1
|
Schneider A, Won S, Armstrong EA, Cooper AJ, Suresh A, Rivera R, Barrett-Wilt G, Denu JM, Simcox JA, Svaren J. The role of ATP citrate lyase in myelin formation and maintenance. Glia 2024. [PMID: 39318247 DOI: 10.1002/glia.24620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/05/2024] [Accepted: 09/14/2024] [Indexed: 09/26/2024]
Abstract
Formation of myelin by Schwann cells is tightly coupled to peripheral nervous system development and is important for neuronal function and long-term maintenance. Perturbation of myelin causes a number of specific disorders that are among the most prevalent diseases affecting the nervous system. Schwann cells synthesize myelin lipids de novo rather than relying on uptake of circulating lipids, yet one unresolved matter is how acetyl CoA, a central metabolite in lipid formation is generated during myelin formation and maintenance. Recent studies have shown that glucose-derived acetyl CoA itself is not required for myelination. However, the importance of mitochondrially-derived acetyl CoA has never been tested for myelination in vivo. Therefore, we have developed a Schwann cell-specific knockout of the ATP citrate lyase (Acly) gene to determine the importance of mitochondrial metabolism to supply acetyl CoA in nerve development. Intriguingly, the ACLY pathway is important for myelin maintenance rather than myelin formation. In addition, ACLY is required to maintain expression of a myelin-associated gene program and to inhibit activation of the latent Schwann cell injury program.
Collapse
Affiliation(s)
- Andrew Schneider
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Seongsik Won
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Eric A Armstrong
- Wisconsin Institute of Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Aaron J Cooper
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Amulya Suresh
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Rachell Rivera
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | - John M Denu
- Wisconsin Institute of Discovery, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Judith A Simcox
- Howard Hughes Medical Institute, Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - John Svaren
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
2
|
Salzer J, Feltri ML, Jacob C. Schwann Cell Development and Myelination. Cold Spring Harb Perspect Biol 2024; 16:a041360. [PMID: 38503507 PMCID: PMC11368196 DOI: 10.1101/cshperspect.a041360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024]
Abstract
Glial cells in the peripheral nervous system (PNS), which arise from the neural crest, include axon-associated Schwann cells (SCs) in nerves, synapse-associated SCs at the neuromuscular junction, enteric glia, perikaryon-associated satellite cells in ganglia, and boundary cap cells at the border between the central nervous system (CNS) and the PNS. Here, we focus on axon-associated SCs. These SCs progress through a series of formative stages, which culminate in the generation of myelinating SCs that wrap large-caliber axons and of nonmyelinating (Remak) SCs that enclose multiple, small-caliber axons. In this work, we describe SC development, extrinsic signals from the axon and extracellular matrix (ECM) and the intracellular signaling pathways they activate that regulate SC development, and the morphogenesis and organization of myelinating SCs and the myelin sheath. We review the impact of SCs on the biology and integrity of axons and their emerging role in regulating peripheral nerve architecture. Finally, we explain how transcription and epigenetic factors control and fine-tune SC development and myelination.
Collapse
Affiliation(s)
- James Salzer
- Neuroscience Institute, New York University Grossman School of Medicine, New York, New York 10016, USA
| | - M Laura Feltri
- Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York 14203, USA
- IRCCS Neurological Institute Carlo Besta, Milano 20133, Italy
- Department of Biotechnology and Translational Sciences, Universita' Degli Studi di Milano, Milano 20133, Italy
| | - Claire Jacob
- Faculty of Biology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz 55128, Germany
| |
Collapse
|
3
|
Chen Y, Shang T, Sun J, Ji Y, Gong L, Li A, Ding F, Shen M, Zhang Q. Characterization of sciatic nerve myelin sheath during development in C57BL/6 mice. Eur J Neurosci 2024; 60:4503-4517. [PMID: 38951719 DOI: 10.1111/ejn.16457] [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: 11/19/2023] [Revised: 06/05/2024] [Accepted: 06/16/2024] [Indexed: 07/03/2024]
Abstract
Myelin sheath plays important roles in information conduction and nerve injury repair in the peripheral nerve system (PNS). Enhancing comprehension of the structure and components of the myelin sheath in the PNS during development would contribute to a more comprehensive understanding of the developmental and regenerative processes. In this research, the structure of sciatic nerve myelin sheath in C57BL/6 mice from embryonic day 14 (E14) to postnatal 12 months (12M) was observed with transmission electron microscopy. Myelin structure appeared in the sciatic nerve as early as E14, and the number and thickness of myelin lamellar gradually increased with the development until 12M. Transcriptome analysis was performed to show the expressions of myelin-associated genes and transcriptional factors involved in myelin formation. The genes encoding myelin proteins (Mag, Pmp22, Mpz, Mbp, Cnp and Prx) showed the same expression pattern, peaking at postnatal day 7 (P7) and P28 after birth, whereas the negative regulators of myelination (c-Jun, Tgfb1, Tnc, Cyr61, Ngf, Egr1, Hgf and Bcl11a) showed an opposite expression pattern. In addition, the expression of myelin-associated proteins and transcriptional factors was measured by Western blot and immunofluorescence staining. The protein expressions of MAG, PMP22, MPZ, CNPase and PRX increased from E20 to P14. The key transcriptional factor c-Jun co-localized with the Schwann cells Marker S100β and decreased after birth, whereas Krox20/Egr2 increased during development. Our data characterized the structure and components of myelin sheath during the early developmental stages, providing insights for further understanding of PNS development.
Collapse
Affiliation(s)
- Yuhan Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Medical School, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Tongxin Shang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Medical School, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Junjie Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Medical School, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Yuhua Ji
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Medical School, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Leilei Gong
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Medical School, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
- Research and Development Center for E-Learning, Ministry of Education, Beijing, China
| | - Aihong Li
- Department of Neurology, Affiliated Hospital of Nantong University, Medical School, Nantong University, Nantong, China
| | - Fei Ding
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Medical School, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Mi Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Medical School, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| | - Qi Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Medical School, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Nantong University, Nantong, China
| |
Collapse
|
4
|
Wei C, Guo Y, Ci Z, Li M, Zhang Y, Zhou Y. Advances of Schwann cells in peripheral nerve regeneration: From mechanism to cell therapy. Biomed Pharmacother 2024; 175:116645. [PMID: 38729050 DOI: 10.1016/j.biopha.2024.116645] [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: 01/30/2024] [Revised: 04/18/2024] [Accepted: 04/24/2024] [Indexed: 05/12/2024] Open
Abstract
Peripheral nerve injuries (PNIs) frequently occur due to various factors, including mechanical trauma such as accidents or tool-related incidents, as well as complications arising from diseases like tumor resection. These injuries frequently result in persistent numbness, impaired motor and sensory functions, neuropathic pain, or even paralysis, which can impose a significant financial burden on patients due to outcomes that often fall short of expectations. The most frequently employed clinical treatment for PNIs involves either direct sutures of the severed ends or bridging the proximal and distal stumps using autologous nerve grafts. However, autologous nerve transplantation may result in sensory and motor functional loss at the donor site, as well as neuroma formation and scarring. Transplantation of Schwann cells/Schwann cell-like cells has emerged as a promising cellular therapy to reconstruct the microenvironment and facilitate peripheral nerve regeneration. In this review, we summarize the role of Schwann cells and recent advances in Schwann cell therapy in peripheral nerve regeneration. We summarize current techniques used in cell therapy, including cell injection, 3D-printed scaffolds for cell delivery, cell encapsulation techniques, as well as the cell types employed in experiments, experimental models, and research findings. At the end of the paper, we summarize the challenges and advantages of various cells (including ESCs, iPSCs, and BMSCs) in clinical cell therapy. Our goal is to provide the theoretical and experimental basis for future treatments targeting peripheral nerves, highlighting the potential of cell therapy and tissue engineering as invaluable resources for promoting nerve regeneration.
Collapse
Affiliation(s)
- Chuqiao Wei
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yuanxin Guo
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Zhen Ci
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Mucong Li
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China
| | - Yidi Zhang
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China.
| | - Yanmin Zhou
- Department of Oral Implantology, Hospital of Stomatology, Jilin University, Changchun, China; Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun, China.
| |
Collapse
|
5
|
Grove M, Kim H, Pang S, Amaya JP, Hu G, Zhou J, Lemay M, Son YJ. TEAD1 is crucial for developmental myelination, Remak bundles, and functional regeneration of peripheral nerves. eLife 2024; 13:e87394. [PMID: 38456457 PMCID: PMC10959528 DOI: 10.7554/elife.87394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 03/06/2024] [Indexed: 03/09/2024] Open
Abstract
Previously we showed that the hippo pathway transcriptional effectors, YAP and TAZ, are essential for Schwann cells (SCs) to develop, maintain and regenerate myelin . Although TEAD1 has been implicated as a partner transcription factor, the mechanisms by which it mediates YAP/TAZ regulation of SC myelination are unclear. Here, using conditional and inducible knockout mice, we show that TEAD1 is crucial for SCs to develop and regenerate myelin. It promotes myelination by both positively and negatively regulating SC proliferation, enabling Krox20/Egr2 to upregulate myelin proteins, and upregulating the cholesterol biosynthetic enzymes FDPS and IDI1. We also show stage-dependent redundancy of TEAD1 and that non-myelinating SCs have a unique requirement for TEAD1 to enwrap nociceptive axons in Remak bundles. Our findings establish TEAD1 as a major partner of YAP/TAZ in developmental myelination and functional nerve regeneration and as a novel transcription factor regulating Remak bundle integrity.
Collapse
Affiliation(s)
- Matthew Grove
- Department of Neural Sciences, Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple UniversityPhiladelphiaUnited States
| | - Hyukmin Kim
- Department of Neural Sciences, Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple UniversityPhiladelphiaUnited States
| | - Shuhuan Pang
- Department of Neural Sciences, Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple UniversityPhiladelphiaUnited States
| | - Jose Paz Amaya
- Department of Bioengineering, Temple UniversityPhiladelphiaUnited States
| | - Guoqing Hu
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta UniversityAugustaUnited States
| | - Jiliang Zhou
- Department of Pharmacology & Toxicology, Medical College of Georgia, Augusta UniversityAugustaUnited States
| | - Michel Lemay
- Department of Bioengineering, Temple UniversityPhiladelphiaUnited States
| | - Young-Jin Son
- Department of Neural Sciences, Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple UniversityPhiladelphiaUnited States
| |
Collapse
|
6
|
Grove M, Kim H, Pang S, Amaya JP, Hu G, Zhou J, Lemay M, Son YJ. TEAD1 is crucial for developmental myelination, Remak bundles, and functional regeneration of peripheral nerves. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.02.27.530298. [PMID: 38293102 PMCID: PMC10827063 DOI: 10.1101/2023.02.27.530298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
Previously we showed that the hippo pathway transcriptional effectors, YAP and TAZ, are essential for Schwann cells (SCs) to develop, maintain and regenerate myelin (Grove et al., 2017; Grove, Lee, Zhao, & Son, 2020). Although TEAD1 has been implicated as a partner transcription factor, the mechanisms by which it mediates YAP/TAZ regulation of SC myelination are unclear. Here, using conditional and inducible knockout mice, we show that TEAD1 is crucial for SCs to develop and regenerate myelin. It promotes myelination by both positively and negatively regulating SC proliferation, enabling Krox20/Egr2 to upregulate myelin proteins, and upregulating the cholesterol biosynthetic enzymes FDPS and IDI1. We also show stage-dependent redundancy of TEAD1 and that non-myelinating SCs have a unique requirement for TEAD1 to enwrap nociceptive axons in Remak bundles. Our findings establish TEAD1 as a major partner of YAP/TAZ in developmental myelination and functional nerve regeneration and as a novel transcription factor regulating Remak bundle integrity.
Collapse
|
7
|
Doan RA, Monk KR. Dock1 acts cell-autonomously in Schwann cells to regulate the development, maintenance, and repair of peripheral myelin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.26.564271. [PMID: 37961336 PMCID: PMC10634861 DOI: 10.1101/2023.10.26.564271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Schwann cells, the myelinating glia of the peripheral nervous system (PNS), are critical for myelin development, maintenance, and repair. Rac1 is a known regulator of radial sorting, a key step in developmental myelination, and we previously showed in zebrafish that loss of Dock1, a Rac1-specific guanine nucleotide exchange factor, results in delayed peripheral myelination in development. We demonstrate here that Dock1 is necessary for myelin maintenance and remyelination after injury in adult zebrafish. Furthermore, it performs an evolutionary conserved role in mice, acting cell-autonomously in Schwann cells to regulate peripheral myelin development, maintenance, and repair. Additionally, manipulating Rac1 levels in larval zebrafish reveals that dock1 mutants are sensitized to inhibition of Rac1, suggesting an interaction between the two proteins during PNS development. We propose that the interplay between Dock1 and Rac1 signaling in Schwann cells is required to establish, maintain, and facilitate repair and remyelination within the peripheral nervous system.
Collapse
Affiliation(s)
- Ryan A Doan
- The Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Kelly R Monk
- The Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
8
|
Zhang Y, Shen Y, Zhao L, Zhao Q, Zhao L, Yi S. Transcription Factor BCL11A Regulates Schwann Cell Behavior During Peripheral Nerve Regeneration. Mol Neurobiol 2023; 60:5352-5365. [PMID: 37316757 DOI: 10.1007/s12035-023-03432-6] [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: 02/16/2023] [Accepted: 06/05/2023] [Indexed: 06/16/2023]
Abstract
Nerve injury-induced Schwann cell dedifferentiation helps to construct a favorable microenvironment for axon growth. Transcription factors regulate cell reprogramming and thus may be critical for Schwann cell phenotype switch during peripheral nerve regeneration. Here, we show that transcription factor B-cell lymphoma/leukemia 11A (BCL11A) is up-regulated in Schwann cells of injured peripheral nerves. Bcl11a silencing suppresses Schwann cell viability, decreases Schwann cell proliferation and migration rates, and impairs the debris clearance ability of Schwann cells. Reduced Bcl11a in injured peripheral nerves results in restricted axon elongation and myelin wrapping, leading to recovery failure. Mechanistically, we demonstrate that BCL11A may mediate Schwann cell activity through binding to the promoter of nuclear receptor subfamily 2 group F member 2 (Nr2f2) and regulating Nr2f2 expression. Collectively, we conclude that BCL11A is essential for Schwann cell activation and peripheral nerve regeneration, providing a potential therapeutic target for the treatment of peripheral nerve injury.
Collapse
Affiliation(s)
- Yunsong Zhang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Yinying Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Li Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Qian Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China
| | - Lili Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China.
| | - Sheng Yi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
9
|
Maita KC, Garcia JP, Avila FR, Ricardo A TG, Ho OA, Claudia C S C, Eduardo N C, Forte AJ. Evaluation of the Aging Effect on Peripheral Nerve Regeneration: A Systematic Review. J Surg Res 2023; 288:329-340. [PMID: 37060859 DOI: 10.1016/j.jss.2023.03.017] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/18/2023] [Accepted: 03/16/2023] [Indexed: 04/17/2023]
Abstract
INTRODUCTION Peripheral nerve injuries have been associated with increased healthcare costs and decreased patients' quality of life. Aging represents one factor that slows the speed of peripheral nervous system (PNS) regeneration. Since cellular homeostasis imbalance associated with aging lead to an increased failure in nerve regeneration in mammals of advanced age, this systematic review aims to determine the main molecular and cellular mechanisms involved in peripheral nerve regeneration in aged murine models after a peripheral nerve injuries. METHODS Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, a literature search of 4 databases was conducted in July 2022 for studies comparing the peripheral nerve regeneration capability between young and aged murine models. RESULTS After the initial search yielded 744 publications, ten articles fulfilled the inclusion criteria. These studies show that age-related changes such as chronic inflammatory state, delayed macrophages' response to injury, dysfunctional Schwann Cells (SCs), and microenvironment alterations cause a reduction in the regenerative capability of the PNS in murine models. Furthermore, identifying altered gene expression patterns of SC after nerve damage can contribute to the understanding of physiological modifications produced by aging. CONCLUSIONS The interaction between macrophages and SC plays a crucial role in the nerve regeneration of aged models. Therefore, studies aimed at developing new and promising therapies for nerve regeneration should focus on these cellular groups to enhance the regenerative capabilities of the PNS in elderly populations.
Collapse
Affiliation(s)
- Karla C Maita
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, Florida
| | - John P Garcia
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, Florida
| | | | | | - Olivia A Ho
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, Florida
| | - Chini Claudia C S
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida
| | - Chini Eduardo N
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, Florida
| | - Antonio J Forte
- Division of Plastic Surgery, Mayo Clinic, Jacksonville, Florida.
| |
Collapse
|
10
|
Willows JW, Gunsch G, Paradie E, Blaszkiewicz M, Tonniges JR, Pino MF, Smith SR, Sparks LM, Townsend KL. Schwann cells contribute to demyelinating diabetic neuropathy and nerve terminal structures in white adipose tissue. iScience 2023; 26:106189. [PMID: 36895649 PMCID: PMC9989657 DOI: 10.1016/j.isci.2023.106189] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 12/09/2022] [Accepted: 02/08/2023] [Indexed: 02/15/2023] Open
Abstract
Peripheral neuropathy, which can include axonal degeneration and/or demyelination, impacts adipose tissues with obesity, diabetes, and aging. However, the presence of demyelinating neuropathy had not yet been explored in adipose. Both demyelinating neuropathies and axonopathies implicate Schwann cells (SCs), a glial support cell that myelinates axons and contributes to nerve regeneration after injury. We performed a comprehensive assessment of SCs and myelination patterns of subcutaneous white adipose tissue (scWAT) nerves, and changes across altered energy balance states. We found that mouse scWAT contains both myelinated and unmyelinated nerves and is populated by SCs, including SCs that were associated with synaptic vesicle-containing nerve terminals. BTBR ob/ob mice, a model of diabetic peripheral neuropathy, exhibited small fiber demyelinating neuropathy and alterations in SC marker gene expression in adipose that were similar to obese human adipose. These data indicate that adipose SCs regulate the plasticity of tissue nerves and become dysregulated in diabetes.
Collapse
Affiliation(s)
- Jake W Willows
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Gilian Gunsch
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | - Emma Paradie
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| | | | - Jeffrey R Tonniges
- Campus Microscopy and Imaging Facility, The Ohio State University, Columbus, OH, USA
| | - Maria F Pino
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Steven R Smith
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Lauren M Sparks
- Translational Research Institute, AdventHealth, Orlando, FL, USA
| | - Kristy L Townsend
- Department of Neurological Surgery, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
11
|
Low-intensity pulsed ultrasound promotes proliferation and myelinating genes expression of Schwann cells through NRG1/ErbB signaling pathway. Tissue Cell 2023; 80:101985. [PMID: 36459840 DOI: 10.1016/j.tice.2022.101985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/15/2022] [Accepted: 11/17/2022] [Indexed: 11/22/2022]
Abstract
Schwann cells (SCs) are the major component of myelin sheath in the peripheral nervous system, which are necessary in the development, function maintenance, and repair of peripheral nerves. This study aimed to investigate the potential mechanism of low-intensity pulsed ultrasound (LIPUS) affecting the proliferation and myelinating activity of SCs. Rat Schwann cell line RSC96 were cultured and exposed to LIPUS of different duty ratios (control, 20 %, 50 %, 80 %). Results demonstrated that LIPUS with a duty ratio of 50 % showing the maximal effect in facilitating proliferation of SCs. The expressions of Krox20 and myelin basic protein (MBP), the key molecules of SC myelination, and the potent inducer of myelination neuregulin 1 (NRG1) and its receptors ErbB2 and ErbB3 increased significantly by LIPUS. The reaction of these factors to LIPUS were both time- and duty ratio-dependent: namely LIPUS with higher duty ratios took effects when applied repeatedly over more consecutive days. These observations indicated that NRG1/ErbB signaling pathway might contribute to the effects of LIPUS on the proliferation and myelinating status of SCs, which could be one of the mechanisms in the protective role of LIPUS in nerve repair and regeneration. Our work provided novel insights for promising strategies of nerve repair therapy.
Collapse
|
12
|
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: 19] [Impact Index Per Article: 9.5] [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
|
13
|
Marechal E, Poliard A, Henry K, Moreno M, Legrix M, Macagno N, Mondielli G, Fauquier T, Barlier A, Etchevers HC. Multiple congenital malformations arise from somatic mosaicism for constitutively active Pik3ca signaling. Front Cell Dev Biol 2022; 10:1013001. [PMID: 36353506 PMCID: PMC9637999 DOI: 10.3389/fcell.2022.1013001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 10/07/2022] [Indexed: 11/17/2022] Open
Abstract
Recurrent missense mutations of the PIK3CA oncogene are among the most frequent drivers of human cancers. These often lead to constitutive activation of its product p110α, a phosphatidylinositol 3-kinase (PI3K) catalytic subunit. In addition to causing a broad range of cancers, the H1047R mutation is also found in affected tissues of a distinct set of congenital tumors and malformations. Collectively termed PIK3CA-related disorders (PRDs), these lead to overgrowth of brain, adipose, connective and musculoskeletal tissues and/or blood and lymphatic vessel components. Vascular malformations are frequently observed in PRD, due to cell-autonomous activation of PI3K signaling within endothelial cells. These, like most muscle, connective tissue and bone, are derived from the embryonic mesoderm. However, important organ systems affected in PRDs are neuroectodermal derivatives. To further examine their development, we drove the most common post-zygotic activating mutation of Pik3ca in neural crest and related embryonic lineages. Outcomes included macrocephaly, cleft secondary palate and more subtle skull anomalies. Surprisingly, Pik3ca-mutant subpopulations of neural crest origin were also associated with widespread cephalic vascular anomalies. Mesectodermal neural crest is a major source of non-endothelial connective tissue in the head, but not the body. To examine the response of vascular connective tissues of the body to constitutive Pik3ca activity during development, we expressed the mutation by way of an Egr2 (Krox20) Cre driver. Lineage tracing led us to observe new lineages that had normally once expressed Krox20 and that may be co-opted in pathogenesis, including vascular pericytes and perimysial fibroblasts. Finally, Schwann cell precursors having transcribed either Krox20 or Sox10 and induced to express constitutively active PI3K were associated with vascular and other tumors. These murine phenotypes may aid discovery of new candidate human PRDs affecting craniofacial and vascular smooth muscle development as well as the reciprocal paracrine signaling mechanisms leading to tissue overgrowth.
Collapse
Affiliation(s)
- Elise Marechal
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
| | - Anne Poliard
- URP 2496 Orofacial Pathologies, Imagery and Biotherapies, CNRS, GDR 2031 CREST-NET, Université Paris Cité, Montrouge, France
- School of Dentistry, Université Paris Cité, Montrouge, France
| | - Kilian Henry
- School of Dentistry, Université Paris Cité, Montrouge, France
| | - Mathias Moreno
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
| | - Mathilde Legrix
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
| | - Nicolas Macagno
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
| | - Grégoire Mondielli
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
| | - Teddy Fauquier
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
| | - Anne Barlier
- INSERM, MMG, U1251, MarMaRa Institute, Aix Marseille University, Marseille, France
- AP-HM, MMG, MarMaRa Institute, La Conception Hospital Laboratory of Molecular Biology, Marseille, France
| | - Heather C. Etchevers
- INSERM, MMG, U1251, CNRS, GDR 2031 CREST-NET, MarMaRa Institute, Aix Marseille University, Marseille, France
- *Correspondence: Heather C. Etchevers,
| |
Collapse
|
14
|
McLean JW, Wilson JA, Tian T, Watson JA, VanHart M, Bean AJ, Scherer SS, Crossman DK, Ubogu E, Wilson SM. Disruption of Endosomal Sorting in Schwann Cells Leads to Defective Myelination and Endosomal Abnormalities Observed in Charcot-Marie-Tooth Disease. J Neurosci 2022; 42:5085-5101. [PMID: 35589390 PMCID: PMC9233440 DOI: 10.1523/jneurosci.2481-21.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 04/24/2022] [Accepted: 05/03/2022] [Indexed: 12/24/2022] Open
Abstract
Endosomal sorting plays a fundamental role in directing neural development. By altering the temporal and spatial distribution of membrane receptors, endosomes regulate signaling pathways that control the differentiation and function of neural cells. Several genes linked to inherited demyelinating peripheral neuropathies, known as Charcot-Marie-Tooth (CMT) disease, encode proteins that directly interact with components of the endosomal sorting complex required for transport (ESCRT). Our previous studies demonstrated that a point mutation in the ESCRT component hepatocyte growth-factor-regulated tyrosine kinase substrate (HGS), an endosomal scaffolding protein that identifies internalized cargo to be sorted by the endosome, causes a peripheral neuropathy in the neurodevelopmentally impaired teetering mice. Here, we constructed a Schwann cell-specific deletion of Hgs to determine the role of endosomal sorting during myelination. Inactivation of HGS in Schwann cells resulted in motor and sensory deficits, slowed nerve conduction velocities, delayed myelination and hypomyelinated axons, all of which occur in demyelinating forms of CMT. Consistent with a delay in Schwann cell maturation, HGS-deficient sciatic nerves displayed increased mRNA levels for several promyelinating genes and decreased mRNA levels for genes that serve as markers of myelinating Schwann cells. Loss of HGS also altered the abundance and activation of the ERBB2/3 receptors, which are essential for Schwann cell development. We therefore hypothesize that HGS plays a critical role in endosomal sorting of the ERBB2/3 receptors during Schwann cell maturation, which further implicates endosomal dysfunction in inherited peripheral neuropathies.SIGNIFICANCE STATEMENT Schwann cells myelinate peripheral axons, and defects in Schwann cell function cause inherited demyelinating peripheral neuropathies known as CMT. Although many CMT-linked mutations are in genes that encode putative endosomal proteins, little is known about the requirements of endosomal sorting during myelination. In this study, we demonstrate that loss of HGS disrupts the endosomal sorting pathway in Schwann cells, resulting in hypomyelination, aberrant myelin sheaths, and impairment of the ERBB2/3 receptor pathway. These findings suggest that defective endosomal trafficking of internalized cell surface receptors may be a common mechanism contributing to demyelinating CMT.
Collapse
Affiliation(s)
- John W McLean
- Department of Neurobiology, Evelyn F. McKnight Brain Institute, Civitan International Research Center, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Julie A Wilson
- Department of Neurobiology, Evelyn F. McKnight Brain Institute, Civitan International Research Center, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Tina Tian
- Department of Neurobiology, Evelyn F. McKnight Brain Institute, Civitan International Research Center, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Jennifer A Watson
- Department of Neurobiology, Evelyn F. McKnight Brain Institute, Civitan International Research Center, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Mary VanHart
- Department of Neurobiology, Evelyn F. McKnight Brain Institute, Civitan International Research Center, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Andrew J Bean
- Graduate College, Rush University, Chicago, Illinois 60612
| | - Steven S Scherer
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - David K Crossman
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, 35294
| | - Eroboghene Ubogu
- Department of Neurobiology, Evelyn F. McKnight Brain Institute, Civitan International Research Center, University of Alabama at Birmingham, Birmingham, Alabama 35294
- Division of Neuromuscular Disease, Department of Neurology, University of Alabama at Birmingham, Birmingham, Alabama 35294
| | - Scott M Wilson
- Department of Neurobiology, Evelyn F. McKnight Brain Institute, Civitan International Research Center, University of Alabama at Birmingham, Birmingham, Alabama 35294
| |
Collapse
|
15
|
Guo S, Moore RM, Charlesworth MC, Johnson KL, Spinner RJ, Windebank AJ, Wang H. The proteome of distal nerves: implication in delayed repair and poor functional recovery. Neural Regen Res 2022; 17:1998-2006. [PMID: 35142689 PMCID: PMC8848594 DOI: 10.4103/1673-5374.335159] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Chronic denervation is one of the key factors that affect nerve regeneration. Chronic axotomy deteriorates the distal nerve stump, causes protein changes, and renders the microenvironment less permissive for regeneration. Some of these factors/proteins have been individually studied. To better delineate the comprehensive protein expression profiles and identify proteins that contribute to or are associated with this detrimental effect, we carried out a proteomic analysis of the distal nerve using an established delayed rat sciatic nerve repair model. Four rats that received immediate repair after sciatic nerve transection served as control, whereas four rats in the experimental group (chronic denervation) had their sciatic nerve repaired after a 12-week delay. All the rats were sacrificed after 16 weeks to harvest the distal nerves for extracting proteins. Twenty-five micrograms of protein from each sample were fractionated in SDS-PAGE gels. NanoLC-MS/MS analysis was applied to the gels. Protein expression levels of nerves on the surgery side were compared to those on the contralateral side. Any protein with a P value of less than 0.05 and a fold change of 4 or higher was deemed differentially expressed. All the differentially expressed proteins in both groups were further stratified according to the biological processes. A PubMed search was also conducted to identify the differentially expressed proteins that have been reported to be either beneficial or detrimental to nerve regeneration. Ingenuity Pathway Analysis (IPA) software was used for pathway analysis. The results showed that 709 differentially expressed proteins were identified in the delayed repair group, with a bigger proportion of immune and inflammatory process-related proteins and a smaller proportion of proteins related to axon regeneration and lipid metabolism in comparison to the control group where 478 differentially expressed proteins were identified. The experimental group also had more beneficial proteins that were downregulated and more detrimental proteins that were upregulated. IPA revealed that protective pathways such as LXR/RXR, acute phase response, RAC, ERK/MAPK, CNTF, IL-6, and FGF signaling were inhibited in the delayed repair group, whereas three detrimental pathways, including the complement system, PTEN, and apoptosis signaling, were activated. An available database of the adult rodent sciatic nerve was used to assign protein changes to specific cell types. The poor regeneration seen in the delayed repair group could be associated with the down-regulation of beneficial proteins and up-regulation of detrimental proteins. The proteins and pathways identified in this study may offer clues for future studies to identify therapeutic targets.
Collapse
Affiliation(s)
- Song Guo
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Raymond M Moore
- Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | | | | | - Robert J Spinner
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Huan Wang
- Department of Neurologic Surgery, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
16
|
Taïb S, Lamandé N, Martin S, Coulpier F, Topilko P, Brunet I. Myelinating Schwann cells and Netrin-1 control intra-nervous vascularization of the developing mouse sciatic nerve. eLife 2022; 11:64773. [PMID: 35019839 PMCID: PMC8782568 DOI: 10.7554/elife.64773] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/11/2022] [Indexed: 11/13/2022] Open
Abstract
Peripheral nerves are vascularized by a dense network of blood vessels to guarantee their complex function. Despite the crucial role of vascularization to ensure nerve homeostasis and regeneration, the mechanisms governing nerve invasion by blood vessels remain poorly understood. We found, in mice, that the sciatic nerve invasion by blood vessels begins around embryonic day 16 and continues until birth. Interestingly, intra-nervous blood vessel density significantly decreases during post-natal period, starting from P10. We show that, while the axon guidance molecule Netrin-1 promotes nerve invasion by blood vessels via the endothelial receptor UNC5B during embryogenesis, myelinated Schwann cells negatively control intra-nervous vascularization during post-natal period.
Collapse
Affiliation(s)
- Sonia Taïb
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France
| | - Noël Lamandé
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France
| | - Sabrina Martin
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France
| | - Fanny Coulpier
- UMR U955 INSERM UPEC, Institut Mondor de Recherche Biomédicale, Créteil, France
| | - Piotr Topilko
- UMR U955 INSERM UPEC, Institut Mondor de Recherche Biomédicale, Créteil, France
| | - Isabelle Brunet
- Center for Interdisciplinary Research in Biology, Collège de France, Paris, France
| |
Collapse
|
17
|
Rosso G, Wehner D, Schweitzer C, Möllmert S, Sock E, Guck J, Shahin V. Matrix stiffness mechanosensing modulates the expression and distribution of transcription factors in Schwann cells. Bioeng Transl Med 2022; 7:e10257. [PMID: 35079632 PMCID: PMC8780053 DOI: 10.1002/btm2.10257] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/04/2021] [Accepted: 09/06/2021] [Indexed: 12/22/2022] Open
Abstract
After peripheral nerve injury, mature Schwann cells (SCs) de-differentiate and undergo cell reprogramming to convert into a specialized cell repair phenotype that promotes nerve regeneration. Reprogramming of SCs into the repair phenotype is tightly controlled at the genome level and includes downregulation of pro-myelinating genes and activation of nerve repair-associated genes. Nerve injuries induce not only biochemical but also mechanical changes in the tissue architecture which impact SCs. Recently, we showed that SCs mechanically sense the stiffness of the extracellular matrix and that SC mechanosensitivity modulates their morphology and migratory behavior. Here, we explore the expression levels of key transcription factors and myelin-associated genes in SCs, and the outgrowth of primary dorsal root ganglion (DRG) neurites, in response to changes in the stiffness of generated matrices. The selected stiffness range matches the physiological conditions of both utilized cell types as determined in our previous investigations. We find that stiffer matrices induce upregulation of the expression of transcription factors Sox2, Oct6, and Krox20, and concomitantly reduce the expression of the repair-associated transcription factor c-Jun, suggesting a link between SC substrate mechanosensing and gene expression regulation. Likewise, DRG neurite outgrowth correlates with substrate stiffness. The remarkable intrinsic physiological plasticity of SCs, and the mechanosensitivity of SCs and neurites, may be exploited in the design of bioengineered scaffolds that promote nerve regeneration upon injury.
Collapse
Affiliation(s)
- Gonzalo Rosso
- Max Planck Institute for the Science of LightErlangenGermany
- Max‐Planck‐Zentrum für Physik und MedizinErlangenGermany
- Institute of Physiology II, University of MünsterMünsterGermany
| | - Daniel Wehner
- Max Planck Institute for the Science of LightErlangenGermany
- Max‐Planck‐Zentrum für Physik und MedizinErlangenGermany
| | - Christine Schweitzer
- Max Planck Institute for the Science of LightErlangenGermany
- Max‐Planck‐Zentrum für Physik und MedizinErlangenGermany
| | - Stephanie Möllmert
- Max Planck Institute for the Science of LightErlangenGermany
- Max‐Planck‐Zentrum für Physik und MedizinErlangenGermany
| | - Elisabeth Sock
- Institute of Biochemistry, FAU Erlangen‐NürnbergErlangenGermany
| | - Jochen Guck
- Max Planck Institute for the Science of LightErlangenGermany
- Max‐Planck‐Zentrum für Physik und MedizinErlangenGermany
- Department of PhysicsFAU Erlangen‐NürnbergErlangenGermany
| | - Victor Shahin
- Institute of Physiology II, University of MünsterMünsterGermany
| |
Collapse
|
18
|
Intisar A, Kim WH, Shin HY, Kim MY, Kim YS, Lim H, Kang HG, Mo YJ, Aly MAS, Lee YI, Kim MS. An electroceutical approach enhances myelination via upregulation of lipid biosynthesis in the dorsal root ganglion. Biofabrication 2021; 14. [PMID: 34933294 DOI: 10.1088/1758-5090/ac457c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 12/21/2021] [Indexed: 11/12/2022]
Abstract
As the myelin sheath is crucial for neuronal saltatory conduction, loss of myelin in the peripheral nervous system (PNS) leads to demyelinating neuropathies causing muscular atrophy, numbness, foot deformities and paralysis. Unfortunately, few interventions are available for such neuropathies, because previous pharmaceuticals have shown severe side effects and failed in clinical trials. Therefore, exploring new strategies to enhance PNS myelination is critical to provide solution for such intractable diseases. This study aimed to investigate the effectiveness of electrical stimulation (ES) to enhance myelination in the mouse dorsal root ganglion (DRG) - an ex vivo model of the PNS. Mouse embryonic DRGs were extracted at E13 and seeded onto Matrigel-coated surfaces. After sufficient growth and differentiation, screening was carried out by applying ES in the 1-100 Hz range at the beginning of the myelination process. DRG myelination was evaluated via immunostaining at the intermediate (19 DIV) and mature (30 DIV) stages. Further biochemical analyses were carried out by utilizing RNA sequencing, qPCR and biochemical assays at both intermediate and mature myelination stages. Imaging of DRG myelin lipids was carried out via time-of-flight secondary ion mass spectrometry (ToF-SIMS). With screening ES conditions, optimal condition was identified at 20 Hz, which enhanced the percentage of myelinated neurons and average myelin length not only at intermediate (129% and 61%) but also at mature (72% and 17%) myelination stages. Further biochemical analyses elucidated that ES promoted lipid biosynthesis in the DRG. ToF-SIMS imaging showed higher abundance of the structural lipids, cholesterol and sphingomyelin, in the myelin membrane. Therefore, promotion of lipid biosynthesis and higher abundance of myelin lipids led to ES-mediated myelination enhancement. Given that myelin lipid deficiency is culpable for most demyelinating PNS neuropathies, the results might pave a new way to treat such diseases via electroceuticals.
Collapse
Affiliation(s)
- Aseer Intisar
- New Biology, DGIST, 333 Techno Jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu, 42988, Korea (the Republic of)
| | - Woon-Hae Kim
- CTCELLS Corp., 333 Techno Jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu, 42988, Korea (the Republic of)
| | - Hyun Young Shin
- CTCELLS Corp., 333 Techno Jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu, 42988, Korea (the Republic of)
| | - Min Young Kim
- New Biology, DGIST, 333 Techno Jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu, 42988, Korea (the Republic of)
| | - Yu Seon Kim
- Well Aging Research Center, DGIST, 333 Techno Jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu, 42988, Korea (the Republic of)
| | - Heejin Lim
- New Biology, DGIST, 333 Techno Jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu, 42988, Korea (the Republic of)
| | - Hyun Gyu Kang
- New Biology, DGIST, 333 Techno Jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu, 42988, Korea (the Republic of)
| | - Yun Jeoung Mo
- Well Aging Research Center, DGIST, 333 Techno Jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu, 42988, Korea (the Republic of)
| | - Mohamed Aly Saad Aly
- New Biology, DGIST, 333 Techno Jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu, 42988, Korea (the Republic of)
| | - Yun-Il Lee
- Well Aging Research Center, DGIST, 333 Techno Jungang-daero, Hyeonpung-eup, Dalseong-gun, Daegu, 42988, Korea (the Republic of)
| | - Minseok S Kim
- New Biology, DGIST, Room 313, Building E5, DGIST, Daegu, 42988, Korea (the Republic of)
| |
Collapse
|
19
|
Sardella-Silva G, Mietto BS, Ribeiro-Resende VT. Four Seasons for Schwann Cell Biology, Revisiting Key Periods: Development, Homeostasis, Repair, and Aging. Biomolecules 2021; 11:1887. [PMID: 34944531 PMCID: PMC8699407 DOI: 10.3390/biom11121887] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 01/28/2023] Open
Abstract
Like the seasons of the year, all natural things happen in stages, going through adaptations when challenged, and Schwann cells are a great example of that. During maturation, these cells regulate several steps in peripheral nervous system development. The Spring of the cell means the rise and bloom through organized stages defined by time-dependent regulation of factors and microenvironmental influences. Once matured, the Summer of the cell begins: a high energy stage focused on maintaining adult homeostasis. The Schwann cell provides many neuron-glia communications resulting in the maintenance of synapses. In the peripheral nervous system, Schwann cells are pivotal after injuries, balancing degeneration and regeneration, similarly to when Autumn comes. Their ability to acquire a repair phenotype brings the potential to reconnect axons to targets and regain function. Finally, Schwann cells age, not only by growing old, but also by imposed environmental cues, like loss of function induced by pathologies. The Winter of the cell presents as reduced activity, especially regarding their role in repair; this reflects on the regenerative potential of older/less healthy individuals. This review gathers essential information about Schwann cells in different stages, summarizing important participation of this intriguing cell in many functions throughout its lifetime.
Collapse
Affiliation(s)
- Gabriela Sardella-Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
- Núcleo Multidisciplinar de Pesquisa em Biologia (Numpex-Bio), Campus de Duque de Caxias Geraldo Guerra Cidade, Universidade Federal do Rio de Janeiro, Duque de Caxias 25255-030, RJ, Brazil
| | - Bruno Siqueira Mietto
- Instituto de Ciências Biológicas, Universidade Federal de Juiz de Fora, Juiz de Fora 36036-900, MG, Brazil;
| | - Victor Túlio Ribeiro-Resende
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, RJ, Brazil;
- Núcleo Multidisciplinar de Pesquisa em Biologia (Numpex-Bio), Campus de Duque de Caxias Geraldo Guerra Cidade, Universidade Federal do Rio de Janeiro, Duque de Caxias 25255-030, RJ, Brazil
| |
Collapse
|
20
|
Della-Flora Nunes G, Wilson ER, Hurley E, He B, O'Malley BW, Poitelon Y, Wrabetz L, Feltri ML. Activation of mTORC1 and c-Jun by Prohibitin1 loss in Schwann cells may link mitochondrial dysfunction to demyelination. eLife 2021; 10:e66278. [PMID: 34519641 PMCID: PMC8478418 DOI: 10.7554/elife.66278] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 09/13/2021] [Indexed: 12/26/2022] Open
Abstract
Schwann cell (SC) mitochondria are quickly emerging as an important regulator of myelin maintenance in the peripheral nervous system (PNS). However, the mechanisms underlying demyelination in the context of mitochondrial dysfunction in the PNS are incompletely understood. We recently showed that conditional ablation of the mitochondrial protein Prohibitin 1 (PHB1) in SCs causes a severe and fast progressing demyelinating peripheral neuropathy in mice, but the mechanism that causes failure of myelin maintenance remained unknown. Here, we report that mTORC1 and c-Jun are continuously activated in the absence of Phb1, likely as part of the SC response to mitochondrial damage. Moreover, we demonstrate that these pathways are involved in the demyelination process, and that inhibition of mTORC1 using rapamycin partially rescues the demyelinating pathology. Therefore, we propose that mTORC1 and c-Jun may play a critical role as executioners of demyelination in the context of perturbations to SC mitochondria.
Collapse
Affiliation(s)
- Gustavo Della-Flora Nunes
- Hunter James Kelly Research Institute, University at BuffaloBuffaloUnited States
- Department of Biochemistry, University at BuffaloBuffaloUnited States
| | - Emma R Wilson
- Hunter James Kelly Research Institute, University at BuffaloBuffaloUnited States
- Department of Biochemistry, University at BuffaloBuffaloUnited States
| | - Edward Hurley
- Hunter James Kelly Research Institute, University at BuffaloBuffaloUnited States
| | - Bin He
- Immunobiology & Transplant Science Center and Department of Surgery, Houston Methodist HospitalHoustonUnited States
| | - Bert W O'Malley
- Department of Medicine and Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Yannick Poitelon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical CollegeAlbanyUnited States
| | - Lawrence Wrabetz
- Hunter James Kelly Research Institute, University at BuffaloBuffaloUnited States
- Department of Biochemistry, University at BuffaloBuffaloUnited States
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at BuffaloBuffaloUnited States
| | - M Laura Feltri
- Hunter James Kelly Research Institute, University at BuffaloBuffaloUnited States
- Department of Biochemistry, University at BuffaloBuffaloUnited States
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at BuffaloBuffaloUnited States
| |
Collapse
|
21
|
Yan W, Wu Z, Zhang Y, Hong D, Dong X, Liu L, Rao Y, Huang L, Zhang X, Wu J. The molecular and cellular insight into the toxicology of bortezomib-induced peripheral neuropathy. Biomed Pharmacother 2021; 142:112068. [PMID: 34463262 DOI: 10.1016/j.biopha.2021.112068] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/04/2021] [Accepted: 08/17/2021] [Indexed: 12/26/2022] Open
Abstract
The proteasome inhibitor bortezomib (BTZ) is a first-line antitumor drug, mainly used for multiple myeloma treatment. However, BTZ shows prominent toxicity in the peripheral nervous system, termed BTZ-induced peripheral neuropathy (BIPN). BIPN is characterized by neuropathic pain, resulting in a dose reduction or even treatment withdrawal. To date, the pathological mechanism of BIPN has not been elucidated. There is still no effective strategy to prevent or treat BIPN. This review summarizes the pathological mechanisms of BIPN, which involves the pathological changes of Schwann cells, neurons, astrocytes and macrophages. A better knowledge of the pathological mechanisms of BIPN would provide new ideas for therapeutic interventions of BIPN patients.
Collapse
Affiliation(s)
- Wenping Yan
- Department of Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhanxun Wu
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yuyu Zhang
- Department of Pharmacy, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| | - Dongsheng Hong
- Department of Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xihao Dong
- Department of Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lin Liu
- Department of Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuefeng Rao
- Department of Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lili Huang
- Department of Pharmacy, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
| | - Xiangnan Zhang
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Jiaying Wu
- Department of Pharmacy, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| |
Collapse
|
22
|
Zhao Y, Tian C, Wu P, Chen F, Xiao A, Ye Q, Shi X, Wang Z, Han X, Chen Y. Hydroxypropyl chitosan/soy isolate protein conduits promote peripheral nerve regeneration. Tissue Eng Part A 2021; 28:225-238. [PMID: 34375147 DOI: 10.1089/ten.tea.2021.0068] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Designing scaffolds, with optimized micro-structure and function for promoting the release of neuro-related factors, is significant in peripheral nerve regeneration. Herein, a series of hydroxypropyl chitosan/soy protein isolate composite sponges (HCSS) were fabricated by a freeze-drying technique. The physicochemical properties of the resultant HCSS were examined by a Fourier infrared spectrometer, X-ray diffractometer, scanning electron microscope, water absorption assay, water retention assay, and compressive strength assay. The results indicated that HCSS exhibited an interconnected porous micro-structure and a high water retention ratio with the increase in SPI content. The biological characterization found that the HCSS-50 containing 50% SPI content profoundly promoted the proliferation of RSC96 cells and the secretion of neuro-related factors without excessive ROS production. In addition, HCSS-50 could significantly promote the expression of neuro-related factors; for example, the expression of TGF-β was 3 times higher than that of the control group. Finally, an optimized HCSS-based conduit was fabricated from HCSS-50 to repair sciatic nerve injury in rats with the combination of BMSCs or BMSC-derived Schwann cells. The results suggested that the constructed HCSS-based conduit accompanying BMSC-derived Schwann cells could effectively promote axonal regeneration and upregulate expression of neuro-related factors such as Krox20, Zeb2, and GAP43. Collectively, a newly engineered nerve conduit system was developed by incorporating HCSS-50 and BMSC-derived Schwann cells, which could be an alternative candidate for peripheral nerve regeneration.
Collapse
Affiliation(s)
- Yanan Zhao
- Zhengzhou University First Affiliated Hospital, 191599, Zhengzhou, Henan, China.,Wuhan University School of Basic Medical Sciences, 36841, Wuhan, China;
| | - Chuan Tian
- Zhengzhou University First Affiliated Hospital, 191599, Zhengzhou, Henan, China;
| | - Ping Wu
- Wuhan University School of Basic Medical Sciences, 36841, Wuhan, China;
| | - Feixiang Chen
- Wuhan University School of Basic Medical Sciences, 36841, Wuhan, China;
| | - Ao Xiao
- Wuhan University School of Basic Medical Sciences, 36841, Wuhan, China;
| | - Qifa Ye
- Wuhan University Zhongnan Hospital, 89674, Wuhan, Hubei , China;
| | - Xiaowen Shi
- Wuhan University, 12390, School of Resource and Environmental Science, Wuhan, Hubei , China;
| | - Zijian Wang
- Wuhan University School of Basic Medical Sciences, 36841, Wuhan, China;
| | - Xinwei Han
- Zhengzhou University First Affiliated Hospital, 191599, Zhengzhou, Henan, China;
| | - Yun Chen
- Wuhan University School of Basic Medical Sciences, 36841, 115 Donghu Road, Wuchang District, Wuhan, China, Wuhan, China, 430071;
| |
Collapse
|
23
|
Li X, Li K, Chen Y, Fang F. The Role of Hippo Signaling Pathway in the Development of the Nervous System. Dev Neurosci 2021; 43:263-270. [PMID: 34350875 DOI: 10.1159/000515633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 02/26/2021] [Indexed: 11/19/2022] Open
Abstract
Hippo signaling pathway is a highly conserved and crucial signaling pathway that controls the size of tissues and organs by regulating the proliferation, differentiation, and apoptosis of cells. The nervous system is a complicated system that participates in information collection, integration, and procession. The balance of various aspects of the nervous system is vital for the normal regulation of physiological conditions of the body, like the population and distribution of nerve cells, nerve connections, and so on. Defects in these aspects may lead to cognitive, behavioral, and neurological dysfunction, resulting in various nervous system diseases. Recently, accumulating evidence proposes that Hippo pathway maintains numerous biological functions in the nervous system development, including modulating the proliferation and differentiation of nerve cells and promoting the development of synapse, corpus callosum, and cortex. In this review, we will summarize recent findings of Hippo pathway in the nervous system to improve our understanding on its function and to provide potential therapeutic strategies of nervous system diseases in the future.
Collapse
Affiliation(s)
- Xifan Li
- Department of Human Anatomy, School of Basic Medicine Sciences, Guilin Medical University, Guilin, China
| | - Kaixuan Li
- Department of Human Anatomy, School of Basic Medicine Sciences, Guilin Medical University, Guilin, China
| | - Yu Chen
- Department of Human Anatomy, School of Basic Medicine Sciences, Guilin Medical University, Guilin, China
| | - Fang Fang
- Department of Human Anatomy, School of Basic Medicine Sciences, Guilin Medical University, Guilin, China
| |
Collapse
|
24
|
Sencar L, Coşkun G, Şaker D, Sapmaz T, Kara S, Çelenk A, Polat S, Yılmaz DM, Dağlıoğlu YK, Polat S. Effects of Theranekron and alpha-lipoic acid combined treatment on GAP-43 and Krox-20 gene expressions and inflammation markers in peripheral nerve injury. Ultrastruct Pathol 2021; 45:167-181. [PMID: 34184615 DOI: 10.1080/01913123.2021.1923600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Peripheral nerve injury (PNI) is a major health problem that results in loss of motor and sensory functions. In treatment of PNI, various methods such as anastomosis, nerve grafts, nonneural tissue grafts, and nerve conduits are applied. In the present study, it was aimed to investigate the effects of Theranekron and Alpha-lipoic acid (ALA) combined treatment on nerve healing in experimental PNI by using histomorphometric, electron microscopic, immunohistochemical and molecular biological methods. Sixty-two Wistar rats were divided into six groups; the normal control group, sham operation group, experimental control group having a crush type injury with no treatment, Theranekron treatment group, ALA treatment group and Theranekron+ALA combined treatment group. Sciatic nerve tissue samples were obtained on days 1, 7 and 14 following injury in all groups. GAP-43 expression was upregulated in all PNI received groups compared to the control group. Krox-20 expression was downregulated in all groups that received PNI compared to the control group. While intensely positive TNF-α and IL-6 expressions were observed up to the 1st to the 14th day for the experimental control group, these expressions were seen as "weakly positive" in the treatment groups from the 1st day to the 14th day. The number of myelinated fibers was higher in the control and sham operation groups. Additionally, the number of myelinated nerve fibers increased in the combined treatment group. In conclusion, these findings suggest that combined therapy of Theranekron and ALA promotes structural recovery and it should be considered as an effective treatment protocol following PNI.
Collapse
Affiliation(s)
- Leman Sencar
- Department of Histology and Embryology, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Gülfidan Coşkun
- Department of Histology and Embryology, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Dilek Şaker
- Department of Histology and Embryology, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Tuğçe Sapmaz
- Department of Histology and Embryology, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Samet Kara
- Department of Histology and Embryology, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Alper Çelenk
- Department of Histology and Embryology, Faculty of Medicine, Çukurova University, Adana, Turkey
| | - Sema Polat
- Department of Anatomy, Faculty of Medicine, Çukurova University, Adana, Turkey
| | | | - Y Kenan Dağlıoğlu
- Medical Sciences and Experimental Research and Application Center of Çukurova University, Adana, Turkey
| | - Sait Polat
- Department of Histology and Embryology, Faculty of Medicine, Çukurova University, Adana, Turkey
| |
Collapse
|
25
|
Hui TK, Lai XS, Dong X, Jing H, Liu Z, Fei E, Chen WB, Wang S, Ren D, Zou S, Wu HT, Pan BX. Ablation of Lrp4 in Schwann Cells Promotes Peripheral Nerve Regeneration in Mice. BIOLOGY 2021; 10:biology10060452. [PMID: 34063992 PMCID: PMC8223976 DOI: 10.3390/biology10060452] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 04/16/2021] [Accepted: 05/18/2021] [Indexed: 11/16/2022]
Abstract
Low-density lipoprotein receptor-related protein 4 (Lrp4) is a critical protein involved in the Agrin-Lrp4-MuSK signaling pathway that drives the clustering of acetylcholine receptors (AChRs) at the neuromuscular junction (NMJ). Many studies have shown that Lrp4 also functions in kidney development, bone formation, nervous system development, etc. However, whether Lrp4 participates in nerve regeneration in mammals remains unknown. Herein, we show that Lrp4 is expressed in SCs and that conditional knockout (cKO) of Lrp4 in SCs promotes peripheral nerve regeneration. In Lrp4 cKO mice, the demyelination of SCs was accelerated, and the proliferation of SCs was increased in the injured nerve. Furthermore, we identified that two myelination-related genes, Krox-20 and Mpz, were downregulated more dramatically in the cKO group than in the control group. Our results elucidate a novel role of Lrp4 in peripheral nerve regeneration and thereby provide a potential therapeutic target for peripheral nerve recovery.
Collapse
Affiliation(s)
- Tian-Kun Hui
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
| | - Xin-Sheng Lai
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
| | - Xia Dong
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| | - Hongyang Jing
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
| | - Ziyang Liu
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
| | - Erkang Fei
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
| | - Wen-Bing Chen
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
| | - Shunqi Wang
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
| | - Dongyan Ren
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
| | - Suqi Zou
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
- Correspondence: (S.Z.); (H.-T.W.); (B.-X.P.)
| | - Hai-Tao Wu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing 100850, China
- Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226019, China
- Correspondence: (S.Z.); (H.-T.W.); (B.-X.P.)
| | - Bing-Xing Pan
- School of Life Sciences, Nanchang University, Nanchang 330031, China; (T.-K.H.); (X.-S.L.); (H.J.); (Z.L.); (E.F.); (W.-B.C.); (S.W.); (D.R.)
- Institute of Life Science, Nanchang University, Nanchang 330031, China;
- Correspondence: (S.Z.); (H.-T.W.); (B.-X.P.)
| |
Collapse
|
26
|
Tyler EJ, Gutierrez del Arroyo A, Hughes BK, Wallis R, Garbe JC, Stampfer MR, Koh J, Lowe R, Philpott MP, Bishop CL. Early growth response 2 (EGR2) is a novel regulator of the senescence programme. Aging Cell 2021; 20:e13318. [PMID: 33547862 PMCID: PMC7963333 DOI: 10.1111/acel.13318] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 12/16/2020] [Accepted: 12/31/2020] [Indexed: 12/14/2022] Open
Abstract
Senescence, a state of stable growth arrest, plays an important role in ageing and age-related diseases in vivo. Although the INK4/ARF locus is known to be essential for senescence programmes, the key regulators driving p16 and ARF transcription remain largely underexplored. Using siRNA screening for modulators of the p16/pRB and ARF/p53/p21 pathways in deeply senescent human mammary epithelial cells (DS HMECs) and fibroblasts (DS HMFs), we identified EGR2 as a novel regulator of senescence. EGR2 expression is up-regulated during senescence, and its ablation by siRNA in DS HMECs and HMFs transiently reverses the senescent phenotype. We demonstrate that EGR2 activates the ARF and p16 promoters and directly binds to both the ARF and p16 promoters. Loss of EGR2 down-regulates p16 levels and increases the pool of p16- p21- 'reversed' cells in the population. Moreover, EGR2 overexpression is sufficient to induce senescence. Our data suggest that EGR2 is a direct transcriptional activator of the p16/pRB and ARF/p53/p21 pathways in senescence and a novel marker of senescence.
Collapse
Affiliation(s)
- Eleanor J. Tyler
- Blizard InstituteBarts and The London School of Medicine and Dentistry, Queen Mary University of LondonLondonUK
| | - Ana Gutierrez del Arroyo
- Translational Medicine & TherapeuticsWilliam Harvey Research InstituteBarts and The London School of Medicine and DentistryQueen Mary University of LondonLondonUK
| | - Bethany K. Hughes
- Blizard InstituteBarts and The London School of Medicine and Dentistry, Queen Mary University of LondonLondonUK
| | - Ryan Wallis
- Blizard InstituteBarts and The London School of Medicine and Dentistry, Queen Mary University of LondonLondonUK
| | - James C. Garbe
- Biological Systems and Engineering DivisionLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Martha R. Stampfer
- Biological Systems and Engineering DivisionLawrence Berkeley National LaboratoryBerkeleyCaliforniaUSA
| | - Jim Koh
- Division of General SurgeryDepartment of SurgeryUCSFSan FranciscoCaliforniaUSA
| | - Robert Lowe
- Blizard InstituteBarts and The London School of Medicine and Dentistry, Queen Mary University of LondonLondonUK
| | - Michael P. Philpott
- Blizard InstituteBarts and The London School of Medicine and Dentistry, Queen Mary University of LondonLondonUK
| | - Cleo L. Bishop
- Blizard InstituteBarts and The London School of Medicine and Dentistry, Queen Mary University of LondonLondonUK
| |
Collapse
|
27
|
Choi SJ, Park SY, Shin YH, Heo SH, Kim KH, Lee HI, Kim JK. Mesenchymal Stem Cells Derived from Wharton's Jelly Can Differentiate into Schwann Cell-Like Cells and Promote Peripheral Nerve Regeneration in Acellular Nerve Grafts. Tissue Eng Regen Med 2021; 18:467-478. [PMID: 33515168 DOI: 10.1007/s13770-020-00329-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 12/08/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Schwann cells (SCs) secrete neurotrophic factors and provide structural support and guidance during axonal regeneration. However, nearby nerves may be damaged to obtain primary SCs, and there is a lack of nervous tissue donors. We investigated the potential of Wharton's Jelly-derived mesenchymal stem cells (WJ-MSCs) in differentiating into Schwann cell-like cells (WJ-SCLCs) as an alternative to SCs. We also examined whether implantation of WJ-SCLCs-laden acellular nerve grafts (ANGs) are effective in inducing functional recovery and nerve regeneration in an animal model of peripheral nerve injury. METHODS The differentiation of WJ-MSCs into WJ-SCLCs was determined by analyzing SC-specific markers. The secretion of neurotrophic factors was assessed by the Neuro Discovery antibody array. Neurite outgrowth and myelination of axons were found in a co-culture system involving motor neuron cell lines. The effects of ANGs on repairing sciatic nerves were evaluated using video gait angle test, isometric tetanic force analysis, and toluidine blue staining. RESULTS Compared with undifferentiated WJ-MSCs, WJ-SCLCs showed higher expression levels of SC-specific markers such as S100β, GFAP, KROX20, and NGFR. WJ-SCLCs also showed higher secreted amounts of brain-derived neurotrophic factor, glial cell-derived neurotrophic factor, and granulocyte-colony stimulating factor than did WJ-MSCs. WJ-SCLCs effectively promoted the outgrowth and myelination of neurites in motor neuron cells, and WJ-SCLCs laden ANGs significantly facilitated peripheral nerve regeneration in an animal model of sciatic nerve injury. CONCLUSION WJ-MSCs were readily differentiated into WJ-SCLCs, which effectively promoted the regeneration of peripheral nerves. Transplantation of WJ-SCLCs with ANGs might be useful for assisting peripheral nerve regeneration.
Collapse
Affiliation(s)
- Soon Jin Choi
- Asan Peripheral Nerve Regeneration Lab Institute for Life Sciences, Seoul, South Korea
| | - Suk Young Park
- Asan Peripheral Nerve Regeneration Lab Institute for Life Sciences, Seoul, South Korea
| | - Young Ho Shin
- Department of Orthopedic Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic Road 43-gil, Songpa-gu, Seoul, 05505, South Korea
| | - Seung-Ho Heo
- Convergence Medicine Research Center, Asan Medical Center, Seoul, South Korea
| | - Kang-Hyun Kim
- Convergence Medicine Research Center, Asan Medical Center, Seoul, South Korea
| | - Hyo In Lee
- Convergence Medicine Research Center, Asan Medical Center, Seoul, South Korea
| | - Jae Kwang Kim
- Asan Peripheral Nerve Regeneration Lab Institute for Life Sciences, Seoul, South Korea. .,Department of Orthopedic Surgery, Asan Medical Center, University of Ulsan College of Medicine, 88, Olympic Road 43-gil, Songpa-gu, Seoul, 05505, South Korea.
| |
Collapse
|
28
|
Soluble dimeric prion protein ligand activates Adgrg6 receptor but does not rescue early signs of demyelination in PrP-deficient mice. PLoS One 2020; 15:e0242137. [PMID: 33180885 PMCID: PMC7660510 DOI: 10.1371/journal.pone.0242137] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023] Open
Abstract
The adhesion G-protein coupled receptor Adgrg6 (formerly Gpr126) is instrumental in the development, maintenance and repair of peripheral nervous system myelin. The prion protein (PrP) is a potent activator of Adgrg6 and could be used as a potential therapeutic agent in treating peripheral demyelinating and dysmyelinating diseases. We designed a dimeric Fc-fusion protein comprising the myelinotrophic domain of PrP (FT2Fc), which activated Adgrg6 in vitro and exhibited favorable pharmacokinetic properties for in vivo treatment of peripheral neuropathies. While chronic FT2Fc treatment elicited specific transcriptomic changes in the sciatic nerves of PrP knockout mice, no amelioration of the early molecular signs demyelination was detected. Instead, RNA sequencing of sciatic nerves revealed downregulation of cytoskeletal and sarcomere genes, akin to the gene expression changes seen in myopathic skeletal muscle of PrP overexpressing mice. These results call for caution when devising myelinotrophic therapies based on PrP-derived Adgrg6 ligands. While our treatment approach was not successful, Adgrg6 remains an attractive therapeutic target to be addressed in other disease models or by using different biologically active Adgrg6 ligands.
Collapse
|
29
|
Muppirala AN, Limbach LE, Bradford EF, Petersen SC. Schwann cell development: From neural crest to myelin sheath. WILEY INTERDISCIPLINARY REVIEWS-DEVELOPMENTAL BIOLOGY 2020; 10:e398. [PMID: 33145925 DOI: 10.1002/wdev.398] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/16/2022]
Abstract
Vertebrate nervous system function requires glial cells, including myelinating glia that insulate axons and provide trophic support that allows for efficient signal propagation by neurons. In vertebrate peripheral nervous systems, neural crest-derived glial cells known as Schwann cells (SCs) generate myelin by encompassing and iteratively wrapping membrane around single axon segments. SC gliogenesis and neurogenesis are intimately linked and governed by a complex molecular environment that shapes their developmental trajectory. Changes in this external milieu drive developing SCs through a series of distinct morphological and transcriptional stages from the neural crest to a variety of glial derivatives, including the myelinating sublineage. Cues originate from the extracellular matrix, adjacent axons, and the developing SC basal lamina to trigger intracellular signaling cascades and gene expression changes that specify stages and transitions in SC development. Here, we integrate the findings from in vitro neuron-glia co-culture experiments with in vivo studies investigating SC development, particularly in zebrafish and mouse, to highlight critical factors that specify SC fate. Ultimately, we connect classic biochemical and mutant studies with modern genetic and visualization tools that have elucidated the dynamics of SC development. This article is categorized under: Signaling Pathways > Cell Fate Signaling Nervous System Development > Vertebrates: Regional Development.
Collapse
Affiliation(s)
- Anoohya N Muppirala
- Program in Neuroscience, Harvard Medical School, Boston, Massachusetts, USA.,Department of Neuroscience, Kenyon College, Gambier, Ohio, USA
| | | | | | - Sarah C Petersen
- Department of Neuroscience, Kenyon College, Gambier, Ohio, USA.,Department of Biology, Kenyon College, Gambier, Ohio, USA
| |
Collapse
|
30
|
Wüst HM, Wegener A, Fröb F, Hartwig AC, Wegwitz F, Kari V, Schimmel M, Tamm ER, Johnsen SA, Wegner M, Sock E. Egr2-guided histone H2B monoubiquitination is required for peripheral nervous system myelination. Nucleic Acids Res 2020; 48:8959-8976. [PMID: 32672815 PMCID: PMC7498331 DOI: 10.1093/nar/gkaa606] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/03/2020] [Accepted: 07/07/2020] [Indexed: 12/30/2022] Open
Abstract
Schwann cells are the nerve ensheathing cells of the peripheral nervous system. Absence, loss and malfunction of Schwann cells or their myelin sheaths lead to peripheral neuropathies such as Charcot-Marie-Tooth disease in humans. During Schwann cell development and myelination chromatin is dramatically modified. However, impact and functional relevance of these modifications are poorly understood. Here, we analyzed histone H2B monoubiquitination as one such chromatin modification by conditionally deleting the Rnf40 subunit of the responsible E3 ligase in mice. Rnf40-deficient Schwann cells were arrested immediately before myelination or generated abnormally thin, unstable myelin, resulting in a peripheral neuropathy characterized by hypomyelination and progressive axonal degeneration. By combining sequencing techniques with functional studies we show that H2B monoubiquitination does not influence global gene expression patterns, but instead ensures selective high expression of myelin and lipid biosynthesis genes and proper repression of immaturity genes. This requires the specific recruitment of the Rnf40-containing E3 ligase by Egr2, the central transcriptional regulator of peripheral myelination, to its target genes. Our study identifies histone ubiquitination as essential for Schwann cell myelination and unravels new disease-relevant links between chromatin modifications and transcription factors in the underlying regulatory network.
Collapse
Affiliation(s)
- Hannah M Wüst
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, D-91054 Erlangen, Germany
| | - Amélie Wegener
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, D-91054 Erlangen, Germany
| | - Franziska Fröb
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, D-91054 Erlangen, Germany
| | - Anna C Hartwig
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, D-91054 Erlangen, Germany
| | - Florian Wegwitz
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, D-37075 Göttingen, Germany
| | - Vijayalakshmi Kari
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, D-37075 Göttingen, Germany
| | - Margit Schimmel
- Institut für Humananatomie und Embryologie, Universität Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | - Ernst R Tamm
- Institut für Humananatomie und Embryologie, Universität Regensburg, Universitätsstrasse 31, D-93053 Regensburg, Germany
| | - Steven A Johnsen
- Department of General, Visceral, and Pediatric Surgery, University Medical Center Göttingen, D-37075 Göttingen, Germany.,Gene Regulatory Mechanisms and Molecular Epigenetics Lab, Division of Gastroenterology and Hepatology, Mayo Clinic, 200 First St SW, Rochester, MN, USA
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, D-91054 Erlangen, Germany
| | - Elisabeth Sock
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, D-91054 Erlangen, Germany
| |
Collapse
|
31
|
Oct-6 transcriptional factor a possible biomarker for leprosy diagnosis. Diagn Microbiol Infect Dis 2020; 99:115232. [PMID: 33130505 DOI: 10.1016/j.diagmicrobio.2020.115232] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 09/11/2020] [Accepted: 09/26/2020] [Indexed: 11/22/2022]
Abstract
Leprosy is an infectious disease caused by Mycobacterium leprae that affects the skin and nerves. The nerve damage in leprosy may be related to alterations in transcriptional factors, such as Krox-20, Oct-6, Sox-10. Thirty skin biopsies in leprosy patients and 15 non-leprosy skin biopsies were evaluated using RT-qPCR to assess Krox-20, Oct-6, and Sox-10 and these data was related with S-100 immunohistochemistry. Changes in gene expression were observed in the skin and dermal nerves of leprosy patients in Oct-6 and Sox-10. When comparing Oct-6 with S-100 IHC as diagnostic tests for leprosy, Oct-6 showed a sensitivity of 73.3%, and specificity of 100%, while S-100 IHC showed a sensitivity of 96.6% and specificity of 100%. Our data suggest Oct-6 could be an auxiliary biomarker specific to detecting changes in dermal nerves in leprosy and thus useful to health workers and pathologists with no expertise to observe nerve injuries in leprosy.
Collapse
|
32
|
EEF1A1 deacetylation enables transcriptional activation of remyelination. Nat Commun 2020; 11:3420. [PMID: 32647127 PMCID: PMC7347577 DOI: 10.1038/s41467-020-17243-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 06/19/2020] [Indexed: 12/14/2022] Open
Abstract
Remyelination of the peripheral and central nervous systems (PNS and CNS, respectively) is a prerequisite for functional recovery after lesion. However, this process is not always optimal and becomes inefficient in the course of multiple sclerosis. Here we show that, when acetylated, eukaryotic elongation factor 1A1 (eEF1A1) negatively regulates PNS and CNS remyelination. Acetylated eEF1A1 (Ac-eEF1A1) translocates into the nucleus of myelinating cells where it binds to Sox10, a key transcription factor for PNS and CNS myelination and remyelination, to drag Sox10 out of the nucleus. We show that the lysine acetyltransferase Tip60 acetylates eEF1A1, whereas the histone deacetylase HDAC2 deacetylates eEF1A1. Promoting eEF1A1 deacetylation maintains the activation of Sox10 target genes and increases PNS and CNS remyelination efficiency. Taken together, these data identify a major mechanism of Sox10 regulation, which appears promising for future translational studies on PNS and CNS remyelination. The molecular mechanisms regulating remyelination are unclear. Here, the authors show that promoting deacetylation of eEF1A1 prevents the translocation of Sox10 outside the nucleus, contributing to maintaining the expression of Sox10 target genes and increasing remyelination efficiency.
Collapse
|
33
|
Patchett AL, Flies AS, Lyons AB, Woods GM. Curse of the devil: molecular insights into the emergence of transmissible cancers in the Tasmanian devil (Sarcophilus harrisii). Cell Mol Life Sci 2020; 77:2507-2525. [PMID: 31900624 PMCID: PMC11104928 DOI: 10.1007/s00018-019-03435-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 12/17/2019] [Accepted: 12/19/2019] [Indexed: 12/22/2022]
Abstract
The Tasmanian devil (Sarcophilus harrisii) is the only mammalian species known to be affected by multiple transmissible cancers. Devil facial tumours 1 and 2 (DFT1 and DFT2) are independent neoplastic cell lineages that produce large, disfiguring cancers known as devil facial tumour disease (DFTD). The long-term persistence of wild Tasmanian devils is threatened due to the ability of DFTD cells to propagate as contagious allografts and the high mortality rate of DFTD. Recent studies have demonstrated that both DFT1 and DFT2 cancers originated from founder cells of the Schwann cell lineage, an uncommon origin of malignant cancer in humans. This unprecedented finding has revealed a potential predisposition of Tasmanian devils to transmissible cancers of the Schwann cell lineage. In this review, we compare the molecular nature of human Schwann cells and nerve sheath tumours with DFT1 and DFT2 to gain insights into the emergence of transmissible cancers in the Tasmanian devil. We discuss a potential mechanism, whereby Schwann cell plasticity and frequent wounding in Tasmanian devils combine with an inherent cancer predisposition and low genetic diversity to give rise to transmissible Schwann cell cancers in devils on rare occasions.
Collapse
Affiliation(s)
- Amanda L Patchett
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS, 7000, Australia
| | - Andrew S Flies
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS, 7000, Australia
| | - A Bruce Lyons
- School of Medicine, University of Tasmania, Hobart, TAS, 7000, Australia
| | - Gregory M Woods
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool Street, Hobart, TAS, 7000, Australia.
| |
Collapse
|
34
|
Grove M, Lee H, Zhao H, Son YJ. Axon-dependent expression of YAP/TAZ mediates Schwann cell remyelination but not proliferation after nerve injury. eLife 2020; 9:50138. [PMID: 32436841 PMCID: PMC7259960 DOI: 10.7554/elife.50138] [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: 07/16/2019] [Accepted: 05/19/2020] [Indexed: 12/18/2022] Open
Abstract
Previously we showed that YAP/TAZ promote not only proliferation but also differentiation of immature Schwann cells (SCs), thereby forming and maintaining the myelin sheath around peripheral axons (Grove et al., 2017). Here we show that YAP/TAZ are required for mature SCs to restore peripheral myelination, but not to proliferate, after nerve injury. We find that YAP/TAZ dramatically disappear from SCs of adult mice concurrent with axon degeneration after nerve injury. They reappear in SCs only if axons regenerate. YAP/TAZ ablation does not impair SC proliferation or transdifferentiation into growth promoting repair SCs. SCs lacking YAP/TAZ, however, fail to upregulate myelin-associated genes and completely fail to remyelinate regenerated axons. We also show that both YAP and TAZ are redundantly required for optimal remyelination. These findings suggest that axons regulate transcriptional activity of YAP/TAZ in adult SCs and that YAP/TAZ are essential for functional regeneration of peripheral nerve.
Collapse
Affiliation(s)
- Matthew Grove
- Shriners Hospitals Pediatric Research Center and Center for Neural Repair and Rehabilitation, Temple University, Philadelphia, United States.,Department of Anatomy and Cell Biology, Temple University, Philadelphia, United States
| | - Hyunkyoung Lee
- Shriners Hospitals Pediatric Research Center and Center for Neural Repair and Rehabilitation, Temple University, Philadelphia, United States.,Department of Anatomy and Cell Biology, Temple University, Philadelphia, United States
| | - Huaqing Zhao
- Department of Clinical Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Young-Jin Son
- Shriners Hospitals Pediatric Research Center and Center for Neural Repair and Rehabilitation, Temple University, Philadelphia, United States.,Department of Anatomy and Cell Biology, Temple University, Philadelphia, United States
| |
Collapse
|
35
|
Duman M, Martinez-Moreno M, Jacob C, Tapinos N. Functions of histone modifications and histone modifiers in Schwann cells. Glia 2020; 68:1584-1595. [PMID: 32034929 DOI: 10.1002/glia.23795] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/24/2020] [Accepted: 01/28/2020] [Indexed: 01/25/2023]
Abstract
Schwann cells (SCs) are the main glial cells present in the peripheral nervous system (PNS). Their primary functions are to insulate peripheral axons to protect them from the environment and to enable fast conduction of electric signals along big caliber axons by enwrapping them in a thick myelin sheath rich in lipids. In addition, SCs have the peculiar ability to foster axonal regrowth after a lesion by demyelinating and converting into repair cells that secrete neurotrophic factors and guide axons back to their former target to finally remyelinate regenerated axons. The different steps of SC development and their role in the maintenance of PNS integrity and regeneration after lesion are controlled by various factors among which transcription factors and chromatin-remodeling enzymes hold major functions. In this review, we discussed how histone modifications and histone-modifying enzymes control SC development, maintenance of PNS integrity and response to injury. The functions of histone modifiers as part of chromatin-remodeling complexes are discussed in another review published in the same issue of Glia.
Collapse
Affiliation(s)
- Mert Duman
- Faculty of Biology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Margot Martinez-Moreno
- Department of Neurosurgery, Molecular Neuroscience & Neuro-Oncology Laboratory, Brown University, Providence, Rhode Island
| | - Claire Jacob
- Faculty of Biology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Nikos Tapinos
- Department of Neurosurgery, Molecular Neuroscience & Neuro-Oncology Laboratory, Brown University, Providence, Rhode Island
| |
Collapse
|
36
|
Fröb F, Wegner M. The role of chromatin remodeling complexes in Schwann cell development. Glia 2019; 68:1596-1603. [DOI: 10.1002/glia.23766] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/27/2019] [Accepted: 12/03/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Franziska Fröb
- Institut für Biochemie, Emil‐Fischer‐Zentrum Friedrich‐Alexander‐Universität Erlangen‐Nürnberg Erlangen Germany
| | - Michael Wegner
- Institut für Biochemie, Emil‐Fischer‐Zentrum Friedrich‐Alexander‐Universität Erlangen‐Nürnberg Erlangen Germany
| |
Collapse
|
37
|
Sabag E, Halperin E, Liron T, Hiram-Bab S, Frenkel B, Gabet Y. Hormone-Independent Sexual Dimorphism in the Regulation of Bone Resorption by Krox20. J Bone Miner Res 2019; 34:2277-2286. [PMID: 31398266 DOI: 10.1002/jbmr.3847] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Revised: 06/28/2019] [Accepted: 07/28/2019] [Indexed: 12/22/2022]
Abstract
Krox20/EGR2 is a zinc finger transcription factor, implicated in the development of the hindbrain, nerve myelination, and tumor suppression. In skeletal biology, we have demonstrated that Krox20 also regulates adult bone metabolism. We and others have characterized several functions of Krox20 in the osteoclast lineage, namely, preosteoclast proliferation and differentiation, and mature osteoclast apoptosis. We have previously reported that systemically Krox20-haploinsufficient mice have a low bone mass with increased bone resorption. However, new data have now revealed that this phenotype is restricted to females. In addition, we discovered that conditional knockout of Krox20 (cKO) restricted to osteoclast progenitors is sufficient to induce the same female-specific bone loss observed in systemic mutants. To test whether this sexual dimorphism results from an interaction between Krox20 and sex hormones, we examined the sex- and hormone-dependent role of Krox20 deficiency on proliferation and apoptosis in osteoclastic cells. Our results indicate that male and female sex hormones (dihydrotestosterone [DHT] and estradiol [E2], respectively) as well as Krox20 inhibit preosteoclast proliferation and augment osteoclast apoptosis. The observation that Krox20 expression is inhibited by DHT and E2 negates the hypothesis that the effect of sex hormones is mediated by an increase in Krox20 expression. Interestingly, the effect of Krox20 deficiency was observed only with cells derived from female animals, regardless of any sex hormones added in vitro. In addition, we have identified sexual dimorphism in the expression of several Krox20-related genes, including NAB2. This sex-specific epigenetic profile was established at puberty, maintained in the absence of sex hormones, and explains the female-specific skeletal importance of Krox20. The findings described in this study emphasize the medical importance of sex differences, which may be determined at the epigenetic level. © 2019 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Elias Sabag
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Elinor Halperin
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tamar Liron
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sahar Hiram-Bab
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Baruch Frenkel
- Department of Biochemistry and Molecular Medicine, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA.,Department of Orthopedic Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Yankel Gabet
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
38
|
c-Jun in Schwann Cells: Stay Away from Extremes. J Neurosci 2019; 38:3388-3390. [PMID: 29618544 DOI: 10.1523/jneurosci.0028-18.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 02/15/2018] [Accepted: 02/22/2018] [Indexed: 11/21/2022] Open
|
39
|
Chen WA, Luo TD, Barnwell JC, Smith TL, Li Z. Age-Dependent Schwann Cell Phenotype Regulation Following Peripheral Nerve Injury. J Hand Surg Asian Pac Vol 2019; 22:464-471. [PMID: 29117831 DOI: 10.1142/s0218810417500514] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND Schwann cells are integral to the regenerative capacity of the peripheral nervous system, which declines after adolescence. The mechanisms underlying this decline are poorly understood. This study sought to compare the protein expression of Notch, c-Jun, and Krox-20 after nerve crush injury in adolescent and young adult rats. We hypothesized that these Schwann cell myelinating regulatory factors are down-regulated after nerve injury in an age-dependent fashion. METHODS Adolescent (2 months old) and young adult (12 months old) rats (n = 48) underwent sciatic nerve crush injury. Protein expression of Notch, c-Jun, and Krox-20 was quantified by Western blot analysis at 1, 3, and 7 days post-injury. Functional recovery was assessed in a separate group of animals (n = 8) by gait analysis (sciatic functional index) and electromyography (compound motor action potential) over an 8-week post-injury period. RESULTS Young adult rats demonstrated a trend of delayed onset of the dedifferentiating regulatory factors, Notch and c-Jun, corresponding to the delayed functional recovery observed in young adult rats compared to adolescent rats. Compound motor action potential area was significantly greater in adolescent rats relative to young adult rats, while amplitude and velocity trended toward statistical significance. CONCLUSIONS The process of Schwann cell dedifferentiation following peripheral nerve injury shows different trends with age. These trends of delayed onset of key regulatory factors responsible for Schwann cell myelination may be one of many possible factors mediating the significant differences in functional recovery between adolescent and young adult rats following peripheral nerve injury.
Collapse
Affiliation(s)
- Wayne A Chen
- 1 Department of Orthopaedic Surgery, Wake Forest Baptist Medical Center, Medical Center Boulevard, NC, USA
| | - T David Luo
- 1 Department of Orthopaedic Surgery, Wake Forest Baptist Medical Center, Medical Center Boulevard, NC, USA
| | - Jonathan C Barnwell
- 1 Department of Orthopaedic Surgery, Wake Forest Baptist Medical Center, Medical Center Boulevard, NC, USA
| | - Thomas L Smith
- 1 Department of Orthopaedic Surgery, Wake Forest Baptist Medical Center, Medical Center Boulevard, NC, USA
| | - Zhongyu Li
- 1 Department of Orthopaedic Surgery, Wake Forest Baptist Medical Center, Medical Center Boulevard, NC, USA
| |
Collapse
|
40
|
Sock E, Wegner M. Transcriptional control of myelination and remyelination. Glia 2019; 67:2153-2165. [PMID: 31038810 DOI: 10.1002/glia.23636] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/01/2019] [Accepted: 04/11/2019] [Indexed: 12/11/2022]
Abstract
Myelination is an evolutionary recent differentiation program that has been independently acquired in vertebrates by Schwann cells in the peripheral nervous system and oligodendrocytes in the central nervous system. Therefore, it is not surprising that regulating transcription factors differ substantially between both cell types. However, overall principles are similar as transcriptional control in Schwann cells and oligodendrocytes combines lineage determining and stage-specific factors in complex regulatory networks. Myelination does not only occur during development, but also as remyelination in the adult. In line with the different conditions during developmental myelination and remyelination and the distinctive properties of Schwann cells and oligodendrocytes, transcriptional regulation of remyelination exhibits unique features and differs between the two cell types. This review gives an overview of the current state in the field.
Collapse
Affiliation(s)
- Elisabeth Sock
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
41
|
Svaren J, Moran JJ, Wu X, Zuccarino R, Bacon C, Bai Y, Ramesh R, Gutmann L, Anderson DM, Pavelec D, Shy ME. Schwann cell transcript biomarkers for hereditary neuropathy skin biopsies. Ann Neurol 2019; 85:887-898. [PMID: 30945774 DOI: 10.1002/ana.25480] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/02/2019] [Accepted: 04/03/2019] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Charcot-Marie-Tooth (CMT) disease is most commonly caused by duplication of a chromosomal segment surrounding Peripheral Myelin Protein 22, or PMP22 gene, which is classified as CMT1A. Several candidate therapies reduce Pmp22 mRNA levels in CMT1A rodent models, but development of biomarkers for clinical trials in CMT1A is a challenge given its slow progression and difficulty in obtaining nerve samples. Quantitative PCR measurements of PMP22 mRNA in dermal nerves were performed using skin biopsies in human clinical trials for CMT1A, but this approach did not show increased PMP22 mRNA in CMT1A patients compared to controls. One complicating factor is the variable amounts of Schwann cells (SCs) in skin. The objective of the study was to develop a novel method for precise evaluation of PMP22 levels in skin biopsies that can discriminate CMT1A patients from controls. METHODS We have developed methods to normalize PMP22 transcript levels to SC-specific genes that are not altered by CMT1A status. Several CMT1A-associated genes were assembled into a custom Nanostring panel to enable precise transcript measurements that can be normalized to variable SC content. RESULTS The digital expression data from Nanostring analysis showed reproducible elevation of PMP22 levels in CMT1A versus control skin biopsies, particularly after normalization to SC-specific genes. INTERPRETATION This platform should be useful in clinical trials for CMT1A as a biomarker of target engagement that can be used to optimize dosing, and the same normalization framework is applicable to other types of CMT. ANN NEUROL 2019;85:887-898.
Collapse
Affiliation(s)
- John Svaren
- Waisman Center, University of Wisconsin-Madison, Madison, WI.,Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI
| | - John J Moran
- Waisman Center, University of Wisconsin-Madison, Madison, WI
| | - Xingyao Wu
- Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Riccardo Zuccarino
- Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, IA.,Neuromuscular Omnicentre (NEMO)-Fondazione Serena Onlus, Arenzano, Italy
| | - Chelsea Bacon
- Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Yunhong Bai
- Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Raghu Ramesh
- Waisman Center, University of Wisconsin-Madison, Madison, WI
| | - Laurie Gutmann
- Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Daniel M Anderson
- Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, IA
| | - Derek Pavelec
- Biotechnology Center, University of Wisconsin-Madison, Madison, WI
| | - Michael E Shy
- Department of Neurology, University of Iowa Carver College of Medicine, Iowa City, IA
| |
Collapse
|
42
|
Casalenovo MB, Rosa PS, de Faria Bertoluci DF, Barbosa ASAA, do Nascimento DC, de Souza VNB, Nogueira MRS. Myelination key factor krox-20 is downregulated in Schwann cells and murine sciatic nerves infected by Mycobacterium leprae. Int J Exp Pathol 2019; 100:83-93. [PMID: 31090128 PMCID: PMC6540694 DOI: 10.1111/iep.12309] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 01/28/2019] [Accepted: 02/06/2019] [Indexed: 12/14/2022] Open
Abstract
Schwann cells (SCs) critically maintain the plasticity of the peripheral nervous system. Peripheral nerve injuries and infections stimulate SCs in order to retrieve homeostasis in neural tissues. Previous studies indicate that Mycobacterium leprae (ML) regulates the expression of key factors related to SC identity, suggesting that alterations in cell phenotype may be involved in the pathogenesis of neural damage in leprosy. To better understand whether ML restricts the plasticity of peripheral nerves, the present study sought to determine the expression of Krox-20, Sox-10, c-Jun and p75NTR in SC culture and mice sciatic nerves, both infected by ML Thai-53 strain. Primary SC cultures were stimulated with two different multiplicities of infection (MOI 100:1; MOI 50:1) and assessed after 7 and 14 days. Sciatic nerves of nude mice (NU-Foxn1nu ) infected with ML were evaluated after 6 and 9 months. In vitro results demonstrate downregulation of Krox-20 and Sox-10 along with the increase in p75NTR-immunolabelled cells. Concurrently, sciatic nerves of infected mice showed a significant decrease in Krox-20 and increase in p75NTR. Our results corroborate previous findings on the interference of ML in the expression of factors involved in cell maturation, favouring the maintenance of a non-myelinating phenotype in SCs, with possible implications for the repair of adult peripheral nerves.
Collapse
Affiliation(s)
- Mariane Bertolucci Casalenovo
- School of Medicine of BotucatuSão Paulo State UniversityBotucatuBrazil
- Lauro de Souza Lima InstituteSecretariat of Health of São PauloBauruSão PauloBrazil
| | | | | | | | | | - Vânia Nieto Brito de Souza
- School of Medicine of BotucatuSão Paulo State UniversityBotucatuBrazil
- Lauro de Souza Lima InstituteSecretariat of Health of São PauloBauruSão PauloBrazil
| | | |
Collapse
|
43
|
de Faria O, Dhaunchak AS, Kamen Y, Roth AD, Kuhlmann T, Colman DR, Kennedy TE. TMEM10 Promotes Oligodendrocyte Differentiation and is Expressed by Oligodendrocytes in Human Remyelinating Multiple Sclerosis Plaques. Sci Rep 2019; 9:3606. [PMID: 30837646 PMCID: PMC6400977 DOI: 10.1038/s41598-019-40342-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Accepted: 01/25/2019] [Indexed: 11/09/2022] Open
Abstract
Oligodendrocyte precursor cells (OPCs) differentiate during postnatal development into myelin-forming oligodendrocytes, in a process distinguished by substantial changes in morphology and the onset of myelin gene expression. A mammalian-specific CNS myelin gene, tmem10, also called Opalin, encodes a type 1 transmembrane protein that is highly upregulated during early stages of OPC differentiation; however, a function for TMEM10 has not yet been identified. Here, consistent with previous studies, we detect TMEM10 protein in mouse brain beginning at ~P10 and show that protein levels continue to increase as oligodendrocytes differentiate and myelinate axons in vivo. We show that constitutive TMEM10 overexpression in the Oli-neu oligodendroglial cell line promotes the expression of the myelin-associated genes MAG, CNP and CGT, whereas TMEM10 knock down in primary OPCs reduces CNP mRNA expression and decreases the percentage of MBP-positive oligodendrocytes that differentiate in vitro. Ectopic TMEM10 expression evokes an increase in process extension and branching, and blocking endogenous TMEM10 expression results in oligodendrocytes with abnormal cell morphology. These findings may have implications for human demyelinating disorders, as oligodendrocytes expressing TMEM10 are detected in human remyelinating multiple sclerosis lesions. Together, our findings provide evidence that TMEM10 promotes oligodendrocyte terminal differentiation and may represent a novel target to promote remyelination in demyelinating disorders.
Collapse
Affiliation(s)
- Omar de Faria
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University St., Montreal, Quebec, H3A 2B4, Canada
| | - Ajit S Dhaunchak
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University St., Montreal, Quebec, H3A 2B4, Canada
| | - Yasmine Kamen
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University St., Montreal, Quebec, H3A 2B4, Canada
| | - Alejandro D Roth
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University St., Montreal, Quebec, H3A 2B4, Canada.,Departamento de Biología, Facultad de Ciencias, Universidad de Chile, Santiago, Chile
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, D-48149, Münster, Germany
| | - David R Colman
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University St., Montreal, Quebec, H3A 2B4, Canada
| | - Timothy E Kennedy
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, 3801 University St., Montreal, Quebec, H3A 2B4, Canada.
| |
Collapse
|
44
|
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: 3.6] [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
|
45
|
Rosenberg LH, Cattin AL, Fontana X, Harford-Wright E, Burden JJ, White IJ, Smith JG, Napoli I, Quereda V, Policarpi C, Freeman J, Ketteler R, Riccio A, Lloyd AC. HDAC3 Regulates the Transition to the Homeostatic Myelinating Schwann Cell State. Cell Rep 2018; 25:2755-2765.e5. [PMID: 30517863 PMCID: PMC6293966 DOI: 10.1016/j.celrep.2018.11.045] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 10/16/2018] [Accepted: 11/09/2018] [Indexed: 12/29/2022] Open
Abstract
The formation of myelinating Schwann cells (mSCs) involves the remarkable biogenic process, which rapidly generates the myelin sheath. Once formed, the mSC transitions to a stable homeostatic state, with loss of this stability associated with neuropathies. The histone deacetylases histone deacetylase 1 (HDAC1) and HDAC2 are required for the myelination transcriptional program. Here, we show a distinct role for HDAC3, in that, while dispensable for the formation of mSCs, it is essential for the stability of the myelin sheath once formed-with loss resulting in progressive severe neuropathy in adulthood. This is associated with the prior failure to downregulate the biogenic program upon entering the homeostatic state leading to hypertrophy and hypermyelination of the mSCs, progressing to the development of severe myelination defects. Our results highlight distinct roles of HDAC1/2 and HDAC3 in controlling the differentiation and homeostatic states of a cell with broad implications for the understanding of this important cell-state transition.
Collapse
Affiliation(s)
- Laura H Rosenberg
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK; CRUK Therapeutic Discovery Laboratories, Babraham Research Campus, Cambridge CB22 3AT, UK
| | - Anne-Laure Cattin
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Xavier Fontana
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Elizabeth Harford-Wright
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Jemima J Burden
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Ian J White
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Jacob G Smith
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Ilaria Napoli
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Victor Quereda
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK; The Scripps Research Institute, 130 Scripps Way, Jupiter, FL 33458, USA
| | - Cristina Policarpi
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Jamie Freeman
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK; Horizon Discovery, 8100 Cambridge Research Park, Cambridge CB25 9TL, UK
| | - Robin Ketteler
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Antonella Riccio
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Alison C Lloyd
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK; UCL Cancer Institute, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
46
|
Fischbach F, Nedelcu J, Leopold P, Zhan J, Clarner T, Nellessen L, Beißel C, van Heuvel Y, Goswami A, Weis J, Denecke B, Schmitz C, Hochstrasser T, Nyamoya S, Victor M, Beyer C, Kipp M. Cuprizone-induced graded oligodendrocyte vulnerability is regulated by the transcription factor DNA damage-inducible transcript 3. Glia 2018; 67:263-276. [PMID: 30511355 DOI: 10.1002/glia.23538] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 09/05/2018] [Accepted: 09/07/2018] [Indexed: 12/28/2022]
Abstract
Oligodendrocytes are integral to efficient neuronal signaling. Loss of myelinating oligodendrocytes is a central feature of many neurological diseases, including multiple sclerosis (MS). The results of neuropathological studies suggest that oligodendrocytes react with differing sensitivity to toxic insults, with some cells dying early during lesion development and some cells being resistant for weeks. This proposed graded vulnerability has never been demonstrated but provides an attractive window for therapeutic interventions. Furthermore, the biochemical pathways associated with graded oligodendrocyte vulnerability have not been well explored. We used immunohistochemistry and serial block-face scanning electron microscopy (3D-SEM) to show that cuprizone-induced metabolic stress results in an "out of phase" degeneration of oligodendrocytes. Although expression induction of stress response transcription factors in oligodendrocytes occurs within days, subsequent oligodendrocyte apoptosis continues for weeks. In line with the idea of an out of phase degeneration of oligodendrocytes, detailed ultrastructural reconstructions of the axon-myelin unit demonstrate demyelination of single internodes. In parallel, genome wide array analyses revealed an active unfolded protein response early after initiation of the cuprizone intoxication. In addition to the cytoprotective pathways, the pro-apoptotic transcription factor DNA damage-inducible transcript 3 (DDIT3) was induced early in oligodendrocytes. In advanced lesions, DDIT3 was as well expressed by activated astrocytes. Toxin-induced oligodendrocyte apoptosis, demyelination, microgliosis, astrocytosis, and acute axonal damage were less intense in the Ddit3-null mutants. This study identifies DDIT3 as an important regulator of graded oligodendrocyte vulnerability in a MS animal model. Interference with this stress cascade might offer a promising therapeutic approach for demyelinating disorders.
Collapse
Affiliation(s)
- Felix Fischbach
- Faculty of Medicine, LMU Munich, Chair of Neuroanatomy, Institute of Anatomy, Munich, Germany
| | - Julia Nedelcu
- Faculty of Medicine, LMU Munich, Chair of Neuroanatomy, Institute of Anatomy, Munich, Germany
| | - Patrizia Leopold
- Faculty of Medicine, LMU Munich, Chair of Neuroanatomy, Institute of Anatomy, Munich, Germany
| | - Jiangshan Zhan
- Faculty of Medicine, LMU Munich, Chair of Neuroanatomy, Institute of Anatomy, Munich, Germany
| | - Tim Clarner
- Faculty of Medicine, RWTH Aachen University, Institute of Neuroanatomy, Aachen, Germany
| | - Lara Nellessen
- Faculty of Medicine, RWTH Aachen University, Institute of Neuroanatomy, Aachen, Germany
| | - Christian Beißel
- Faculty of Medicine, RWTH Aachen University, Institute of Neuroanatomy, Aachen, Germany
| | - Yasemin van Heuvel
- Faculty of Medicine, RWTH Aachen University, Institute of Neuroanatomy, Aachen, Germany
| | - Anand Goswami
- Institute of Neuropathology, RWTH Aachen University, Aachen, Germany
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University, Aachen, Germany
| | - Bernd Denecke
- Interdisciplinary Center for Clinical Research Aachen (IZKF Aachen), RWTH Aachen University, Aachen, Germany
| | - Christoph Schmitz
- Faculty of Medicine, LMU Munich, Chair of Neuroanatomy, Institute of Anatomy, Munich, Germany
| | - Tanja Hochstrasser
- Faculty of Medicine, LMU Munich, Chair of Neuroanatomy, Institute of Anatomy, Munich, Germany
| | - Stella Nyamoya
- Faculty of Medicine, LMU Munich, Chair of Neuroanatomy, Institute of Anatomy, Munich, Germany.,Faculty of Medicine, RWTH Aachen University, Institute of Neuroanatomy, Aachen, Germany
| | - Marion Victor
- Faculty of Medicine, RWTH Aachen University, Institute of Neuroanatomy, Aachen, Germany
| | - Cordian Beyer
- Faculty of Medicine, RWTH Aachen University, Institute of Neuroanatomy, Aachen, Germany
| | - Markus Kipp
- Faculty of Medicine, LMU Munich, Chair of Neuroanatomy, Institute of Anatomy, Munich, Germany.,Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| |
Collapse
|
47
|
Gökbuget D, Pereira JA, Opitz L, Christe D, Kessler T, Marchais A, Suter U. The miRNA biogenesis pathway prevents inappropriate expression of injury response genes in developing and adult Schwann cells. Glia 2018; 66:2632-2644. [PMID: 30295958 PMCID: PMC6585637 DOI: 10.1002/glia.23516] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 07/18/2018] [Accepted: 07/18/2018] [Indexed: 12/24/2022]
Abstract
Proper function of the nervous system depends on myelination. In peripheral nerves, Schwann cells (SCs) myelinate axons and the miRNA biogenesis pathway is required for developmental myelination and myelin maintenance. However, regulatory roles of this pathway at different stages of myelination are only partially understood. We addressed the requirement of the core miRNA biogenesis pathway components Dgcr8, Drosha, and Dicer in developing and adult SCs using mouse mutants with a comparative genetics and transcriptomics approach. We found that the microprocessor components Dgcr8 and Drosha are crucial for axonal radial sorting and to establish correct SC numbers upon myelination. Transcriptome analyses revealed a requirement of the microprocessor to prevent aberrantly increased expression of injury-response genes. Those genes are predicted targets of abundant miRNAs in sciatic nerves (SNs) during developmental myelination. In agreement, Dgcr8 and Dicer are required for proper maintenance of the myelinated SC state, where abundant miRNAs in adult SNs are predicted to target injury-response genes. We conclude that the miRNA biogenesis pathway in SCs is crucial for preventing inappropriate activity of injury-response genes in developing and adult SCs.
Collapse
Affiliation(s)
- Deniz Gökbuget
- ETH Zurich, Department of BiologyInstitute of Molecular Health SciencesZurichSwitzerland
| | - Jorge A. Pereira
- ETH Zurich, Department of BiologyInstitute of Molecular Health SciencesZurichSwitzerland
| | - Lennart Opitz
- ETH Zurich/University of ZurichFunctional Genomics Center ZurichZurichSwitzerland
| | - Dominik Christe
- ETH Zurich, Department of BiologyInstitute of Molecular Health SciencesZurichSwitzerland
| | - Tobias Kessler
- ETH Zurich, Department of BiologyInstitute of Molecular Health SciencesZurichSwitzerland
| | - Antonin Marchais
- ETH Zurich, Department of BiologyInstitute of Agricultural SciencesZurichSwitzerland
| | - Ueli Suter
- ETH Zurich, Department of BiologyInstitute of Molecular Health SciencesZurichSwitzerland
| |
Collapse
|
48
|
Tammia M, Mi R, Sluch VM, Zhu A, Chung T, Shinn D, Zack DJ, Höke A, Mao HQ. Egr2 overexpression in Schwann cells increases myelination frequency in vitro. Heliyon 2018; 4:e00982. [PMID: 30761371 PMCID: PMC6275687 DOI: 10.1016/j.heliyon.2018.e00982] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 10/31/2018] [Accepted: 11/26/2018] [Indexed: 12/20/2022] Open
Abstract
Schwann cells are key players in peripheral nerve regeneration, and are uniquely capable of remyelinating axons in this context. Schwann cells orchestrate this process via a set of transcription factors. While it has been shown that overexpression of specific genes, e.g. Egr2, upregulates myelin-related transcripts, it remains unknown if such manipulation can functionalize the cells and enhance their myelination frequency. The ability to do so could have implications in the use of human stem cell-derived Schwann cells, where myelination is hard to achieve. After screening four candidate transcription factors (Sox10, Oct6, Brn2 and Egr2), we found that overexpression of Egr2 in rat Schwann cells co-cultured with sensory neurons enhanced myelination frequency and reduced cell proliferation. However, in a mouse model of sciatic nerve repair with cells engrafted within a nerve guide, myelination frequency in the engrafted cells was reduced upon Egr2 overexpression. Our results show that while overexpression of Egr2 can enhance the myelination frequency in vitro, it is context-dependent, potentially influenced by the microenvironment, timing of association with axons, expression level, species differences, or other factors.
Collapse
Affiliation(s)
- Markus Tammia
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA.,Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ruifa Mi
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Valentin M Sluch
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Allen Zhu
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tiffany Chung
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Daniel Shinn
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Donald J Zack
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Ophthalmology, Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Ahmet Höke
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.,Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hai-Quan Mao
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD 21218, USA.,Department of Materials Science and Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.,Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.,Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
49
|
LaVallee J, Grant T, D'Angelo-Early S, Kletsov S, Berry NA, Abt KM, Bloch CP, Muscedere ML, Adams KW. Refining the nuclear localization signal within the Egr transcriptional coregulator NAB2. FEBS Lett 2018; 593:107-118. [PMID: 30411343 DOI: 10.1002/1873-3468.13288] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/29/2018] [Accepted: 11/02/2018] [Indexed: 01/09/2023]
Abstract
NAB1 and 2 are coregulators for early growth response (Egr) transcription factors. The NAB1 nuclear localization signal (NLS) was previously described as a bipartite NLS of sequence R(X2 )K(X11 )KRXK. The sequence is conserved in NAB2 as K(X2 )R(X11 )KKXK; however, whether it functions as the NAB2 NLS has not been tested. We show that the KKXK motif in NAB2 is necessary and sufficient to mediate nuclear localization. Mutation of the KKXK motif to AAXA causes cytoplasmic localization of NAB2, while Lys/Arg-to-Ala mutations of the upstream K(X2 )R motif have no effect. Fusion of the KKXK motif to cytoplasmic protein eIF2Bε causes nuclear localization. Altogether, this study refines our knowledge of the NAB2 NLS, demonstrating that KKXK343-346 is necessary and sufficient for nuclear localization.
Collapse
Affiliation(s)
- Jacquelyn LaVallee
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Terrain Grant
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | | | - Sergey Kletsov
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Nicole A Berry
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Kimberly M Abt
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | - Christopher P Bloch
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| | | | - Kenneth W Adams
- Department of Biological Sciences, Bridgewater State University, Bridgewater, MA, USA
| |
Collapse
|
50
|
Ma KH, Duong P, Moran JJ, Junaidi N, Svaren J. Polycomb repression regulates Schwann cell proliferation and axon regeneration after nerve injury. Glia 2018; 66:2487-2502. [PMID: 30306639 PMCID: PMC6289291 DOI: 10.1002/glia.23500] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Revised: 07/02/2018] [Accepted: 07/03/2018] [Indexed: 01/01/2023]
Abstract
The transition of differentiated Schwann cells to support of nerve repair after injury is accompanied by remodeling of the Schwann cell epigenome. The EED-containing polycomb repressive complex 2 (PRC2) catalyzes histone H3K27 methylation and represses key nerve repair genes such as Shh, Gdnf, and Bdnf, and their activation is accompanied by loss of H3K27 methylation. Analysis of nerve injury in mice with a Schwann cell-specific loss of EED showed the reversal of polycomb repression is required and a rate limiting step in the increased transcription of Neuregulin 1 (type I), which is required for efficient remyelination. However, mouse nerves with EED-deficient Schwann cells display slow axonal regeneration with significantly low expression of axon guidance genes, including Sema4f and Cntf. Finally, EED loss causes impaired Schwann cell proliferation after injury with significant induction of the Cdkn2a cell cycle inhibitor gene. Interestingly, PRC2 subunits and CDKN2A are commonly co-mutated in the transition from benign neurofibromas to malignant peripheral nerve sheath tumors (MPNST's). RNA-seq analysis of EED-deficient mice identified PRC2-regulated molecular pathways that may contribute to the transition to malignancy in neurofibromatosis.
Collapse
Affiliation(s)
- Ki H. Ma
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Phu Duong
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - John J. Moran
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Nabil Junaidi
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - John Svaren
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|