1
|
Doan RA, Monk KR. Dock1 functions in Schwann cells to regulate development, maintenance, and repair. J Cell Biol 2025; 224:e202311041. [PMID: 40105697 PMCID: PMC11921805 DOI: 10.1083/jcb.202311041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 12/05/2024] [Accepted: 02/10/2025] [Indexed: 03/20/2025] Open
Abstract
Schwann cells, the myelinating glia of the peripheral nervous system (PNS), are critical for myelin development, maintenance, and repair. Rac1 is a known regulator of radial sorting, a key step in developmental myelination. Previously, in zebrafish, we showed that the loss of Dock1, a Rac1-specific guanine nucleotide exchange factor, resulted in delayed peripheral myelination during development. Here, we demonstrate that Dock1 is necessary for myelin maintenance and remyelination after injury in adult zebrafish. Furthermore, Dock1 performs an evolutionarily conserved role in mice, functioning cell autonomously in Schwann cells to regulate the development, maintenance, and repair of peripheral myelin. Pharmacological and genetic manipulation of Rac1 in larval zebrafish, along with the analysis of active Rac1 levels in developing Dock1 mutant mouse nerves, revealed an interaction between these two proteins. We propose that the interplay between Dock1 and Rac1 signaling in Schwann cells is required to establish, maintain, and facilitate repair and remyelination within the PNS.
Collapse
Affiliation(s)
- Ryan A. Doan
- The Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Kelly R. Monk
- The Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
2
|
Negro S, Baggio C, Tonellato M, Stazi M, D’Este G, Megighian A, Montecucco C, Rigoni M. Hydrogen Peroxide Modulates the Timely Activation of Jun and Erk in Schwann Cells at the Injury Site and Is Required for Motor Axon Regeneration. Cells 2025; 14:671. [PMID: 40358195 PMCID: PMC12072069 DOI: 10.3390/cells14090671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/16/2025] [Accepted: 05/02/2025] [Indexed: 05/15/2025] Open
Abstract
Peripheral nervous system (PNS) neurons, including motor neurons (MNs), possess a remarkable ability to regenerate and reinnervate target muscles following nerve injury. This process is orchestrated by a combination of intrinsic neuronal properties and extrinsic factors, with Schwann cells (SCs) playing a central role. Upon injury, SCs transition into a repair phenotype that allows axonal regeneration through molecular signaling and structural guidance. However, the identity of the SCs' reprogramming factors is only partially known. We previously identified hydrogen peroxide (H2O2) as an early and key driver of nerve repair, inducing gene expression rewiring in SCs to support nerve re-growth. In this study, we quantitatively assessed the role of H2O2 in the activation of key pro-regenerative signaling pathways in SCs following sciatic nerve compression, specifically the extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun, which are essential for functional nerve recovery. Notably, we found that H2O2 neutralization does not impact degeneration, but it significantly affects the regenerative response. Collectively, our findings establish H2O2 as a promising regulator of the Schwann cell injury response at the injury site, linking oxidative signaling to the molecular mechanisms governing nerve regeneration.
Collapse
Affiliation(s)
- Samuele Negro
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Chiara Baggio
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Marika Tonellato
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
| | - Marco Stazi
- Cancer Neuroscience Laboratory, Francis Crick Institute, London NW1 1ATK, UK
| | - Giorgia D’Este
- Neurobiology Lab, IRCCS San Camillo Hospital, 30126 Venice, Italy
| | - Aram Megighian
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
- Padua Neuroscience Center, University of Padua, 35129 Padua, Italy
| | | | - Michela Rigoni
- Department of Biomedical Sciences, University of Padua, 35131 Padua, Italy
- Myology Center (CIR-Myo), University of Padua, 35131 Padua, Italy
| |
Collapse
|
3
|
Leibovit-Reiben Z, Godfrey H, Jedlowski P, Thiede R. Neurologic adverse events associated with BRAF and MEK inhibitor therapy in patients with malignant melanoma: a disproportionality analysis using the Food and Drug Administration Adverse Event Reporting System. Melanoma Res 2025; 35:122-129. [PMID: 39656586 DOI: 10.1097/cmr.0000000000001015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
BRAF and MEK inhibitor (BRAFi + MEKi) therapy has improved the treatment of solid tumors with BRAF mutation. However, their neurologic adverse events (nAEs) have been largely unexplored. This study aimed to provide clinicians with more updated knowledge on nAEs associated with BRAFi + MEKi therapy in patients with malignant melanoma compared with nonmelanoma cancers. The United States Food and Drug Administration Adverse Event Reporting System was queried from 2011 to 2022 to capture nAEs reported for the BRAFi + MEKi therapies, vemurafenib plus cobimetinib (V + C), dabrafenib plus trametinib (D + T), and encorafenib plus binimetinib (E + B). A disproportionality analysis was performed to calculate their reporting odds ratios (RORs) and 95% confidence intervals (CIs) using a control group of antineoplastic medications. There were 2881 BRAFi + MEKi therapy-associated nAE cases, the majority of which listed malignant melanoma as the reason for use (87.5, 66.7, and 62.0% for V + C, D + T, and E + B, respectively). Several novel associations were identified; including epidural lipomatosis (ROR: 320.07, 95% CI: 123.76-827.77 for V + C), peripheral nerve lesion (ROR: 185.64, 95% CI: 73.95-466.03 for V + C), Guillain-Barre syndrome (RORs: 8.80, 2.94, and 11.79, 95% CIs: 3.65-21.22, 1.40-6.19, and 5.87-23.66 for V + C, D + T, and E + B), demyelinating polyneuropathy (RORs: 24.72 and 78.98, 95% CI: 8.16-74.86 and 24.84-251.13 for D + T and E + B), and multiple sclerosis (ROR: 5.90, 95% CI: 3.06-11.40 for D + T) in melanoma patients. nAEs in the setting of BRAFi + MEKi therapy should be a safety consideration when utilizing these medications.
Collapse
Affiliation(s)
| | | | - Patrick Jedlowski
- Department of Dermatology, University of Arizona College of Medicine - Tucson, Tucson, Arizona, USA
| | - Rebecca Thiede
- Department of Dermatology, University of Arizona College of Medicine - Tucson, Tucson, Arizona, USA
| |
Collapse
|
4
|
Guzman SD, Brooks SV. Skeletal muscle innervation: Reactive oxygen species as regulators of neuromuscular junction dynamics and motor unit remodeling. Free Radic Biol Med 2025; 230:58-65. [PMID: 39892501 PMCID: PMC11893230 DOI: 10.1016/j.freeradbiomed.2025.01.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/21/2025] [Accepted: 01/29/2025] [Indexed: 02/03/2025]
Abstract
This review explores the intricate processes of motor unit remodeling with a specific focus on the influence of reactive oxygen species (ROS) and oxidative stress on the primary cellular components: nerves/axons, muscle fibers, and muscle-resident glial cells. Emphasizing the role of redox biology, we highlight how oxidative stress impacts motor unit adaptation, injury response, and aging. By synthesizing findings from recent studies with seminal works, including investigations of myelin and terminal Schwann cells and neuromuscular junction (NMJ) dynamics, this review provides a comprehensive understanding of the molecular mechanisms underpinning motor unit maintenance and repair. The goal is to elucidate how oxidative stress influences these processes and to explore potential therapeutic strategies for neuromuscular disorders.
Collapse
Affiliation(s)
- Steve D Guzman
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Susan V Brooks
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
5
|
Van Emmenis L, Mòdol-Caballero G, Harford-Wright E, Power A, Cattin AL, White IJ, Casal G, Boal-Carvalho I, Bennett CL, Lloyd AC. Identification of CCL3 as a Schwann cell chemotactic factor essential for nerve regeneration. Cell Rep 2025; 44:115322. [PMID: 39960833 DOI: 10.1016/j.celrep.2025.115322] [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: 09/11/2024] [Revised: 12/14/2024] [Accepted: 01/27/2025] [Indexed: 02/28/2025] Open
Abstract
Peripheral nerves regenerate following injury, in contrast to those of the central nervous system. This involves the collective migration of Schwann cell (SC) cords, which transport regrowing axons across the wound site. The SC cords migrate along a newly formed vasculature, which bridges the wound site in response to vascular endothelial growth factor, secreted by hypoxic macrophages. However, the directional signals by which SC cords navigate the long distances across the wound, in the absence of those that guide axons during development, remain unknown. Here, we identify CCL3 as the SC chemotactic factor, secreted by hypoxic macrophages, responsible for this process. We show that CCL3 promotes collective SC migration and axonal regrowth in vivo and, using genetic mouse models and widely used CCL3 inhibitors, that CCL3 is essential for effective nerve regeneration. These findings have therapeutic implications for both promoting nerve repair and inhibiting the aberrant nerve growth associated with trauma and disease.
Collapse
Affiliation(s)
- Lucie Van Emmenis
- UCL Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Guillem Mòdol-Caballero
- UCL Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Elizabeth Harford-Wright
- UCL Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Alex Power
- UCL Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Anne-Laure Cattin
- UCL Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Ian J White
- UCL Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Giulia Casal
- UCL Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK
| | - Inês Boal-Carvalho
- Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Clare L Bennett
- Department of Haematology, UCL Cancer Institute, University College London, London WC1E 6DD, UK
| | - Alison C Lloyd
- UCL Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK.
| |
Collapse
|
6
|
Han S, Gao L, Dou X, Wang Z, Yang K, Li D, Yuan Y, Xing C, Jiang B, Tian Y, Feng CL, Zhang P. Chiral Hydrogel Nerve Conduit Boosts Peripheral Nerve Regeneration via Regulation of Schwann Cell Reprogramming. ACS NANO 2024; 18:28358-28370. [PMID: 39403973 DOI: 10.1021/acsnano.4c10653] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/01/2025]
Abstract
The Schwann cell (SC) is essential in peripheral nerve regeneration by reprogramming into a stem-like "repair Schwann cell" (rSC) phenotype; however, maintaining the rSC stemness remains an unmet challenge. Chirality is a fundamental factor controlling cell fate, and its potential role in regulation of SC reprogramming has long been ignored and remains poorly understood. Herein, inspired by natural chiral components in the SC microenvironment, chiral hydrogel nerve conduits are prepared by supramolecular assembly of l/d-phenylalanine derivatives (l/d-Phe) in polymeric chitosan-gelatin conduits. Right-handed l-Phe fibers within hydrogel conduits maintain the stemness of rSC through enhanced stereoselective interaction between collagen IV and l-Phe fibers triggered by collagen IV-Integrin α1β1, MAPK, and YAP/TAZ signaling pathways and finally activate the key regulator of SC reprogramming, the c-Jun pathway. In the rat model of a sciatic nerve defect, the l-Phe hydrogel nerve conduit significantly enhances nerve regeneration, exhibiting markedly improved histological, electrophysiological, and functional outcomes. The findings reveal the chirality-dependent regulation of SC reprogramming in a pioneering way, offering potential strategies for nerve regeneration therapies.
Collapse
Affiliation(s)
- Shuai Han
- Department of Orthopedics, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing 100191, People's Republic of China
- Department of Trauma and Orthopedics, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, People's Republic of China
| | - Laiben Gao
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang District, Shanghai 200230, People's Republic of China
| | - Xiaoqiu Dou
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang District, Shanghai 200230, People's Republic of China
| | - Zhengguang Wang
- Department of Orthopedics, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing 100191, People's Republic of China
| | - Kaikai Yang
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang District, Shanghai 200230, People's Republic of China
| | - Dongdong Li
- Department of Trauma and Orthopedics, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, People's Republic of China
| | - Yusong Yuan
- Department of Trauma and Orthopedics, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, People's Republic of China
| | - Chao Xing
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang District, Shanghai 200230, People's Republic of China
| | - Baoguo Jiang
- Department of Trauma and Orthopedics, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, People's Republic of China
| | - Yun Tian
- Department of Orthopedics, Peking University Third Hospital, 49 North Garden Rd., Haidian District, Beijing 100191, People's Republic of China
| | - Chuan-Liang Feng
- State Key Lab of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, 800 Dongchuan Rd., Minhang District, Shanghai 200230, People's Republic of China
| | - Peixun Zhang
- Department of Trauma and Orthopedics, Peking University People's Hospital, No. 11 Xizhimen South Street, Xicheng District, Beijing 100044, People's Republic of China
| |
Collapse
|
7
|
Qi F, He H, Zhu Y. Neural Development and Repair Induced by Femtosecond Laser Stimulation. ACS Chem Neurosci 2024; 15:3106-3112. [PMID: 39163542 DOI: 10.1021/acschemneuro.4c00310] [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] [Indexed: 08/22/2024] Open
Abstract
Dendritic spines function as postsynaptic sites, receiving excitatory signals from presynaptic axons. The synaptic plasticity of spines underlies the refinement of neuronal circuits. Neural cognitive disorders are commonly associated with the impairment and elimination of dendritic spines. In this study, we report an all-optical method to activate dendritic spine growth and regeneration by a single short flash of femtosecond laser stimulation. The inhibited development and loss of spines can be rescued by a transient illumination of the laser inside a micrometer region of the soma by activating the extracellular signal-regulated kinase (ERK) signaling pathway. The rescued neurons exhibit function. Hence we provide a potential noninvasive method for the regeneration of dendritic spines.
Collapse
Affiliation(s)
- Fan Qi
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, Zhejiang 311121, P. R. China
| | - Hao He
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200031, P. R. China
| | - Yujie Zhu
- Department of Laser and Aesthetic Medicine and Department of Dermatology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200041, P. R. China
| |
Collapse
|
8
|
Perrin S, Protic S, Bretegnier V, Laurendeau I, de Lageneste OD, Panara N, Ruckebusch O, Luka M, Masson C, Maillard T, Coulpier F, Pannier S, Wicart P, Hadj-Rabia S, Radomska KJ, Zarhrate M, Ménager M, Vidaud D, Topilko P, Parfait B, Colnot C. MEK-SHP2 inhibition prevents tibial pseudarthrosis caused by NF1 loss in Schwann cells and skeletal stem/progenitor cells. Sci Transl Med 2024; 16:eadj1597. [PMID: 38924432 DOI: 10.1126/scitranslmed.adj1597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 01/15/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024]
Abstract
Congenital pseudarthrosis of the tibia (CPT) is a severe pathology marked by spontaneous bone fractures that fail to heal, leading to fibrous nonunion. Half of patients with CPT are affected by the multisystemic genetic disorder neurofibromatosis type 1 (NF1) caused by mutations in the NF1 tumor suppressor gene, a negative regulator of RAS-mitogen-activated protein kinase (MAPK) signaling pathway. Here, we analyzed patients with CPT and Prss56-Nf1 knockout mice to elucidate the pathogenic mechanisms of CPT-related fibrous nonunion and explored a pharmacological approach to treat CPT. We identified NF1-deficient Schwann cells and skeletal stem/progenitor cells (SSPCs) in pathological periosteum as affected cell types driving fibrosis. Whereas NF1-deficient SSPCs adopted a fibrotic fate, NF1-deficient Schwann cells produced critical paracrine factors including transforming growth factor-β and induced fibrotic differentiation of wild-type SSPCs. To counteract the elevated RAS-MAPK signaling in both NF1-deficient Schwann cells and SSPCs, we used MAPK kinase (MEK) and Src homology 2 containing protein tyrosine phosphatase 2 (SHP2) inhibitors. Combined MEK-SHP2 inhibition in vivo prevented fibrous nonunion in the Prss56-Nf1 knockout mouse model, providing a promising therapeutic strategy for the treatment of fibrous nonunion in CPT.
Collapse
Affiliation(s)
- Simon Perrin
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| | - Sanela Protic
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| | | | - Ingrid Laurendeau
- INSERM UMR S1016, Institut Cochin, Université Paris Cité, 75014 Paris, France
| | | | - Nicolas Panara
- INSERM UMR S1016, Institut Cochin, Université Paris Cité, 75014 Paris, France
| | - Odile Ruckebusch
- Université Paris Est Creteil, INSERM, IMRB, Plateforme de Cytométrie en flux, 94000 Creteil, France
| | - Marine Luka
- Paris Cité University, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Cécile Masson
- Bioinformatics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163, 75015 Paris, France
- INSERM US24/CNRS UAR3633, Paris Cité University, 75015 Paris, France
| | - Théodora Maillard
- Service de Médecine Génomique des Maladies de Système et d'Organe, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université Paris Cité, F-75014 Paris, France
| | - Fanny Coulpier
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| | - Stéphanie Pannier
- Department of Pediatric Orthopedic Surgery and Traumatology, Necker-Enfants Malades Hospital, AP-HP, Paris Cité University, 75015 Paris, France
| | - Philippe Wicart
- Department of Pediatric Orthopedic Surgery and Traumatology, Necker-Enfants Malades Hospital, AP-HP, Paris Cité University, 75015 Paris, France
| | - Smail Hadj-Rabia
- Department of Dermatology, Reference Center for Rare Skin Diseases (MAGEC), Imagine Institute, Necker-Enfants Malades Hospital, AP-HP, Paris Cité University, 75015 Paris, France
| | | | - Mohammed Zarhrate
- INSERM US24/CNRS UAR3633, Paris Cité University, 75015 Paris, France
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163, 75015 Paris, France
| | - Mickael Ménager
- Paris Cité University, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Dominique Vidaud
- INSERM UMR S1016, Institut Cochin, Université Paris Cité, 75014 Paris, France
- Service de Médecine Génomique des Maladies de Système et d'Organe, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université Paris Cité, F-75014 Paris, France
| | - Piotr Topilko
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| | - Béatrice Parfait
- INSERM UMR S1016, Institut Cochin, Université Paris Cité, 75014 Paris, France
- Service de Médecine Génomique des Maladies de Système et d'Organe, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université Paris Cité, F-75014 Paris, France
| | - Céline Colnot
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| |
Collapse
|
9
|
Stassart RM, Gomez-Sanchez JA, Lloyd AC. Schwann Cells as Orchestrators of Nerve Repair: Implications for Tissue Regeneration and Pathologies. Cold Spring Harb Perspect Biol 2024; 16:a041363. [PMID: 38199866 PMCID: PMC11146315 DOI: 10.1101/cshperspect.a041363] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2024]
Abstract
Peripheral nerves exist in a stable state in adulthood providing a rapid bidirectional signaling system to control tissue structure and function. However, following injury, peripheral nerves can regenerate much more effectively than those of the central nervous system (CNS). This multicellular process is coordinated by peripheral glia, in particular Schwann cells, which have multiple roles in stimulating and nurturing the regrowth of damaged axons back to their targets. Aside from the repair of damaged nerves themselves, nerve regenerative processes have been linked to the repair of other tissues and de novo innervation appears important in establishing an environment conducive for the development and spread of tumors. In contrast, defects in these processes are linked to neuropathies, aging, and pain. In this review, we focus on the role of peripheral glia, especially Schwann cells, in multiple aspects of nerve regeneration and discuss how these findings may be relevant for pathologies associated with these processes.
Collapse
Affiliation(s)
- Ruth M Stassart
- Paul-Flechsig-Institute of Neuropathology, University Clinic Leipzig, Leipzig 04103, Germany
| | - Jose A Gomez-Sanchez
- Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante 03010, Spain
- Instituto de Neurociencias CSIC-UMH, Sant Joan de Alicante 03550, Spain
| | - Alison C Lloyd
- UCL Laboratory for Molecular Cell Biology, University College London, London WC1E 6BT, United Kingdom
| |
Collapse
|
10
|
Hazard M, Duval F, Dutriaux C, Beylot-Barry M, Pham-Ledard A, Quereux G, Amini-Adle M, Heidelberger V, Aubin F, Saint-Jean M, Nardin C, Abed S, Leccia MT, Mansard S, Prey S, Khammari A, Dréno B, Gérard E. Neurological toxicities of targeted therapies in melanoma: a multicenter national observational study of the French Group of Skin Cancers (Groupe de Cancérologie Cutanée, GCC). J Neurol 2024; 271:1473-1482. [PMID: 38052771 DOI: 10.1007/s00415-023-12105-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 11/06/2023] [Accepted: 11/08/2023] [Indexed: 12/07/2023]
Affiliation(s)
- Madeleine Hazard
- Department of Dermatology, Hospital Dupuytren 2, UHC of Limoges, Limoges, France
| | - Fanny Duval
- Department of Medical Neurology, Hospital Pellegrin, UHC of Bordeaux, Bordeaux, France
| | - Caroline Dutriaux
- Department of Dermatology, Hôpital Saint-André, CHU de Bordeaux, Bordeaux, France
- BRIC (Bordeaux Institute of Oncology), UMR1312, INSERM, Univ. Bordeaux, 33000, Bordeaux, France
| | - Marie Beylot-Barry
- Department of Dermatology, Hôpital Saint-André, CHU de Bordeaux, Bordeaux, France
- BRIC (Bordeaux Institute of Oncology), UMR1312, INSERM, Univ. Bordeaux, 33000, Bordeaux, France
| | - Anne Pham-Ledard
- Department of Dermatology, Hôpital Saint-André, CHU de Bordeaux, Bordeaux, France
- BRIC (Bordeaux Institute of Oncology), UMR1312, INSERM, Univ. Bordeaux, 33000, Bordeaux, France
| | - Gaelle Quereux
- Department of Dermatology, UHC of Nantes, France, CIC 1413, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302, 44000, Nantes, France
| | | | | | - François Aubin
- Department of Dermatology, UHC of Besançon, Besançon, France
| | - Mélanie Saint-Jean
- Department of Oncology, Cancer Comprehensive Center, Saint-Herblain, France
| | - Charlée Nardin
- Department of Dermatology, UHC of Besançon, Besançon, France
| | - Safia Abed
- Department of Dermatology, Military Training Hospital of Sainte Anne, Toulon, France
| | | | - Sandrine Mansard
- Department of Dermatology, Estaing Hospital, Clermont-Ferrand, France
| | - Sorilla Prey
- Department of Dermatology, Hôpital Saint-André, CHU de Bordeaux, Bordeaux, France
- BRIC (Bordeaux Institute of Oncology), UMR1312, INSERM, Univ. Bordeaux, 33000, Bordeaux, France
| | - Amir Khammari
- Department of Dermatology, UHC of Nantes, France, CIC 1413, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302, 44000, Nantes, France
| | - Brigitte Dréno
- Department of Dermatology, UHC of Nantes, France, CIC 1413, INSERM, CNRS, Immunology and New Concepts in ImmunoTherapy, INCIT, UMR 1302, 44000, Nantes, France
| | - Emilie Gérard
- Department of Dermatology, Hôpital Saint-André, CHU de Bordeaux, Bordeaux, France.
- BRIC (Bordeaux Institute of Oncology), UMR1312, INSERM, Univ. Bordeaux, 33000, Bordeaux, France.
| |
Collapse
|
11
|
Gordon T. Brief Electrical Stimulation Promotes Recovery after Surgical Repair of Injured Peripheral Nerves. Int J Mol Sci 2024; 25:665. [PMID: 38203836 PMCID: PMC10779324 DOI: 10.3390/ijms25010665] [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: 10/13/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 01/12/2024] Open
Abstract
Injured peripheral nerves regenerate their axons in contrast to those in the central nervous system. Yet, functional recovery after surgical repair is often disappointing. The basis for poor recovery is progressive deterioration with time and distance of the growth capacity of the neurons that lose their contact with targets (chronic axotomy) and the growth support of the chronically denervated Schwann cells (SC) in the distal nerve stumps. Nonetheless, chronically denervated atrophic muscle retains the capacity for reinnervation. Declining electrical activity of motoneurons accompanies the progressive fall in axotomized neuronal and denervated SC expression of regeneration-associated-genes and declining regenerative success. Reduced motoneuronal activity is due to the withdrawal of synaptic contacts from the soma. Exogenous neurotrophic factors that promote nerve regeneration can replace the endogenous factors whose expression declines with time. But the profuse axonal outgrowth they provoke and the difficulties in their delivery hinder their efficacy. Brief (1 h) low-frequency (20 Hz) electrical stimulation (ES) proximal to the injury site promotes the expression of endogenous growth factors and, in turn, dramatically accelerates axon outgrowth and target reinnervation. The latter ES effect has been demonstrated in both rats and humans. A conditioning ES of intact nerve days prior to nerve injury increases axonal outgrowth and regeneration rate. Thereby, this form of ES is amenable for nerve transfer surgeries and end-to-side neurorrhaphies. However, additional surgery for applying the required electrodes may be a hurdle. ES is applicable in all surgeries with excellent outcomes.
Collapse
Affiliation(s)
- Tessa Gordon
- Division of Reconstructive Surgery, Department of Surgery, University of Toronto, Toronto, ON M4G 1X8, Canada
| |
Collapse
|
12
|
Krishnan A, Verge VMK, Zochodne DW. Hallmarks of peripheral nerve injury and regeneration. HANDBOOK OF CLINICAL NEUROLOGY 2024; 201:1-17. [PMID: 38697733 DOI: 10.1016/b978-0-323-90108-6.00014-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2024]
Abstract
Peripheral nerves are functional networks in the body. Disruption of these networks induces varied functional consequences depending on the types of nerves and organs affected. Despite the advances in microsurgical repair and understanding of nerve regeneration biology, restoring full functions after severe traumatic nerve injuries is still far from achieved. While a blunted growth response from axons and errors in axon guidance due to physical barriers may surface as the major hurdles in repairing nerves, critical additional cellular and molecular aspects challenge the orderly healing of injured nerves. Understanding the systematic reprogramming of injured nerves at the cellular and molecular levels, referred to here as "hallmarks of nerve injury regeneration," will offer better ideas. This chapter discusses the hallmarks of nerve injury and regeneration and critical points of failures in the natural healing process. Potential pharmacological and nonpharmacological intervention points for repairing nerves are also discussed.
Collapse
Affiliation(s)
- Anand Krishnan
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada; Cameco MS Neuroscience Research Centre (CMSNRC), Saskatoon, SK, Canada.
| | - Valerie M K Verge
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada; Cameco MS Neuroscience Research Centre (CMSNRC), Saskatoon, SK, Canada.
| | - Douglas W Zochodne
- Neuroscience and Mental Health Institute and Division of Neurology, Department of Medicine, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
13
|
Hromada C, Szwarc-Hofbauer D, Quyen Nguyen M, Tomasch J, Purtscher M, Hercher D, Teuschl-Woller AH. Strain-induced bands of Büngner formation promotes axon growth in 3D tissue-engineered constructs. J Tissue Eng 2024; 15:20417314231220396. [PMID: 38249993 PMCID: PMC10798132 DOI: 10.1177/20417314231220396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 11/28/2023] [Indexed: 01/23/2024] Open
Abstract
Treatment of peripheral nerve lesions remains a major challenge due to poor functional recovery; hence, ongoing research efforts strive to enhance peripheral nerve repair. In this study, we aimed to establish three-dimensional tissue-engineered bands of Büngner constructs by subjecting Schwann cells (SCs) embedded in fibrin hydrogels to mechanical stimulation. We show for the first time that the application of strain induces (i) longitudinal alignment of SCs resembling bands of Büngner, and (ii) the expression of a pronounced repair SC phenotype as evidenced by upregulation of BDNF, NGF, and p75NTR. Furthermore, we show that mechanically aligned SCs provide physical guidance for migrating axons over several millimeters in vitro in a co-culture model with rat dorsal root ganglion explants. Consequently, these constructs hold great therapeutic potential for transplantation into patients and might also provide a physiologically relevant in vitro peripheral nerve model for drug screening or investigation of pathologic or regenerative processes.
Collapse
Affiliation(s)
- Carina Hromada
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Dorota Szwarc-Hofbauer
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Mai Quyen Nguyen
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, Austria
| | - Janine Tomasch
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Michaela Purtscher
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - David Hercher
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
- Ludwig Boltzmann Institute for Traumatology, The Research Centre in Cooperation with AUVA, Vienna, Austria
| | - Andreas Herbert Teuschl-Woller
- Department Life Science Engineering, University of Applied Sciences Technikum Wien, Vienna, Austria
- The Austrian Cluster for Tissue Regeneration, Vienna, Austria
| |
Collapse
|
14
|
Janku F, Kim TM, Iyer G, Spreafico A, Elez E, de Jonge M, Yamamoto N, van der Wekken AJ, Ascierto PA, Maur M, Marmé F, Kiladjian JJ, Basu S, Baffert F, Buigues A, Chen C, Cooke V, Giorgetti E, Kim J, McCarthy F, Moschetta M, Dummer R. First-in-human study of naporafenib (LXH254) with or without spartalizumab in adult patients with advanced solid tumors harboring MAPK signaling pathway alterations. Eur J Cancer 2024; 196:113458. [PMID: 38039779 PMCID: PMC11380116 DOI: 10.1016/j.ejca.2023.113458] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 12/03/2023]
Abstract
BACKGROUND We investigated naporafenib (LXH254), a pan-RAF kinase inhibitor, with or without spartalizumab, in patients with advanced solid tumors harboring MAPK pathway alterations. METHODS This first-in-human phase 1 study had two dose-escalation arms: single-agent naporafenib (starting at 100 mg once-daily [QD]) and naporafenib (starting at the recommended dose/regimen)/spartalizumab (400 mg every 4 weeks). The naporafenib/spartalizumab dose-expansion part enrolled patients with KRAS-mutated non-small cell lung cancer (NSCLC) and NRAS-mutated melanoma. The primary objectives were to establish the maximum tolerated doses (MTD)/recommended doses for expansion (RDE) and evaluate tolerability and safety. RESULTS A total of 142 patients were included in the naporafenib dose-escalation (n = 87), naporafenib/spartalizumab dose-escalation (n = 12) and naporafenib/spartalizumab dose-expansion (n = 43) arms. The MTD/RDE of naporafenib was 600 mg twice-daily (BID). In naporafenib escalation, five patients experienced 7 dose-limiting toxicities: decreased platelet count (1200 mg QD); neuralgia, maculopapular rash, pruritus (600 mg BID); increased blood bilirubin, hyponatremia, peripheral sensory neuropathy (800 mg BID). No DLTs occurred in the naporafenib/spartalizumab arm: the RDE was established at 400 mg BID. The most common treatment-related adverse events were rash and dermatitis acneiform (each 24.1%; naporafenib), nausea and pruritus (each 33.3%; naporafenib/spartalizumab; escalation) and rash (39.5%; naporafenib/spartalizumab; expansion). Naporafenib reduced DUSP6 expression in tumors. Two partial responses (PRs) occurred in naporafenib escalation, and 1 complete response and 3 PRs in the naporafenib/spartalizumab NRAS-mutated melanoma and KRAS-mutated NSCLC arms, respectively. CONCLUSIONS Naporafenib, with or without spartalizumab, showed an acceptable safety profile, pharmacodynamic activity and limited antitumor activity. Additional naporafenib combination therapies are currently under investigation.
Collapse
Affiliation(s)
- Filip Janku
- The University of Texas, MD Anderson Cancer Center, Houston, TX, USA.
| | - Tae Min Kim
- Seoul National University Hospital, Seoul, South Korea
| | - Gopakumar Iyer
- Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anna Spreafico
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Elena Elez
- Vall d'Hebron University Hospital, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | | | | | | | - Michela Maur
- Oncology Unit, AOU Policlinico di Modena, Modena, Italy
| | - Frederik Marmé
- Medical Faculty Mannheim, Heidelberg University, University Hospital Mannheim, Mannheim, Germany
| | | | - Sumit Basu
- Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Fabienne Baffert
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Amparo Buigues
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Chi Chen
- China Novartis Institutes for BioMedical Research, Shanghai, China
| | - Vesselina Cooke
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Elisa Giorgetti
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Jaeyeon Kim
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | | | - Michele Moschetta
- Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Reinhard Dummer
- University of Zürich, Skin Cancer Center, Zürich, Switzerland.
| |
Collapse
|
15
|
Barbieri MA, Russo G, Sorbara EE, Cicala G, Franchina T, Santarpia M, Speranza D, Spina E, Silvestris N. Neuropsychiatric adverse drug reactions with oral tyrosine kinase inhibitors in metastatic colorectal cancer: an analysis from the FDA Adverse Event Reporting System. Front Oncol 2023; 13:1268672. [PMID: 38023143 PMCID: PMC10646181 DOI: 10.3389/fonc.2023.1268672] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/09/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction New oral tyrosine kinase inhibitors (TKIs) are approved for metastatic colorectal cancer (mCRC). The aim of this study was to assess the neuropsychiatric adverse drug reactions (ADRs) of these drugs reported in the FDA Adverse Event Reporting System (FAERS) database. Methods All reports with regorafenib (REG) and encorafenib (ENC) as the primary suspect, and reported in the FAERS between 2012 and 2022, were collected. A descriptive and disproportionality analyses were conducted. Results Out of 4,984 cases, 1,357 (30.2%) reported at least one neuropsychiatric ADR. New potential signals for REG included neuropathy peripheral (n = 265; reporting odds ratio, ROR = 19.48, 95% confidence interval, CI 95% = 17.52-22.47; information component, IC = 2.89, IC025-IC075 = 2.77-3.02), hyperesthesia (n = 18; ROR = 12.56, CI 95% = 7.90-19.96; IC = 2.25, IC025-IC075 = 1.79-2.72), taste disorder (n = 41; ROR = 9.91, CI 95% = 7.29-13.49; IC = 2.18, IC025-IC075 = 1.88-2.49), poor quality sleep (n = 18; ROR = 6.56, CI 95% = 4.13-10.42; IC = 1.74, IC025-IC075 = 1.27-2.20), altered state of consciousness (n = 15; ROR = 5.50, CI 95% = 3.31-9.14; IC = 1.57, IC025-IC075 = 1.06-2.07), depressed mood (n = 13; ROR = 1.85, CI 95% = 1.07-3.19; IC = 0.58, IC025-IC075 = 0.04-1.13) and insomnia (n = 63; ROR = 1.48, CI 95% = 1.15-1.89; IC = 0.38, IC025-IC075 = 0.13-0.63). For ENC comprised depressed mood (n = 4; ROR = 5.75, CI 95% = 2.15-15.39; IC = 1.74, IC025-IC075 = 0.76-2.73) and cognitive disorders (n = 3; ROR = 4.71, CI 95% = 1.51-14.66; IC = 1.54, IC025-IC075 = 0.41-2.68). Discussion This study identified new unknown potential neuropsychiatric ADRs. Further investigations are required to better define the neurotoxicity of TKIs in mCRC patients.
Collapse
Affiliation(s)
| | - Giulia Russo
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | | | - Giuseppe Cicala
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Tindara Franchina
- Department of Human Pathology in Adulthood and Childhood Gaetano Barresi, University of Messina, Messina, Italy
| | - Mariacarmela Santarpia
- Department of Human Pathology in Adulthood and Childhood Gaetano Barresi, University of Messina, Messina, Italy
| | - Desirèe Speranza
- Department of Human Pathology in Adulthood and Childhood Gaetano Barresi, University of Messina, Messina, Italy
| | - Edoardo Spina
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy
| | - Nicola Silvestris
- Department of Human Pathology in Adulthood and Childhood Gaetano Barresi, University of Messina, Messina, Italy
| |
Collapse
|
16
|
Doan RA, Monk KR. Dock1 acts cell-autonomously in Schwann cells to regulate the development, maintenance, and repair of peripheral myelin. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.26.564271. [PMID: 37961336 PMCID: PMC10634861 DOI: 10.1101/2023.10.26.564271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Schwann cells, the myelinating glia of the peripheral nervous system (PNS), are critical for myelin development, maintenance, and repair. Rac1 is a known regulator of radial sorting, a key step in developmental myelination, and we previously showed in zebrafish that loss of Dock1, a Rac1-specific guanine nucleotide exchange factor, results in delayed peripheral myelination in development. We demonstrate here that Dock1 is necessary for myelin maintenance and remyelination after injury in adult zebrafish. Furthermore, it performs an evolutionary conserved role in mice, acting cell-autonomously in Schwann cells to regulate peripheral myelin development, maintenance, and repair. Additionally, manipulating Rac1 levels in larval zebrafish reveals that dock1 mutants are sensitized to inhibition of Rac1, suggesting an interaction between the two proteins during PNS development. We propose that the interplay between Dock1 and Rac1 signaling in Schwann cells is required to establish, maintain, and facilitate repair and remyelination within the peripheral nervous system.
Collapse
Affiliation(s)
- Ryan A Doan
- The Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| | - Kelly R Monk
- The Vollum Institute, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
17
|
Bai X, Zhao N, Koupourtidou C, Fang LP, Schwarz V, Caudal LC, Zhao R, Hirrlinger J, Walz W, Bian S, Huang W, Ninkovic J, Kirchhoff F, Scheller A. In the mouse cortex, oligodendrocytes regain a plastic capacity, transforming into astrocytes after acute injury. Dev Cell 2023:S1534-5807(23)00192-2. [PMID: 37220747 DOI: 10.1016/j.devcel.2023.04.016] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 02/16/2023] [Accepted: 04/25/2023] [Indexed: 05/25/2023]
Abstract
Acute brain injuries evoke various response cascades directing the formation of the glial scar. Here, we report that acute lesions associated with hemorrhagic injuries trigger a re-programming of oligodendrocytes. Single-cell RNA sequencing highlighted a subpopulation of oligodendrocytes activating astroglial genes after acute brain injuries. By using PLP-DsRed1/GFAP-EGFP and PLP-EGFPmem/GFAP-mRFP1 transgenic mice, we visualized this population of oligodendrocytes that we termed AO cells based on their concomitant activity of astro- and oligodendroglial genes. By fate mapping using PLP- and GFAP-split Cre complementation and repeated chronic in vivo imaging with two-photon laser-scanning microscopy, we observed the conversion of oligodendrocytes into astrocytes via the AO cell stage. Such conversion was promoted by local injection of IL-6 and was diminished by IL-6 receptor-neutralizing antibody as well as by inhibiting microglial activation with minocycline. In summary, our findings highlight the plastic potential of oligodendrocytes in acute brain trauma due to microglia-derived IL-6.
Collapse
Affiliation(s)
- Xianshu Bai
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, 66421 Homburg, Germany.
| | - Na Zhao
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, 66421 Homburg, Germany
| | - Christina Koupourtidou
- Department of Cell Biology and Anatomy, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany; Institute of Stem Cell Research, Helmholtz Zentrum Munich, 85764 Neuherberg-Munich, Germany
| | - Li-Pao Fang
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, 66421 Homburg, Germany
| | - Veronika Schwarz
- Department of Cell Biology and Anatomy, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany; Institute of Stem Cell Research, Helmholtz Zentrum Munich, 85764 Neuherberg-Munich, Germany
| | - Laura C Caudal
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, 66421 Homburg, Germany
| | - Renping Zhao
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, 66421 Homburg, Germany
| | - Johannes Hirrlinger
- Carl-Ludwig-Institute for Physiology, Leipzig University, 04103 Leipzig, Germany; Department of Neurogenetics, Max-Planck-Institute for Multidisciplinary Sciences, 37075 Göttingen, Germany
| | - Wolfgang Walz
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, 66421 Homburg, Germany; Department of Psychiatry, University of Saskatchewan, 107 Wiggins Road, Saskatoon, SK S7N 5E5, Canada
| | - Shan Bian
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, 200092 Shanghai, China; Frontier Science Center for Stem Cell Research, Tongji University, 200092 Shanghai, China
| | - Wenhui Huang
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, 66421 Homburg, Germany
| | - Jovica Ninkovic
- Department of Cell Biology and Anatomy, Ludwig-Maximilians-Universität München, 82152 Planegg-Martinsried, Germany; Institute of Stem Cell Research, Helmholtz Zentrum Munich, 85764 Neuherberg-Munich, Germany
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, 66421 Homburg, Germany; Experimental Research Center for Normal and Pathological Aging, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania
| | - Anja Scheller
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, 66421 Homburg, Germany.
| |
Collapse
|
18
|
Zhang H, Chen Y, Hua W, Gu W, Zhuang H, Li H, Jiang X, Mao Y, Liu Y, Jin D, Bu W. Heterostructures with Built-in Electric Fields for Long-lasting Chemodynamic Therapy. Angew Chem Int Ed Engl 2023; 62:e202300356. [PMID: 36780170 DOI: 10.1002/anie.202300356] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/14/2023]
Abstract
Sustained signal activation by hydroxyl radicals (⋅OH) has great significance, especially for tumor treatment, but remains challenging. Here, a built-in electric field (BIEF)-driven strategy was proposed for sustainable generation of ⋅OH, thereby achieving long-lasting chemodynamic therapy (LCDT). As a proof of concept, a novel Janus-like Fe@Fe3 O4 -Cu2 O heterogeneous catalyst was designed and synthesized, in which the BIEF induced the transfer of electrons in the Fe core to the surface, reducing ≡Cu2+ to ≡Cu+ , thus achieving continuous Fenton-like reactions and ⋅OH release for over 18 h, which is approximately 12 times longer than that of Fe3 O4 -Cu2 O and 72 times longer than that of Cu2 O nanoparticles. In vitro and in vivo antitumor results indicated that sustained ⋅OH levels led to persistent extracellular regulated protein kinases (ERK) signal activation and irreparable oxidative damage to tumor cells, which promoted irreversible tumor apoptosis. Importantly, this strategy provides ideas for developing long-acting nanoplatforms for various applications.
Collapse
Affiliation(s)
- Huilin Zhang
- Departments of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, P. R. China
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, P. R. China
| | - Yang Chen
- School of Life Sciences and Technology, Tongji University, Shanghai, 200092, P. R. China
| | - Wei Hua
- Departments of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, P. R. China
| | - Wenjun Gu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, P. R. China
| | - Hongjun Zhuang
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, P. R. China
- Departments of Rehabilitation, Zhongshan Hospital, Fudan University, Shanghai, 200032, P. R. China
| | - Huiyan Li
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, P. R. China
| | - Xingwu Jiang
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, P. R. China
| | - Ying Mao
- Departments of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, P. R. China
| | - Yanyan Liu
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, P. R. China
| | - Dayong Jin
- Institute for Biomedical Materials & Devices (IBMD), Faculty of Science, University of Technology Sydney, New South Wales, 2007, Australia
| | - Wenbo Bu
- Departments of Neurosurgery, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, 200040, P. R. China
- Department of Materials Science and State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai, 200438, P. R. China
| |
Collapse
|
19
|
Intisar A, Woo H, Kang HG, Kim WH, Shin HY, Kim MY, Kim YS, Mo YJ, Lee YI, Kim MS. Electroceutical approach ameliorates intracellular PMP22 aggregation and promotes pro-myelinating pathways in a CMT1A in vitro model. Biosens Bioelectron 2023; 224:115055. [PMID: 36630746 DOI: 10.1016/j.bios.2022.115055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/20/2022] [Accepted: 12/28/2022] [Indexed: 01/01/2023]
Abstract
Charcot-Marie-Tooth disease subtype 1A (CMT1A) is one of the most prevalent demyelinating peripheral neuropathies worldwide, caused by duplication of the peripheral myelin protein 22 (PMP22) gene, which is expressed primarily in Schwann cells (SCs). PMP22 overexpression in SCs leads to intracellular aggregation of the protein, which eventually results in demyelination. Unfortunately, previous biochemical approaches have not resulted in an approved treatment for CMT1A disease, compelling the pursuit for a biophysical approach such as electrical stimulation (ES). However, the effects of ES on CMT1A SCs have remained unexplored. In this study, we established PMP22-overexpressed Schwannoma cells as a CMT1A in vitro model, and investigated the biomolecular changes upon applying ES via a custom-made high-throughput ES platform, screening for the condition that delivers optimal therapeutic effects. While PMP22-overexpressed Schwannoma exhibited intracellular PMP22 aggregation, ES at 20 Hz for 1 h improved this phenomenon, bringing PMP22 distribution closer to healthy condition. ES at this condition also enhanced the expression of the genes encoding myelin basic protein (MBP) and myelin-associated glycoprotein (MAG), which are essential for assembling myelin sheath. Furthermore, ES altered the gene expression for myelination-regulating transcription factors Krox-20, Oct-6, c-Jun and Sox10, inducing pro-myelinating effects in PMP22-overexpressed Schwannoma. While electroceuticals has previously been applied in the peripheral nervous system towards acquired peripheral neuropathies such as pain and nerve injury, this study demonstrates its effectiveness towards ameliorating biomolecular abnormalities in an in vitro model of CMT1A, an inherited peripheral neuropathy. These findings will facilitate the clinical translation of an electroceutical treatment for CMT1A.
Collapse
Affiliation(s)
- Aseer Intisar
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | - Hanwoong Woo
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | - Hyun Gyu Kang
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | - Woon-Hae Kim
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea; CTCELLS Corp., Daegu, 42988, Republic of Korea
| | - Hyun Young Shin
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea; CTCELLS Corp., Daegu, 42988, Republic of Korea; SBCure Corp., Daegu, 43017, Republic of Korea
| | - Min Young Kim
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea
| | - Yu Seon Kim
- Well Aging Research Center, DGIST, Daegu, 42988, Republic of Korea
| | - Yun Jeoung Mo
- Well Aging Research Center, DGIST, Daegu, 42988, Republic of Korea
| | - Yun-Il Lee
- Well Aging Research Center, DGIST, Daegu, 42988, Republic of Korea
| | - Minseok S Kim
- Department of New Biology, DGIST, Daegu, 42988, Republic of Korea; CTCELLS Corp., Daegu, 42988, Republic of Korea; Translational Responsive Medicine Center (TRMC), DGIST, Daegu, 42988, Republic of Korea; New Biology Research Center (NBRC), DGIST, Daegu, 42988, Republic of Korea.
| |
Collapse
|
20
|
Liu M, He F, Shao M, Li T, Wang L, Wang Y, Xu W. PACAP inhibition alleviates neuropathic pain by modulating Nav1.7 through the MAPK/ERK signaling pathway in a rat model of chronic constriction injury. Neuropeptides 2023; 99:102327. [PMID: 36842389 DOI: 10.1016/j.npep.2023.102327] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 02/08/2023] [Accepted: 02/12/2023] [Indexed: 02/17/2023]
Abstract
BACKGROUND Trigeminal neuralgia is a common chronic maxillofacial neuropathic pain disorder, and voltage-gated sodium channels (VSGCs) are likely involved in its pathology. Prior studies report that pituitary adenylate cyclase-activating polypeptide (PACAP), a neuropeptide highly expressed in the trigeminal ganglion, may contribute to dorsal root ganglion neuron excitability by modulating the Nav1.7. OBJECTIVE We investigated whether PACAP can regulate Nav1.7 through the mitogen-activated protein kinase/ERK kinase/extracellular-signal-regulated kinase (MEK/ERK) pathway in the trigeminal ganglion after chronic constriction injury of the infraorbital nerve (ION-CCI) in rats. STUDY DESIGN Sprague-Dawley rats underwent ION-CCI, followed by intrathecal injection of PACAP 6-38 (PAC1 receptor antagonist) and PD98059 (MEK/ERK antagonist). Quantitative real-time PCR and western blot were used to quantify ATF3, PACAP, ERK, p-ERK, and Nav1.7 expression. RESULTS The mechanical pain threshold decreased from day 3 to day 21 after ION-CCI and reached the lowest testing value by day 14; however, it increased after PACAP 6-38 and PD98059 injections. Additionally, ION-CCI surgery increased ATF3, PACAP, and p-ERK expression in the rat trigeminal ganglion and decreased Nav1.7 and PAC1 receptor expression; however, there was no difference in ERK expression. PACAP 6-38 injection significantly decreased PACAP, p-ERK, and Nav1.7 expression and increased the PAC1 receptor expression, with no change in ERK expression. Moreover, PD98059 injection decreased PACAP, p-ERK, and Nav1.7 expression and increased the expression of PAC1 receptor. CONCLUSION After ION-CCI, PACAP in the rat trigeminal ganglion can modulate Nav1.7 through the MEK/ERK pathway via the PAC1 receptor. Further, PACAP inhibition alleviates allodynia in ION-CCI rats.
Collapse
Affiliation(s)
- Mingzheng Liu
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Fan He
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Mengci Shao
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Tianyuan Li
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China
| | - Liecheng Wang
- School of Basic Medical Sciences, Department of Physiology, Anhui Medical University, Hefei 230032, China
| | - Yuanyin Wang
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China.
| | - Wenhua Xu
- Stomatologic Hospital & College, Anhui Medical University, Key Lab. of Oral Diseases Research of Anhui Province, Hefei 230032, China.
| |
Collapse
|
21
|
Malong L, Napoli I, Casal G, White IJ, Stierli S, Vaughan A, Cattin AL, Burden JJ, Hng KI, Bossio A, Flanagan A, Zhao HT, Lloyd AC. Characterization of the structure and control of the blood-nerve barrier identifies avenues for therapeutic delivery. Dev Cell 2023; 58:174-191.e8. [PMID: 36706755 DOI: 10.1016/j.devcel.2023.01.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 10/26/2022] [Accepted: 01/04/2023] [Indexed: 01/27/2023]
Abstract
The blood barriers of the nervous system protect neural environments but can hinder therapeutic accessibility. The blood-brain barrier (BBB) is well characterized, consisting of endothelial cells with specialized tight junctions and low levels of transcytosis, properties conferred by contacting pericytes and astrocytes. In contrast, the blood-nerve barrier (BNB) of the peripheral nervous system is poorly defined. Here, we characterize the structure of the mammalian BNB, identify the processes that confer barrier function, and demonstrate how the barrier can be opened in response to injury. The homeostatic BNB is leakier than the BBB, which we show is due to higher levels of transcytosis. However, the barrier is reinforced by macrophages that specifically engulf leaked materials, identifying a role for resident macrophages as an important component of the BNB. Finally, we demonstrate the exploitation of these processes to effectively deliver RNA-targeting therapeutics to peripheral nerves, indicating new treatment approaches for nervous system pathologies.
Collapse
Affiliation(s)
- Liza Malong
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Ilaria Napoli
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Giulia Casal
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Ian J White
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Salome Stierli
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Andrew Vaughan
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Anne-Laure Cattin
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Jemima J Burden
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Keng I Hng
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Alessandro Bossio
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Adrienne Flanagan
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK
| | - Hien T Zhao
- IONIS, 2855 Gazelle Court, Carlsbad, CA 92010, USA
| | - Alison C Lloyd
- UCL Laboratory for Molecular Cell Biology and UCL Cancer Institute, University College London, Gower Street, London, WC1E 6BT, UK.
| |
Collapse
|
22
|
Park Y, Gaddy M, Hyun M, Jones ME, Aslam HM, Lee MH. Genetic and Chemical Controls of Sperm Fate and Spermatocyte Dedifferentiation via PUF-8 and MPK-1 in Caenorhabditis elegans. Cells 2023; 12:cells12030434. [PMID: 36766775 PMCID: PMC9913519 DOI: 10.3390/cells12030434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/21/2023] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
Using the nematode C. elegans germline as a model system, we previously reported that PUF-8 (a PUF RNA-binding protein) and LIP-1 (a dual-specificity phosphatase) repress sperm fate at 20 °C and the dedifferentiation of spermatocytes into mitotic cells (termed "spermatocyte dedifferentiation") at 25 °C. Thus, double mutants lacking both PUF-8 and LIP-1 produce excess sperm at 20 °C, and their spermatocytes return to mitotically dividing cells via dedifferentiation at 25 °C, resulting in germline tumors. To gain insight into the molecular competence for spermatocyte dedifferentiation, we compared the germline phenotypes of three mutant strains that produce excess sperm-fem-3(q20gf), puf-8(q725); fem-3(q20gf), and puf-8(q725); lip-1(zh15). Spermatocyte dedifferentiation was not observed in fem-3(q20gf) mutants, but it was more severe in puf-8(q725); lip-1(zh15) than in puf-8(q725); fem-3(q20gf) mutants. These results suggest that MPK-1 (the C. elegans ERK1/2 MAPK ortholog) activation in the absence of PUF-8 is required to promote spermatocyte dedifferentiation. This idea was confirmed using Resveratrol (RSV), a potential activator of MPK-1 and ERK1/2 in C. elegans and human cells, respectively. Notably, spermatocyte dedifferentiation was significantly enhanced by RSV treatment in the absence of PUF-8, and its effect was blocked by mpk-1 RNAi. We, therefore, conclude that PUF-8 and MPK-1 are essential regulators for spermatocyte dedifferentiation and tumorigenesis. Since these regulators are broadly conserved, we suggest that similar regulatory circuitry may control cellular dedifferentiation and tumorigenesis in other organisms, including humans.
Collapse
Affiliation(s)
- Youngyong Park
- Division of Hematology/Oncology, Department of Internal Medicine, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
| | - Matthew Gaddy
- Division of Hematology/Oncology, Department of Internal Medicine, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
| | - Moonjung Hyun
- Biological Resources Research Group, Bioenvironmental Science & Toxicology Division, Korea Institute of Toxicology, Jinju 52834, Gyeongsangnam-do, Republic of Korea
| | - Mariah E. Jones
- Division of Hematology/Oncology, Department of Internal Medicine, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
| | - Hafiz M. Aslam
- Division of Hematology/Oncology, Department of Internal Medicine, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
| | - Myon Hee Lee
- Division of Hematology/Oncology, Department of Internal Medicine, Brody School of Medicine at East Carolina University, Greenville, NC 27834, USA
- Department of Biology, East Carolina University, Greenville, NC 27858, USA
- Correspondence:
| |
Collapse
|
23
|
Yuan Y, Wang Y, Wu S, Zhao MY. Review: Myelin clearance is critical for regeneration after peripheral nerve injury. Front Neurol 2022; 13:908148. [PMID: 36588879 PMCID: PMC9801717 DOI: 10.3389/fneur.2022.908148] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 11/17/2022] [Indexed: 12/23/2022] Open
Abstract
Traumatic peripheral nerve injury occurs frequently and is a major clinical and public health problem that can lead to functional impairment and permanent disability. Despite the availability of modern diagnostic procedures and advanced microsurgical techniques, active recovery after peripheral nerve repair is often unsatisfactory. Peripheral nerve regeneration involves several critical events, including the recreation of the microenvironment and remyelination. Results from previous studies suggest that the peripheral nervous system (PNS) has a greater capacity for repair than the central nervous system. Thus, it will be important to understand myelin and myelination specifically in the PNS. This review provides an update on myelin biology and myelination in the PNS and discusses the mechanisms that promote myelin clearance after injury. The roles of Schwann cells and macrophages are considered at length, together with the possibility of exogenous intervention.
Collapse
Affiliation(s)
- YiMing Yuan
- Laboratory of Brain Function and Neurorehabilitation, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yan Wang
- Laboratory of Brain Function and Neurorehabilitation, Heilongjiang University of Chinese Medicine, Harbin, China,Department of Rehabilitation, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China,*Correspondence: Yan Wang
| | - ShanHong Wu
- Laboratory of Brain Function and Neurorehabilitation, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Ming Yue Zhao
- Laboratory of Brain Function and Neurorehabilitation, Heilongjiang University of Chinese Medicine, Harbin, China,Department of Rehabilitation, The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
24
|
Guo SW. Cracking the enigma of adenomyosis: an update on its pathogenesis and pathophysiology. Reproduction 2022; 164:R101-R121. [PMID: 36099328 DOI: 10.1530/rep-22-0224] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 09/12/2022] [Indexed: 11/08/2022]
Abstract
In brief Traditionally viewed as enigmatic and elusive, adenomyosis is a fairly common gynecological disease but is under-recognized and under-researched. This review summarizes the latest development on the pathogenesis and pathophysiology of adenomyosis, which have important implications for imaging diagnosis of the disease and for the development of non-hormonal therapeutics. Abstract Traditionally considered as an enigmatic disease, adenomyosis is a uterine disease that affects many women of reproductive age and is a contributing factor for pelvic pain, heavy menstrual bleeding (HMB), and subfertility. In this review, the new development in the pathogenesis and pathophysiology of adenomyosis has been summarized, along with their clinical implications. After reviewing the progress in our understanding of the pathogenesis and describing the prevailing theories, in conjunction with their deficiencies, a new hypothesis, called endometrial-myometrial interface disruption (EMID), which is backed by extensive epidemiologic data and demonstrated by a mouse model, is reviewed, along with recent data implicating the role of Schwann cells in the EMI area in the genesis of adenomyosis. Additionally, the natural history of adenomyotic lesions is elaborated and underscores that, in essence, adenomyotic lesions are fundamentally wounds undergoing repeated tissue injury and repair (ReTIAR), which progress to fibrosis through epithelial-mesenchymal transition, fibroblast-to-myofibroblast transdifferentiation, and smooth muscle metaplasia. Increasing lesional fibrosis propagates into the neighboring EMI and endometrium. The increased endometrial fibrosis, with ensuing greater tissue stiffness, results in attenuated prostaglandin E2, hypoxia signaling and glycolysis, impairing endometrial repair and causing HMB. Compared with adenomyosis-associated HMB, the mechanisms underlying adenomyosis-associated pain are less understood but presumably involve increased uterine contractility, hyperinnervation, increased lesional production of pain mediators, and central sensitization. Viewed through the prism of ReTIAR, a new imaging technique can be used to diagnose adenomyosis more accurately and informatively and possibly help to choose the best treatment modality.
Collapse
Affiliation(s)
- Sun-Wei Guo
- Shanghai Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China.,Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, China
| |
Collapse
|
25
|
Klymenko A, Lutz D. Melatonin signalling in Schwann cells during neuroregeneration. Front Cell Dev Biol 2022; 10:999322. [PMID: 36299487 PMCID: PMC9589221 DOI: 10.3389/fcell.2022.999322] [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: 07/20/2022] [Accepted: 09/23/2022] [Indexed: 11/13/2022] Open
Abstract
It has widely been thought that in the process of nerve regeneration Schwann cells populate the injury site with myelinating, non–myelinating, phagocytic, repair, and mesenchyme–like phenotypes. It is now clear that the Schwann cells modify their shape and basal lamina as to accommodate re–growing axons, at the same time clear myelin debris generated upon injury, and regulate expression of extracellular matrix proteins at and around the lesion site. Such a remarkable plasticity may follow an intrinsic functional rhythm or a systemic circadian clock matching the demands of accurate timing and precision of signalling cascades in the regenerating nervous system. Schwann cells react to changes in the external circadian clock clues and to the Zeitgeber hormone melatonin by altering their plasticity. This raises the question of whether melatonin regulates Schwann cell activity during neurorepair and if circadian control and rhythmicity of Schwann cell functions are vital aspects of neuroregeneration. Here, we have focused on different schools of thought and emerging concepts of melatonin–mediated signalling in Schwann cells underlying peripheral nerve regeneration and discuss circadian rhythmicity as a possible component of neurorepair.
Collapse
|
26
|
González-Cubero E, González-Fernández ML, Rodríguez-Díaz M, Palomo-Irigoyen M, Woodhoo A, Villar-Suárez V. Application of adipose-derived mesenchymal stem cells in an in vivo model of peripheral nerve damage. Front Cell Neurosci 2022; 16:992221. [PMID: 36159399 PMCID: PMC9493127 DOI: 10.3389/fncel.2022.992221] [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: 07/12/2022] [Accepted: 08/19/2022] [Indexed: 11/13/2022] Open
Abstract
Background Neuropathic pain is one of the most difficult to treat chronic pain syndromes. It has significant effects on patients’ quality of life and substantially adds to the burden of direct and indirect medical costs. There is a critical need to improve therapies for peripheral nerve regeneration. The aim of this study is to address this issue by performing a detailed analysis of the therapeutic benefits of two treatment options: adipose tissue derived-mesenchymal stem cells (ASCs) and ASC-conditioned medium (CM). Methods To this end, we used an in vivo rat sciatic nerve damage model to investigate the molecular mechanisms involved in the myelinating capacity of ASCs and CM. Furthermore, effect of TNF and CM on Schwann cells (SCs) was evaluated. For our in vivo model, biomaterial surgical implants containing TNF were used to induce peripheral neuropathy in rats. Damaged nerves were also treated with either ASCs or CM and molecular methods were used to collect evidence of nerve regeneration. Post-operatively, rats were subjected to walking track analysis and their sciatic functional index was evaluated. Morphological data was gathered through transmission electron microscopy (TEM) of sciatic nerves harvested from the experimental rats. We also evaluated the effect of TNF on Schwann cells (SCs) in vitro. Genes and their correspondent proteins associated with nerve regeneration were analyzed by qPCR, western blot, and confocal microscopy. Results Our data suggests that both ASCs and CM are potentially beneficial treatments for promoting myelination and axonal regeneration. After TNF-induced nerve damage we observed an upregulation of c-Jun along with a downregulation of Krox-20 myelin-associated transcription factor. However, when CM was added to TNF-treated nerves the opposite effect occurred and also resulted in increased expression of myelin-related genes and their corresponding proteins. Conclusion Findings from our in vivo model showed that both ASCs and CM aided the regeneration of axonal myelin sheaths and the remodeling of peripheral nerve morphology.
Collapse
Affiliation(s)
- Elsa González-Cubero
- Department of Anatomy, Faculty of Veterinary Sciences, University of León-Universidad de León, León, Spain
| | | | - María Rodríguez-Díaz
- Department of Anatomy, Faculty of Veterinary Sciences, University of León-Universidad de León, León, Spain
| | - Marta Palomo-Irigoyen
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
- Genes and Disease Group, Department of Dermatology, Medical University of Vienna, Anna Spiegel Center of Translational Research, Vienna, Austria
| | - Ashwin Woodhoo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, Derio, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Gene Regulatory Control in Disease Group, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain
| | - Vega Villar-Suárez
- Department of Anatomy, Faculty of Veterinary Sciences, University of León-Universidad de León, León, Spain
- Institute of Biomedicine (IBIOMED), University of León-Universidad de León, León, Spain
- *Correspondence: Vega Villar-Suárez,
| |
Collapse
|
27
|
McMorrow LA, Kosalko A, Robinson D, Saiani A, Reid AJ. Advancing Our Understanding of the Chronically Denervated Schwann Cell: A Potential Therapeutic Target? Biomolecules 2022; 12:1128. [PMID: 36009023 PMCID: PMC9406133 DOI: 10.3390/biom12081128] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 08/04/2022] [Accepted: 08/11/2022] [Indexed: 11/25/2022] Open
Abstract
Outcomes for patients following major peripheral nerve injury are extremely poor. Despite advanced microsurgical techniques, the recovery of function is limited by an inherently slow rate of axonal regeneration. In particular, a time-dependent deterioration in the ability of the distal stump to support axonal growth is a major determinant to the failure of reinnervation. Schwann cells (SC) are crucial in the orchestration of nerve regeneration; their plasticity permits the adoption of a repair phenotype following nerve injury. The repair SC modulates the initial immune response, directs myelin clearance, provides neurotrophic support and remodels the distal nerve. These functions are critical for regeneration; yet the repair phenotype is unstable in the setting of chronic denervation. This phenotypic instability accounts for the deteriorating regenerative support offered by the distal nerve stump. Over the past 10 years, our understanding of the cellular machinery behind this repair phenotype, in particular the role of c-Jun, has increased exponentially, creating opportunities for therapeutic intervention. This review will cover the activation of the repair phenotype in SC, the effects of chronic denervation on SC and current strategies to 'hack' these cellular pathways toward supporting more prolonged periods of neural regeneration.
Collapse
Affiliation(s)
- Liam A. McMorrow
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK
- Department of Plastic Surgery & Burns, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M23 9LT, UK
| | - Adrian Kosalko
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK
| | - Daniel Robinson
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK
| | - Alberto Saiani
- School of Materials & Manchester Institute of Biotechnology, Faculty of Science and Engineering, University of Manchester, Manchester M13 9PL, UK
| | - Adam J. Reid
- Blond McIndoe Laboratories, Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester M13 9PL, UK
- Department of Plastic Surgery & Burns, Wythenshawe Hospital, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M23 9LT, UK
| |
Collapse
|
28
|
Perioperative Suppression of Schwann Cell Dedifferentiation Reduces the Risk of Adenomyosis Resulting from Endometrial–Myometrial Interface Disruption in Mice. Biomedicines 2022; 10:biomedicines10061218. [PMID: 35740240 PMCID: PMC9219744 DOI: 10.3390/biomedicines10061218] [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: 04/28/2022] [Revised: 05/21/2022] [Accepted: 05/23/2022] [Indexed: 11/16/2022] Open
Abstract
We have recently demonstrated that endometrial–myometrial interface (EMI) disruption (EMID) can cause adenomyosis in mice, providing experimental evidence for the well-documented epidemiological finding that iatrogenic uterine procedures increase the risk of adenomyosis. To further elucidate its underlying mechanisms, we designed this study to test the hypothesis that Schwann cells (SCs) dedifferentiating after EMID facilitate the genesis of adenomyosis, but the suppression of SC dedifferentiation perioperatively reduces the risk. We treated mice perioperatively with either mitogen-activated protein kinase kinase (MEK)/extracellular-signal regulated protein kinase (ERK) or c-Jun N-terminal kinase (JNK) inhibitors or a vehicle 4 h before and 24 h, 48 h and 72 h after the EMID procedure. We found that EMID resulted in progressive SCs dedifferentiation, concomitant with an increased abundance of epithelial cells in the myometrium and a subsequent epithelial–mesenchymal transition (EMT). This EMID-induced change was abrogated significantly with perioperative administration of JNK or MEK/ERK inhibitors. Consistently, perioperative administration of a JNK or a MEK/ERK inhibitor reduced the incidence by nearly 33.5% and 14.3%, respectively, in conjunction with reduced myometrial infiltration of adenomyosis and alleviation of adenomyosis-associated hyperalgesia. Both treatments significantly decelerated the establishment of adenomyosis and progression of EMT, fibroblast-to-myofibroblast trans-differentiation and fibrogenesis in adenomyotic lesions. Thus, we provide the first piece of evidence strongly implicating the involvement of SCs in the pathogenesis of adenomyosis induced by EMID.
Collapse
|
29
|
Gundlach JP, Kerber J, Hendricks A, Bernsmeier A, Halske C, Röder C, Becker T, Röcken C, Braun F, Sebens S, Heits N. Paracrine Interaction of Cholangiocellular Carcinoma with Cancer-Associated Fibroblasts and Schwann Cells Impact Cell Migration. J Clin Med 2022; 11:jcm11102785. [PMID: 35628911 PMCID: PMC9145811 DOI: 10.3390/jcm11102785] [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: 04/13/2022] [Revised: 05/11/2022] [Accepted: 05/13/2022] [Indexed: 11/16/2022] Open
Abstract
Although the Mitogen-activated protein kinase (MAPK) pathway is enriched in cholangiocarcinoma (CCA), treatment with the multityrosine kinase-inhibitor Sorafenib is disappointing. While cancer-associated fibroblasts (CAF) are known to contribute to treatment resistance in CCA, knowledge is lacking for Schwann cells (SC). We investigated the impact of stromal cells on CCA cells and whether this is affected by Sorafenib. Immunohistochemistry revealed elevated expression of CAF and SC markers significantly correlating with reduced tumor-free survival. In co-culture with CAF, CCA cells mostly migrated, which could be diminished by Sorafenib, while in SC co-cultures, SC predominantly migrated towards CCA cells, unaffected by Sorafenib. Moreover, increased secretion of pro-inflammatory cytokines MCP-1, CXCL-1, IL-6 and IL-8 was determined in CAF mono- and co-cultures, which could be reduced by Sorafenib. Corresponding to migration results, an increased expression of phospho-AKT was measured in CAF co-cultured HuCCT-1 cells, although was unaffected by Sorafenib. Intriguingly, CAF co-cultured TFK-1 cells showed increased activation of STAT3, JNK, ERK and AKT pathways, which was partly reduced by Sorafenib. This study indicates that CAF and SC differentially impact CCA cells and Sorafenib partially reverts these stroma-mediated effects. These findings contribute to a better understanding of the paracrine interplay of CAF and SC with CCA cells.
Collapse
Affiliation(s)
- Jan-Paul Gundlach
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, Arnold-Heller-Str. 3, Building C, 24105 Kiel, Germany; (J.K.); (A.B.); (T.B.); (F.B.); (N.H.)
- Institute for Experimental Cancer Research, Kiel University and University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, Arnold-Heller-Str. 3, Building U30, 24105 Kiel, Germany; (C.R.); (S.S.)
- Correspondence:
| | - Jannik Kerber
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, Arnold-Heller-Str. 3, Building C, 24105 Kiel, Germany; (J.K.); (A.B.); (T.B.); (F.B.); (N.H.)
- Institute for Experimental Cancer Research, Kiel University and University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, Arnold-Heller-Str. 3, Building U30, 24105 Kiel, Germany; (C.R.); (S.S.)
| | - Alexander Hendricks
- Department of General, Visceral-, Vascular-, and Transplantation Surgery, Medical University Rostock, Schillingallee 35, 18057 Rostock, Germany;
| | - Alexander Bernsmeier
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, Arnold-Heller-Str. 3, Building C, 24105 Kiel, Germany; (J.K.); (A.B.); (T.B.); (F.B.); (N.H.)
| | - Christine Halske
- Institute of Pathology, University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, Arnold-Heller-Str. 3, Building U33, 24105 Kiel, Germany; (C.H.); (C.R.)
| | - Christian Röder
- Institute for Experimental Cancer Research, Kiel University and University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, Arnold-Heller-Str. 3, Building U30, 24105 Kiel, Germany; (C.R.); (S.S.)
| | - Thomas Becker
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, Arnold-Heller-Str. 3, Building C, 24105 Kiel, Germany; (J.K.); (A.B.); (T.B.); (F.B.); (N.H.)
| | - Christoph Röcken
- Institute of Pathology, University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, Arnold-Heller-Str. 3, Building U33, 24105 Kiel, Germany; (C.H.); (C.R.)
| | - Felix Braun
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, Arnold-Heller-Str. 3, Building C, 24105 Kiel, Germany; (J.K.); (A.B.); (T.B.); (F.B.); (N.H.)
| | - Susanne Sebens
- Institute for Experimental Cancer Research, Kiel University and University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, Arnold-Heller-Str. 3, Building U30, 24105 Kiel, Germany; (C.R.); (S.S.)
| | - Nils Heits
- Department of General, Visceral-, Thoracic-, Transplantation- and Pediatric Surgery, University Medical Center Schleswig-Holstein (UKSH), Campus Kiel, Arnold-Heller-Str. 3, Building C, 24105 Kiel, Germany; (J.K.); (A.B.); (T.B.); (F.B.); (N.H.)
- Gesundheitszentrum Kiel-Mitte, Prüner Gang 15, 24103 Kiel, Germany
| |
Collapse
|
30
|
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
|
31
|
Su Q, Nasser MI, He J, Deng G, Ouyang Q, Zhuang D, Deng Y, Hu H, Liu N, Li Z, Zhu P, Li G. Engineered Schwann Cell-Based Therapies for Injury Peripheral Nerve Reconstruction. Front Cell Neurosci 2022; 16:865266. [PMID: 35602558 PMCID: PMC9120533 DOI: 10.3389/fncel.2022.865266] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 04/04/2022] [Indexed: 12/12/2022] Open
Abstract
Compared with the central nervous system, the adult peripheral nervous system possesses a remarkable regenerative capacity, which is due to the strong plasticity of Schwann cells (SCs) in peripheral nerves. After peripheral nervous injury, SCs de-differentiate and transform into repair phenotypes, and play a critical role in axonal regeneration, myelin formation, and clearance of axonal and myelin debris. In view of the limited self-repair capability of SCs for long segment defects of peripheral nerve defects, it is of great clinical value to supplement SCs in necrotic areas through gene modification or stem cell transplantation or to construct tissue-engineered nerve combined with bioactive scaffolds to repair such tissue defects. Based on the developmental lineage of SCs and the gene regulation network after peripheral nerve injury (PNI), this review summarizes the possibility of using SCs constructed by the latest gene modification technology to repair PNI. The therapeutic effects of tissue-engineered nerve constructed by materials combined with Schwann cells resembles autologous transplantation, which is the gold standard for PNI repair. Therefore, this review generalizes the research progress of biomaterials combined with Schwann cells for PNI repair. Based on the difficulty of donor sources, this review also discusses the potential of “unlimited” provision of pluripotent stem cells capable of directing differentiation or transforming existing somatic cells into induced SCs. The summary of these concepts and therapeutic strategies makes it possible for SCs to be used more effectively in the repair of PNI.
Collapse
Affiliation(s)
- Qisong Su
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial People’s Hospital, Guangdong Cardiovascular Institute, Guangzhou, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Moussa Ide Nasser
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial People’s Hospital, Guangdong Cardiovascular Institute, Guangzhou, China
| | - Jiaming He
- School of Basic Medical Science, Shandong University, Jinan, China
| | - Gang Deng
- Guangdong Provincial People’s Hospital, Guangdong Cardiovascular Institute, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Qing Ouyang
- Guangdong Provincial People’s Hospital, Guangdong Cardiovascular Institute, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Donglin Zhuang
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yuzhi Deng
- Guangdong Provincial People’s Hospital, Guangdong Cardiovascular Institute, Guangzhou, China
- The First Clinical College, Guangdong Medical University, Zhanjiang, China
| | - Haoyun Hu
- Guangdong Provincial People’s Hospital, Guangdong Cardiovascular Institute, Guangzhou, China
- The First Clinical College, Guangdong Medical University, Zhanjiang, China
| | - Nanbo Liu
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
| | - Zhetao Li
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Ping Zhu
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial People’s Hospital, Guangdong Cardiovascular Institute, Guangzhou, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
- The First Clinical College, Guangdong Medical University, Zhanjiang, China
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Structural Heart Disease, Guangzhou, China
- *Correspondence: Ping Zhu,
| | - Ge Li
- Medical Research Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Guangdong Provincial People’s Hospital, Guangdong Cardiovascular Institute, Guangzhou, China
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Structural Heart Disease, Guangzhou, China
- Ge Li,
| |
Collapse
|
32
|
Sahu R, Mehan S, Kumar S, Prajapati A, Alshammari A, Alharbi M, Assiri MA, Narula AS. Effect of alpha-mangostin in the prevention of behavioural and neurochemical defects in methylmercury-induced neurotoxicity in experimental rats. Toxicol Rep 2022; 9:977-998. [PMID: 35783250 PMCID: PMC9247835 DOI: 10.1016/j.toxrep.2022.04.023] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 04/17/2022] [Accepted: 04/20/2022] [Indexed: 12/11/2022] Open
Abstract
Methylmercury (MeHg+) is a known neurotoxin that causes progressive motor neuron degeneration in the central nervous system. Axonal degeneration, oligodendrocyte degeneration, and myelin basic protein (MBP) deficits are among the neuropathological abnormalities caused by MeHg+ in amyotrophic lateral sclerosis (ALS). This results in demyelination and motor neuron death in both humans and animals. Previous experimental studies have confirmed that overexpression of the extracellular signalling regulated kinase (ERK1/2) signalling contributes to glutamate excitotoxicity, inflammatory response of microglial cells, and oligodendrocyte (OL) dysfunction that promotes myelin loss. Alpha-mangostin (AMG), an active ingredient obtained from the tree "Garcinia mangostana Linn," has been used in experimental animals to treat a variety of brain disorders, including Parkinson's and Huntington's disease memory impairment, Alzheimer's disease, and schizophrenia, including Parkinson's disease and Huntington's disease memory impairment, Alzheimer's disease, and schizophrenia. AMG has traditionally been used as an antioxidant, anti-inflammatory, and neuroprotective agent.Accordingly, we investigated the therapeutic potential of AMG (100 and 200 mg/kg) in experimental rats with methylmercury (MeHg+)-induced neurotoxicity. The neuroprotective effect of AMG on behavioural, cellular, molecular, and other gross pathological changes, such as histopathological alterations in MeHg+ -treated rat brains, is presented. The neurological behaviour of experimental rats was evaluated using a Morris water maze (MWM), open field test (OFT), grip strength test (GST), and force swim test (FST). In addition, we investigate AMG's neuroprotective effect by restoring MBP levels in cerebral spinal fluid and whole rat brain homogenate. The apoptotic, pro-inflammatory, and oxidative stress markers were measured in rat blood plasma samples and brain homogenate. According to the findings of this study, AMG decreases ERK-1/2 levels and modulates neurochemical alterations in rat brains, minimising MeHg+ -induced neurotoxicity.
Collapse
Affiliation(s)
- Rakesh Sahu
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sumit Kumar
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Aradhana Prajapati
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Abdulrahman Alshammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh 11451, Saudi Arabia
| | - Metab Alharbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh 11451, Saudi Arabia
| | - Mohammed A. Assiri
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Post Box 2455, Riyadh 11451, Saudi Arabia
| | | |
Collapse
|
33
|
English AW, Carrasco D, Hoffman D, Isaacson R, Kang SS, Khan S, Liu X, Ye K. Oral Treatments With the TrkB Ligand Prodrug, R13, Promote Enhanced Axon Regeneration Following Peripheral Nerve Injury. Front Cell Neurosci 2022; 16:857664. [PMID: 35496909 PMCID: PMC9051483 DOI: 10.3389/fncel.2022.857664] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Accepted: 03/24/2022] [Indexed: 11/16/2022] Open
Abstract
Axon regeneration after peripheral nerve injury is slow and inefficient, leading to generally poor functional recovery. Activity-dependent experimental therapies that increase expression of brain-derived neurotrophic factor (BDNF) and its TrkB receptors enhance regeneration, suggesting that treatments with BDNF might also be effective. However, recombinant human BDNF (rhBDNF), as well as 7,8-dihydroxyflavone (7,8-DHF), a small molecular BDNF mimetic, may have limited treatment applications because of their modest oral bioavailability and pharmacokinetic profile. R13 is a 7,8-DHF prodrug. Upon oral administration, it is converted in the liver to 7,8-DHF. In immunoblots from tissues at the site of nerve injury, a single oral treatment with R13 to mice following sciatic nerve transection and repair produced a rapid and prolonged increase in immunoreactivity to phosphorylated TrkB, prolonged phosphorylation of mitogen activated protein kinase (MAPK/Erk1/2), and a rapid but transient increase in phosphorylated AKT (protein kinase B). Intramuscular injections of fluorescent retrograde tracers into the gastrocnemius and tibialis anterior muscles 4 weeks after nerve injury resulted in significantly greater numbers of labeled motoneurons and dorsal root ganglion neurons in R13-treated mice than in vehicle-treated controls. Direct electromyographic (EMG) responses (M waves) were significantly larger in R13-treated mice 4 weeks after injury than vehicle-treated controls or mice treated with oral 7,8-DHF. Oral treatments with the prodrug, R13, are a potent therapy for stimulating axon regeneration and functional recovery after peripheral nerve injury.
Collapse
Affiliation(s)
- Arthur W. English
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
- Rehabilitation Medicine, Emory University School of Medicine, Atlanta, GA, United States
- *Correspondence: Arthur W. English,
| | - Dario Carrasco
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Dustin Hoffman
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Robin Isaacson
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Seong Su Kang
- Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Samia Khan
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, United States
| | - Xia Liu
- Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States
| | - Keqiang Ye
- Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, United States
- Faculty of Life and Health Sciences, Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
34
|
Gong Y, Jiang X, Yang S, Huang Y, Hong J, Ma Y, Fang X, Fang Y, Wu J. The Biological Activity of 3-O-Acetyl-11-keto-β-Boswellic Acid in Nervous System Diseases. Neuromolecular Med 2022; 24:374-384. [PMID: 35303275 PMCID: PMC8931781 DOI: 10.1007/s12017-022-08707-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 03/02/2022] [Indexed: 12/29/2022]
Abstract
Frankincense is a hard gelatinous resin exuded by Boswellia serrata. It contains a complex array of components, of which acetyl-11-keto-beta-boswellic acid (AKBA), a pentacyclic triterpenoid of the resin class, is the main active component. AKBA has a variety of physiological actions, including anti-infection, anti-tumor, and antioxidant effects. The use of AKBA for the treatment of mental diseases has been documented as early as ancient Greece. Recent studies have found that AKBA has anti-aging and other neurological effects, suggesting its potential for the treatment of neurological diseases. This review focuses on nervous system-related diseases, summarizes the functions and mechanisms of AKBA in promoting nerve repair and regeneration after injury, protecting against ischemic brain injury and aging, inhibiting neuroinflammation, ameliorating memory deficits, and alleviating neurotoxicity, as well as having anti-glioma effects and relieving brain edema. The mechanisms by which AKBA functions in different diseases and the relationships between dosage and biological effects are discussed in depth with the aim of increasing understanding of AKBA and guiding its use for the treatment of nervous system diseases.
Collapse
Affiliation(s)
- Yuqing Gong
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, and Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, No. 2318, Yuhang Tang Road, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Xinyi Jiang
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, and Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, No. 2318, Yuhang Tang Road, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Suibi Yang
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, and Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, No. 2318, Yuhang Tang Road, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Yue Huang
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, and Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, No. 2318, Yuhang Tang Road, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Jinhui Hong
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, and Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, No. 2318, Yuhang Tang Road, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Yanxiu Ma
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, and Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, No. 2318, Yuhang Tang Road, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Xin Fang
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, and Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, No. 2318, Yuhang Tang Road, Hangzhou, 310000, Zhejiang, People's Republic of China
| | - Yong Fang
- Department of Microbiology, WU Lien-Teh Institute, Harbin Medical University, Harbin, 150081, China.
| | - Jing Wu
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, and Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, No. 2318, Yuhang Tang Road, Hangzhou, 310000, Zhejiang, People's Republic of China.
| |
Collapse
|
35
|
Wen X, Jiao L, Tan H. MAPK/ERK Pathway as a Central Regulator in Vertebrate Organ Regeneration. Int J Mol Sci 2022; 23:ijms23031464. [PMID: 35163418 PMCID: PMC8835994 DOI: 10.3390/ijms23031464] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/24/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023] Open
Abstract
Damage to organs by trauma, infection, diseases, congenital defects, aging, and other injuries causes organ malfunction and is life-threatening under serious conditions. Some of the lower order vertebrates such as zebrafish, salamanders, and chicks possess superior organ regenerative capacity over mammals. The extracellular signal-regulated kinases 1 and 2 (ERK1/2), as key members of the mitogen-activated protein kinase (MAPK) family, are serine/threonine protein kinases that are phylogenetically conserved among vertebrate taxa. MAPK/ERK signaling is an irreplaceable player participating in diverse biological activities through phosphorylating a broad variety of substrates in the cytoplasm as well as inside the nucleus. Current evidence supports a central role of the MAPK/ERK pathway during organ regeneration processes. MAPK/ERK signaling is rapidly excited in response to injury stimuli and coordinates essential pro-regenerative cellular events including cell survival, cell fate turnover, migration, proliferation, growth, and transcriptional and translational activities. In this literature review, we recapitulated the multifaceted MAPK/ERK signaling regulations, its dynamic spatio-temporal activities, and the profound roles during multiple organ regeneration, including appendages, heart, liver, eye, and peripheral/central nervous system, illuminating the possibility of MAPK/ERK signaling as a critical mechanism underlying the vastly differential regenerative capacities among vertebrate species, as well as its potential applications in tissue engineering and regenerative medicine.
Collapse
|
36
|
Han W, Pan Y, Han Z, Cheng L, Jiang L. Advanced Maternal Age Impairs Myelination in Offspring Rats. Front Pediatr 2022; 10:850213. [PMID: 35311052 PMCID: PMC8927774 DOI: 10.3389/fped.2022.850213] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 02/07/2022] [Indexed: 11/13/2022] Open
Abstract
The effects of advanced maternal age (AMA) on the neurodevelopment of offspring are becoming increasingly important. Myelination is an important aspect of brain development; however, a limited number of studies have focused on the effects of AMA on myelination in offspring. The current study aims to evaluate the association between AMA and myelin sheath development in offspring. We studied the learning and memory function of immature offspring using the novel object recognition test. Then, we investigated the expression of myelin basic protein (MBP) in the immature offspring of young (3-month-old) and old (12-month-old) female rats at different time points (14, 28, and 60 days) after birth with immunofluorescence and western blotting. The myelin sheath ultrastructure was observed with transmission electron microscopy in immature and mature offspring. Extracellular signal-regulated kinase 1 and 2 (ERK1/2) and phosphorylated ERK1/2 (p-ERK1/2) were investigated by western blot in immature offspring at the above time points. AMA impaired the memory function of offspring during early postnatal days. The MBP expression level gradually increased with postnatal development in the offspring of both the AMA and Control (Ctl) groups, but the MBP level in the offspring of the AMA group was lower than that of the Ctl group at 14 days after birth. In addition, the ultrastructure of the myelin sheath was defective in AMA offspring during the early postnatal period; however, the myelin sheath was not significantly affected in offspring during adulthood. Interestingly, ERK phosphorylation at 14 days after birth was lower in AMA offspring than in Ctl offspring. However, ERK phosphorylation at 28 days after birth was higher in AMA offspring than in Ctl offspring. The peak of ERK phosphorylation in the AMA group was abnormal and delayed. Our results indicated that AMA is associated with poor developmental myelin formation in offspring. The ERK signaling pathway may play an essential role in the adverse effects of AMA on the offspring myelin sheath development.
Collapse
Affiliation(s)
- Wei Han
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Ya'nan Pan
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Ziyao Han
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Li Cheng
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| | - Li Jiang
- Department of Neurology Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
37
|
Li B, Tang H, Bian X, Ma K, Chang J, Fu X, Zhang C. Calcium silicate accelerates cutaneous wound healing with enhanced re-epithelialization through EGF/EGFR/ERK-mediated promotion of epidermal stem cell functions. BURNS & TRAUMA 2021; 9:tkab029. [PMID: 34604395 PMCID: PMC8484206 DOI: 10.1093/burnst/tkab029] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 11/23/2020] [Accepted: 06/16/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND Human epidermal stem cells (hESCs) play an important role in re-epithelialization and thereby in facilitating wound healing, while an effective way to activate hESCs remains to be explored. Calcium silicate (CS) is a form of bioceramic that can alter cell behavior and promote tissue regeneration. Here, we have observed the effect of CS on hESCs and investigated its possible mechanism. METHODS Using a mouse full-thickness skin excision model, we explored the therapeutic effect of CS on wound healing and re-epithelialization. In vitro, hESCs were cultured with diluted CS ion extracts (CSIEs), and the proliferation, migration ability and stemness of hESCs were evaluated. The effects of CS on the epidermal growth factor (EGF), epidermal growth factor receptor (EGFR) and extracellular signal-related kinase (ERK) signaling pathway were also explored. RESULTS In vivo, CS accelerated wound healing and re-epithelialization. Immunohistochemistry demonstrated that CS upregulated cytokeratin 19 and integrin β1 expression, indicating that CS improved hESCs stemness. In vitro studies confirmed that CS improved the biological function of hESCs. And the possible mechanism could be due to the activation of the EGF/EGFR/ERK signaling pathway. CONCLUSION CS can promote re-epithelialization and improve the biological functions of hESCs via activating the EGF/EGFR/ERK signaling pathway.
Collapse
Affiliation(s)
- Bingmin Li
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Division and Fourth Medical Center of Chinese PLA General Hospital, 100048, Beijing, China
- Department of Dermatology, Fourth Medical Center of Chinese PLA General Hospital, 100048, Beijing, China
| | - Haowen Tang
- Faculty of Hepato-Biliary-Pancreatic Surgery, Chinese PLA General Hospital, 100853, Beijing, China
| | - Xiaowei Bian
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Division and Fourth Medical Center of Chinese PLA General Hospital, 100048, Beijing, China
| | - Kui Ma
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Division and Fourth Medical Center of Chinese PLA General Hospital, 100048, Beijing, China
| | - Jiang Chang
- Shanghai Institute of Ceramics, Chinese Academy of Sciences, 200050, Shanghai, China
| | - Xiaobing Fu
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Division and Fourth Medical Center of Chinese PLA General Hospital, 100048, Beijing, China
- Research Unit of Trauma Care, Tissue Repair and Regeneration, Chinese Academy of Medical Sciences, 100048, Beijing, China
| | - Cuiping Zhang
- Research Center for Tissue Repair and Regeneration affiliated to the Medical Innovation Research Division and Fourth Medical Center of Chinese PLA General Hospital, 100048, Beijing, China
| |
Collapse
|
38
|
Sahu R, Upadhayay S, Mehan S. Inhibition of extracellular regulated kinase (ERK)-1/2 signaling pathway in the prevention of ALS: Target inhibitors and influences on neurological dysfunctions. Eur J Cell Biol 2021; 100:151179. [PMID: 34560374 DOI: 10.1016/j.ejcb.2021.151179] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/18/2021] [Accepted: 09/15/2021] [Indexed: 12/13/2022] Open
Abstract
Cell signal transduction pathways are essential modulators of several physiological and pathological processes in the brain. During overactivation, these signaling processes may lead to disease progression. Abnormal protein kinase activation is associated with several biological dysfunctions that facilitate neurodegeneration under different biological conditions. As a result, these signaling pathways are essential in understanding brain disorders' development or progression. Recent research findings indicate the crucial role of extracellular signal-regulated kinase-1/2 (ERK-1/2) signaling during the neuronal development process. ERK-1/2 is a key component of its mitogen-activated protein kinase (MAPK) group, controlling certain neurological activities by regulating metabolic pathways, cell proliferation, differentiation, and apoptosis. ERK-1/2 also influences neuronal elastic properties, nerve growth, and neurological and cognitive processing during brain injuries. The primary goal of this review is to elucidate the activation of ERK1/2 signaling, which is involved in the development of several ALS-related neuropathological dysfunctions. ALS is a rare neurological disorder category that mainly affects the nerve cells responsible for regulating voluntary muscle activity. ALS is progressive, which means that the symptoms are getting worse over time, and there is no cure for ALS and no effective treatment to avoid or reverse. Genetic abnormalities, oligodendrocyte degradation, glial overactivation, and immune deregulation are associated with ALS progression. Furthermore, the current review also identifies ERK-1/2 signaling inhibitors that can promote neuroprotection and neurotrophic effects against the clinical-pathological presentation of ALS. As a result, in the future, the potential ERK-1/2 signaling inhibitors could be used in the treatment of ALS and related neurocomplications.
Collapse
Affiliation(s)
- Rakesh Sahu
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Shubham Upadhayay
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India.
| |
Collapse
|
39
|
Yow YY, Goh TK, Nyiew KY, Lim LW, Phang SM, Lim SH, Ratnayeke S, Wong KH. Therapeutic Potential of Complementary and Alternative Medicines in Peripheral Nerve Regeneration: A Systematic Review. Cells 2021; 10:cells10092194. [PMID: 34571842 PMCID: PMC8472132 DOI: 10.3390/cells10092194] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 08/20/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
Despite the progressive advances, current standards of treatments for peripheral nerve injury do not guarantee complete recovery. Thus, alternative therapeutic interventions should be considered. Complementary and alternative medicines (CAMs) are widely explored for their therapeutic value, but their potential use in peripheral nerve regeneration is underappreciated. The present systematic review, designed according to guidelines of Preferred Reporting Items for Systematic Review and Meta-Analysis Protocols, aims to present and discuss the current literature on the neuroregenerative potential of CAMs, focusing on plants or herbs, mushrooms, decoctions, and their respective natural products. The available literature on CAMs associated with peripheral nerve regeneration published up to 2020 were retrieved from PubMed, Scopus, and Web of Science. According to current literature, the neuroregenerative potential of Achyranthes bidentata, Astragalus membranaceus, Curcuma longa, Panax ginseng, and Hericium erinaceus are the most widely studied. Various CAMs enhanced proliferation and migration of Schwann cells in vitro, primarily through activation of MAPK pathway and FGF-2 signaling, respectively. Animal studies demonstrated the ability of CAMs to promote peripheral nerve regeneration and functional recovery, which are partially associated with modulations of neurotrophic factors, pro-inflammatory cytokines, and anti-apoptotic signaling. This systematic review provides evidence for the potential use of CAMs in the management of peripheral nerve injury.
Collapse
Affiliation(s)
- Yoon-Yen Yow
- Department of Biological Sciences, School of Medicine and Life Sciences, Sunway University, Petaling Jaya 47500, Malaysia; (T.-K.G.); (K.-Y.N.); (S.R.)
- Correspondence: (Y.-Y.Y.); (L.-W.L.); (K.-H.W.); Tel.: +603-7491-8622 (Y.-Y.Y.); +852-3917-6830 (L.-W.L.); +603-7967-4729 (K.-H.W.)
| | - Tiong-Keat Goh
- Department of Biological Sciences, School of Medicine and Life Sciences, Sunway University, Petaling Jaya 47500, Malaysia; (T.-K.G.); (K.-Y.N.); (S.R.)
| | - Ke-Ying Nyiew
- Department of Biological Sciences, School of Medicine and Life Sciences, Sunway University, Petaling Jaya 47500, Malaysia; (T.-K.G.); (K.-Y.N.); (S.R.)
| | - Lee-Wei Lim
- Neuromodulation Laboratory, School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 21 Sassoon Road, L4 Laboratory Block, Hong Kong
- Correspondence: (Y.-Y.Y.); (L.-W.L.); (K.-H.W.); Tel.: +603-7491-8622 (Y.-Y.Y.); +852-3917-6830 (L.-W.L.); +603-7967-4729 (K.-H.W.)
| | - Siew-Moi Phang
- Institute of Ocean and Earth Sciences, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
- Faculty of Applied Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia
| | - Siew-Huah Lim
- Department of Chemistry, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia;
| | - Shyamala Ratnayeke
- Department of Biological Sciences, School of Medicine and Life Sciences, Sunway University, Petaling Jaya 47500, Malaysia; (T.-K.G.); (K.-Y.N.); (S.R.)
| | - Kah-Hui Wong
- Department of Anatomy, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
- Correspondence: (Y.-Y.Y.); (L.-W.L.); (K.-H.W.); Tel.: +603-7491-8622 (Y.-Y.Y.); +852-3917-6830 (L.-W.L.); +603-7967-4729 (K.-H.W.)
| |
Collapse
|
40
|
Park K, Shin Y, Lee G, Park H, Choi Y. Dabrafenib Promotes Schwann Cell Differentiation by Inhibition of the MEK-ERK Pathway. Molecules 2021; 26:2141. [PMID: 33917810 PMCID: PMC8068149 DOI: 10.3390/molecules26082141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 04/04/2021] [Accepted: 04/07/2021] [Indexed: 11/16/2022] Open
Abstract
Schwann cell differentiation involves a dynamic interaction of signaling cascades. However, much remains to be elucidated regarding the function of signaling molecules that differ depending on the context in which the molecules are engaged. Here, we identified a small molecule, dabrafenib, which promotes Schwann cell differentiation in vitro and exploited this compound as a pharmacological tool to understand the molecular mechanisms regulating Schwann cell differentiation. The results indicated that dabrafenib inhibited ERK phosphorylation and enhanced ErbB2 autophosphorylation and Akt phosphorylation, and the effects of dabrafenib on ErbB2 and Akt phosphorylation were phenocopied by pharmacological inhibition of the MEK-ERK signaling pathway. However, the small molecule inhibitors of MEK and ERK had no effect on the expression of Oct6 and EGR2, which are key transcription factors that drive Schwann cell differentiation. In addition, pharmacological inhibition of phosphatidylinositol-3-kinase (PI3K) almost completely interfered with dabrafenib-induced Schwann cell differentiation. These results suggest that the ErbB2-PI3K-Akt axis is required for the induction of Schwann cell differentiation by dabrafenib in vitro. Although additional molecules targeted by dabrafenib remain to be identified, our data provides insights into the crosstalk that exists between the MEK-ERK signaling pathway and the PI3K-Akt axis in Schwann cell differentiation.
Collapse
Affiliation(s)
- Kyuhee Park
- Biocenter, Gyeonggido Business and Science Accelerator, Suwon 16229, Korea; (K.P.); (G.L.)
| | - Yoonkyoung Shin
- Department of Physiology, Peripheral Neuropathy Research Center, Donga University Medical School, Busan 49201, Korea; (Y.S.); (H.P.)
| | - Gyeongbeen Lee
- Biocenter, Gyeonggido Business and Science Accelerator, Suwon 16229, Korea; (K.P.); (G.L.)
| | - Hwantae Park
- Department of Physiology, Peripheral Neuropathy Research Center, Donga University Medical School, Busan 49201, Korea; (Y.S.); (H.P.)
| | - Yongmun Choi
- Biocenter, Gyeonggido Business and Science Accelerator, Suwon 16229, Korea; (K.P.); (G.L.)
| |
Collapse
|
41
|
Pituitary Adenylate Cyclase-Activating Polypeptide: A Potent Therapeutic Agent in Oxidative Stress. Antioxidants (Basel) 2021; 10:antiox10030354. [PMID: 33653014 PMCID: PMC7996859 DOI: 10.3390/antiox10030354] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/13/2021] [Accepted: 02/22/2021] [Indexed: 12/13/2022] Open
Abstract
Stroke is a life-threatening condition that is characterized by secondary cell death processes that occur after the initial disruption of blood flow to the brain. The inability of endogenous repair mechanisms to sufficiently support functional recovery in stroke patients and the inadequate treatment options available are cause for concern. The pathology behind oxidative stress in stroke is of particular interest due to its detrimental effects on the brain. The oxidative stress caused by ischemic stroke overwhelms the neutralization capacity of the body's endogenous antioxidant system, which leads to an overproduction of reactive oxygen species (ROS) and reactive nitrogen species (RNS) and eventually results in cell death. The overproduction of ROS compromises the functional and structural integrity of brain tissue. Therefore, it is essential to investigate the mechanisms involved in oxidative stress to help obtain adequate treatment options for stroke. Here, we focus on the latest preclinical research that details the mechanisms behind secondary cell death processes that cause many central nervous system (CNS) disorders, as well as research that relates to how the neuroprotective molecular mechanisms of pituitary adenylate cyclase-activating polypeptides (PACAPs) could make these molecules an ideal candidate for the treatment of stroke.
Collapse
|
42
|
Fornaro M, Marcus D, Rattin J, Goral J. Dynamic Environmental Physical Cues Activate Mechanosensitive Responses in the Repair Schwann Cell Phenotype. Cells 2021; 10:cells10020425. [PMID: 33671410 PMCID: PMC7922665 DOI: 10.3390/cells10020425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/09/2021] [Accepted: 02/11/2021] [Indexed: 01/10/2023] Open
Abstract
Schwann cells plastically change in response to nerve injury to become a newly reconfigured repair phenotype. This cell is equipped to sense and interact with the evolving and unusual physical conditions characterizing the injured nerve environment and activate intracellular adaptive reprogramming as a consequence of external stimuli. Summarizing the literature contributions on this matter, this review is aimed at highlighting the importance of the environmental cues of the regenerating nerve as key factors to induce morphological and functional changes in the Schwann cell population. We identified four different microenvironments characterized by physical cues the Schwann cells sense via interposition of the extracellular matrix. We discussed how the physical cues of the microenvironment initiate changes in Schwann cell behavior, from wrapping the axon to becoming a multifunctional denervated repair cell and back to reestablishing contact with regenerated axons.
Collapse
Affiliation(s)
- Michele Fornaro
- Department of Anatomy, College of Graduate Studies (CGS), Midwestern University, Downers Grove, IL 60515, USA;
- Department of Anatomy, Chicago College of Osteopathic Medicine (CCOM), Midwestern University, Downers Grove, IL 60515, USA; (D.M.); (J.R.)
- Correspondence: ; Tel.: +001-630-515-6055
| | - Dominic Marcus
- Department of Anatomy, Chicago College of Osteopathic Medicine (CCOM), Midwestern University, Downers Grove, IL 60515, USA; (D.M.); (J.R.)
| | - Jacob Rattin
- Department of Anatomy, Chicago College of Osteopathic Medicine (CCOM), Midwestern University, Downers Grove, IL 60515, USA; (D.M.); (J.R.)
| | - Joanna Goral
- Department of Anatomy, College of Graduate Studies (CGS), Midwestern University, Downers Grove, IL 60515, USA;
- Department of Anatomy, Chicago College of Osteopathic Medicine (CCOM), Midwestern University, Downers Grove, IL 60515, USA; (D.M.); (J.R.)
| |
Collapse
|
43
|
Balakrishnan A, Belfiore L, Chu TH, Fleming T, Midha R, Biernaskie J, Schuurmans C. Insights Into the Role and Potential of Schwann Cells for Peripheral Nerve Repair From Studies of Development and Injury. Front Mol Neurosci 2021; 13:608442. [PMID: 33568974 PMCID: PMC7868393 DOI: 10.3389/fnmol.2020.608442] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 12/31/2020] [Indexed: 12/13/2022] Open
Abstract
Peripheral nerve injuries arising from trauma or disease can lead to sensory and motor deficits and neuropathic pain. Despite the purported ability of the peripheral nerve to self-repair, lifelong disability is common. New molecular and cellular insights have begun to reveal why the peripheral nerve has limited repair capacity. The peripheral nerve is primarily comprised of axons and Schwann cells, the supporting glial cells that produce myelin to facilitate the rapid conduction of electrical impulses. Schwann cells are required for successful nerve regeneration; they partially “de-differentiate” in response to injury, re-initiating the expression of developmental genes that support nerve repair. However, Schwann cell dysfunction, which occurs in chronic nerve injury, disease, and aging, limits their capacity to support endogenous repair, worsening patient outcomes. Cell replacement-based therapeutic approaches using exogenous Schwann cells could be curative, but not all Schwann cells have a “repair” phenotype, defined as the ability to promote axonal growth, maintain a proliferative phenotype, and remyelinate axons. Two cell replacement strategies are being championed for peripheral nerve repair: prospective isolation of “repair” Schwann cells for autologous cell transplants, which is hampered by supply challenges, and directed differentiation of pluripotent stem cells or lineage conversion of accessible somatic cells to induced Schwann cells, with the potential of “unlimited” supply. All approaches require a solid understanding of the molecular mechanisms guiding Schwann cell development and the repair phenotype, which we review herein. Together these studies provide essential context for current efforts to design glial cell-based therapies for peripheral nerve regeneration.
Collapse
Affiliation(s)
- Anjali Balakrishnan
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Lauren Belfiore
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Tak-Ho Chu
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Taylor Fleming
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada
| | - Rajiv Midha
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Jeff Biernaskie
- Department of Comparative Biology and Experimental Medicine, Hotchkiss Brain Institute, University of Calgary, Calgary, AB, Canada
| | - Carol Schuurmans
- Biological Sciences Platform, Sunnybrook Research Institute (SRI), Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
44
|
Xu Y, Sheng X, Zhao T, Zhang L, Ruan Y, Lu H. O-GlcNAcylation of MEK2 promotes the proliferation and migration of breast cancer cells. Glycobiology 2020; 31:571-581. [PMID: 33226073 DOI: 10.1093/glycob/cwaa103] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 11/06/2020] [Accepted: 11/14/2020] [Indexed: 12/13/2022] Open
Abstract
Mitogen-activated protein kinase kinases are an important part of evolutionary conserved signaling modules that are involved in a variety of cellular processes in response to environmental stimuli. Among them, mitogen-activated protein kinase kinase 2 (MEK2) is the most crucial upstream signaling pathway of ERK1/2 cascade as a therapeutic target for overcoming Ras-driven cancers. However, the mechanisms of MEK2 regulation during tumor progression remain not fully elucidated. Herein, we identified that MEK2 was post-translationally regulated by O-GlcNAcylation. We found that MEK2 associated with OGT and was modified by O-GlcNAc. Mass spectrometry analysis further verified that O-GlcNAcylation of MEK2 occurred at Thr13, which was in the docking domain for specifically identifying its target proteins. While total O-GlcNAcylation stimulated the protein stability and phosphorylation of MEK2, Thr13 O-GlcNAcylation of MEK2 specifically enhanced its Thr394 phosphorylation as well as downstream ERK1/2 activation. Genetic ablation of MEK2 O-GlcNAcylation at Thr13 abrogated its ability to promote the proliferation and migration of breast cancer cells. Together, our data demonstrate that O-GlcNAcylation of MEK2 might be a key regulatory mechanism during tumorigenesis and is a potential therapeutic target for tumor treatment.
Collapse
Affiliation(s)
- Yaoyao Xu
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Xiangying Sheng
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.,Department of Chemistry, Fudan University, Shanghai 200433, China
| | - Ting Zhao
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Lei Zhang
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Yuanyuan Ruan
- NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai 200032, China.,Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Haojie Lu
- Shanghai Cancer Center and Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.,Department of Chemistry, Fudan University, Shanghai 200433, China.,NHC Key Laboratory of Glycoconjugates Research, Fudan University, Shanghai 200032, China
| |
Collapse
|
45
|
Effects of Pacap on Schwann Cells: Focus on Nerve Injury. Int J Mol Sci 2020; 21:ijms21218233. [PMID: 33153152 PMCID: PMC7663204 DOI: 10.3390/ijms21218233] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/23/2020] [Accepted: 11/02/2020] [Indexed: 12/27/2022] Open
Abstract
Schwann cells, the most abundant glial cells of the peripheral nervous system, represent the key players able to supply extracellular microenvironment for axonal regrowth and restoration of myelin sheaths on regenerating axons. Following nerve injury, Schwann cells respond adaptively to damage by acquiring a new phenotype. In particular, some of them localize in the distal stump to form the Bungner band, a regeneration track in the distal site of the injured nerve, whereas others produce cytokines involved in recruitment of macrophages infiltrating into the nerve damaged area for axonal and myelin debris clearance. Several neurotrophic factors, including pituitary adenylyl cyclase-activating peptide (PACAP), promote survival and axonal elongation of injured neurons. The present review summarizes the evidence existing in the literature demonstrating the autocrine and/or paracrine action exerted by PACAP to promote remyelination and ameliorate the peripheral nerve inflammatory response following nerve injury.
Collapse
|
46
|
Scapin C, Ferri C, Pettinato E, Bianchi F, Del Carro U, Feltri ML, Kaufman RJ, Wrabetz L, D'Antonio M. Phosphorylation of eIF2α Promotes Schwann Cell Differentiation and Myelination in CMT1B Mice with Activated UPR. J Neurosci 2020; 40:8174-8187. [PMID: 32973043 PMCID: PMC7574653 DOI: 10.1523/jneurosci.0957-20.2020] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 07/09/2020] [Accepted: 07/27/2020] [Indexed: 01/01/2023] Open
Abstract
Myelin Protein Zero (MPZ/P0) is the most abundant glycoprotein of peripheral nerve myelin. P0 is synthesized by myelinating Schwann cells, processed in the endoplasmic reticulum (ER) and delivered to myelin via the secretory pathway. The mutant P0S63del (deletion of serine 63 in the extracellular domain of P0), that causes Charcot-Marie-Tooth type 1B (CMT1B) neuropathy in humans and a similar demyelinating neuropathy in transgenic mice, is instead retained the ER where it activates an unfolded protein response. Under ER-stress conditions, protein kinase R-like endoplasmic reticulum kinase (PERK) phosphorylates eukaryotic initiation factor 2α (eIF2α) to attenuate global translation, thus reducing the misfolded protein overload in the ER. Genetic and pharmacological inactivation of Gadd34 (damage-inducible protein 34), a subunit of the PP1 phosphatase complex that promotes the dephosphorylation of eIF2α, prolonged eIF2α phosphorylation and improved motor, neurophysiological, and morphologic deficits in S63del mice. However, PERK ablation in S63del Schwann cells ameliorated, rather than worsened, S63del neuropathy despite reduced levels of phosphorylated eIF2α. These contradictory findings prompted us to genetically explore the role of eIF2α phosphorylation in P0S63del-CMT1B neuropathy through the generation of mice in which eIF2α cannot be phosphorylated specifically in Schwann cells. Morphologic and electrophysiological analysis of male and female S63del mice showed a worsening of the neuropathy in the absence of eIF2α phosphorylation. However, we did not detect significant changes in ER stress levels, but rather a dramatic increase of the MEK/ERK/c-Jun pathway accompanied by a reduction in expression of myelin genes and a delay in Schwann cell differentiation. Our results support the hypothesis that eIF2α phosphorylation is protective in CMT1B and unveil a possible cross talk between eIF2α and the MEK/ERK pathway in neuropathic nerves.SIGNIFICANCE STATEMENT In the P0S63del (deletion of serine 63 in the extracellular domain of P0) mouse model of Charcot-Marie-Tooth type 1B (CMT1B), the genetic and pharmacological inhibition of Gadd34 (damage-inducible protein 34) prolonged eukaryotic initiation factor 2α (eIF2α) phosphorylation, leading to a proteostatic rebalance that significantly ameliorated the neuropathy. Yet, ablation of protein kinase R-like endoplasmic reticulum kinase (PERK) also ameliorated the S63del neuropathy, despite reduced levels of eIF2α phosphorylation (P-eIF2α). In this study, we provide genetic evidence that eIF2α phosphorylation has a protective role in CMT1B Schwann cells by limiting ERK/c-Jun hyperactivation. Our data support the targeting of the P-eIF2α/Gadd34 complex as a therapeutic avenue in CMT1B and also suggest that PERK may hamper myelination via mechanisms outside its role in the unfolded protein response.
Collapse
Affiliation(s)
- Cristina Scapin
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Cinzia Ferri
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Emanuela Pettinato
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Francesca Bianchi
- Institute of Experimental Neurology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Ubaldo Del Carro
- Institute of Experimental Neurology, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - M Laura Feltri
- Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, New York 14203
- Department of Neurology, State University of New York at Buffalo, Buffalo, New York 14203
- Department of Biochemistry, Jacob School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York 14203
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, California 92130
| | - Lawrence Wrabetz
- Hunter James Kelly Research Institute, State University of New York at Buffalo, Buffalo, New York 14203
- Department of Neurology, State University of New York at Buffalo, Buffalo, New York 14203
- Department of Biochemistry, Jacob School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York 14203
| | - Maurizio D'Antonio
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, 20132 Milan, Italy
| |
Collapse
|
47
|
Nocera G, Jacob C. Mechanisms of Schwann cell plasticity involved in peripheral nerve repair after injury. Cell Mol Life Sci 2020; 77:3977-3989. [PMID: 32277262 PMCID: PMC7532964 DOI: 10.1007/s00018-020-03516-9] [Citation(s) in RCA: 249] [Impact Index Per Article: 49.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 03/09/2020] [Accepted: 03/30/2020] [Indexed: 01/01/2023]
Abstract
The great plasticity of Schwann cells (SCs), the myelinating glia of the peripheral nervous system (PNS), is a critical feature in the context of peripheral nerve regeneration following traumatic injuries and peripheral neuropathies. After a nerve damage, SCs are rapidly activated by injury-induced signals and respond by entering the repair program. During the repair program, SCs undergo dynamic cell reprogramming and morphogenic changes aimed at promoting nerve regeneration and functional recovery. SCs convert into a repair phenotype, activate negative regulators of myelination and demyelinate the damaged nerve. Moreover, they express many genes typical of their immature state as well as numerous de-novo genes. These genes modulate and drive the regeneration process by promoting neuronal survival, damaged axon disintegration, myelin clearance, axonal regrowth and guidance to their former target, and by finally remyelinating the regenerated axon. Many signaling pathways, transcriptional regulators and epigenetic mechanisms regulate these events. In this review, we discuss the main steps of the repair program with a particular focus on the molecular mechanisms that regulate SC plasticity following peripheral nerve injury.
Collapse
Affiliation(s)
- Gianluigi Nocera
- Faculty of Biology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University, Mainz, Germany
| | - Claire Jacob
- Faculty of Biology, Institute of Developmental Biology and Neurobiology, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
48
|
Sun P, Zhang G, Su X, Jin C, Yu B, Yu X, Lv Z, Ma H, Zhang M, Wei W, Li W. Maintenance of Primary Hepatocyte Functions In Vitro by Inhibiting Mechanical Tension-Induced YAP Activation. Cell Rep 2020; 29:3212-3222.e4. [PMID: 31801084 DOI: 10.1016/j.celrep.2019.10.128] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 04/30/2019] [Accepted: 10/29/2019] [Indexed: 02/07/2023] Open
Abstract
Hepatocytes are the primary functional cells of the liver, performing its metabolic, detoxification, and endocrine functions. Functional hepatocytes are extremely valuable in drug discovery and evaluation, as well as in cell therapy for liver diseases. However, it has been a long-standing challenge to maintain the functions of hepatocytes in vitro. Even freshly isolated hepatocytes lose essential functions after short-term culture for reasons that are still not well understood. In the present study, we find that mechanical tension-induced yes-associated protein activation triggers hepatocyte dedifferentiation. Alleviation of mechanical tension by confining cell spreading is sufficient to inhibit hepatocyte dedifferentiation. Based on this finding, we identify a small molecular cocktail through reiterative chemical screening that can maintain hepatocyte functions over the long term and in vivo repopulation capacity by targeting actin polymerization and actomyosin contraction. Our work reveals the mechanisms underlying hepatocyte dedifferentiation and establishes feasible approaches to maintain hepatocyte functions.
Collapse
Affiliation(s)
- Pingxin Sun
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Guanyu Zhang
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Xiaohui Su
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China; Stem Cell and Regenerative Medicine Center, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Caixia Jin
- Department of Regenerative Medicine, College of Medicine, Tongji University, Shanghai 200433, China
| | - Bing Yu
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Xinlu Yu
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Zhuman Lv
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Haoxin Ma
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China
| | - Mingliang Zhang
- Department of Histoembryology, Genetics, and Developmental Biology, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Wanguo Wei
- Stem Cell and Regenerative Medicine Center, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai 201210, China
| | - Wenlin Li
- Department of Cell Biology, Second Military Medical University, Shanghai 200433, China; Department of Regenerative Medicine, College of Medicine, Tongji University, Shanghai 200433, China; Shanghai Key Laboratory of Cell Engineering, Second Military Medical University, Shanghai 200433, China.
| |
Collapse
|
49
|
Tiong YL, Ng KY, Koh RY, Ponnudurai G, Chye SM. Melatonin promotes Schwann cell dedifferentiation and proliferation through the Ras/Raf/ERK and MAPK pathways, and glial cell-derived neurotrophic factor expression. Exp Ther Med 2020; 20:16. [PMID: 32934681 PMCID: PMC7471953 DOI: 10.3892/etm.2020.9143] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 11/22/2019] [Indexed: 12/24/2022] Open
Abstract
Upon peripheral nerve injury (PNI), continuous proliferation of Schwann cells is critical for axon regeneration and tubular reconstruction for nerve regeneration. Melatonin is a hormone that is able to induce proliferation in various cell types. In the present study, the effects of melatonin on promoting Schwann cell proliferation and the molecular mechanism involved were investigated. The present results showed that melatonin enhanced the melatonin receptors (MT1 and MT2) expression in Schwann cells. Melatonin induced Schwann cell dedifferentiation into progenitor-like Schwann cells, as observed by immunofluorescence staining, which showed Sox2 marker expression. In addition, melatonin enhanced Schwann cell proliferation, mediated by the upregulation of glial cell-derived neurotropic factor (GNDF) and protein kinase C (PKC). Furthermore, the Ras/Raf/ERK and MAPK signaling pathways were also involved in Schwann cell dedifferentiation and proliferation. In conclusion, melatonin induced Schwann cell dedifferentiation and proliferation via the Ras/Raf/ERK, MAPK and GDNF/PKC pathways. The present results suggested that melatonin could be used to enhance the recovery of PNI.
Collapse
Affiliation(s)
- Yee Lian Tiong
- School of Postgraduate, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University Malaysia, Subang Jaya, Selangor 47500, Malaysia
| | - Rhun Yian Koh
- School of Health Science, International Medical University, Kuala Lumpur 57000, Malaysia
| | | | - Soi Moi Chye
- School of Health Science, International Medical University, Kuala Lumpur 57000, Malaysia
| |
Collapse
|
50
|
Mungroo MR, Khan NA, Siddiqui R. Mycobacterium leprae: Pathogenesis, diagnosis, and treatment options. Microb Pathog 2020; 149:104475. [PMID: 32931893 DOI: 10.1016/j.micpath.2020.104475] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 09/02/2020] [Accepted: 09/04/2020] [Indexed: 01/14/2023]
Abstract
Mycobacterium leprae is known to cause leprosy, a neurological and dermatological disease. In the past 20 years, 16 million leprosy cases have been recorded and more than 200,000 new cases were registered each year, indicating that the disease is still progressing without hindrance. M. leprae, an intracellular bacterium, infects the Schwann cells of the peripheral nervous system. Several types of leprosy have been described, including indeterminate, tuberculoid, borderline tuberculoid, mid-borderline, borderline lepromatous and lepromatous, and three different forms of leprosy reactions, namely type 1, 2 and 3, have been designated. Microscopic detection, serological diagnostic test, polymerase chain reaction and flow tests are employed in the diagnosis of leprosy. The recommended treatment for leprosy consists of rifampicin, dapsone, clofazimine, ofloxacin and minocycline and vaccines are also available. However, relapse may occur after treatment has been halted and hence patients must be educated on the signs of relapse to allow proper treatment and reduce severity. In this review, we depict the current understanding of M. leprae pathogenicity, clinical aspects and manifestations. Transmission of leprosy, diagnosis and treatment are also discussed.
Collapse
Affiliation(s)
- Mohammad Ridwane Mungroo
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, University City, Sharjah, United Arab Emirates
| | - Naveed Ahmed Khan
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, University City, Sharjah, United Arab Emirates.
| | - Ruqaiyyah Siddiqui
- Department of Biology, Chemistry and Environmental Sciences, College of Arts and Sciences, American University of Sharjah, University City, Sharjah, United Arab Emirates
| |
Collapse
|