1
|
Chung HJ, Nguyen TNC, Lee JW, Huh Y, Ko S, Lim H, Seo H, Ha YG, Chang JH, Woo JS, Song JJ, Kim SW, Lee JS, Mo JS, Park B, Min KW, Yoon JH, Kim MS, Jung J, Jeong NY. Targeting the Hippo pathway in Schwann cells ameliorates peripheral nerve degeneration via a polypharmacological mechanism. Neurotherapeutics 2024; 21:e00458. [PMID: 39384453 PMCID: PMC11585884 DOI: 10.1016/j.neurot.2024.e00458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 09/03/2024] [Accepted: 09/22/2024] [Indexed: 10/11/2024] Open
Abstract
Peripheral neuropathies (PNs) are common diseases in elderly individuals characterized by Schwann cell (SC) dysfunction and irreversible Wallerian degeneration (WD). Although the molecular mechanisms of PN onset and progression have been widely studied, therapeutic opportunities remain limited. In this study, we investigated the pharmacological inhibition of Mammalian Ste20-like kinase 1/2 (MST1/2) by using its chemical inhibitor, XMU-MP-1 (XMU), against WD. XMU treatment suppressed the proliferation, dedifferentiation, and demyelination of SCs in models of WD in vitro, in vivo, and ex vivo. As a downstream mediator of canonical and noncanonical Hippo/MST1 pathway activation, the mature microRNA (miRNA) let-7b and its binding partners quaking homolog (QKI)/nucleolin (NCL) modulated miRNA-mediated silencing of genes involved in protein transport. Hence, direct phosphorylation of QKI and NCL by MST1 might be critical for WD onset and pathogenesis. Moreover, p38α/mitogen-activated protein kinase 14 (p38α) showed a strong affinity for XMU, and therefore, it may be an alternative XMU target for controlling WD in SCs. Taken together, our findings provide new insights into the Hippo/MST pathway function in PNs and suggest that XMU is a novel multitargeted therapeutic for elderly individuals with PNs.
Collapse
Affiliation(s)
- Hyung-Joo Chung
- Department of Anesthesiology and Pain Medicine, College of Medicine, Kosin University, Busan 49267, South Korea
| | - Thy N C Nguyen
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, South Korea
| | - Ji Won Lee
- Department of Biology, College of Natural Sciences, Gangneung-Wonju National University, Gangneung 25457, South Korea
| | - Youngbuhm Huh
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, South Korea
| | - Seungbeom Ko
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Heejin Lim
- Center for Scientific Instrumentation, Korea Basic Science Institute (KBSI), Cheongju 28119, South Korea
| | - Hyewon Seo
- New Drug Development Center (NDDC), Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI hub), Daegu 41061, South Korea
| | - Young-Geun Ha
- Department of Chemistry, College of Convergence Science, Kyonggi University, Suwon 16227, South Korea
| | - Jeong Ho Chang
- Department of Biology Education, Kyungpook National University, Daegu 41566, South Korea
| | - Jae-Sung Woo
- Department of Life Sciences, Korea University, Seongbuk-gu, Seoul 02841, South Korea
| | - Ji-Joon Song
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Yuseong-gu, Daejeon 34141, South Korea
| | - So-Woon Kim
- Department of Pathology, College of Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul 02447, South Korea
| | - Jin San Lee
- Department of Neurology, College of Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul 02447, South Korea
| | - Jung-Soon Mo
- Institute of Medical Science, School of Medicine, Ajou University, Suwon 16499, South Korea
| | - Boyoun Park
- Department of Systems Biology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, South Korea
| | - Kyung-Won Min
- Department of Biology, College of Natural Sciences, Gangneung-Wonju National University, Gangneung 25457, South Korea
| | - Je-Hyun Yoon
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC 29425, USA; Department of Oncology Science, College of Medicine, The University of Oklahoma, Oklahoma City, OK 73104, USA.
| | - Min-Sik Kim
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 42988, South Korea.
| | - Junyang Jung
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul 02447, South Korea.
| | - Na Young Jeong
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Busan 49201, South Korea.
| |
Collapse
|
2
|
Piñero G, Vence M, Aranda ML, Cercato MC, Soto PA, Usach V, Setton-Avruj PC. All the PNS is a Stage: Transplanted Bone Marrow Cells Play an Immunomodulatory Role in Peripheral Nerve Regeneration. ASN Neuro 2023; 15:17590914231167281. [PMID: 37654230 PMCID: PMC10475269 DOI: 10.1177/17590914231167281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 02/28/2023] [Accepted: 03/16/2023] [Indexed: 09/02/2023] Open
Abstract
SUMMARY STATEMENT Bone marrow cell transplant has proven to be an effective therapeutic approach to treat peripheral nervous system injuries as it not only promoted regeneration and remyelination of the injured nerve but also had a potent effect on neuropathic pain.
Collapse
Affiliation(s)
- Gonzalo Piñero
- Departamento de Química Biológica, Cátedra de Química Biológica Patalógica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Universidad de Buenos Aires-CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Ciudad Autónoma de Buenos Aires, Argentina
- Department of Pathology, Mount Sinai Hospital, New York, NY, USA
| | - Marianela Vence
- Universidad de Buenos Aires-CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Ciudad Autónoma de Buenos Aires, Argentina
| | - Marcos L. Aranda
- Universidad de Buenos Aires-CONICET, Centro de Estudios Farmacológicos y Botánicos (CEFYBO), Ciudad Autónoma de Buenos Aires, Argentina
- Department of Neurobiology, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA
| | - Magalí C. Cercato
- Universidad de Buenos Aires-CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Ciudad Autónoma de Buenos Aires, Argentina
| | - Paula A. Soto
- Departamento de Química Biológica, Cátedra de Química Biológica Patalógica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Universidad de Buenos Aires-CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Ciudad Autónoma de Buenos Aires, Argentina
| | - Vanina Usach
- Departamento de Química Biológica, Cátedra de Química Biológica Patalógica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Universidad de Buenos Aires-CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Ciudad Autónoma de Buenos Aires, Argentina
| | - Patricia C. Setton-Avruj
- Departamento de Química Biológica, Cátedra de Química Biológica Patalógica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
- Universidad de Buenos Aires-CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
3
|
Cristobal CD, Lee HK. Development of myelinating glia: An overview. Glia 2022; 70:2237-2259. [PMID: 35785432 PMCID: PMC9561084 DOI: 10.1002/glia.24238] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 06/24/2022] [Accepted: 06/24/2022] [Indexed: 01/07/2023]
Abstract
Myelin is essential to nervous system function, playing roles in saltatory conduction and trophic support. Oligodendrocytes (OLs) and Schwann cells (SCs) form myelin in the central and peripheral nervous systems respectively and follow different developmental paths. OLs are neural stem-cell derived and follow an intrinsic developmental program resulting in a largely irreversible differentiation state. During embryonic development, OL precursor cells (OPCs) are produced in distinct waves originating from different locations in the central nervous system, with a subset developing into myelinating OLs. OPCs remain evenly distributed throughout life, providing a population of responsive, multifunctional cells with the capacity to remyelinate after injury. SCs derive from the neural crest, are highly dependent on extrinsic signals, and have plastic differentiation states. SC precursors (SCPs) are produced in early embryonic nerve structures and differentiate into multipotent immature SCs (iSCs), which initiate radial sorting and differentiate into myelinating and non-myelinating SCs. Differentiated SCs retain the capacity to radically change phenotypes in response to external signals, including becoming repair SCs, which drive peripheral regeneration. While several transcription factors and myelin components are common between OLs and SCs, their differentiation mechanisms are highly distinct, owing to their unique lineages and their respective environments. In addition, both OLs and SCs respond to neuronal activity and regulate nervous system output in reciprocal manners, possibly through different pathways. Here, we outline their basic developmental programs, mechanisms regulating their differentiation, and recent advances in the field.
Collapse
Affiliation(s)
- Carlo D. Cristobal
- Integrative Program in Molecular and Biomedical SciencesBaylor College of MedicineHoustonTexasUSA,Jan and Dan Duncan Neurological Research InstituteTexas Children's HospitalHoustonTexasUSA
| | - Hyun Kyoung Lee
- Integrative Program in Molecular and Biomedical SciencesBaylor College of MedicineHoustonTexasUSA,Jan and Dan Duncan Neurological Research InstituteTexas Children's HospitalHoustonTexasUSA,Department of PediatricsBaylor College of MedicineHoustonTexasUSA,Department of NeuroscienceBaylor College of MedicineHoustonTexasUSA
| |
Collapse
|
4
|
Ye Z, Wei J, Zhan C, Hou J. Role of Transforming Growth Factor Beta in Peripheral Nerve Regeneration: Cellular and Molecular Mechanisms. Front Neurosci 2022; 16:917587. [PMID: 35769702 PMCID: PMC9234557 DOI: 10.3389/fnins.2022.917587] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/11/2022] [Indexed: 11/24/2022] Open
Abstract
Peripheral nerve injury (PNI) is one of the most common concerns in trauma patients. Despite significant advances in repair surgeries, the outcome can still be unsatisfactory, resulting in morbidities such as loss of sensory or motor function and reduced quality of life. This highlights the need for more supportive strategies for nerve regrowth and adequate recovery. Multifunctional cytokine transforming growth factor-β (TGF-β) is essential for the development of the nervous system and is known for its neuroprotective functions. Accumulating evidence indicates its involvement in multiple cellular and molecular responses that are critical to peripheral nerve repair. Following PNI, TGF-β is released at the site of injury where it can initiate a series of phenotypic changes in Schwann cells (SCs), modulate immune cells, activate neuronal intrinsic growth capacity, and regulate blood nerve barrier (BNB) permeability, thus enhancing the regeneration of the nerves. Notably, TGF-β has already been applied experimentally in the treatment of PNI. These treatments with encouraging outcomes further demonstrate its regeneration-promoting capacity. Herein, we review the possible roles of TGF-β in peripheral nerve regeneration and discuss the underlying mechanisms, thus providing new cues for better treatment of PNI.
Collapse
Affiliation(s)
- Zhiqian Ye
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junbin Wei
- The First School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Chaoning Zhan
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jin Hou
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- *Correspondence: Jin Hou,
| |
Collapse
|
5
|
Jessen KR, Mirsky R. The Role of c-Jun and Autocrine Signaling Loops in the Control of Repair Schwann Cells and Regeneration. Front Cell Neurosci 2022; 15:820216. [PMID: 35221918 PMCID: PMC8863656 DOI: 10.3389/fncel.2021.820216] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 12/30/2021] [Indexed: 12/12/2022] Open
Abstract
After nerve injury, both Schwann cells and neurons switch to pro-regenerative states. For Schwann cells, this involves reprogramming of myelin and Remak cells to repair Schwann cells that provide the signals and mechanisms needed for the survival of injured neurons, myelin clearance, axonal regeneration and target reinnervation. Because functional repair cells are essential for regeneration, it is unfortunate that their phenotype is not robust. Repair cell activation falters as animals get older and the repair phenotype fades during chronic denervation. These malfunctions are important reasons for the poor outcomes after nerve damage in humans. This review will discuss injury-induced Schwann cell reprogramming and the concept of the repair Schwann cell, and consider the molecular control of these cells with emphasis on c-Jun. This transcription factor is required for the generation of functional repair cells, and failure of c-Jun expression is implicated in repair cell failures in older animals and during chronic denervation. Elevating c-Jun expression in repair cells promotes regeneration, showing in principle that targeting repair cells is an effective way of improving nerve repair. In this context, we will outline the emerging evidence that repair cells are sustained by autocrine signaling loops, attractive targets for interventions aimed at promoting regeneration.
Collapse
Affiliation(s)
- Kristjan R. Jessen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | | |
Collapse
|
6
|
Owen RS, Ramarathinam SH, Bailey A, Gastaldello A, Hussey K, Skipp PJ, Purcell AW, Siddle HV. The differentiation state of the Schwann cell progenitor drives phenotypic variation between two contagious cancers. PLoS Pathog 2021; 17:e1010033. [PMID: 34780568 PMCID: PMC8629380 DOI: 10.1371/journal.ppat.1010033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/29/2021] [Accepted: 10/13/2021] [Indexed: 01/04/2023] Open
Abstract
Contagious cancers are a rare pathogenic phenomenon in which cancer cells gain the ability to spread between genetically distinct hosts. Nine examples have been identified across marine bivalves, dogs and Tasmanian devils, but the Tasmanian devil is the only mammalian species known to have given rise to two distinct lineages of contagious cancer, termed Devil Facial Tumour 1 (DFT1) and 2 (DFT2). Remarkably, DFT1 and DFT2 arose independently from the same cell type, a Schwann cell, and while their ultra-structural features are highly similar they exhibit variation in their mutational signatures and infection dynamics. As such, DFT1 and DFT2 provide a unique framework for investigating how a common progenitor cell can give rise to distinct contagious cancers. Using a proteomics approach, we show that DFT1 and DFT2 are derived from Schwann cells in different differentiation states, with DFT2 carrying a molecular signature of a less well differentiated Schwann cell. Under inflammatory signals DFT1 and DFT2 have different gene expression profiles, most notably involving Schwann cell markers of differentiation, reflecting the influence of their distinct origins. Further, DFT2 cells express immune cell markers typically expressed during nerve repair, consistent with an ability to manipulate their extracellular environment, facilitating the cell’s ability to transmit between individuals. The emergence of two contagious cancers in the Tasmanian devil suggests that the inherent plasticity of Schwann cells confers a vulnerability to the formation of contagious cancers. Cancer can be an infectious pathogen, with nine known cases, infecting bivalves, dogs and two independent tumours circulating in the endangered Tasmanian devil. These cancers, known as Devil Facial Tumour 1 (DFT1) and Devil Facial Tumour 2 (DFT2), spread through the wild population much like parasites, moving between genetically distinct hosts during social biting behaviours and persisting in the population. As DFT1 and DFT2 are independent contagious cancers that arose from the same cell type, a Schwann cell, they provide a unique model system for studying the emergence of phenotypic variation in cancers derived from a single progenitor cell. In this study, we have shown that these two remarkably similar tumours have emerged from Schwann cells in different differentiation states. The differentiation state of the progenitor has altered the characteristics of each tumour, resulting in different responses to external signals. This work demonstrates that the cellular origin of infection can direct the phenotype of a contagious cancer and how it responds to signals from the host environment. Further, the plasticity of Schwann cells may make these cells more prone to forming contagious cancers, raising the possibility that further parasitic cancers could emerge from this cell type.
Collapse
Affiliation(s)
- Rachel S. Owen
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Sri H. Ramarathinam
- Department of Biochemistry and Molecular Biology and the Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Alistair Bailey
- Centre for Cancer Immunology, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Annalisa Gastaldello
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Kathryn Hussey
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Paul J. Skipp
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Anthony W. Purcell
- Department of Biochemistry and Molecular Biology and the Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Hannah V. Siddle
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
- * E-mail:
| |
Collapse
|
7
|
Fuertes-Alvarez S, Izeta A. Terminal Schwann Cell Aging: Implications for Age-Associated Neuromuscular Dysfunction. Aging Dis 2021; 12:494-514. [PMID: 33815879 PMCID: PMC7990373 DOI: 10.14336/ad.2020.0708] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022] Open
Abstract
Action potential is transmitted to muscle fibers through specialized synaptic interfaces called neuromuscular junctions (NMJs). These structures are capped by terminal Schwann cells (tSCs), which play essential roles during formation and maintenance of the NMJ. tSCs are implicated in the correct communication between nerves and muscles, and in reinnervation upon injury. During aging, loss of muscle mass and strength (sarcopenia and dynapenia) are due, at least in part, to the progressive loss of contacts between muscle fibers and nerves. Despite the important role of tSCs in NMJ function, very little is known on their implication in the NMJ-aging process and in age-associated denervation. This review summarizes the current knowledge about the implication of tSCs in the age-associated degeneration of NMJs. We also speculate on the possible mechanisms underlying the observed phenotypes.
Collapse
Affiliation(s)
- Sandra Fuertes-Alvarez
- 1Biodonostia, Tissue Engineering Group, Paseo Dr. Begiristain, s/n, San Sebastian 20014, Spain
| | - Ander Izeta
- 1Biodonostia, Tissue Engineering Group, Paseo Dr. Begiristain, s/n, San Sebastian 20014, Spain.,2Tecnun-University of Navarra, School of Engineering, Department of Biomedical Engineering and Science, Paseo Mikeletegi, 48, San Sebastian 20009, Spain
| |
Collapse
|
8
|
Fornaro M, Marcus D, Rattin J, Goral J. Dynamic Environmental Physical Cues Activate Mechanosensitive Responses in the Repair Schwann Cell Phenotype. Cells 2021; 10:cells10020425. [PMID: 33671410 PMCID: PMC7922665 DOI: 10.3390/cells10020425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/09/2021] [Accepted: 02/11/2021] [Indexed: 01/10/2023] Open
Abstract
Schwann cells plastically change in response to nerve injury to become a newly reconfigured repair phenotype. This cell is equipped to sense and interact with the evolving and unusual physical conditions characterizing the injured nerve environment and activate intracellular adaptive reprogramming as a consequence of external stimuli. Summarizing the literature contributions on this matter, this review is aimed at highlighting the importance of the environmental cues of the regenerating nerve as key factors to induce morphological and functional changes in the Schwann cell population. We identified four different microenvironments characterized by physical cues the Schwann cells sense via interposition of the extracellular matrix. We discussed how the physical cues of the microenvironment initiate changes in Schwann cell behavior, from wrapping the axon to becoming a multifunctional denervated repair cell and back to reestablishing contact with regenerated axons.
Collapse
Affiliation(s)
- Michele Fornaro
- Department of Anatomy, College of Graduate Studies (CGS), Midwestern University, Downers Grove, IL 60515, USA;
- Department of Anatomy, Chicago College of Osteopathic Medicine (CCOM), Midwestern University, Downers Grove, IL 60515, USA; (D.M.); (J.R.)
- Correspondence: ; Tel.: +001-630-515-6055
| | - Dominic Marcus
- Department of Anatomy, Chicago College of Osteopathic Medicine (CCOM), Midwestern University, Downers Grove, IL 60515, USA; (D.M.); (J.R.)
| | - Jacob Rattin
- Department of Anatomy, Chicago College of Osteopathic Medicine (CCOM), Midwestern University, Downers Grove, IL 60515, USA; (D.M.); (J.R.)
| | - Joanna Goral
- Department of Anatomy, College of Graduate Studies (CGS), Midwestern University, Downers Grove, IL 60515, USA;
- Department of Anatomy, Chicago College of Osteopathic Medicine (CCOM), Midwestern University, Downers Grove, IL 60515, USA; (D.M.); (J.R.)
| |
Collapse
|
9
|
Catignas KK, Frick LR, Pellegatta M, Hurley E, Kolb Z, Addabbo K, McCarty JH, Hynes RO, van der Flier A, Poitelon Y, Wrabetz L, Feltri ML. α V integrins in Schwann cells promote attachment to axons, but are dispensable in vivo. Glia 2021; 69:91-108. [PMID: 32744761 PMCID: PMC8491627 DOI: 10.1002/glia.23886] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 06/22/2020] [Accepted: 06/23/2020] [Indexed: 12/22/2022]
Abstract
In the developing peripheral nervous system, Schwann cells (SCs) extend their processes to contact, sort, and myelinate axons. The mechanisms that contribute to the interaction between SCs and axons are just beginning to be elucidated. Using a SC-neuron coculture system, we demonstrate that Arg-Gly-Asp (RGD) peptides that inhibit αV -containing integrins delay the extension of SCs elongating on axons. αV integrins in SC localize to sites of contact with axons and are expressed early in development during radial sorting and myelination. Short interfering RNA-mediated knockdown of the αV integrin subunit also delays SC extension along axons in vitro, suggesting that αV -containing integrins participate in axo-glial interactions. However, mice lacking the αV subunit in SCs, alone or in combination with the potentially compensating α5 subunit, or the αV partners β3 or β8 , myelinate normally during development and remyelinate normally after nerve crush, indicating that overlapping or compensatory mechanisms may hide the in vivo role of RGD-binding integrins.
Collapse
Affiliation(s)
- Kathleen K. Catignas
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York
- Department of Biochemistry, University at Buffalo, Buffalo, New York
| | - Luciana R. Frick
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - Marta Pellegatta
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York
- IRCCS San Raffaele Scientific Institute and Vita Salute San Raffaele University, Milan, Italy
| | - Edward Hurley
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - Zachary Kolb
- Department of Biochemistry, University at Buffalo, Buffalo, New York
| | - Kathryn Addabbo
- Department of Biochemistry, University at Buffalo, Buffalo, New York
| | - Joseph H. McCarty
- Department of Neurosurgery, The University of Texas M.D. Anderson Cancer Center, Houston, Texas
| | - Richard O. Hynes
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Boston, Massachusetts
| | - Arjan van der Flier
- Howard Hughes Medical Institute, Massachusetts Institute of Technology, Boston, Massachusetts
- Sanofi, Boston, Massachusetts
| | - Yannick Poitelon
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York
- Department of Biochemistry, University at Buffalo, Buffalo, New York
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York
| | - Lawrence Wrabetz
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York
- Department of Biochemistry, University at Buffalo, Buffalo, New York
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| | - Maria Laura Feltri
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, New York
- Department of Biochemistry, University at Buffalo, Buffalo, New York
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York
| |
Collapse
|
10
|
Wang Z, Mudalal M, Sun Y, Liu Y, Wang J, Wang Y, Sun X, Zhou Y. The Effects of Leukocyte-Platelet Rich Fibrin (L-PRF) on Suppression of the Expressions of the Pro-Inflammatory Cytokines, and Proliferation of Schwann Cell, and Neurotrophic Factors. Sci Rep 2020; 10:2421. [PMID: 32051476 PMCID: PMC7016122 DOI: 10.1038/s41598-020-59319-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 01/27/2020] [Indexed: 11/21/2022] Open
Abstract
This study evaluates the use of L-PRF as an autologous scaffold in nerve regeneration, and Schwann cells (SCs) proliferation and secretion of neurotrophic factors and its anti-inflammatory effect on SC Porphyromonas Gingivalis-Lipopolysaccharide (PG-LPS)-induced inflammatory responses in vitro. SEM was done to investigate various features of L-PRF. L-PRF-extracts was used to investigate the release of growth factors and treatment of SCs line. ELISA was applied to examine the release of IGF-1. The proliferative effect of L-PRF on SCs was assessed with CCK-8 assay. The effect of L-PRF on the mRNA and protein expression of SC neurotrophic factors were analyzed by RT-qPCR and ELISA. CCK-8 assay and RT-qPCR were used to determine the required concentration and the action time of PG-LPS before the anti-inflammatory effect of L-PRF was determined by measuring the changes in IL-1β, IL-6, and TNF-a with RT-qPCR and ELISA. There are different features in L-PRF. Fourteen days was sufficient to release adequate GF. The mRNA expressions of the pro-inflammatory cytokines were notably raised by PG-LPS in 3-hours treatment. L-PRF can increase SC proliferation, neurotrophic factors secretion, and suppress SC PG-LPS-induced inflammatory responses in vitro. L-PRF has the potential as an autologous biological additive for peripheral nerve regeneration in the event of nerve inflammation and injuries.
Collapse
Affiliation(s)
- Zhanqi Wang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China
- Provincial Key Laboratory of Dental Development, Jaw Remodeling and Regeneration, Jilin University, Changchun, 130021, China
| | - Mahmoud Mudalal
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China
- Provincial Key Laboratory of Dental Development, Jaw Remodeling and Regeneration, Jilin University, Changchun, 130021, China
- Department of Oral and Maxillofacial Surgery and Periodontology, Faculty of Dentistry, The Arab American University, Jenin, 240, Palestine
| | - Yue Sun
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China
- Provincial Key Laboratory of Dental Development, Jaw Remodeling and Regeneration, Jilin University, Changchun, 130021, China
| | - Yiping Liu
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China
- Provincial Key Laboratory of Dental Development, Jaw Remodeling and Regeneration, Jilin University, Changchun, 130021, China
| | - Jia Wang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China
- Provincial Key Laboratory of Dental Development, Jaw Remodeling and Regeneration, Jilin University, Changchun, 130021, China
| | - Yao Wang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China
- Provincial Key Laboratory of Dental Development, Jaw Remodeling and Regeneration, Jilin University, Changchun, 130021, China
| | - Xiaolin Sun
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Provincial Key Laboratory of Dental Development, Jaw Remodeling and Regeneration, Jilin University, Changchun, 130021, China.
| | - Yanmin Zhou
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, 130021, China.
- Provincial Key Laboratory of Dental Development, Jaw Remodeling and Regeneration, Jilin University, Changchun, 130021, China.
| |
Collapse
|
11
|
Fledrich R, Kungl T, Nave KA, Stassart RM. Axo-glial interdependence in peripheral nerve development. Development 2019; 146:146/21/dev151704. [PMID: 31719044 DOI: 10.1242/dev.151704] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
During the development of the peripheral nervous system, axons and myelinating Schwann cells form a unique symbiotic unit, which is realized by a finely tuned network of molecular signals and reciprocal interactions. The importance of this complex interplay becomes evident after injury or in diseases in which aspects of axo-glial interaction are perturbed. This Review focuses on the specific interdependence of axons and Schwann cells in peripheral nerve development that enables axonal outgrowth, Schwann cell lineage progression, radial sorting and, finally, formation and maintenance of the myelin sheath.
Collapse
Affiliation(s)
- Robert Fledrich
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany .,Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Theresa Kungl
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany.,Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Ruth M Stassart
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany .,Department of Neuropathology, University Clinic Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
12
|
Comparison of three congruent patient-specific cell types for the modelling of a human genetic Schwann-cell disorder. Nat Biomed Eng 2019; 3:571-582. [PMID: 30962586 PMCID: PMC6612317 DOI: 10.1038/s41551-019-0381-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 03/05/2019] [Indexed: 12/15/2022]
Abstract
Patient-specific human induced pluripotent stem cells (hiPSCs) hold great promise for the modelling of genetic disorders. However, these cells display wide intra-individual and inter-individual variations in gene expression, making it challenging to distinguish true-positive and false-positive phenotypes. Also, data from hiPSC phenotypes and from human embryonic stem cells (hESCs) harbouring the same disease mutation are lacking. Here, we report a comparison of molecular, cellular and functional characteristics of three congruent patient-specific cell types ― hiPSCs, hESCs, and direct lineage-converted cells ― derived from currently available differentiation and direct-reprogramming technologies, for the modelling of Charcot Marie Tooth 1A, a human genetic Schwann-cell disorder featuring a 1.4 megabase chromosomal duplication. In particular, we find that the chemokines CXCL1 and MCP1 are commonly upregulated in all three congruent models and in clinical patient samples. The development of congruent models of a single genetic disease by using somatic cells from a common patient will facilitate the search for convergent phenotypes.
Collapse
|
13
|
Jessen KR, Mirsky R. Schwann Cell Precursors; Multipotent Glial Cells in Embryonic Nerves. Front Mol Neurosci 2019; 12:69. [PMID: 30971890 PMCID: PMC6443887 DOI: 10.3389/fnmol.2019.00069] [Citation(s) in RCA: 105] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 03/04/2019] [Indexed: 12/20/2022] Open
Abstract
The cells of the neural crest, often referred to as neural crest stem cells, give rise to a number of sub-lineages, one of which is Schwann cells, the glial cells of peripheral nerves. Crest cells transform to adult Schwann cells through the generation of two well defined intermediate stages, the Schwann cell precursors (SCP) in early embryonic nerves, and immature Schwann cells (iSch) in late embryonic and perinatal nerves. SCP are formed when neural crest cells enter nascent nerves and form intimate relationships with axons, a diagnostic feature of glial cells. This involves large-scale changes in gene expression, including the activation of established glial cell markers. Like early glia in the CNS, radial glia, SCP retain developmental multipotency and contribute to other crest-derived lineages during embryonic development. SCP, as well as closely related cells termed boundary cap cells, and later stages of the Schwann cell lineage have all been implicated as the tumor initiating cell in NF1 associated neurofibromas. iSch are formed from SCP in a process that involves the appearance of additional differentiation markers, autocrine survival circuits, cellular elongation, a formation of endoneurial connective tissue and basal lamina. Finally, in peri- and post-natal nerves, iSch are reversibly induced by axon-associated signals to form the myelin and non-myelin Schwann cells of adult nerves. This review article discusses early Schwann cell development in detail and describes a large number of molecular signaling systems that control glial development in embryonic nerves.
Collapse
Affiliation(s)
- Kristjan R. Jessen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | | |
Collapse
|
14
|
Jessen KR, Arthur-Farraj P. Repair Schwann cell update: Adaptive reprogramming, EMT, and stemness in regenerating nerves. Glia 2019; 67:421-437. [PMID: 30632639 DOI: 10.1002/glia.23532] [Citation(s) in RCA: 212] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 08/20/2018] [Accepted: 09/05/2018] [Indexed: 12/16/2022]
Abstract
Schwann cells respond to nerve injury by cellular reprogramming that generates cells specialized for promoting regeneration and repair. These repair cells clear redundant myelin, attract macrophages, support survival of damaged neurons, encourage axonal growth, and guide axons back to their targets. There are interesting parallels between this response and that found in other tissues. At the cellular level, many other tissues also react to injury by cellular reprogramming, generating cells specialized to promote tissue homeostasis and repair. And at the molecular level, a common feature possessed by Schwann cells and many other cells is the injury-induced activation of genes associated with epithelial-mesenchymal transitions and stemness, differentiation states that are linked to cellular plasticity and that help injury-induced tissue remodeling. The number of signaling systems regulating Schwann cell plasticity is rapidly increasing. Importantly, this includes mechanisms that are crucial for the generation of functional repair Schwann cells and nerve regeneration, although they have no or a minor role elsewhere in the Schwann cell lineage. This encourages the view that selective tools can be developed to control these particular cells, amplify their repair supportive functions and prevent their deterioration. In this review, we discuss the emerging similarities between the injury response seen in nerves and in other tissues and survey the transcription factors, epigenetic mechanisms, and signaling cascades that control repair Schwann cells, with emphasis on systems that selectively regulate the Schwann cell injury response.
Collapse
Affiliation(s)
- Kristjan R Jessen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Peter Arthur-Farraj
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
15
|
Gerber D, Ghidinelli M, Tinelli E, Somandin C, Gerber J, Pereira JA, Ommer A, Figlia G, Miehe M, Nägeli LG, Suter V, Tadini V, Sidiropoulos PNM, Wessig C, Toyka KV, Suter U. Schwann cells, but not Oligodendrocytes, Depend Strictly on Dynamin 2 Function. eLife 2019; 8:e42404. [PMID: 30648534 PMCID: PMC6335055 DOI: 10.7554/elife.42404] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 12/31/2018] [Indexed: 12/13/2022] Open
Abstract
Myelination requires extensive plasma membrane rearrangements, implying that molecules controlling membrane dynamics play prominent roles. The large GTPase dynamin 2 (DNM2) is a well-known regulator of membrane remodeling, membrane fission, and vesicular trafficking. Here, we genetically ablated Dnm2 in Schwann cells (SCs) and in oligodendrocytes of mice. Dnm2 deletion in developing SCs resulted in severely impaired axonal sorting and myelination onset. Induced Dnm2 deletion in adult SCs caused a rapidly-developing peripheral neuropathy with abundant demyelination. In both experimental settings, mutant SCs underwent prominent cell death, at least partially due to cytokinesis failure. Strikingly, when Dnm2 was deleted in adult SCs, non-recombined SCs still expressing DNM2 were able to remyelinate fast and efficiently, accompanied by neuropathy remission. These findings reveal a remarkable self-healing capability of peripheral nerves that are affected by SC loss. In the central nervous system, however, we found no major defects upon Dnm2 deletion in oligodendrocytes.
Collapse
Affiliation(s)
- Daniel Gerber
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Monica Ghidinelli
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Elisa Tinelli
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Christian Somandin
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Joanne Gerber
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Jorge A Pereira
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Andrea Ommer
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Gianluca Figlia
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Michaela Miehe
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Lukas G Nägeli
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Vanessa Suter
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Valentina Tadini
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Páris NM Sidiropoulos
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Carsten Wessig
- Department of NeurologyUniversity Hospital of Würzburg, University of WürzburgWürzburgGermany
| | - Klaus V Toyka
- Department of NeurologyUniversity Hospital of Würzburg, University of WürzburgWürzburgGermany
| | - Ueli Suter
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| |
Collapse
|
16
|
Jiang M, Rao R, Wang J, Wang J, Xu L, Wu LM, Chan JR, Wang H, Lu QR. The TSC1-mTOR-PLK axis regulates the homeostatic switch from Schwann cell proliferation to myelination in a stage-specific manner. Glia 2018; 66:1947-1959. [PMID: 29722913 PMCID: PMC6185760 DOI: 10.1002/glia.23449] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 04/10/2018] [Accepted: 04/10/2018] [Indexed: 12/17/2022]
Abstract
Proper peripheral myelination depends upon the balance between Schwann cell proliferation and differentiation programs. The serine/threonine kinase mTOR integrates various environmental cues to serve as a central regulator of cell growth, metabolism, and function. We report here that tuberous sclerosis complex 1 (TSC1), a negative regulator of mTOR activity, establishes a stage-dependent program for Schwann cell lineage progression and myelination by controlling cell proliferation and myelin homeostasis. Tsc1 ablation in Schwann cell progenitors in mice resulted in activation of mTOR signaling, and caused over-proliferation of Schwann cells and blocked their differentiation, leading to hypomyelination. Transcriptome profiling analysis revealed that mTOR activation in Tsc1 mutants resulted in upregulation of a polo-like kinase (PLK)-dependent pathway and cell cycle regulators. Attenuation of mTOR or pharmacological inhibition of polo-like kinases partially rescued hypomyelination caused by Tsc1 loss in the developing peripheral nerves. In contrast, deletion of Tsc1 in mature Schwann cells led to redundant and overgrown myelin sheaths in adult mice. Together, our findings indicate stage-specific functions for the TSC1-mTOR-PLK signaling axis in controlling the transition from proliferation to differentiation and myelin homeostasis during Schwann cell development.
Collapse
Affiliation(s)
- Minqing Jiang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
- The Institute of Cognitive Neuroscience, East China Normal University, Shanghai, China
| | - Rohit Rao
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jincheng Wang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jiajia Wang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Lingli Xu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Lai Man Wu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jonah R. Chan
- Department of Neurology and Programs in Biomedical and Neurosciences, University of California, San Francisco, CA 94158
| | - Huimin Wang
- The Institute of Cognitive Neuroscience, East China Normal University, Shanghai, China
| | - Q. Richard Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
17
|
Miller SR, Benito C, Mirsky R, Jessen KR, Baker CVH. Neural crest Notch/Rbpj signaling regulates olfactory gliogenesis and neuronal migration. Genesis 2018; 56:e23215. [PMID: 30134068 PMCID: PMC6099236 DOI: 10.1002/dvg.23215] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 05/02/2018] [Accepted: 05/04/2018] [Indexed: 12/13/2022]
Abstract
The neural crest-derived ensheathing glial cells of the olfactory nerve (OECs) are unique in spanning both the peripheral and central nervous systems: they ensheathe bundles of axons projecting from olfactory receptor neurons in the nasal epithelium to their targets in the olfactory bulb. OECs are clinically relevant as a promising autologous cell transplantation therapy for promoting central nervous system repair. They are also important for fertility, being required for the migration of embryonic gonadotropin-releasing hormone (GnRH) neurons from the olfactory placode along terminal nerve axons to the medial forebrain, which they enter caudal to the olfactory bulbs. Like Schwann cell precursors, OEC precursors associated with the developing olfactory nerve express the glial marker myelin protein zero and the key peripheral glial transcription factor Sox10. The transition from Schwann cell precursors to immature Schwann cells is accelerated by canonical Notch signaling via the Rbpj transcription factor. Here, we aimed to test the role of Notch/Rbpj signaling in developing OECs by blocking the pathway in both chicken and mouse. Our results suggest that Notch/Rbpj signaling prevents the cranial neural crest cells that colonize the olfactory nerve from differentiating as neurons, and at later stages contributes to the guidance of GnRH neurons.
Collapse
Affiliation(s)
- Sophie R. Miller
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeCB2 3DYUnited Kingdom
| | - Cristina Benito
- Department of Cell and Developmental BiologyUniversity College London, Gower StreetLondonWC1E 6BTUnited Kingdom
| | - Rhona Mirsky
- Department of Cell and Developmental BiologyUniversity College London, Gower StreetLondonWC1E 6BTUnited Kingdom
| | - Kristján R. Jessen
- Department of Cell and Developmental BiologyUniversity College London, Gower StreetLondonWC1E 6BTUnited Kingdom
| | - Clare V. H. Baker
- Department of Physiology, Development and NeuroscienceUniversity of CambridgeCambridgeCB2 3DYUnited Kingdom
| |
Collapse
|
18
|
Sustained Expression of Negative Regulators of Myelination Protects Schwann Cells from Dysmyelination in a Charcot-Marie-Tooth 1B Mouse Model. J Neurosci 2018; 38:4275-4287. [PMID: 29610440 DOI: 10.1523/jneurosci.0201-18.2018] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Revised: 03/02/2018] [Accepted: 03/16/2018] [Indexed: 12/16/2022] Open
Abstract
Schwann cell differentiation and myelination in the PNS are the result of fine-tuning of positive and negative transcriptional regulators. As myelination starts, negative regulators are downregulated, whereas positive ones are upregulated. Fully differentiated Schwann cells maintain an extraordinary plasticity and can transdifferentiate into "repair" Schwann cells after nerve injury. Reactivation of negative regulators of myelination is essential to generate repair Schwann cells. Negative regulators have also been implicated in demyelinating neuropathies, although their role in disease remains elusive. Here, we used a mouse model of Charcot-Marie-Tooth neuropathy type 1B (CMT1B), the P0S63del mouse characterized by ER stress and the activation of the unfolded protein response, to show that adult Schwann cells are in a partial differentiation state because they overexpress transcription factors that are normally expressed only before myelination. We provide evidence that two of these factors, Sox2 and Id2, act as negative regulators of myelination in vivo However, their sustained expression in neuropathy is protective because ablation of Sox2 or/and Id2 from S63del mice of both sexes results in worsening of the dysmyelinating phenotype. This is accompanied by increased levels of mutant P0 expression and exacerbation of ER stress, suggesting that limited differentiation may represent a novel adaptive mechanism through which Schwann cells counter the toxic effect of a mutant terminal differentiation protein.SIGNIFICANCE STATEMENT In many neuropathies, Schwann cells express high levels of early differentiation genes, but the significance of these altered expression remained unclear. Because many of these factors may act as negative regulators of myelination, it was suggested that their misexpression could contribute to dysmyelination. Here, we show that the transcription factors Sox2 and Id2 act as negative regulators of myelination in vivo, but that their sustained expression in Charcot-Marie-Tooth type 1B (CMT1B) represents an adaptive response activated by the Schwann cells to reduce mutant protein toxicity and prevent demyelination.
Collapse
|
19
|
Shin YK, Jang SY, Yun SH, Choi YY, Yoon BA, Jo YR, Park SY, Pak MG, Park JI, Park HT. Cooperative interaction of hepatocyte growth factor and neuregulin regulates Schwann cell migration and proliferation through Grb2-associated binder-2 in peripheral nerve repair. Glia 2017; 65:1794-1808. [PMID: 28722233 DOI: 10.1002/glia.23195] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/29/2017] [Accepted: 06/30/2017] [Indexed: 12/27/2022]
Abstract
The sequential reactive changes in Schwann cell phenotypes in transected peripheral nerves, including dedifferentiation, proliferation and migration, are essential for nerve repair. Even though the injury-induced migratory and proliferative behaviors of Schwann cells resemble epithelial and mesenchymal transition (EMT) in tumors, the molecular mechanisms underlying this phenotypic change of Schwann cells are still unclear. Here we show that the reactive Schwann cells exhibit migratory features dependent on the expression of a scaffolding oncoprotein Grb2-associated binder-2 (Gab2), which was transcriptionally induced by neuregulin 1-ErbB2 signaling following nerve injury. Injury-induced Gab2 expression was dependent on c-Jun, a transcription factor critical to a Schwann cell reprograming into a repair-type cell. Interestingly, the injury-induced activation (tyrosine phosphorylation) of Gab2 in Schwann cells was regulated by an EMT signal, the hepatocyte growth factor-c-Met signaling, but not by neuregulin 1. Gab2 knockout mice exhibited a deficit in nerve repair after nerve transection due to limited Schwann cell migration. Furthermore, Gab2 was required for the proliferation of Schwann cells following nerve injury and in vitro, and was over-expressed in human Schwann cell-derived tumors. In contrast, the tyrosine phosphorylation of Gab1 after nerve injury was principally regulated by the neuregulin 1-ErbB2 signaling and was indispensable for remyelination after crush injury, but not for the proliferation and migration of Schwann cells. Our findings indicate that Gab1 and Gab2 in Schwann cells are nonredundant and play a crucial role in peripheral nerve repair.
Collapse
Affiliation(s)
- Yoon Kyoung Shin
- Department of Physiology, Peripheral Neuropathy Research Center (PNRC), College of Medicine, Dong-A University, Busan, South Korea
| | - So Young Jang
- Department of Physiology, Peripheral Neuropathy Research Center (PNRC), College of Medicine, Dong-A University, Busan, South Korea
| | - Seoug Hoon Yun
- Department of Biochemistry, Peripheral Neuropathy Research Center (PNRC), College of Medicine, Dong-A University, Busan, South Korea
| | - Yun Young Choi
- Department of Physiology, Peripheral Neuropathy Research Center (PNRC), College of Medicine, Dong-A University, Busan, South Korea
| | - Byeol-A Yoon
- Department of Physiology, Peripheral Neuropathy Research Center (PNRC), College of Medicine, Dong-A University, Busan, South Korea
| | - Young Rae Jo
- Department of Physiology, Peripheral Neuropathy Research Center (PNRC), College of Medicine, Dong-A University, Busan, South Korea
| | - So Young Park
- Department of Physiology, Peripheral Neuropathy Research Center (PNRC), College of Medicine, Dong-A University, Busan, South Korea
| | - Min Gyoung Pak
- Department of Pathology, College of Medicine, Dong-A University, Busan, South Korea
| | - Joo In Park
- Department of Biochemistry, Peripheral Neuropathy Research Center (PNRC), College of Medicine, Dong-A University, Busan, South Korea
| | - Hwan Tae Park
- Department of Physiology, Peripheral Neuropathy Research Center (PNRC), College of Medicine, Dong-A University, Busan, South Korea
| |
Collapse
|
20
|
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: 220] [Impact Index Per Article: 27.5] [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
|
21
|
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: 10.1] [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
|
22
|
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: 0.5] [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
|
23
|
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
|
24
|
KAJIGAYA H, ISHIBASHI T, HAYASHI A, YAMAGUCHI Y, BABA H. Concentration of neddylation-related molecules in paranodal myelin of the peripheral nervous system. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2016; 92:56-68. [PMID: 26860454 PMCID: PMC4906812 DOI: 10.2183/pjab.92.56] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/09/2015] [Indexed: 06/05/2023]
Abstract
Neddylation is a reversible post-translational modification in which a small ubiquitin-like molecule called NEDD8 covalently binds to substrate proteins. Although a recent study suggests that neddylation is essential for formation and maintenance of dendritic spines in the brain, the role of this protein modification in the peripheral nerves is wholly unknown. In this study, we demonstrate that neddylation-related molecules, NEDD8 and DCUN1D2 (defective in cullin neddylation 1, domain containing 2), were concentrated at the paranode of peripheral myelin, in addition to the myelinated and unmyelinated Schwann cell bodies. These proteins were localized mainly within larger fibers, but not in fibers with small diameters. Developmental analyses showed that these molecules first appeared at the paranode during later stages of myelination, and this characteristic distribution disappeared in sulfatide-deficient mice in which paranodal axo-glial junctions were disrupted. These results suggest that the myelin paranode may be one of the regions where neddylation occurs within the peripheral nerves.
Collapse
Affiliation(s)
- Hitoshi KAJIGAYA
- Department of Molecular Neurobiology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
- Tokai University School of Medicine, Kanagawa, Japan
| | - Tomoko ISHIBASHI
- Department of Molecular Neurobiology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Akiko HAYASHI
- Department of Molecular Neurobiology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yoshihide YAMAGUCHI
- Department of Molecular Neurobiology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Hiroko BABA
- Department of Molecular Neurobiology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| |
Collapse
|
25
|
Qin J, Wang L, Sun Y, Sun X, Wen C, Shahmoradi M, Zhou Y. Concentrated growth factor increases Schwann cell proliferation and neurotrophic factor secretion and promotes functional nerve recovery in vivo. Int J Mol Med 2015; 37:493-500. [PMID: 26709397 DOI: 10.3892/ijmm.2015.2438] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 12/11/2015] [Indexed: 02/07/2023] Open
Abstract
Concentrated growth factor (CGF) is a newly generated complex that comprises a fibrin matrix incorporating growth factors and plasmatic and leukocyte cytokines. It has been widely used in bone regenerative medicine. However, the effect of CGF on peripheral nerve regeneration had not been previously investigated. The aim of the present study was to evaluate the possibility of using CGF for nerve regeneration by i) investigating the effect of CGF on the proliferation of Schwann cells (SCs) and secretion of neurotrophic factors nerve growth factor (NGF) and glial cell line‑derived neurotrophic factor (GDNF) in vitro; and ii) analyzing the effect of CGF on functional nerve recovery after nerve injury in vivo. CGF was prepared from venous blood taken from rats, and using scanning electron microscopy (SEM) we noted that it featured a fiber‑like appearance with pore size ranging from 0.1 to 1.0 µm. The soluble component of CGF was used to produce conditioned media with which to treat the Schwann cell line. A cell counting kit-8 assay and cell cycle analysis were both used to study the proliferative effect of CGF on SCs. Reverse transcription-quantitative PCR and western blot analysis demonstrated that there was an increase in the mRNA and protein expression of NGF and GDNF, both of which are markers of SC neurotrophic secretion. A model of sciatic nerve crush injury was established for the in vivo experiment, and CGF was found to increase the sciatic functional index (indicative of nerve function). We noted that CGF increased SC proliferation and secretion of neurotrophic factors in vitro, and promoted functional recovery after peripheral nerve injuries in vivo. These results suggest that CGF is a promising candidate biomaterial for peripheral nerve regeneration, and may potentially be utilized to repair nerve injuries.
Collapse
Affiliation(s)
- Jie Qin
- Department of Dental Implantology, School and Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Lin Wang
- Department of Very Important People, School and Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yue Sun
- Department of Dental Implantology, School and Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xiaolin Sun
- Department of Dental Implantology, School and Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Chaoju Wen
- Department of Dental Implantology, School and Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Mahdi Shahmoradi
- Department of Bioengineering, School of Dentistry, The University of Sydney, NSW 2006, Australia
| | - Yanmin Zhou
- Department of Dental Implantology, School and Hospital of Stomatology, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
26
|
Suarez-Mier GB, Buckwalter MS. Glial Fibrillary Acidic Protein-Expressing Glia in the Mouse Lung. ASN Neuro 2015; 7:7/5/1759091415601636. [PMID: 26442852 PMCID: PMC4601129 DOI: 10.1177/1759091415601636] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Autonomic nerves regulate important functions in visceral organs, including the lung. The postganglionic portion of these nerves is ensheathed by glial cells known as non-myelinating Schwann cells. In the brain, glia play important functional roles in neurotransmission, neuroinflammation, and maintenance of the blood brain barrier. Similarly, enteric glia are now known to have analogous roles in gastrointestinal neurotransmission, inflammatory response, and barrier formation. In contrast to this, very little is known about the function of glia in other visceral organs. Like the gut, the lung forms a barrier between airborne pathogens and the bloodstream, and autonomic lung innervation is known to affect pulmonary inflammation and lung function. Lung glia are described as non-myelinating Schwann cells but their function is not known, and indeed no transgenic tools have been validated to study them in vivo. The primary goal of this research was, therefore, to investigate the relationship between non-myelinating Schwann cells and pulmonary nerves in the airways and vasculature and to validate existing transgenic mouse tools that would be useful for studying their function. We focused on the glial fibrillary acidic protein promoter, which is a cognate marker of astrocytes that is expressed by enteric glia and non-myelinating Schwann cells. We describe the morphology of non-myelinating Schwann cells in the lung and verify that they express glial fibrillary acidic protein and S100, a classic glial marker. Furthermore, we characterize the relationship of non-myelinating Schwann cells to pulmonary nerves. Finally, we report tools for studying their function, including a commercially available transgenic mouse line.
Collapse
Affiliation(s)
- Gabriela B Suarez-Mier
- Department of Neurology and Neurological Sciences, Stanford Medical School, Stanford, CA, USA Stanford Neurosciences Institute, Stanford, CA, USA
| | - Marion S Buckwalter
- Department of Neurology and Neurological Sciences, Stanford Medical School, Stanford, CA, USA Department of Neurosurgery, Stanford Medical School, Stanford, CA, USA
| |
Collapse
|
27
|
Abstract
Schwann cells develop from the neural crest in a well-defined sequence of events. This involves the formation of the Schwann cell precursor and immature Schwann cells, followed by the generation of the myelin and nonmyelin (Remak) cells of mature nerves. This review describes the signals that control the embryonic phase of this process and the organogenesis of peripheral nerves. We also discuss the phenotypic plasticity retained by mature Schwann cells, and explain why this unusual feature is central to the striking regenerative potential of the peripheral nervous system (PNS).
Collapse
Affiliation(s)
- Kristján R Jessen
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom
| | - Rhona Mirsky
- Department of Cell and Developmental Biology, University College London, London WC1E 6BT, United Kingdom
| | - Alison C Lloyd
- MRC Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
28
|
Bone marrow Schwann cells induce hematopoietic stem cell hibernation. Int J Hematol 2014; 99:695-8. [PMID: 24817152 DOI: 10.1007/s12185-014-1588-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2014] [Accepted: 04/25/2014] [Indexed: 01/09/2023]
Abstract
Hematopoietic stem cells (HSCs) are clonogenic cells capable of both self-renewal and multilineage differentiation. In adult mouse bone marrow (BM), most HSCs remain in the non-dividing G0-phase of cell cycle, in close contact with supporting cells known as the HSC "niche". In the present study, we focused on signaling mechanisms that regulate stem cell dormancy in the BM niche. We show that TGF-β type II receptor deficiency causes reduced phosphorylation of Smad2/3 and impairs long-term repopulating activity in HSCs, suggesting a significant role for TGF-β/Smad signaling in hematopoiesis. Furthermore, we aimed at defining the candidate BM niche responsible for homeostasis of hematopoiesis, and revealed that non-myelinating Schwann cells sustain HSC hibernation by converting TGF-β from its latent to its active form.
Collapse
|
29
|
Tzekova N, Heinen A, Küry P. Molecules involved in the crosstalk between immune- and peripheral nerve Schwann cells. J Clin Immunol 2014; 34 Suppl 1:S86-104. [PMID: 24740512 DOI: 10.1007/s10875-014-0015-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2014] [Accepted: 03/19/2014] [Indexed: 12/13/2022]
Abstract
Schwann cells are the myelinating glial cells of the peripheral nervous system and establish myelin sheaths on large caliber axons in order to accelerate their electrical signal propagation. Apart from this well described function, these cells revealed to exhibit a high degree of differentiation plasticity as they were shown to re- and dedifferentiate upon injury and disease as well as to actively participate in regenerative- and inflammatory processes. This review focuses on the crosstalk between glial- and immune cells observed in many peripheral nerve pathologies and summarizes functional evidences of molecules, regulators and factors involved in this process. We summarize data on Schwann cell's role presenting antigens, on interactions with the complement system, on Schwann cell surface molecules/receptors and on secreted factors involved in immune cell interactions or para-/autocrine signaling events, thus strengthening the view for a broader (patho) physiological role of this cell lineage.
Collapse
Affiliation(s)
- Nevena Tzekova
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Moorenstrasse 5, D-40225, Düsseldorf, Germany
| | | | | |
Collapse
|
30
|
Cerri F, Salvatore L, Memon D, Boneschi FM, Madaghiele M, Brambilla P, Del Carro U, Taveggia C, Riva N, Trimarco A, Lopez ID, Comi G, Pluchino S, Martino G, Sannino A, Quattrini A. Peripheral nerve morphogenesis induced by scaffold micropatterning. Biomaterials 2014; 35:4035-4045. [PMID: 24559639 DOI: 10.1016/j.biomaterials.2014.01.069] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 01/26/2014] [Indexed: 10/25/2022]
Abstract
Several bioengineering approaches have been proposed for peripheral nervous system repair, with limited results and still open questions about the underlying molecular mechanisms. We assessed the biological processes that occur after the implantation of collagen scaffold with a peculiar porous micro-structure of the wall in a rat sciatic nerve transection model compared to commercial collagen conduits and nerve crush injury using functional, histological and genome wide analyses. We demonstrated that within 60 days, our conduit had been completely substituted by a normal nerve. Gene expression analysis documented a precise sequential regulation of known genes involved in angiogenesis, Schwann cells/axons interactions and myelination, together with a selective modulation of key biological pathways for nerve morphogenesis induced by porous matrices. These data suggest that the scaffold's micro-structure profoundly influences cell behaviors and creates an instructive micro-environment to enhance nerve morphogenesis that can be exploited to improve recovery and understand the molecular differences between repair and regeneration.
Collapse
Affiliation(s)
- Federica Cerri
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Luca Salvatore
- Department of Innovation Engineering, University of Lecce, Via per Monteroni, 73100 Lecce, Italy
| | - Danish Memon
- Department of Clinical Neurosciences, Centre for Brain Repair, University of Cambridge, Robinson Way CB2 0PY, UK
| | | | - Marta Madaghiele
- Department of Innovation Engineering, University of Lecce, Via per Monteroni, 73100 Lecce, Italy
| | - Paola Brambilla
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Ubaldo Del Carro
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Carla Taveggia
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Nilo Riva
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Amelia Trimarco
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Ignazio D Lopez
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Giancarlo Comi
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Stefano Pluchino
- Department of Clinical Neurosciences, Centre for Brain Repair, University of Cambridge, Robinson Way CB2 0PY, UK
| | - Gianvito Martino
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| | - Alessandro Sannino
- Department of Innovation Engineering, University of Lecce, Via per Monteroni, 73100 Lecce, Italy
| | - Angelo Quattrini
- Division of Neuroscience and INSPE, San Raffaele Scientific Institute, via Olgettina 60, 20132 Milan, Italy
| |
Collapse
|
31
|
Wang Y, Long L, Yang J, Wu Y, Wu H, Wei H, Deng X, Cheng X, Lou D, Chen H, Wen H. Spatiotemporal expression of SKIP after rat sciatic nerve crush. Neurochem Res 2013; 38:857-65. [PMID: 23389663 DOI: 10.1007/s11064-013-0990-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 01/05/2013] [Accepted: 01/29/2013] [Indexed: 11/26/2022]
Abstract
Ski-interacting protein (SKIP) is a highly conserved protein from yeast to Human. As an essential spliceosomal component and transcriptional co-regulator it plays an important role in preinitiation, splicing and polyadenylation. SKIP can also combine with Ski to overcome the G1 arrest and the growth-suppressive activities of pRb. Furthermore SKIP has the capacity to augment TGF-β dependent transcription. While the distribution and function of SKIP in peripheral nervous system lesion and regeneration remain unclear. Here, we investigated the spatiotemporal expression of SKIP in an acute sciatic nerve crush model in adult rats. Western Blot analysis revealed that SKIP was expressed in normal sciatic nerves. It gradually increased, reached a peak at 1 week after crush, and then returned to the normal level at 4 weeks. Besides, we observed that up-regulation of SKIP was approximately in parallel with Proliferating cell nuclear antigen (PCNA), and numerous Schwann cells (SCs) expressing SKIP were PCNA and Ki-67 positive. Collectively, we hypothesized peripheral nerve crush induced up-regulation of SKIP in the sciatic nerve, which was associated with SCs proliferation.
Collapse
Affiliation(s)
- Youhua Wang
- Department of Orthopaedics, Affiliated Hospital of Nantong University, 20 Xi-Si Road, Nantong, 226001 Jiangsu, People's Republic of China.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Doddrell RDS, Dun XP, Moate RM, Jessen KR, Mirsky R, Parkinson DB. Regulation of Schwann cell differentiation and proliferation by the Pax-3 transcription factor. Glia 2012; 60:1269-78. [PMID: 22532290 PMCID: PMC5722199 DOI: 10.1002/glia.22346] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2011] [Accepted: 03/30/2012] [Indexed: 12/13/2022]
Abstract
Pax-3 is a paired domain transcription factor that plays many roles during vertebrate development. In the Schwann cell lineage, Pax-3 is expressed at an early stage in Schwann cells precursors of the embryonic nerve, is maintained in the nonmyelinating cells of the adult nerve, and is upregulated in Schwann cells after peripheral nerve injury. Consistent with this expression pattern, Pax-3 has previously been shown to play a role in repressing the expression of the myelin basic protein gene in Schwann cells. We have studied the role of Pax-3 in Schwann cells and have found that it controls not only the regulation of cell differentiation but also the survival and proliferation of Schwann cells. Pax-3 expression blocks both the induction of Oct-6 and Krox-20 (K20) by cyclic AMP and completely inhibits the ability of K20, the physiological regulator of myelination in the peripheral nervous system, to induce myelin gene expression in Schwann cells. In contrast to other inhibitors of myelination, we find that Pax-3 represses myelin gene expression in a c-Jun-independent manner. In addition to this, we find that Pax-3 expression alone is sufficient to inhibit the induction of apoptosis by TGFβ1 in Schwann cells. Expression of Pax-3 is also sufficient to induce the proliferation of Schwann cells in the absence of added growth factors and to reverse K20-induced exit from the cell cycle. These findings indicate new roles for the Pax-3 transcription factor in controlling the differentiation and proliferation of Schwann cells during development and after peripheral nerve injury.
Collapse
Affiliation(s)
- Robin D. S. Doddrell
- Peninsula College of Medicine and Dentistry, University of Exeter, Plymouth, Devon, United Kingdom
| | - Xin-Peng Dun
- Peninsula College of Medicine and Dentistry, University of Exeter, Plymouth, Devon, United Kingdom
| | - Roy M. Moate
- School of Biomedical and Biological Sciences, University of Plymouth, Plymouth, Devon, United Kingdom
| | - Kristjan R. Jessen
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Rhona Mirsky
- Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - David B. Parkinson
- Peninsula College of Medicine and Dentistry, University of Exeter, Plymouth, Devon, United Kingdom
| |
Collapse
|
33
|
Patmore DM, Welch S, Fulkerson PC, Wu J, Choi K, Eaves D, Kordich JJ, Collins MH, Cripe TP, Ratner N. In vivo regulation of TGF-β by R-Ras2 revealed through loss of the RasGAP protein NF1. Cancer Res 2012; 72:5317-27. [PMID: 22918885 DOI: 10.1158/0008-5472.can-12-1972] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Ras superfamily proteins participate in TGF-β-mediated developmental pathways that promote either tumor suppression or progression. However, the specific Ras proteins, which integrate in vivo with TGF-β signaling pathways, are unknown. As a general approach to this question, we activated all Ras proteins in vivo by genetic deletion of the RasGAP protein Nf1 and examined mice doubly deficient in a Ras protein to determine its requirement in formation of TGF-β-dependent neurofibromas that arise in Nf1-deficient mice. Animals lacking Nf1 and the Ras-related protein R-Ras2/TC21 displayed a delay in formation of neurofibromas but an acceleration in formation of brain tumors and sarcomas. Loss of R-Ras2 was associated with elevated expression of TGF-β in Nf1-deficient Schwann cell precursors, blockade of a Nf1/TGFβRII/AKT-dependent autocrine survival loop in tumor precursor cells, and decreased precursor cell numbers. Furthermore, the increase in size of sarcomas from xenografts doubly deficient in these genes was also found to be TGF-β-dependent, in this case resulting from cell nonautonomous effects on endothelial cells and myofibroblasts. Extending these findings in clinical specimens, we documented an increase in TGF-β ligands and an absence of TGF-β receptor II in malignant peripheral nerve sheath tumors, which correspond to tumors in the Nf1-deficient mouse model. Together, our findings reveal R-Ras2 as a critical regulator of TGF-β signaling in vivo.
Collapse
Affiliation(s)
- Deanna M Patmore
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Heermann S, Spittau B, Zajzon K, Schwab MH, Krieglstein K. Schwann cells migrate along axons in the absence of GDNF signaling. BMC Neurosci 2012; 13:92. [PMID: 22863354 PMCID: PMC3445819 DOI: 10.1186/1471-2202-13-92] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 07/19/2012] [Indexed: 01/23/2023] Open
Abstract
Background During development neural crest derived Schwann Cell (SC) precursors migrate to nerve trunks and populate nascent nerves. Axonal ensheathment by SC is a prerequisite for normal nerve function and the integrity of myelinated as well as nonmyelinated axons. To provide adequate support functions, SC colonize entire nerves. One important prerequisite for this is their migration into distal axonal regions. Results Here, we studied the role of Glial cell line derived neurotrophic factor (GDNF), a TGF-beta related growth factor, for SC migration. To this end we used a superior cervical ganglion (SCG) explant-SC migration assay, GDNF null mutant mouse embryos and a chemical inhibitor for GDNF signaling in combination with time-lapse imaging. We found that GDNF signaling is dispensable for SC migration along murine embryonic sympathetic axons. Furthermore, in vivo analyzes revealed that SC migration along the sciatic nerve is also not dependent on GDNF. Conclusions In contrast to previous in vitro findings in the sciatic nerve and a SC precursor cell line, our results clearly indicate that GDNF is dispensable for embryonic SC migration. This is demonstrated for the sympathetic nervous system and also for the sciatic nerve in mouse.
Collapse
Affiliation(s)
- Stephan Heermann
- Department of Neuroanatomy, University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | |
Collapse
|
35
|
The RNA-binding protein human antigen R controls global changes in gene expression during Schwann cell development. J Neurosci 2012; 32:4944-58. [PMID: 22492050 DOI: 10.1523/jneurosci.5868-11.2012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
An important prerequisite to myelination in peripheral nerves is the establishment of one-to-one relationships between axons and Schwann cells. This patterning event depends on immature Schwann cell proliferation, apoptosis, and morphogenesis, which are governed by coordinated changes in gene expression. Here, we found that the RNA-binding protein human antigen R (HuR) was highly expressed in immature Schwann cells, where genome-wide identification of its target mRNAs in vivo in mouse sciatic nerves using ribonomics showed an enrichment of functionally related genes regulating these processes. HuR coordinately regulated expression of several genes to promote proliferation, apoptosis, and morphogenesis in rat Schwann cells, in response to NRG1, TGFβ, and laminins, three major signals implicated in this patterning event. Strikingly, HuR also binds to several mRNAs encoding myelination-related proteins but, contrary to its typical function, negatively regulated their expression, likely to prevent ectopic myelination during development. These functions of HuR correlated with its abundance and subcellular localization, which were regulated by different signals in Schwann cells.
Collapse
|
36
|
Yamazaki S, Ema H, Karlsson G, Yamaguchi T, Miyoshi H, Shioda S, Taketo MM, Karlsson S, Iwama A, Nakauchi H. Nonmyelinating Schwann cells maintain hematopoietic stem cell hibernation in the bone marrow niche. Cell 2012; 147:1146-58. [PMID: 22118468 DOI: 10.1016/j.cell.2011.09.053] [Citation(s) in RCA: 561] [Impact Index Per Article: 43.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2010] [Revised: 04/12/2011] [Accepted: 09/06/2011] [Indexed: 01/02/2023]
Abstract
Hematopoietic stem cells (HSCs) reside and self-renew in the bone marrow (BM) niche. Overall, the signaling that regulates stem cell dormancy in the HSC niche remains controversial. Here, we demonstrate that TGF-β type II receptor-deficient HSCs show low-level Smad activation and impaired long-term repopulating activity, underlining the critical role of TGF-β/Smad signaling in HSC maintenance. TGF-β is produced as a latent form by a variety of cells, so we searched for those that express activator molecules for latent TGF-β. Nonmyelinating Schwann cells in BM proved responsible for activation. These glial cells ensheathed autonomic nerves, expressed HSC niche factor genes, and were in contact with a substantial proportion of HSCs. Autonomic nerve denervation reduced the number of these active TGF-β-producing cells and led to rapid loss of HSCs from BM. We propose that glial cells are components of a BM niche and maintain HSC hibernation by regulating activation of latent TGF-β.
Collapse
Affiliation(s)
- Satoshi Yamazaki
- Japan Science and Technology Agency, ERATO, Chiyoda-ku, Tokyo 102-0075, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Heermann S, Schmücker J, Hinz U, Rickmann M, Unterbarnscheidt T, Schwab MH, Krieglstein K. Neuregulin 1 type III/ErbB signaling is crucial for Schwann cell colonization of sympathetic axons. PLoS One 2011; 6:e28692. [PMID: 22194888 PMCID: PMC3241675 DOI: 10.1371/journal.pone.0028692] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 11/14/2011] [Indexed: 11/24/2022] Open
Abstract
Analysis of Schwann cell (SC) development has been hampered by the lack of growing axons in many commonly used in vitro assays. As a consequence, the molecular signals and cellular dynamics of SC development along peripheral axons are still only poorly understood. Here we use a superior cervical ganglion (SCG) explant assay, in which axons elongate after treatment with nerve growth factor (NGF). Migration as well as proliferation and apoptosis of endogenous SCG-derived SCs along sympathetic axons were studied in these cultures using pharmacological interference and time-lapse imaging. Inhibition of ErbB receptor tyrosine kinases leads to reduced SC proliferation, increased apoptosis and thereby severely interfered with SC migration to distal axonal sections and colonization of axons. Furthermore we demonstrate that SC colonization of axons is also strongly impaired in a specific null mutant of an ErbB receptor ligand, Neuregulin 1 (NRG1) type III. Taken together, using a novel SC development assay, we demonstrate that NRG1 type III serves as a critical axonal signal for glial ErbB receptors that drives SC development along sympathetic axons.
Collapse
Affiliation(s)
- Stephan Heermann
- Department of Neuroanatomy, University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | | | |
Collapse
|
38
|
Berti C, Bartesaghi L, Ghidinelli M, Zambroni D, Figlia G, Chen ZL, Quattrini A, Wrabetz L, Feltri ML. Non-redundant function of dystroglycan and β1 integrins in radial sorting of axons. Development 2011; 138:4025-37. [PMID: 21862561 DOI: 10.1242/dev.065490] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Radial sorting allows the segregation of axons by a single Schwann cell (SC) and is a prerequisite for myelination during peripheral nerve development. Radial sorting is impaired in models of human diseases, congenital muscular dystrophy (MDC) 1A, MDC1D and Fukuyama, owing to loss-of-function mutations in the genes coding for laminin α2, Large or fukutin glycosyltransferases, respectively. It is not clear which receptor(s) are activated by laminin 211, or glycosylated by Large and fukutin during sorting. Candidates are αβ1 integrins, because their absence phenocopies laminin and glycosyltransferase deficiency, but the topography of the phenotypes is different and β1 integrins are not substrates for Large and fukutin. By contrast, deletion of the Large and fukutin substrate dystroglycan does not result in radial sorting defects. Here, we show that absence of dystroglycan in a specific genetic background causes sorting defects with topography identical to that of laminin 211 mutants, and recapitulating the MDC1A, MDC1D and Fukuyama phenotypes. By epistasis studies in mice lacking one or both receptors in SCs, we show that only absence of β1 integrins impairs proliferation and survival, and arrests radial sorting at early stages, that β1 integrins and dystroglycan activate different pathways, and that the absence of both molecules is synergistic. Thus, the function of dystroglycan and β1 integrins is not redundant, but is sequential. These data identify dystroglycan as a functional laminin 211 receptor during axonal sorting and the key substrate relevant to the pathogenesis of glycosyltransferase congenital muscular dystrophies.
Collapse
Affiliation(s)
- Caterina Berti
- Divisions of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milano, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Fei W, Aixi Y, Danmou X, Wusheng K, Zhengren P, Ting R. The mood stabilizer valproic acid induces proliferation and myelination of rat Schwann cells. Neurosci Res 2011; 70:383-90. [PMID: 21530595 DOI: 10.1016/j.neures.2011.04.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2011] [Revised: 04/06/2011] [Accepted: 04/08/2011] [Indexed: 01/26/2023]
Abstract
Schwann cells (SCs) within peripheral nerve respond robustly after exposure to neurotrophic factors. Recent results have revealed that valproic acid (VPA), at a clinically relevant therapeutic concentration, produces effects similar to neurotrophic factors, and promotes neurite growth and cell survival. We hypothesized that VPA could also induce Schwann cell response. In this study, we sought to determine how pure Schwann cells responded to VPA by evaluating for proliferation, expression of S-100, growth cone-associated protein 43 (GAP-43), myelin-associated glycoprotein (MAG), and myelin basic protein (MBP). Immunohistochemistry demonstrated that the Schwann cells were positive for S-100, GAP-43, MAG, and MBP greater than 99% of the experimental cells. The rate of proliferation was increased in experimental cells from MTT assay and Bromodeoxyuridine/DAPI double staining. Furthermore, Western blot showed an up-regulation in GAP-43, MAG and MBP protein expression in experimental cells, respectively. We also found that mitogen-activated protein kinase (MAPK)/extracellular signal-regulated kinase (ERK) 1/2 pathway was involved in the enhanced cell proliferation of Schwann cells evoked by VPA. This study provides novel information regarding Schwann cell response to VPA, which might help the understanding of VPA-based treatment for peripheral nerve injury.
Collapse
Affiliation(s)
- Wu Fei
- Department of Hand Surgery & Microsurgery, Affiliated Pu Ai Hospital of Tongji Medical College of Huazhong University of Science and Technology, Wuhan City, Hu Bei Province, People's Republic of China
| | | | | | | | | | | |
Collapse
|
40
|
Zhou S, Yu B, Qian T, Yao D, Wang Y, Ding F, Gu X. Early changes of microRNAs expression in the dorsal root ganglia following rat sciatic nerve transection. Neurosci Lett 2011; 494:89-93. [PMID: 21371527 DOI: 10.1016/j.neulet.2011.02.064] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Revised: 02/19/2011] [Accepted: 02/23/2011] [Indexed: 12/13/2022]
Abstract
MicroRNAs (miRNAs) are a novel class of small non-coding RNAs that regulate gene expression at the post-transcriptional level. Here we report early alterations of miRNAs expression following rat sciatic nerve injury using microarray analysis. We harvested dorsal root ganglia (DRG) tissues and identified 19 miRNAs that showed significant changes at four early time points after sciatic nerve transection. Subsequently, miR-188 and miR-500 microarray results were verified by real-time quantitative reverse transcriptase polymerase chain reaction (qRT-PCR). The bioinformatics analysis indicated that the potential targets for these miRNAs were involved in the intracellular signaling cascade, the regulation of signal transduction, the regulation of cellular process and the response to cAMP that were known to play important roles in mobilizing the inherent capacity for neurite outgrowth and promoting regeneration during the early phase of sciatic nerve injury. Our results show that abnormal expression of miRNAs may contribute to illustrate the molecular mechanisms of nerve regeneration and miRNAs are potential targets for therapeutic interventions that may enhance intrinsic regenerative ability.
Collapse
Affiliation(s)
- Songlin Zhou
- Jiangsu Key Laboratory of Neuroregeneration, Nantong University, 19 Qixiu Road, Nantong, JS 226001, PR China
| | | | | | | | | | | | | |
Collapse
|
41
|
Arthur-Farraj P, Wanek K, Hantke J, Davis CM, Jayakar A, Parkinson DB, Mirsky R, Jessen KR. Mouse schwann cells need both NRG1 and cyclic AMP to myelinate. Glia 2011; 59:720-33. [PMID: 21322058 DOI: 10.1002/glia.21144] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Accepted: 12/20/2010] [Indexed: 12/13/2022]
Abstract
Genetically modified mice have been a major source of information about the molecular control of Schwann-cell myelin formation, and the role of β-neuregulin 1 (NRG1) in this process in vivo. In vitro, on the other hand, Schwann cells from rats have been used in most analyses of the signaling pathways involved in myelination. To correlate more effectively in vivo and in vitro data, we used purified cultures of mouse Schwann cells in addition to rat Schwann cells to examine two important myelin-related signals, cyclic adenosine monophosphate (cAMP), and NRG1 and to determine whether they interact to control myelin differentiation. We find that in mouse Schwann cells, neither cAMP nor NRG1, when used separately, induced markers of myelin differentiation. When combined, however, they induced strong protein expression of the myelin markers, Krox-20 and P(0) . Importantly, the level of cAMP signaling was crucial in switching NRG1 from a proliferative signal to a myelin differentiation signal. Also in cultured rat Schwann cells, NRG1 promoted cAMP-induced Krox-20 and P(0) expression. Finally, we found that cAMP/NRG1-induced Schwann-cell differentiation required the activity of the cAMP response element binding family of transcription factors in both mouse and rat cells. These observations reconcile observations in vivo and on neuron-Schwann-cell cultures with studies on purified Schwann cells. They demonstrate unambiguously the promyelin effects of NRG1 in purified cells, and they show that the cAMP pathway determines whether NRG1 drives proliferation or induces myelin differentiation.
Collapse
Affiliation(s)
- Peter Arthur-Farraj
- Department of Cell and Developmental Biology, University College London, Gower Street, London WC1E 6BT, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Chaballe L, Close P, Sempels M, Delstanche S, Fanielle J, Moons L, Carmeliet P, Schoenen J, Chariot A, Franzen R. Involvement of placental growth factor in Wallerian degeneration. Glia 2010; 59:379-96. [PMID: 21264946 DOI: 10.1002/glia.21108] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 10/18/2010] [Indexed: 01/13/2023]
Abstract
Wallerian degeneration (WD) is an inflammatory process of nerve degeneration, which occurs more rapidly in the peripheral nervous system compared with the central nervous system, resulting, respectively in successful and aborted axon regeneration. In the peripheral nervous system, Schwann cells (SCs) and macrophages, under the control of a network of cytokines and chemokines, represent the main cell types involved in this process. Within this network, the role of placental growth factor (PlGF) remains totally unknown. However, properties like monocyte activation/attraction, ability to increase expression of pro-inflammatory molecules, as well as neuroprotective effects, make it a candidate likely implicated in this process. Also, nothing is described about the expression and localization of this molecule in the peripheral nervous system. To address these original questions, we decided to study PlGF expression under physiological and degenerative conditions and to explore its role in WD, using a model of sciatic nerve transection in wild-type and Pgf(-/-) mice. Our data show dynamic changes of PlGF expression, from periaxonal in normal nerve to SCs 24h postinjury, in parallel with a p65/NF-κB recruitment on Pgf promoter. After injury, SC proliferation is reduced by 30% in absence of PlGF. Macrophage invasion is significantly delayed in Pgf(-/-) mice compared with wild-type mice, which results in worse functional recovery. MCP-1 and proMMP-9 exhibit a 3-fold reduction of their relative expressions in Pgf(-/-) injured nerves, as demonstrated by cytokine array. In conclusion, this work originally describes PlGF as a novel member of the cytokine network of WD.
Collapse
Affiliation(s)
- Linda Chaballe
- GIGA Neurosciences, Axonal Regeneration and Cephalic Pain unit, University of Liege, Avenue de l'Hopital, Liege, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Sun J, Zhou W, Sha B, Yang Y. Ischemia induced neural stem cell proliferation and differentiation in neonatal rat involved vascular endothelial growth factor and transforming growth factor-beta pathways. Brain Dev 2010; 32:191-200. [PMID: 19232476 DOI: 10.1016/j.braindev.2009.01.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2008] [Revised: 12/30/2008] [Accepted: 01/16/2009] [Indexed: 11/24/2022]
Abstract
Brain ischemia is a leading cause of mortality and morbidity in premature infants. Knowing the fate of neural stem cells in the subventricular zone (SVZ) after ischemia and the mechanisms that determine this fate would be useful in manipulating neural stem cell proliferation and differentiation and possibly in reversing ischemic damage. We sought to identify the genes involved in the proliferation and differentiation of neural stem cells after exposure to ischemia in a 3-day-old rat model that approximates ischemia in premature infants. Proliferating cells were labeled by bromodeoxyuridine (BrdU) through intraperitoneal injection. Using immunfluorescence assays, we observed the proliferation and differentiation of neural stem cells. Genes were identified with GeneChip and real-time quantitative polymerase chain reaction analysis. Ischemic rats had more BrdU-positive cells in the SVZ at all four time points and more neural stem cells differentiation into neurons, astrocytes, and oligodendrocytes. GeneChip analysis showed a 3- to 10-fold increase in the mRNA expression of vascular endothelial growth factor, transforming growth factor-beta, and their receptors in the SVZ. PCR assays and Western blot analyses confirmed these results, indicating that vascular endothelial growth factor and transforming growth factor-beta might be two of the factors that involve post-ischemic neural stem cell proliferation and differentiation.
Collapse
Affiliation(s)
- Jinqiao Sun
- Institute of Pediatrics, Children's Hospital, Fudan University, Shanghai, China
| | | | | | | |
Collapse
|
44
|
Smad3 deficiency reduces neurogenesis in adult mice. J Mol Neurosci 2010; 41:383-96. [PMID: 20155334 DOI: 10.1007/s12031-010-9329-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Accepted: 01/06/2010] [Indexed: 12/11/2022]
Abstract
Transforming growth factor-beta signaling through Smad3 inhibits cell proliferation in many cell types. As cell proliferation in the brain is an integral part of neurogenesis, we sought to determine the role of Smad3 in adult neurogenesis through examining processes and structures important to neurogenesis in adult Smad3 null mice. We find that there are fewer proliferating cells in neurogenic regions of adult Smad3 null mouse brains and reduced migration of neuronal precursor cells from the subventricular zone to the olfactory bulb. Alterations in astrocyte number and distribution within the rostral migratory stream of Smad3 null mice give rise to a smaller and more disorganized structure that may impact on neuronal precursor cell migration. However, the proportion of proliferating cells that become neurons is similar in wild type and Smad3 null mice. Our results suggest that signaling through Smad3 is needed to maintain the rate of cell division of neuronal precursors in the adult brain and hence the amount of neurogenesis, without altering neuronal cell fate.
Collapse
|
45
|
Kritis A, Kapoukranidou D, Michailidou B, Hatzisotiriou A, Albani M. Sciatic nerve crush evokes a biphasic TGF-beta and decorin modulation in the rat spinal cord. Hippokratia 2010; 14:37-41. [PMID: 20411058 PMCID: PMC2843569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
BACKGROUND AND AIM Inherent property of the motoneurons of the peripheral nervous system is their ability to recover, at least in part, upon injury. To this end different factors are expressed and are thought to play important role in the regeneration processes. These factors are diverse, and range from transcription factors and chemokines, to molecules of the extracellular matrix. Transforming growth factor beta (TGF-beta) is a protein with diverse actions controlling cell growth and proliferation. In the extracellular matrix it is found bound to decorin a proteoglycan involved in cell adhesion and cell signaling. In the present study we investigate the expression of TGF-beta and decorin at different time points, in the regenerating sciatic nerve of a seven day old rat, having suffered nerve crush injury, over a period of one month. MATERIALS AND METHODS To achieve this, we evoked injury to male Wistar rats by exposing and applying pressure to the sciatic nerve using watchmaker's forceps. After that at 12 h, 24 h, 48 h, 72 h, one week, and one month intervals we investigated the gene expression of decorin using RT-PCR, and followed the expression of TGF-beta molecule by immunohistochemistry in frozen sections of the L4-L5 region of the rat spinal cord. RESULTS We report that both decorin mRNA and TGF- protein exhibit a concerted, biphasic expression after 12 hours and one month having the animal suffered the nerve crush. DISCUSSION Our data reveal a biphasic modulation of TGF-beta protein and decorin mRNA expression at lumbar segment of the spinal cord of animals having suffered unilateral sciatic nerve crush. We postulate that their concerted expression both at an early and a late phase after the nerve injury is of importance and can be part of a repair or neuroprotective mechanism as yet unclarified.
Collapse
Affiliation(s)
- A Kritis
- Laboratory of Physiology, Department of Physiology and Pharmacology, Medical School, Aristotle University of Thessaloniki, and 2nd Department of Neurology, AHEPA University Hospital, Thessaloniki, Greece.
| | | | | | | | | |
Collapse
|
46
|
Ribeiro-Resende VT, Koenig B, Nichterwitz S, Oberhoffner S, Schlosshauer B. Strategies for inducing the formation of bands of Büngner in peripheral nerve regeneration. Biomaterials 2009; 30:5251-9. [PMID: 19632717 DOI: 10.1016/j.biomaterials.2009.07.007] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Accepted: 07/06/2009] [Indexed: 11/18/2022]
Abstract
Peripheral human nerves fail to regenerate across longer tube implants (>2 cm), most likely because implants lack the microarchitecture of native nerves, including bands of Büngner. Bands of Büngner comprise longitudinally aligned Schwann cell strands that guide selectively regrowing axons. We aim to optimize tubular implants by integrating artificial bands of Büngner. Three principle strategies for inducing the formation of bands of Büngner were investigated: (a) an aligned extracellular matrix, (b) polarizing differentiation factors, and (c) microstructured biomaterial filaments. In vitro oriented collagen and a combination of differentiation factors (NGF, neuregulin-1, TGF-beta) induced Schwann cell alignment to some extent. The most pronounced Schwann cell alignment was evident on ultrathin, endless poly-epsilon-caprolactone (PCL) filaments with longitudinal microgrooves. Precoated PCL filaments proved to be non-cytotoxic, displayed good cell attachment, and supported Schwann cell proliferation as well as guided axonal outgrowth. In vitro on PCL filaments Schwann cells displayed a polarized expression of the cell adhesion molecule L1 similar to that seen in vivo in bands of Büngner after sciatic nerve crush in adult rats. In summary, the integration of bioengineered bands of Büngner based on microstructured polymer filaments in nerve conduits promises to be the most valuable approach to initiating a more efficient regeneration across longer nerve lesions.
Collapse
Affiliation(s)
- Victor T Ribeiro-Resende
- NMI Naturwissenschaftliches und Medizinisches, Institut an der Universität Tübingen, Markwiesenstr. 55, D-72770 Reutlingen, Germany
| | | | | | | | | |
Collapse
|
47
|
Woodhoo A, Alonso MBD, Droggiti A, Turmaine M, D'Antonio M, Parkinson DB, Wilton DK, Al-Shawi R, Simons P, Shen J, Guillemot F, Radtke F, Meijer D, Feltri ML, Wrabetz L, Mirsky R, Jessen KR. Notch controls embryonic Schwann cell differentiation, postnatal myelination and adult plasticity. Nat Neurosci 2009; 12:839-47. [PMID: 19525946 PMCID: PMC2782951 DOI: 10.1038/nn.2323] [Citation(s) in RCA: 239] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2009] [Accepted: 03/30/2009] [Indexed: 11/09/2022]
Abstract
Notch signaling is central to vertebrate development, and analysis of Notch has provided important insights into pathogenetic mechanisms in the CNS and many other tissues. However, surprisingly little is known about the role of Notch in the development and pathology of Schwann cells and peripheral nerves. Using transgenic mice and cell cultures, we found that Notch has complex and extensive regulatory functions in Schwann cells. Notch promoted the generation of Schwann cells from Schwann cell precursors and regulated the size of the Schwann cell pool by controlling proliferation. Notch inhibited myelination, establishing that myelination is subject to negative transcriptional regulation that opposes forward drives such as Krox20. Notably, in the adult, Notch dysregulation resulted in demyelination; this finding identifies a signaling pathway that induces myelin breakdown in vivo. These findings are relevant for understanding the molecular mechanisms that control Schwann cell plasticity and underlie nerve pathology, including demyelinating neuropathies and tumorigenesis.
Collapse
Affiliation(s)
- Ashwin Woodhoo
- Department of Cell and Developmental Biology, University College London, UK
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Woodhoo A, Sommer L. Development of the Schwann cell lineage: from the neural crest to the myelinated nerve. Glia 2009; 56:1481-1490. [PMID: 18803317 DOI: 10.1002/glia.20723] [Citation(s) in RCA: 166] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The myelinating and nonmyelinating Schwann cells in peripheral nerves are derived from the neural crest, which is a transient and multipotent embryonic structure that also generates the other main glial subtypes of the peripheral nervous system (PNS). Schwann cell development occurs through a series of transitional embryonic and postnatal phases, which are tightly regulated by a number of signals. During the early embryonic phases, neural crest cells are specified to give rise to Schwann cell precursors, which represent the first transitional stage in the Schwann cell lineage, and these then generate the immature Schwann cells. At birth, the immature Schwann cells differentiate into either the myelinating or nonmyelinating Schwann cells that populate the mature nerve trunks. In this review, we will discuss the biology of the transitional stages in embryonic and early postnatal Schwann cell development, including the phenotypic differences between them and the recently identified signaling pathways, which control their differentiation and maintenance. In addition, the role and importance of the microenvironment in which glial differentiation takes place will be discussed.
Collapse
Affiliation(s)
- Ashwin Woodhoo
- Department of Anatomy and Developmental Biology, University College London, London, United Kingdom.
| | | |
Collapse
|
49
|
Mirsky R, Woodhoo A, Parkinson DB, Arthur-Farraj P, Bhaskaran A, Jessen KR. Novel signals controlling embryonic Schwann cell development, myelination and dedifferentiation. J Peripher Nerv Syst 2008; 13:122-35. [PMID: 18601657 DOI: 10.1111/j.1529-8027.2008.00168.x] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Immature Schwann cells found in perinatal rodent nerves are generated from Schwann cell precursors (SCPs) that originate from the neural crest. Immature Schwann cells generate the myelinating and non-myelinating Schwann cells of adult nerves. When axons degenerate following injury, Schwann cells demyelinate, proliferate and dedifferentiate to assume a molecular phenotype similar to that of immature cells, a process essential for successful nerve regeneration. Increasing evidence indicates that Schwann cell dedifferentiation involves activation of specific receptors, intracellular signalling pathways and transcription factors in a manner analogous to myelination. We have investigated the roles of Notch and the transcription factor c-Jun in development and after nerve transection. In vivo, Notch signalling regulates the transition from SCP to Schwann cell, times Schwann cell generation, controls Schwann cell proliferation and acts as a brake on myelination. Notch is elevated in injured nerves where it accelerates the rate of dedifferentiation. Likewise, the transcription factor c-Jun is required for Schwann cell proliferation and death and is down-regulated by Krox-20 on myelination. Forced expression of c-Jun in Schwann cells prevents myelination, and in injured nerves, c-Jun is required for appropriate dedifferentiation, the re-emergence of the immature Schwann cell state and nerve regeneration. Thus, both Notch and c-Jun are negative regulators of myelination. The growing realisation that myelination is subject to negative as well as positive controls and progress in molecular identification of negative regulators is likely to impact on our understanding of demyelinating disease and mechanisms that control nerve repair.
Collapse
Affiliation(s)
- Rhona Mirsky
- Department of Cell and Developmental Biology, University College London, London, UK.
| | | | | | | | | | | |
Collapse
|
50
|
Jacob C, Grabner H, Atanasoski S, Suter U. Expression and localization of Ski determine cell type-specific TGFbeta signaling effects on the cell cycle. ACTA ACUST UNITED AC 2008; 182:519-30. [PMID: 18695043 PMCID: PMC2500137 DOI: 10.1083/jcb.200710161] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Transforming growth factor β (TGFβ) promotes epithelial cell differentiation but induces Schwann cell proliferation. We show that the protooncogene Ski (Sloan-Kettering viral oncogene homologue) is an important regulator of these effects. TGFβ down-regulates Ski in epithelial cells but not in Schwann cells. In Schwann cells but not in epithelial cells, retinoblastoma protein (Rb) is up-regulated by TGFβ. Additionally, both Ski and Rb move to the cytoplasm, where they partially colocalize. In vivo, Ski and phospho-Rb (pRb) appear to interact in the Schwann cell cytoplasm of developing sciatic nerves. Ski overexpression induces Rb hyperphosphorylation, proliferation, and colocalization of both proteins in Schwann cell and epithelial cell cytoplasms independently of TGFβ treatment. Conversely, Ski knockdown in Schwann cells blocks TGFβ-induced proliferation and pRb cytoplasmic relocalization. Our findings reveal a critical function of fine-tuned Ski levels in the control of TGFβ effects on the cell cycle and suggest that at least a part of Ski regulatory effects on TGFβ-induced proliferation of Schwann cells is caused by its concerted action with Rb.
Collapse
Affiliation(s)
- Claire Jacob
- Department of Biology, Institute of Cell Biology, ETH Zurich, CH-8093 Zurich, Switzerland.
| | | | | | | |
Collapse
|