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/15/2021] [Accepted: 07/19/2021] [Indexed: 11/25/2022] Open
Abstract
The human natural killer (HNK-1) carbohydrate plays important roles during nervous system development, regeneration after trauma and synaptic plasticity. Four proteins have been identified as receptors for HNK-1: the laminin adhesion molecule, high-mobility group box 1 and 2 (also called amphoterin) and cadherin 2 (also called N-cadherin). Because of HNK-1′s importance, we asked whether additional receptors for HNK-1 exist and whether the four identified proteins share any similarity in their primary structures. A set of 40,000 sequences homologous to the known HNK-1 receptors was selected and used for large-scale sequence alignments and motif searches. Although there are conserved regions and highly conserved sites within each of these protein families, there was no sequence similarity or conserved sequence motifs found to be shared by all families. Since HNK-1 receptors have not been compared regarding binding constants and since it is not known whether the sulfated or non-sulfated part of HKN-1 represents the structurally crucial ligand, the receptors are more heterogeneous in primary structure than anticipated, possibly involving different receptor or ligand regions. We thus conclude that the primary protein structure may not be the sole determinant for a bona fide HNK-1 receptor, rendering receptor structure more complex than originally assumed.
Collapse
Affiliation(s)
- Gaston Castillo
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany; (G.C.); (R.K.)
| | - Ralf Kleene
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany; (G.C.); (R.K.)
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience, Department of Cell Biology and Neuroscience, Rutgers University, 604 Allison Road, Piscataway, NJ 08854, USA;
| | - Gabriele Loers
- Center for Molecular Neurobiology, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany; (G.C.); (R.K.)
- Correspondence: (G.L.); (A.E.T.); Tel.: +49-40741056292 (G.L.); +49-40428387331 (A.E.T.)
| | - Andrew E. Torda
- Centre for Bioinformatics, University of Hamburg, Bundesstr. 43, 20146 Hamburg, Germany
- Correspondence: (G.L.); (A.E.T.); Tel.: +49-40741056292 (G.L.); +49-40428387331 (A.E.T.)
| |
Collapse
|
5
|
Schmitt-Ulms G, Mehrabian M, Williams D, Ehsani S. The IDIP framework for assessing protein function and its application to the prion protein. Biol Rev Camb Philos Soc 2021; 96:1907-1932. [PMID: 33960099 DOI: 10.1111/brv.12731] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 04/22/2021] [Accepted: 04/26/2021] [Indexed: 01/06/2023]
Abstract
The quest to determine the function of a protein can represent a profound challenge. Although this task is the mandate of countless research groups, a general framework for how it can be approached is conspicuously lacking. Moreover, even expectations for when the function of a protein can be considered to be 'known' are not well defined. In this review, we begin by introducing concepts pertinent to the challenge of protein function assignments. We then propose a framework for inferring a protein's function from four data categories: 'inheritance', 'distribution', 'interactions' and 'phenotypes' (IDIP). We document that the functions of proteins emerge at the intersection of inferences drawn from these data categories and emphasise the benefit of considering them in an evolutionary context. We then apply this approach to the cellular prion protein (PrPC ), well known for its central role in prion diseases, whose function continues to be considered elusive by many investigators. We document that available data converge on the conclusion that the function of the prion protein is to control a critical post-translational modification of the neural cell adhesion molecule in the context of epithelial-to-mesenchymal transition and related plasticity programmes. Finally, we argue that this proposed function of PrPC has already passed the test of time and is concordant with the IDIP framework in a way that other functions considered for this protein fail to achieve. We anticipate that the IDIP framework and the concepts analysed herein will aid the investigation of other proteins whose primary functional assignments have thus far been intractable.
Collapse
Affiliation(s)
- Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, M5T 0S8, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | | | - Declan Williams
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, M5T 0S8, Canada
| | - Sepehr Ehsani
- Theoretical and Philosophical Biology, Department of Philosophy, University College London, Bloomsbury, London, WC1E 6BT, U.K.,Ronin Institute for Independent Scholarship, Montclair, NJ, 07043, U.S.A
| |
Collapse
|
6
|
Gerber D, Pereira JA, Gerber J, Tan G, Dimitrieva S, Yángüez E, Suter U. Transcriptional profiling of mouse peripheral nerves to the single-cell level to build a sciatic nerve ATlas (SNAT). eLife 2021; 10:e58591. [PMID: 33890853 PMCID: PMC8064760 DOI: 10.7554/elife.58591] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
Peripheral nerves are organ-like structures containing diverse cell types to optimize function. This interactive assembly includes mostly axon-associated Schwann cells, but also endothelial cells of supporting blood vessels, immune system-associated cells, barrier-forming cells of the perineurium surrounding and protecting nerve fascicles, and connective tissue-resident cells within the intra-fascicular endoneurium and inter-fascicular epineurium. We have established transcriptional profiles of mouse sciatic nerve-inhabitant cells to foster the fundamental understanding of peripheral nerves. To achieve this goal, we have combined bulk RNA sequencing of developing sciatic nerves up to the adult with focused bulk and single-cell RNA sequencing of Schwann cells throughout postnatal development, extended by single-cell transcriptome analysis of the full sciatic nerve both perinatally and in the adult. The results were merged in the transcriptome resource Sciatic Nerve ATlas (SNAT: https://www.snat.ethz.ch). We anticipate that insights gained from our multi-layered analysis will serve as valuable interactive reference point to guide future studies.
Collapse
Affiliation(s)
- Daniel Gerber
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Jorge A Pereira
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Joanne Gerber
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Ge Tan
- Functional Genomics Center Zurich, ETH Zurich/University of ZurichZurichSwitzerland
| | - Slavica Dimitrieva
- Functional Genomics Center Zurich, ETH Zurich/University of ZurichZurichSwitzerland
| | - Emilio Yángüez
- Functional Genomics Center Zurich, ETH Zurich/University of ZurichZurichSwitzerland
| | - Ueli Suter
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| |
Collapse
|
7
|
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: 41] [Impact Index Per Article: 8.2] [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
|
8
|
Gregory LC, Shah P, Sanner JRF, Arancibia M, Hurst J, Jones WD, Spoudeas H, Le Quesne Stabej P, Williams HJ, Ocaka LA, Loureiro C, Martinez-Aguayo A, Dattani MT. Mutations in MAGEL2 and L1CAM Are Associated With Congenital Hypopituitarism and Arthrogryposis. J Clin Endocrinol Metab 2019; 104:5737-5750. [PMID: 31504653 PMCID: PMC6916815 DOI: 10.1210/jc.2019-00631] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 07/18/2019] [Indexed: 12/29/2022]
Abstract
CONTEXT Congenital hypopituitarism (CH) is rarely observed in combination with severe joint contractures (arthrogryposis). Schaaf-Yang syndrome (SHFYNG) phenotypically overlaps with Prader-Willi syndrome, with patients also manifesting arthrogryposis. L1 syndrome, a group of X-linked disorders that include hydrocephalus and lower limb spasticity, also rarely presents with arthrogryposis. OBJECTIVE We investigated the molecular basis underlying the combination of CH and arthrogryposis in five patients. PATIENTS The heterozygous p.Q666fs*47 mutation in the maternally imprinted MAGEL2 gene, previously described in multiple patients with SHFYNG, was identified in patients 1 to 4, all of whom manifested growth hormone deficiency and variable SHFYNG features, including dysmorphism, developmental delay, sleep apnea, and visual problems. Nonidentical twins (patients 2 and 3) had diabetes insipidus and macrocephaly, and patient 4 presented with ACTH insufficiency. The hemizygous L1CAM variant p.G452R, previously implicated in patients with L1 syndrome, was identified in patient 5, who presented with antenatal hydrocephalus. RESULTS Human embryonic expression analysis revealed MAGEL2 transcripts in the developing hypothalamus and ventral diencephalon at Carnegie stages (CSs) 19, 20, and 23 and in the Rathke pouch at CS20 and CS23. L1CAM was expressed in the developing hypothalamus, ventral diencephalon, and hindbrain (CS19, CS20, CS23), but not in the Rathke pouch. CONCLUSION We report MAGEL2 and L1CAM mutations in four pedigrees with variable CH and arthrogryposis. Patients presenting early in life with this combined phenotype should be examined for features of SHFYNG and/or L1 syndrome. This study highlights the association of hypothalamo-pituitary disease with MAGEL2 and L1CAM mutations.
Collapse
Affiliation(s)
- Louise C Gregory
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Pratik Shah
- Great Ormond Street Hospital, London, United Kingdom
| | | | - Monica Arancibia
- Division de Pediatria, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Jane Hurst
- NE Thames Genetics Service, Great Ormond Street Hospital, London, United Kingdom
| | - Wendy D Jones
- NE Thames Genetics Service, Great Ormond Street Hospital, London, United Kingdom
| | | | - Polona Le Quesne Stabej
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Hywel J Williams
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Louise A Ocaka
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Carolina Loureiro
- Division de Pediatria, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Alejandro Martinez-Aguayo
- Division de Pediatria, Escuela de Medicina, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Mehul T Dattani
- Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- Great Ormond Street Hospital, London, United Kingdom
- Correspondence and Reprint Requests: Mehul T. Dattani, MD, Paediatric Endocrinology, Genetics and Genomic Medicine Programme, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London WC1N 1EH, United Kingdom. E-mail:
| |
Collapse
|
9
|
Endo T, Kadoya K, Kawamura D, Iwasaki N. Evidence for cell-contact factor involvement in neurite outgrowth of dorsal root ganglion neurons stimulated by Schwann cells. Exp Physiol 2019; 104:1447-1454. [PMID: 31294871 DOI: 10.1113/ep087634] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 07/08/2019] [Indexed: 12/14/2022]
Abstract
NEW FINDINGS What is the central question of this study? Although the factors secreted from Schwann cells that promote axonal growth in the peripheral nervous system have been well studied, the effect of cell-contact factors on Schwann cells remains to be determined. What is the main finding and its importance? This study demonstrates that Schwann cells stimulate neurite outgrowth by direct contact with neurites and by secreting factors. Notably, the effect of cell-contact factors in neurite outgrowth is comparable to that of secreted factors, indicating that the identification of cell surface molecules on Schwann cells that promote neurite outgrowth could lead to development of a new therapy for peripheral nervous system injury. ABSTRACT Schwann cells (SCs) play a variety of roles in the regeneration process after injury to the peripheral nervous system. The factors secreted from SCs that promote axonal growth have been well studied. However, the involvement of cell-contact factors on SCs remains to be determined. Here, we demonstrate a significant contribution of a cell-contact mechanism in the effect of SCs on promotion of neuronal outgrowth. Neurite outgrowth of adult sensory neurons from dorsal root ganglia was quantified during co-culture with adult SCs. Direct contact of SCs with neurons was eliminated by culturing SCs on an insert placed in the same well; this resulted in a 51% reduction in the length of neurite outgrowth. In addition, when dorsal root ganglion neurons were cultured on sparsely seeded SCs, neurons that made contact with SCs on their neurites had 118% longer neurites than neurons that lacked contacts with SCs. Collectively, these findings provide evidence that SCs stimulate neurite outgrowth via direct contact with neurites in addition to secreting factors. The identification of cell surface molecules on SCs that promote neurite outgrowth could lead to development of a new therapy for peripheral nervous system injury.
Collapse
Affiliation(s)
- Takeshi Endo
- Department of Orthopaedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Ken Kadoya
- Department of Orthopaedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Daisuke Kawamura
- Department of Orthopaedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| | - Norimasa Iwasaki
- Department of Orthopaedic Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Hokkaido, Japan
| |
Collapse
|
10
|
Sahu S, Li R, Kadeyala PK, Liu S, Schachner M. The human natural killer-1 (HNK-1) glycan mimetic ursolic acid promotes functional recovery after spinal cord injury in mouse. J Nutr Biochem 2018; 55:219-228. [PMID: 29567576 DOI: 10.1016/j.jnutbio.2018.01.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 11/17/2017] [Accepted: 01/19/2018] [Indexed: 02/05/2023]
Abstract
Human natural killer-1 (HNK-1) cell antigen is a glycan epitope involved in several neural events, such as neuritogenesis, myelination, synaptic plasticity and regeneration of the nervous system after injury. We have recently identified the small organic compound ursolic acid (UA) as a HNK-1 mimetic with the aim to test its therapeutic potential in the central nervous system. UA, a plant-derived pentacyclic triterpenoid, is well known for its multiple biological functions, including neuroprotective, antioxidant and anti-inflammatory activities. In the present study, we evaluated its functions in a mouse model of spinal cord injury (SCI) and explored the molecular mechanisms underlying its positive effects. Oral administration of UA to mice 1 h after SCI and thereafter once daily for 6 weeks enhanced the regaining of motor functions and axonal regrowth, and decreased astrogliosis. UA administration decreased levels of proinflammatory markers, including interleukin-6 and tumor necrosis factor-α, in the injured spinal cord at the acute phase of inflammation and activated the mitogen-activated protein kinase and phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin pathways in the injured spinal cord. Taken together, these results suggest that UA may be a candidate for treatment of nervous system injuries.
Collapse
Affiliation(s)
- Sudhanshu Sahu
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Rong Li
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Praveen Kumar Kadeyala
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Shisong Liu
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong 515041, China; Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ 08554, USA.
| |
Collapse
|
11
|
Inaguma S, Wang Z, Lasota JP, Miettinen MM. Expression of neural cell adhesion molecule L1 (CD171) in neuroectodermal and other tumors: An immunohistochemical study of 5155 tumors and critical evaluation of CD171 prognostic value in gastrointestinal stromal tumors. Oncotarget 2018; 7:55276-55289. [PMID: 27419370 PMCID: PMC5338914 DOI: 10.18632/oncotarget.10527] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/26/2016] [Indexed: 12/13/2022] Open
Abstract
The neural cell adhesion molecule L1 (CD171) is a multidomain type 1 membrane glycoprotein of the immunoglobulin superfamily important in the nervous system development, kidney morphogenesis, and maintenance of the immune system. Recent studies reported CD171 expression being associated with adverse clinical outcome in different types of cancer and there has been a growing interest in targeting this cell membrane molecule on neoplastic cells by chimeric antigen receptor redirected T lymphocytes or specific antibodies. Nevertheless, conflicting results regarding the prognostic value of CD171 expression in renal cell carcinomas and gastrointestinal stromal tumors were published. In this study, CD171 expression was immunohistochemically analyzed in 5155 epithelial, mesenchymal, melanocytic, and lymphohematopoietic tumors to assess its utility in diagnostic pathology and to pinpoint potential targets for CD171-targeting therapy. A newly developed anti-CD171 rabbit monoclonal antibody, clone 014, was selected from the panel of commercially available CD171 antibodies. Immunohistochemistry was performed using Leica Bond Max automation and multitumor blocks containing up to 60 tumor samples. CD171 was constitutively and strongly expressed in neuroectodermal tumors such as schwannoma, neuroblastoma, and paraganglioma, whereas other mesenchymal tumors including schwannoma mimics showed only rarely CD171 positivity. Frequent CD171-expression was also detected in ovarian serous carcinoma, malignant mesothelioma, and testicular embryonal carcinoma. CD171 immunohistochemistry may have some role in immunophenotypic differential diagnosis of neurogenic tumors and pinpointing potential candidates for anti-CD171 therapy. Though, because of its rare expression and lack of predictive value, CD171 is neither a diagnostic nor prognostic marker for gastrointestinal stromal tumors.
Collapse
Affiliation(s)
- Shingo Inaguma
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA.,Department of Pathology, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Zengfeng Wang
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | - Jerzy P Lasota
- Laboratory of Pathology, National Cancer Institute, Bethesda, MD, USA
| | | |
Collapse
|
12
|
Höftberger R, Lassmann H. Inflammatory demyelinating diseases of the central nervous system. HANDBOOK OF CLINICAL NEUROLOGY 2018; 145:263-283. [PMID: 28987175 PMCID: PMC7149979 DOI: 10.1016/b978-0-12-802395-2.00019-5] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Inflammatory demyelinating diseases are a heterogeneous group of disorders, which occur against the background of an acute or chronic inflammatory process. The pathologic hallmark of multiple sclerosis (MS) is the presence of focal demyelinated lesions with partial axonal preservation and reactive astrogliosis. Demyelinated plaques are present in the white as well as gray matter, such as the cerebral or cerebellar cortex and brainstem nuclei. Activity of the disease process is reflected by the presence of lesions with ongoing myelin destruction. Axonal and neuronal destruction in the lesions is a major substrate for permanent neurologic deficit in MS patients. The MS pathology is qualitatively similar in different disease stages, such as relapsing remitting MS or secondary or primary progressive MS, but the prevalence of different lesion types differs quantitatively. Acute MS and Balo's type of concentric sclerosis appear to be variants of classic MS. In contrast, neuromyelitis optica (NMO) and spectrum disorders (NMOSD) are inflammatory diseases with primary injury of astrocytes, mediated by aquaporin-4 antibodies. Finally, we discuss the histopathology of other inflammatory demyelinating diseases such as acute disseminated encephalomyelitis and myelin oligodendrocyte glycoprotein antibody-associated demyelination. Knowledge of the heterogenous immunopathology in demyelinating diseases is important, to understand the clinical presentation and disease course and to find the optimal treatment for an individual patient.
Collapse
Affiliation(s)
- Romana Höftberger
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Vienna, Austria,Correspondence to: Hans Lassmann, MD, Center for Brain Research, Medical University of Vienna, Spitalgasse, 1090 Vienna, Austria
| |
Collapse
|
13
|
Peschl P, Bradl M, Höftberger R, Berger T, Reindl M. Myelin Oligodendrocyte Glycoprotein: Deciphering a Target in Inflammatory Demyelinating Diseases. Front Immunol 2017; 8:529. [PMID: 28533781 PMCID: PMC5420591 DOI: 10.3389/fimmu.2017.00529] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 04/19/2017] [Indexed: 12/23/2022] Open
Abstract
Myelin oligodendrocyte glycoprotein (MOG), a member of the immunoglobulin (Ig) superfamily, is a myelin protein solely expressed at the outermost surface of myelin sheaths and oligodendrocyte membranes. This makes MOG a potential target of cellular and humoral immune responses in inflammatory demyelinating diseases. Due to its late postnatal developmental expression, MOG is an important marker for oligodendrocyte maturation. Discovered about 30 years ago, it is one of the best-studied autoantigens for experimental autoimmune models for multiple sclerosis (MS). Human studies, however, have yielded controversial results on the role of MOG, especially MOG antibodies (Abs), as a biomarker in MS. But with improved detection methods using different expression systems to detect Abs in patients' samples, this is meanwhile no longer the case. Using cell-based assays with recombinant full-length, conformationally intact MOG, several recent studies have revealed that MOG Abs can be found in a subset of predominantly pediatric patients with acute disseminated encephalomyelitis (ADEM), aquaporin-4 (AQP4) seronegative neuromyelitis optica spectrum disorders (NMOSD), monophasic or recurrent isolated optic neuritis (ON), or transverse myelitis, in atypical MS and in N-methyl-d-aspartate receptor-encephalitis with overlapping demyelinating syndromes. Whereas MOG Abs are only transiently observed in monophasic diseases such as ADEM and their decline is associated with a favorable outcome, they are persistent in multiphasic ADEM, NMOSD, recurrent ON, or myelitis. Due to distinct clinical features within these diseases it is controversially disputed to classify MOG Ab-positive cases as a new disease entity. Neuropathologically, the presence of MOG Abs is characterized by MS-typical demyelination and oligodendrocyte pathology associated with Abs and complement. However, it remains unclear whether MOG Abs are a mere inflammatory bystander effect or truly pathogenetic. This article provides deeper insight into recent developments, the clinical relevance of MOG Abs and their role in the immunpathogenesis of inflammatory demyelinating disorders.
Collapse
Affiliation(s)
- Patrick Peschl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Monika Bradl
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Romana Höftberger
- Institute of Neurology, Medical University of Vienna, Vienna, Austria
| | - Thomas Berger
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Markus Reindl
- Clinical Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
14
|
McFerrin J, Patton BL, Sunderhaus ER, Kretzschmar D. NTE/PNPLA6 is expressed in mature Schwann cells and is required for glial ensheathment of Remak fibers. Glia 2017; 65:804-816. [PMID: 28206686 PMCID: PMC5357176 DOI: 10.1002/glia.23127] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 01/26/2017] [Accepted: 01/26/2017] [Indexed: 11/08/2022]
Abstract
Neuropathy target esterase (NTE) or patatin-like phospholipase domain containing 6 (PNPLA6) was first linked with a neuropathy occurring after organophosphate poisoning and was later also found to cause complex syndromes when mutated, which can include mental retardation, spastic paraplegia, ataxia, and blindness. NTE/PNPLA6 is widely expressed in neurons but experiments with its Drosophila orthologue Swiss-cheese (SWS) suggested that it may also have glial functions. Investigating whether NTE/PNPLA6 is expressed in glia, we found that NTE/PNPLA6 is expressed by Schwann cells in the sciatic nerve of adult mice with the most prominent expression in nonmyelinating Schwann cells. Within Schwann cells, NTE/PNPLA6 is enriched at the Schmidt-Lanterman incisures and around the nucleus. When analyzing postnatal expression patterns, we did not detect NTE/PNPLA6 in promyelinating Schwann cells, while weak expression was detectable at postnatal day 5 in Schwann cells and increased with their maturation. Interestingly, NTE/PNPLA6 levels were upregulated after nerve crush and localized to ovoids forming along the nerve fibers. Using a GFAP-based knock-out of NTE/PNPLA6, we detected an incomplete ensheathment of Remak fibers whereas myelination did not appear to be affected. These results suggest that NTE/PNPLA6 is involved in the maturation of nonmyelinating Schwann cells during development and de-/remyelination after neuronal injury. Since Schwann cells play an important role in maintaining axonal viability and function, it is therefore likely that changes in Schwann cells contribute to the locomotory deficits and neuropathy observed in patients carrying mutations in NTE.
Collapse
Affiliation(s)
- Janis McFerrin
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Bruce L. Patton
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Elizabeth R. Sunderhaus
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
- Molecular and Medical Genetics, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Doris Kretzschmar
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
- Molecular and Medical Genetics, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| |
Collapse
|
15
|
Moyon S, Casaccia P. DNA methylation in oligodendroglial cells during developmental myelination and in disease. NEUROGENESIS 2017; 4:e1270381. [PMID: 28203606 DOI: 10.1080/23262133.2016.1270381] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 11/23/2016] [Accepted: 12/01/2016] [Indexed: 10/20/2022]
Abstract
Oligodendrocyte progenitor cells (OPC) are the myelinating cells of the central nervous system (CNS). During development, they differentiate into mature oligodendrocytes (OL) and ensheath axons, providing trophic and functional support to the neurons. This process is regulated by the dynamic expression of specific transcription factors, which, in turn, is controlled by epigenetic marks such as DNA methylation. Here we discuss recent findings showing that DNA methylation levels are differentially regulated in the oligodendrocyte lineage during developmental myelination, affecting both genes expression and alternative splicing events. Based on the phenotypic characterization of mice with genetic ablation of DNA methyltransferase 1 (Dnmt1) we conclude that DNA methylation is critical for efficient OPC expansion and for developmental myelination. Previous work suggests that in the context of diseases such as multiple sclerosis (MS) or gliomas, DNA methylation is differentially regulated in the CNS of affected individuals compared with healthy controls. In this commentary, based on the results of previous work, we propose the potential role of DNA methylation in adult oligodendroglial lineage cells in physiologic and pathological conditions, and delineate potential research approaches to be undertaken to test this hypothesis. A better understanding of this epigenetic modification in adult oligodendrocyte progenitor cells is essential, as it can potentially result in the design of new therapeutic strategies to enhance remyelination in MS patients or reduce proliferation in glioma patients.
Collapse
Affiliation(s)
- Sarah Moyon
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai , New York, NY, USA
| | - Patrizia Casaccia
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Neuroscience Initiative Advanced Science Research Center, CUNY, New York, NY, USA
| |
Collapse
|
16
|
Mehrabian M, Hildebrandt H, Schmitt-Ulms G. NCAM1 Polysialylation: The Prion Protein's Elusive Reason for Being? ASN Neuro 2016; 8:8/6/1759091416679074. [PMID: 27879349 PMCID: PMC5122176 DOI: 10.1177/1759091416679074] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 09/08/2016] [Accepted: 10/02/2016] [Indexed: 01/06/2023] Open
Abstract
Much confusion surrounds the physiological function of the cellular prion protein (PrPC). It is, however, anticipated that knowledge of its function will shed light on its contribution to neurodegenerative diseases and suggest ways to interfere with the cellular toxicity central to them. Consequently, efforts to elucidate its function have been all but exhaustive. Building on earlier work that uncovered the evolutionary descent of the prion founder gene from an ancestral ZIP zinc transporter, we recently investigated a possible role of PrPC in a morphogenetic program referred to as epithelial-to-mesenchymal transition (EMT). By capitalizing on PrPC knockout cell clones in a mammalian cell model of EMT and using a comparative proteomics discovery strategy, neural cell adhesion molecule-1 emerged as a protein whose upregulation during EMT was perturbed in PrPC knockout cells. Follow-up work led us to observe that PrPC regulates the polysialylation of the neural cell adhesion molecule NCAM1 in cells undergoing morphogenetic reprogramming. In addition to governing cellular migration, polysialylation modulates several other cellular plasticity programs PrPC has been phenotypically linked to. These include neurogenesis in the subventricular zone, controlled mossy fiber sprouting and trimming in the hippocampal formation, hematopoietic stem cell renewal, myelin repair and maintenance, integrity of the circadian rhythm, and glutamatergic signaling. This review revisits this body of literature and attempts to present it in light of this novel contextual framework. When approached in this manner, a coherent model of PrPC acting as a regulator of polysialylation during specific cell and tissue morphogenesis events comes into focus.
Collapse
Affiliation(s)
- Mohadeseh Mehrabian
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Herbert Hildebrandt
- Institute for Cellular Chemistry, Hannover Medical School, Hannover, Germany
| | - Gerold Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Ontario, Canada .,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
17
|
Abstract
A variety of anatomical features suggest that functional activity in the nervous system can influence the process of myelination, yet direct evidence of this is lacking. Research by Zalc and colleagues shows that myelination of optic nerve is inhibited by a neurotoxin that blocks action potential activity and is stimulated by a toxin that increases impulse activity, suggesting that impulse activity is necessary for initiating myelination during development of the optic nerve. Research by Fields and colleagues, using electrical stimulation of axons, shows that low frequency impulse activity inhibits myelination of dorsal root ganglion neurons, but high frequency impulse activity has no effect. This results from reduced expression of a cell adhesion molecule on the stimulated axons that is critical for inducing myelination. Together these studies support the conclusion that impulse activity can influence the process of myelination, probably through more than one molecular mechanism operating during discrete steps in the myelination process.
Collapse
Affiliation(s)
- Bernard Zalc
- Biologie des Interactions Neurones-Glie, Institut National de la Santé et de la Recherche Médicale U-495, Université P. M. Curie, Hôpital de la Salpêtrière, Paris, France (BZ), Laboratory of Developmental Neurobiology, National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland (RDF)
| | | |
Collapse
|
18
|
Jennings AR, Carroll WM. Oligodendrocyte Lineage Cells in Chronic Demyelination of Multiple Sclerosis Optic Nerve. Brain Pathol 2014; 25:517-30. [PMID: 25175564 PMCID: PMC8028859 DOI: 10.1111/bpa.12193] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 08/26/2014] [Indexed: 11/27/2022] Open
Abstract
Reports that chronically demyelinated multiple sclerosis brain and spinal cord lesions contained immature oligodendrocyte lineage cells have generated major interest aimed at the potential for promotion of endogenous repair. Despite the prominence of the optic nerve as a lesion site and its importance in clinical disease assessment, no detailed studies of multiple sclerosis‐affected optic nerve exist. This study aims to provide insight into the cellular pathology of chronic demyelination in multiple sclerosis through direct morphological and immunohistochemical analysis of optic nerve in conjunction with observations from an experimental cat optic nerve model of successful remyelination. Myelin staining was followed by immunohistochemistry to differentially label neuroglia. Digitally immortalized sections were then analyzed to generate quantification data and antigenic phenotypes including maturational stages within the oligodendrocyte lineage. It was found that some chronically demyelinated multiple sclerosis optic nerve lesions contained oligodendroglial cells and that heterogeneity existed in the presence of myelin sheaths, oligodendrocyte maturational stages and extent of axonal investment. The findings advance our understanding of oligodendrocyte activity in chronically demyelinated human optic nerve and may have implications for studies aimed at enhancement of endogenous repair in multiple sclerosis.
Collapse
Affiliation(s)
- Alison Ruth Jennings
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, WA, Australia
| | - William M Carroll
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, WA, Australia.,Department of Neurology, Sir Charles Gairdner Hospital, Nedlands, WA, Australia
| |
Collapse
|
19
|
Leinders M, Koehrn FJ, Bartok B, Boyle DL, Shubayev V, Kalcheva I, Yu NK, Park J, Kaang BK, Hefferan MP, Firestein GS, Sorkin LS. Differential distribution of PI3K isoforms in spinal cord and dorsal root ganglia: potential roles in acute inflammatory pain. Pain 2014; 155:1150-1160. [PMID: 24631588 PMCID: PMC4128246 DOI: 10.1016/j.pain.2014.03.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2013] [Revised: 03/03/2014] [Accepted: 03/05/2014] [Indexed: 11/30/2022]
Abstract
PI3-kinases (PI3Ks) participate in nociception within spinal cord, dorsal root ganglion (DRG), and peripheral nerves. To extend our knowledge, we immunohistochemically stained for each of the 4 class I PI3K isoforms along with several cell-specific markers within the lumbar spinal cord, DRG, and sciatic nerve of naive rats. Intrathecal and intraplantar isoform specific antagonists were given as pretreatments before intraplantar carrageenan; pain behavior was then assessed over time. The α-isoform was localized to central terminals of primary afferent fibers in spinal cord laminae IIi to IV as well as to neurons in ventral horn and DRG. The PI3Kβ isoform was the only class I isoform seen in dorsal horn neurons; it was also observed in DRG, Schwann cells, and axonal paranodes. The δ-isoform was found in spinal cord white matter oligodendrocytes and radial astrocytes, and the γ-isoform was seen in a subpopulation of IB4-positive DRG neurons. No isoform co-localized with microglial markers or satellite cells in naive tissue. Only the PI3Kβ antagonist, but none of the other antagonists, had anti-allodynic effects when administered intrathecally; coincident with reduced pain behavior, this agent completely blocked paw carrageenan-induced dorsal horn 2-amino-3-(3-hydroxy-5-methyl-isoxazol-4-yl) propanoic acid (AMPA) receptor trafficking to plasma membranes. Intraplantar administration of the γ-antagonist prominently reduced pain behavior. These data suggest that each isoform displays specificity with regard to neuronal type as well as to specific tissues. Furthermore, each PI3K isoform has a unique role in development of nociception and tissue inflammation.
Collapse
Affiliation(s)
- Mathias Leinders
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Fred J. Koehrn
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Beatrix Bartok
- Deprtment of Medicine, Division of Rheumatology, University of California, San Diego, La Jolla, CA
| | - David L. Boyle
- Deprtment of Medicine, Division of Rheumatology, University of California, San Diego, La Jolla, CA
| | - Veronica Shubayev
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA 92093, USA
- San Diego VA Healthcare System, La Jolla, CA
| | - Iveta Kalcheva
- Deprtment of Medicine, Division of Rheumatology, University of California, San Diego, La Jolla, CA
| | - Nam-Kyung Yu
- Department of Biological Sciences and Brain and Cognitive Sciences, Seoul National University, Seoul 151-747, Korea
| | - Jihye Park
- Department of Biological Sciences and Brain and Cognitive Sciences, Seoul National University, Seoul 151-747, Korea
| | - Bong-Kiun Kaang
- Department of Biological Sciences and Brain and Cognitive Sciences, Seoul National University, Seoul 151-747, Korea
| | | | - Gary S. Firestein
- Deprtment of Medicine, Division of Rheumatology, University of California, San Diego, La Jolla, CA
| | - Linda S. Sorkin
- Department of Anesthesiology, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
20
|
Klein D, Groh J, Wettmarshausen J, Martini R. Nonuniform molecular features of myelinating Schwann cells in models for CMT1: Distinct disease patterns are associated with NCAM and c-Jun upregulation. Glia 2014; 62:736-50. [DOI: 10.1002/glia.22638] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 01/21/2014] [Accepted: 01/22/2014] [Indexed: 11/06/2022]
Affiliation(s)
- Dennis Klein
- Department of Neurology; Developmental Neurobiology; University of Würzburg; Josef-Schneider-Str. 11 D-97080 Würzburg Germany
| | - Janos Groh
- Department of Neurology; Developmental Neurobiology; University of Würzburg; Josef-Schneider-Str. 11 D-97080 Würzburg Germany
| | - Jennifer Wettmarshausen
- Department of Neurology; Developmental Neurobiology; University of Würzburg; Josef-Schneider-Str. 11 D-97080 Würzburg Germany
| | - Rudolf Martini
- Department of Neurology; Developmental Neurobiology; University of Würzburg; Josef-Schneider-Str. 11 D-97080 Würzburg Germany
| |
Collapse
|
21
|
Koppes AN, Nordberg AL, Paolillo GM, Goodsell NM, Darwish HA, Zhang L, Thompson DM. Electrical stimulation of schwann cells promotes sustained increases in neurite outgrowth. Tissue Eng Part A 2014; 20:494-506. [PMID: 24063574 PMCID: PMC3926181 DOI: 10.1089/ten.tea.2013.0012] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Accepted: 09/03/2013] [Indexed: 12/20/2022] Open
Abstract
Endogenous electric fields are instructive during embryogenesis by acting to direct cell migration, and postnatally, they can promote axonal growth after injury (McCaig 1991, Al-Majed 2000). However, the mechanisms for these changes are not well understood. Application of an appropriate electrical stimulus may increase the rate and success of nerve repair by directly promoting axonal growth. Previously, DC electrical stimulation at 50 mV/mm (1 mA, 8 h duration) was shown to promote neurite outgrowth and a more pronounced effect was observed if both peripheral glia (Schwann cells) and neurons were co-stimulated. If electrical stimulation is delivered to an injury site, both the neurons and all resident non-neuronal cells [e.g., Schwann cells, endothelial cells, fibroblasts] will be treated and this biophysical stimuli can influence axonal growth directly or indirectly via changes to the resident, non-neuronal cells. In this work, non-neuronal cells were electrically stimulated, and changes in morphology and neuro-supportive cells were evaluated. Schwann cell response (morphology and orientation) was examined after an 8 h stimulation over a range of DC fields (0-200 mV/mm, DC 1 mA), and changes in orientation were observed. Electrically prestimulating Schwann cells (50 mV/mm) promoted 30% more neurite outgrowth relative to co-stimulating both Schwann cells with neurons, suggesting that electrical stimulation modifies Schwann cell phenotype. Conditioned medium from the electrically prestimulated Schwann cells promoted a 20% increase in total neurite outgrowth and was sustained for 72 h poststimulation. An 11-fold increase in nerve growth factor but not brain-derived neurotrophic factor or glial-derived growth factor was found in the electrically prestimulated Schwann cell-conditioned medium. No significant changes in fibroblast or endothelial morphology and neuro-supportive behavior were observed poststimulation. Electrical stimulation is widely used in clinical settings; however, the rational application of this cue may directly impact and enhance neuro-supportive behavior, improving nerve repair.
Collapse
Affiliation(s)
- Abigail N Koppes
- Department of Biomedical Engineering and Center for Biotechnology and Interdisciplinary Studies, Renssalaer Polytechnic Institute , Troy, New York
| | | | | | | | | | | | | |
Collapse
|
22
|
Guseva D, Loers G, Schachner M. Function-triggering antibodies to the adhesion molecule L1 enhance recovery after injury of the adult mouse femoral nerve. PLoS One 2014; 9:e112984. [PMID: 25393007 PMCID: PMC4231121 DOI: 10.1371/journal.pone.0112984] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Accepted: 10/19/2014] [Indexed: 02/05/2023] Open
Abstract
L1 is among the few adhesion molecules that favors repair after trauma in the adult central nervous system of vertebrates by promoting neuritogenesis and neuronal survival, among other beneficial features. In the peripheral nervous system, L1 is up-regulated in Schwann cells and regrowing axons after nerve damage, but the functional consequences of this expression remain unclear. Our previous study of L1-deficient mice in a femoral nerve injury model showed an unexpected improved functional recovery, attenuated motoneuronal cell death, and enhanced Schwann cell proliferation, being attributed to the persistent synthesis of neurotrophic factors. On the other hand, transgenic mice over-expressing L1 in neurons led to improved remyelination, but not improved functional recovery. The present study was undertaken to investigate whether the monoclonal L1 antibody 557 that triggers beneficial L1 functions in vitro would trigger these also in femoral nerve repair. We analyzed femoral nerve regeneration in C57BL/6J mice that received this antibody in a hydrogel filled conduit connecting the cut and sutured nerve before its bifurcation, leading to short-term release of antibody by diffusion. Video-based quantitative analysis of motor functions showed improved recovery when compared to mice treated with conduits containing PBS in the hydrogel scaffold, as a vehicle control. This improved recovery was associated with attenuated motoneuron loss, remyelination and improved precision of preferential motor reinnervation. We suggest that function-triggering L1 antibodies applied to the lesion site at the time of injury over a limited time period will not only be beneficial in peripheral, but also central nervous system regeneration.
Collapse
Affiliation(s)
- Daria Guseva
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
- Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, China
- W. M. Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
23
|
Wang Y, Loers G, Pan HC, Gouveia R, Zhao WJ, Shen YQ, Kleene R, Costa J, Schachner M. Antibody fragments directed against different portions of the human neural cell adhesion molecule L1 act as inhibitors or activators of L1 function. PLoS One 2012; 7:e52404. [PMID: 23272240 PMCID: PMC3525558 DOI: 10.1371/journal.pone.0052404] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 11/16/2012] [Indexed: 02/05/2023] Open
Abstract
The neural cell adhesion molecule L1 plays important roles in neuronal migration and survival, neuritogenesis and synaptogenesis. L1 has also been found in tumors of different origins, with levels of L1 expression correlating positively with the metastatic potential of tumors. To select antibodies targeting the varied functions of L1, we screened the Tomlinson library of recombinant human antibody fragments to identify antibodies binding to recombinant human L1 protein comprising the entire extracellular domain of human L1. We obtained four L1 binding single-chain variable fragment antibodies (scFvs), named I4, I6, I13, and I27 and showed by enzyme-linked immunosorbent assay (ELISA) that scFvs I4 and I6 have high affinity to the immunoglobulin-like (Ig) domains 1-4 of L1, while scFvs I13 and I27 bind strongly to the fibronectin type III homologous (Fn) domains 1-3 of L1. Application of scFvs I4 and I6 to human SK-N-SH neuroblastoma cells reduced proliferation and transmigration of these cells. Treatment of SK-N-SH cells with scFvs I13 and I27 enhanced cell proliferation and migration, neurite outgrowth, and protected against the toxic effects of H(2)O(2) by increasing the ratio of Bcl-2/Bax. In addition, scFvs I4 and I6 inhibited and scFvs I13 and I27 promoted phosphorylation of src and Erk. Our findings indicate that scFvs reacting with the immunoglobulin-like domains 1-4 inhibit L1 functions, whereas scFvs interacting with the fibronectin type III domains 1-3 trigger L1 functions of cultured neuroblastoma cells.
Collapse
Affiliation(s)
- Yan Wang
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Gabriele Loers
- Zentrum für Molekulare Neurobiologie Hamburg, Universität Hamburg, Hamburg, Germany
| | - Hong-Chao Pan
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Ricardo Gouveia
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Wei-Jiang Zhao
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Yan-Qin Shen
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
| | - Ralf Kleene
- Zentrum für Molekulare Neurobiologie Hamburg, Universität Hamburg, Hamburg, Germany
| | - Julia Costa
- Instituto de Tecnologia Química e Biológica, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Melitta Schachner
- Center for Neuroscience, Shantou University Medical College, Shantou, Guangdong Province, People's Republic of China
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey, United States of America
| |
Collapse
|
24
|
Kinter J, Lazzati T, Schmid D, Zeis T, Erne B, Lützelschwab R, Steck AJ, Pareyson D, Peles E, Schaeren-Wiemers N. An essential role of MAG in mediating axon-myelin attachment in Charcot-Marie-Tooth 1A disease. Neurobiol Dis 2012; 49:221-31. [PMID: 22940629 DOI: 10.1016/j.nbd.2012.08.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Revised: 07/30/2012] [Accepted: 08/16/2012] [Indexed: 10/28/2022] Open
Abstract
Charcot-Marie-Tooth disease type 1A (CMT1A) is a hereditary demyelinating peripheral neuropathy caused by the duplication of the PMP22 gene. Demyelination precedes the occurrence of clinical symptoms that correlate with axonal degeneration. It was postulated that a disturbed axon-glia interface contributes to altered myelination consequently leading to axonal degeneration. In this study, we examined the expression of MAG and Necl4, two critical adhesion molecules that are present at the axon-glia interface, in sural nerve biopsies of CMT1A patients and in peripheral nerves of mice overexpressing human PMP22, an animal model for CMT1A. We show an increase in the expression of MAG and a strong decrease of Necl4 in biopsies of CMT1A patients as well as in CMT1A mice. Expression analysis revealed that MAG is strongly upregulated during peripheral nerve maturation, whereas Necl4 expression remains very low. Ablating MAG in CMT1A mice results in separation of axons from their myelin sheath. Our data show that MAG is important for axon-glia contact in a model for CMT1A, and suggest that its increased expression in CMT1A disease has a compensatory role in the pathology of the disease. Thus, we demonstrate that MAG together with other adhesion molecules such as Necl4 is important in sustaining axonal integrity.
Collapse
Affiliation(s)
- Jochen Kinter
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.
| | - Thomas Lazzati
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.
| | - Daniela Schmid
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.
| | - Thomas Zeis
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.
| | - Beat Erne
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.
| | - Roland Lützelschwab
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland; Department of Neurology, University Hospital Basel, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.
| | - Andreas J Steck
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland; Department of Neurology, University Hospital Basel, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.
| | - Davide Pareyson
- IRCCS Foundation, C. Besta Neurological Institute, Milan, Italy.
| | - Elior Peles
- Department of Molecular Cell Biology, The Weizmann Institute of Science, POB 26, Rehovot 76100, Israel.
| | - Nicole Schaeren-Wiemers
- Neurobiology, Department of Biomedicine, University Hospital Basel, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland; Department of Neurology, University Hospital Basel, University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland.
| |
Collapse
|
25
|
Satar B, Hidir Y, Serdar MA, Kucuktag Z, Ural AU, Avcu F, Safali M, Oguztuzun S. Protein profiling of anastomosed facial nerve treated with mesenchymal stromal cells. Cytotherapy 2012; 14:522-8. [PMID: 22268520 DOI: 10.3109/14653249.2011.651530] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND AIMS The types of proteins released from mesenchymal stromal cells (MSC) are still unclear. Our aim was to compare apoptosis scores and the expression of myelin-associated glycoprotein (MAG), myelin basic protein (MBP), neural cell adhesion molecule (NCAM)-1,matrix metalloproteinase (MMP)-1A, tissue inhibitor of metalloproteinase (TIMP)-1, TIMP-1/MMP-1A ratio, nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), ciliary neurotrophic factor (CNTF), neurotrophin (NT)-3, NT-4, glial cell-derived neurotropic factor (GDNF), leukemia inhibitory factor (LIF), basic fibroblast growth factor (FGF)-2, insulin-like growth factor (IGF)-1, platelet-derived growth factor (PDGF)-α and transforming growth factor (TGF)-β1 in anastomosed facial nerves that had been treated with or without MSC. METHODS In seven rats, the buccal branch of the right facial nerve was transected, anastomosed and treated with MSC (anastomosed + MSC group). The left buccal branch was anastomosed only (anastomosed-only group). The left mandibular branch served as an intact nerve group. On days 18-20, the distal segments of the branches were examined in terms of expression of the mentioned proteins and apoptosis scores using polymerase chain reaction (PCR) and terminal deoxynucleotidyl transferase-mediated digoxigenin-UTP nick end labeling (TUNEL) assays. RESULTS MSC application significantly increased CNTF, PDGF-α, LIF, TGF-β1, BDNF and NT-3 expression (P < 0.05). MAG expression slightly decreased whereas NCAM-1, MMP-1A and FGF-2 slightly increased(P > 0.05). Changes in other proteins and apoptosis scores were not significant. CONCLUSIONS These results suggest that MSC increases expression of CNTF, PDGF-α, LIF,TGF-β1, BDNF and NT-3. MAG, NCAM-1, MMP-1A and FGF-2 expressions were slightly changed in this stage of nerve regeneration. The comparison of apoptotic activity was not conclusive. Overall, it appears that MSC might have differential effects on the mentioned tissue-related proteins and trophic/growth factors.
Collapse
Affiliation(s)
- Bulent Satar
- Department of Otolaryngology, Head and Neck Surgery, Gulhane Military Medical Academy, Ankara, Turkey.
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Carlson KB, Singh P, Feaster MM, Ramnarain A, Pavlides C, Chen ZL, Yu WM, Feltri ML, Strickland S. Mesenchymal stem cells facilitate axon sorting, myelination, and functional recovery in paralyzed mice deficient in Schwann cell-derived laminin. Glia 2011; 59:267-77. [PMID: 21125647 DOI: 10.1002/glia.21099] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Peripheral nerve function depends on a regulated process of axon and Schwann cell development. Schwann cells interact with peripheral neurons to sort and ensheath individual axons. Ablation of laminin γ1 in the peripheral nervous system (PNS) arrests Schwann cell development prior to radial sorting of axons. Peripheral nerves of laminin-deficient animals are disorganized and hypomyelinated. In this study, sciatic nerves of laminin-deficient mice were treated with syngenic murine adipose-derived stem cells (ADSCs). ADSCs expressed laminin in vitro and in vivo following transplant into mutant sciatic nerves. ADSC-treatment of mutant nerves caused endogenous Schwann cells to differentiate past the point of developmental arrest to sort and myelinate axons. This was shown by (1) functional, (2) ultrastructural, and (3) immunohistochemical analysis. Treatment of laminin-deficient nerves with either soluble laminin or the immortalized laminin-expressing cell line 3T3/L1 did not overcome endogenous Schwann cell developmental arrest. In summary, these results indicate that (1) laminin-deficient Schwann cells can be rescued, (2) a cell-based approach is beneficial in comparison with soluble protein treatment, and (3) mesenchymal stem cells modify sciatic nerve function via trophic effects rather than transdifferentiation in this system.
Collapse
Affiliation(s)
- Karen B Carlson
- Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, New York 10065, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Chang CY, Lee YH, Jiang-Shieh YF, Chien HF, Pai MH, Chen HM, Fong TH, Wu CH. Novel distribution of cluster of differentiation 200 adhesion molecule in glial cells of the peripheral nervous system of rats and its modulation after nerve injury. Neuroscience 2011; 183:32-46. [PMID: 21453758 DOI: 10.1016/j.neuroscience.2011.03.049] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2010] [Revised: 03/21/2011] [Accepted: 03/23/2011] [Indexed: 02/02/2023]
Abstract
This study examined CD200 expression in different peripheral nerves and ganglia. Intense CD200 immunoreactivity was consistently localized in unmyelinated nerve fibers as opposed to a faint immunostaining in the myelinated nerve fibers. By light microscopy, structures resembling the node of Ranvier and Schmidt-Lanterman incisures in the myelinated nerve fibers displayed CD200 immunoreactivity. Ultrastructural study revealed CD200 expression on the neurilemma of Schwann cells whose microvilli and paranodal loops at the node of Ranvier were immunoreactive. The CD200 immunoexpression was also localized in the satellite glial cells of sensory and autonomic ganglia and in the enteric glial cells. Double labeling of CD200 with specific antigens of satellite glia or Schwann cells in the primary cultures of dorsal root ganglia had shown a differential expression of CD200 in the peripheral glial cells. The existence of CD200 in glial cells in the peripheral nervous system (PNS) was corroborated by the expression of CD200 mRNA and protein in a rat Schwann cell line RSC96. Using the model of crush or transected sciatic nerve, it was found that CD200 expression was attenuated or diminished at the site of lesion. A remarkable feature, however, was an increase in incidence of CD200-labelled Schmidt-Lanterman incisures proximal to the injured site at 7 days postlesion. Because CD200 has been reported to impart immunosuppressive signal, we suggest that its localization in PNS glial cells may play a novel inhibitory role in immune homeostasis in both normal and pathological conditions.
Collapse
Affiliation(s)
- C-Y Chang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Irintchev A. Potentials and limitations of peripheral nerve injury models in rodents with particular reference to the femoral nerve. Ann Anat 2011; 193:276-85. [PMID: 21481575 DOI: 10.1016/j.aanat.2011.02.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2011] [Revised: 02/16/2011] [Accepted: 02/24/2011] [Indexed: 12/11/2022]
Abstract
Restoration of function after peripheral nerve repair in humans is unsatisfactory. Various causes of poor recovery have been proposed. Still, we do not understand which of these potential factors are indeed detrimental and do not know how to manipulate them experimentally in a clinically feasible way. Future success largely depends on methodological improvement in rodent models. An example of recent progress is the introduction of new functional and anatomical outcome measures in the facial nerve injury paradigm which led to novel insights into facial nerve regeneration and a new therapeutic concept. Less success can be ascribed to the use of the classical spinal nerve model, the sciatic nerve paradigm, not least because of its anatomical and functional complexity making assessment of recovery challenging. A simpler alternative to the sciatic nerve is the femoral nerve model. It offers, alongside with its known usefulness for studies on precision of motor reinnervation, the possibility of reliable functional assessments and a straightforward search of anatomical substrates of dysfunction. The structure-function approach in the femoral nerve paradigm has been useful for testing of novel therapeutic means and analyses of regeneration in mutant mice. The potential of the method has still not been really exploited and its more extensive use may contribute to better understanding of nerve regeneration.
Collapse
Affiliation(s)
- Andrey Irintchev
- Neuroscience Laboratory, Department of Otorhinolaryngology, Friedrich Schiller University Jena, Lessingstrasse 2, Jena, Germany.
| |
Collapse
|
29
|
Guseva D, Zerwas M, Xiao MF, Jakovcevski I, Irintchev A, Schachner M. Adhesion molecule L1 overexpressed under the control of the neuronal Thy-1 promoter improves myelination after peripheral nerve injury in adult mice. Exp Neurol 2011; 229:339-52. [PMID: 21376041 DOI: 10.1016/j.expneurol.2011.02.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Revised: 02/21/2011] [Accepted: 02/24/2011] [Indexed: 01/11/2023]
Abstract
L1 is an adhesion molecule favorably influencing the functional and anatomical recoveries after central nervous system (CNS) injuries. Its roles in peripheral nervous system (PNS) regeneration are less well understood. Studies using knockout mice have surprisingly revealed that L1 has a negative impact on functional nerve regeneration by inhibiting Schwann cell proliferation. To further elucidate the roles of L1 in PNS regeneration, here we used a novel transgenic mouse overexpressing L1 in neurons, but not in PNS or CNS glial cells, under the control of a neuron-specific Thy-1 promoter. Without nerve injury, the transgene expression, as compared to wild-type mice, had no effect on femoral nerve function, numbers of quadriceps motoneurons and myelinated axons in the femoral nerve but resulted in slightly reduced myelination in the sensory saphenous nerve and increased neurofilament density in myelinated axons of the quadriceps motor nerve branch. After femoral nerve injury, L1 overexpression had no impact on the time course and degree of functional recovery. Unaffected were also numbers of regenerated quadriceps motoneurons, precision of muscle reinnervation, axon numbers and internodal lengths in the regenerated nerves. Despite the lack of functional effects, myelination in the motor and sensory femoral nerve branches was significantly improved and loss of perisomatic inhibitory terminals on motoneurons was attenuated in the transgenic mice. Our results indicate that L1 is a regulator of myelination in the injured PNS and warrant studies aiming to improve function in demyelinating PNS and CNS disorders using exogenous L1.
Collapse
Affiliation(s)
- Daria Guseva
- Zentrum für Molekulare Neurobiologie, University Hospital Hamburg-Eppendorf, Martinistrasse 52, D-20246 Hamburg, Germany
| | | | | | | | | | | |
Collapse
|
30
|
Webb AA, Ngan S, Fowler JD. Spinal cord injury I: A synopsis of the basic science. THE CANADIAN VETERINARY JOURNAL = LA REVUE VETERINAIRE CANADIENNE 2010; 51:485-492. [PMID: 20676289 PMCID: PMC2857426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
Substantial knowledge has been gained in the pathological findings following naturally occurring spinal cord injury (SCI) in dogs and cats. The molecular mechanisms involved in failure of neural regeneration within the central nervous system, potential therapeutics including cellular transplantation therapy, neural plasticity, and prognostic indicators of recovery from SCI have been studied. This 2-part review summarizes 1) basic science perspectives regarding treating and curing spinal cord injury, 2) recent studies that shed light on prognosis and recovery from SCI, 3) current thinking regarding standards of care for dogs with SCI, 4) experimental approaches in the laboratory setting, and 5) current clinical trials being conducted in veterinary medicine. Part I presents timely information on the pathophysiology of spinal cord injury, challenges associated with promoting regeneration of neurons of the central nervous system, and experimental approaches aimed at developing treatments for spinal cord injury.
Collapse
Affiliation(s)
- Aubrey A Webb
- Hotchkiss Brain Institute, Department of Comparative Biology and Experimental Medicine, University of Calgary, 3330 Hospital Drive, NW, Calgary, Alberta T2N 4N1.
| | | | | |
Collapse
|
31
|
Chipman PH, Franz CK, Nelson A, Schachner M, Rafuse VF. Neural cell adhesion molecule is required for stability of reinnervated neuromuscular junctions. Eur J Neurosci 2010; 31:238-49. [PMID: 20074227 DOI: 10.1111/j.1460-9568.2009.07049.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Studies examining the etiology of motoneuron diseases usually focus on motoneuron death as the defining pathophysiology of the disease. However, impaired neuromuscular transmission and synapse withdrawal often precede cell death, raising the possibility that abnormalities in synaptic function contribute to disease onset. Although little is known about the mechanisms maintaining the synaptic integrity of neuromuscular junctions (NMJs), Drosophila studies suggest that Fasciclin II plays an important role. Inspired by these studies we used a reinnervation model of synaptogenesis to analyze neuromuscular function in mice lacking neural cell adhesion molecule (NCAM), the Fasciclin II vertebrate homolog. Our results showed that the recovery of contractile force was the same in wild-type and NCAM-/- mice at 1 month after nerve injury, indicating that endplates were appropriately reformed. This normality was only transient because the contractile force and myofiber number decreased at 3 months after injury in NCAM-/- mice. Both declined further 3 months later. Myofibers degenerated, not because motoneurons died but because synapses were withdrawn. Although neurotransmission was initially normal at reinnervated NCAM-/- NMJs, it was significantly compromised 3 months later. Interestingly, the selective ablation of NCAM from motoneurons, or muscle fibers, did not mimic the deficits observed in reinnervated NCAM-/- mice. Taken together, these results indicate that NCAM is required to maintain normal synaptic function at reinnervated NMJs, although its loss pre-synaptically or post-synaptically is not sufficient to induce synaptic destabilization. Consideration is given to the role of NCAM in terminal Schwann cells for maintaining synaptic integrity and how NCAM dysfunction may contribute to motoneuron disorders.
Collapse
Affiliation(s)
- Peter H Chipman
- Department of Anatomy and Neurobiology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | | | | | | | |
Collapse
|
32
|
Nielsen J, Kulahin N, Walmod PS. Extracellular protein interactions mediated by the neural cell adhesion molecule, NCAM: heterophilic interactions between NCAM and cell adhesion molecules, extracellular matrix proteins, and viruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 663:23-53. [PMID: 20017013 DOI: 10.1007/978-1-4419-1170-4_2] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Janne Nielsen
- Protein Laboratory, Department of Neuroscience and Pharmacology, University of Copenhagen, Copenhagen, Denmark
| | | | | |
Collapse
|
33
|
An integrin-contactin complex regulates CNS myelination by differential Fyn phosphorylation. J Neurosci 2009; 29:9174-85. [PMID: 19625508 DOI: 10.1523/jneurosci.5942-08.2009] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The understanding of how adhesion molecules mediate the axon-glial interactions in the CNS that ensure target-dependent survival of oligodendrocytes and initiate myelination remains incomplete. Here, we investigate how signals from adhesion molecules can be integrated to regulate these initial steps of myelination. We first demonstrate that the Ig superfamily molecule contactin is associated in oligodendrocytes with integrins, extracellular matrix receptors that regulate target-dependent survival by amplification of growth factor signaling. This amplification is inhibited by small interfering RNA-mediated knockdown of contactin in oligodendrocytes. In contrast, the presence of L1-Fc, the extracellular portion of a contactin ligand expressed on axons, enhanced survival and additionally promoted myelination in cocultures of neurons and oligodendrocytes. We further demonstrate that the signals from contactin and integrin are integrated by differential phosphorylation of the Src family kinase Fyn. Integrin induced dephosphorylation of the inhibitory Tyr-531, whereas contactin increased phosphorylation of both Tyr-531 and the activating Tyr-420. The combined effect is an enhanced activity of Fyn and also a dynamic regulation of the phosphorylation/dephosphorylation balance of Fyn, as required for normal cell adhesion and spreading. We conclude, therefore, that a novel integrin/contactin complex coordinates signals from extracellular matrix and the axonal surface to regulate both oligodendrocyte survival and myelination by controlling Fyn activity.
Collapse
|
34
|
Yu WM, Yu H, Chen ZL, Strickland S. Disruption of laminin in the peripheral nervous system impedes nonmyelinating Schwann cell development and impairs nociceptive sensory function. Glia 2009; 57:850-9. [PMID: 19053061 DOI: 10.1002/glia.20811] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The mechanisms controlling the differentiation of immature Schwann cells (SCs) into nonmyelinating SCs is not known. Laminins are extracellular matrix proteins critical for myelinating SC differentiation, but their roles in nonmyelinating SC development have not been established. Here, we show that the peripheral nerves of mutant mice with laminin-deficient SCs do not form Remak bundles, which consist of a single nonmyelinating SC interacting with multiple unmyelinated axons. These mutant nerves show aberrant L1 and neural cell adhesion molecule (N-CAM) expression pattern during development. The homophilic and heterophilic interactions of N-CAM are also impaired in the mutant nerves. Other molecular markers for nonmyelinating SCs, including Egr-1, glial fibrillary acidic protein, and AN2/NG2, are all absent in adult mutant nerves. Analysis of expression of SC lineage markers demonstrates that nonmyelinating SCs do not develop in mutant nerves. Additionally, mutant mice are insensitive to heat stimuli and show a decreased number of C-fiber sensory neurons, indicating reduced nociceptive sensory function. These results show that laminin participates in nonmyelinating SC development and Remak bundle formation and suggest a possible role for laminin deficiency in peripheral sensory neuropathies.
Collapse
Affiliation(s)
- Wei-Ming Yu
- Laboratory of Neurobiology and Genetics, The Rockefeller University, New York, NY, USA
| | | | | | | |
Collapse
|
35
|
Guseva D, Angelov DN, Irintchev A, Schachner M. Ablation of adhesion molecule L1 in mice favours Schwann cell proliferation and functional recovery after peripheral nerve injury. ACTA ACUST UNITED AC 2009; 132:2180-95. [PMID: 19541848 DOI: 10.1093/brain/awp160] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The adhesion molecule L1 is one of the few adhesion molecules known to be beneficial for repair processes in the adult central nervous system of vertebrates by promoting axonal growth and neuronal survival. In the peripheral nervous system, L1 is up-regulated by myelination-competent Schwann cells and regenerating axons after nerve damage but its functional role has remained unknown. Here we tested the hypothesis that L1 is, as in the central nervous system, beneficial for nerve regeneration in the peripheral nervous system by performing combined functional and histological analyses of adult L1-deficient mice (L1y/-) and wild-type (L1y/+) littermates. Contrary to our hypothesis, quantitative video-based motion analysis revealed better locomotor recovery in L1y/- than in L1y/+ mice at 4-12 weeks after transection and surgical repair of the femoral nerve. Motoneuron regeneration in L1y/- mice was also enhanced as indicated by attenuated post-traumatic loss of motoneurons, enhanced precision of motor reinnervation, larger cell bodies of regenerated motoneurons and diminished loss of inhibitory synaptic input to motoneurons. In search of mechanisms underlying the observed effects, we analysed peripheral nerves at short time-periods (3-14 days) after transection and found that Schwann cell proliferation is strongly augmented in L1y/- versus L1y/+ mice. L1-deficient Schwann cells showed increased proliferation than wild-type Schwann cells, both in vivo and in vitro. These findings suggest a novel role for L1 in nerve regeneration. We propose that L1 negatively regulates Schwann cell proliferation after nerve damage, which in turn restricts functional recovery by limiting the trophic support for regenerating motoneurons.
Collapse
Affiliation(s)
- Daria Guseva
- Zentrum für Molekulare Neurobiologie, Universität Hamburg, Hamburg, Germany
| | | | | | | |
Collapse
|
36
|
Abstract
BACKGROUND L1-cell adhesion molecule (L1-CAM) is a cell adhesion receptor of the immunoglobulin superfamily, known for its roles in nerve cell function. While originally believed to be present only in brain cells, in recent years L1-CAM has been detected in other tissues, and in a variety of cancer cells, including some common types of human cancer. OBJECTIVE/METHODS We review the prevalence of L1-CAM in human cancer, the possible mechanisms involved in L1-CAM-mediated tumorigenesis, and cancer therapies based upon L1-CAM antibody treatment. RESULTS/CONCLUSIONS In colon cancer cells, the L1-CAM gene was identified as a target of the Wnt/beta-catenin-TCF signaling pathway, and L1-CAM was exclusively detected at the invasive front of colon and ovarian cancer tissue. The expression of L1-CAM in normal and cancer cells enhanced tumorigenesis and conferred metastasis in colon cancer cells. Antibodies against the L1-CAM ectodomain severely inhibited the proliferation of a variety of cancer cells in culture and reduced tumor burden when injected into mice harboring cancer cells expressing L1-CAM. These results, in addition to the presence of L1-CAM on the cell surface and its restricted distribution in normal tissues, make it an ideal target for tumor therapy.
Collapse
Affiliation(s)
- Nancy Gavert
- Weizmann Institute of Science, Department of Molecular Cell Biology, Rehovot, Israel
| | | | | | | |
Collapse
|
37
|
Abstract
Oligodendrocytes and Schwann cells are highly specialized glial cells that wrap axons with a multilayered myelin membrane for rapid impulse conduction. Investigators have recently identified axonal signals that recruit myelin-forming Schwann cells from an alternate fate of simple axonal engulfment. This is the evolutionary oldest form of axon-glia interaction, and its function is unknown. Recent observations suggest that oligodendrocytes and Schwann cells not only myelinate axons but also maintain their long-term functional integrity. Mutations in the mouse reveal that axonal support by oligodendrocytes is independent of myelin assembly. The underlying mechanisms are still poorly understood; we do know that to maintain axonal integrity, mammalian myelin-forming cells require the expression of some glia-specific proteins, including CNP, PLP, and MAG, as well as intact peroxisomes, none of which is necessary for myelin assembly. Loss of glial support causes progressive axon degeneration and possibly local inflammation, both of which are likely to contribute to a variety of neuronal diseases in the central and peripheral nervous systems.
Collapse
Affiliation(s)
- Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, D-37075 Göttingen, Germany.
| | | |
Collapse
|
38
|
Nielsen J, Kulahin N, Walmod PS. Extracellular Protein Interactions Mediated by the Neural Cell Adhesion Molecule, NCAM: Heterophilic Interactions Between NCAM and Cell Adhesion Molecules, Extracellular Matrix Proteins, and Viruses. Neurochem Res 2008. [DOI: 10.1007/s11064-008-9761-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
39
|
Woolley AG, Tait KJ, Hurren BJ, Fisher L, Sheard PW, Duxson MJ. Developmental loss of NT-3 in vivo results in reduced levels of myelin-specific proteins, a reduced extent of myelination and increased apoptosis of Schwann cells. Glia 2008; 56:306-17. [PMID: 18080292 DOI: 10.1002/glia.20614] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
This work investigates the role of NT-3 in peripheral myelination. Recent articles, based in vitro, propose that NT-3 acting through its high-affinity receptor TrkC may act to inhibit myelin formation by enhancing Schwann cell motility and/or migration. Here, we investigate this hypothesis in vivo by examining myelination formation in NT-3 mutant mice. On the day of birth, soon after the onset of myelination, axons showed normal ensheathment by Schwann cells, no change in the proportion of axons which had begun to myelinate, and no change in either myelin thickness or number of myelin lamellae. However in postnatal day 21 mice, when myelination is substantially complete, we observed an unexpected reduction in mRNA and protein levels for MAG and P(0), and in myelin thickness. This is the opposite result to that predicted from previous in vitro studies, where removal of an inhibitory NT-3 signal would have been expected to enhance myelination. These results suggest that, in vivo, the importance of NT-3 as a major support factor for Schwann cells (Meier et al., (1999) J Neurosci 19:3847-3859) over-rides its potential role as an myelin inhibitor, with the net effect that loss of NT-3 results in degradation of Schwann cell functions, including myelination. In support of this idea, Schwann cells of NT-3 null mutants showed increased expression of activated caspase-3. Finally, we observed significant reduction in width of the Schwann cell periaxonal collar in NT-3 mutant animals suggesting that loss of NT-3 and resulting reduction in MAG levels may alter signaling at the axon-glial interface.
Collapse
Affiliation(s)
- Adele G Woolley
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin, New Zealand
| | | | | | | | | | | |
Collapse
|
40
|
Cheng SM, Carr CE. Functional delay of myelination of auditory delay lines in the nucleus laminaris of the barn owl. Dev Neurobiol 2008; 67:1957-74. [PMID: 17918244 DOI: 10.1002/dneu.20541] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
In the barn owl, maps of interaural time difference (ITD) are created in the nucleus laminaris (NL) by interdigitating axons that act as delay lines. Adult delay line axons are myelinated, and this myelination is timely, coinciding with the attainment of adult head size, and stable ITD cues. The proximal portions of the axons become myelinated in late embryonic life, but the delay line portions of the axon in NL remain unmyelinated until the first postnatal week. Myelination of the delay lines peaks at the third week posthatch, and myelinating oligodendrocyte density approaches adult levels by one month, when the head reaches its adult width. Migration of oligodendrocyte progenitors into NL and the subsequent onset of myelination may be restricted by a glial barrier in late embryonic stages and the first posthatch week, since the loss of tenascin-C immunoreactivity in NL is correlated with oligodendrocyte progenitor migration into NL.
Collapse
Affiliation(s)
- Shih-Min Cheng
- Department of Biology, University of Maryland, College Park, MD 20742-4415, USA.
| | | |
Collapse
|
41
|
Bock P, Beineke A, Techangamsuwan S, Baumgärtner W, Wewetzer K. Differential expression of HNK-1 and p75(NTR) in adult canine Schwann cells and olfactory ensheathing cells in situ but not in vitro. J Comp Neurol 2008; 505:572-85. [PMID: 17924534 DOI: 10.1002/cne.21519] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Olfactory ensheathing cells (OECs) are promising candidates for autologous cell transplantation therapies of nervous system injury and disease. Large animal models are relevant for transferring experimental data into clinical practice. In vivo studies have suggested that adult canine OECs may display similar regenerating capacities as their rodent counterpart. However, data on their molecular phenotype required for generating pure cell preparations are still scarce. In the present study, we comparatively analyzed expression of the carbohydrate HNK-1 epitope and the neurotrophin receptor p75(NTR) in adult canine Schwann cells and olfactory ensheathing cells in situ and in vitro. Myelinating and nonmyelinating Schwann cells in situ exclusively expressed HNK-1 and p75(NTR), respectively, whereas OECs were negative for both markers. In vitro, OECs and Schwann cells shared cell surface expression of p75(NTR) but not of HNK-1, which could be detected transiently in intracellular vesicles. This suggests that Schwann cells and OECs in vitro phagozytose HNK-1+ cellular debris. The cultivation-induced downregulation of HNK-1 expression in Schwann cells and upregulation of p75(NTR) in OECs argues for the possibility that axonal signals control the expression of both markers in situ. Whereas HNK-1 expression in Schwann cells is most likely controlled by signals inducing myelination, e.g., neuregulin, the mechanisms that may suppress p75(NTR) expression in OECs in situ remain to be elucidated. Interestingly, HNK-1 expression in the adult dog was found in both sensory and motor nerve myelinating Schwann cells. This is reminiscent of humans and differs from rodents; it also underscores the importance of large animal models for translational research.
Collapse
Affiliation(s)
- Patricia Bock
- Department of Pathology, University of Veterinary Medicine Hannover, 30559 Hannover, Germany
| | | | | | | | | |
Collapse
|
42
|
Baculovirus expression and bioactivity of a soluble 140kDa extracellular cleavage fragment of L1 neural cell adhesion molecule. Protein Expr Purif 2008; 57:172-9. [DOI: 10.1016/j.pep.2007.10.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2007] [Revised: 10/08/2007] [Accepted: 10/11/2007] [Indexed: 11/21/2022]
|
43
|
Desarnaud F, Jakovcevski M, Morellini F, Schachner M. Stress downregulates hippocampal expression of the adhesion molecules NCAM and CHL1 in mice by mechanisms independent of DNA methylation of their promoters. Cell Adh Migr 2008; 2:38-44. [PMID: 19262122 DOI: 10.4161/cam.2.1.6013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Stress is an important physiological regulator of brain function in young and adult mammals. The mechanisms underlying regulation of the consequences of stress, and in particular severe chronic stress, are thus important to investigate. These consequences most likely involve changes in synaptic function of brain areas being part of neural networks that regulate responses to stress. Cell adhesion molecules have been shown to regulate synaptic function in the adult and we were thus interested to investigate a regulatory mechanism that could influence expression of three adhesion molecules of the immunoglobulin superfamily (NCAM, L1 and CHL1) after exposure of early postnatal and adult mice to repeated stress. We hypothesized that reduction of adhesion molecule expression after chronic stress, as observed previously in vivo, could be due to gene silencing of the three molecules by DNA methylation. Although adhesion molecule expression was reduced after exposure of C57BL/6 mice to stress, thus validating our stress paradigm as imposing changes in adhesion molecule expression, we did not observe differences in methylation of CpG islands in the promoter regions of NCAM, L1 and CHL1, nor in the promoter region of the glucocorticoid receptor in the hippocampus, the expression of which at the protein level was also reduced after stress. We must therefore infer that severe stress in mice of the C57BL/6 strain downregulates adhesion molecule levels by mechanisms that do not relate to DNA methylation.
Collapse
Affiliation(s)
- Frank Desarnaud
- Department of Cell Biology and Neuroscience, WM Keck Center for Collaborative Neuroscience, Rutgers University, Piscataway, New Jersey, USA
| | | | | | | |
Collapse
|
44
|
Maurel P, Einheber S, Galinska J, Thaker P, Lam I, Rubin MB, Scherer SS, Murakami Y, Gutmann DH, Salzer JL. Nectin-like proteins mediate axon Schwann cell interactions along the internode and are essential for myelination. ACTA ACUST UNITED AC 2007; 178:861-74. [PMID: 17724124 PMCID: PMC2064549 DOI: 10.1083/jcb.200705132] [Citation(s) in RCA: 131] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Axon-glial interactions are critical for the induction of myelination and the domain organization of myelinated fibers. Although molecular complexes that mediate these interactions in the nodal region are known, their counterparts along the internode are poorly defined. We report that neurons and Schwann cells express distinct sets of nectin-like (Necl) proteins: axons highly express Necl-1 and -2, whereas Schwann cells express Necl-4 and lower amounts of Necl-2. These proteins are strikingly localized to the internode, where Necl-1 and -2 on the axon are directly apposed by Necl-4 on the Schwann cell; all three proteins are also enriched at Schmidt-Lanterman incisures. Binding experiments demonstrate that the Necl proteins preferentially mediate heterophilic rather than homophilic interactions. In particular, Necl-1 on axons binds specifically to Necl-4 on Schwann cells. Knockdown of Necl-4 by short hairpin RNA inhibits Schwann cell differentiation and subsequent myelination in cocultures. These results demonstrate a key role for Necl-4 in initiating peripheral nervous system myelination and implicate the Necl proteins as mediators of axo-glial interactions along the internode.
Collapse
Affiliation(s)
- Patrice Maurel
- Department of Cell Biology and Neurology, Smilow Neuroscience Program, New York University School of Medicine, New York, NY 10016, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Schachner M. Families of neural adhesion molecules. CIBA FOUNDATION SYMPOSIUM 2007; 145:156-69, discussion 169-72. [PMID: 2477202 DOI: 10.1002/9780470513828.ch10] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The neural cell adhesion molecules L1 and N-CAM share a common carbohydrate epitope that is recognized by the monoclonal antibodies L2 and HNK-1. The L2/HNK-1 epitope is also present on the myelin-associated glycoprotein (MAG) and secreted J1 glycoprotein, both of which have been identified as cell adhesion molecules. Each of the four adhesion molecules is differentially expressed during development on distinct cell types. Expression of the L2/HNK-1 epitope is regulated independently of the protein backbone, is phylogenetically conserved, and plays a role in cell-cell and, particularly, cell-substrate interactions. Another set of glycoproteins shares a common carbohydrate epitope designated L3. This epitope is present on the novel adhesion molecule on glia (AMOG), L1 and MAG, but not on J1 and N-CAM. As in the L2/HNK-1 family, the number of glycoproteins expressing this epitope is not yet known. It is therefore possible that heterogeneities in carbohydrate structures are associated with different sets of adhesion molecules and may have functional implications.
Collapse
Affiliation(s)
- M Schachner
- Department of Neurobiology, University of Heidelberg, Federal Republic of Germany
| |
Collapse
|
46
|
Hannila SS, Filbin MT. The role of cyclic AMP signaling in promoting axonal regeneration after spinal cord injury. Exp Neurol 2007; 209:321-32. [PMID: 17720160 PMCID: PMC2692909 DOI: 10.1016/j.expneurol.2007.06.020] [Citation(s) in RCA: 207] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2007] [Accepted: 06/28/2007] [Indexed: 01/08/2023]
Abstract
The failure of axons to regenerate after spinal cord injury remains one of the greatest challenges facing both medicine and neuroscience, but in the last 20 years there have been tremendous advances in the field of spinal cord injury repair. One of the most important of these has been the identification of inhibitory proteins in CNS myelin, and this has led to the development of strategies that will enable axons to overcome myelin inhibition. Elevation of intracellular cyclic AMP (cAMP) has been one of the most successful of these strategies, and in this review we examine how cAMP signaling promotes axonal regeneration in the CNS. Intracellular cAMP levels can be increased through a peripheral conditioning lesion, administration of cAMP analogues, priming with neurotrophins or treatment with the phosphodiesterase inhibitor rolipram, and each of these methods has been shown to overcome myelin inhibition both in vitro and in vivo. It is now known that the effects of cAMP are transcription dependent, and that cAMP-mediated activation of CREB leads to upregulated expression of genes such as arginase I and interleukin-6. The products of these genes have been shown to directly promote axonal regeneration, which raises the possibility that other cAMP-regulated genes could yield additional agents that would be beneficial in the treatment of spinal cord injury. Further study of these genes, in combination with human clinical trials of existing agents such as rolipram, would allow the therapeutic potential of cAMP to be fully realized.
Collapse
Affiliation(s)
| | - Marie T. Filbin
- Correspondence should be addressed to Dr. Marie T. Filbin at the above address.
| |
Collapse
|
47
|
Päiväläinen S, Heape AM. Myelin-associated glycoprotein and galactosylcerebroside expression in Schwann cells during myelination. Mol Cell Neurosci 2007; 35:436-46. [PMID: 17513122 DOI: 10.1016/j.mcn.2007.04.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2007] [Revised: 04/11/2007] [Accepted: 04/17/2007] [Indexed: 01/06/2023] Open
Abstract
The temporo-spatial expression profiles of the myelin-associated glycoprotein (MAG) and galactosylcerebrosides (Gal-CBs) were analysed during Schwann cell differentiation and myelination in Schwann cell cultures and in dorsal root ganglion/Schwann cell cocultures. The temporal expression profiles demonstrate that the mechanisms triggering and regulating MAG and Gal-CBs are different and independent. They also provide further support for a role of MAG, but not Gal-CBs, in the establishment of the Schwann cell/axon interaction and in the formation of the primary mesaxon, while both MAG and Gal-CBs may participate in the further maturation of the mesaxon. The spatial expression profiles demonstrate that although Gal-CBs are concentrated in the same structures (mesaxonal spirals, paranodes and Schmidt-Lanterman incisures) as MAG in both developing and mature sheaths, where it is therefore possible that the two might have complementary functions, they are also present in regions from which MAG is excluded.
Collapse
Affiliation(s)
- Satu Päiväläinen
- The Myelin Group, Department of Anatomy & Cell Biology, P.O. Box 5000 (Aapistie 7A), 90014 University of Oulu, Finland
| | | |
Collapse
|
48
|
Amici SA, Dunn WA, Notterpek L. Developmental abnormalities in the nerves of peripheral myelin protein 22-deficient mice. J Neurosci Res 2007; 85:238-49. [PMID: 17131416 DOI: 10.1002/jnr.21118] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Peripheral myelin protein 22 (PMP22) is a tetraspan glycoprotein whose misexpression is associated with a family of hereditary peripheral neuropathies. In a recent report, we have characterized a novel PMP22-deficient mouse model in which the first two coding exons were replaced by the lacZ reporter. To investigate further the myelin abnormalities in the absence of PMP22, sciatic nerves and dorsal root ganglion (DRG) neuron explant cultures from PMP22-deficient mice were studied at various stages of myelination. Throughout the first 3 months of postnatal development, myelin protein and beta4 integrin levels are dramatically reduced, whereas p75 and beta1 integrin remain elevated. By immunostaining, the distributions of several glial proteins, including beta4 integrin, the voltage-gated potassium channel Kv1.1, and E-cadherin, are altered. Schwann cells from PMP22-deficient mice are able to produce limited amounts of myelin in DRG explant cultures, yet the internodal segments are dramatically fewer and shorter. The comparison of PMP22-deficient mice with other PMP22 mutant models reveals that the decrease in beta4 integrin is specific to an absence of PMP22. Furthermore, whereas lysosome-associated membrane protein 1 and ubiquitin are notably up-regulated in nerves of PMP22-deficient mice, heat shock protein 70 levels remain constant or decrease compared with wild-type or PMP22 mutant samples. Together these results support a role for PMP22 in the early events of peripheral nerve myelination. Additionally, although myelin abnormalities are a commonality among PMP22 neuropathic models, the underlying subcellular mechanisms are distinct and depend on the specific genetic abnormality.
Collapse
Affiliation(s)
- Stephanie A Amici
- Department of Neuroscience, College of Medicine, McKnight Brain Institute, University of Florida, Gainesville, Florida 32610, USA
| | | | | |
Collapse
|
49
|
Hannila SS, Siddiq MM, Filbin MT. Therapeutic Approaches to Promoting Axonal Regeneration in the Adult Mammalian Spinal Cord. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2007; 77:57-105. [PMID: 17178472 DOI: 10.1016/s0074-7742(06)77003-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Sari S Hannila
- Department of Biological Sciences, Hunter College, City University of New York, New York 10021, USA
| | | | | |
Collapse
|
50
|
Gingras M, Beaulieu MM, Gagnon V, Durham HD, Berthod F. In vitro study of axonal migration and myelination of motor neurons in a three-dimensional tissue-engineered model. Glia 2007; 56:354-64. [DOI: 10.1002/glia.20617] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|