101
|
Shin JE, Ha H, Cho EH, Kim YK, Cho Y. Comparative analysis of the transcriptome of injured nerve segments reveals spatiotemporal responses to neural damage in mice. J Comp Neurol 2018; 526:1195-1208. [PMID: 29405296 DOI: 10.1002/cne.24404] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 01/22/2018] [Accepted: 01/23/2018] [Indexed: 02/02/2023]
Abstract
Nerves are particularly vulnerable to damage due to their unique structure with meter-long axons. In the peripheral nervous system neurons and Schwann cells can activate the injury-response program that directs axons to either regenerate or degenerate after traumatic nerve injury. However, the differences between the genetic programs driving nerve regeneration and degeneration have not yet been described extensively. To understand these differences, in this study we have compared the injury-induced transcriptomic changes between the regenerating proximal segment and the degenerating distal segment of a transected nerve, at different post-injury time points. We analyzed the spatiotemporal dynamics of the mouse transcriptome using a sciatic nerve-injury model by means of RNA sequencing. The results of the differentially regulated genes (DEGs) analysis show that some DEG groups are similarly regulated in both proximal and distal segments, and primarily display a positive correlation. However, some DEG groups are exclusively regulated in either the proximal or the distal segment, suggesting that these DEG groups constitute a genetic network for distinguishing the regenerative and degenerative responses. In addition, our gene ontology analysis revealed an enrichment of particular biological processes in different phases and locations. Thus, our data provide a spatiotemporal profile of the transcriptomes that are differentially regulated in either regenerating or degenerating nerves, in vivo. The specific biological processes enriched in the DEG groups might delineate the injury-responsive program that induces contrasting regenerative and degenerative responses in different nerve segments.
Collapse
Affiliation(s)
- Jung Eun Shin
- The Research Institute of Basic Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Hongseok Ha
- Division of Life Sciences, Korea University, Seoul, 02841, Republic of Korea.,Creative Research Initiatives Center for Molecular Biology of Translation, Korea University, Seoul, 02841, Republic of Korea
| | - Eun Hye Cho
- Division of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| | - Yoon Ki Kim
- Division of Life Sciences, Korea University, Seoul, 02841, Republic of Korea.,Creative Research Initiatives Center for Molecular Biology of Translation, Korea University, Seoul, 02841, Republic of Korea
| | - Yongcheol Cho
- Division of Life Sciences, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
102
|
Furlan A, Adameyko I. Schwann cell precursor: a neural crest cell in disguise? Dev Biol 2018; 444 Suppl 1:S25-S35. [PMID: 29454705 DOI: 10.1016/j.ydbio.2018.02.008] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/14/2018] [Accepted: 02/14/2018] [Indexed: 01/19/2023]
Abstract
Schwann cell precursors (SCPs) are multipotent embryonic progenitors covering all developing peripheral nerves. These nerves grow and navigate with unprecedented precision, delivering SCP progenitors to almost all locations in the embryonic body. Within specific developing tissues, SCPs detach from nerves and generate neuroendocrine cells, autonomic neurons, mature Schwann cells, melanocytes and other cell types. These properties of SCPs evoke resemblances between them and their parental population, namely, neural crest cells. Neural crest cells are incredibly multipotent migratory cells that revolutionized the course of evolution in the lineage of early chordate animals. Given this similarity and recent data, it is possible to hypothesize that proto-neural crest cells are similar to SCPs spreading along the nerves. Here, we review the multipotency of SCPs, the signals that govern them, their potential therapeutic value, SCP's embryonic origin and their evolutionary connections. We dedicate this article to the memory of Wilhelm His, the father of the microtome and "Zwischenstrang", currently known as the neural crest.
Collapse
Affiliation(s)
- Alessandro Furlan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, New York 11724 USA
| | - Igor Adameyko
- Department of Physiology and Pharmacology, Karolinska Institutet, 17177 Stockholm, Sweden; Center for Brain Research, Medical University Vienna, 1090 Vienna, Austria.
| |
Collapse
|
103
|
Abstract
The journey of Schwann cells from their origin in the neural crest to their ensheathment and myelination of peripheral nerves is a remarkable one. Their apparent static function in enabling saltatory conduction of mature nerve is not only vital for long-term health of peripheral nerve but also belies an innate capacity of terminally differentiated Schwann cells to radically alter their differentiation status in the face of nerve injury. The transition from migrating neural crest cells to nerve ensheathment, and then myelination of large diameter axons has been characterized extensively and several of the transcriptional networks have been identified. However, transcription factors must also modify chromatin structure during Schwann cell maturation and this review will focus on chromatin modification machinery that is involved in promoting the transition to, and maintenance of, myelinating Schwann cells. In addition, Schwann cells are known to play important regenerative roles after peripheral nerve injury, and information on epigenomic reprogramming of the Schwann cell genome has emerged. Characterization of epigenomic requirements for myelin maintenance and Schwann cell responses to injury will be vital in understanding how the various Schwann cell functions can be optimized to maintain and repair peripheral nerve function.
Collapse
Affiliation(s)
- Ki H Ma
- 1 Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - John Svaren
- 1 Waisman Center, University of Wisconsin-Madison, Madison, WI, USA.,2 Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
104
|
Abstract
Schwann cells are the main glial cells of the peripheral nervous system (PNS) and play key roles in peripheral nerve development and function, including providing myelin that is essential for normal movement and sensation in the adult. Schwann cells can be readily destabilized by a wide variety of distinct conditions that range from nerve injury to immune assaults, metabolic disturbances, microbial infections, or genetic defects, leading to the breakdown of myelin (demyelination) and a subsequent switch in phenotypic states. This striking feature of Schwann cells forms the cornerstone of several debilitating and even fatal PNS neurological disorders that include the demyelinating neuropathies Guillain Barré syndrome (GBS) and Charcot-Marie-Tooth disease (CMT), and PNS cancers, including Neurofibromatosis.Primary Schwann cell cultures have proved a valuable tool to dissect key mechanisms that regulate proliferation, survival, differentiation, and myelination of these glial cell types. In this chapter, we describe the steps involved in the isolation and purification of Schwann cells from rodent peripheral nerves and the use of these cultures to model myelination in vitro.
Collapse
|
105
|
The Pseudopod System for Axon-Glia Interactions: Stimulation and Isolation of Schwann Cell Protrusions that Form in Response to Axonal Membranes. Methods Mol Biol 2018; 1739:233-253. [PMID: 29546711 PMCID: PMC7373153 DOI: 10.1007/978-1-4939-7649-2_15] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
In the peripheral nervous system, axons dictate the differentiation state of Schwann cells. Most of this axonal influence on Schwann cells is due to juxtacrine interactions between axonal transmembrane molecules (e.g., the neuregulin growth factor) and receptors on the Schwann cell (e.g., the ErbB2/ErbB3 receptor). The fleeting nature of this interaction together with the lack of synchronicity in the development of the Schwann cell population limits our capability to study this phenomenon in vivo. Here we present a simple Boyden Chamber-based method to study this important cell-cell interaction event. We isolate the early protrusions of Schwann cells that are generated in response to juxtacrine stimulation by sensory neuronal membranes. This method is compatible with a large array of current biochemical analyses and provides an effective approach to study biomolecules that are differentially localized in Schwann cell protrusions and cell bodies in response to axonal signals. A similar approach can be extended to different kinds of cell-cell interactions.
Collapse
|
106
|
Suh JKF, Hyung S. Primary Motor Neuron Culture to Promote Cellular Viability and Myelination. Methods Mol Biol 2018; 1727:403-411. [PMID: 29222800 DOI: 10.1007/978-1-4939-7571-6_32] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A culture system that can recapitulate myelination in vitro will not only help us to better understand the mechanism of myelination and demyelination but also identify possible therapeutic interventions for treating demyelinating diseases. Here, we introduce a simple and reproducible myelination culture system using mouse motor neurons (MNs) and Schwann cells (SCs). Dissociated motor neurons are plated on a feeder layer of SCs, which interact with and wrap around the axons of MNs as they differentiate in culture. In our MN-SC co-culture system, MNs survive over 3 weeks and extend long axons. Both viability and axon growth of MNs in the co-culture are markedly enhanced as compared to those of MN monocultures. Co-labeling of myelin basic proteins and neuronal cell microtubules reveals that SCs form myelin sheaths by wrapping around the axons of MNs.
Collapse
Affiliation(s)
| | - Sujin Hyung
- Multiscale Mechanical Design Laboratory, School of Mechanical and Aerospace Engineering, Seoul National University, Seoul, South Korea
| |
Collapse
|
107
|
Kastriti ME, Adameyko I. Specification, plasticity and evolutionary origin of peripheral glial cells. Curr Opin Neurobiol 2017; 47:196-202. [DOI: 10.1016/j.conb.2017.11.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Revised: 08/30/2017] [Accepted: 11/03/2017] [Indexed: 12/31/2022]
|
108
|
Aquino JB, Sierra R. Schwann cell precursors in health and disease. Glia 2017; 66:465-476. [PMID: 29124786 DOI: 10.1002/glia.23262] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 10/07/2017] [Accepted: 10/26/2017] [Indexed: 12/25/2022]
Abstract
Schwann cell precursors (SCPs) are frequently regarded as neural crest-derived cells (NCDCs) found in contact with axons during nerve formation. Nevertheless, cells with SCPs properties can be found up to the adulthood. They are well characterized with regard to both gene expression profile and cellular behavior -for instance, proliferation, migratory capabilities and survival requirements-. They differ in origin regarding their anatomic location: even though most of them are derived from migratory NCCs, there is also contribution of the boundary cap neural crest cells (bNCCs) to the skin and other tissues. Many functions are known for SCPs in normal development, including nerve fasciculation and target innervation, arterial branching patterning and differentiation, and other morphogenetic processes. In addition, SCPs are now known to be a source of many neural (glia, endoneural fibroblasts, melanocytes, visceral neurons, and chromaffin cells) and non-neural-like (mesenchymal stromal cells, able e.g., to generate dentine-producing odontoblasts) cell types. Until now no reports of endoderm-like derivatives were reported so far. Interestingly, in the Schwann cell lineage only early SCPs are likely able to differentiate into melanocytes and bone marrow mesenchymal stromal cells. We have also herein discussed the literature regarding their role in repair as well as in disease mechanisms, such as in diverse cancers. Moreover, many caveats in our knowledge of SCPs biology are highlighted all through this article. Future research should expand more into the relevance of SCPs in pathologies and in other regenerative mechanisms which might bring new unexpected clinically-relevant knowledge.
Collapse
Affiliation(s)
- Jorge B Aquino
- Developmental Biology & Regenerative Medicine Laboratory, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Derqui-Pilar, Buenos Aires, Argentina
| | - Romina Sierra
- Developmental Biology & Regenerative Medicine Laboratory, Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Derqui-Pilar, Buenos Aires, Argentina
| |
Collapse
|
109
|
Chan WH, Anderson CR, Gonsalvez DG. From proliferation to target innervation: signaling molecules that direct sympathetic nervous system development. Cell Tissue Res 2017; 372:171-193. [PMID: 28971249 DOI: 10.1007/s00441-017-2693-x] [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: 06/06/2017] [Accepted: 08/30/2017] [Indexed: 02/07/2023]
Abstract
The sympathetic division of the autonomic nervous system includes a variety of cells including neurons, endocrine cells and glial cells. A recent study (Furlan et al. 2017) has revised thinking about the developmental origin of these cells. It now appears that sympathetic neurons and chromaffin cells of the adrenal medulla do not have an immediate common ancestor in the form a "sympathoadrenal cell", as has been long believed. Instead, chromaffin cells arise from Schwann cell precursors. This review integrates the new findings with the expanding body of knowledge on the signalling pathways and transcription factors that regulate the origin of cells of the sympathetic division of the autonomic nervous system.
Collapse
Affiliation(s)
- W H Chan
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, The University of Melbourne, Parkville, 3010, Australia
| | - C R Anderson
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, The University of Melbourne, Parkville, 3010, Australia
| | - David G Gonsalvez
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, The University of Melbourne, Parkville, 3010, Australia.
| |
Collapse
|
110
|
Clements MP, Byrne E, Camarillo Guerrero LF, Cattin AL, Zakka L, Ashraf A, Burden JJ, Khadayate S, Lloyd AC, Marguerat S, Parrinello S. The Wound Microenvironment Reprograms Schwann Cells to Invasive Mesenchymal-like Cells to Drive Peripheral Nerve Regeneration. Neuron 2017; 96:98-114.e7. [PMID: 28957681 PMCID: PMC5626803 DOI: 10.1016/j.neuron.2017.09.008] [Citation(s) in RCA: 211] [Impact Index Per Article: 30.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2016] [Revised: 07/07/2017] [Accepted: 09/06/2017] [Indexed: 01/05/2023]
Abstract
Schwann cell dedifferentiation from a myelinating to a progenitor-like cell underlies the remarkable ability of peripheral nerves to regenerate following injury. However, the molecular identity of the differentiated and dedifferentiated states in vivo has been elusive. Here, we profiled Schwann cells acutely purified from intact nerves and from the wound and distal regions of severed nerves. Our analysis reveals novel facets of the dedifferentiation response, including acquisition of mesenchymal traits and a Myc module. Furthermore, wound and distal dedifferentiated Schwann cells constitute different populations, with wound cells displaying increased mesenchymal character induced by localized TGFβ signaling. TGFβ promotes invasion and crosstalks with Eph signaling via N-cadherin to drive collective migration of the Schwann cells across the wound. Consistently, Tgfbr2 deletion in Schwann cells resulted in misdirected and delayed reinnervation. Thus, the wound microenvironment is a key determinant of Schwann cell identity, and it promotes nerve repair through integration of multiple concerted signals. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Melanie P Clements
- Cell Interactions and Cancer Group, MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom
| | - Elizabeth Byrne
- Cell Interactions and Cancer Group, MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom
| | - Luis F Camarillo Guerrero
- Cell Interactions and Cancer Group, MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom; Quantitative Gene Expression Group, MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, United Kingdom
| | - Anne-Laure Cattin
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Leila Zakka
- Cell Interactions and Cancer Group, MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom
| | - Azhaar Ashraf
- Cell Interactions and Cancer Group, MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom
| | - Jemima J Burden
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | - Sanjay Khadayate
- Cell Interactions and Cancer Group, MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom
| | - Alison C Lloyd
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, United Kingdom; UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, United Kingdom
| | - Samuel Marguerat
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom; Quantitative Gene Expression Group, MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, United Kingdom
| | - Simona Parrinello
- Cell Interactions and Cancer Group, MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, United Kingdom; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, Du Cane Road, London W12 0NN, United Kingdom.
| |
Collapse
|
111
|
Arthur-Farraj PJ, Morgan CC, Adamowicz M, Gomez-Sanchez JA, Fazal SV, Beucher A, Razzaghi B, Mirsky R, Jessen KR, Aitman TJ. Changes in the Coding and Non-coding Transcriptome and DNA Methylome that Define the Schwann Cell Repair Phenotype after Nerve Injury. Cell Rep 2017; 20:2719-2734. [PMID: 28903050 PMCID: PMC5608958 DOI: 10.1016/j.celrep.2017.08.064] [Citation(s) in RCA: 137] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Revised: 06/17/2017] [Accepted: 08/18/2017] [Indexed: 12/12/2022] Open
Abstract
Repair Schwann cells play a critical role in orchestrating nerve repair after injury, but the cellular and molecular processes that generate them are poorly understood. Here, we perform a combined whole-genome, coding and non-coding RNA and CpG methylation study following nerve injury. We show that genes involved in the epithelial-mesenchymal transition are enriched in repair cells, and we identify several long non-coding RNAs in Schwann cells. We demonstrate that the AP-1 transcription factor C-JUN regulates the expression of certain micro RNAs in repair Schwann cells, in particular miR-21 and miR-34. Surprisingly, unlike during development, changes in CpG methylation are limited in injury, restricted to specific locations, such as enhancer regions of Schwann cell-specific genes (e.g., Nedd4l), and close to local enrichment of AP-1 motifs. These genetic and epigenomic changes broaden our mechanistic understanding of the formation of repair Schwann cell during peripheral nervous system tissue repair.
Collapse
Affiliation(s)
- Peter J Arthur-Farraj
- Department of Clinical Neurosciences, Addenbrooke's Hospital, University of Cambridge, Cambridge CB2 0QQ, UK; Department of Medicine, Imperial College, London W12 0NN, UK; Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK.
| | - Claire C Morgan
- Department of Medicine, Imperial College, London W12 0NN, UK
| | - Martyna Adamowicz
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH16 2XU, UK
| | - Jose A Gomez-Sanchez
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Shaline V Fazal
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Anthony Beucher
- Department of Medicine, Imperial College, London W12 0NN, UK
| | - Bonnie Razzaghi
- Department of Medicine, Imperial College, London W12 0NN, UK
| | - Rhona Mirsky
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Kristjan R Jessen
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, UK
| | - Timothy J Aitman
- Department of Medicine, Imperial College, London W12 0NN, UK; Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh EH16 2XU, UK.
| |
Collapse
|
112
|
Directed Differentiation of Human Bone Marrow Stromal Cells to Fate-Committed Schwann Cells. Stem Cell Reports 2017; 9:1097-1108. [PMID: 28890164 PMCID: PMC5639182 DOI: 10.1016/j.stemcr.2017.08.004] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 08/11/2017] [Accepted: 08/11/2017] [Indexed: 12/14/2022] Open
Abstract
Our ultimate goal of in vitro derivation of Schwann cells (SCs) from adult bone marrow stromal cells (BMSCs) is such that they may be used autologously to assist post-traumatic nerve regeneration. Existing protocols for derivation of SC-like cells from BMSCs fall short in the stability of the acquired phenotype and the functional capacity to myelinate axons. Our experiments indicated that neuro-ectodermal progenitor cells among the human hBMSCs could be selectively expanded and then induced to differentiate into SC-like cells. Co-culture of the SC-like cells with embryonic dorsal root ganglion neurons facilitated contact-mediated signaling that accomplished the switch to fate-committed SCs. Microarray analysis and in vitro myelination provided evidence that the human BMSC-derived SCs were functionally mature. This was reinforced by repair and myelination phenotypes observable in vivo with the derived SCs seeded into a nerve guide as an implant across a critical gap in a rat model of sciatic nerve injury. A protocol for in vitro derivation of fate-committed SCs from human BMSCs The derived human SCs were functionally capable of myelination in vitro The derived human SCs guided axonal regrowth and formed compact myelin in vivo
Collapse
|
113
|
Roberts SL, Dun XP, Doddrell RDS, Mindos T, Drake LK, Onaitis MW, Florio F, Quattrini A, Lloyd AC, D'Antonio M, Parkinson DB. Sox2 expression in Schwann cells inhibits myelination in vivo and induces influx of macrophages to the nerve. Development 2017; 144:3114-3125. [PMID: 28743796 PMCID: PMC5611958 DOI: 10.1242/dev.150656] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 07/13/2017] [Indexed: 12/25/2022]
Abstract
Correct myelination is crucial for the function of the peripheral nervous system. Both positive and negative regulators within the axon and Schwann cell function to ensure the correct onset and progression of myelination during both development and following peripheral nerve injury and repair. The Sox2 transcription factor is well known for its roles in the development and maintenance of progenitor and stem cell populations, but has also been proposed in vitro as a negative regulator of myelination in Schwann cells. We wished to test fully whether Sox2 regulates myelination in vivo and show here that, in mice, sustained Sox2 expression in vivo blocks myelination in the peripheral nerves and maintains Schwann cells in a proliferative non-differentiated state, which is also associated with increased inflammation within the nerve. The plasticity of Schwann cells allows them to re-myelinate regenerated axons following injury and we show that re-myelination is also blocked by Sox2 expression in Schwann cells. These findings identify Sox2 as a physiological regulator of Schwann cell myelination in vivo and its potential to play a role in disorders of myelination in the peripheral nervous system.
Collapse
Affiliation(s)
- Sheridan L Roberts
- Plymouth University Peninsula Schools of Medicine and Dentistry, John Bull Building, Plymouth Science Park, Plymouth PL6 8BU, UK
| | - Xin-Peng Dun
- Plymouth University Peninsula Schools of Medicine and Dentistry, John Bull Building, Plymouth Science Park, Plymouth PL6 8BU, UK
| | - Robin D S Doddrell
- Plymouth University Peninsula Schools of Medicine and Dentistry, John Bull Building, Plymouth Science Park, Plymouth PL6 8BU, UK
| | - Thomas Mindos
- Plymouth University Peninsula Schools of Medicine and Dentistry, John Bull Building, Plymouth Science Park, Plymouth PL6 8BU, UK
| | | | - Mark W Onaitis
- Department of Thoracic Surgery, University of California, San Diego, CA 92103, USA
| | - Francesca Florio
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, DIBIT, 20132 Milan, Italy
| | - Angelo Quattrini
- Division of Neuroscience, San Raffaele Scientific Institute, DIBIT, 20132 Milan, Italy
| | - Alison C Lloyd
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, UK
| | - Maurizio D'Antonio
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, DIBIT, 20132 Milan, Italy
| | - David B Parkinson
- Plymouth University Peninsula Schools of Medicine and Dentistry, John Bull Building, Plymouth Science Park, Plymouth PL6 8BU, UK
| |
Collapse
|
114
|
Boerboom A, Reusch C, Pieltain A, Chariot A, Franzen R. KIAA1199: A novel regulator of MEK/ERK-induced Schwann cell dedifferentiation. Glia 2017; 65:1682-1696. [PMID: 28699206 DOI: 10.1002/glia.23188] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Revised: 06/22/2017] [Accepted: 06/23/2017] [Indexed: 12/14/2022]
Abstract
The molecular mechanisms that regulate Schwann cell (SC) plasticity and the role of the Nrg1/ErbB-induced MEK1/ERK1/2 signalling pathway in SC dedifferentiation or in myelination remain unclear. It is currently believed that different levels of MEK1/ERK1/2 activation define the state of SC differentiation. Thus, the identification of new regulators of MEK1/ERK1/2 signalling could help to decipher the context-specific aspects driving the effects of this pathway on SC plasticity. In this perspective, we have investigated the potential role of KIAA1199, a protein that promotes ErbB and MEK1/ERK1/2 signalling in cancer cells, in SC plasticity. We depleted KIAA1199 in the SC-derived MSC80 cell line with RNA-interference-based strategy and also generated Tamoxifen-inducible and conditional mouse models in which KIAA1199 is inactivated through homologous recombination, using the Cre-lox technology. We show that the invalidation of KIAA1199 in SC decreases the expression of cJun and other negative regulators of myelination and elevates Krox20, driving them towards a pro-myelinating phenotype. We further show that in dedifferentiation conditions, SC invalidated for KIAA1199 exhibit lower myelin clearance as well as increased myelination capacity. Finally, the Nrg1-induced activation of the MEK/ERK/1/2 pathway is severely reduced when KIAA1199 is absent, indicating that KIAA1199 promotes Nrg1-dependent MEK1 and ERK1/2 activation in SCs. In conclusion, this work identifies KIAA1199 as a novel regulator of MEK/ERK-induced SC dedifferentiation and contributes to a better understanding of the molecular control of SC dedifferentiation.
Collapse
Affiliation(s)
| | - Céline Reusch
- GIGA-Molecular Biology of Diseases, University of Liège, Belgium
| | | | - Alain Chariot
- GIGA-Molecular Biology of Diseases, University of Liège, Belgium.,Walloon Excellence in Lifesciences and Biotechnology (WELBIO), Wavre, Belgium
| | | |
Collapse
|
115
|
Tricaud N, Park HT. Wallerian demyelination: chronicle of a cellular cataclysm. Cell Mol Life Sci 2017; 74:4049-4057. [PMID: 28600652 PMCID: PMC5641270 DOI: 10.1007/s00018-017-2565-2] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Revised: 05/10/2017] [Accepted: 06/01/2017] [Indexed: 12/23/2022]
Abstract
Wallerian demyelination is characteristic of peripheral nerve degeneration after traumatic injury. After axonal degeneration, the myelinated Schwann cell undergoes a stereotypical cellular program that results in the disintegration of the myelin sheath, a process termed demyelination. In this review, we chronologically describe this program starting from the late and visible features of myelin destruction and going backward to the initial molecular steps that trigger the nuclear reprogramming few hours after injury. Wallerian demyelination is a wonderful model for myelin degeneration occurring in the diverse forms of demyelinating peripheral neuropathies that plague human beings.
Collapse
Affiliation(s)
- Nicolas Tricaud
- INSERM U1051, Institut des Neurosciences de Montpellier (INM), Université de Montpellier, Montpellier, France.
| | - Hwan Tae Park
- Peripheral Neuropathy Research Center, Department of Physiology, College of Medicine, Dong-A University, Busan, South Korea
| |
Collapse
|
116
|
Hu X, Hou H, Bastian C, He W, Qiu S, Ge Y, Yin X, Kidd GJ, Brunet S, Trapp BD, Baltan S, Yan R. BACE1 regulates the proliferation and cellular functions of Schwann cells. Glia 2017; 65:712-726. [PMID: 28191691 PMCID: PMC5357169 DOI: 10.1002/glia.23122] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2016] [Revised: 12/16/2016] [Accepted: 01/18/2017] [Indexed: 12/31/2022]
Abstract
BACE1 is an indispensable enzyme for generating β-amyloid peptides, which are excessively accumulated in brains of Alzheimer's patients. However, BACE1 is also required for proper myelination of peripheral nerves, as BACE1-null mice display hypomyelination. To determine the precise effects of BACE1 on myelination, here we have uncovered a role of BACE1 in the control of Schwann cell proliferation during development. We demonstrate that BACE1 regulates the cleavage of Jagged-1 and Delta-1, two membrane-bound ligands of Notch. BACE1 deficiency induces elevated Jag-Notch signaling activity, which in turn facilitates proliferation of Schwann cells. This increase in proliferation leads to shortened internodes and decreased Schmidt-Lanterman incisures. Functionally, evoked compound action potentials in BACE1-null nerves were significantly smaller and slower, with a clear decrease in excitability. BACE1-null nerves failed to effectively use lactate as an alternative energy source under conditions of increased physiological activity. Correlatively, BACE1-null mice showed reduced performance on rotarod tests. Collectively, our data suggest that BACE1 deficiency enhances proliferation of Schwann cell due to the elevated Jag1/Delta1-Notch signaling, but fails to myelinate axons efficiently due to impaired the neuregulin1-ErbB signaling, which has been documented.
Collapse
Affiliation(s)
- Xiangyou Hu
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Hailong Hou
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Chinthasagar Bastian
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Wanxia He
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Shupeng Qiu
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Yingying Ge
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Xinhua Yin
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Grahame J. Kidd
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Sylvain Brunet
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Bruce D. Trapp
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Selva Baltan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Riqiang Yan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| |
Collapse
|
117
|
Comparison of DNA Methylation in Schwann Cells before and after Peripheral Nerve Injury in Rats. BIOMED RESEARCH INTERNATIONAL 2017; 2017:5393268. [PMID: 28459064 PMCID: PMC5385226 DOI: 10.1155/2017/5393268] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 02/23/2017] [Accepted: 03/08/2017] [Indexed: 01/13/2023]
Abstract
This study aims to find the difference of genomewide DNA methylation in Schwann cells (SCs) before and after peripheral nerve system (PNS) injury by Methylated DNA Immunoprecipitation Sequencing (MeDIP-Seq) and seek meaningful differentially methylated genes related to repairment of injured PNS. SCs harvested from sciatic nerve were named as activated Schwann cells (ASCs), and the ones harvested from brachial plexus were named as normal Schwann cells (NSCs). Genomic DNA of ASCs and NSCs were isolated and MeDIP-Seq was conducted. Differentially methylated genes and regions were discovered and analyzed by bioinformatic methods. MeDIP-Seq analysis showed methylation differences were identified between ASCs and NSCs. The distribution of differentially methylated regions (DMRs) peaks in different components of genome was mainly located in distal intergenic regions. GO and KEGG analysis of these methylated genes were also conducted. The expression patterns of hypermethylated genes (Dgcr8, Zeb2, Dixdc1, Sox2, and Shh) and hypomethylated genes (Gpr126, Birc2) detected by qRT-PCR were opposite to the MeDIP analysis data with significance (p < 0.05), which proved MeDIP analysis data were real and believable. Our data serve as a basis for understanding the injury-induced epigenetic changes in SCs and the foundation for further studies on repair of PNS injury.
Collapse
|
118
|
STAT3 Controls the Long-Term Survival and Phenotype of Repair Schwann Cells during Nerve Regeneration. J Neurosci 2017; 37:4255-4269. [PMID: 28320842 PMCID: PMC5413174 DOI: 10.1523/jneurosci.3481-16.2017] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 01/11/2017] [Accepted: 01/19/2017] [Indexed: 11/21/2022] Open
Abstract
After nerve injury, Schwann cells convert to a phenotype specialized to promote repair. But during the slow process of axonal regrowth, these repair Schwann cells gradually lose their regeneration-supportive features and eventually die. Although this is a key reason for the frequent regeneration failures in humans, the transcriptional mechanisms that control long-term survival and phenotype of repair cells have not been studied, and the molecular signaling underlying their decline is obscure. We show, in mice, that Schwann cell STAT3 has a dual role. It supports the long-term survival of repair Schwann cells and is required for the maintenance of repair Schwann cell properties. In contrast, STAT3 is less important for the initial generation of repair Schwann cells after injury. In repair Schwann cells, we find that Schwann cell STAT3 activation by Tyr705 phosphorylation is sustained during long-term denervation. STAT3 is required for maintaining autocrine Schwann cell survival signaling, and inactivation of Schwann cell STAT3 results in a striking loss of repair cells from chronically denervated distal stumps. STAT3 inactivation also results in abnormal morphology of repair cells and regeneration tracks, and failure to sustain expression of repair cell markers, including Shh, GDNF, and BDNF. Because Schwann cell development proceeds normally without STAT3, the function of this factor appears restricted to Schwann cells after injury. This identification of transcriptional mechanisms that support long-term survival and differentiation of repair cells will help identify, and eventually correct, the failures that lead to the deterioration of this important cell population. SIGNIFICANCE STATEMENT Although injured peripheral nerves contain repair Schwann cells that provide signals and spatial clues for promoting regeneration, the clinical outcome after nerve damage is frequently poor. A key reason for this is that, during the slow growth of axons through the proximal parts of injured nerves repair, Schwann cells gradually lose regeneration-supporting features and eventually die. Identification of signals that sustain repair cells is therefore an important goal. We have found that in mice the transcription factor STAT3 protects these cells from death and contributes to maintaining the molecular and morphological repair phenotype that promotes axonal regeneration. Defining the molecular mechanisms that maintain repair Schwann cells is an essential step toward developing therapeutic strategies that improve nerve regeneration and functional recovery.
Collapse
|
119
|
Miller SR, Perera SN, Baker CVH. Constitutively active Notch1 converts cranial neural crest-derived frontonasal mesenchyme to perivascular cells in vivo. Biol Open 2017; 6:317-325. [PMID: 28183698 PMCID: PMC5374403 DOI: 10.1242/bio.023887] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Perivascular/mural cells originate from either the mesoderm or the cranial neural crest. Regardless of their origin, Notch signalling is necessary for their formation. Furthermore, in both chicken and mouse, constitutive Notch1 activation (via expression of the Notch1 intracellular domain) is sufficient in vivo to convert trunk mesoderm-derived somite cells to perivascular cells, at the expense of skeletal muscle. In experiments originally designed to investigate the effect of premature Notch1 activation on the development of neural crest-derived olfactory ensheathing glial cells (OECs), we used in ovo electroporation to insert a tetracycline-inducible NotchΔE construct (encoding a constitutively active mutant of mouse Notch1) into the genome of chicken cranial neural crest cell precursors, and activated NotchΔE expression by doxycycline injection at embryonic day 4. NotchΔE-targeted cells formed perivascular cells within the frontonasal mesenchyme, and expressed a perivascular marker on the olfactory nerve. Hence, constitutively activating Notch1 is sufficient in vivo to drive not only somite cells, but also neural crest-derived frontonasal mesenchyme and perhaps developing OECs, to a perivascular cell fate. These results also highlight the plasticity of neural crest-derived mesenchyme and glia. Summary: Sustained Notch1 activation is sufficient to drive cranial neural crest-derived frontonasal mesenchyme to adopt a perivascular (mural) cell fate in developing chick embryos.
Collapse
Affiliation(s)
- Sophie R Miller
- Department of Physiology, Development and Neuroscience, University of Cambridge, Anatomy Building, Downing Street, Cambridge CB2 3DY, UK
| | - Surangi N Perera
- Department of Physiology, Development and Neuroscience, University of Cambridge, Anatomy Building, Downing Street, Cambridge CB2 3DY, UK
| | - Clare V H Baker
- Department of Physiology, Development and Neuroscience, University of Cambridge, Anatomy Building, Downing Street, Cambridge CB2 3DY, UK
| |
Collapse
|
120
|
Soto J, Monje PV. Axon contact-driven Schwann cell dedifferentiation. Glia 2017; 65:864-882. [PMID: 28233923 DOI: 10.1002/glia.23131] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 01/24/2017] [Accepted: 02/02/2017] [Indexed: 12/26/2022]
Abstract
Mature Schwann cells (SCs) retain dedifferentiation potential throughout adulthood. Still, how dedifferentiation occurs remains uncertain. Results from a variety of cell-based assays using in vitro cultured cAMP-differentiated and myelinating SCs revealed the existence of a novel dedifferentiating activity expressed on the surface of dorsal root ganglion (DRG) axons. This activity had the capacity to prevent SC differentiation and elicit dedifferentiation through direct SC-axon contact. Evidence is provided showing that a rapid loss of myelinating SC markers concomitant to proliferation occurred even in the presence of elevated cAMP, a signal that is required to drive and maintain a differentiated state. The dedifferentiating activity was a membrane-bound protein found exclusively in DRG neurons, as judged by its subcellular partitioning, sensitivity to proteolytic degradation and cell-type specificity, and remained active even after disruption of cellular organization. It differed from the membrane-anchored neuregulin-1 isoforms that are responsible for axon contact-induced SC proliferation and exerted its action independently of mitogenic signaling emanating from receptor tyrosine kinases and mitogen-activated protein kinases such as ERK and JNK. Interestingly, dedifferentiation occurred without concomitant changes in the expression of Krox-20, a transcriptional enhancer of myelination, and c-Jun, an inhibitor of myelination. In sum, our data indicated the existence of cell surface axon-derived signals that override pro-differentiating cues, drive dedifferentiation and allow SCs to proliferate in response to axonal mitogens. This axonal signal may negatively regulate myelination at the onset or reversal of the differentiated state. GLIA 2017;65:851-863.
Collapse
Affiliation(s)
- Jennifer Soto
- Miami Project to Cure Paralysis and Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, 33136
| | - Paula V Monje
- Miami Project to Cure Paralysis and Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, 33136
| |
Collapse
|
121
|
Boerboom A, Dion V, Chariot A, Franzen R. Molecular Mechanisms Involved in Schwann Cell Plasticity. Front Mol Neurosci 2017; 10:38. [PMID: 28261057 PMCID: PMC5314106 DOI: 10.3389/fnmol.2017.00038] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2016] [Accepted: 01/31/2017] [Indexed: 01/09/2023] Open
Abstract
Schwann cell incredible plasticity is a hallmark of the utmost importance following nerve damage or in demyelinating neuropathies. After injury, Schwann cells undergo dedifferentiation before redifferentiating to promote nerve regeneration and complete functional recovery. This review updates and discusses the molecular mechanisms involved in the negative regulation of myelination as well as in the reprogramming of Schwann cells taking place early following nerve lesion to support repair. Significant advance has been made on signaling pathways and molecular components that regulate SC regenerative properties. These include for instance transcriptional regulators such as c-Jun or Notch, the MAPK and the Nrg1/ErbB2/3 pathways. This comprehensive overview ends with some therapeutical applications targeting factors that control Schwann cell plasticity and highlights the need to carefully modulate and balance this capacity to drive nerve repair.
Collapse
Affiliation(s)
| | - Valérie Dion
- GIGA-Neurosciences, University of Liège Liège, Belgium
| | - Alain Chariot
- GIGA-Molecular Biology of Diseases, University of LiègeLiège, Belgium; Walloon Excellence in Lifesciences and Biotechnology (WELBIO)Wavre, Belgium
| | | |
Collapse
|
122
|
Brinkmann BG, Quintes S. Zeb2: Inhibiting the inhibitors in Schwann cells. NEUROGENESIS 2017; 4:e1271495. [PMID: 28203609 DOI: 10.1080/23262133.2016.1271495] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 12/02/2016] [Accepted: 12/06/2016] [Indexed: 10/20/2022]
Abstract
Development of Schwann cells is tightly regulated by concerted action of activating and inhibiting factors. Most of the regulatory feedback loops identified to date are transcriptional activators promoting induction of genes coding for integral myelin proteins and lipids. The mechanisms by which inhibitory factors are silenced during Schwann cell maturation are less well understood. We could recently show a pivotal function for the transcription factor zinc finger E-box binding homeobox 2 (Zeb2) during Schwann cell development and myelination as a transcriptional repressor of maturation inhibitors. Zeb2 belongs to a family of highly conserved 2-handed zinc-finger proteins and represses gene transcription by binding to E-box sequences in the regulatory region of target genes. The protein is known to repress E-cadherin during epithelial to mesenchymal transition (EMT) in tumor malignancy and mediates its functions by interacting with multiple co-factors. During nervous system development, Zeb2 is expressed in neural crest cells, the precursors of Schwann cells, the myelinating glial cells of peripheral nerves. Schwann cells lacking Zeb2 fail to fully differentiate and are unable to sort and myelinate peripheral nerve axons. The maturation inhibitors Sox2, Ednrb and Hey2 emerge as targets for Zeb2-mediated transcriptional repression and show persistent aberrant expression in Zeb2-deficient Schwann cells. While dispensible for adult Schwann cells, re-activation of Zeb2 is essential after nerve injury to allow remyelination and functional recovery. In summary, Zeb2 emerges as an "inhibitor of inhibitors," a novel concept in Schwann cell development and nerve repair.
Collapse
Affiliation(s)
- Bastian G Brinkmann
- Max Planck Institute of Experimental Medicine, Department of Neurogenetics , Göttingen, Germany
| | - Susanne Quintes
- Max Planck Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany; University Medical Center Göttingen (UMG), Department of Clinical Neurophysiology, Göttingen, Germany
| |
Collapse
|
123
|
Lackington WA, Ryan AJ, O'Brien FJ. Advances in Nerve Guidance Conduit-Based Therapeutics for Peripheral Nerve Repair. ACS Biomater Sci Eng 2017; 3:1221-1235. [PMID: 33440511 DOI: 10.1021/acsbiomaterials.6b00500] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Peripheral nerve injuries have high incidence rates, limited treatment options and poor clinical outcomes, rendering a significant socioeconomic burden. For effective peripheral nerve repair, the gap or site of injury must be structurally bridged to promote correct reinnervation and functional regeneration. However, effective repair becomes progressively more difficult with larger gaps. Autologous nerve grafting remains the best clinical option for the repair of large gaps (20-80 mm) despite being associated with numerous limitations including permanent donor site morbidity, a lack of available tissue and the formation of neuromas. To meet the clinical demand of large gap repair and overcome these limitations, tissue engineering has led to the development of nerve guidance conduit-based therapeutics. This review focuses on the advances of nerve guidance conduit-based therapeutics in terms of their structural properties including biomimetic composition, permeability, architecture, and surface modifications. Associated biochemical properties, pertaining to the incorporation of cells and neurotrophic factors, are also reviewed. After reviewing the progress in the field, we conclude by presenting an outlook on their clinical translatability and the next generation of therapeutics.
Collapse
Affiliation(s)
- William A Lackington
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons in Ireland, Dublin 2, Ireland.,Trinity Centre for Bioengineering (TCBE), Trinity College Dublin, Dublin 2, Ireland
| | - Alan J Ryan
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons in Ireland, Dublin 2, Ireland.,Trinity Centre for Bioengineering (TCBE), Trinity College Dublin, Dublin 2, Ireland
| | - Fergal J O'Brien
- Tissue Engineering Research Group (TERG), Department of Anatomy, Royal College of Surgeons in Ireland, Dublin 2, Ireland.,Advanced Materials and Bioengineering Research Centre (AMBER), Trinity College Dublin and Royal College of Surgeons in Ireland, Dublin 2, Ireland.,Trinity Centre for Bioengineering (TCBE), Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
124
|
Abstract
Axonal degeneration is a pivotal feature of many neurodegenerative conditions and substantially accounts for neurological morbidity. A widely used experimental model to study the mechanisms of axonal degeneration is Wallerian degeneration (WD), which occurs after acute axonal injury. In the peripheral nervous system (PNS), WD is characterized by swift dismantling and clearance of injured axons with their myelin sheaths. This is a prerequisite for successful axonal regeneration. In the central nervous system (CNS), WD is much slower, which significantly contributes to failed axonal regeneration. Although it is well-documented that Schwann cells (SCs) have a critical role in the regenerative potential of the PNS, to date we have only scarce knowledge as to how SCs ‘sense’ axonal injury and immediately respond to it. In this regard, it remains unknown as to whether SCs play the role of a passive bystander or an active director during the execution of the highly orchestrated disintegration program of axons. Older reports, together with more recent studies, suggest that SCs mount dynamic injury responses minutes after axonal injury, long before axonal breakdown occurs. The swift SC response to axonal injury could play either a pro-degenerative role, or alternatively a supportive role, to the integrity of distressed axons that have not yet committed to degenerate. Indeed, supporting the latter concept, recent findings in a chronic PNS neurodegeneration model indicate that deactivation of a key molecule promoting SC injury responses exacerbates axonal loss. If this holds true in a broader spectrum of conditions, it may provide the grounds for the development of new glia-centric therapeutic approaches to counteract axonal loss.
Collapse
Affiliation(s)
- Keit Men Wong
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Elisabetta Babetto
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.,Department of Pharmacology and Toxicology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Bogdan Beirowski
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
125
|
Quintes S, Brinkmann BG. Transcriptional inhibition in Schwann cell development and nerve regeneration. Neural Regen Res 2017; 12:1241-1246. [PMID: 28966633 PMCID: PMC5607813 DOI: 10.4103/1673-5374.213537] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Schwann cells, the myelinating glial cells of the peripheral nervous system are remarkably plastic after nerve trauma. Their transdifferentiation into specialized repair cells after injury shares some features with their development from the neural crest. Both processes are governed by a tightly regulated balance between activators and inhibitors to ensure timely lineage progression and allow re-maturation after nerve injury. Functional recovery after injury is very successful in rodents, however, in humans, lack of regeneration after nerve trauma and loss of function as the result of peripheral neuropathies represents a significant problem. Our understanding of the basic molecular machinery underlying Schwann cell maturation and plasticity has made significant progress in recent years and novel players have been discovered. While the transcriptional activators of Schwann cell development and nerve repair have been well defined, the mechanisms counteracting negative regulation of (re-)myelination are less well understood. Recently, transcriptional inhibition has emerged as a new regulatory mechanism in Schwann cell development and nerve repair. This mini-review summarizes some of the regulatory mechanisms controlling both processes and the novel concept of “inhibiting the inhibitors” in the context of Schwann cell plasticity.
Collapse
Affiliation(s)
- Susanne Quintes
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany.,Department of Clinical Neurophysiology, University Medical Center Göttingen (UMG), Göttingen, Germany
| | - Bastian G Brinkmann
- Max-Planck-Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany
| |
Collapse
|
126
|
Castelnovo LF, Bonalume V, Melfi S, Ballabio M, Colleoni D, Magnaghi V. Schwann cell development, maturation and regeneration: a focus on classic and emerging intracellular signaling pathways. Neural Regen Res 2017; 12:1013-1023. [PMID: 28852375 PMCID: PMC5558472 DOI: 10.4103/1673-5374.211172] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The development, maturation and regeneration of Schwann cells (SCs), the main glial cells of the peripheral nervous system, require the coordinate and complementary interaction among several factors, signals and intracellular pathways. These regulatory molecules consist of integrins, neuregulins, growth factors, hormones, neurotransmitters, as well as entire intracellular pathways including protein-kinase A, C, Akt, Erk/MAPK, Hippo, mTOR, etc. For instance, Hippo pathway is overall involved in proliferation, apoptosis, regeneration and organ size control, being crucial in cancer proliferation process. In SCs, Hippo is linked to merlin and YAP/TAZ signaling and it seems to respond to mechanic/physical challenges. Recently, among factors regulating SCs, also the signaling intermediates Src tyrosine kinase and focal adhesion kinase (FAK) proved relevant for SC fate, participating in the regulation of adhesion, motility, migration and in vitro myelination. In SCs, the factors Src and FAK are regulated by the neuroactive steroid allopregnanolone, thus corroborating the importance of this steroid in the control of SC maturation. In this review, we illustrate some old and novel signaling pathways modulating SC biology and functions during the different developmental, mature and regenerative states.
Collapse
Affiliation(s)
- Luca Franco Castelnovo
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Veronica Bonalume
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Simona Melfi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Marinella Ballabio
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Deborah Colleoni
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| | - Valerio Magnaghi
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
127
|
Zhang SJ, Wu WL, Yang KY, Chen YZ, Liu HC. Phenotypic changes of Schwann cells on the proximal stump of injured peripheral nerve during repair using small gap conduit tube. Neural Regen Res 2017; 12:1538-1543. [PMID: 29090001 PMCID: PMC5649476 DOI: 10.4103/1673-5374.215266] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Dedifferentiation of Schwann cells is an important feature of the response to peripheral nerve injury and specific negative myelination regulators are considered to have a major role in this process. However, most experiments have focused on the distal nerve stump, where the Notch signaling pathway is strongly associated with Schwann cell dedifferentiation and repair of the nerve. We observed the phenotypic changes of Schwann cells and changes of active Notch signaling on the proximal stump during peripheral nerve repair using small gap conduit tubulization. Eighty rats, with right sciatic nerve section of 4 mm, were randomly assigned to conduit bridging group and control group (epineurium suture). Glial fibrillary acidic protein expression, in myelinating Schwann cells on the proximal stump, began to up-regulate at 1 day after injury and was still evident at 5 days. Compared with the control group, Notch1 mRNA was expressed at a higher level in the conduit bridging group during the first week on the proximal stump. Hes1 mRNA levels in the conduit bridging group significantly increased compared with the control group at 3, 5, 7 and 14 days post-surgery. The change of the Notch intracellular domain shared a similar trend as Hes1 mRNA expression. Our results confirmed that phenotypic changes of Schwann cells occurred in the proximal stump. The differences in these changes between the conduit tubulization and epineurium suture groups correlate with changes in Notch signaling. This suggests that active Notch signaling might be a key mechanism during the early stage of neural regeneration in the proximal nerve stump.
Collapse
Affiliation(s)
- Shi-Jun Zhang
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Wen-Liang Wu
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Kai-Yun Yang
- Laboratory of Ears, Nose, and Throat, Oral and Maxillofacial Surgery, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Yun-Zhen Chen
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| | - Hai-Chun Liu
- Department of Orthopedics, Qilu Hospital of Shandong University, Jinan, Shandong Province, China
| |
Collapse
|
128
|
Fontenas L, De Santis F, Di Donato V, Degerny C, Chambraud B, Del Bene F, Tawk M. Neuronal Ndrg4 Is Essential for Nodes of Ranvier Organization in Zebrafish. PLoS Genet 2016; 12:e1006459. [PMID: 27902705 PMCID: PMC5130175 DOI: 10.1371/journal.pgen.1006459] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 11/03/2016] [Indexed: 11/19/2022] Open
Abstract
Axon ensheathment by specialized glial cells is an important process for fast propagation of action potentials. The rapid electrical conduction along myelinated axons is mainly due to its saltatory nature characterized by the accumulation of ion channels at the nodes of Ranvier. However, how these ion channels are transported and anchored along axons is not fully understood. We have identified N-myc downstream-regulated gene 4, ndrg4, as a novel factor that regulates sodium channel clustering in zebrafish. Analysis of chimeric larvae indicates that ndrg4 functions autonomously within neurons for sodium channel clustering at the nodes. Molecular analysis of ndrg4 mutants shows that expression of snap25 and nsf are sharply decreased, revealing a role of ndrg4 in controlling vesicle exocytosis. This uncovers a previously unknown function of ndrg4 in regulating vesicle docking and nodes of Ranvier organization, at least through its ability to finely tune the expression of the t-SNARE/NSF machinery. Myelination is an important process that enables fast propagation of action potential along the axons. Schwann cells (SCs) are the specialized glial cells that ensure the ensheathment of the corresponding axons in the Peripheral Nervous System. In order to do so, SCs and axons need to communicate to organize the myelinating segments and the clustering of sodium channels at the nodes of Ranvier. We have investigated the early events of myelination in the zebrafish embryo. We here identify ndrg4 as a novel neuronal factor essential for sodium channel clustering at the nodes. Immuno-labeling analysis show defective vesicle patterning along the axons of ndrg4 mutants, while timelapse experiments monitoring the presence and the transport of these vesicles reveal a normal behavior. Molecular analysis unravels a novel function of ndrg4 in controlling the expression of the t-SNARE/NSF machinery required for vesicle docking and release. However, inhibiting specifically regulated synaptic vesicle release does not lead to sodium channel clustering defects. We thus propose that ndrg4 can regulate this process, at least partially, through its ability to regulate the expression of key components of the t-SNARE/NSF machinery, responsible for clustering of sodium channels along myelinated axons.
Collapse
Affiliation(s)
- Laura Fontenas
- U1195, Inserm, University Paris Sud, University Paris-Saclay, Kremlin-Bicêtre, France
| | | | | | - Cindy Degerny
- U1195, Inserm, University Paris Sud, University Paris-Saclay, Kremlin-Bicêtre, France
| | - Béatrice Chambraud
- U1195, Inserm, University Paris Sud, University Paris-Saclay, Kremlin-Bicêtre, France
| | | | - Marcel Tawk
- U1195, Inserm, University Paris Sud, University Paris-Saclay, Kremlin-Bicêtre, France
- * E-mail:
| |
Collapse
|
129
|
Schwann cells: a new player in the tumor microenvironment. Cancer Immunol Immunother 2016; 66:959-968. [PMID: 27885383 DOI: 10.1007/s00262-016-1929-z] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 11/14/2016] [Indexed: 02/07/2023]
Abstract
Cancerous cells must cooperate with the surrounding stroma and non-malignant cells within the microenvironment to support the growth and invasion of the tumor. The nervous system is a component of every organ system of the body, and therefore, is invariably at the front line of the tumor invasion. Due to the complexity of the nervous system physiology, this review separately discusses the contributions of the central and peripheral nervous systems to the tumorigenesis and tumor progression. We further focus the discussion on the evidence that Schwann cells aid in tumor growth and invasion. Schwann cells, a largely unexplored element of the tumor microenvironment, may participate in the creation of tumor-favorable conditions through both bi-directional interaction with cancer cells and the facilitation of the immune-suppressive microenvironment through the mechanism of neural repair and immunomodulation.
Collapse
|
130
|
Gennarini G, Bizzoca A, Picocci S, Puzzo D, Corsi P, Furley AJW. The role of Gpi-anchored axonal glycoproteins in neural development and neurological disorders. Mol Cell Neurosci 2016; 81:49-63. [PMID: 27871938 DOI: 10.1016/j.mcn.2016.11.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 11/10/2016] [Accepted: 11/14/2016] [Indexed: 01/06/2023] Open
Abstract
This review article focuses on the Contactin (CNTN) subset of the Immunoglobulin supergene family (IgC2/FNIII molecules), whose components share structural properties (the association of Immunoglobulin type C2 with Fibronectin type III domains), as well as a general role in cell contact formation and axonal growth control. IgC2/FNIII molecules include 6 highly related components (CNTN 1-6), associated with the cell membrane via a Glycosyl Phosphatidyl Inositol (GPI)-containing lipid tail. Contactin 1 and Contactin 2 share ~50 (49.38)% identity at the aminoacid level. They are components of the cell surface, from which they may be released in soluble forms. They bind heterophilically to multiple partners in cis and in trans, including members of the related L1CAM family and of the Neurexin family Contactin-associated proteins (CNTNAPs or Casprs). Such interactions are important for organising the neuronal membrane, as well as for modulating the growth and pathfinding of axon tracts. In addition, they also mediate the functional maturation of axons by promoting their interactions with myelinating cells at the nodal, paranodal and juxtaparanodal regions. Such interactions also mediate differential ionic channels (both Na+ and K+) distribution, which is of critical relevance in the generation of the peak-shaped action potential. Indeed, thanks to their interactions with Ankyrin G, Na+ channels map within the nodal regions, where they drive axonal depolarization. However, no ionic channels are found in the flanking Contactin1-containing paranodal regions, where CNTN1 interactions with Caspr1 and with the Ig superfamily component Neurofascin 155 in cis and in trans, respectively, build a molecular barrier between the node and the juxtaparanode. In this region K+ channels are clustered, depending upon molecular interactions with Contactin 2 and with Caspr2. In addition to these functions, the Contactins appear to have also a role in degenerative and inflammatory disorders: indeed Contactin 2 is involved in neurodegenerative disorders with a special reference to the Alzheimer disease, given its ability to work as a ligand of the Alzheimer Precursor Protein (APP), which results in increased Alzheimer Intracellular Domain (AICD) release in a γ-secretase-dependent manner. On the other hand Contactin 1 drives Notch signalling activation via the Hes pathway, which could be consistent with its ability to modulate neuroinflammation events, and with the possibility that Contactin 1-dependent interactions may participate to the pathogenesis of the Multiple Sclerosis and of other inflammatory disorders.
Collapse
Affiliation(s)
- Gianfranco Gennarini
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy.
| | - Antonella Bizzoca
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy
| | - Sabrina Picocci
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy
| | - Daniela Puzzo
- Department of Biomedical and Biotechnological Sciences, University of Catania, Italy
| | - Patrizia Corsi
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, Medical School, University of Bari Policlinico. Piazza Giulio Cesare. I-70124 Bari, Italy
| | - Andrew J W Furley
- Department of Biomedical Science, University of Sheffield, Western Bank, Sheffield S10 2NT, UK
| |
Collapse
|
131
|
Gopinath C, Law WD, Rodríguez-Molina JF, Prasad AB, Song L, Crawford GE, Mullikin JC, Svaren J, Antonellis A. Stringent comparative sequence analysis reveals SOX10 as a putative inhibitor of glial cell differentiation. BMC Genomics 2016; 17:887. [PMID: 27821050 PMCID: PMC5100263 DOI: 10.1186/s12864-016-3167-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 10/18/2016] [Indexed: 01/22/2023] Open
Abstract
Background The transcription factor SOX10 is essential for all stages of Schwann cell development including myelination. SOX10 cooperates with other transcription factors to activate the expression of key myelin genes in Schwann cells and is therefore a context-dependent, pro-myelination transcription factor. As such, the identification of genes regulated by SOX10 will provide insight into Schwann cell biology and related diseases. While genome-wide studies have successfully revealed SOX10 target genes, these efforts mainly focused on myelinating stages of Schwann cell development. We propose that less-biased approaches will reveal novel functions of SOX10 outside of myelination. Results We developed a stringent, computational-based screen for genome-wide identification of SOX10 response elements. Experimental validation of a pilot set of predicted binding sites in multiple systems revealed that SOX10 directly regulates a previously unreported alternative promoter at SOX6, which encodes a transcription factor that inhibits glial cell differentiation. We further explored the utility of our computational approach by combining it with DNase-seq analysis in cultured Schwann cells and previously published SOX10 ChIP-seq data from rat sciatic nerve. Remarkably, this analysis enriched for genomic segments that map to loci involved in the negative regulation of gliogenesis including SOX5, SOX6, NOTCH1, HMGA2, HES1, MYCN, ID4, and ID2. Functional studies in Schwann cells revealed that: (1) all eight loci are expressed prior to myelination and down-regulated subsequent to myelination; (2) seven of the eight loci harbor validated SOX10 binding sites; and (3) seven of the eight loci are down-regulated upon repressing SOX10 function. Conclusions Our computational strategy revealed a putative novel function for SOX10 in Schwann cells, which suggests a model where SOX10 activates the expression of genes that inhibit myelination during non-myelinating stages of Schwann cell development. Importantly, the computational and functional datasets we present here will be valuable for the study of transcriptional regulation, SOX protein function, and glial cell biology. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3167-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chetna Gopinath
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - William D Law
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - José F Rodríguez-Molina
- Cellular and Molecular Pathology Program, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Arjun B Prasad
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lingyun Song
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Gregory E Crawford
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA.,Department of Pediatrics, Duke University Medical Center, Durham, NC, 27708, USA
| | - James C Mullikin
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - John Svaren
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53706, USA.,Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Anthony Antonellis
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA. .,Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA. .,Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
132
|
Altmann C, Vasic V, Hardt S, Heidler J, Häussler A, Wittig I, Schmidt MHH, Tegeder I. Progranulin promotes peripheral nerve regeneration and reinnervation: role of notch signaling. Mol Neurodegener 2016; 11:69. [PMID: 27770818 PMCID: PMC5075406 DOI: 10.1186/s13024-016-0132-1] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 09/28/2016] [Indexed: 01/11/2023] Open
Abstract
Background Peripheral nerve injury is a frequent cause of lasting motor deficits and chronic pain. Although peripheral nerves are capable of regrowth they often fail to re-innervate target tissues. Results Using newly generated transgenic mice with inducible neuronal progranulin overexpression we show that progranulin accelerates axonal regrowth, restoration of neuromuscular synapses and recovery of sensory and motor functions after injury of the sciatic nerve. Oppositely, progranulin deficient mice have long-lasting deficits in motor function tests after nerve injury due to enhanced losses of motor neurons and stronger microglia activation in the ventral horn of the spinal cord. Deep proteome and gene ontology (GO) enrichment analysis revealed that the proteins upregulated in progranulin overexpressing mice were involved in ‘regulation of transcription’ and ‘response to insulin’ (GO terms). Transcription factor prediction pointed to activation of Notch signaling and indeed, co-immunoprecipitation studies revealed that progranulin bound to the extracellular domain of Notch receptors, and this was functionally associated with higher expression of Notch target genes in the dorsal root ganglia of transgenic mice with neuronal progranulin overexpression. Functionally, these transgenic mice recovered normal gait and running, which was not achieved by controls and was stronger impaired in progranulin deficient mice. Conclusion We infer that progranulin activates Notch signaling pathways, enhancing thereby the regenerative capacity of partially injured neurons, which leads to improved motor function recovery. Graphical abstract ![]()
Electronic supplementary material The online version of this article (doi:10.1186/s13024-016-0132-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Christine Altmann
- Institute of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany
| | - Verica Vasic
- Molecular Signal Transduction Laboratories, Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Stefanie Hardt
- Institute of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany
| | - Juliana Heidler
- Functional Proteomics, SFB815 Core Unit, Goethe-University, Frankfurt, Germany
| | - Annett Häussler
- Institute of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany
| | - Ilka Wittig
- Functional Proteomics, SFB815 Core Unit, Goethe-University, Frankfurt, Germany
| | - Mirko H H Schmidt
- Molecular Signal Transduction Laboratories, Institute for Microscopic Anatomy and Neurobiology, Focus Program Translational Neuroscience (FTN), Rhine Main Neuroscience Network (rmn2), University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Goethe-University Hospital, Frankfurt, Germany.
| |
Collapse
|
133
|
Alemany S, Vilor-Tejedor N, Bustamante M, Pujol J, Macià D, Martínez-Vilavella G, Fenoll R, Alvárez-Pedrerol M, Forns J, Júlvez J, Suades-González E, Llop S, Rebagliato M, Sunyer J. A Genome-Wide Association Study of Attention Function in a Population-Based Sample of Children. PLoS One 2016; 11:e0163048. [PMID: 27656889 PMCID: PMC5033492 DOI: 10.1371/journal.pone.0163048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Accepted: 09/01/2016] [Indexed: 01/19/2023] Open
Abstract
Background Attention function filters and selects behaviorally relevant information. This capacity is impaired in some psychiatric disorders and has been proposed as an endophenotype for Attention-Deficit/Hyperactivity Disorder; however, its genetic basis remains largely unknown. This study aimed to identify single nucleotide polymorphism (SNPs) associated with attention function. Materials and Methods The discovery sample included 1655 children (7–12 years) and the replication sample included 546 children (5–8 years). Five attention outcomes were assessed using the computerized Attentional Network Test (ANT): alerting, orienting, executive attention, Hit Reaction time (HRT) and the standard error of HRT (HRTSE). A Genome-wide Association Study was conducted for each outcome. Gene set enrichment analyses were performed to detect biological pathways associated with attention outcomes. Additional neuroimaging analyses were conducted to test neural effects of detected SNPs of interest. Results Thirteen loci showed suggestive evidence of association with attention function (P<10−5) in the discovery sample. One of them, the rs4321351 located in the PID1 gene, was nominally significant in the replication sample although it did not survive multiple testing correction. Neuroimaging analysis revealed a significant association between this SNP and brain structure and function involving the frontal-basal ganglia circuits. The mTOR signaling and Alzheimer disease-amyloid secretase pathways were significantly enriched for alerting, orienting and HRT respectively (FDR<5%). Conclusion These results suggest for the first time the involvement of the PID1 gene, mTOR signaling and Alzheimer disease-amyloid secretase pathways, in attention function during childhood. These genes and pathways have been proposed to play a role in neuronal plasticity, memory and neurodegenerative disease.
Collapse
Affiliation(s)
- Silvia Alemany
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiology and Public Health (CIBERESP), Barcelona, Spain
- * E-mail:
| | - Natàlia Vilor-Tejedor
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiology and Public Health (CIBERESP), Barcelona, Spain
| | - Mariona Bustamante
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiology and Public Health (CIBERESP), Barcelona, Spain
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Jesús Pujol
- MRI Research Unit, Department of Radiology, Hospital del Mar, Barcelona, Spain
- Centro Investigación Biomédica en Red de Salud Mental, CIBERSAM G21, Barcelona, Spain
| | - Dídac Macià
- MRI Research Unit, Department of Radiology, Hospital del Mar, Barcelona, Spain
| | | | - Raquel Fenoll
- MRI Research Unit, Department of Radiology, Hospital del Mar, Barcelona, Spain
| | - Mar Alvárez-Pedrerol
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiology and Public Health (CIBERESP), Barcelona, Spain
| | - Joan Forns
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiology and Public Health (CIBERESP), Barcelona, Spain
- Department of Genes and Environment, Division of Epidemiology, Norwegian Institute of Public Health, Oslo, Norway
| | - Jordi Júlvez
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiology and Public Health (CIBERESP), Barcelona, Spain
| | - Elisabet Suades-González
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiology and Public Health (CIBERESP), Barcelona, Spain
- Learning Disabilities Unit (UTAE); Neuropediatrics Department, Hospital de Sant Joan de Déu, Universitat de Barcelona, Barcelona, Spain
| | - Sabrina Llop
- CIBER Epidemiology and Public Health (CIBERESP), Barcelona, Spain
- Epidemiology and Environmental Health Joint Research Unit, FISABIO−Universitat Jaume I−Universitat de València, Valencia, Spain
| | - Marisa Rebagliato
- CIBER Epidemiology and Public Health (CIBERESP), Barcelona, Spain
- Epidemiology and Environmental Health Joint Research Unit, FISABIO−Universitat Jaume I−Universitat de València, Valencia, Spain
- University Jaime I (UJI), Castellón, Spain
| | - Jordi Sunyer
- ISGlobal, Centre for Research in Environmental Epidemiology (CREAL), Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiology and Public Health (CIBERESP), Barcelona, Spain
- IMIM (Hospital del Mar Medical Research Institute), Barcelona, Spain
| |
Collapse
|
134
|
Miller SR, Perera SN, Benito C, Stott SRW, Baker CVH. Evidence for a Notch1-mediated transition during olfactory ensheathing cell development. J Anat 2016; 229:369-83. [PMID: 27271278 PMCID: PMC4974551 DOI: 10.1111/joa.12494] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2016] [Indexed: 01/19/2023] Open
Abstract
Olfactory ensheathing cells (OECs) are a unique glial population found in both the peripheral and central nervous system: they ensheath bundles of unmyelinated olfactory axons from their peripheral origin in the olfactory epithelium to their central synaptic targets in the glomerular layer of the olfactory bulb. Like all other peripheral glia (Schwann cells, satellite glia, enteric glia), OECs are derived from the embryonic neural crest. However, in contrast to Schwann cells, whose development has been extensively characterised, relatively little is known about their normal development in vivo. In the Schwann cell lineage, the transition from multipotent Schwann cell precursor to immature Schwann cell is promoted by canonical Notch signalling. Here, in situ hybridisation and immunohistochemistry data from chicken, mouse and human embryos are presented that suggest a canonical Notch-mediated transition also occurs during OEC development.
Collapse
Affiliation(s)
- Sophie R. Miller
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
- Present address: DanStemUniversity of Copenhagen3B BlegdamsvejDK‐2200Copenhagen NDenmark
| | - Surangi N. Perera
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| | - Cristina Benito
- Department of Cell and Developmental BiologyUniversity College LondonLondonUK
| | - Simon R. W. Stott
- John van Geest Centre for Brain RepairUniversity of CambridgeCambridgeUK
| | - Clare V. H. Baker
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeUK
| |
Collapse
|
135
|
Andersen ND, Srinivas S, Piñero G, Monje PV. A rapid and versatile method for the isolation, purification and cryogenic storage of Schwann cells from adult rodent nerves. Sci Rep 2016; 6:31781. [PMID: 27549422 PMCID: PMC4994039 DOI: 10.1038/srep31781] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 07/27/2016] [Indexed: 01/04/2023] Open
Abstract
We herein developed a protocol for the rapid procurement of adult nerve-derived Schwann cells (SCs) that was optimized to implement an immediate enzymatic dissociation of fresh nerve tissue while maintaining high cell viability, improving yields and minimizing fibroblast and myelin contamination. This protocol introduces: (1) an efficient method for enzymatic cell release immediately after removal of the epineurium and extensive teasing of the nerve fibers; (2) an adaptable drop-plating method for selective cell attachment, removal of myelin debris, and expansion of the initial SC population in chemically defined medium; (3) a magnetic-activated cell sorting purification protocol for rapid and effective fibroblast elimination; and (4) an optional step of cryopreservation for the storage of the excess of cells. Highly proliferative SC cultures devoid of myelin and fibroblast growth were obtained within three days of nerve processing. Characterization of the initial, expanded, and cryopreserved cell products confirmed maintenance of SC identity, viability and growth rates throughout the process. Most importantly, SCs retained their sensitivity to mitogens and potential for differentiation even after cryopreservation. To conclude, this easy-to-implement and clinically relevant protocol allows for the preparation of expandable homogeneous SC cultures while minimizing time, manipulation of the cells, and exposure to culture variables.
Collapse
Affiliation(s)
- Natalia D. Andersen
- The Miami Project to Cure Paralysis and Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Shruthi Srinivas
- The Miami Project to Cure Paralysis and Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| | - Gonzalo Piñero
- The Miami Project to Cure Paralysis and Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
- Universidad de Buenos Aires, CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Buenos Aires, Argentina
| | - Paula V. Monje
- The Miami Project to Cure Paralysis and Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida 33136, USA
| |
Collapse
|
136
|
Neal JW, Gasque P. The role of primary infection of Schwann cells in the aetiology of infective inflammatory neuropathies. J Infect 2016; 73:402-418. [PMID: 27546064 DOI: 10.1016/j.jinf.2016.08.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 08/10/2016] [Accepted: 08/11/2016] [Indexed: 02/08/2023]
Abstract
Numerous different pathogens are responsible for infective peripheral neuropathies and this is generally the result of the indirect effects of pathogen infection, namely anti pathogen antibodies cross reacting with epitopes on peripheral nerve, auto reactive T cells attacking myelin, circulating immune complexes and complement fixation. Primary infection of Schwann cells (SC) associated with peripheral nerve inflammation is rare requiring pathogens to cross the Blood Peripheral Nerve Barrier (BPNB) evade anti-pathogen innate immune pathways and invade the SC. Spirochetes Borrelia bourgdorferi and Trepomema pallidum are highly invasive, express surface lipo proteins, but despite this SC are rarely infected. However, Trypanosoma cruzi (Chaga's disease) and Mycobacterium leprae. Leprosy are two important causes of peripheral nerve infection and both demonstrate primary infection of SC. This is due to two novel strategies; T. cruzi express a trans-silalidase that mimics host neurotrophic factors and infects SC via tyrosine kinase receptors. M. leprae demonstrates multi receptor SC tropism and subsequent infection promotes nuclear reprogramming and dedifferentiation of host SC into progenitor stem like cells (pSLC) that are vulnerable to M. leprae infection. These two novel pathogen evasion strategies, involving stem cells and receptor mimicry, provide potential therapeutic targets relevant to the prevention of peripheral nerve inflammation by inhibiting primary SC infection.
Collapse
Affiliation(s)
- J W Neal
- Infection and Immunity, Henry Wellcome Building, Cardiff University, Cardiff CF14 4XN, United Kingdom.
| | - P Gasque
- Laboratoire d'Immunologie Clinique et Expérimentale de l'OI (LICE-OI), Centre recherche Immuno-clinique des agents pathogènes de l'OI (CRIC-AP OI) Pôle Biologie Santé, Hôpital Félix Guyon, CHU de la Réunion, Reunion.
| |
Collapse
|
137
|
Guha I, Slamova I, Chun S, Clegg A, Golos M, Thrasivoulou C, Simons JP, Al-Shawi R. The effects of short-term JNK inhibition on the survival and growth of aged sympathetic neurons. Neurobiol Aging 2016; 46:138-48. [PMID: 27490965 DOI: 10.1016/j.neurobiolaging.2016.06.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2016] [Revised: 05/30/2016] [Accepted: 06/24/2016] [Indexed: 11/27/2022]
Abstract
During the course of normal aging, certain populations of nerve growth factor (NGF)-responsive neurons become selectively vulnerable to cell death. Studies using dissociated neurons isolated from neonates have shown that c-Jun N-terminal kinases (JNKs) are important in regulating the survival and neurite outgrowth of NGF-responsive sympathetic neurons. Unlike neonatal neurons, adult sympathetic neurons are not dependent on NGF for their survival. Moreover, the NGF precursor, proNGF, is neurotoxic for aging but not young adult NGF-responsive neurons. Because of these age-related differences, the effects of JNK inhibition on the survival and growth of sympathetic neurons isolated from aged mice were studied. Aged neurons, as well as glia, were found to be dependent on JNK for their growth but not their survival. Conversely, proNGF neurotoxicity was JNK-dependent and mediated by the p75-interacting protein NRAGE, whereas neurite outgrowth was independent of NRAGE. These results have implications for the potential use of JNK inhibitors as therapies for ameliorating age-related neurodegenerative disease.
Collapse
Affiliation(s)
- Isa Guha
- Genetics Unit and Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, Royal Free Campus, London, UK
| | - Ivana Slamova
- Genetics Unit and Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, Royal Free Campus, London, UK
| | - Soyon Chun
- Genetics Unit and Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, Royal Free Campus, London, UK
| | - Arthur Clegg
- Genetics Unit and Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, Royal Free Campus, London, UK
| | - Michal Golos
- Genetics Unit and Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, Royal Free Campus, London, UK
| | - Chris Thrasivoulou
- Research Department of Cell and Developmental Biology, University College London, London, UK
| | - J Paul Simons
- Genetics Unit and Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, Royal Free Campus, London, UK.
| | - Raya Al-Shawi
- Genetics Unit and Wolfson Drug Discovery Unit, Centre for Amyloidosis and Acute Phase Proteins, Division of Medicine, University College London, Royal Free Campus, London, UK.
| |
Collapse
|
138
|
Kangas SM, Ohlmeier S, Sormunen R, Jouhilahti EM, Peltonen S, Peltonen J, Heape AM. An approach to comprehensive genome and proteome expression analyses in Schwann cells and neurons during peripheral nerve myelin formation. J Neurochem 2016; 138:830-44. [PMID: 27364987 DOI: 10.1111/jnc.13722] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Revised: 06/09/2016] [Accepted: 06/27/2016] [Indexed: 12/01/2022]
Abstract
Peripheral nerve myelination is a complex event resulting from spatially and temporally regulated reciprocal interactions between the neuron and myelin-forming Schwann cells. The dynamic process and the protein functional modules and networks that operate throughout the myelination process are poorly understood because of a lack of methodologies suitable for observing specific changes in the Schwann cell/neuron-unit. The identification of the precise roles for the proteins participating in the functional modules and networks that participate in the myelination process is hindered by the cellular and molecular complexity of the nervous tissue itself. We have developed an approach based on a myelinating dorsal root ganglion explant model that allows distinguishing clear, reproducible and predictable differences between the biochemical properties and the genomic and proteomic expression profiles of both cellular components of the Schwann cell/neuron unit at different stages of the myelination process. This model, derived from E13.5 C57BL/6J mouse embryos, is sufficiently robust for use in identifying the protein functional networks and modules related to peripheral nerve myelin formation. The genomic expression profiles of the selected neuronal, Schwann cell and myelin-specific proteins in the cultures reflect in vivo profiles reported in the literature, and the structural and ultrastructural properties of the myelin, as well as the myelination schedule of the cultures, closely resemble those observed in peripheral nerves in situ. The RNA expression data set is available through NCBI gene expression omnibus accession GSE60345. We have developed a reproducible and robust cell culture-based approach, accompanied by a genome-wide expression data set, which allows studying myelination in the peripheral nervous system at the proteomic and transcriptomic levels in Schwann cells and neurons. Myelinating dorsal root explant cultures, prepared from C57BL/6J mouse embryos, present distinct developmental stages comparable to those observed in a peripheral nerve in situ. This model can be used for identifying the protein functional networks and modules related to peripheral nerve myelin formation.
Collapse
Affiliation(s)
- Salla M Kangas
- Cancer and Translational Medicine Research Unit (Anatomy and Cell Biology), University of Oulu, Oulu, Finland.
| | - Steffen Ohlmeier
- Proteomics Core Facility, Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Raija Sormunen
- Biocenter Oulu and Departments of Pathology, University of Oulu, Oulu, Finland
| | - Eeva-Mari Jouhilahti
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Sirkku Peltonen
- Department of Dermatology, University of Turku and Turku University Hospital, Turku, Finland
| | - Juha Peltonen
- Department of Cell Biology and Anatomy, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Anthony M Heape
- Cancer and Translational Medicine Research Unit (Anatomy and Cell Biology), University of Oulu, Oulu, Finland.
| |
Collapse
|
139
|
Schwann cells–axon interaction in myelination. Curr Opin Neurobiol 2016; 39:24-9. [DOI: 10.1016/j.conb.2016.03.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 03/15/2016] [Accepted: 03/16/2016] [Indexed: 01/01/2023]
|
140
|
Quintes S, Brinkmann BG, Ebert M, Fröb F, Kungl T, Arlt FA, Tarabykin V, Huylebroeck D, Meijer D, Suter U, Wegner M, Sereda MW, Nave KA. Zeb2 is essential for Schwann cell differentiation, myelination and nerve repair. Nat Neurosci 2016; 19:1050-1059. [PMID: 27294512 PMCID: PMC4964942 DOI: 10.1038/nn.4321] [Citation(s) in RCA: 109] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 05/04/2016] [Indexed: 12/12/2022]
Abstract
Schwann cell development and peripheral nerve myelination require the serial expression of transcriptional activators, such as Sox10, Oct6 (also called Scip or Pou3f1) and Krox20 (also called Egr2). Here we show that transcriptional repression, mediated by the zinc-finger protein Zeb2 (also known as Sip1), is essential for differentiation and myelination. Mice lacking Zeb2 in Schwann cells develop a severe peripheral neuropathy, caused by failure of axonal sorting and virtual absence of myelin membranes. Zeb2-deficient Schwann cells continuously express repressors of lineage progression. Moreover, genes for negative regulators of maturation such as Sox2 and Ednrb emerge as Zeb2 target genes, supporting its function as an 'inhibitor of inhibitors' in myelination control. When Zeb2 is deleted in adult mice, Schwann cells readily dedifferentiate following peripheral nerve injury and become repair cells. However, nerve regeneration and remyelination are both perturbed, demonstrating that Zeb2, although undetectable in adult Schwann cells, has a latent function throughout life.
Collapse
Affiliation(s)
- Susanne Quintes
- Max Planck Institute of Experimental Medicine, Department of
Neurogenetics, Göttingen, Germany
- University Medical Center Göttingen (UMG), Department of
Clinical Neurophysiology, Göttingen, Germany
| | - Bastian G Brinkmann
- Max Planck Institute of Experimental Medicine, Department of
Neurogenetics, Göttingen, Germany
| | - Madlen Ebert
- Max Planck Institute of Experimental Medicine, Department of
Neurogenetics, Göttingen, Germany
| | - Franziska Fröb
- Institut für Biochemie, Emil-Fischer-Zentrum,
Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen,
Germany
| | - Theresa Kungl
- Max Planck Institute of Experimental Medicine, Department of
Neurogenetics, Göttingen, Germany
| | - Friederike A Arlt
- Max Planck Institute of Experimental Medicine, Department of
Neurogenetics, Göttingen, Germany
| | - Victor Tarabykin
- Institute for Cell and Neurobiology, Center for Anatomy,
Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Danny Huylebroeck
- Laboratory of Molecular Biology (Celgen), Department of Development
and Regeneration, KU Leuven, Leuven, Belgium
- Department of Cell Biology, Erasmus University Medical Center,
Rotterdam, The Netherlands
| | - Dies Meijer
- Centre for Neuroregeneration, University of Edinburgh, Edinburgh,
United Kingdom
| | - Ueli Suter
- Institute of Molecular Health Sciences, Department of Biology, ETH
Zürich, Zürich, Switzerland
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum,
Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen,
Germany
| | - Michael W Sereda
- Max Planck Institute of Experimental Medicine, Department of
Neurogenetics, Göttingen, Germany
- University Medical Center Göttingen (UMG), Department of
Clinical Neurophysiology, Göttingen, Germany
| | - Klaus-Armin Nave
- Max Planck Institute of Experimental Medicine, Department of
Neurogenetics, Göttingen, Germany
| |
Collapse
|
141
|
Elbaz B, Traka M, Kunjamma RB, Dukala D, Brosius Lutz A, Anton ES, Barres BA, Soliven B, Popko B. Adenomatous polyposis coli regulates radial axonal sorting and myelination in the PNS. Development 2016; 143:2356-66. [PMID: 27226321 PMCID: PMC4958326 DOI: 10.1242/dev.135913] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 05/09/2016] [Indexed: 11/20/2022]
Abstract
The tumor suppressor protein adenomatous polyposis coli (APC) is multifunctional - it participates in the canonical Wnt/β-catenin signal transduction pathway as well as modulating cytoskeleton function. Although APC is expressed by Schwann cells, the role that it plays in these cells and in the myelination of the peripheral nervous system (PNS) is unknown. Therefore, we used the Cre-lox approach to generate a mouse model in which APC expression is specifically eliminated from Schwann cells. These mice display hindlimb weakness and impaired axonal conduction in sciatic nerves. Detailed morphological analyses revealed that APC loss delays radial axonal sorting and PNS myelination. Furthermore, APC loss delays Schwann cell differentiation in vivo, which correlates with persistent activation of the Wnt signaling pathway and results in perturbed extension of Schwann cell processes and disrupted lamellipodia formation. In addition, APC-deficient Schwann cells display a transient diminution of proliferative capacity. Our data indicate that APC is required by Schwann cells for their timely differentiation to mature, myelinating cells and plays a crucial role in radial axonal sorting and PNS myelination.
Collapse
Affiliation(s)
- Benayahu Elbaz
- Department of Neurology, Center for Peripheral Neuropathy, University of Chicago, Chicago, IL 60637, USA
| | - Maria Traka
- Department of Neurology, Center for Peripheral Neuropathy, University of Chicago, Chicago, IL 60637, USA
| | - Rejani B Kunjamma
- Department of Neurology, Center for Peripheral Neuropathy, University of Chicago, Chicago, IL 60637, USA
| | - Danuta Dukala
- Department of Neurology, Center for Peripheral Neuropathy, University of Chicago, Chicago, IL 60637, USA
| | - Amanda Brosius Lutz
- Stanford University School of Medicine, Department of Neurobiology, Fairchild Building Room D235, 299 Campus Drive, Stanford, CA 94305-5125, USA
| | - E S Anton
- UNC Neuroscience Center and the Department of Cell and Molecular Physiology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Ben A Barres
- Stanford University School of Medicine, Department of Neurobiology, Fairchild Building Room D235, 299 Campus Drive, Stanford, CA 94305-5125, USA
| | - Betty Soliven
- Department of Neurology, Center for Peripheral Neuropathy, University of Chicago, Chicago, IL 60637, USA
| | - Brian Popko
- Department of Neurology, Center for Peripheral Neuropathy, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
142
|
Zeb2 recruits HDAC-NuRD to inhibit Notch and controls Schwann cell differentiation and remyelination. Nat Neurosci 2016; 19:1060-72. [PMID: 27294509 PMCID: PMC4961522 DOI: 10.1038/nn.4322] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 05/10/2016] [Indexed: 12/12/2022]
Abstract
The mechanisms that coordinate and balance a complex network of opposing regulators to control Schwann cell (SC) differentiation remain elusive. Here we demonstrate that zinc-finger E-box binding-homeobox 2 (Zeb2/Sip1) transcription factor is a critical intrinsic timer that controls the onset of Schwann cell (SC) differentiation by recruiting HDAC1/2-NuRD co-repressor complexes. Zeb2 deletion arrests SCs at an undifferentiated state during peripheral nerve development and inhibits remyelination after injury. Zeb2 antagonizes inhibitory effectors including Notch and Sox2. Importantly, genome-wide transcriptome analysis reveals a Zeb2 target gene, encoding the Notch effector Hey2, as a potent inhibitor for SC differentiation. Strikingly, a genetic Zeb2 variant, which is associated with Mowat-Wilson syndrome, disrupts the interaction with HDAC1/2-NuRD and abolishes Zeb2 activity for SC differentiation. Therefore, Zeb2 controls SC maturation by recruiting HDAC1/2-NuRD complexes and inhibiting a novel Notch-Hey2 signaling axis, pointing to the critical role of HDAC1/2-NuRD activity in peripheral neuropathies caused by ZEB2 mutations.
Collapse
|
143
|
Glu-tubulin is a marker for Schwann cells and can distinguish between schwannomas and neurofibromas. Histochem Cell Biol 2016; 146:467-77. [PMID: 27278446 DOI: 10.1007/s00418-016-1455-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/25/2016] [Indexed: 10/21/2022]
Abstract
Schwann cells generate myelin sheaths around the axons of the peripheral nervous system, thus facilitating efficient nerve impulse propagation. Two main tumor types can arise from peripheral nerves, schwannomas and neurofibromas, which are sometimes difficult to distinguish and may require the use of diagnostic biomarkers. Here, we characterize a new marker for Schwann cells and its potential use as a diagnostic marker for schwannomas. Immunohistochemistry for Glu-tubulin, a posttranslational modification of α-tubulin, was performed in mouse and human tissues. This technique labels Schwann cells but not oligodendrocytes. All peripheral nerves were immunoreactive for this antibody, including large nerve trunks, thin myelinated nerves, as well as the myenteric and submucous plexus of the digestive tract. In the mouse brain, many neurons were immunoreactive for Glu-tubulin but oligodendrocytes were negative. During embryo development, immunoreactive nerves were already found at E10. In Schwann cells, the staining is restricted to the myelin sheaths and is not present in the perinuclear cytoplasm or the Ranvier nodes. Primary cultures of fibroblasts and Schwann cells were established from mouse sciatic nerves, and Western blot analysis showed that Glu-tubulin immunoreactivity was found in the Schwann cells but not in the fibroblasts. Clinical specimens of schwannomas (n = 20) and neurofibromas (n = 20) were stained with anti-Glu-tubulin antibodies. Schwannomas presented a strong staining in all tumor cells, whereas neurofibromas had a light speckled staining pattern, easily distinguishable from the one found in schwannomas. In conclusion, Glu-tubulin can be used as a marker of Schwann cells and can help in diagnosing peripheral nerve tumors.
Collapse
|
144
|
Rao SNR, Pearse DD. Regulating Axonal Responses to Injury: The Intersection between Signaling Pathways Involved in Axon Myelination and The Inhibition of Axon Regeneration. Front Mol Neurosci 2016; 9:33. [PMID: 27375427 PMCID: PMC4896923 DOI: 10.3389/fnmol.2016.00033] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 05/02/2016] [Indexed: 01/06/2023] Open
Abstract
Following spinal cord injury (SCI), a multitude of intrinsic and extrinsic factors adversely affect the gene programs that govern the expression of regeneration-associated genes (RAGs) and the production of a diversity of extracellular matrix molecules (ECM). Insufficient RAG expression in the injured neuron and the presence of inhibitory ECM at the lesion, leads to structural alterations in the axon that perturb the growth machinery, or form an extraneous barrier to axonal regeneration, respectively. Here, the role of myelin, both intact and debris, in antagonizing axon regeneration has been the focus of numerous investigations. These studies have employed antagonizing antibodies and knockout animals to examine how the growth cone of the re-growing axon responds to the presence of myelin and myelin-associated inhibitors (MAIs) within the lesion environment and caudal spinal cord. However, less attention has been placed on how the myelination of the axon after SCI, whether by endogenous glia or exogenously implanted glia, may alter axon regeneration. Here, we examine the intersection between intracellular signaling pathways in neurons and glia that are involved in axon myelination and axon growth, to provide greater insight into how interrogating this complex network of molecular interactions may lead to new therapeutics targeting SCI.
Collapse
Affiliation(s)
- Sudheendra N R Rao
- The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine Miami, FL, USA
| | - Damien D Pearse
- The Miami Project to Cure Paralysis, University of Miami Miller School of MedicineMiami, FL, USA; The Department of Neurological Surgery, University of Miami Miller School of MedicineMiami, FL, USA; The Neuroscience Program, University of Miami Miller School of MedicineMiami, FL, USA; The Interdisciplinary Stem Cell Institute, University of Miami Miller School of MedicineMiami, FL, USA; Bruce W. Carter Department of Veterans Affairs Medical CenterMiami, FL, USA
| |
Collapse
|
145
|
Balakrishnan A, Stykel MG, Touahri Y, Stratton JA, Biernaskie J, Schuurmans C. Temporal Analysis of Gene Expression in the Murine Schwann Cell Lineage and the Acutely Injured Postnatal Nerve. PLoS One 2016; 11:e0153256. [PMID: 27058953 PMCID: PMC4826002 DOI: 10.1371/journal.pone.0153256] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Accepted: 03/26/2016] [Indexed: 01/09/2023] Open
Abstract
Schwann cells (SCs) arise from neural crest cells (NCCs) that first give rise to SC precursors (SCPs), followed by immature SCs, pro-myelinating SCs, and finally, non-myelinating or myelinating SCs. After nerve injury, mature SCs ‘de-differentiate’, downregulating their myelination program while transiently re-activating early glial lineage genes. To better understand molecular parallels between developing and de-differentiated SCs, we characterized the expression profiles of a panel of 12 transcription factors from the onset of NCC migration through postnatal stages, as well as after acute nerve injury. Using Sox10 as a pan-glial marker in co-expression studies, the earliest transcription factors expressed in E9.0 Sox10+ NCCs were Sox9, Pax3, AP2α and Nfatc4. E10.5 Sox10+ NCCs coalescing in the dorsal root ganglia differed slightly, expressing Sox9, Pax3, AP2α and Etv5. E12.5 SCPs continued to express Sox10, Sox9, AP2α and Pax3, as well as initiating Sox2 and Egr1 expression. E14.5 immature SCs were similar to SCPs, except that they lost Pax3 expression. By E18.5, AP2α, Sox2 and Egr1 expression was turned off in the nerve, while Jun, Oct6 and Yy1 expression was initiated in pro-myelinating Sox9+/Sox10+ SCs. Early postnatal and adult SCs continued to express Sox9, Jun, Oct6 and Yy1 and initiated Nfatc4 and Egr2 expression. Notably, at all stages, expression of each marker was observed only in a subset of Sox10+ SCs, highlighting the heterogeneity of the SC pool. Following acute nerve injury, Egr1, Jun, Oct6, and Sox2 expression was upregulated, Egr2 expression was downregulated, while Sox9, Yy1, and Nfatc4 expression was maintained at similar frequencies. Notably, de-differentiated SCs in the injured nerve did not display a transcription factor profile corresponding to a specific stage in the SC lineage. Taken together, we demonstrate that uninjured and injured SCs are heterogeneous and distinct from one another, and de-differentiation recapitulates transcriptional aspects of several different embryonic stages.
Collapse
Affiliation(s)
- Anjali Balakrishnan
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children’s Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Morgan G. Stykel
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, Alberta Children’s Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Yacine Touahri
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children’s Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, Alberta Children’s Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Jo Anne Stratton
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, Alberta Children’s Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
- Department of Clinical Neurosciences, Cumming School of Medicine, Alberta Children’s Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Jeff Biernaskie
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, Alberta Children’s Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
- * E-mail: (CS); (JB)
| | - Carol Schuurmans
- Department of Biochemistry and Molecular Biology, Cumming School of Medicine, Alberta Children’s Hospital Research Institute and Hotchkiss Brain Institute, University of Calgary, Calgary, AB, T2N 4N1, Canada
- * E-mail: (CS); (JB)
| |
Collapse
|
146
|
Zhou S, Ding F, Gu X. Non-coding RNAs as Emerging Regulators of Neural Injury Responses and Regeneration. Neurosci Bull 2016; 32:253-64. [PMID: 27037691 DOI: 10.1007/s12264-016-0028-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 03/14/2016] [Indexed: 02/06/2023] Open
Abstract
Non-coding RNAs (ncRNAs) are a large cluster of RNAs that do not encode proteins, but have multiple functions in diverse cellular processes. Mounting evidence indicates the involvement of ncRNAs in the physiology and pathophysiology of the central and peripheral nervous systems. It has been shown that numerous ncRNAs, especially microRNAs and long non-coding RNAs, are differentially expressed after insults such as acquired brain injury, spinal cord injury, and peripheral nerve injury. These ncRNAs affect neuronal survival, neurite regrowth, and glial phenotype primarily by targeting specific mRNAs, resulting in translation repression or degradation of the mRNAs. An increasing number of studies have investigated the regulatory roles of microRNAs and long non-coding RNAs in neural injury and regeneration, and thus a new research field is emerging. In this review, we highlight current progress in the field in an attempt to provide further insight into post-transcriptional changes occurring after neural injury, and to facilitate the potential use of ncRNAs for improving neural regeneration. We also suggest potential directions for future studies.
Collapse
Affiliation(s)
- Songlin Zhou
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Fei Ding
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xiaosong Gu
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
147
|
Predegenerated Schwann cells--a novel prospect for cell therapy for glaucoma: neuroprotection, neuroregeneration and neuroplasticity. Sci Rep 2016; 6:23187. [PMID: 27034151 PMCID: PMC4817039 DOI: 10.1038/srep23187] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 01/26/2016] [Indexed: 02/07/2023] Open
Abstract
Glaucoma is an optic neuropathy that leads to irreversible blindness. Because the current therapies are not sufficient to protect against glaucoma-induced visual impairment, new treatment approaches are necessary to prevent disease progression. Cell transplantation techniques are currently considered to be among the most promising opportunities for nervous system damage treatment. The beneficial effects of undifferentiated cells have been investigated in experimental models of glaucoma, however experiments were accompanied by various barriers, which would make putative treatment difficult or even impossible to apply in a clinical setting. The novel therapy proposed in our study creates conditions to eliminate some of the identified barriers described for precursor cells transplantation and allows us to observe direct neuroprotective and pro-regenerative effects in ongoing optic neuropathy without additional modifications to the transplanted cells. We demonstrated that the proposed novel Schwann cell therapy might be promising, effective and easy to apply, and is safer than the alternative cell therapies for the treatment of glaucoma.
Collapse
|
148
|
Jessen KR, Mirsky R. The repair Schwann cell and its function in regenerating nerves. J Physiol 2016; 594:3521-31. [PMID: 26864683 PMCID: PMC4929314 DOI: 10.1113/jp270874] [Citation(s) in RCA: 735] [Impact Index Per Article: 91.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2015] [Accepted: 11/28/2015] [Indexed: 01/05/2023] Open
Abstract
Nerve injury triggers the conversion of myelin and non‐myelin (Remak) Schwann cells to a cell phenotype specialized to promote repair. Distal to damage, these repair Schwann cells provide the necessary signals and spatial cues for the survival of injured neurons, axonal regeneration and target reinnervation. The conversion to repair Schwann cells involves de‐differentiation together with alternative differentiation, or activation, a combination that is typical of cell type conversions often referred to as (direct or lineage) reprogramming. Thus, injury‐induced Schwann cell reprogramming involves down‐regulation of myelin genes combined with activation of a set of repair‐supportive features, including up‐regulation of trophic factors, elevation of cytokines as part of the innate immune response, myelin clearance by activation of myelin autophagy in Schwann cells and macrophage recruitment, and the formation of regeneration tracks, Bungner's bands, for directing axons to their targets. This repair programme is controlled transcriptionally by mechanisms involving the transcription factor c‐Jun, which is rapidly up‐regulated in Schwann cells after injury. In the absence of c‐Jun, damage results in the formation of a dysfunctional repair cell, neuronal death and failure of functional recovery. c‐Jun, although not required for Schwann cell development, is therefore central to the reprogramming of myelin and non‐myelin (Remak) Schwann cells to repair cells after injury. In future, the signalling that specifies this cell requires further analysis so that pharmacological tools that boost and maintain the repair Schwann cell phenotype can be developed.
![]()
Collapse
Affiliation(s)
- K R Jessen
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| | - R Mirsky
- Department of Cell and Developmental Biology, University College London, Gower Street, London, WC1E 6BT, UK
| |
Collapse
|
149
|
Musa G, Engel FB, Niaudet C. Heart Development, Angiogenesis, and Blood-Brain Barrier Function Is Modulated by Adhesion GPCRs. Handb Exp Pharmacol 2016; 234:351-368. [PMID: 27832496 DOI: 10.1007/978-3-319-41523-9_16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The cardiovascular system in adult organisms forms a network of interconnected endothelial cells, supported by mural cells and displaying a high degree of hierarchy: arteries emerging from the heart ramify into arterioles and then capillaries, which return to the venous systems through venules and veins. The cardiovascular system allows blood circulation, which in turn is essential for hemostasis through gas diffusion, nutrient distribution, and cell trafficking. In this chapter, we have summarized the current knowledge on how adhesion GPCRs (aGPCRs) impact heart development, followed by their role in modulating vascular angiogenesis.
Collapse
Affiliation(s)
- Gentian Musa
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 12, Erlangen, 91054, Germany
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Schwabachanlage 12, Erlangen, 91054, Germany.
| | - Colin Niaudet
- Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, Dag Hammarskjölds väg 20, Uppsala, 751 85, Sweden.
| |
Collapse
|
150
|
The Lin28/let-7 axis is critical for myelination in the peripheral nervous system. Nat Commun 2015; 6:8584. [PMID: 26466203 PMCID: PMC4634210 DOI: 10.1038/ncomms9584] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 09/04/2015] [Indexed: 12/31/2022] Open
Abstract
MicroRNAs (miRNAs) are crucial regulators of myelination in the peripheral nervous system (PNS). However, the miRNAs species involved and the underlying mechanisms are largely unknown. We found that let-7 miRNAs are highly abundant during PNS myelination and that their levels are inversely correlated to the expression of lin28 homolog B (Lin28B), an antagonist of let-7 accumulation. Sustained expression of Lin28B and consequently reduced levels of let-7 miRNAs results in a failure of Schwann cell myelination in transgenic mouse models and in cell culture. Subsequent analyses revealed that let-7 miRNAs promote expression of the myelination-driving master transcription factor Krox20 (also known as Egr2) through suppression of myelination inhibitory Notch signalling. We conclude that the Lin28B/let-7 axis acts as a critical driver of PNS myelination, in particular by regulating myelination onset, identifying this pathway also as a potential therapeutic target in demyelinating diseases.
Collapse
|