1
|
Kasi PB, Opoku H, Novikova LN, Wiberg M, Kingham PJ, Wang J, Novikov LN. Quercetin-derived carbon dots promote proliferation and migration of Schwann cells and enhance neurite outgrowth. Colloids Surf B Biointerfaces 2025; 251:114609. [PMID: 40073625 DOI: 10.1016/j.colsurfb.2025.114609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/02/2024] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
Quercetin, a flavonoid known for its antioxidant properties, has recently garnered attention as a potential neuroprotective agent for treatment of the injured nervous system. The repair of peripheral nerve injuries hinges on the proliferation and migration of Schwann cells, which play a crucial role in supporting axonal growth and myelination. In this study we synthesized Quercetin-derived carbon dots (QCDs) and investigated their effects on cultured Schwann cells and the NG108-15 cell line. QCDs was obtained by solvothermal synthesis and characterized via UV-vis absorption spectroscopy, transmission electron microscopy, Fourier transform infrared spectroscopy, and X-ray diffraction analysis. The particles demonstrated significant dose-dependent free radical scavenging activity in DPPH and ABTS radical scavenging assays, supported in vitro proliferation and migration of Schwann cells, expression of neurotrophic and angiogenic growth factors, and stimulated neurite outgrowth from NG108-15 cells. Thus, QCDs could serve as a potential novel treatment strategy to promote regeneration in the injured peripheral nervous system.
Collapse
Affiliation(s)
- Phanindra Babu Kasi
- Department of Medical and Translational Biology, Umeå University, Umeå SE-901 87, Sweden
| | - Henry Opoku
- The Organic Photonics and Electronics Group, Department of Physics, Umeå University, Umeå SE-901 87, Sweden
| | - Liudmila N Novikova
- Department of Medical and Translational Biology, Umeå University, Umeå SE-901 87, Sweden
| | - Mikael Wiberg
- Department of Medical and Translational Biology, Umeå University, Umeå SE-901 87, Sweden; Department of Diagnostics and Intervention, Section of Hand and Plastic Surgery, Umeå University, Umeå SE-901 87, Sweden
| | - Paul J Kingham
- Department of Medical and Translational Biology, Umeå University, Umeå SE-901 87, Sweden
| | - Jia Wang
- The Organic Photonics and Electronics Group, Department of Physics, Umeå University, Umeå SE-901 87, Sweden.
| | - Lev N Novikov
- Department of Medical and Translational Biology, Umeå University, Umeå SE-901 87, Sweden.
| |
Collapse
|
2
|
Jorge KTDOS, Braga MP, Cazzaniga RA, Santos CNO, Teixeira MM, Gomes KB, de Jesus AMR, Soriani FM. The role of neurotrophin polymorphisms and susceptibility to neural damage in leprosy. Int J Infect Dis 2024; 142:106946. [PMID: 38278287 DOI: 10.1016/j.ijid.2024.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 12/20/2023] [Accepted: 01/21/2024] [Indexed: 01/28/2024] Open
Abstract
OBJECTIVES Mycobacterium leprae is able to infect Schwann cells leading to neural damage. Neurotrophins are involved in nervous system plasticity and impact neural integrity during diseases. Investigate the association between single nucleotide polymorphisms in neurotrophin genes and leprosy phenotypes, especially neural damage. DESIGN We selected single nucleotide polymorphisms in neurotrophins or their receptors genes associated with neural disorders: rs6265 and rs11030099 of brain-derived neurotrophic factor (BDNF), rs6330 of BDNF, rs6332 in NT3 and rs2072446 of P75NTR. The association of genetic frequencies with leprosy phenotypes was investigated in a case-control study. RESULTS An association of the BDNF single nucleotide polymorphism rs11030099 with the number of affected nerves was demonstrated. The "AA+AC" genotypes were demonstrated to be protective against nerve impairment. However, this variation does not affect BDNF serum levels. BDNF is an important factor for myelination of Schwann cells and polymorphisms in this gene can be associated with leprosy outcome. Moreover, rs11030099 is located in the binding region for micro-RNA (miRNA) 26a that could be involved in control of BDNF expression. We demonstrated different expression levels of this miRNA in polar forms of leprosy. CONCLUSION Our findings demonstrate for the first time an association between the polymorphism rs11030099 in the BDNF gene and neural commitment in leprosy and may indicate a possible role of miRNA-26a acting synergistically to these genetic variants in neural damage development.
Collapse
Affiliation(s)
| | - Marina Pimenta Braga
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | | | - Mauro Martins Teixeira
- Laboratory of Immunopharmacology, Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Karina Braga Gomes
- Department of Clinical and Toxicological Analyzes - Faculty of Pharmacy, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | | | - Frederico Marianetti Soriani
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
3
|
Berardo A, Bacaglio CR, Báez BB, Sambuelli R, Sheikh KA, Lopez PHH. Blockade of Rho-associated kinase prevents inhibition of axon regeneration of peripheral nerves induced by anti-ganglioside antibodies. Neural Regen Res 2024; 19:895-899. [PMID: 37843226 PMCID: PMC10664126 DOI: 10.4103/1673-5374.382258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 06/08/2023] [Accepted: 07/19/2023] [Indexed: 10/17/2023] Open
Abstract
Anti-ganglioside antibodies are associated with delayed/poor clinical recovery in Guillain-Barrè syndrome, mostly related to halted axon regeneration. Cross-linking of cell surface gangliosides by anti-ganglioside antibodies triggers inhibition of nerve repair in in vitro and in vivo paradigms of axon regeneration. These effects involve the activation of the small GTPase RhoA/ROCK signaling pathways, which negatively modulate growth cone cytoskeleton, similarly to well stablished inhibitors of axon regeneration described so far. The aim of this work was to perform a proof of concept study to demonstrate the effectiveness of Y-27632, a selective pharmacological inhibitor of ROCK, in a mouse model of axon regeneration of peripheral nerves, where the passive immunization with a monoclonal antibody targeting gangliosides GD1a and GT1b was previously reported to exert a potent inhibitory effect on regeneration of both myelinated and unmyelinated fibers. Our results demonstrate a differential sensitivity of myelinated and unmyelinated axons to the pro-regenerative effect of Y-27632. Treatment with a total dosage of 9 mg/kg of Y-27632 resulted in a complete prevention of anti-GD1a/GT1b monoclonal antibody-mediated inhibition of axon regeneration of unmyelinated fibers to skin and the functional recovery of mechanical cutaneous sensitivity. In contrast, the same dose showed toxic effects on the regeneration of myelinated fibers. Interestingly, scale down of the dosage of Y-27632 to 5 mg/kg resulted in a significant although not complete recovery of regenerated myelinated axons exposed to anti-GD1a/GT1b monoclonal antibody in the absence of toxicity in animals exposed to only Y-27632. Overall, these findings confirm the in vivo participation of RhoA/ROCK signaling pathways in the molecular mechanisms associated with the inhibition of axon regeneration induced by anti-GD1a/GT1b monoclonal antibody. Our findings open the possibility of therapeutic pharmacological intervention targeting RhoA/Rock pathway in immune neuropathies associated with the presence of anti-ganglioside antibodies and delayed or incomplete clinical recovery after injury in the peripheral nervous system.
Collapse
Affiliation(s)
- Andrés Berardo
- Laboratorio de Neurobiología, Instituto de Investigación Médica Mercedes y Martin Ferreyra, (INIMEC)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Cristian R. Bacaglio
- Laboratorio de Neurobiología, Instituto de Investigación Médica Mercedes y Martin Ferreyra, (INIMEC)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Química Biológica-Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Cs. Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Bárbara B. Báez
- Laboratorio de Neurobiología, Instituto de Investigación Médica Mercedes y Martin Ferreyra, (INIMEC)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Química Biológica-Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Cs. Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Rubén Sambuelli
- Servicio de Anatomía Patológica, Clínica Universitaria Reina Fabiola, Universidad Católica de Córdoba, Córdoba, Argentina
| | - Kazim A. Sheikh
- Department of Neurology, University of Texas Medical School at Houston, Houston, TX, USA
| | - Pablo H. H. Lopez
- Laboratorio de Neurobiología, Instituto de Investigación Médica Mercedes y Martin Ferreyra, (INIMEC)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET)-Universidad Nacional de Córdoba, Córdoba, Argentina
- Departamento de Química Biológica-Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC)- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Facultad de Cs. Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
| |
Collapse
|
4
|
Gérard A, Owen RS, Dujon AM, Roche B, Hamede R, Thomas F, Ujvari B, Siddle HV. In vitro competition between two transmissible cancers and potential implications for their host, the Tasmanian devil. Evol Appl 2024; 17:e13670. [PMID: 38468711 PMCID: PMC10925828 DOI: 10.1111/eva.13670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 02/02/2024] [Accepted: 02/09/2024] [Indexed: 03/13/2024] Open
Abstract
Since the emergence of a transmissible cancer, devil facial tumour disease (DFT1), in the 1980s, wild Tasmanian devil populations have been in decline. In 2016, a second, independently evolved transmissible cancer (DFT2) was discovered raising concerns for survival of the host species. Here, we applied experimental and modelling frameworks to examine competition dynamics between the two transmissible cancers in vitro. Using representative cell lines for DFT1 and DFT2, we have found that in monoculture, DFT2 grows twice as fast as DFT1 but reaches lower maximum cell densities. Using co-cultures, we demonstrate that DFT2 outcompetes DFT1: the number of DFT1 cells decreasing over time, never reaching exponential growth. This phenomenon could not be replicated when cells were grown separated by a semi-permeable membrane, consistent with exertion of mechanical stress on DFT1 cells by DFT2. A logistic model and a Lotka-Volterra competition model were used to interrogate monoculture and co-culture growth curves, respectively, suggesting DFT2 is a better competitor than DFT1, but also showing that competition outcomes might depend on the initial number of cells, at least in the laboratory. We provide theories how the in vitro results could be translated to observations in the wild and propose that these results may indicate that although DFT2 is currently in a smaller geographic area than DFT1, it could have the potential to outcompete DFT1. Furthermore, we provide a framework for improving the parameterization of epidemiological models applied to these cancer lineages, which will inform future disease management.
Collapse
Affiliation(s)
- Anne‐Lise Gérard
- School of Life and Environmental SciencesDeakin UniversityWaurn PondsVictoriaAustralia
- CREEC/MIVEGEC, CNRS, IRDUniversité de MontpellierMontpellierFrance
| | - Rachel S. Owen
- School of Biological SciencesUniversity of SouthamptonSouthamptonUK
- Institute for Life SciencesUniversity of SouthamptonSouthamptonUK
- The Roslin InstituteThe University of EdinburghEdinburghUK
| | - Antoine M. Dujon
- School of Life and Environmental SciencesDeakin UniversityWaurn PondsVictoriaAustralia
| | - Benjamin Roche
- CREEC/MIVEGEC, CNRS, IRDUniversité de MontpellierMontpellierFrance
| | - Rodrigo Hamede
- School of Natural SciencesUniversity of TasmaniaHobartTasmaniaAustralia
| | - Frédéric Thomas
- CREEC/MIVEGEC, CNRS, IRDUniversité de MontpellierMontpellierFrance
| | - Beata Ujvari
- School of Life and Environmental SciencesDeakin UniversityWaurn PondsVictoriaAustralia
| | - Hannah V. Siddle
- School of Biological SciencesUniversity of SouthamptonSouthamptonUK
- Institute for Life SciencesUniversity of SouthamptonSouthamptonUK
| |
Collapse
|
5
|
Zeng X, Wei QS, Ye JC, Rao JH, Zheng MG, Ma YH, Peng LZ, Ding Y, Lai BQ, Li G, Cheng SX, Ling EA, Han I, Zeng YS. A biocompatible gelatin sponge scaffold confers robust tissue remodeling after spinal cord injury in a non-human primate model. Biomaterials 2023; 299:122161. [PMID: 37236138 DOI: 10.1016/j.biomaterials.2023.122161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 04/09/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023]
Abstract
We previously constructed a three-dimensional gelatin sponge (3D-GS) scaffold as a delivery vehicle for therapeutic cells and trophic factors in the treatment of spinal cord injury (SCI), and this study aimed to assess the biosafety and efficacy of the scaffold in a non-human primate SCI model. However, because it has only been tested in rodent and canine models, the biosafety and efficacy of the scaffold should ideally be assessed in a non-human primate SCI model before its use in the clinic. No adverse reactions were observed over 8 weeks following 3D-GS scaffold implantation into in a Macaca fascicularis with hemisected SCI. Scaffold implantation also did not add to neuroinflammatory or astroglial responses already present at the injured site, suggesting good biocompatibility. Notably, there was a significant reduction in α-smooth muscle actin (αSMA)-positive cells at the injury/implantation interface, leading to alleviation of fibrotic compression of the residual spinal cord tissue. The regenerating tissue in the scaffold showed numerous cells migrating into the implant secreting abundant extracellular matrix, resulting in a pro-regenerative microenvironment. Consequently, nerve fiber regeneration, myelination, vascularization, neurogenesis, and electrophysiological improvements were achieved. These results indicated that the 3D-GS scaffold had good histocompatibility and effectiveness in the structural repair of injured spinal cord tissue in a non-human primate and is suitable for use in the treatment of patients with SCI.
Collapse
Affiliation(s)
- Xiang Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China; Lab of Stem Cell Biology and Innovative Research of Chinese Medicine; National Institute for Stem Cell Clinical Research, Guangdong Provincial Hospital of Chinese Medicine/The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510120, China; Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510080, China
| | - Qing-Shuai Wei
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ji-Chao Ye
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China; Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jun-Hua Rao
- Guangdong Key Laboratory of Animal Conservation and Resource Utilization, Guangdong Public Laboratory of Wild Animal Conservation and Utilization, Institute of Zoology, Guangdong Academy of Sciences, Guangzhou, 510260, China
| | - Mei-Guang Zheng
- Department of Neurosurgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China
| | - Yuan-Huan Ma
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Li-Zhi Peng
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China
| | - Ying Ding
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Bi-Qin Lai
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Ge Li
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shi-Xiang Cheng
- Healthina Academy of Biomedicine, Tianjin Economic-Technological Development Area (HAB-TEDA) and XinCheng Hospital of Tianjin University, Tianjin, 301999, China
| | - Eng-Ang Ling
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore
| | - Inbo Han
- Department of Neurosurgery, CHA University, CHA Bundang Medical Center, Seongnam-si, Gyeonggi-do, 13496, Republic of Korea
| | - Yuan-Shan Zeng
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, 510080, China; Department of Histology and Embryology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China; Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China; Institute of Spinal Cord Injury, Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
6
|
Kalotay E, Klugmann M, Housley GD, Fröhlich D. Dominant aminoacyl-tRNA synthetase disorders: lessons learned from in vivo disease models. Front Neurosci 2023; 17:1182845. [PMID: 37274211 PMCID: PMC10234151 DOI: 10.3389/fnins.2023.1182845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/05/2023] [Indexed: 06/06/2023] Open
Abstract
Aminoacyl-tRNA synthetases (ARSs) play an essential role in protein synthesis, being responsible for ligating tRNA molecules to their corresponding amino acids in a reaction known as 'tRNA aminoacylation'. Separate ARSs carry out the aminoacylation reaction in the cytosol and in mitochondria, and mutations in almost all ARS genes cause pathophysiology most evident in the nervous system. Dominant mutations in multiple cytosolic ARSs have been linked to forms of peripheral neuropathy including Charcot-Marie-Tooth disease, distal hereditary motor neuropathy, and spinal muscular atrophy. This review provides an overview of approaches that have been employed to model each of these diseases in vivo, followed by a discussion of the existing animal models of dominant ARS disorders and key mechanistic insights that they have provided. In summary, ARS disease models have demonstrated that loss of canonical ARS function alone cannot fully account for the observed disease phenotypes, and that pathogenic ARS variants cause developmental defects within the peripheral nervous system, despite a typically later onset of disease in humans. In addition, aberrant interactions between mutant ARSs and other proteins have been shown to contribute to the disease phenotypes. These findings provide a strong foundation for future research into this group of diseases, providing methodological guidance for studies on ARS disorders that currently lack in vivo models, as well as identifying candidate therapeutic targets.
Collapse
Affiliation(s)
- Elizabeth Kalotay
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Matthias Klugmann
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
- Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Gary D. Housley
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Dominik Fröhlich
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
7
|
Zhang J, Li Y. Propofol-Induced Developmental Neurotoxicity: From Mechanisms to Therapeutic Strategies. ACS Chem Neurosci 2023; 14:1017-1032. [PMID: 36854650 DOI: 10.1021/acschemneuro.2c00755] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2023] Open
Abstract
Propofol is the most commonly used intravenous general anesthetic in clinical anesthesia, and it is also widely used in general anesthesia for pregnant women and infants. Some clinical and preclinical studies have found that propofol causes damage to the immature nervous system, which may lead to neurodevelopmental disorders and cognitive dysfunction in infants and children. However, its potential molecular mechanism has not been fully elucidated. Recent in vivo and in vitro studies have found that some exogenous drugs and interventions can effectively alleviate propofol-induced neurotoxicity. In this review, we focus on the relevant preclinical studies and summarize the latest findings on the potential mechanisms and therapeutic strategies of propofol-induced developmental neurotoxicity.
Collapse
Affiliation(s)
- Jing Zhang
- Department of Anesthesiology, Affiliated Hospital of Qingdao University, Qingdao 266000, China.,Department of Medicine, Qingdao University, Qingdao 266000, China
| | - Yu Li
- Department of Anesthesiology, Affiliated Hospital of Qingdao University, Qingdao 266000, China
| |
Collapse
|
8
|
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
|
9
|
Zanin JP, Friedman WJ. p75NTR prevents the onset of cerebellar granule cell migration via RhoA activation. eLife 2022; 11:e79934. [PMID: 36040414 PMCID: PMC9427104 DOI: 10.7554/elife.79934] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 08/07/2022] [Indexed: 12/20/2022] Open
Abstract
Neuronal migration is one of the fundamental processes during brain development. Several neurodevelopmental disorders can be traced back to dysregulated migration. Although substantial efforts have been placed in identifying molecular signals that stimulate migration, little is known about potential mechanisms that restrict migration. These restrictive mechanisms are essential for proper development since it helps coordinate the timing for each neuronal population to arrive and establish proper connections. Moreover, preventing migration away from a proliferative niche is necessary in maintaining a pool of proliferating cells until the proper number of neuronal progenitors is attained. Here, using mice and rats, we identify an anti-migratory role for the p75 neurotrophin receptor (p75NTR) in cerebellar development. Our results show that granule cell precursors (GCPs) robustly express p75NTR in the external granule layer (EGL) when they are proliferating during postnatal development, however, they do not express p75NTR when they migrate either from the rhombic lip during embryonic development or from the EGL during postnatal development. We show that p75NTR prevented GCP migration by maintaining elevated levels of active RhoA. The expression of p75NTR was sufficient to prevent the migration of the granule cells even in the presence of BDNF (brain-derived neurotrophic factor), a well-established chemotactic signal for this cell population. Our findings suggest that the expression of p75NTR might be a critical signal that stops and maintains the GCPs in the proliferative niche of the EGL, by promoting the clonal expansion of cerebellar granule neurons.
Collapse
Affiliation(s)
- Juan P Zanin
- Department of Biological Sciences, Rutgers UniversityNewarkUnited States
| | - Wilma J Friedman
- Department of Biological Sciences, Rutgers UniversityNewarkUnited States
| |
Collapse
|
10
|
Gong C, Hu B, Chen H, Zhu J, Nie J, Hua L, Chen L, Fang Y, Hang C, Lu Y. β2-adrenergic receptor drives the metastasis and invasion of pancreatic ductal adenocarcinoma through activating Cdc42 signaling pathway. J Mol Histol 2022; 53:645-655. [PMID: 35717490 DOI: 10.1007/s10735-022-10076-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2021] [Accepted: 05/23/2022] [Indexed: 10/18/2022]
Abstract
Recent investigations indicate that β2-adrenergic receptor (β2-AR) signaling may facilitate the progression of various tumors, whose underlying mechanisms remain largely elusive. In the present study, we showed that β2-AR recruited Cdc42 in response to isoproterenol (ISO, a β-AR selective agonist) exposure in pancreatic ductal adenocarcinoma (PDAC) cells. The association of β2-AR and Cdc42 promoted the activation of Cdc42, as revealed by increased levels of Cdc42-GTP, and co-incubation with β2-AR antagonist abrogated ISO-induced activation of Cdc42. β2-AR-mediated Cdc42 activation further led to the phosphorylation of downstream PAK1, LIMK1 and Merlin. Furthermore, we showed that the activation of β2-AR/Cdc42 signaling facilitated the migration and invasion of PDAC cells. In addition, β2-AR and Cdc42 were overexpressed in PDAC specimens, compared with adjacent non-tumor tissues. High expression of β2-AR and Cdc42 were correlated with lymph node metastasis and TNM stage in PDAC patients. Finally, we showed that overexpression of β2-AR and Cdc42 were indicative of unfavorable prognosis in PDAC patients. Taken together, our findings suggested that β2-AR might facilitate Cdc42 signaling to drive the migration and invasion of PDAC cells, consequently resulting in the metastasis and dismal prognosis of PDAC. These studies highlight targeting β2-AR/Cdc42 signaling as a therapeutic strategy against PDAC.
Collapse
Affiliation(s)
- Chen Gong
- Department of Gastroenterology, Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, Taicang, 215400, Jiangsu Province, China
| | - Baoying Hu
- Department of Immunology, Nantong University School of Medicine, Nantong, 226001, Jiangsu Province, China
| | - Haifeng Chen
- Department of Gastroenterology, Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, Taicang, 215400, Jiangsu Province, China
| | - Jianxin Zhu
- Department of Gastroenterology, Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, Taicang, 215400, Jiangsu Province, China
| | - Jinshan Nie
- Department of Gastroenterology, Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, Taicang, 215400, Jiangsu Province, China
| | - Lu Hua
- Department of Oncology, Taizhou People's Hospital, Taizhou, 225300, Jiangsu Province, China
| | - Long Chen
- Department of Gastroenterology, Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, Taicang, 215400, Jiangsu Province, China
| | - Yanfei Fang
- Department of Gastroenterology, Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, Taicang, 215400, Jiangsu Province, China
| | - Cheng Hang
- Department of Gastroenterology, Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, Taicang, 215400, Jiangsu Province, China.
| | - Ye Lu
- Department of Hematology and Oncology, Taicang Affiliated Hospital of Soochow University, The First People's Hospital of Taicang, Taicang, 215400, Jiangsu Province, China.
| |
Collapse
|
11
|
Abstract
BACKGROUND Acellular nerve allograft (ANA) occupies an increasingly prominent role in the treatment of peripheral nerve reconstruction. There is demonstrable efficacy; however, some grafts fail to support axonal regrowth and the reasons for this are unclear. This study examines the ANA experience in a specialized peripheral nerve surgery department to discuss the clinical and histological findings in failed cases. METHOD Failed ANA grafts were identified from a prospective database using Medical Research Council Classification (MRCC) S3 and M3 as thresholds for success. Cases in which ANA grafting was indicated for nerve related pain and dysesthesia but where no subjective improvement in symptoms occurred were also included. Patients requiring revision surgery after ANA grafting were also considered failures. Cases were then examined in conjunction with a literature review to identify possible mechanisms of failure, including detailed histological analysis in 2 cases. RESULTS Eight failed procedures were identified from a database of 99 separate allograft records on 74 patients. This included procedures for 2 tibial nerves, 2 superficial radial nerves, 2 median nerves, 1 digital nerve and a lateral cord brachial plexus injury (male/female, 5:3; age range, 24-54 years). Allograft length range 25 to 120 mm. One postoperative infection was identified. Histological findings in 2 cases included adequate vascularization of allograft material without subsequent axonal regeneration, a reduction of large myelinated fibers proximal to a tibial nerve allograft in the setting of a chronic injury, and a preference for small rather than large fiber regeneration. CONCLUSIONS This article reports instances of ANA graft failure in a variety of contexts, for which the primary reasons for failure remain unclear. The etiology is likely to be multifactorial with both patient, graft and surgeon factors contributing to failure. Further clinical and histological analysis of ANA failures will improve our understanding of the mechanisms of graft failure.
Collapse
Affiliation(s)
- Calum Thomson
- From the Department of Peripheral Nerve Surgery, Queen Elizabeth Hospital
| | | | - Ute Pohl
- Department of Cellular Pathology
| | - Dominic M Power
- The Birmingham Peripheral Nerve Injury Service, Queen Elizabeth Hospital, Birmingham, United Kingdom
| |
Collapse
|
12
|
Ulrichsen M, Gonçalves NP, Mohseni S, Hjæresen S, Lisle TL, Molgaard S, Madsen NK, Andersen OM, Svenningsen ÅF, Glerup S, Nykjær A, Vægter CB. Sortilin Modulates Schwann Cell Signaling and Remak Bundle Regeneration Following Nerve Injury. Front Cell Neurosci 2022; 16:856734. [PMID: 35634462 PMCID: PMC9130554 DOI: 10.3389/fncel.2022.856734] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Peripheral nerve regeneration relies on the ability of Schwann cells to support the regrowth of damaged axons. Schwann cells re-differentiate when reestablishing contact with the sprouting axons, with large fibers becoming remyelinated and small nociceptive fibers ensheathed and collected into Remak bundles. We have previously described how the receptor sortilin facilitates neurotrophin signaling in peripheral neurons via regulated trafficking of Trk receptors. This study aims to characterize the effects of sortilin deletion on nerve regeneration following sciatic crush injury. We found that Sort1–/– mice displayed functional motor recovery like that of WT mice, with no detectable differences in relation to nerve conduction velocities and morphological aspects of myelinated fibers. In contrast, we found abnormal ensheathment of regenerated C-fibers in injured Sort1–/– mice, demonstrating a role of sortilin for Remak bundle formation following injury. Further studies on Schwann cell signaling pathways showed a significant reduction of MAPK/ERK, RSK, and CREB phosphorylation in Sort1–/– Schwann cells after stimulation with neurotrophin-3 (NT-3), while Schwann cell migration and myelination remained unaffected. In conclusion, our results demonstrate that loss of sortilin blunts NT-3 signaling in Schwann cells which might contribute to the impaired Remak bundle regeneration after sciatic nerve injury.
Collapse
Affiliation(s)
- Maj Ulrichsen
- Danish Research Institute of Translational Neuroscience – DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Nádia P. Gonçalves
- Danish Research Institute of Translational Neuroscience – DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Simin Mohseni
- Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Simone Hjæresen
- Neurobiological Research, Faculty of Health Sciences, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Thomas L. Lisle
- Danish Research Institute of Translational Neuroscience – DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Simon Molgaard
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Niels K. Madsen
- Danish Research Institute of Translational Neuroscience – DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Olav M. Andersen
- Danish Research Institute of Translational Neuroscience – DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Åsa F. Svenningsen
- Neurobiological Research, Faculty of Health Sciences, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Simon Glerup
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Anders Nykjær
- Danish Research Institute of Translational Neuroscience – DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark
- Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
- Center of Excellence PROMEMO, Aarhus University, Aarhus, Denmark
| | - Christian B. Vægter
- Danish Research Institute of Translational Neuroscience – DANDRITE, Nordic EMBL Partnership for Molecular Medicine, Department of Biomedicine, Aarhus University, Aarhus, Denmark
- *Correspondence: Christian B. Vægter,
| |
Collapse
|
13
|
Transcriptome Analysis of Schwann Cells at Various Stages of Myelination Implicates Chromatin Regulator Sin3A in Control of Myelination Identity. Neurosci Bull 2022; 38:720-740. [DOI: 10.1007/s12264-022-00850-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 12/13/2021] [Indexed: 10/18/2022] Open
|
14
|
Analysis of Signal Transduction Pathways Downstream M2 Receptor Activation: Effects on Schwann Cell Migration and Morphology. Life (Basel) 2022; 12:life12020211. [PMID: 35207498 PMCID: PMC8875146 DOI: 10.3390/life12020211] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 01/14/2023] Open
Abstract
Background: Schwann cells (SCs) express cholinergic receptors, suggesting a role of cholinergic signaling in the control of SC proliferation, differentiation and/or myelination. Our previous studies largely demonstrated that the pharmacological activation of the M2 muscarinic receptor subtype caused an inhibition of cell proliferation and promoted the expression of pro-myelinating differentiation genes. In order to elucidate the molecular signaling activated downstream the M2 receptor activation, in the present study we investigated the signal transduction pathways activated by the M2 orthosteric agonist arecaidine propargyl ester (APE) in SCs. Methods: Using Western blot we analyzed some components of the noncanonical pathways involving β1-arrestin and PI3K/AKT/mTORC1 signaling. A wound healing assay was used to evaluate SC migration. Results: Our results demonstrated that M2 receptor activation negatively modulated the PI3K/Akt/mTORC1 axis, possibly through β1-arrestin downregulation. The involvement of the mTORC1 complex was also supported by the decreased expression of its specific target p-p70 S6KThr389. Then, we also analyzed the expression of p-AMPKαthr172, a negative regulator of myelination that resulted in reduced levels after M2 agonist treatment. The analysis of cell migration and morphology allowed us to demonstrate that M2 receptor activation caused an arrest of SC migration and modified cell morphology probably by the modulation of β1-arrestin/cofilin-1 and PKCα expression, respectively. Conclusions: The data obtained demonstrated that M2 receptor activation in addition to the canonical Gi protein-coupled pathway modulates noncanonical pathways involving the mTORC1 complex and other kinases whose activation may contribute to the inhibition of SC proliferation and migration and address SC differentiation.
Collapse
|
15
|
Owen RS, Ramarathinam SH, Bailey A, Gastaldello A, Hussey K, Skipp PJ, Purcell AW, Siddle HV. The differentiation state of the Schwann cell progenitor drives phenotypic variation between two contagious cancers. PLoS Pathog 2021; 17:e1010033. [PMID: 34780568 PMCID: PMC8629380 DOI: 10.1371/journal.ppat.1010033] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 11/29/2021] [Accepted: 10/13/2021] [Indexed: 01/04/2023] Open
Abstract
Contagious cancers are a rare pathogenic phenomenon in which cancer cells gain the ability to spread between genetically distinct hosts. Nine examples have been identified across marine bivalves, dogs and Tasmanian devils, but the Tasmanian devil is the only mammalian species known to have given rise to two distinct lineages of contagious cancer, termed Devil Facial Tumour 1 (DFT1) and 2 (DFT2). Remarkably, DFT1 and DFT2 arose independently from the same cell type, a Schwann cell, and while their ultra-structural features are highly similar they exhibit variation in their mutational signatures and infection dynamics. As such, DFT1 and DFT2 provide a unique framework for investigating how a common progenitor cell can give rise to distinct contagious cancers. Using a proteomics approach, we show that DFT1 and DFT2 are derived from Schwann cells in different differentiation states, with DFT2 carrying a molecular signature of a less well differentiated Schwann cell. Under inflammatory signals DFT1 and DFT2 have different gene expression profiles, most notably involving Schwann cell markers of differentiation, reflecting the influence of their distinct origins. Further, DFT2 cells express immune cell markers typically expressed during nerve repair, consistent with an ability to manipulate their extracellular environment, facilitating the cell’s ability to transmit between individuals. The emergence of two contagious cancers in the Tasmanian devil suggests that the inherent plasticity of Schwann cells confers a vulnerability to the formation of contagious cancers. Cancer can be an infectious pathogen, with nine known cases, infecting bivalves, dogs and two independent tumours circulating in the endangered Tasmanian devil. These cancers, known as Devil Facial Tumour 1 (DFT1) and Devil Facial Tumour 2 (DFT2), spread through the wild population much like parasites, moving between genetically distinct hosts during social biting behaviours and persisting in the population. As DFT1 and DFT2 are independent contagious cancers that arose from the same cell type, a Schwann cell, they provide a unique model system for studying the emergence of phenotypic variation in cancers derived from a single progenitor cell. In this study, we have shown that these two remarkably similar tumours have emerged from Schwann cells in different differentiation states. The differentiation state of the progenitor has altered the characteristics of each tumour, resulting in different responses to external signals. This work demonstrates that the cellular origin of infection can direct the phenotype of a contagious cancer and how it responds to signals from the host environment. Further, the plasticity of Schwann cells may make these cells more prone to forming contagious cancers, raising the possibility that further parasitic cancers could emerge from this cell type.
Collapse
Affiliation(s)
- Rachel S. Owen
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Sri H. Ramarathinam
- Department of Biochemistry and Molecular Biology and the Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Alistair Bailey
- Centre for Cancer Immunology, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Annalisa Gastaldello
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Kathryn Hussey
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
| | - Paul J. Skipp
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
| | - Anthony W. Purcell
- Department of Biochemistry and Molecular Biology and the Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Australia
| | - Hannah V. Siddle
- School of Biological Sciences, University of Southampton, Southampton, United Kingdom
- Institute for Life Sciences, University of Southampton, Southampton, United Kingdom
- * E-mail:
| |
Collapse
|
16
|
Ki SM, Jeong HS, Lee JE. Primary Cilia in Glial Cells: An Oasis in the Journey to Overcoming Neurodegenerative Diseases. Front Neurosci 2021; 15:736888. [PMID: 34658775 PMCID: PMC8514955 DOI: 10.3389/fnins.2021.736888] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/31/2021] [Indexed: 12/29/2022] Open
Abstract
Many neurodegenerative diseases have been associated with defects in primary cilia, which are cellular organelles involved in diverse cellular processes and homeostasis. Several types of glial cells in both the central and peripheral nervous systems not only support the development and function of neurons but also play significant roles in the mechanisms of neurological disease. Nevertheless, most studies have focused on investigating the role of primary cilia in neurons. Accordingly, the interest of recent studies has expanded to elucidate the role of primary cilia in glial cells. Correspondingly, several reports have added to the growing evidence that most glial cells have primary cilia and that impairment of cilia leads to neurodegenerative diseases. In this review, we aimed to understand the regulatory mechanisms of cilia formation and the disease-related functions of cilia, which are common or specific to each glial cell. Moreover, we have paid close attention to the signal transduction and pathological mechanisms mediated by glia cilia in representative neurodegenerative diseases. Finally, we expect that this field of research will clarify the mechanisms involved in the formation and function of glial cilia to provide novel insights and ideas for the treatment of neurodegenerative diseases in the future.
Collapse
Affiliation(s)
- Soo Mi Ki
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea
| | - Hui Su Jeong
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea
| | - Ji Eun Lee
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Sciences and Technology, Sungkyunkwan University, Seoul, South Korea
- Samsung Medical Center, Samsung Biomedical Research Institute, Seoul, South Korea
| |
Collapse
|
17
|
Binamé F, Pham-Van LD, Bagnard D. Manipulating oligodendrocyte intrinsic regeneration mechanism to promote remyelination. Cell Mol Life Sci 2021; 78:5257-5273. [PMID: 34019104 PMCID: PMC11073109 DOI: 10.1007/s00018-021-03852-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 04/14/2021] [Accepted: 05/08/2021] [Indexed: 02/06/2023]
Abstract
In demyelinated lesions, astrocytes, activated microglia and infiltrating macrophages secrete several factors regulating oligodendrocyte precursor cells' behaviour. What appears to be the initiation of an intrinsic mechanism of myelin repair is only leading to partial recovery and inefficient remyelination, a process worsening over the course of the disease. This failure is largely due to the concomitant accumulation of inhibitory cues in and around the lesion sites opposing to growth promoting factors. Here starts a complex game of interactions between the signalling pathways controlling oligodendrocytes migration or differentiation. Receptors of positive or negative cues are modulating Ras, PI3K or RhoGTPases pathways acting on oligodendrocyte cytoskeleton remodelling. From the description of this intricate signalling network, this review addresses the extent to which the modulation of the global response to inhibitory cues may pave the route towards novel therapeutic approaches for myelin repair.
Collapse
Affiliation(s)
- Fabien Binamé
- INSERM U1119, Biopathology of Myelin, Neuroprotection and Therapeutic Strategy (BMNST Lab), Labex Medalis, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Pôle API, Ecole Supérieure de Biotechnologie, 300 Boulevard Sébastien Brant, 67412, Illkirch, France
| | - Lucas D Pham-Van
- INSERM U1119, Biopathology of Myelin, Neuroprotection and Therapeutic Strategy (BMNST Lab), Labex Medalis, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Pôle API, Ecole Supérieure de Biotechnologie, 300 Boulevard Sébastien Brant, 67412, Illkirch, France
| | - Dominique Bagnard
- INSERM U1119, Biopathology of Myelin, Neuroprotection and Therapeutic Strategy (BMNST Lab), Labex Medalis, Fédération de Médecine Translationnelle de Strasbourg (FMTS), Pôle API, Ecole Supérieure de Biotechnologie, 300 Boulevard Sébastien Brant, 67412, Illkirch, France.
| |
Collapse
|
18
|
Yan Z, Qian Y, Fan C. Biomimicry in 3D printing design: implications for peripheral nerve regeneration. Regen Med 2021; 16:683-701. [PMID: 34189955 DOI: 10.2217/rme-2020-0182] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Nerve guide conduits (NGCs) connect dissected nerve stumps and effectively repair short-range peripheral nerve defects. However, for long-range defects, autografts show better therapeutic effects, despite intrinsic limitations. Recent evidence shows that biomimetic design is essential for high-performance NGCs, and 3D printing is a promising fabricating technique. The current work includes a brief review of the challenges for peripheral nerve regeneration. The authors propose a potential solution using biomimetic 3D-printed NGCs as alternative therapies. The assessment of biomimetic designs includes microarchitecture, mechanical property, electrical conductivity and biologics inclusion. The applications of 3D printing in preparing NGCs and present strategies to improve therapeutic effects are also discussed.
Collapse
Affiliation(s)
- Zhiwen Yan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 200233, China.,Youth Science and Technology Innovation Studio, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yun Qian
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 200233, China.,Youth Science and Technology Innovation Studio, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Cunyi Fan
- Department of Orthopedics, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.,Shanghai Engineering Research Center for Orthopaedic Material Innovation and Tissue Regeneration, Shanghai, 200233, China.,Youth Science and Technology Innovation Studio, Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| |
Collapse
|
19
|
Xu Z, Orkwis JA, Harris GM. Cell Shape and Matrix Stiffness Impact Schwann Cell Plasticity via YAP/TAZ and Rho GTPases. Int J Mol Sci 2021; 22:ijms22094821. [PMID: 34062912 PMCID: PMC8124465 DOI: 10.3390/ijms22094821] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 01/01/2023] Open
Abstract
Schwann cells (SCs) are a highly plastic cell type capable of undergoing phenotypic changes following injury or disease. SCs are able to upregulate genes associated with nerve regeneration and ultimately achieve functional recovery. During the regeneration process, the extracellular matrix (ECM) and cell morphology play a cooperative, critical role in regulating SCs, and therefore highly impact nerve regeneration outcomes. However, the roles of the ECM and mechanotransduction relating to SC phenotype are largely unknown. Here, we describe the role that matrix stiffness and cell morphology play in SC phenotype specification via known mechanotransducers YAP/TAZ and RhoA. Using engineered microenvironments to precisely control ECM stiffness, cell shape, and cell spreading, we show that ECM stiffness and SC spreading downregulated SC regenerative associated proteins by the activation of RhoA and YAP/TAZ. Additionally, cell elongation promoted a distinct SC regenerative capacity by the upregulation of Rac1/MKK7/JNK, both necessary for the ECM and morphology changes found during nerve regeneration. These results confirm the role of ECM signaling in peripheral nerve regeneration as well as provide insight to the design of future biomaterials and cellular therapies for peripheral nerve regeneration.
Collapse
Affiliation(s)
- Zhenyuan Xu
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (Z.X.); (J.A.O.)
| | - Jacob A. Orkwis
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (Z.X.); (J.A.O.)
| | - Greg M. Harris
- Department of Chemical and Environmental Engineering, University of Cincinnati, Cincinnati, OH 45221, USA; (Z.X.); (J.A.O.)
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, USA
- Neuroscience Graduate Program, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
- Correspondence: ; Tel.: +1-(513)-556-4167
| |
Collapse
|
20
|
Qu WR, Zhu Z, Liu J, Song DB, Tian H, Chen BP, Li R, Deng LX. Interaction between Schwann cells and other cells during repair of peripheral nerve injury. Neural Regen Res 2021; 16:93-98. [PMID: 32788452 PMCID: PMC7818858 DOI: 10.4103/1673-5374.286956] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Peripheral nerve injury (PNI) is common and, unlike damage to the central nervous system injured nerves can effectively regenerate depending on the location and severity of injury. Peripheral myelinating glia, Schwann cells (SCs), interact with various cells in and around the injury site and are important for debris elimination, repair, and nerve regeneration. Following PNI, Wallerian degeneration of the distal stump is rapidly initiated by degeneration of damaged axons followed by morphologic changes in SCs and the recruitment of circulating macrophages. Interaction with fibroblasts from the injured nerve microenvironment also plays a role in nerve repair. The replication and migration of injury-induced dedifferentiated SCs are also important in repairing the nerve. In particular, SC migration stimulates axonal regeneration and subsequent myelination of regenerated nerve fibers. This mobility increases SC interactions with other cells in the nerve and the exogenous environment, which influence SC behavior post-injury. Following PNI, SCs directly and indirectly interact with other SCs, fibroblasts, and macrophages. In addition, the inter- and intracellular mechanisms that underlie morphological and functional changes in SCs following PNI still require further research to explain known phenomena and less understood cell-specific roles in the repair of the injured peripheral nerve. This review provides a basic assessment of SC function post-PNI, as well as a more comprehensive evaluation of the literature concerning the SC interactions with macrophages and fibroblasts that can influence SC behavior and, ultimately, repair of the injured nerve.
Collapse
Affiliation(s)
- Wen-Rui Qu
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Zhe Zhu
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Jun Liu
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - De-Biao Song
- Department of Emergency and Critical Medicine, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Heng Tian
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Bing-Peng Chen
- Orthopedic Medical Center, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Rui Li
- Department of Hand Surgery, the Second Hospital of Jilin University, Changchun, Jilin Province, China
| | - Ling-Xiao Deng
- Spinal Cord and Brain Injury Research Group, Stark Neurosciences Research Institute, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
21
|
Puhl DL, Funnell JL, Nelson DW, Gottipati MK, Gilbert RJ. Electrospun Fiber Scaffolds for Engineering Glial Cell Behavior to Promote Neural Regeneration. Bioengineering (Basel) 2020; 8:4. [PMID: 33383759 PMCID: PMC7823609 DOI: 10.3390/bioengineering8010004] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023] Open
Abstract
Electrospinning is a fabrication technique used to produce nano- or micro- diameter fibers to generate biocompatible, biodegradable scaffolds for tissue engineering applications. Electrospun fiber scaffolds are advantageous for neural regeneration because they mimic the structure of the nervous system extracellular matrix and provide contact guidance for regenerating axons. Glia are non-neuronal regulatory cells that maintain homeostasis in the healthy nervous system and regulate regeneration in the injured nervous system. Electrospun fiber scaffolds offer a wide range of characteristics, such as fiber alignment, diameter, surface nanotopography, and surface chemistry that can be engineered to achieve a desired glial cell response to injury. Further, electrospun fibers can be loaded with drugs, nucleic acids, or proteins to provide the local, sustained release of such therapeutics to alter glial cell phenotype to better support regeneration. This review provides the first comprehensive overview of how electrospun fiber alignment, diameter, surface nanotopography, surface functionalization, and therapeutic delivery affect Schwann cells in the peripheral nervous system and astrocytes, oligodendrocytes, and microglia in the central nervous system both in vitro and in vivo. The information presented can be used to design and optimize electrospun fiber scaffolds to target glial cell response to mitigate nervous system injury and improve regeneration.
Collapse
Affiliation(s)
- Devan L. Puhl
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; (D.L.P.); (J.L.F.); (D.W.N.); (M.K.G.)
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA
| | - Jessica L. Funnell
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; (D.L.P.); (J.L.F.); (D.W.N.); (M.K.G.)
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA
| | - Derek W. Nelson
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; (D.L.P.); (J.L.F.); (D.W.N.); (M.K.G.)
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA
| | - Manoj K. Gottipati
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; (D.L.P.); (J.L.F.); (D.W.N.); (M.K.G.)
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA
- Center for Brain and Spinal Cord Repair, Department of Neuroscience, The Ohio State University, 460 W. 12th Avenue, Columbus, OH 43210, USA
| | - Ryan J. Gilbert
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA; (D.L.P.); (J.L.F.); (D.W.N.); (M.K.G.)
- Center for Biotechnology and Interdisciplinary Studies, Rensselaer Polytechnic Institute, 110 8th Street, Troy, NY 12180, USA
| |
Collapse
|
22
|
Stewart CE, Kan CFK, Stewart BR, Sanicola HW, Jung JP, Sulaiman OAR, Wang D. Machine intelligence for nerve conduit design and production. J Biol Eng 2020; 14:25. [PMID: 32944070 PMCID: PMC7487837 DOI: 10.1186/s13036-020-00245-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Accepted: 08/13/2020] [Indexed: 02/08/2023] Open
Abstract
Nerve guidance conduits (NGCs) have emerged from recent advances within tissue engineering as a promising alternative to autografts for peripheral nerve repair. NGCs are tubular structures with engineered biomaterials, which guide axonal regeneration from the injured proximal nerve to the distal stump. NGC design can synergistically combine multiple properties to enhance proliferation of stem and neuronal cells, improve nerve migration, attenuate inflammation and reduce scar tissue formation. The aim of most laboratories fabricating NGCs is the development of an automated process that incorporates patient-specific features and complex tissue blueprints (e.g. neurovascular conduit) that serve as the basis for more complicated muscular and skin grafts. One of the major limitations for tissue engineering is lack of guidance for generating tissue blueprints and the absence of streamlined manufacturing processes. With the rapid expansion of machine intelligence, high dimensional image analysis, and computational scaffold design, optimized tissue templates for 3D bioprinting (3DBP) are feasible. In this review, we examine the translational challenges to peripheral nerve regeneration and where machine intelligence can innovate bottlenecks in neural tissue engineering.
Collapse
Affiliation(s)
- Caleb E. Stewart
- Current Affiliation: Department of Neurosurgery, Louisiana State University Health Sciences Center, Shreveport Louisiana, USA
| | - Chin Fung Kelvin Kan
- Current Affiliation: Department of General Surgery, Brigham and Women’s Hospital, Boston, MA 02115 USA
| | - Brody R. Stewart
- Current Affiliation: Department of Surgery, Mayo Clinic College of Medicine, Rochester, MN 55905 USA
| | - Henry W. Sanicola
- Current Affiliation: Department of Neurosurgery, Louisiana State University Health Sciences Center, Shreveport Louisiana, USA
| | - Jangwook P. Jung
- Department of Biological Engineering, Louisiana State University, Baton Rouge, LA 70803 USA
| | - Olawale A. R. Sulaiman
- Ochsner Neural Injury & Regeneration Laboratory, Ochsner Clinic Foundation, New Orleans, LA 70121 USA
- Department of Neurosurgery, Ochsner Clinic Foundation, New Orleans, 70121 USA
| | - Dadong Wang
- Quantitative Imaging Research Team, Data 61, Commonwealth Scientific and Industrial Research Organization, Marsfield, NSW 2122 Australia
| |
Collapse
|
23
|
Li Z, Yu Y, Kang J, Zheng Y, Xu J, Xu K, Hou K, Hou Y, Chi G. MicroRNA-124 Overexpression in Schwann Cells Promotes Schwann Cell-Astrocyte Integration and Inhibits Glial Scar Formation Ability. Front Cell Neurosci 2020; 14:144. [PMID: 32714149 PMCID: PMC7347021 DOI: 10.3389/fncel.2020.00144] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/28/2020] [Indexed: 11/13/2022] Open
Abstract
Schwann cell (SC) transplantation is a promising approach for the treatment of spinal cord injury (SCI); however, SC grafts show a low migratory capacity within the astrocytic environment, which inevitably hampers their therapeutic efficacy. The purpose of this study was to explore mechanisms to modify the characteristics of SCs and astrocytes (ASs), as well as to adjust the SC-AS interface to break the SC-AS boundary, thus improving the benefits of SCI treatment. We observed that the expression levels of miR-124 in SCs and ASs were significantly lower than those in the normal spinal cord. Furthermore, overexpressing miR-124 in SCs (miR-124-SCs) significantly inhibited gene and protein expression levels of SC-specific markers, such as GFAP and Krox20. The expression of neurotrophic factors, Bdnf and Nt-3, was up-regulated in miR-124-SCs without affecting their proliferation. Further, the boundary assay showed an increased number of miR-124-SCs that had actively migrated and entered the astrocytic region to intermingle with ASs, compared with normal SCs. In addition, although Krox20 protein expression was down-regulated in miR-124-SCs, the luciferase assay showed that Krox20 is not a direct target of miR-124. RNA sequencing of miR-124-SCs revealed seven upregulated and eleven downregulated genes involved in cell migration and motility. Based on KEGG pathway and KOG functional analyses, changes in these genes corresponded to the activation of Hippo, FoxO, and TGF-beta signaling pathways, cytokine-cytokine receptor interactions, and the cell cycle. Finally, co-culturing of miR-124-SCs and ASs in a transwell system revealed that GFAP and p-STAT3 protein expression in ASs was significantly reduced. Collectively, these results show that overexpression of miR-124 in SCs promotes SC-AS integration in vitro and may attenuate the capacity of ASs to form glial scars. Thus, this study provides novel insights into modifying SCs by overexpressing miR-124 to improve their therapeutic potential in SCI.
Collapse
Affiliation(s)
- Zhijun Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yifei Yu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Juanjuan Kang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Yangyang Zheng
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jinying Xu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Kan Xu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Kun Hou
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, China
| | - Yi Hou
- Department of Regeneration Medicine, School of Pharmaceutical Sciences, Jilin University, Changchun, China
| | - Guangfan Chi
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
24
|
Min Q, Parkinson DB, Dun XP. Migrating Schwann cells direct axon regeneration within the peripheral nerve bridge. Glia 2020; 69:235-254. [PMID: 32697392 DOI: 10.1002/glia.23892] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 07/03/2020] [Accepted: 07/08/2020] [Indexed: 12/12/2022]
Abstract
Schwann cells within the peripheral nervous system possess a remarkable regenerative potential. Current research shows that peripheral nerve-associated Schwann cells possess the capacity to promote repair of multiple tissues including peripheral nerve gap bridging, skin wound healing, digit tip repair as well as tooth regeneration. One of the key features of the specialized repair Schwann cells is that they become highly motile. They not only migrate into the area of damaged tissue and become a key component of regenerating tissue but also secrete signaling molecules to attract macrophages, support neuronal survival, promote axonal regrowth, activate local mesenchymal stem cells, and interact with other cell types. Currently, the importance of migratory Schwann cells in tissue regeneration is most evident in the case of a peripheral nerve transection injury. Following nerve transection, Schwann cells from both proximal and distal nerve stumps migrate into the nerve bridge and form Schwann cell cords to guide axon regeneration. The formation of Schwann cell cords in the nerve bridge is key to successful peripheral nerve repair following transection injury. In this review, we first examine nerve bridge formation and the behavior of Schwann cell migration in the nerve bridge, and then discuss how migrating Schwann cells direct regenerating axons into the distal nerve. We also review the current understanding of signals that could activate Schwann cell migration and signals that Schwann cells utilize to direct axon regeneration. Understanding the molecular mechanism of Schwann cell migration could potentially offer new therapeutic strategies for peripheral nerve repair.
Collapse
Affiliation(s)
- Qing Min
- School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei Province, People's Republic of China
| | - David B Parkinson
- Peninsula Medical School, Faculty of Health, Plymouth University, Plymouth, Devon, UK
| | - Xin-Peng Dun
- School of Pharmacy, Hubei University of Science and Technology, Xianning, Hubei Province, People's Republic of China
- Peninsula Medical School, Faculty of Health, Plymouth University, Plymouth, Devon, UK
- The Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, People's Republic of China
| |
Collapse
|
25
|
Ma T, Yang Y, Quan X, Lu L, Xia B, Gao J, Qi F, Li S, Zhao L, Mei L, Zheng Y, Shen Y, Luo Z, Jin Y, Huang J. Oxygen carrier in core-shell fibers synthesized by coaxial electrospinning enhances Schwann cell survival and nerve regeneration. Am J Cancer Res 2020; 10:8957-8973. [PMID: 32802174 PMCID: PMC7415813 DOI: 10.7150/thno.45035] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 06/17/2020] [Indexed: 12/11/2022] Open
Abstract
Rationale: Local hypoxia is a challenge for fabrication of cellular grafts and treatment of peripheral nerve injury. In our previous studies, we demonstrated that perfluorotributylamine (PFTBA) could provide short term oxygen supply to Schwann cells (SCs) and counteract the detrimental effects of hypoxia on SCs during the early stages of nerve injury. However, the quick release of oxygen in PFTBA compromised its ability to counteract hypoxia over an extended time, limiting its performance in peripheral nerve injury. Methods: In this study, PFTBA-based oxygen carrier systems were prepared through coaxial electrospinning to prolong the time course of oxygen release. Core-shell structures were fabricated, optimized, and the oxygen kinetics of PFTBA-enriched core-shell fibers evaluated. The effect of core-shells on the survival and function of SCs was examined in both 2D and 3D systems as well as in vivo. The system was used to bridge large sciatic nerve defects in rats. Results: PFTBA core-shell fibers provided high levels of oxygen to SCs in vitro, enhancing their survival, and increasing NGF, BDNF, and VEGF expression in 2D and 3D culture systems under hypoxic condition. In vivo analysis showed that the majority of GFP-expressing SCs in the PFTBA conduit remained viable 14 days post-implantation. We found that axons in PFTBA oxygen carrier scaffold improved axonal regeneration, remyelination, and recovery. Conclusion: A synthetic oxygen carrier in core-shell fibers was fabricated by the coaxial electrospinning technique and was capable of enhancing SC survival and nerve regeneration by prolonged oxygen supply. These findings provide a new strategy for fabricating cellular scaffolds to achieve regeneration in peripheral nerve injury treatment and other aerobic tissue injuries.
Collapse
|
26
|
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
|
27
|
Ferdoushi A, Li X, Jamaluddin MFB, Hondermarck H. Proteomic Profile of Human Schwann Cells. Proteomics 2019; 20:e1900294. [PMID: 31820567 DOI: 10.1002/pmic.201900294] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/22/2019] [Indexed: 12/29/2022]
Abstract
Schwann cells (SC) are essential for the growth, maintenance, and regeneration of peripheral nerves, but the proteome of normal human SC is poorly defined. Here, a proteomic analysis by LC-MS/MS is performed to define the protein expression profile of primary human SC. A total of 19 557 unique peptides corresponding to 1553 individual proteins are identified. Ingenuity Pathway Analysis (IPA), Gene Ontology (GO), and Database for Annotation, Visualization, and Integrated Discovery (DAVID) are used to assign protein localization and function, and to define enriched pathways. EIF2, mTOR, and integrin signaling are among the most enriched pathways and the most enriched biological function is cell-cell adhesion, which is in agreement with the supportive role of SC in peripheral nerves. In addition, several nociceptors and synaptic proteins are identified and may contribute to the recently discovered role of SC in pain sensation and cancer progression. This proteome analysis of normal human SC constitutes a reference for future molecular explorations of physiological and pathological processes where SC are involved.
Collapse
Affiliation(s)
- Aysha Ferdoushi
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, University of Newcastle, New Lambton, NSW, 2305, Australia
| | - Xiang Li
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, University of Newcastle, New Lambton, NSW, 2305, Australia
| | - Muhammad Fairuz Bin Jamaluddin
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, University of Newcastle, New Lambton, NSW, 2305, Australia
| | - Hubert Hondermarck
- School of Biomedical Sciences and Pharmacy, Faculty of Health and Medicine, University of Newcastle, Callaghan, NSW, 2308, Australia.,Hunter Medical Research Institute, University of Newcastle, New Lambton, NSW, 2305, Australia
| |
Collapse
|
28
|
Torii T, Miyamoto Y, Yamauchi J. Cellular Signal-Regulated Schwann Cell Myelination and Remyelination. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1190:3-22. [PMID: 31760634 DOI: 10.1007/978-981-32-9636-7_1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Increasing studies have demonstrated multiple signaling molecules responsible for oligodendrocytes and Schwann cells development such as migration, differentiation, myelination, and axo-glial interaction. However, complicated roles in these events are still poorly understood. This chapter focuses on well established intracellular signaling transduction and recent topics that control myelination and are elucidated from accumulating evidences. The underlying molecular mechanisms, which involved in membrane trafficking through small GTPase Arf6 and its activator cytohesins, demonstrate the crosstalk between well established intracellular signaling transduction and a new finding signaling pathway in glial cells links to physiological phenotype and essential role in peripheral nerve system (PNS). Since Arf family proteins affect the expression levels of myelin protein zero (MPZ) and Krox20, which is a transcription factor regulatory factor in early developmental stages of Schwann cells, Arf proteins likely to be key regulator for Schwann cells development. Herein, we discuss how intracellular signaling transductions in Schwann cells associate with myelination in CNS and PNS.
Collapse
Affiliation(s)
- Tomohiro Torii
- Graduate School of Brain Science, Doshisha University, Kyotanabe-shi, Kyoto, Japan
| | - Yuki Miyamoto
- Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya, Tokyo, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Science, Hachioji, Tokyo, Japan.
| |
Collapse
|
29
|
Li H, Yang Z, Wang W, Wang J, Zhang J, Liu J, Yang T, Yang Y, Wei J, Lei D, Yang X. NT-3/TrkC Axis Contributes to the Perineural Invasion and the Poor Prognosis in Human Salivary Adenoid Cystic Carcinoma. J Cancer 2019; 10:6065-6073. [PMID: 31762816 PMCID: PMC6856580 DOI: 10.7150/jca.33635] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Accepted: 08/25/2019] [Indexed: 12/19/2022] Open
Abstract
The present study was aimed to investigate the role and mechanism of neurotrophin-3 (NT-3) and its specific receptor tropomyosin receptor kinase C (TrkC) in the perineural invasion (PNI) process of the salivary adenoid cystic carcinoma (SACC). The co-cultured system between SACC cells and Schwann cells (SCs) was employed to detect the expression of NT-3 and TrkC. The results of ELISA, qRT-PCR and western blot showed that NT-3 was noticeably elevated in the co-cultured SACC-83 cells, while TrkC was increased in the co-cultured SCs. The results of scratch wound healing, migration, and 3D co-culture assays showed that the directional migration abilities of the co-cultured SACC-83 cells and SCs were significantly increased. Under the stimulation of NT-3, the directional motor ability of SACC-83 cells and SCs was significantly improved, and the apoptosis of SACC-83 cells and SCs were obviously inhibited. In addition, blocking TrkC by its specific inhibitor AZD7451 could significantly inhibit these effects. Immunohistochemistry staining showed that the positive expression of NT-3 (88.5%) and TrkC (92.3%) was significantly correlated with the PNI in SACC specimens (P < 0.05). Additionally, the high expression of NT-3 was significantly associated with the poor prognosis of SACC patients (P < 0.05). The present study indicated that NT-3/TrkC axis contributed to the PNI progression and the poor prognosis of SACC via regulating the interaction between SACC cells and SCs. Interruption of the interaction between SACC cells and SCs by blocking the NT-3/TrkC axis might be an effective strategy for anti-PNI therapy in SACC.
Collapse
Affiliation(s)
- Huan Li
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Zihui Yang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Weiqi Wang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Jun Wang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Jianying Zhang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Junye Liu
- Department of Radiation Medicine, Fourth Military Medical University, Xi'an 710032, China
| | - Tao Yang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Yaowu Yang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Jianhua Wei
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Delin Lei
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| | - Xinjie Yang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases, and Shaanxi Clinical Research Center for Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, Fourth Military Medical University, Xi'an 710032, China
| |
Collapse
|
30
|
Das J, Verma D, Gustafsson M, Lerm M. Identification of DNA methylation patterns predisposing for an efficient response to BCG vaccination in healthy BCG-naïve subjects. Epigenetics 2019; 14:589-601. [PMID: 31010371 PMCID: PMC6557603 DOI: 10.1080/15592294.2019.1603963] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/19/2019] [Accepted: 04/02/2019] [Indexed: 12/17/2022] Open
Abstract
The protection against tuberculosis induced by the Bacille Calmette Guérin (BCG) vaccine is unpredictable. In our previous study, altered DNA methylation pattern in peripheral blood mononuclear cells (PBMCs) in response to BCG was observed in a subgroup of individuals, whose macrophages killed mycobacteria effectively ('responders'). These macrophages also showed production of Interleukin-1β (IL-1β) in response to mycobacterial stimuli before vaccination. Here, we hypothesized that the propensity to respond to the BCG vaccine is reflected in the DNA methylome. We mapped the differentially methylated genes (DMGs) in PBMCs isolated from responders/non-responders at the time point before vaccination aiming to identify possible predictors of BCG responsiveness. We identified 43 DMGs and subsequent bioinformatic analyses showed that these were enriched for actin-modulating pathways, predicting differences in phagocytosis. This could be validated by experiments showing that phagocytosis of mycobacteria, which is an event preceding mycobacteria-induced IL-1β production, was strongly correlated with the DMG pattern.
Collapse
Affiliation(s)
- Jyotirmoy Das
- Department of Clinical and Experimental Medicine (IKE), Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| | - Deepti Verma
- Department of Clinical and Experimental Medicine (IKE), Division of Cell Biology (CELLB), Linköping University, Linköping, Sweden
| | - Mika Gustafsson
- Department of Physics, Chemistry and Biology (IFM) Bioinformatics (BION), Linköping University, Linköping, Sweden
| | - Maria Lerm
- Department of Clinical and Experimental Medicine (IKE), Faculty of Medicine and Health Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
31
|
Lu G, Zhang M, Wang J, Zhang K, Wu S, Zhao X. Epigenetic regulation of myelination in health and disease. Eur J Neurosci 2019; 49:1371-1387. [DOI: 10.1111/ejn.14337] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 12/22/2018] [Accepted: 01/02/2019] [Indexed: 12/20/2022]
Affiliation(s)
- Guozhen Lu
- Department of Neurobiology and Collaborative Innovation Center for Brain ScienceSchool of Basic MedicineFourth Military Medical University Xi'an China
| | - Ming Zhang
- Department of Neurobiology and Collaborative Innovation Center for Brain ScienceSchool of Basic MedicineFourth Military Medical University Xi'an China
| | - Jian Wang
- Department of Neurobiology and Collaborative Innovation Center for Brain ScienceSchool of Basic MedicineFourth Military Medical University Xi'an China
| | - Kaixiang Zhang
- Department of Neurobiology and Collaborative Innovation Center for Brain ScienceSchool of Basic MedicineFourth Military Medical University Xi'an China
| | - Shengxi Wu
- Department of Neurobiology and Collaborative Innovation Center for Brain ScienceSchool of Basic MedicineFourth Military Medical University Xi'an China
| | - Xianghui Zhao
- Department of Neurobiology and Collaborative Innovation Center for Brain ScienceSchool of Basic MedicineFourth Military Medical University Xi'an China
| |
Collapse
|
32
|
Dixon AR, Jariwala SH, Bilis Z, Loverde JR, Pasquina PF, Alvarez LM. Bridging the gap in peripheral nerve repair with 3D printed and bioprinted conduits. Biomaterials 2018; 186:44-63. [DOI: 10.1016/j.biomaterials.2018.09.010] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Revised: 09/06/2018] [Accepted: 09/07/2018] [Indexed: 01/14/2023]
|
33
|
Labroo P, Hilgart D, Davis B, Lambert C, Sant H, Gale B, Shea JE, Agarwal J. Drug-delivering nerve conduit improves regeneration in a critical-sized gap. Biotechnol Bioeng 2018; 116:143-154. [PMID: 30229866 DOI: 10.1002/bit.26837] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2018] [Revised: 09/04/2018] [Accepted: 09/05/2018] [Indexed: 01/03/2023]
Abstract
Autologous nerve grafts are the current "gold standard" for repairing large nerve gaps. However, they cause morbidity at the donor nerve site and only a limited amount of nerve can be harvested. Nerve conduits are a promising alternative to autografts and can act as guidance cues for the regenerating axons, without the need to harvest donor nerve. Separately, it has been shown that localized delivery of GDNF can enhance axon growth and motor recovery. FK506, an FDA approved small molecule, has also been shown to enhance peripheral nerve regeneration. This paper describes the design of a novel hole-based drug delivery apparatus integrated with a polytetrafluoroethylene (PTFE) nerve conduit for controlled local delivery of a protein such as GDNF or a small molecule such as FK506. The PTFE devices were tested in a diffusion chamber, and the bioactivity of the released media was evaluated by measuring neurite growth of dorsal root ganglions (DRGs) exposed to the released drugs. The drug delivering nerve guide was able to release bioactive concentrations of FK506 or GDNF. Following these tests, optimized drug releasing nerve conduits were implanted across 10 mm sciatic nerve gaps in a BL6 yellow fluorescent protein (YFP) mouse model, where they demonstrated significant improvement in muscle mass, compound muscle action potential, and axon myelination in vivo as compared with nerve conduits without the drug. The drug delivery nerve guide could release drug for extended periods of time and enhance axon growth in vitro and in vivo.
Collapse
Affiliation(s)
- Pratima Labroo
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah
| | - David Hilgart
- Department of Bioengineering, University of Utah, Salt Lake City, Utah
| | - Brett Davis
- Department of Bioengineering, University of Utah, Salt Lake City, Utah
| | - Christopher Lambert
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah
| | - Himanshu Sant
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah
| | - Bruce Gale
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah
| | - Jill E Shea
- Department of Surgery, University of Utah, Salt Lake City, Utah
| | - Jayant Agarwal
- Department of Surgery, University of Utah, Salt Lake City, Utah
| |
Collapse
|
34
|
Gu Y, Wu Y, Su W, Xing L, Shen Y, He X, Li L, Yuan Y, Tang X, Chen G. 17β-Estradiol Enhances Schwann Cell Differentiation via the ERβ-ERK1/2 Signaling Pathway and Promotes Remyelination in Injured Sciatic Nerves. Front Pharmacol 2018; 9:1026. [PMID: 30356713 PMCID: PMC6189327 DOI: 10.3389/fphar.2018.01026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Accepted: 08/23/2018] [Indexed: 01/08/2023] Open
Abstract
Remyelination is critical for nerve regeneration. However, the molecular mechanism involved in remyelination is poorly understood. To explore the roles of 17β-estradiol (E2) for myelination in the peripheral nervous system, we used a co-culture model of rat dorsal root ganglion (DRG) explants and Schwann cells (SCs) and a regeneration model of the crushed sciatic nerves in ovariectomized (OVX) and non-ovariectomized (non-OVX) rats for in vitro and in vivo analysis. E2 promoted myelination by facilitating the differentiation of SCs in vitro, which could be inhibited by the estrogen receptors (ER) antagonist ICI182780, ERβ antagonist PHTPP, or ERK1/2 antagonist PD98059. This suggests that E2 accelerates SC differentiation via the ERβ-ERK1/2 signaling. Furthermore, E2 promotes remyelination in crushed sciatic nerves of both OVX and non-OVX rats. Interestingly, E2 also significantly increased the expression of the lysosome membrane proteins LAMP1 and myelin protein P0 in the regenerating nerves. Moreover, P0 has higher degree of colocalization with LAMP1 in the regenerating nerves. Taking together, our results suggest that E2 enhances Schwann cell differentiation and further myelination via the ERβ-ERK1/2 signaling and that E2 increases the expression of myelin proteins and lysosomes in SCs to promotes remyelination in regenerating sciatic nerves.
Collapse
Affiliation(s)
- Yun Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Affiliated Hospital of Nantong University, Nantong, China
| | - Yumen Wu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Wenfeng Su
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - LingYan Xing
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Yuntian Shen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Xiaowen He
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Lilan Li
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Ying Yuan
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Affiliated Hospital of Nantong University, Nantong, China
| | - Xin Tang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China
| | - Gang Chen
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
35
|
Sachana M, Rolaki A, Bal-Price A. Development of the Adverse Outcome Pathway (AOP): Chronic binding of antagonist to N-methyl-d-aspartate receptors (NMDARs) during brain development induces impairment of learning and memory abilities of children. Toxicol Appl Pharmacol 2018; 354:153-175. [PMID: 29524501 PMCID: PMC6095943 DOI: 10.1016/j.taap.2018.02.024] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/26/2018] [Accepted: 02/28/2018] [Indexed: 01/06/2023]
Abstract
The Adverse Outcome Pathways (AOPs) are designed to provide mechanistic understanding of complex biological systems and pathways of toxicity that result in adverse outcomes (AOs) relevant to regulatory endpoints. AOP concept captures in a structured way the causal relationships resulting from initial chemical interaction with biological target(s) (molecular initiating event) to an AO manifested in individual organisms and/or populations through a sequential series of key events (KEs), which are cellular, anatomical and/or functional changes in biological processes. An AOP provides the mechanistic detail required to support chemical safety assessment, the development of alternative methods and the implementation of an integrated testing strategy. An example of the AOP relevant to developmental neurotoxicity (DNT) is described here following the requirements of information defined by the OECD Users' Handbook Supplement to the Guidance Document for developing and assessing AOPs. In this AOP, the binding of an antagonist to glutamate receptor N-methyl-d-aspartate (NMDAR) receptor is defined as MIE. This MIE triggers a cascade of cellular KEs including reduction of intracellular calcium levels, reduction of brain derived neurotrophic factor release, neuronal cell death, decreased glutamate presynaptic release and aberrant dendritic morphology. At organ level, the above mentioned KEs lead to decreased synaptogenesis and decreased neuronal network formation and function causing learning and memory deficit at organism level, which is defined as the AO. There are in vitro, in vivo and epidemiological data that support the described KEs and their causative relationships rendering this AOP relevant to DNT evaluation in the context of regulatory purposes.
Collapse
Affiliation(s)
| | | | - Anna Bal-Price
- European Commission, Joint Research Centre, Ispra, Italy.
| |
Collapse
|
36
|
Sachana M, Rolaki A, Bal-Price A. Development of the Adverse Outcome Pathway (AOP): Chronic binding of antagonist to N-methyl-d-aspartate receptors (NMDARs) during brain development induces impairment of learning and memory abilities of children. Toxicol Appl Pharmacol 2018; 354:153-175. [PMID: 29524501 DOI: 10.1787/5jlsqs5hcrmq-en] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Revised: 02/26/2018] [Accepted: 02/28/2018] [Indexed: 05/20/2023]
Abstract
The Adverse Outcome Pathways (AOPs) are designed to provide mechanistic understanding of complex biological systems and pathways of toxicity that result in adverse outcomes (AOs) relevant to regulatory endpoints. AOP concept captures in a structured way the causal relationships resulting from initial chemical interaction with biological target(s) (molecular initiating event) to an AO manifested in individual organisms and/or populations through a sequential series of key events (KEs), which are cellular, anatomical and/or functional changes in biological processes. An AOP provides the mechanistic detail required to support chemical safety assessment, the development of alternative methods and the implementation of an integrated testing strategy. An example of the AOP relevant to developmental neurotoxicity (DNT) is described here following the requirements of information defined by the OECD Users' Handbook Supplement to the Guidance Document for developing and assessing AOPs. In this AOP, the binding of an antagonist to glutamate receptor N-methyl-d-aspartate (NMDAR) receptor is defined as MIE. This MIE triggers a cascade of cellular KEs including reduction of intracellular calcium levels, reduction of brain derived neurotrophic factor release, neuronal cell death, decreased glutamate presynaptic release and aberrant dendritic morphology. At organ level, the above mentioned KEs lead to decreased synaptogenesis and decreased neuronal network formation and function causing learning and memory deficit at organism level, which is defined as the AO. There are in vitro, in vivo and epidemiological data that support the described KEs and their causative relationships rendering this AOP relevant to DNT evaluation in the context of regulatory purposes.
Collapse
Affiliation(s)
| | | | - Anna Bal-Price
- European Commission, Joint Research Centre, Ispra, Italy.
| |
Collapse
|
37
|
Abstract
This study was designed to characterize morphologic stages during neuroma development post amputation with an eye toward developing better treatment strategies that intervene before neuromas are fully formed. Right forelimbs of 30 Sprague Dawley rats were amputated and limb stumps were collected at 3, 7, 28, 60 and 90 Days Post Amputation (DPA). Morphology of newly formed nerves and neuromas were assessed via general histology and neurofilament protein antibody staining. Analysis revealed six morphological characteristics during nerve and neuroma development; 1) normal nerve, 2) degenerating axons, 3) axonal sprouts, 4) unorganized bundles of axons, 5) unorganized axon growth into muscles, and 6) unorganized axon growth into fibrotic tissue (neuroma). At early stages (3 & 7 DPA) after amputation, normal nerves could be identified throughout the limb stump and small areas of axonal sprouts were present near the site of injury. Signs of degenerating axons were evident from 7 to 90 DPA. From day 28 on, variability of nerve characteristics with signs of unorganized axon growth into muscle and fibrotic tissue and neuroma formation became visible in multiple areas of stump tissue. These pathological features became more evident on days 60 and 90. At 90 DPA frank neuroma formation was present in all stump tissue. By following nerve regrowth and neuroma formation after amputation we were able to identify 6 separate histological stages of nerve regrowth and neuroma development. Axonal regrowth was observed as early as 3 DPA and signs of unorganized axonal growth and neuroma formation were evident by 28 DPA. Based on these observations we speculate that neuroma treatment and or prevention strategies might be more successful if targeted at the initial stages of development and not after 28 DPA.
Collapse
|
38
|
Wright AA, Todorovic M, Tello-Velasquez J, Rayfield AJ, St John JA, Ekberg JA. Enhancing the Therapeutic Potential of Olfactory Ensheathing Cells in Spinal Cord Repair Using Neurotrophins. Cell Transplant 2018; 27:867-878. [PMID: 29852748 PMCID: PMC6050907 DOI: 10.1177/0963689718759472] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Autologous olfactory ensheathing cell (OEC) transplantation is a promising therapy for
spinal cord injury; however, the efficacy varies between trials in both animals and
humans. The main reason for this variability is that the purity and phenotype of the
transplanted cells differs between studies. OECs are susceptible to modulation with
neurotrophic factors, and thus, neurotrophins can be used to manipulate the transplanted
cells into an optimal, consistent phenotype. OEC transplantation can be divided into 3
phases: (1) cell preparation, (2) cell administration, and (3) continuous support to the
transplanted cells in situ. The ideal behaviour of OECs differs between these 3 phases; in
the cell preparation phase, rapid cell expansion is desirable to decrease the time between
damage and transplantation. In the cell administration phase, OEC survival and integration
at the injury site, in particular migration into the glial scar, are the most critical
factors, along with OEC-mediated phagocytosis of cellular debris. Finally, continuous
support needs to be provided to the transplantation site to promote survival of both
transplanted cells and endogenous cells within injury site and to promote long-term
integration of the transplanted cells and angiogenesis. In this review, we define the 3
phases of OEC transplantation into the injured spinal cord and the optimal cell behaviors
required for each phase. Optimising functional outcomes of OEC transplantation can be
achieved by modulation of cell behaviours with neurotrophins. We identify the key growth
factors that exhibit the strongest potential for optimizing the OEC phenotype required for
each phase.
Collapse
Affiliation(s)
- A A Wright
- 1 Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| | - M Todorovic
- 1 Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia.,2 Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia
| | - J Tello-Velasquez
- 1 Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia
| | - A J Rayfield
- 1 Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia.,2 Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia
| | - J A St John
- 1 Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia.,2 Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia
| | - J A Ekberg
- 1 Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith Institute for Drug Discovery, Griffith University, Nathan, Queensland, Australia.,2 Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia
| |
Collapse
|
39
|
Influence of Genetically Modified Human Umbilical Cord Blood Mononuclear Cells on the Expression of Schwann Cell Molecular Determinants in Spinal Cord Injury. Stem Cells Int 2018. [PMID: 29531538 PMCID: PMC5835253 DOI: 10.1155/2018/4695275] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Spinal cord injury (SCI) unavoidably results in death of not only neurons but also glial cells. In particular, the death of oligodendrocytes leads to impaired nerve impulse conduction in intact axons. However, after SCI, the Schwann cells (SCs) are capable of migrating towards an area of injury and participating in the formation of functional myelin. In addition to SCI, cell-based therapy can influence the migration of SCs and the expression of their molecular determinants. In a number of cases, it can be explained by the ability of implanted cells to secrete neurotrophic factors (NTFs). Genetically modified stem and progenitor cells overexpressing NTFs have recently attracted special attention of researchers and are most promising for the purposes of regenerative medicine. Therefore, we have studied the effect of genetically modified human umbilical cord blood mononuclear cells on the expression of SC molecular determinants in SCI.
Collapse
|
40
|
Li Z, Yang A, Yin X, Dong S, Luo F, Dou C, Lan X, Xie Z, Hou T, Xu J, Xing J. Mesenchymal stem cells promote endothelial progenitor cell migration, vascularization, and bone repair in tissue‐engineered constructs
via
activating CXCR2‐Src‐PKL/Vav2‐Rac1. FASEB J 2018; 32:2197-2211. [DOI: 10.1096/fj.201700895r] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Zhilin Li
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing CityChongqingChina
- Tissue Engineering Laboratory of Chongqing CityChongqingChina
- Department of SpineLanzhou General Hospital, Lanzhou Command of the Chinese People's Liberation Army (CPLA)LanzhouChina
| | - Aijun Yang
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing CityChongqingChina
- Tissue Engineering Laboratory of Chongqing CityChongqingChina
| | - Xiaolong Yin
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing CityChongqingChina
- Tissue Engineering Laboratory of Chongqing CityChongqingChina
| | - Shiwu Dong
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Department of Biomedical Materials ScienceCollege of Biomedical Engineering, Third Military Medical UniversityChongqingChina
| | - Fei Luo
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing CityChongqingChina
- Tissue Engineering Laboratory of Chongqing CityChongqingChina
| | - Ce Dou
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing CityChongqingChina
- Tissue Engineering Laboratory of Chongqing CityChongqingChina
| | - Xu Lan
- Department of SpineLanzhou General Hospital, Lanzhou Command of the Chinese People's Liberation Army (CPLA)LanzhouChina
| | - Zhao Xie
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing CityChongqingChina
- Tissue Engineering Laboratory of Chongqing CityChongqingChina
| | - Tianyong Hou
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing CityChongqingChina
- Tissue Engineering Laboratory of Chongqing CityChongqingChina
| | - Jianzhong Xu
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing CityChongqingChina
- Tissue Engineering Laboratory of Chongqing CityChongqingChina
| | - Junchao Xing
- National and Regional United Engineering Laboratory of Tissue EngineeringDepartment of OrthopedicsSouthwest Hospital, and Third Military Medical UniversityChongqingChina
- Center of Regenerative and Reconstructive Engineering Technology in Chongqing CityChongqingChina
- Tissue Engineering Laboratory of Chongqing CityChongqingChina
| |
Collapse
|
41
|
Zhang D, Xu S, Wu S, Gao C. Micropatterned poly(d,l-lactide-co-caprolactone) films entrapped with gelatin for promoting the alignment and directional migration of Schwann cells. J Mater Chem B 2018; 6:1226-1237. [DOI: 10.1039/c7tb03073h] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Gelatin entrapped and micropatterned poly(d,l-lactide-co-caprolactone) (PLCL) film promotes the alignment and directional migration of Schwann cells.
Collapse
Affiliation(s)
- Deteng Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization
- Department of Polymer Science and Engineering
- Zhejiang University
- Hangzhou 310027
- China
| | - Shengjun Xu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization
- Department of Polymer Science and Engineering
- Zhejiang University
- Hangzhou 310027
- China
| | - Sai Wu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization
- Department of Polymer Science and Engineering
- Zhejiang University
- Hangzhou 310027
- China
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization
- Department of Polymer Science and Engineering
- Zhejiang University
- Hangzhou 310027
- China
| |
Collapse
|
42
|
Ulc A, Gottschling C, Schäfer I, Wegrzyn D, van Leeuwen S, Luft V, Reinhard J, Faissner A. Involvement of the guanine nucleotide exchange factor Vav3 in central nervous system development and plasticity. Biol Chem 2017; 398:663-675. [DOI: 10.1515/hsz-2016-0275] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 02/10/2017] [Indexed: 12/13/2022]
Abstract
Abstract
Small GTP-hydrolyzing enzymes (GTPases) of the RhoA family play manifold roles in cell biology and are regulated by upstream guanine nucleotide exchange factors (GEFs). Herein, we focus on the GEFs of the Vav subfamily. Vav1 was originally described as a proto-oncogene of the hematopoietic lineage. The GEFs Vav2 and Vav3 are more broadly expressed in various tissues. In particular, the GEF Vav3 may play important roles in the developing nervous system during the differentiation of neural stem cells into the major lineages, namely neurons, oligodendrocytes and astrocytes. We discuss its putative regulatory roles for progenitor differentiation in the developing retina, polarization of neurons and formation of synapses, migration of oligodendrocyte progenitors and establishment of myelin sheaths. We propose that Vav3 mediates the response of various neural cell types to environmental cues.
Collapse
|
43
|
Barton MJ, John JS, Clarke M, Wright A, Ekberg J. The Glia Response after Peripheral Nerve Injury: A Comparison between Schwann Cells and Olfactory Ensheathing Cells and Their Uses for Neural Regenerative Therapies. Int J Mol Sci 2017; 18:E287. [PMID: 28146061 PMCID: PMC5343823 DOI: 10.3390/ijms18020287] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Revised: 01/14/2017] [Accepted: 01/17/2017] [Indexed: 01/04/2023] Open
Abstract
The peripheral nervous system (PNS) exhibits a much larger capacity for regeneration than the central nervous system (CNS). One reason for this difference is the difference in glial cell types between the two systems. PNS glia respond rapidly to nerve injury by clearing debris from the injury site, supplying essential growth factors and providing structural support; all of which enhances neuronal regeneration. Thus, transplantation of glial cells from the PNS is a very promising therapy for injuries to both the PNS and the CNS. There are two key types of PNS glia: olfactory ensheathing cells (OECs), which populate the olfactory nerve, and Schwann cells (SCs), which are present in the rest of the PNS. These two glial types share many similar morphological and functional characteristics but also exhibit key differences. The olfactory nerve is constantly turning over throughout life, which means OECs are continuously stimulating neural regeneration, whilst SCs only promote regeneration after direct injury to the PNS. This review presents a comparison between these two PNS systems in respect to normal physiology, developmental anatomy, glial functions and their responses to injury. A thorough understanding of the mechanisms and differences between the two systems is crucial for the development of future therapies using transplantation of peripheral glia to treat neural injuries and/or disease.
Collapse
Affiliation(s)
- Matthew J Barton
- Menzies Health Institute Queensland, Griffith University, Nathan QLD 4111, Australia.
- Clem Jones Centre for Neurobiology & Stem Cell Research, Griffith University, Nathan QLD 4111, Australia.
| | - James St John
- Menzies Health Institute Queensland, Griffith University, Nathan QLD 4111, Australia.
- Clem Jones Centre for Neurobiology & Stem Cell Research, Griffith University, Nathan QLD 4111, Australia.
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia.
| | - Mary Clarke
- Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD 4111, Australia.
| | - Alison Wright
- Faculty of Health and Medical Science, Bond University, Robina, QLD 4226, Australia.
| | - Jenny Ekberg
- Clem Jones Centre for Neurobiology & Stem Cell Research, Griffith University, Nathan QLD 4111, Australia.
- Faculty of Health and Medical Science, Bond University, Robina, QLD 4226, Australia.
| |
Collapse
|
44
|
Labroo P, Shea J, Sant H, Gale B, Agarwal J. Effect Of combining FK506 and neurotrophins on neurite branching and elongation. Muscle Nerve 2016; 55:570-581. [PMID: 27503321 DOI: 10.1002/mus.25370] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 07/28/2016] [Accepted: 08/05/2016] [Indexed: 12/21/2022]
Abstract
INTRODUCTION There is a clinical need to improve the outcomes of peripheral nerve regeneration and repair after injury. In addition to its immunosuppressive effects, FK506 (tacrolimus) has been shown to have neuroregenerative properties. To determine biologically relevant local FK506 and growth factor concentrations, we performed an in vitro bioassay using dorsal root ganglion (DRG) from chicken embryos. METHODS Neurite elongation and neurite branching were analyzed microscopically after addition of FK506, glial cell line-derived neurotrophic factor (GDNF), and nerve growth factor (NGF), each alone and in combination. RESULTS FK506 induced modest neurite elongation (∼500-800 µm) without improving neurite branching significantly. The combination of FK506 with NGF, GDNF, or both, exerted a potentiating or competitive effect on neurite elongation (∼700-1100 µm) based on dosage and competitive effect on neurite branching (∼0.2-0.4). CONCLUSIONS These results strongly suggest that the interaction of FK506 with GDNF and NGF mediates distinct enhancement of neurite growth. Muscle Nerve 55: 570-581, 2017.
Collapse
Affiliation(s)
- Pratima Labroo
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Jill Shea
- Department of Surgery, University of Utah, 30 N 1900 E, 3b400, Salt Lake City, Utah, 84132, USA
| | - Himanshu Sant
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Bruce Gale
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Jayant Agarwal
- Department of Surgery, University of Utah, 30 N 1900 E, 3b400, Salt Lake City, Utah, 84132, USA
| |
Collapse
|
45
|
Ding YQ, Li XY, Xia GN, Ren HY, Zhou XF, Su BY, Qi JG. ProBDNF inhibits collective migration and chemotaxis of rat Schwann cells. Tissue Cell 2016; 48:503-10. [DOI: 10.1016/j.tice.2016.07.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 06/15/2016] [Accepted: 07/13/2016] [Indexed: 01/23/2023]
|
46
|
Wang Y, Teng HL, Gao Y, Zhang F, Ding YQ, Huang ZH. Brain-derived Neurotrophic Factor Promotes the Migration of Olfactory Ensheathing Cells Through TRPC Channels. Glia 2016; 64:2154-2165. [PMID: 27534509 DOI: 10.1002/glia.23049] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 07/24/2016] [Accepted: 08/02/2016] [Indexed: 01/15/2023]
Abstract
Olfactory ensheathing cells (OECs) are a unique type of glial cells with axonal growth-promoting properties in the olfactory system. Organized migration of OECs is essential for neural regeneration and olfactory development. However, the molecular mechanism of OEC migration remains unclear. In the present study, we examined the effects of brain-derived neurotrophic factor (BDNF) on OEC migration. Initially, the "scratch" migration assay, the inverted coverslip and Boyden chamber migration assays showed that BDNF could promote the migration of primary cultured OECs. Furthermore, BDNF gradient attracted the migration of OECs in single-cell migration assays. Mechanistically, TrkB receptor expressed in OECs mediated BDNF-induced OEC migration, and BDNF triggered calcium signals in OECs. Finally, transient receptor potential cation channels (TRPCs) highly expressed in OECs were responsible for BDNF-induced calcium signals, and required for BDNF-induced OEC migration. Taken together, these results demonstrate that BDNF promotes the migration of cultured OECs and an unexpected finding is that TRPCs are required for BDNF-induced OEC migration. GLIA 2016;64:2154-2165.
Collapse
Affiliation(s)
- Ying Wang
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Collaborative Innovation Center for Brain Science, Tongji University School of Medicine, Shanghai, 200092, China.,Institute of Neuroscience and Institute of Hypoxia Medicine, Department of Basic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Hong-Lin Teng
- Department of Spine Surgery, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuan Gao
- Institute of Neuroscience and Institute of Hypoxia Medicine, Department of Basic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Fan Zhang
- Institute of Neuroscience and Institute of Hypoxia Medicine, Department of Basic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Yu-Qiang Ding
- Key Laboratory of Arrhythmias, Ministry of Education, East Hospital, and Department of Anatomy and Neurobiology, Collaborative Innovation Center for Brain Science, Tongji University School of Medicine, Shanghai, 200092, China. .,Institute of Neuroscience and Institute of Hypoxia Medicine, Department of Basic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Zhi-Hui Huang
- Institute of Neuroscience and Institute of Hypoxia Medicine, Department of Basic Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| |
Collapse
|
47
|
Stimulating the proliferation, migration and lamellipodia of Schwann cells using low-dose curcumin. Neuroscience 2016; 324:140-50. [PMID: 26955781 DOI: 10.1016/j.neuroscience.2016.02.073] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 02/23/2016] [Accepted: 02/29/2016] [Indexed: 11/24/2022]
Abstract
Transplantation of peripheral glia is being trialled for neural repair therapies, and identification of compounds that enhance the activity of glia is therefore of therapeutic interest. We have previously shown that curcumin potently stimulates the activity of olfactory glia. We have now examined the effect of curcumin on Schwann cell (SC) activities including proliferation, migration and the expression of protein markers. SCs were treated with control media and with different concentrations of curcumin (0.02-20 μM). Cell proliferation was determined by MTS assay and migration changes were determined by single live cell migration tracking. We found that small doses of curcumin (40 nM) dramatically increased the proliferation and migration in SCs within just one day. When compared with olfactory glia, curcumin stimulated SC proliferation more rapidly and at lower concentrations. Curcumin significantly increased the migration of SCs, and also increased the dynamic activity of lamellipodial waves which are essential for SC migration. Expression of the activated form of the MAP kinase p38 (p-p38) was significantly decreased in curcumin-treated SCs. These results show that curcumin's effects on SCs differ remarkably to its effects on olfactory glia, suggesting that subtypes of closely related glia can be differentially stimulated by curcumin. Overall these results demonstrate that the therapeutically beneficial activities of glia can be differentially enhanced by curcumin which could be used to improve outcomes of neural repair therapies.
Collapse
|
48
|
Lentivirus-Mediated RNA Interference Targeting RhoA Slacks the Migration, Proliferation, and Myelin Formation of Schwann Cells. Mol Neurobiol 2016; 54:1229-1239. [DOI: 10.1007/s12035-016-9733-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2015] [Accepted: 01/19/2016] [Indexed: 10/24/2022]
|
49
|
Fitzpatrick ER, Hu T, Ciccarelli BT, Whitehead IP. Regulation of vesicle transport and cell motility by Golgi-localized Dbs. Small GTPases 2015; 5:1-12. [PMID: 25483302 DOI: 10.4161/sgtp.28570] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
DBS/MCF2L has been recently identified as a risk locus for osteoarthritis. It encodes a guanine nucleotide exchange factor (Dbs) that has been shown to regulate both normal and tumor cell motility. In the current study, we have determined that endogenous Dbs is predominantly expressed as 2 isoforms, a 130 kDa form (Dbs-130) that is localized to the Golgi complex, and an 80 kDa form (Dbs-80) that is localized to the endoplasmic reticulum (ER). We have previously described an inhibitor that binds to the RhoGEF domain of Dbs and blocks its transforming activity. Here we show that the inhibitor localizes to the Golgi, where it specifically interacts with Dbs-130. Inhibition of endogenous Dbs-130 activity is associated with reduced levels of activated Cdc42, enlarged Golgi, and resistance to Brefeldin A-mediated Golgi dispersal, suggesting a role for Dbs in vesicle transport. Cells treated with the inhibitor exhibit normal protein transport from the ER to the Golgi, but are defective in transport from the Golgi to the plasma membrane. Inhibition of Dbs-130 in MDA-MB-231 human breast tumor cells limits motility in both transwell and wound healing assays, but appears to have no effect on the organization of the microtubule cytoskeleton. The reduced motility is associated with a failure to reorient the Golgi toward the leading edge. This is consistent with the Golgi localization, and suggests that the Dbs-130 regulates aspects of the secretory pathway that are required to support cell polarization during directed migration.
Collapse
Key Words
- BFA, Brefeldin A
- COP1, coat protein
- DAPI, 4’, 6-diamidino-2-phenylindole
- DH, Dbl homology
- Dbs
- Dbs, Dbl's big sister
- ECL, electrochemiluminescence
- ER, endoplasmic reticulum
- FACS, fluorescence-activated cell sorting
- FGD1, faciogenital dysplasia 1 protein
- GEF, guanine nucleotide exchange factor
- GFP, green fluorescent protein
- HA, hemagglutinin
- HM, homogenization medium
- MCF2L
- MTOC, microtubule organizing center
- NF-1, neurofibromatosis type 1
- PAK3, p21 protein-activated kinase 3
- PBS, phosphate buffered saline
- PH, pleckstrin homology
- Rho
- SH3, Src homology 3
- VSVG, vesicular stomatitis virus-G
- WGA, wheat germ agglutinin
- breast cancer
- cell motility
- golgi complex
- guanine nucleotide exchange factor
- osteoarthritis
- rtPCR, real-time polymerase chain reaction
- siRNA, small inhibitory RNA
Collapse
Affiliation(s)
- Ethan R Fitzpatrick
- a Department of Microbiology and Molecular Genetics; The New Jersey Medical School-Cancer Center; Rutgers Biomedical and Health Sciences ; Newark , NJ USA
| | | | | | | |
Collapse
|
50
|
Abstract
Dendritic protein synthesis and actin cytoskeleton reorganization are important events required for the consolidation of hippocampal LTP and memory. However, the temporal and spatial relationships between these two processes remain unclear. Here, we report that treatment of adult rat hippocampal slices with BDNF or with tetraethylammonium (TEA), which induces a chemical form of LTP, produces a rapid and transient increase in RhoA protein levels. Changes in RhoA were restricted to dendritic spines of CA3 and CA1 and require de novo protein synthesis regulated by mammalian target of rapamycin (mTOR). BDNF-mediated stimulation of RhoA activity, cofilin phosphorylation, and actin polymerization were completely suppressed by protein synthesis inhibitors. Furthermore, intrahippocampal injections of RhoA antisense oligodeoxynucleotides inhibited theta burst stimulation (TBS)-induced RhoA upregulation in dendritic spines and prevented LTP consolidation. Addition of calpain inhibitors after BDNF or TEA treatment maintained RhoA levels elevated and prolonged the effects of BDNF and TEA on actin polymerization. Finally, the use of isoform-selective calpain inhibitors revealed that calpain-2 was involved in RhoA synthesis, whereas calpain-1 mediated RhoA degradation. Overall, this mechanism provides a novel link between dendritic protein synthesis and reorganization of the actin cytoskeleton in hippocampal dendritic spines during LTP consolidation.
Collapse
|