1
|
Weaver MR, Shkoruta D, Pellegatta M, Berti C, Palmisano M, Ferguson S, Hurley E, French J, Patel S, Belin S, Selbach M, Paul FE, Sim F, Poitelon Y, Feltri ML. The STRIPAK complex is required for radial sorting and laminin receptor expression in Schwann cells. Cell Rep 2025; 44:115401. [PMID: 40056414 PMCID: PMC12035956 DOI: 10.1016/j.celrep.2025.115401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 01/19/2025] [Accepted: 02/14/2025] [Indexed: 03/10/2025] Open
Abstract
During peripheral nervous system development, Schwann cells undergo Rac1-dependent cytoskeletal reorganization as they insert cytoplasmic extensions into axon bundles to sort and myelinate individual axons. However, our understanding of the direct effectors targeted by Rac1 is limited. Here, we demonstrate that striatin-3 and MOB4 are Rac1 interactors. We show that Schwann-cell-specific ablation of striatin-3 causes defects in lamellipodia formation, and conditional Schwann cell knockout for striatins presents a severe delay in radial sorting. Finally, we demonstrate that deletion of Rac1 or striatin-1/3 in Schwann cells causes defects in the activation of Hippo pathway effectors YAP and TAZ and the expression of genes co-regulated by YAP and TAZ, such as extracellular matrix receptors. In summary, our results indicate that striatin-3 is a Rac1 interactor and that striatins are required for peripheral nervous system development and reveal a role for Rac1 in the regulation of the Hippo pathway in Schwann cells.
Collapse
Affiliation(s)
| | - Dominika Shkoruta
- Ivano-Frankivsk National Medical University, Ivano-Frankivsk, Ivano-Frankivsk Oblast, Ukraine
| | - Marta Pellegatta
- Institute for Myelin and Glia Exploration, Buffalo, NY, USA; Department of Biochemistry, Buffalo, NY, USA
| | - Caterina Berti
- Institute for Myelin and Glia Exploration, Buffalo, NY, USA; Department of Biochemistry, Buffalo, NY, USA
| | - Marilena Palmisano
- Institute for Myelin and Glia Exploration, Buffalo, NY, USA; Department of Biochemistry, Buffalo, NY, USA
| | - Scott Ferguson
- Institute for Myelin and Glia Exploration, Buffalo, NY, USA; Department of Pharmaceutical Sciences, Buffalo, NY, USA
| | - Edward Hurley
- Institute for Myelin and Glia Exploration, Buffalo, NY, USA
| | - Julianne French
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Shreya Patel
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | - Sophie Belin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | | | | | - Fraser Sim
- Department of Pharmacology and Toxicology, Buffalo, NY, USA
| | - Yannick Poitelon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA.
| | - M Laura Feltri
- Institute for Myelin and Glia Exploration, Buffalo, NY, USA; Department of Biochemistry, Buffalo, NY, USA; Department of Neurology, State University of New York at Buffalo Jacob's School of Medicine and Biomedical Sciences, Buffalo, NY, USA
| |
Collapse
|
2
|
Schumacher N, Vandenbosch R, Franzen R. Peripheral myelin: From development to maintenance. J Neurochem 2025; 169:e16268. [PMID: 39655795 DOI: 10.1111/jnc.16268] [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: 05/15/2024] [Revised: 10/24/2024] [Accepted: 10/28/2024] [Indexed: 12/18/2024]
Abstract
Peripheral myelin is synthesized by glial cells called Schwann cells (SCs). SC development and differentiation must be tightly regulated to avoid any pathological consequence affecting peripheral nerve function. Neuropathic symptoms can arise from developmental issues in SCs, as well as in adult life through processes affecting mature SCs. In this review we focus on SC differentiation from the immature towards the myelinating and non-myelinating SC stages, defining molecular mechanisms outlining radial sorting, a multi-stepped event essential for immature SC differentiation and myelination. We also describe mechanisms regulating myelin sheath maintenance and SC homeostasis during aging. Finally, we will conclude with some remaining questions in the field of SC biology.
Collapse
Affiliation(s)
- Nathalie Schumacher
- Laboratory of Nervous System Disorders and Therapies, GIGA Institute, University of Liège, Liège, Belgium
| | - Renaud Vandenbosch
- Laboratory of Developmental Neurobiology, GIGA Institute, University of Liège, Liège, Belgium
| | - Rachelle Franzen
- Laboratory of Nervous System Disorders and Therapies, GIGA Institute, University of Liège, Liège, Belgium
| |
Collapse
|
3
|
Weaver MR, Shkoruta D, Pellegatta M, Berti C, Palmisano M, Ferguson S, Hurley E, French J, Patel S, Belin S, Selbach M, Paul FE, Sim F, Poitelon Y, Feltri ML. The STRIPAK complex is required for radial sorting and laminin receptor expression in Schwann cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.30.620661. [PMID: 39554194 PMCID: PMC11565846 DOI: 10.1101/2024.10.30.620661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
During peripheral nervous system development, Schwann cells undergo Rac1-dependent cytoskeletal reorganization as they insert cytoplasmic extensions into axon bundles to radially sort, ensheath, and myelinate individual axons. However, our understanding of the direct effectors targeted by Rac1 is limited. Here, we demonstrate that striatin-3 and MOB4 are novel Rac1 interactors. We show that, similar to Rac1-null Schwann cells, Schwann cell specific ablation of striatin-3 causes defects in lamellipodia formation. In addition, conditional Schwann cell knockout of multiple striatin proteins presents a severe delay in radial sorting. Finally, we demonstrate here that deletion of Rac1 or striatin-1/3 in Schwann cells causes defects in Hippo pathway regulation, phosphorylation of the Hippo pathway effectors YAP and TAZ, and expression of genes co-regulated by YAP and TAZ, such as extracellular matrix receptors. In summary, our results indicate that striatin-3 is a novel Rac1 interactor, show that striatin proteins are required for peripheral nervous system development, and reveal a role for Rac1 in regulation of the Hippo pathway in Schwann cells.
Collapse
|
4
|
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
|
5
|
Zhou W, Rahman MSU, Sun C, Li S, Zhang N, Chen H, Han CC, Xu S, Liu Y. Perspectives on the Novel Multifunctional Nerve Guidance Conduits: From Specific Regenerative Procedures to Motor Function Rebuilding. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307805. [PMID: 37750196 DOI: 10.1002/adma.202307805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 09/19/2023] [Indexed: 09/27/2023]
Abstract
Peripheral nerve injury potentially destroys the quality of life by inducing functional movement disorders and sensory capacity loss, which results in severe disability and substantial psychological, social, and financial burdens. Autologous nerve grafting has been commonly used as treatment in the clinic; however, its rare donor availability limits its application. A series of artificial nerve guidance conduits (NGCs) with advanced architectures are also proposed to promote injured peripheral nerve regeneration, which is a complicated process from axon sprouting to targeted muscle reinnervation. Therefore, exploring the interactions between sophisticated NGC complexes and versatile cells during each process including axon sprouting, Schwann cell dedifferentiation, nerve myelination, and muscle reinnervation is necessary. This review highlights the contribution of functional NGCs and the influence of microscale biomaterial architecture on biological processes of nerve repair. Progressive NGCs with chemical molecule induction, heterogenous topographical morphology, electroactive, anisotropic assembly microstructure, and self-powered electroactive and magnetic-sensitive NGCs are also collected, and they are expected to be pioneering features in future multifunctional and effective NGCs.
Collapse
Affiliation(s)
- Weixian Zhou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Muhammad Saif Ur Rahman
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education Guangdong province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Chengmei Sun
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education Guangdong province, College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Shilin Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| | - Nuozi Zhang
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Hao Chen
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
| | - Charles C Han
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Materials Science and Engineering, University of Maryland, College Park, MD, 20742, USA
| | - Shanshan Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen, 518060, P. R. China
- Materials Science and Engineering, University of Maryland, College Park, MD, 20742, USA
| | - Ying Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety & CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, P. R. China
- University of Chinese Academy of Sciences, Beijing, 100049, P. R. China
| |
Collapse
|
6
|
Li Q, Huo A, Li M, Wang J, Yin Q, Chen L, Chu X, Qin Y, Qi Y, Li Y, Cui H, Cong Q. Structure, ligands, and roles of GPR126/ADGRG6 in the development and diseases. Genes Dis 2024; 11:294-305. [PMID: 37588228 PMCID: PMC10425801 DOI: 10.1016/j.gendis.2023.02.016] [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: 10/27/2022] [Revised: 12/24/2022] [Accepted: 02/05/2023] [Indexed: 03/29/2023] Open
Abstract
Adhesion G protein-coupled receptors (aGPCRs) are the second largest diverse group within the GPCR superfamily, which play critical roles in many physiological and pathological processes through cell-cell and cell-extracellular matrix interactions. The adhesion GPCR Adgrg6, also known as GPR126, is one of the better-characterized aGPCRs. GPR126 was previously found to have critical developmental roles in Schwann cell maturation and its mediated myelination in the peripheral nervous system in both zebrafish and mammals. Current studies have extended our understanding of GPR126-mediated roles during development and in human diseases. In this review, we highlighted these recent advances in GPR126 in expression profile, molecular structure, ligand-receptor interactions, and associated physiological and pathological functions in development and diseases.
Collapse
Affiliation(s)
- Qi Li
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Anran Huo
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Mengqi Li
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Jiali Wang
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Qiao Yin
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Lumiao Chen
- Department of Nephrology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| | - Xin Chu
- Department of Emergency Center, The Second Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Yuan Qin
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yuwan Qi
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
| | - Yang Li
- Department of Neurology, Huzhou Central Hospital, The Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, Zhejiang 313000, China
| | - Hengxiang Cui
- Institute of Neuroscience, State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Qifei Cong
- Institute of Neuroscience and Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, Jiangsu 215123, China
- Department of Neurology and Clinical Research Center of Neurological Disease, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215004, China
| |
Collapse
|
7
|
Wang Y, Zhang Y, Wang Y, Chen H, Pan L, Liao X, Wang S. A Novel Form of Neuregulin 1 Type III Caused by N-Terminal Processing. Biomolecules 2023; 13:1756. [PMID: 38136627 PMCID: PMC10741733 DOI: 10.3390/biom13121756] [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: 11/22/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Nrg1 (Neuregulin 1) type III, a susceptible gene of schizophrenia, exhibits a critical role in the central nervous system and is essential at each stage of Schwann's cell development. Nrg1 type III comprises double-pass transmembrane domains, with the N-terminal and C-terminal localizing inside the cells. The N-terminal transmembrane helix partially overlaps with the cysteine-rich domain (CRD). In this study, Nrg1 type III constructs with different tags were transformed into cultured cells to verify whether CRD destroyed the transmembrane helix formation. We took advantage of immunofluorescent and immunoprecipitation assays on whole cells and analyzed the N-terminal distribution. Astonishingly, we found that a novel form of Nrg1 type III, about 10% of Nrg1 type III, omitted the N-terminal transmembrane helix, with the N-terminal positioning outside the membrane. The results indicated that the novel single-pass transmembrane status was a minor form of Nrg1 type III caused by N-terminal processing, while the major form was a double-pass transmembrane status.
Collapse
Affiliation(s)
- Yukai Wang
- School of Life Sciences, Nanchang University, Nanchang 330031, China
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Yu Zhang
- School of Life Sciences, Nanchang University, Nanchang 330031, China
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Yingxing Wang
- School of Life Sciences, Nanchang University, Nanchang 330031, China
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Hong Chen
- School of Life Sciences, Nanchang University, Nanchang 330031, China
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Liangjing Pan
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Xufeng Liao
- School of Life Sciences, Nanchang University, Nanchang 330031, China
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| | - Shunqi Wang
- School of Life Sciences, Nanchang University, Nanchang 330031, China
- Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330031, China
| |
Collapse
|
8
|
Abstract
ABSTRACT Peripheral nerve injury is a common injury disease. Understanding of the mechanisms of periphery nerve repair and regeneration after injury is an essential prerequisite for treating related diseases. Although the biological mechanisms of peripheral nerve injury and regeneration have been studied comprehensively, the clinical treatment methods are still limited. The bottlenecks of the treatments are the shortage of donor nerves and the limited surgical precision. Apart from the knowledge regarding the fundamental characteristics and physical processes of peripheral nerve injury, numerous studies have found that Schwann cells, growth factors, and extracellular matrix are main factors affecting the repair and regeneration process of injured nerves. At present, the therapeutical methods of the disease include microsurgery, autologous nerve transplantation, allograft nerve transplantation and tissue engineering technology. Tissue engineering technology, which combines seed cells, neurotrophic factors, and scaffold materials together, is promising for treating the patients with long-gapped and large nerve damage. With the development of neuron science and technology, the treatment of peripheral nerve injury diseases will continue being improved.
Collapse
|
9
|
Han SH, Kim YH, Park SJ, Cho JG, Shin YK, Hong YB, Yun J, Han JY, Park HT, Park JI. COUP-TFII plays a role in cAMP-induced Schwann cell differentiation and in vitro myelination by up-regulating Krox20. J Neurochem 2023; 165:660-681. [PMID: 36648143 DOI: 10.1111/jnc.15764] [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: 12/16/2022] [Accepted: 12/23/2022] [Indexed: 01/18/2023]
Abstract
Schwann cells (SCs) are known to produce myelin for saltatory nerve conduction in the peripheral nervous system (PNS). Schwann cell differentiation and myelination processes are controlled by several transcription factors including Sox10, Oct6/Pou3f1, and Krox20/Egr2. Chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII/NR2F2) is an orphan receptor that plays a role in the development and differentiation. However, the role of COUP-TFII in the transcriptional regulatory network of SC differentiation has not been fully identified yet. Thus, the objective of this study was to investigate the role and molecular hierarchy of COUP-TFII during cAMP-induced SC differentiation. Our results showed that dibutyryl-cAMP (db-cAMP) increased expression levels of COUP-TFII along with the expressions of Oct6, Krox20, and myelin-related genes known to be related to SC differentiation. Our mechanistic studies showed that COUP-TFII acted downstream of Hsp90/ErbB2/Gab1/ERK-AKT pathway during db-cAMP-induced SC differentiation. In addition, we found that COUP-TFII induced Krox20 expression by directly binding to Krox20-MSE8 as revealed by chromatin immunoprecipitation assay and promoter activity assay. In line with this, the expression of COUP-TFII was increased before up-regulation of Oct6, Krox20, and myelin-related genes in the sciatic nerves during early postnatal myelination period. Finally, COUP-TFII knockdown by COUP-TFII siRNA or via AAV-COUP-TFII shRNA in SCs inhibited db-cAMP-induced SC differentiation and in vitro myelination of sensory axons, respectively. Taken together, these findings indicate that COUP-TFII might be involved in postnatal myelination through induction of Krox20 in SCs. Our results present a new insight into the transcriptional regulatory mechanism in SC differentiation and myelination.
Collapse
Affiliation(s)
- Sang-Heum Han
- Department of Biochemistry, Dong-A University College of Medicine, Busan, South Korea.,Peripheral Neuropathy Research Center, Dong-A University, Busan, South Korea.,Department of Translational Biomedical Sciences, Dong-A University Graduate School, Busan, South Korea
| | - Young Hee Kim
- Peripheral Neuropathy Research Center, Dong-A University, Busan, South Korea
| | - Su-Jeong Park
- Department of Biochemistry, Dong-A University College of Medicine, Busan, South Korea.,Peripheral Neuropathy Research Center, Dong-A University, Busan, South Korea.,Department of Translational Biomedical Sciences, Dong-A University Graduate School, Busan, South Korea
| | - Jun-Gi Cho
- Department of Biochemistry, Dong-A University College of Medicine, Busan, South Korea.,Peripheral Neuropathy Research Center, Dong-A University, Busan, South Korea.,Department of Translational Biomedical Sciences, Dong-A University Graduate School, Busan, South Korea
| | - Yoon Kyung Shin
- Peripheral Neuropathy Research Center, Dong-A University, Busan, South Korea
| | - Young Bin Hong
- Department of Biochemistry, Dong-A University College of Medicine, Busan, South Korea.,Peripheral Neuropathy Research Center, Dong-A University, Busan, South Korea.,Department of Translational Biomedical Sciences, Dong-A University Graduate School, Busan, South Korea
| | - Jeanho Yun
- Department of Biochemistry, Dong-A University College of Medicine, Busan, South Korea.,Peripheral Neuropathy Research Center, Dong-A University, Busan, South Korea.,Department of Translational Biomedical Sciences, Dong-A University Graduate School, Busan, South Korea
| | - Jin-Yeong Han
- Department of Laboratory Medicine, Dong-A University College of Medicine, Busan, South Korea
| | - Hwan Tae Park
- Peripheral Neuropathy Research Center, Dong-A University, Busan, South Korea.,Department of Translational Biomedical Sciences, Dong-A University Graduate School, Busan, South Korea.,Department of Molecular Neuroscience, Dong-A University College of Medicine, Busan, South Korea
| | - Joo-In Park
- Department of Biochemistry, Dong-A University College of Medicine, Busan, South Korea.,Peripheral Neuropathy Research Center, Dong-A University, Busan, South Korea.,Department of Translational Biomedical Sciences, Dong-A University Graduate School, Busan, South Korea
| |
Collapse
|
10
|
Yuan Y, Wang Y, Wu S, Zhao MY. Review: Myelin clearance is critical for regeneration after peripheral nerve injury. Front Neurol 2022; 13:908148. [PMID: 36588879 PMCID: PMC9801717 DOI: 10.3389/fneur.2022.908148] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 11/17/2022] [Indexed: 12/23/2022] Open
Abstract
Traumatic peripheral nerve injury occurs frequently and is a major clinical and public health problem that can lead to functional impairment and permanent disability. Despite the availability of modern diagnostic procedures and advanced microsurgical techniques, active recovery after peripheral nerve repair is often unsatisfactory. Peripheral nerve regeneration involves several critical events, including the recreation of the microenvironment and remyelination. Results from previous studies suggest that the peripheral nervous system (PNS) has a greater capacity for repair than the central nervous system. Thus, it will be important to understand myelin and myelination specifically in the PNS. This review provides an update on myelin biology and myelination in the PNS and discusses the mechanisms that promote myelin clearance after injury. The roles of Schwann cells and macrophages are considered at length, together with the possibility of exogenous intervention.
Collapse
Affiliation(s)
- YiMing Yuan
- Laboratory of Brain Function and Neurorehabilitation, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yan Wang
- Laboratory of Brain Function and Neurorehabilitation, Heilongjiang University of Chinese Medicine, Harbin, China,Department of Rehabilitation, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China,*Correspondence: Yan Wang
| | - ShanHong Wu
- Laboratory of Brain Function and Neurorehabilitation, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ming Yue Zhao
- Laboratory of Brain Function and Neurorehabilitation, Heilongjiang University of Chinese Medicine, Harbin, China,Department of Rehabilitation, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
11
|
Hörner SJ, Couturier N, Gueiber DC, Hafner M, Rudolf R. Development and In Vitro Differentiation of Schwann Cells. Cells 2022; 11:3753. [PMID: 36497014 PMCID: PMC9739763 DOI: 10.3390/cells11233753] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/16/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022] Open
Abstract
Schwann cells are glial cells of the peripheral nervous system. They exist in several subtypes and perform a variety of functions in nerves. Their derivation and culture in vitro are interesting for applications ranging from disease modeling to tissue engineering. Since primary human Schwann cells are challenging to obtain in large quantities, in vitro differentiation from other cell types presents an alternative. Here, we first review the current knowledge on the developmental signaling mechanisms that determine neural crest and Schwann cell differentiation in vivo. Next, an overview of studies on the in vitro differentiation of Schwann cells from multipotent stem cell sources is provided. The molecules frequently used in those protocols and their involvement in the relevant signaling pathways are put into context and discussed. Focusing on hiPSC- and hESC-based studies, different protocols are described and compared, regarding cell sources, differentiation methods, characterization of cells, and protocol efficiency. A brief insight into developments regarding the culture and differentiation of Schwann cells in 3D is given. In summary, this contribution provides an overview of the current resources and methods for the differentiation of Schwann cells, it supports the comparison and refinement of protocols and aids the choice of suitable methods for specific applications.
Collapse
Affiliation(s)
- Sarah Janice Hörner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
| | - Nathalie Couturier
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
| | - Daniele Caroline Gueiber
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Department of Electronics Engineering, Federal University of Technology Paraná, Ponta Grossa 84017-220, Brazil
| | - Mathias Hafner
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Institute of Medical Technology, Heidelberg University and Mannheim University of Applied Sciences, 69117 Heidelberg, Germany
| | - Rüdiger Rudolf
- Institute of Molecular and Cell Biology, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
- Center for Mass Spectrometry and Optical Spectroscopy, Mannheim University of Applied Sciences, 68163 Mannheim, Germany
- Institute of Medical Technology, Heidelberg University and Mannheim University of Applied Sciences, 69117 Heidelberg, Germany
| |
Collapse
|
12
|
Negro S, Pirazzini M, Rigoni M. Models and methods to study Schwann cells. J Anat 2022; 241:1235-1258. [PMID: 34988978 PMCID: PMC9558160 DOI: 10.1111/joa.13606] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 12/22/2022] Open
Abstract
Schwann cells (SCs) are fundamental components of the peripheral nervous system (PNS) of all vertebrates and play essential roles in development, maintenance, function, and regeneration of peripheral nerves. There are distinct populations of SCs including: (1) myelinating SCs that ensheath axons by a specialized plasma membrane, called myelin, which enhances the conduction of electric impulses; (2) non-myelinating SCs, including Remak SCs, which wrap bundles of multiple axons of small caliber, and perysinaptic SCs (PSCs), associated with motor axon terminals at the neuromuscular junction (NMJ). All types of SCs contribute to PNS regeneration through striking morphological and functional changes in response to nerve injury, are affected in peripheral neuropathies and show abnormalities and a diminished plasticity during aging. Therefore, methodological approaches to study and manipulate SCs in physiological and pathophysiological conditions are crucial to expand the present knowledge on SC biology and to devise new therapeutic strategies to counteract neurodegenerative conditions and age-derived denervation. We present here an updated overview of traditional and emerging methodologies for the study of SCs for scientists approaching this research field.
Collapse
Affiliation(s)
- Samuele Negro
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
| | - Marco Pirazzini
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
- CIR‐MyoCentro Interdipartimentale di Ricerca di MiologiaUniversity of PaduaPadovaItaly
| | - Michela Rigoni
- Department of Biomedical SciencesUniversity of PaduaPaduaItaly
- CIR‐MyoCentro Interdipartimentale di Ricerca di MiologiaUniversity of PaduaPadovaItaly
| |
Collapse
|
13
|
Liu Y, Yue W, Yu S, Zhou T, Zhang Y, Zhu R, Song B, Guo T, Liu F, Huang Y, Wu T, Wang H. A physical perspective to understand myelin II: The physical origin of myelin development. Front Neurosci 2022; 16:951998. [PMID: 36263368 PMCID: PMC9574017 DOI: 10.3389/fnins.2022.951998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
The physical principle of myelin development is obtained from our previous study by explaining Peter's quadrant mystery: an externally applied negative and positive E-field can promote and inhibit the growth of the inner tongue of the myelin sheath, respectively. In this study, this principle is considered as a fundamental hypothesis, named Hypothesis-E, to explain more phenomena about myelin development systematically. Specifically, the g-ratio and the fate of the Schwann cell's differentiation are explained in terms of the E-field. Moreover, an experiment is proposed to validate this theory.
Collapse
Affiliation(s)
- Yonghong Liu
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Wenji Yue
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Shoujun Yu
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Tian Zhou
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Yapeng Zhang
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Ran Zhu
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Bing Song
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Tianruo Guo
- Key Laboratory of Health Bioinformatics, Chinese Academy of Sciences, Shenzhen, China
| | - Fenglin Liu
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Yubin Huang
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| | - Tianzhun Wu
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, Australia
| | - Hao Wang
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
14
|
de Almeida V, Seabra G, Reis-de-Oliveira G, Zuccoli GS, Rumin P, Fioramonte M, Smith BJ, Zuardi AW, Hallak JEC, Campos AC, Crippa JA, Martins-de-Souza D. Cannabinoids modulate proliferation, differentiation, and migration signaling pathways in oligodendrocytes. Eur Arch Psychiatry Clin Neurosci 2022; 272:1311-1323. [PMID: 35622101 DOI: 10.1007/s00406-022-01425-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 05/02/2022] [Indexed: 11/03/2022]
Abstract
Cannabinoid signaling, mainly via CB1 and CB2 receptors, plays an essential role in oligodendrocyte health and functions. However, the specific molecular signals associated with the activation or blockade of CB1 and CB2 receptors in this glial cell have yet to be elucidated. Mass spectrometry-based shotgun proteomics and in silico biology tools were used to determine which signaling pathways and molecular mechanisms are triggered in a human oligodendrocytic cell line (MO3.13) by several pharmacological stimuli: the phytocannabinoid cannabidiol (CBD); CB1 and CB2 agonists ACEA, HU308, and WIN55, 212-2; CB1 and CB2 antagonists AM251 and AM630; and endocannabinoids anandamide (AEA) and 2-arachidonoylglycerol (2-AG). The modulation of cannabinoid signaling in MO3.13 was found to affect pathways linked to cell proliferation, migration, and differentiation of oligodendrocyte progenitor cells. Additionally, we found that carbohydrate and lipid metabolism, as well as mitochondrial function, were modulated by these compounds. Comparing the proteome changes and upstream regulators among treatments, the highest overlap was between the CB1 and CB2 antagonists, followed by overlaps between AEA and 2-AG. Our study opens new windows of opportunities, suggesting that cannabinoid signaling in oligodendrocytes might be relevant in the context of demyelinating and neurodegenerative diseases. Proteomics data are available at ProteomeXchange (PXD031923).
Collapse
Affiliation(s)
- Valéria de Almeida
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil.
| | - Gabriela Seabra
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil
| | - Guilherme Reis-de-Oliveira
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil
| | - Giuliana S Zuccoli
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil
| | - Priscila Rumin
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil
| | - Mariana Fioramonte
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil
| | - Bradley J Smith
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil
| | - Antonio W Zuardi
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,National Institute for Science and Technology, Translational Medicine, São Paulo, Brazil
| | - Jaime E C Hallak
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,National Institute for Science and Technology, Translational Medicine, São Paulo, Brazil
| | - Alline C Campos
- National Institute for Science and Technology, Translational Medicine, São Paulo, Brazil.,Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - José A Crippa
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil.,National Institute for Science and Technology, Translational Medicine, São Paulo, Brazil
| | - Daniel Martins-de-Souza
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Rua Monteiro Lobato, Campinas, SP, 255, 13083-862, Brazil. .,Instituto Nacional de Biomarcadores Em Neuropsiquiatria (INBION) Conselho Nacional de Desenvolvimento Científico E Tecnológico, São Paulo, Brazil. .,Experimental Medicine Research Cluster (EMRC), University of Campinas, Campinas, SP, Brazil. .,D'Or Institute for Research and Education (IDOR), São Paulo, Brazil.
| |
Collapse
|
15
|
Abstract
Schwann cells in the peripheral nervous system (PNS) are essential for the support and myelination of axons, ensuring fast and accurate communication between the central nervous system and the periphery. Schwann cells and related glia accompany innervating axons in virtually all tissues in the body, where they exhibit remarkable plasticity and the ability to modulate pathology in extraordinary, and sometimes surprising, ways. Here, we provide a brief overview of the various glial cell types in the PNS and describe the cornerstone cellular and molecular processes that enable Schwann cells to perform their canonical functions. We then dive into discussing exciting noncanonical functions of Schwann cells and related PNS glia, which include their role in organizing the PNS, in regulating synaptic activity and pain, in modulating immunity, in providing a pool of stem cells for different organs, and, finally, in influencing cancer.
Collapse
Affiliation(s)
- Carla Taveggia
- Axo-Glial Interaction Unit, Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy;
| | - M. Laura Feltri
- Institute for Myelin and Glia Exploration, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| |
Collapse
|
16
|
McKee KK, Yurchenco PD. Amelioration of muscle and nerve pathology of Lama2-related dystrophy by AAV9-laminin-αLN-linker protein. JCI Insight 2022; 7:158397. [PMID: 35639486 PMCID: PMC9310540 DOI: 10.1172/jci.insight.158397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 05/25/2022] [Indexed: 11/17/2022] Open
Abstract
LAMA2 deficiency, resulting from a defective or absent laminin α2 subunit, is a common cause of congenital muscular dystrophy. It is characterized by muscle weakness from myofiber degeneration and neuropathy from Schwann cell amyelination. Previously it was shown that transgenic muscle-specific expression of αLNNd, a laminin γ1–binding linker protein that enables polymerization in defective laminins, selectively ameliorates the muscle abnormality in mouse disease models. Here, adeno-associated virus was used to deliver linker mini-genes to dystrophic dy2J/dy2J mice for expression of αLNNd in muscle, or αLNNdΔG2′, a shortened linker, in muscle, nerve, and other tissues. Linker and laminin α2 levels were higher in αLNNdΔG2′-treated mice. Both αLNNd- and αLNNdΔG2′-treated mice exhibited increased forelimb grip strength. Further, αLNNdΔG2′-treated mice achieved hind limb and all-limb grip strength levels approaching those of WT mice as well as ablation of hind limb paresis and contractures. This was accompanied by restoration of sciatic nerve axonal envelopment and myelination. Improvement of muscle histology was evident in the muscle-specific αLNNd-expressing mice but more extensive in the αLNNdΔG2′-expressing mice. The results reveal that an αLN linker mini-gene, driven by a ubiquitous promoter, is superior to muscle-specific delivery because of its higher expression that extends to the peripheral nerve. These studies support a potentially novel approach of somatic gene therapy.
Collapse
Affiliation(s)
- Karen K McKee
- Department of Pathology & Laboratory Medicine, Rutgers University - Robert Wood Johnson Medical School, Piscataway, United States of America
| | - Peter D Yurchenco
- Department of Pathology & Laboratory Medicine, Rutgers University - Robert Wood Johnson Medical School, Piscataway, United States of America
| |
Collapse
|
17
|
Touchant M, Labonté B. Sex-Specific Brain Transcriptional Signatures in Human MDD and Their Correlates in Mouse Models of Depression. Front Behav Neurosci 2022; 16:845491. [PMID: 35592639 PMCID: PMC9110970 DOI: 10.3389/fnbeh.2022.845491] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 04/05/2022] [Indexed: 01/13/2023] Open
Abstract
Major depressive disorder (MDD) is amongst the most devastating psychiatric conditions affecting several millions of people worldwide every year. Despite the importance of this disease and its impact on modern societies, still very little is known about the etiological mechanisms. Treatment strategies have stagnated over the last decades and very little progress has been made to improve the efficiency of current therapeutic approaches. In order to better understand the disease, it is necessary for researchers to use appropriate animal models that reproduce specific aspects of the complex clinical manifestations at the behavioral and molecular levels. Here, we review the current literature describing the use of mouse models to reproduce specific aspects of MDD and anxiety in males and females. We first describe some of the most commonly used mouse models and their capacity to display unique but also shared features relevant to MDD. We then transition toward an integral description, combined with genome-wide transcriptional strategies. The use of these models reveals crucial insights into the molecular programs underlying the expression of stress susceptibility and resilience in a sex-specific fashion. These studies performed on human and mouse tissues establish correlates into the mechanisms mediating the impact of stress and the extent to which different mouse models of chronic stress recapitulate the molecular changes observed in depressed humans. The focus of this review is specifically to highlight the sex differences revealed from different stress paradigms and transcriptional analyses both in human and animal models.
Collapse
Affiliation(s)
- Maureen Touchant
- CERVO Brain Research Centre, Québec, QC, Canada
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Benoit Labonté
- CERVO Brain Research Centre, Québec, QC, Canada
- Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec, QC, Canada
- *Correspondence: Benoit Labonté
| |
Collapse
|
18
|
Park HJ, Tsai E, Huang D, Weaver M, Frick L, Alcantara A, Moran JJ, Patzig J, Melendez-Vasquez CV, Crabtree GR, Feltri M, Svaren J, Casaccia P. ACTL6a coordinates axonal caliber recognition and myelination in the peripheral nerve. iScience 2022; 25:104132. [PMID: 35434551 PMCID: PMC9010646 DOI: 10.1016/j.isci.2022.104132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 01/29/2022] [Accepted: 03/17/2022] [Indexed: 11/12/2022] Open
Abstract
Cells elaborate transcriptional programs in response to external signals. In the peripheral nerves, Schwann cells (SC) sort axons of given caliber and start the process of wrapping their membrane around them. We identify Actin-like protein 6a (ACTL6a), part of SWI/SNF chromatin remodeling complex, as critical for the integration of axonal caliber recognition with the transcriptional program of myelination. Nuclear levels of ACTL6A in SC are increased by contact with large caliber axons or nanofibers, and result in the eviction of repressive histone marks to facilitate myelination. Without Actl6a the SC are unable to coordinate caliber recognition and myelin production. Peripheral nerves in knockout mice display defective radial sorting, hypo-myelination of large caliber axons, and redundant myelin around small caliber axons, resulting in a clinical motor phenotype. Overall, this suggests that ACTL6A is a key component of the machinery integrating external signals for proper myelination of the peripheral nerve.
Collapse
Affiliation(s)
- Hye-Jin Park
- Advanced Science Research Center (ASRC) at The Graduate Center of the City University of New York (CUNY), New York, NY 10031, USA
| | - Eric Tsai
- Advanced Science Research Center (ASRC) at The Graduate Center of the City University of New York (CUNY), New York, NY 10031, USA
- Graduate Program in Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Dennis Huang
- Advanced Science Research Center (ASRC) at The Graduate Center of the City University of New York (CUNY), New York, NY 10031, USA
- Graduate Program in Biology, Graduate Center of CUNY, New York, NY 10016, USA
| | - Michael Weaver
- Hunter James Kelly Research Institute, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Luciana Frick
- Hunter James Kelly Research Institute, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Ace Alcantara
- Graduate Program in Biology, Graduate Center of CUNY, New York, NY 10016, USA
- Hunter College, Department of Biological Sciences, New York, NY 10065, USA
| | - John J. Moran
- Waisman Center and Department of Comparative Biosciences, University of Wisconsin, Madison, WI 53705, USA
| | - Julia Patzig
- Advanced Science Research Center (ASRC) at The Graduate Center of the City University of New York (CUNY), New York, NY 10031, USA
| | - Carmen V. Melendez-Vasquez
- Graduate Program in Biology, Graduate Center of CUNY, New York, NY 10016, USA
- Hunter College, Department of Biological Sciences, New York, NY 10065, USA
| | - Gerald R. Crabtree
- Department of Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - M.L. Feltri
- Hunter James Kelly Research Institute, Departments of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - John Svaren
- Waisman Center and Department of Comparative Biosciences, University of Wisconsin, Madison, WI 53705, USA
| | - Patrizia Casaccia
- Advanced Science Research Center (ASRC) at The Graduate Center of the City University of New York (CUNY), New York, NY 10031, USA
- Graduate Program in Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Graduate Program in Biology, Graduate Center of CUNY, New York, NY 10016, USA
| |
Collapse
|
19
|
Hibbitts AJ, Kočí Z, Kneafsey S, Matsiko A, Žilić L, Dervan A, Hinton P, Chen G, Cavanagh B, Dowling J, McCoy C, Buckley CT, Archibald SJ, O'Brien FJ. Multi-Factorial Nerve Guidance Conduit Engineering Improves Outcomes in Inflammation, Angiogenesis and Large Defect Nerve Repair. Matrix Biol 2022; 106:34-57. [PMID: 35032612 DOI: 10.1016/j.matbio.2022.01.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 11/13/2021] [Accepted: 01/05/2022] [Indexed: 12/20/2022]
Abstract
Nerve guidance conduits (NGCs) are sub-optimal for long-distance injuries with inflammation and poor vascularization related to poor axonal repair. This study used a multi-factorial approach to create an optimized biomaterial NGC to address each of these issues. Through stepwise optimization, a collagen-chondroitin-6-sulphate (Coll-CS) biomaterial was functionalized with extracellular matrix (ECM) components; fibronectin, laminin 1 and laminin 2 (FibL1L2) in specific ratios. A snap-cooled freeze-drying process was then developed with optimal pore architecture and alignment to guide axonal bridging. Culture of adult rat dorsal root ganglia on NGCs demonstrated significant improvements in inflammation, neurogenesis and angiogenesis in the specific Fib:L1:L2 ratio of 1:4:1. In clinically relevant, large 15 mm rat sciatic nerve defects, FibL1L2-NGCs demonstrated significant improvements in axonal density and angiogenesis compared to unmodified NGCs with functional equivalence to autografts. Therefore, a multiparameter ECM-driven strategy can significantly improve axonal repair across large defects, without exogenous cells or growth factors.
Collapse
Affiliation(s)
- Alan J Hibbitts
- Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Zuzana Kočí
- Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Simone Kneafsey
- Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Amos Matsiko
- Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Leyla Žilić
- Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Adrian Dervan
- Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Paige Hinton
- Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Gang Chen
- Department of Physiology and Medical Physics, Centre for the Study of Neurological Disorders, Microsurgical Research and Training Facility (MRTF), RCSI, Dublin, Ireland
| | | | - Jennifer Dowling
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, D02 YN77 Dublin, Ireland
| | - Claire McCoy
- School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, D02 YN77 Dublin, Ireland
| | - Conor T Buckley
- Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | | | - Fergal J O'Brien
- Tissue Engineering Research Group, Dept. of Anatomy & Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin, The University of Dublin, Dublin, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| |
Collapse
|
20
|
Zotter B, Dagan O, Brady J, Baloui H, Samanta J, Salzer JL. Gli1 Regulates the Postnatal Acquisition of Peripheral Nerve Architecture. J Neurosci 2022; 42:183-201. [PMID: 34772739 PMCID: PMC8802940 DOI: 10.1523/jneurosci.3096-20.2021] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 10/17/2021] [Accepted: 10/19/2021] [Indexed: 11/21/2022] Open
Abstract
Peripheral nerves are organized into discrete compartments. Axons, Schwann cells (SCs), and endoneurial fibroblasts (EFs) reside within the endoneurium and are surrounded by the perineurium, a cellular sheath comprised of layers of perineurial glia (PNG). SC secretion of Desert Hedgehog (Dhh) regulates this organization. In Dhh nulls, the perineurium is deficient and the endoneurium is subdivided into small compartments termed minifascicles. Human Dhh mutations cause a neuropathy with similar defects. Here we examine the role of Gli1, a canonical transcriptional effector of hedgehog signaling, in regulating peripheral nerve organization in mice of both genders. We identify PNG, EFs, and pericytes as Gli1-expressing cells by genetic fate mapping. Although expression of Dhh by SCs and Gli1 in target cells is coordinately regulated with myelination, Gli1 expression unexpectedly persists in Dhh null EFs. Thus, Gli1 is expressed in EFs noncanonically (i.e., independent of hedgehog signaling). Gli1 and Dhh also have nonredundant activities. Unlike Dhh nulls, Gli1 nulls have a normal perineurium. Like Dhh nulls, Gli1 nulls form minifascicles, which we show likely arise from EFs. Thus, Dhh and Gli1 are independent signals: Gli1 is dispensable for perineurial development but functions cooperatively with Dhh to drive normal endoneurial development. During development, Gli1 also regulates endoneurial extracellular matrix production, nerve vascular organization, and has modest, nonautonomous effects on SC sorting and myelination of axons. Finally, in adult nerves, induced deletion of Gli1 is sufficient to drive minifascicle formation. Thus, Gli1 regulates the development and is required to maintain the endoneurial architecture of peripheral nerves.SIGNIFICANCE STATEMENT Peripheral nerves are organized into distinct cellular/ECM compartments: the epineurium, perineurium, and endoneurium. This organization, with its associated cellular constituents, is critical for the structural and metabolic support of nerves and their response to injury. Here, we show that Gli1, a transcription factor normally expressed downstream of hedgehog signaling, is required for the proper organization of the endoneurium but not the perineurium. Unexpectedly, Gli1 expression by endoneurial cells is independent of, and functions nonredundantly with, Schwann Cell-derived Desert Hedgehog in regulating peripheral nerve architecture. These results further delineate how peripheral nerves acquire their distinctive organization during normal development, and highlight mechanisms that may regulate their reorganization in pathologic settings, including peripheral neuropathies and nerve injury.
Collapse
Affiliation(s)
- Brendan Zotter
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Langone Medical Center, New York, New York 10016
| | - Or Dagan
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Langone Medical Center, New York, New York 10016
| | - Jacob Brady
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Langone Medical Center, New York, New York 10016
| | - Hasna Baloui
- Departments of Neuroscience and Clinical Neuroscience, Karolinska Institutet, Stockholm, 17177, Sweden
| | - Jayshree Samanta
- Department of Comparative Biosciences, School of Veterinary Medicine, Stem Cell and Regenerative Medicine Center, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - James L Salzer
- Department of Neuroscience and Physiology, Neuroscience Institute, NYU Langone Medical Center, New York, New York 10016
| |
Collapse
|
21
|
Previtali SC. Peripheral Nerve Development and the Pathogenesis of Peripheral Neuropathy: the Sorting Point. Neurotherapeutics 2021; 18:2156-2168. [PMID: 34244926 PMCID: PMC8804061 DOI: 10.1007/s13311-021-01080-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2021] [Indexed: 12/12/2022] Open
Abstract
Nerve development requires a coordinated sequence of events and steps to be accomplished for the generation of functional peripheral nerves to convey sensory and motor signals. Any abnormality during development may result in pathological structure and function of the nerve, which evolves in peripheral neuropathy. In this review, we will briefly describe different steps of nerve development while we will mostly focus on the molecular mechanisms involved in radial sorting of axons, one of these nerve developmental steps. We will summarize current knowledge of molecular pathways so far reported in radial sorting and their possible interactions. Finally, we will describe how disruption of these pathways may result in human neuropathies.
Collapse
Affiliation(s)
- Stefano C Previtali
- Neuromuscular Repair Unit, InSpe (Institute of Experimental Neurology) and Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
22
|
Wrestling and Wrapping: A Perspective on SUMO Proteins in Schwann Cells. Biomolecules 2021; 11:biom11071055. [PMID: 34356679 PMCID: PMC8301837 DOI: 10.3390/biom11071055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 11/20/2022] Open
Abstract
Schwann cell development and peripheral nerve myelination are finely orchestrated multistep processes; some of the underlying mechanisms are well described and others remain unknown. Many posttranslational modifications (PTMs) like phosphorylation and ubiquitination have been reported to play a role during the normal development of the peripheral nervous system (PNS) and in demyelinating neuropathies. However, a relatively novel PTM, SUMOylation, has not been studied in these contexts. SUMOylation involves the covalent attachment of one or more small ubiquitin-like modifier (SUMO) proteins to a substrate, which affects the function, cellular localization, and further PTMs of the conjugated protein. SUMOylation also regulates other proteins indirectly by facilitating non-covalent protein–protein interaction via SUMO interaction motifs (SIM). This pathway has important consequences on diverse cellular processes, and dysregulation of this pathway has been reported in several diseases including neurological and degenerative conditions. In this article, we revise the scarce literature on SUMOylation in Schwann cells and the PNS, we propose putative substrate proteins, and we speculate on potential mechanisms underlying the possible involvement of this PTM in peripheral myelination and neuropathies.
Collapse
|
23
|
Gerber D, Pereira JA, Gerber J, Tan G, Dimitrieva S, Yángüez E, Suter U. Transcriptional profiling of mouse peripheral nerves to the single-cell level to build a sciatic nerve ATlas (SNAT). eLife 2021; 10:e58591. [PMID: 33890853 PMCID: PMC8064760 DOI: 10.7554/elife.58591] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
Peripheral nerves are organ-like structures containing diverse cell types to optimize function. This interactive assembly includes mostly axon-associated Schwann cells, but also endothelial cells of supporting blood vessels, immune system-associated cells, barrier-forming cells of the perineurium surrounding and protecting nerve fascicles, and connective tissue-resident cells within the intra-fascicular endoneurium and inter-fascicular epineurium. We have established transcriptional profiles of mouse sciatic nerve-inhabitant cells to foster the fundamental understanding of peripheral nerves. To achieve this goal, we have combined bulk RNA sequencing of developing sciatic nerves up to the adult with focused bulk and single-cell RNA sequencing of Schwann cells throughout postnatal development, extended by single-cell transcriptome analysis of the full sciatic nerve both perinatally and in the adult. The results were merged in the transcriptome resource Sciatic Nerve ATlas (SNAT: https://www.snat.ethz.ch). We anticipate that insights gained from our multi-layered analysis will serve as valuable interactive reference point to guide future studies.
Collapse
Affiliation(s)
- Daniel Gerber
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Jorge A Pereira
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Joanne Gerber
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| | - Ge Tan
- Functional Genomics Center Zurich, ETH Zurich/University of ZurichZurichSwitzerland
| | - Slavica Dimitrieva
- Functional Genomics Center Zurich, ETH Zurich/University of ZurichZurichSwitzerland
| | - Emilio Yángüez
- Functional Genomics Center Zurich, ETH Zurich/University of ZurichZurichSwitzerland
| | - Ueli Suter
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology, ETH ZurichZurichSwitzerland
| |
Collapse
|
24
|
Park K, Shin Y, Lee G, Park H, Choi Y. Dabrafenib Promotes Schwann Cell Differentiation by Inhibition of the MEK-ERK Pathway. Molecules 2021; 26:2141. [PMID: 33917810 PMCID: PMC8068149 DOI: 10.3390/molecules26082141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/04/2021] [Accepted: 04/07/2021] [Indexed: 11/16/2022] Open
Abstract
Schwann cell differentiation involves a dynamic interaction of signaling cascades. However, much remains to be elucidated regarding the function of signaling molecules that differ depending on the context in which the molecules are engaged. Here, we identified a small molecule, dabrafenib, which promotes Schwann cell differentiation in vitro and exploited this compound as a pharmacological tool to understand the molecular mechanisms regulating Schwann cell differentiation. The results indicated that dabrafenib inhibited ERK phosphorylation and enhanced ErbB2 autophosphorylation and Akt phosphorylation, and the effects of dabrafenib on ErbB2 and Akt phosphorylation were phenocopied by pharmacological inhibition of the MEK-ERK signaling pathway. However, the small molecule inhibitors of MEK and ERK had no effect on the expression of Oct6 and EGR2, which are key transcription factors that drive Schwann cell differentiation. In addition, pharmacological inhibition of phosphatidylinositol-3-kinase (PI3K) almost completely interfered with dabrafenib-induced Schwann cell differentiation. These results suggest that the ErbB2-PI3K-Akt axis is required for the induction of Schwann cell differentiation by dabrafenib in vitro. Although additional molecules targeted by dabrafenib remain to be identified, our data provides insights into the crosstalk that exists between the MEK-ERK signaling pathway and the PI3K-Akt axis in Schwann cell differentiation.
Collapse
Affiliation(s)
- Kyuhee Park
- Biocenter, Gyeonggido Business and Science Accelerator, Suwon 16229, Korea; (K.P.); (G.L.)
| | - Yoonkyoung Shin
- Department of Physiology, Peripheral Neuropathy Research Center, Donga University Medical School, Busan 49201, Korea; (Y.S.); (H.P.)
| | - Gyeongbeen Lee
- Biocenter, Gyeonggido Business and Science Accelerator, Suwon 16229, Korea; (K.P.); (G.L.)
| | - Hwantae Park
- Department of Physiology, Peripheral Neuropathy Research Center, Donga University Medical School, Busan 49201, Korea; (Y.S.); (H.P.)
| | - Yongmun Choi
- Biocenter, Gyeonggido Business and Science Accelerator, Suwon 16229, Korea; (K.P.); (G.L.)
| |
Collapse
|
25
|
Pellegatta M, Berti C, Hurley E, Palmisano M, de Curtis I, Feltri ML, Frick LR. Rac1 and Rac3 have opposite functions in Schwann cells during developmental myelination. Neurosci Lett 2021; 753:135868. [PMID: 33812927 DOI: 10.1016/j.neulet.2021.135868] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 03/14/2021] [Accepted: 03/29/2021] [Indexed: 11/15/2022]
Abstract
Small Rho GTPases such as Cdc42 and Rac1 regulate peripheral myelination during development. Deletion of Rac1 in Schwann cell conditional knockout mice causes a delay in the process of radial sorting, followed by hypomyelination as well as defective PAK1 activation and high number of immature Oct6+ Schwann cells. Rac3 has been shown to have redundant, specific and even opposite functions to Rac1 depending on the cell type, age and other factors. In neuronal cells, evidence suggests that Rac3 may oppose Rac1 by disrupting PAK1-GIT1-Paxillin signaling thus preventing cell differentiation and extension of lamellipodia. Therefore, we tested if these Rho GTPases have similar or opposite functions in Schwann cells, by deleting the genes for both proteins in mice during peripheral myelination. At P30, global deletion of Rac3 alleviates the developmental defects on axonal sorting and hypomyelination that are caused by Schwann cell conditional ablation of Rac1. Moreover, Rac3 deletion also reverses the arrest of Schwann cells at the Oct6+ stage and ameliorates the defects in PAK1 phosphorylation observed in Rac1 deficient mice. This partial rescue of the phenotype declines later on with aging. Since double transgenic animals showed dysmyelination without axonal degeneration at P60, we postulate that this deterioration is not likely due to loss of Rac3 in neurons, but it seems to be a Schwann cell-specific defect in the maintenance of myelin.
Collapse
Affiliation(s)
- Marta Pellegatta
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, United States; Departments of Biochemistry Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, United States
| | - Caterina Berti
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, United States; Departments of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, United States
| | - Edward Hurley
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, United States; Departments of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, United States
| | - Marilena Palmisano
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, United States; Departments of Biochemistry Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, United States
| | - Ivan de Curtis
- San Raffaele Hospital, Division of Neuroscience, Milano, 20132, Italy
| | - M Laura Feltri
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, United States; Departments of Biochemistry Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, United States; Departments of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, United States
| | - Luciana R Frick
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, United States; Departments of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, United States.
| |
Collapse
|
26
|
Feltri ML, Weaver MR, Belin S, Poitelon Y. The Hippo pathway: Horizons for innovative treatments of peripheral nerve diseases. J Peripher Nerv Syst 2021; 26:4-16. [PMID: 33449435 DOI: 10.1111/jns.12431] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/16/2020] [Accepted: 12/20/2020] [Indexed: 12/19/2022]
Abstract
Initially identified in Drosophila, the Hippo signaling pathway regulates how cells respond to their environment by controlling proliferation, migration and differentiation. Many recent studies have focused on characterizing Hippo pathway function and regulation in mammalian cells. Here, we present a brief overview of the major components of the Hippo pathway, as well as their regulation and function. We comprehensively review the studies that have contributed to our understanding of the Hippo pathway in the function of the peripheral nervous system and in peripheral nerve diseases. Finally, we discuss innovative approaches that aim to modulate Hippo pathway components in diseases of the peripheral nervous system.
Collapse
Affiliation(s)
- M Laura Feltri
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA.,Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | - Michael R Weaver
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, Buffalo, New York, USA
| | - Sophie Belin
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Yannick Poitelon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| |
Collapse
|
27
|
Xu W, Zhang S, Feng Y, Zhang C, Xiao Y, Tian F. iTRAQ-based proteomic analysis of the hippocampus of pentylenetetrazole-kindled epileptic rats. Int J Dev Neurosci 2020; 81:125-141. [PMID: 33316100 DOI: 10.1002/jdn.10082] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 11/24/2020] [Accepted: 12/09/2020] [Indexed: 11/06/2022] Open
Abstract
Epilepsy can severely affect the quality of life of patients, who are often at higher risk of mortality. However, the molecular mechanisms and pathogenesis underlying epileptogenesis are poorly understood. In this study, we performed a proteomic analysis of the hippocampus in pentylenetetrazole (PTZ)-kindled epileptic rats to explore the molecular mechanisms of epileptogenesis. We established an epileptic model in Sprague Dawley rats by injecting PTZ intraperitoneally and applied isobaric tags for relative and absolute quantification (iTRAQ) technology integrated with liquid chromatography-tandem mass spectrometry (LC-MS/MS) to identify differentially expressed proteins (DEPs) in the hippocampus. A total of 99 proteins, comprising 93 upregulated and 6 downregulated proteins, were identified based on a fold change >1.2 (or <0.83) and a p-value < .05. A further bioinformatics analysis suggested that the candidate proteins were mainly involved in the ubiquitin ligase complex or metabolite homeostasis or acted as intrinsic components of the membrane. A Kyoto Encyclopedia of Gene and Genomes (KEGG) pathway enrichment analysis identified a series of representative pathological pathways, including the calcium signaling pathway, neuroactive ligand-receptor interaction pathway, and the NF-kappa B pathway. The mass spectrometry results were further confirmed by assessing five representative proteins (Akt1, Syvn1, Amfr, Lamb1, and Cox17) using western blotting and immunohistochemistry. These results may help to reveal the molecular mechanisms underlying epileptogenesis and provide new directions or targets for epilepsy research.
Collapse
Affiliation(s)
- Weiye Xu
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Siyuan Zhang
- Department of Neurology, Hunan Provincial People's Hospital, Changsha, P.R. China
| | - Yanyan Feng
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Chen Zhang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Yeqing Xiao
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, P.R. China
| | - Fafa Tian
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, P.R. China
| |
Collapse
|
28
|
Polo Y, Luzuriaga J, Iturri J, Irastorza I, Toca-Herrera JL, Ibarretxe G, Unda F, Sarasua JR, Pineda JR, Larrañaga A. Nanostructured scaffolds based on bioresorbable polymers and graphene oxide induce the aligned migration and accelerate the neuronal differentiation of neural stem cells. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 31:102314. [PMID: 33059092 DOI: 10.1016/j.nano.2020.102314] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/17/2020] [Accepted: 09/24/2020] [Indexed: 12/30/2022]
Abstract
Within the field of neural tissue engineering, there is a huge need for the development of materials that promote the adhesion, aligned migration and differentiation of stem cells into neuronal and supportive glial cells. In this study, we have fabricated bioresorbable elastomeric scaffolds combining an ordered nanopatterned topography together with a surface functionalization with graphene oxide (GO) in mild conditions. These scaffolds allowed the attachment of murine neural stem cells (NSCs) without the need of any further coating of its surface with extracellular matrix adhesion proteins. The NSCs were able to give rise to both immature neurons and supporting glial cells over the nanostructured scaffolds in vitro, promoting their aligned migration in cell clusters following the nanostructured grooves. This system has the potential to reestablish spatially oriented neural precursor cell connectivity, constituting a promising tool for future cellular therapy including nerve tissue regeneration.
Collapse
Affiliation(s)
- Yurena Polo
- Polimerbio SL, Donostia-San Sebastian, Spain
| | - Jon Luzuriaga
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Jagoba Iturri
- Institute for Biophysics, Department of Nanobiotechnology, BOKU University of Natural Resources and Life Sciences, Vienna, Austria
| | - Igor Irastorza
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - José Luis Toca-Herrera
- Institute for Biophysics, Department of Nanobiotechnology, BOKU University of Natural Resources and Life Sciences, Vienna, Austria
| | - Gaskon Ibarretxe
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Fernando Unda
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain
| | - Jose-Ramon Sarasua
- Group of Science and Engineering of Polymeric Biomaterials (ZIBIO Group), Department of Mining, Metallurgy Engineering and Materials Science & POLYMAT, University of the Basque Country (UPV/EHU), Bilbao, Spain
| | - Jose Ramon Pineda
- Department of Cell Biology and Histology, Faculty of Medicine and Nursing, University of the Basque Country (UPV/EHU), Leioa, Spain; Achucarro Basque Center for Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Spain.
| | - Aitor Larrañaga
- Group of Science and Engineering of Polymeric Biomaterials (ZIBIO Group), Department of Mining, Metallurgy Engineering and Materials Science & POLYMAT, University of the Basque Country (UPV/EHU), Bilbao, Spain.
| |
Collapse
|
29
|
Arreguin AJ, Colognato H. Brain Dysfunction in LAMA2-Related Congenital Muscular Dystrophy: Lessons From Human Case Reports and Mouse Models. Front Mol Neurosci 2020; 13:118. [PMID: 32792907 PMCID: PMC7390928 DOI: 10.3389/fnmol.2020.00118] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/09/2020] [Indexed: 12/26/2022] Open
Abstract
Laminin α2 gene (LAMA2)-related Congenital Muscular Dystrophy (CMD) was distinguished by a defining central nervous system (CNS) abnormality—aberrant white matter signals by MRI—when first described in the 1990s. In the past 25 years, researchers and clinicians have expanded our knowledge of brain involvement in LAMA2-related CMD, also known as Congenital Muscular Dystrophy Type 1A (MDC1A). Neurological changes in MDC1A can be structural, including lissencephaly and agyria, as well as functional, including epilepsy and intellectual disability. Mouse models of MDC1A include both spontaneous and targeted LAMA2 mutations and range from a partial loss of LAMA2 function (e.g., dy2J/dy2J), to a complete loss of LAMA2 expression (dy3K/dy3K). Diverse cellular and molecular changes have been reported in the brains of MDC1A mouse models, including blood-brain barrier dysfunction, altered neuro- and gliogenesis, changes in synaptic plasticity, and decreased myelination, providing mechanistic insight into potential neurological dysfunction in MDC1A. In this review article, we discuss selected studies that illustrate the potential scope and complexity of disturbances in brain development in MDC1A, and as well as highlight mechanistic insights that are emerging from mouse models.
Collapse
Affiliation(s)
- Andrea J Arreguin
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, United States.,Medical Scientist Training Program (MSTP), Stony Brook University, Stony Brook, NY, United States
| | - Holly Colognato
- Department of Pharmacological Sciences, Stony Brook University, Stony Brook, NY, United States
| |
Collapse
|
30
|
Scarpa JR, Fatma M, Loh YHE, Traore SR, Stefan T, Chen TH, Nestler EJ, Labonté B. Shared Transcriptional Signatures in Major Depressive Disorder and Mouse Chronic Stress Models. Biol Psychiatry 2020; 88:159-168. [PMID: 32169281 PMCID: PMC7740570 DOI: 10.1016/j.biopsych.2019.12.029] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 12/03/2019] [Accepted: 12/24/2019] [Indexed: 01/06/2023]
Abstract
BACKGROUND Most of our knowledge of the biological basis of major depressive disorder (MDD) is derived from studies of chronic stress models in rodents. While these models capture certain aspects of the behavioral and neuroendocrine features of MDD, the extent to which they reproduce the molecular pathology of the human syndrome remains unknown. METHODS We systematically compared transcriptional signatures in two brain regions implicated in depression-medial prefrontal cortex and nucleus accumbens-of humans with MDD and of 3 chronic stress models in mice: chronic variable stress, adult social isolation, and chronic social defeat stress. We used differential expression analysis combined with weighted gene coexpression network analysis to create interspecies gene networks and assess the capacity of each stress paradigm to recapitulate the transcriptional organization of gene networks in human MDD. RESULTS Our results show significant overlap between transcriptional alterations in medial prefrontal cortex and nucleus accumbens in human MDD and the 3 mouse chronic stress models, with each of the chronic stress paradigms capturing distinct aspects of MDD abnormalities. Chronic variable stress and adult social isolation better reproduce differentially expressed genes, while chronic social defeat stress and adult social isolation better reproduce gene networks characteristic of human MDD. We also identified several gene networks and their constituent genes that are most significantly associated with human MDD and mouse stress models. CONCLUSIONS This study demonstrates the ability of 3 chronic stress models in mice to recapitulate distinct aspects of the broad molecular pathology of human MDD, with no one mouse model apparently better than another.
Collapse
Affiliation(s)
- Joseph R Scarpa
- Department of Anesthesiology, Weill Cornell Medicine, New York, New York
| | - Mena Fatma
- Department of Psychiatry and Neurosciences, Laval University, Québec, Québec, Canada
| | - Yong-Hwee E Loh
- Norris Medical Library, University of Southern California, Los Angeles, California
| | - Said Romaric Traore
- Department of Psychiatry and Neurosciences, Laval University, Québec, Québec, Canada
| | - Theo Stefan
- Department of Psychiatry and Neurosciences, Laval University, Québec, Québec, Canada
| | - Ting Huei Chen
- Department of Psychiatry and Neurosciences, Laval University, Québec, Québec, Canada
| | - Eric J Nestler
- Nash Family Department of Neuroscience and Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Benoit Labonté
- Department of Psychiatry and Neurosciences, Laval University, Québec, Québec, Canada.
| |
Collapse
|
31
|
Wilson ER, Della-Flora Nunes G, Weaver MR, Frick LR, Feltri ML. Schwann cell interactions during the development of the peripheral nervous system. Dev Neurobiol 2020; 81:464-489. [PMID: 32281247 DOI: 10.1002/dneu.22744] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 03/14/2020] [Accepted: 04/06/2020] [Indexed: 12/21/2022]
Abstract
Schwann cells play a critical role in the development of the peripheral nervous system (PNS), establishing important relationships both with the extracellular milieu and other cell types, particularly neurons. In this review, we discuss various Schwann cell interactions integral to the proper establishment, spatial arrangement, and function of the PNS. We include signals that cascade onto Schwann cells from axons and from the extracellular matrix, bidirectional signals that help to establish the axo-glial relationship and how Schwann cells in turn support the axon. Further, we speculate on how Schwann cell interactions with other components of the developing PNS ultimately promote the complete construction of the peripheral nerve.
Collapse
Affiliation(s)
- Emma R Wilson
- Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, NY, USA.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Gustavo Della-Flora Nunes
- Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, NY, USA.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Michael R Weaver
- Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, NY, USA.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - Luciana R Frick
- Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, NY, USA.,Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| | - M Laura Feltri
- Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, NY, USA.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA.,Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA
| |
Collapse
|
32
|
Previtali SC, Zambon AA. LAMA2 Neuropathies: Human Findings and Pathomechanisms From Mouse Models. Front Mol Neurosci 2020; 13:60. [PMID: 32390798 PMCID: PMC7190814 DOI: 10.3389/fnmol.2020.00060] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 03/26/2020] [Indexed: 12/18/2022] Open
Abstract
Merosin deficient Congenital Muscular Dystrophy (MDC1A), or LAMA2-related muscular dystrophy (LAMA2-RD), is a recessive disorder resulting from mutations in the LAMA2 gene, encoding for the alpha-2 chain of laminin-211. The disease is predominantly characterized by progressive muscular dystrophy affecting patient motor function and reducing life expectancy. However, LAMA2-RD also comprises a developmentally-associated dysmyelinating neuropathy that contributes to the disease progression, in addition to brain abnormalities; the latter often underappreciated. In this brief review, we present data supporting the impact of peripheral neuropathy in the LAMA2-RD phenotype, including both mouse models and human studies. We discuss the molecular mechanisms underlying nerve abnormalities and involved in the laminin-211 pathway, which affects axon sorting, ensheathing and myelination. We conclude with some final considerations of consequences on nerve regeneration and potential therapeutic strategies.
Collapse
Affiliation(s)
- Stefano Carlo Previtali
- Neuromuscular Repair Unit, Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Milan, Italy.,Department of Neurology, IRCCS Ospedale San Raffaele, Milan, Italy
| | | |
Collapse
|
33
|
Siems SB, Jahn O, Eichel MA, Kannaiyan N, Wu LMN, Sherman DL, Kusch K, Hesse D, Jung RB, Fledrich R, Sereda MW, Rossner MJ, Brophy PJ, Werner HB. Proteome profile of peripheral myelin in healthy mice and in a neuropathy model. eLife 2020; 9:e51406. [PMID: 32130108 PMCID: PMC7056269 DOI: 10.7554/elife.51406] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 02/19/2020] [Indexed: 02/07/2023] Open
Abstract
Proteome and transcriptome analyses aim at comprehending the molecular profiles of the brain, its cell-types and subcellular compartments including myelin. Despite the relevance of the peripheral nervous system for normal sensory and motor capabilities, analogous approaches to peripheral nerves and peripheral myelin have fallen behind evolving technical standards. Here we assess the peripheral myelin proteome by gel-free, label-free mass-spectrometry for deep quantitative coverage. Integration with RNA-Sequencing-based developmental mRNA-abundance profiles and neuropathy disease genes illustrates the utility of this resource. Notably, the periaxin-deficient mouse model of the neuropathy Charcot-Marie-Tooth 4F displays a highly pathological myelin proteome profile, exemplified by the discovery of reduced levels of the monocarboxylate transporter MCT1/SLC16A1 as a novel facet of the neuropathology. This work provides the most comprehensive proteome resource thus far to approach development, function and pathology of peripheral myelin, and a straightforward, accurate and sensitive workflow to address myelin diversity in health and disease.
Collapse
Affiliation(s)
- Sophie B Siems
- Department of Neurogenetics, Max Planck Institute of Experimental MedicineGöttingenGermany
| | - Olaf Jahn
- Proteomics Group, Max Planck Institute of Experimental MedicineGöttingenGermany
| | - Maria A Eichel
- Department of Neurogenetics, Max Planck Institute of Experimental MedicineGöttingenGermany
| | - Nirmal Kannaiyan
- Department of Psychiatry and Psychotherapy, University Hospital, LMU MunichMunichGermany
| | - Lai Man N Wu
- Centre for Discovery Brain Sciences, University of EdinburghEdinburghUnited Kingdom
| | - Diane L Sherman
- Centre for Discovery Brain Sciences, University of EdinburghEdinburghUnited Kingdom
| | - Kathrin Kusch
- Department of Neurogenetics, Max Planck Institute of Experimental MedicineGöttingenGermany
| | - Dörte Hesse
- Proteomics Group, Max Planck Institute of Experimental MedicineGöttingenGermany
| | - Ramona B Jung
- Department of Neurogenetics, Max Planck Institute of Experimental MedicineGöttingenGermany
| | - Robert Fledrich
- Department of Neurogenetics, Max Planck Institute of Experimental MedicineGöttingenGermany
- Institute of Anatomy, University of LeipzigLeipzigGermany
| | - Michael W Sereda
- Department of Neurogenetics, Max Planck Institute of Experimental MedicineGöttingenGermany
- Department of Clinical Neurophysiology, University Medical CenterGöttingenGermany
| | - Moritz J Rossner
- Department of Psychiatry and Psychotherapy, University Hospital, LMU MunichMunichGermany
| | - Peter J Brophy
- Centre for Discovery Brain Sciences, University of EdinburghEdinburghUnited Kingdom
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute of Experimental MedicineGöttingenGermany
| |
Collapse
|
34
|
Fledrich R, Kungl T, Nave KA, Stassart RM. Axo-glial interdependence in peripheral nerve development. Development 2019; 146:146/21/dev151704. [PMID: 31719044 DOI: 10.1242/dev.151704] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
During the development of the peripheral nervous system, axons and myelinating Schwann cells form a unique symbiotic unit, which is realized by a finely tuned network of molecular signals and reciprocal interactions. The importance of this complex interplay becomes evident after injury or in diseases in which aspects of axo-glial interaction are perturbed. This Review focuses on the specific interdependence of axons and Schwann cells in peripheral nerve development that enables axonal outgrowth, Schwann cell lineage progression, radial sorting and, finally, formation and maintenance of the myelin sheath.
Collapse
Affiliation(s)
- Robert Fledrich
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany .,Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Theresa Kungl
- Institute of Anatomy, Leipzig University, 04103 Leipzig, Germany.,Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany
| | - Ruth M Stassart
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, 37075 Göttingen, Germany .,Department of Neuropathology, University Clinic Leipzig, 04103 Leipzig, Germany
| |
Collapse
|
35
|
Han SH, Yun SH, Shin YK, Park HT, Park JI. Heat Shock Protein 90 is Required for cAMP-Induced Differentiation in Rat Primary Schwann Cells. Neurochem Res 2019; 44:2643-2657. [PMID: 31606837 DOI: 10.1007/s11064-019-02885-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 08/29/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022]
Abstract
Schwann cells (SCs) play an important role in producing myelin for rapid neurotransmission in the peripheral nervous system. Activation of the differentiation and myelination processes in SCs requires the expression of a series of transcriptional factors including Sox10, Oct6/Pou3f1, and Egr2/Krox20. However, functional interactions among several transcription factors are poorly defined and the important components of the regulatory network are still unknown. Until now, available evidence suggests that SCs require cAMP signaling to initiate the myelination program. Heat shock protein 90 (Hsp90) is known as a chaperone required to stabilize ErbB2 receptor. In recent years, it was reported that cAMP transactivated the ErbB2/ErbB3 signaling in SCs. However, the relationship between Hsp90 and cAMP-induced differentiation in SCs is undefined. Here we investigated the role of Hsp90 during cAMP-induced differentiation of SCs using Hsp90 inhibitor, geldanamycin and Hsp90 siRNA transfection. Our results showed that dibutyryl-cAMP (db-cAMP) treatment upregulated Hsp90 expression and led to nuclear translocation of Gab1/ERK, the downstream signaling pathway of the ErbB2 signaling mechanism in myelination. The expression of myelin-related genes and nuclear translocation of Gab1/ERK following db-cAMP treatment was inhibited by geldanamycin pretreatment and Hsp90 knockdown. These findings suggest that Hsp90 might play a role in cAMP-induced differentiation via stabilization of ErbB2 and nuclear translocation of Gab1/ERK in SCs.
Collapse
Affiliation(s)
- Sang-Heum Han
- Department of Biochemistry, Dong-A University College of Medicine, 32, Daesingongwon-ro, Seo-Gu, Busan, 49201, Republic of Korea.,Peripheral Neuropathy Research Center, Dong-A University, Busan, Republic of Korea
| | - Seong-Hoon Yun
- Department of Biochemistry, Dong-A University College of Medicine, 32, Daesingongwon-ro, Seo-Gu, Busan, 49201, Republic of Korea.,Peripheral Neuropathy Research Center, Dong-A University, Busan, Republic of Korea
| | - Yoon-Kyoung Shin
- Peripheral Neuropathy Research Center, Dong-A University, Busan, Republic of Korea
| | - Hwan-Tae Park
- Peripheral Neuropathy Research Center, Dong-A University, Busan, Republic of Korea. .,Department of Molecular Neuroscience, Dong-A University College of Medicine, 32, Daesingongwon-ro, Seo-Gu, Busan, 49201, Republic of Korea.
| | - Joo-In Park
- Department of Biochemistry, Dong-A University College of Medicine, 32, Daesingongwon-ro, Seo-Gu, Busan, 49201, Republic of Korea. .,Peripheral Neuropathy Research Center, Dong-A University, Busan, Republic of Korea.
| |
Collapse
|
36
|
Sophie B, Jacob H, Jordan VJS, Yungki P, Laura FM, Yannick P. YAP and TAZ Regulate Cc2d1b and Purβ in Schwann Cells. Front Mol Neurosci 2019; 12:177. [PMID: 31379499 PMCID: PMC6650784 DOI: 10.3389/fnmol.2019.00177] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Accepted: 07/04/2019] [Indexed: 12/31/2022] Open
Abstract
Schwann cells (SCs) are exquisitely sensitive to the elasticity of their environment and their differentiation and capacity to myelinate depend on the transduction of mechanical stimuli by YAP and TAZ. YAP/TAZ, in concert with other transcription factors, regulate several pathways including lipid and sterol biosynthesis as well as extracellular matrix receptor expressions such as integrins and G-proteins. Yet, the characterization of the signaling downstream YAP/TAZ in SCs is incomplete. Myelin sheath production by SC coincides with rapid up-regulation of numerous transcription factors. Here, we show that ablation of YAP/TAZ alters the expression of transcription regulators known to regulate SC myelin gene transcription and differentiation. Furthermore, we link YAP/TAZ to two DNA binding proteins, Cc2d1b and Purβ, which have no described roles in myelinating glial cells. We demonstrate that silencing of either Cc2d1b or Purβ limits the formation of myelin segments. These data provide a deeper insight into the myelin gene transcriptional network and the role of YAP/TAZ in myelinating glial cells.
Collapse
Affiliation(s)
- Belin Sophie
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
| | - Herron Jacob
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
| | - VerPlank J S Jordan
- Department of Cell Biology, Harvard Medical School, Boston, MA, United States
| | - Park Yungki
- Department of Biochemistry, Hunter James Kelly Research Institute, University at Buffalo, Buffalo, NY, United States
| | - Feltri M Laura
- Department of Biochemistry, Hunter James Kelly Research Institute, University at Buffalo, Buffalo, NY, United States.,Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Poitelon Yannick
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, United States
| |
Collapse
|
37
|
Ommer A, Figlia G, Pereira JA, Datwyler AL, Gerber J, DeGeer J, Lalli G, Suter U. Ral GTPases in Schwann cells promote radial axonal sorting in the peripheral nervous system. J Cell Biol 2019; 218:2350-2369. [PMID: 31201267 PMCID: PMC6605813 DOI: 10.1083/jcb.201811150] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 04/03/2019] [Accepted: 05/15/2019] [Indexed: 12/11/2022] Open
Abstract
Small GTPases of the Rho and Ras families are important regulators of Schwann cell biology. The Ras-like GTPases RalA and RalB act downstream of Ras in malignant peripheral nerve sheath tumors. However, the physiological role of Ral proteins in Schwann cell development is unknown. Using transgenic mice with ablation of one or both Ral genes, we report that Ral GTPases are crucial for axonal radial sorting. While lack of only one Ral GTPase was dispensable for early peripheral nerve development, ablation of both RalA and RalB resulted in persistent radial sorting defects, associated with hallmarks of deficits in Schwann cell process formation and maintenance. In agreement, ex vivo-cultured Ral-deficient Schwann cells were impaired in process extension and the formation of lamellipodia. Our data indicate further that RalA contributes to Schwann cell process extensions through the exocyst complex, a known effector of Ral GTPases, consistent with an exocyst-mediated function of Ral GTPases in Schwann cells.
Collapse
Affiliation(s)
- Andrea Ommer
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Gianluca Figlia
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Jorge A Pereira
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Anna Lena Datwyler
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Joanne Gerber
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Jonathan DeGeer
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Giovanna Lalli
- Wolfson Centre for Age-Related Diseases, King's College London, London, UK
| | - Ueli Suter
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
38
|
Belin S, Ornaghi F, Shackleford G, Wang J, Scapin C, Lopez-Anido C, Silvestri N, Robertson N, Williamson C, Ishii A, Taveggia C, Svaren J, Bansal R, Schwab MH, Nave K, Fratta P, D’Antonio M, Poitelon Y, Feltri ML, Wrabetz L. Neuregulin 1 type III improves peripheral nerve myelination in a mouse model of congenital hypomyelinating neuropathy. Hum Mol Genet 2019; 28:1260-1273. [PMID: 30535360 PMCID: PMC6452193 DOI: 10.1093/hmg/ddy420] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/06/2018] [Accepted: 12/02/2018] [Indexed: 12/13/2022] Open
Abstract
Myelin sheath thickness is precisely regulated and essential for rapid propagation of action potentials along myelinated axons. In the peripheral nervous system, extrinsic signals from the axonal protein neuregulin 1 (NRG1) type III regulate Schwann cell fate and myelination. Here we ask if modulating NRG1 type III levels in neurons would restore myelination in a model of congenital hypomyelinating neuropathy (CHN). Using a mouse model of CHN, we improved the myelination defects by early overexpression of NRG1 type III. Surprisingly, the improvement was independent from the upregulation of Egr2 or essential myelin genes. Rather, we observed the activation of MAPK/ERK and other myelin genes such as peripheral myelin protein 2 and oligodendrocyte myelin glycoprotein. We also confirmed that the permanent activation of MAPK/ERK in Schwann cells has detrimental effects on myelination. Our findings demonstrate that the modulation of axon-to-glial NRG1 type III signaling has beneficial effects and improves myelination defects during development in a model of CHN.
Collapse
Affiliation(s)
- Sophie Belin
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, NY, USA
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Francesca Ornaghi
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, NY, USA
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
- SR-TIGET, IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | - Ghjuvan’Ghjacumu Shackleford
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, NY, USA
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Jie Wang
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Cristina Scapin
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | | | - Nicholas Silvestri
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Neil Robertson
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Courtney Williamson
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, NY, USA
| | - Akihiro Ishii
- Department of Neuroscience, University of Connecticut Medical School, Farmington, CT, USA
| | - Carla Taveggia
- Division of Neuroscience, San Raffaele Scientific Institute, Milan, Italy
| | - John Svaren
- Waisman Center, University of Wisconsin–Madison, Madison, WI, USA
| | - Rashmi Bansal
- Department of Neuroscience, University of Connecticut Medical School, Farmington, CT, USA
| | - Markus H Schwab
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Klaus Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Pietro Fratta
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, Queen Square, London, UK
| | - Maurizio D’Antonio
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Yannick Poitelon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - M Laura Feltri
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, NY, USA
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| | - Lawrence Wrabetz
- Hunter James Kelly Research Institute, University at Buffalo, Buffalo, NY, USA
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
39
|
Hosseini M, Fattahi Z, Abedini SS, Hu H, Ropers H, Kalscheuer VM, Najmabadi H, Kahrizi K. GPR126
: A novel candidate gene implicated in autosomal recessive intellectual disability. Am J Med Genet A 2018; 179:13-19. [DOI: 10.1002/ajmg.a.40531] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 07/29/2018] [Accepted: 08/10/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Masoumeh Hosseini
- Genetics Research CenterUniversity of Social Welfare and Rehabilitation Sciences Tehran Iran
| | - Zohreh Fattahi
- Genetics Research CenterUniversity of Social Welfare and Rehabilitation Sciences Tehran Iran
| | | | - Hao Hu
- Department Human Molecular GeneticsMax‐Planck‐Institute for Molecular Genetics Berlin Germany
| | - Hans‐H. Ropers
- Department Human Molecular GeneticsMax‐Planck‐Institute for Molecular Genetics Berlin Germany
| | - Vera M. Kalscheuer
- Department Human Molecular GeneticsMax‐Planck‐Institute for Molecular Genetics Berlin Germany
| | - Hossein Najmabadi
- Genetics Research CenterUniversity of Social Welfare and Rehabilitation Sciences Tehran Iran
| | - Kimia Kahrizi
- Genetics Research CenterUniversity of Social Welfare and Rehabilitation Sciences Tehran Iran
| |
Collapse
|
40
|
Gilloteaux J, Subramanian K, Solomon N, Nicaise C. The leptin receptor mutation of the obese Zucker rat causes sciatic nerve demyelination with a centripetal pattern defect. Ultrastruct Pathol 2018; 42:377-408. [PMID: 30339059 DOI: 10.1080/01913123.2018.1522405] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Young male Zucker rats with a leptin receptor mutation are obese, have a non-insulin-dependent diabetes mellitus (NIDDM), and other endocrinopathies. Tibial branches of the sciatic nerve reveal a progressive demyelination that progresses out of the Schwann cells (SCs) where electron-contrast deposits are accumulated while the minor lines or intermembranous SC contacts display exaggerated spacings. Cajal bands contain diversely contrasted vesicles adjacent to the abaxonal myelin layer with blemishes; they appear dispatched centripetally out of many narrow electron densities, regularly spaced around the myelin annulus. These anomalies widen and yield into sectors across the stacked myelin layers. Throughout the worse degradations, the adaxonal membrane remains along the axonal neuroplasm. This peripheral neuropathy with irresponsive leptin cannot modulate hypothalamic-pituitary-adrenal axis and SC neurosteroids, thus exacerbates NIDDM condition. Additionally, the ultrastructure of the progressive myelin alterations may have unraveled a peculiar, centripetal mode of trafficking maintenance of the peripheral nervous system myelin, while some adhesive glycoproteins remain between myelin layers, somewhat hindering the axon mutilation. Heading title: Peripheral neuropathy and myelin.
Collapse
Affiliation(s)
- Jacques Gilloteaux
- a Department of Anatomical Sciences , St George's University School of Medicine, K.B. Taylor Global Scholar's Program at Northumbria University , Newcastle upon Tyne , UK.,b Unité de Recherche en Physiologie Moléculaire (URPhyM), Laboratoire de Neurodégénérescence et Régénération, Département de Médecine , Université de Namur , Namur , Belgium
| | - Kritika Subramanian
- a Department of Anatomical Sciences , St George's University School of Medicine, K.B. Taylor Global Scholar's Program at Northumbria University , Newcastle upon Tyne , UK.,c Department of Clinical and Epidemiological Virology , Rega Institute of Medical Research, Katholiele Universiteit Leuven , Leuven , Belgium
| | - Nadia Solomon
- a Department of Anatomical Sciences , St George's University School of Medicine, K.B. Taylor Global Scholar's Program at Northumbria University , Newcastle upon Tyne , UK
| | - Charles Nicaise
- b Unité de Recherche en Physiologie Moléculaire (URPhyM), Laboratoire de Neurodégénérescence et Régénération, Département de Médecine , Université de Namur , Namur , Belgium
| |
Collapse
|
41
|
Jiang M, Rao R, Wang J, Wang J, Xu L, Wu LM, Chan JR, Wang H, Lu QR. The TSC1-mTOR-PLK axis regulates the homeostatic switch from Schwann cell proliferation to myelination in a stage-specific manner. Glia 2018; 66:1947-1959. [PMID: 29722913 PMCID: PMC6185760 DOI: 10.1002/glia.23449] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 04/10/2018] [Accepted: 04/10/2018] [Indexed: 12/17/2022]
Abstract
Proper peripheral myelination depends upon the balance between Schwann cell proliferation and differentiation programs. The serine/threonine kinase mTOR integrates various environmental cues to serve as a central regulator of cell growth, metabolism, and function. We report here that tuberous sclerosis complex 1 (TSC1), a negative regulator of mTOR activity, establishes a stage-dependent program for Schwann cell lineage progression and myelination by controlling cell proliferation and myelin homeostasis. Tsc1 ablation in Schwann cell progenitors in mice resulted in activation of mTOR signaling, and caused over-proliferation of Schwann cells and blocked their differentiation, leading to hypomyelination. Transcriptome profiling analysis revealed that mTOR activation in Tsc1 mutants resulted in upregulation of a polo-like kinase (PLK)-dependent pathway and cell cycle regulators. Attenuation of mTOR or pharmacological inhibition of polo-like kinases partially rescued hypomyelination caused by Tsc1 loss in the developing peripheral nerves. In contrast, deletion of Tsc1 in mature Schwann cells led to redundant and overgrown myelin sheaths in adult mice. Together, our findings indicate stage-specific functions for the TSC1-mTOR-PLK signaling axis in controlling the transition from proliferation to differentiation and myelin homeostasis during Schwann cell development.
Collapse
Affiliation(s)
- Minqing Jiang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
- The Institute of Cognitive Neuroscience, East China Normal University, Shanghai, China
| | - Rohit Rao
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jincheng Wang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jiajia Wang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Lingli Xu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Lai Man Wu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jonah R. Chan
- Department of Neurology and Programs in Biomedical and Neurosciences, University of California, San Francisco, CA 94158
| | - Huimin Wang
- The Institute of Cognitive Neuroscience, East China Normal University, Shanghai, China
| | - Q. Richard Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
42
|
Tricaud N. Myelinating Schwann Cell Polarity and Mechanically-Driven Myelin Sheath Elongation. Front Cell Neurosci 2018; 11:414. [PMID: 29354031 PMCID: PMC5760505 DOI: 10.3389/fncel.2017.00414] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Accepted: 12/11/2017] [Indexed: 11/13/2022] Open
Abstract
Myelin sheath geometry, encompassing myelin sheath thickness relative to internodal length, is critical to optimize nerve conduction velocity and these parameters are carefully adjusted by the myelinating cells in mammals. In the central nervous system these adjustments could regulate neuronal activities while in the peripheral nervous system they lead to the optimization and the reliability of the nerve conduction velocity. However, the physiological and cellular mechanisms that underlie myelin sheath geometry regulation are not yet fully elucidated. In peripheral nerves the myelinating Schwann cell uses several molecular mechanisms to reach and maintain the correct myelin sheath geometry, such that myelin sheath thickness and internodal length are regulated independently. One of these mechanisms is the epithelial-like cell polarization process that occurs during the early phases of the myelin biogenesis. Epithelial cell polarization factors are known to control cell size and morphology in invertebrates and mammals making these processes critical in the organogenesis. Correlative data indicate that internodal length is regulated by postnatal body growth that elongates peripheral nerves in mammals. In addition, the mechanical stretching of peripheral nerves in adult animals shows that myelin sheath length can be increased by mechanical cues. Recent results describe the important role of YAP/TAZ co-transcription factors during Schwann cell myelination and their functions have linked to the mechanotransduction through the HIPPO pathway and the epithelial polarity factor Crb3. In this review the molecular mechanisms that govern mechanically-driven myelin sheath elongation and how a Schwann cell can modulate internodal myelin sheath length, independent of internodal thickness, will be discussed regarding these recent data. In addition, the potential relevance of these mechanosensitive mechanisms in peripheral pathologies will be highlighted.
Collapse
Affiliation(s)
- Nicolas Tricaud
- Institut National de la Santé et de la Recherche Médicale, Institut des Neurosciences de Montpellier, Université de Montpellier, Montpellier, France
| |
Collapse
|
43
|
Figlia G, Gerber D, Suter U. Myelination and mTOR. Glia 2017; 66:693-707. [PMID: 29210103 PMCID: PMC5836902 DOI: 10.1002/glia.23273] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/08/2017] [Accepted: 11/17/2017] [Indexed: 02/06/2023]
Abstract
Myelinating cells surround axons to accelerate the propagation of action potentials, to support axonal health, and to refine neural circuits. Myelination is metabolically demanding and, consistent with this notion, mTORC1—a signaling hub coordinating cell metabolism—has been implicated as a key signal for myelination. Here, we will discuss metabolic aspects of myelination, illustrate the main metabolic processes regulated by mTORC1, and review advances on the role of mTORC1 in myelination of the central nervous system and the peripheral nervous system. Recent progress has revealed a complex role of mTORC1 in myelinating cells that includes, besides positive regulation of myelin growth, additional critical functions in the stages preceding active myelination. Based on the available evidence, we will also highlight potential nonoverlapping roles between mTORC1 and its known main upstream pathways PI3K‐Akt, Mek‐Erk1/2, and AMPK in myelinating cells. Finally, we will discuss signals that are already known or hypothesized to be responsible for the regulation of mTORC1 activity in myelinating cells. Myelination is metabolically demanding. The metabolic regulator mTORC1 controls differentiation of myelinating cells and promotes myelin
growth. mTORC1‐independent targets of the PI3K‐Akt and Mek‐Erk1/2 pathways may also be significant in myelination.
Collapse
Affiliation(s)
- Gianluca Figlia
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology, ETH Zürich, Zürich, CH 8093, Switzerland
| | - Daniel Gerber
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology, ETH Zürich, Zürich, CH 8093, Switzerland
| | - Ueli Suter
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology, ETH Zürich, Zürich, CH 8093, Switzerland
| |
Collapse
|
44
|
Figlia G, Norrmén C, Pereira JA, Gerber D, Suter U. Dual function of the PI3K-Akt-mTORC1 axis in myelination of the peripheral nervous system. eLife 2017; 6:e29241. [PMID: 28880149 PMCID: PMC5589416 DOI: 10.7554/elife.29241] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 07/31/2017] [Indexed: 01/24/2023] Open
Abstract
Myelination is a biosynthetically demanding process in which mTORC1, the gatekeeper of anabolism, occupies a privileged regulatory position. We have shown previously that loss of mTORC1 function in Schwann cells (SCs) hampers myelination. Here, we genetically disrupted key inhibitory components upstream of mTORC1, TSC1 or PTEN, in mouse SC development, adult homeostasis, and nerve injury. Surprisingly, the resulting mTORC1 hyperactivity led to markedly delayed onset of both developmental myelination and remyelination after injury. However, if mTORC1 was hyperactivated after myelination onset, radial hypermyelination was observed. At early developmental stages, physiologically high PI3K-Akt-mTORC1 signaling suppresses expression of Krox20 (Egr2), the master regulator of PNS myelination. This effect is mediated by S6K and contributes to control mechanisms that keep SCs in a not-fully differentiated state to ensure proper timing of myelination initiation. An ensuing decline in mTORC1 activity is crucial to allow myelination to start, while remaining mTORC1 activity drives myelin growth.
Collapse
Affiliation(s)
- Gianluca Figlia
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| | - Camilla Norrmén
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| | - Jorge A Pereira
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| | - Daniel Gerber
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| | - Ueli Suter
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| |
Collapse
|