1
|
Wilson ER, Nunes GDF, Shen S, Moore S, Gawron J, Maxwell J, Syed U, Hurley E, Lanka M, Qu J, Désaubry L, Wrabetz L, Poitelon Y, Feltri ML. Loss of prohibitin 2 in Schwann cells dysregulates key transcription factors controlling developmental myelination. Glia 2024; 72:2247-2267. [PMID: 39215540 DOI: 10.1002/glia.24610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/18/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024]
Abstract
Schwann cells are critical for the proper development and function of the peripheral nervous system (PNS), where they form a collaborative relationship with axons. Past studies highlighted that a pair of proteins called the prohibitins play major roles in Schwann cell biology. Prohibitins are ubiquitously expressed and versatile proteins. We have previously shown that while prohibitins play a crucial role in Schwann cell mitochondria for long-term myelin maintenance and axon health, they may also be present at the Schwann cell-axon interface during development. Here, we expand on this, showing that drug-mediated modulation of prohibitins in vitro disrupts myelination and confirming that Schwann cell-specific ablation of prohibitin 2 (Phb2) in vivo results in severe defects in radial sorting and myelination. We show in vivo that Phb2-null Schwann cells cannot effectively proliferate and the transcription factors EGR2 (KROX20), POU3F1 (OCT6), and POU3F2 (BRN2), necessary for proper Schwann cell maturation, are dysregulated. Schwann cell-specific deletion of Jun, a transcription factor associated with negative regulation of myelination, confers partial rescue of the developmental defect seen in mice lacking Schwann cell Phb2. Finally, we identify a pool of candidate PHB2 interactors that change their interaction with PHB2 depending on neuronal signals, and thus are potential mediators of PHB2-associated developmental defects. This work develops our understanding of Schwann cell biology, revealing that Phb2 may modulate the timely expression of transcription factors necessary for proper PNS development, and proposing candidates that may play a role in PHB2-mediated integration of axon signals in the Schwann cell.
Collapse
Affiliation(s)
- Emma R Wilson
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, England, UK
| | - Gustavo Della-Flora Nunes
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Shichen Shen
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Seth Moore
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Joseph Gawron
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Jessica Maxwell
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Umair Syed
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Edward Hurley
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Meghana Lanka
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - Jun Qu
- Department of Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Laurent Désaubry
- Center of Research in Biomedicine of Strasbourg, Regenerative Nanomedicine (UMR 1260), INSERM, University of Strasbourg, Strasbourg, France
| | - Lawrence Wrabetz
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| | - Yannick Poitelon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, New York, USA
| | - M Laura Feltri
- Department of Biochemistry, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
- Department of Neurology, Institute for Myelin and Glia Exploration, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, New York, USA
| |
Collapse
|
2
|
Zhang X, Wang X, Yin L, Wang D, Jiao H, Liu X, Zheng J. HACE1 exerts a neuroprotective role against oxidative stress in cerebral ischemia-reperfusion injury by activating the PI3K/AKT/Nrf2 pathway. Neuroscience 2024; 559:249-262. [PMID: 39244008 DOI: 10.1016/j.neuroscience.2024.09.002] [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: 03/22/2024] [Revised: 08/26/2024] [Accepted: 09/01/2024] [Indexed: 09/09/2024]
Abstract
HECT domain and Ankyrin repeat-containing E3 ubiquitin protein ligase 1 (HACE1) is an E3 ubiquitin ligase involving oxidative stress, an important contributor in cerebral ischemia-reperfusion injury (CIRI). It was proposed to be associated with the PI3K/AKT pathway and Nrf2 nuclear translocation, which are important players of oxidative stress. Therefore, we supposed that HACE1 might affect CIRI by regulating the PI3K/AKT/Nrf2 pathway. Here, we used the transient middle cerebral artery occlusion-reperfusion (tMCAO/R) model to induce CIRI in rats and found lower HACE1 expression in ischemic rats compared with the control. To explore the exact role of HACE1, the lentivirus vector carrying the HACE1 sequence was administrated to rats by intracerebroventricular injection (1 × 109 TU/mL, 9 μL) one week before tMCAO/R operation. HACE1 overexpression alleviated tMCAO/R-induced brain damage in rats. Further studies revealed that it reduced oxidative stress via activating the PI3K/AKT/Nrf2 pathway, thereby inhibiting neuronal apoptosis in the ischemic penumbra of rats with CIRI. Then, differentiated PC12 cells were cultured in oxygen-glucose deprivation-reoxygenation (OGD/R) conditions (OGD: 1 % O2, 94 % N2, and 5 % CO2; R: normal atmosphere) to simulate CIRI in vitro. Similarly, HACE1 overexpression inhibited neuronal apoptosis caused by OGD/R treatment. The PI3K inhibitor LY294002 reversed the inhibitory effects of HACE1 overexpression on oxidative stress in OGD/R-injured cells, accompanied by the inactivated AKT/Nrf2 pathway. Altogether, our results suggest that HACE1 protects against oxidative stress-induced neuronal apoptosis in CIRI by activating the PI3K/AKT/Nrf2 pathway, providing a new insight into the CIRI treatment.
Collapse
Affiliation(s)
- Xinyue Zhang
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Xiao Wang
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Le Yin
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Dan Wang
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Hong Jiao
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Xiaodan Liu
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China
| | - Jiaolin Zheng
- Department of Neurology, the Second Affiliated Hospital of Harbin Medical University, Harbin 150001, Heilongjiang, China.
| |
Collapse
|
3
|
Patel AA, Kim H, Ramesh R, Marquez A, Faraj MM, Antikainen H, Lee AS, Torres A, Khawaja IM, Heffernan C, Bonder EM, Maurel P, Svaren J, Son YJ, Dobrowolski R, Kim HA. TFEB/3 Govern Repair Schwann Cell Generation and Function Following Peripheral Nerve Injury. J Neurosci 2024; 44:e0198242024. [PMID: 39054068 PMCID: PMC11358533 DOI: 10.1523/jneurosci.0198-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024] Open
Abstract
TFEB and TFE3 (TFEB/3), key regulators of lysosomal biogenesis and autophagy, play diverse roles depending on cell type. This study highlights a hitherto unrecognized role of TFEB/3 crucial for peripheral nerve repair. Specifically, they promote the generation of progenitor-like repair Schwann cells after axonal injury. In Schwann cell-specific TFEB/3 double knock-out mice of either sex, the TFEB/3 loss disrupts the transcriptomic reprogramming that is essential for the formation of repair Schwann cells. Consequently, mutant mice fail to populate the injured nerve with repair Schwann cells and exhibit defects in axon regrowth, target reinnervation, and functional recovery. TFEB/3 deficiency inhibits the expression of injury-responsive repair Schwann cell genes, despite the continued expression of c-jun, a previously identified regulator of repair Schwann cell function. TFEB/3 binding motifs are enriched in the enhancer regions of injury-responsive genes, suggesting their role in repair gene activation. Autophagy-dependent myelin breakdown is not impaired despite TFEB/3 deficiency. These findings underscore a unique role of TFEB/3 in adult Schwann cells that is required for proper peripheral nerve regeneration.
Collapse
Affiliation(s)
- Akash A Patel
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Hyukmin Kim
- Shriners Hospitals Pediatric Research Center and Department of Neural Science, Temple University, Philadelphia, Pennsylvania 19140
| | - Raghu Ramesh
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin 53705
- Comparative Biomedical Sciences Graduate Program, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Anthony Marquez
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Moler M Faraj
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Henri Antikainen
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Andrew S Lee
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Adriana Torres
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Imran M Khawaja
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Corey Heffernan
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Edward M Bonder
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Patrice Maurel
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - John Svaren
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin 53705
- Comparative Biomedical Sciences Graduate Program, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin 53706
- Department of Comparative Biosciences, School of Veterinary Medicine University of Wisconsin-Madison, Madison, Wisconsin 53705
| | - Young-Jin Son
- Shriners Hospitals Pediatric Research Center and Department of Neural Science, Temple University, Philadelphia, Pennsylvania 19140
- Department of Anatomy and Cell Biology, Temple University, Philadelphia, Pennsylvania 19140
| | - Radek Dobrowolski
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| | - Haesun A Kim
- Department of Biological Sciences, Rutgers University, Newark, New Jersey 07102
| |
Collapse
|
4
|
Wu L, Wang XJ, Luo X, Zhang J, Zhao X, Chen Q. Diabetic peripheral neuropathy based on Schwann cell injury: mechanisms of cell death regulation and therapeutic perspectives. Front Endocrinol (Lausanne) 2024; 15:1427679. [PMID: 39193373 PMCID: PMC11348392 DOI: 10.3389/fendo.2024.1427679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
Diabetic peripheral neuropathy (DPN) is a complication of diabetes mellitus that lacks specific treatment, its high prevalence and disabling neuropathic pain greatly affects patients' physical and mental health. Schwann cells (SCs) are the major glial cells of the peripheral nervous system, which play an important role in various inflammatory and metabolic neuropathies by providing nutritional support, wrapping axons and promoting repair and regeneration. Increasingly, high glucose (HG) has been found to promote the progression of DPN pathogenesis by targeting SCs death regulation, thus revealing the specific molecular process of programmed cell death (PCD) in which SCs are disrupted is an important link to gain insight into the pathogenesis of DPN. This paper is the first to review the recent progress of HG studies on apoptosis, autophagy, pyroptosis, ferroptosis and necroptosis pathways in SCs, and points out the crosstalk between various PCDs and the related therapeutic perspectives, with the aim of providing new perspectives for a deeper understanding of the mechanisms of DPN and the exploration of effective therapeutic targets.
Collapse
Affiliation(s)
- Lijiao Wu
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiang Jin Wang
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, China
| | - Xi Luo
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jingqi Zhang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinyi Zhao
- College of lntegrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Hunan, China
| | - Qiu Chen
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
5
|
Ayuso-García P, Sánchez-Rueda A, Velasco-Avilés S, Tamayo-Caro M, Ferrer-Pinós A, Huarte-Sebastian C, Alvarez V, Riobello C, Jiménez-Vega S, Buendia I, Cañas-Martin J, Fernández-Susavila H, Aparicio-Rey A, Esquinas-Román EM, Ponte CR, Guhl R, Laville N, Pérez-Andrés E, Lavín JL, González-Lopez M, Cámara NM, Aransay AM, Lozano JJ, Sutherland JD, Barrio R, Martinez-Chantar ML, Azkargorta M, Elortza F, Soriano-Navarro M, Matute C, Sánchez-Gómez MV, Bayón-Cordero L, Pérez-Samartín A, Bravo SB, Kurz T, Lama-Díaz T, Blanco MG, Haddad S, Record CJ, van Hasselt PM, Reilly MM, Varela-Rey M, Woodhoo A. Neddylation orchestrates the complex transcriptional and posttranscriptional program that drives Schwann cell myelination. SCIENCE ADVANCES 2024; 10:eadm7600. [PMID: 38608019 PMCID: PMC11014456 DOI: 10.1126/sciadv.adm7600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 03/11/2024] [Indexed: 04/14/2024]
Abstract
Myelination is essential for neuronal function and health. In peripheral nerves, >100 causative mutations have been identified that cause Charcot-Marie-Tooth disease, a disorder that can affect myelin sheaths. Among these, a number of mutations are related to essential targets of the posttranslational modification neddylation, although how these lead to myelin defects is unclear. Here, we demonstrate that inhibiting neddylation leads to a notable absence of peripheral myelin and axonal loss both in developing and regenerating mouse nerves. Our data indicate that neddylation exerts a global influence on the complex transcriptional and posttranscriptional program by simultaneously regulating the expression and function of multiple essential myelination signals, including the master transcription factor EGR2 and the negative regulators c-Jun and Sox2, and inducing global secondary changes in downstream pathways, including the mTOR and YAP/TAZ signaling pathways. This places neddylation as a critical regulator of myelination and delineates the potential pathogenic mechanisms involved in CMT mutations related to neddylation.
Collapse
Affiliation(s)
- Paula Ayuso-García
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Alejandro Sánchez-Rueda
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Sergio Velasco-Avilés
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Miguel Tamayo-Caro
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Aroa Ferrer-Pinós
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Cecilia Huarte-Sebastian
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Vanesa Alvarez
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Cristina Riobello
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Selene Jiménez-Vega
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Izaskun Buendia
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Science Park of UPV/EHU, Sede building, 48940 Leioa, Spain
| | - Jorge Cañas-Martin
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Héctor Fernández-Susavila
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Adrián Aparicio-Rey
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Eva M. Esquinas-Román
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Carlos Rodríguez Ponte
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
| | - Romane Guhl
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
- Université Paris Cité Magistère Européen de Génétique, 85 Boulevard Saint-Germain, 75006 Paris, France
| | - Nicolas Laville
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
- Université Paris Cité Magistère Européen de Génétique, 85 Boulevard Saint-Germain, 75006 Paris, France
| | - Encarni Pérez-Andrés
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - José L. Lavín
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
- NEIKER–Basque Institute for Agricultural Research and Development, Applied Mathematics Department, Bioinformatics Unit, Basque Research and Technology Alliance (BRTA), 48160 Derio, Bizkaia, Spain
| | - Monika González-Lopez
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Nuria Macías Cámara
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Ana M. Aransay
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Juan José Lozano
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - James D. Sutherland
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Rosa Barrio
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - María Luz Martinez-Chantar
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Mikel Azkargorta
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Félix Elortza
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
| | - Mario Soriano-Navarro
- Electron Microscopy Core Facility, Centro de Investigación Príncipe Felipe (CIPF), 46012 Valencia, Spain
| | - Carlos Matute
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Science Park of UPV/EHU, Sede building, 48940 Leioa, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - María Victoria Sánchez-Gómez
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Science Park of UPV/EHU, Sede building, 48940 Leioa, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Laura Bayón-Cordero
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Science Park of UPV/EHU, Sede building, 48940 Leioa, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Alberto Pérez-Samartín
- Laboratory of Neurobiology, Achucarro Basque Center for Neuroscience, Science Park of UPV/EHU, Sede building, 48940 Leioa, Spain
- Department of Neurosciences, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), 28031 Madrid, Spain
| | - Susana B. Bravo
- Proteomic Unit, Health Research Institute of Santiago de Compostela (IDIS), 15705 Santiago de Compostela, A Coruña, Spain
| | - Thimo Kurz
- Evotec SE, Innovation Dr, Milton, Abingdon OX14 4RT, UK and School of Molecular Biosciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Tomas Lama-Díaz
- DNA Repair and Genome Integrity Laboratory, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15706 Santiago de Compostela, A Coruña, Spain
| | - Miguel G. Blanco
- DNA Repair and Genome Integrity Laboratory, CIMUS, University of Santiago de Compostela-Instituto de Investigación Sanitaria, 15706 Santiago de Compostela, A Coruña, Spain
- Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, Plaza do Obradoiro s/n, Santiago de Compostela, Spain
| | - Saif Haddad
- Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Christopher J. Record
- Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Peter M. van Hasselt
- Department of Metabolic Diseases, Division Pediatrics, Wilhelmina Children’s Hospital University Medical Center Utrecht, Utrecht University, 3584 EA, Utrecht, Netherlands
| | - Mary M. Reilly
- Centre for Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
| | - Marta Varela-Rey
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
- Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, Plaza do Obradoiro s/n, Santiago de Compostela, Spain
| | - Ashwin Woodhoo
- Gene Regulatory Control in Disease Laboratory, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, 15706 Santiago de Compostela, A Coruña, Spain
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Bizkaia Technology Park, 48160 Derio, Spain
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Bizkaia, Spain
- Department of Functional Biology, University of Santiago de Compostela, Plaza do Obradoiro s/n, Santiago de Compostela, Spain
- Oportunius Research Professor at CIMUS/USC, Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, A Coruña, Spain
| |
Collapse
|
6
|
Karalis V, Wood D, Teaney NA, Sahin M. The role of TSC1 and TSC2 proteins in neuronal axons. Mol Psychiatry 2024; 29:1165-1178. [PMID: 38212374 DOI: 10.1038/s41380-023-02402-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Revised: 12/20/2023] [Accepted: 12/22/2023] [Indexed: 01/13/2024]
Abstract
Tuberous Sclerosis Complex 1 and 2 proteins, TSC1 and TSC2 respectively, participate in a multiprotein complex with a crucial role for the proper development and function of the nervous system. This complex primarily acts as an inhibitor of the mechanistic target of rapamycin (mTOR) kinase, and mutations in either TSC1 or TSC2 cause a neurodevelopmental disorder called Tuberous Sclerosis Complex (TSC). Neurological manifestations of TSC include brain lesions, epilepsy, autism, and intellectual disability. On the cellular level, the TSC/mTOR signaling axis regulates multiple anabolic and catabolic processes, but it is not clear how these processes contribute to specific neurologic phenotypes. Hence, several studies have aimed to elucidate the role of this signaling pathway in neurons. Of particular interest are axons, as axonal defects are associated with severe neurocognitive impairments. Here, we review findings regarding the role of the TSC1/2 protein complex in axons. Specifically, we will discuss how TSC1/2 canonical and non-canonical functions contribute to the formation and integrity of axonal structure and function.
Collapse
Affiliation(s)
- Vasiliki Karalis
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Delaney Wood
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA
- Human Neuron Core, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Nicole A Teaney
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA
| | - Mustafa Sahin
- Rosamund Stone Zander Translational Neuroscience Center, Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
- FM Kirby Neurobiology Center, Boston Children's Hospital, Boston, MA, 02115, USA.
- Human Neuron Core, Boston Children's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
7
|
Shan F, Zhang N, Yao X, Li Y, Wang Z, Zhang C, Wang Y. Mechanosensitive channel of large conductance enhances the mechanical stretching-induced upregulation of glycolysis and oxidative metabolism in Schwann cells. Cell Commun Signal 2024; 22:93. [PMID: 38302971 PMCID: PMC10835878 DOI: 10.1186/s12964-024-01497-x] [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: 12/29/2023] [Accepted: 01/21/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Physical exercise directly stretching the peripheral nerve promotes nerve regeneration; however, its action mechanism remains elusive. Our present study aimed to investigate the effects of mechanosensitive channel of large conductance (MscL) activated by mechanical stretching on the cultured Schwann cells (SCs) and explore the possible mechanism. METHODS Primary SCs from neonatal mice at 3-5 days of age were derived and transfected with the lentivirus vector expressing a mutant version of MscL, MscL-G22S. We first detected the cell viability and calcium ion (Ca2+) influx in the MscL-G22S-expressing SCs with low-intensity mechanical stretching and the controls. Proteomic and energy metabolomics analyses were performed to investigate the comprehensive effects of MscL-G22S activation on SCs. Measurement of glycolysis- and oxidative phosphorylation-related molecules and ATP production were respectively performed to further validate the effects of MscL-G22S activation on SCs. Finally, the roles of phosphatidylinositol-3-kinase (PI3K)/AKT/mammalian target of rapamycin (mTOR) signaling pathway in the mechanism of energy metabolism modulation of SCs by MscL-G22S activation was investigated. RESULTS Mechanical stretching-induced MscL-G22S activation significantly increased the cell viability and Ca2+ influx into the SCs. Both the proteomic and targeted energy metabolomics analysis indicated the upregulation of energy metabolism as the main action mechanism of MscL-G22S-activation on SCs. MscL-G22S-activated SCs showed significant upregulation of glycolysis and oxidative phosphorylation when SCs with stretching alone had only mild upregulation of energy metabolism than those without stimuli. MscL-G22S activation caused significant phosphorylation of the PI3K/AKT/mTOR signaling pathway and upregulation of HIF-1α/c-Myc. Inhibition of PI3K abolished the MscL-G22S activation-induced upregulation of HIF-1α/c-Myc signaling in SCs and reduced the levels of glycolysis- and oxidative phosphorylation-related substrates and mitochondrial activity. CONCLUSION Mechanical stretching activates MscL-G22S to significantly promote the energy metabolism of SCs and the production of energic substrates, which may be applied to enhance nerve regeneration via the glia-axonal metabolic coupling.
Collapse
Affiliation(s)
- Fangzhen Shan
- Medical Research Centre, Affiliated Hospital of Jining Medical University, Jining, Shandong Province, China
| | - Nannan Zhang
- Department of Respiratory and Critical Care, Affiliated Hospital of Jining Medical University, Jining, Shandong Province, China
| | - Xiaoying Yao
- Medical Research Centre, Affiliated Hospital of Jining Medical University, Jining, Shandong Province, China
| | - Yi Li
- Department of Neurology, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining City, Shandong Province, 272029, China
| | - Zihao Wang
- Cheeloo Medical College, Shandong University, Jinan, Shandong Province, China
| | - Chuanji Zhang
- College of Rehabilitation Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Yuzhong Wang
- Medical Research Centre, Affiliated Hospital of Jining Medical University, Jining, Shandong Province, China.
- Department of Neurology, Affiliated Hospital of Jining Medical University, 89 Guhuai Road, Jining City, Shandong Province, 272029, China.
| |
Collapse
|
8
|
Zhu J, Hu Z, Luo Y, Liu Y, Luo W, Du X, Luo Z, Hu J, Peng S. Diabetic peripheral neuropathy: pathogenetic mechanisms and treatment. Front Endocrinol (Lausanne) 2024; 14:1265372. [PMID: 38264279 PMCID: PMC10803883 DOI: 10.3389/fendo.2023.1265372] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 12/14/2023] [Indexed: 01/25/2024] Open
Abstract
Diabetic peripheral neuropathy (DPN) refers to the development of peripheral nerve dysfunction in patients with diabetes when other causes are excluded. Diabetic distal symmetric polyneuropathy (DSPN) is the most representative form of DPN. As one of the most common complications of diabetes, its prevalence increases with the duration of diabetes. 10-15% of newly diagnosed T2DM patients have DSPN, and the prevalence can exceed 50% in patients with diabetes for more than 10 years. Bilateral limb pain, numbness, and paresthesia are the most common clinical manifestations in patients with DPN, and in severe cases, foot ulcers can occur, even leading to amputation. The etiology and pathogenesis of diabetic neuropathy are not yet completely clarified, but hyperglycemia, disorders of lipid metabolism, and abnormalities in insulin signaling pathways are currently considered to be the initiating factors for a range of pathophysiological changes in DPN. In the presence of abnormal metabolic factors, the normal structure and function of the entire peripheral nervous system are disrupted, including myelinated and unmyelinated nerve axons, perikaryon, neurovascular, and glial cells. In addition, abnormalities in the insulin signaling pathway will inhibit neural axon repair and promote apoptosis of damaged cells. Here, we will discuss recent advances in the study of DPN mechanisms, including oxidative stress pathways, mechanisms of microvascular damage, mechanisms of damage to insulin receptor signaling pathways, and other potential mechanisms associated with neuroinflammation, mitochondrial dysfunction, and cellular oxidative damage. Identifying the contributions from each pathway to neuropathy and the associations between them may help us to further explore more targeted screening and treatment interventions.
Collapse
Affiliation(s)
- Jinxi Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Ziyan Hu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yifan Luo
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yinuo Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Wei Luo
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaohong Du
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Zhenzhong Luo
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Jialing Hu
- Department of Emergency Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Shengliang Peng
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
9
|
Gu X, Rahman FS, Bendale G, Tran B, Miyata JF, Hernandez A, Anand S, Romero-Ortega MI. Pleiotrophin-Neuregulin1 promote axon regeneration and sorting in conduit repair of critical nerve gap injuries. RESEARCH SQUARE 2023:rs.3.rs-3429258. [PMID: 37986821 PMCID: PMC10659554 DOI: 10.21203/rs.3.rs-3429258/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Significant challenges remain in the treatment of critical nerve gap injuries using artificial nerve conduits. We previously reported successful axon regeneration across a 40 mm nerve gap using a biosynthetic nerve implant (BNI) with multi-luminal synergistic growth factor release. However, axon sorting, remyelination, and functional recovery were limited. Neuregulin1 (NRG1) plays a significant role in regulating the proliferation and differentiation of Schwann cells (SCs) during development and after injury. We hypothesize that the release of NRG1 type III combined with pleiotrophin (PTN) in the BNI will enhance axon growth, remyelination, and function of regenerated nerves across a critical gap. A rabbit 40 mm peroneal gap injury model was used to investigate the therapeutic efficacy of BNIs containing either NRG1, PTN, or PTN+NRG1 growth factor release. We found that NRG1 treatment doubled the number of regenerated axons (1276±895) compared to empty controls (633±666) and PTN tripled this number (2270±989). NRG1 also significantly increased the number of SOX10+ Schwann cells in mid-conduit (20.42%±11.78%) and reduced the number of abnormal Remak axon bundles. The combination of PTN+NRG1 increased axon diameter (1.70±1.06) vs control (1.21±0.77) (p<0.01), with 15.35% of axons above 3 μm, comparable to autograft. However, the total number of remyelinated axons was not increased by the added NRG1 release, which correlated with absence of axonal NRG1 type III expression in the regenerated axons. Electrophysiological evaluation showed higher muscle force recruitment (23.8±16.0 mN vs 17.4±1.4 mN) and maximum evoked compound motor action potential (353 μV vs 37 μV) in PTN-NRG1 group versus control, which correlated with the improvement in the toe spread recovery observed in PTN-NRG1 treated animals (0.64±0.02) vs control (0.50±0.01). These results revealed the need of a combination of pro-regenerative and remyelinating growth factor combination therapy for the repair of critical nerve gaps.
Collapse
Affiliation(s)
- Xingjian Gu
- Department of Biomedical Engineering, University of Houston, Houston TX 77204
| | - Farial S. Rahman
- Department of Biomedical Engineering, University of Houston, Houston TX 77204
| | - G Bendale
- Department of Biomedical Engineering, University of Houston, Houston TX 77204
| | - B Tran
- Department of Biomedical Engineering, University of Houston, Houston TX 77204
| | - JF Miyata
- Department of Biomedical Engineering, University of Houston, Houston TX 77204
| | - A Hernandez
- Department of Biomedical Engineering, University of Houston, Houston TX 77204
| | - S Anand
- Department of Biomedical Engineering, University of Houston, Houston TX 77204
| | | |
Collapse
|
10
|
Duong P, Ramesh R, Schneider A, Won S, Cooper AJ, Svaren J. Modulation of Schwann cell homeostasis by the BAP1 deubiquitinase. Glia 2023; 71:1466-1480. [PMID: 36790040 PMCID: PMC10073320 DOI: 10.1002/glia.24351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 01/24/2023] [Accepted: 01/25/2023] [Indexed: 02/16/2023]
Abstract
Schwann cell programming during myelination involves transcriptional networks that activate gene expression but also repress genes that are active in neural crest/embryonic differentiation of Schwann cells. We previously found that a Schwann cell-specific deletion of the EED subunit of the Polycomb Repressive Complex (PRC2) led to inappropriate activation of many such genes. Moreover, some of these genes become re-activated in the pro-regenerative response of Schwann cells to nerve injury, and we found premature activation of the nerve injury program in a Schwann cell-specific knockout of Eed. Polycomb-associated histone modifications include H3K27 trimethylation formed by PRC2 and H2AK119 monoubiquitination (H2AK119ub1), deposited by Polycomb repressive complex 1 (PRC1). We recently found dynamic regulation of H2AK119ub1 in Schwann cell genes after injury. Therefore, we hypothesized that H2AK119 deubiquitination modulates the dynamic polycomb repression of genes involved in Schwann cell maturation. To determine the role of H2AK119 deubiquitination, we generated a Schwann cell-specific knockout of the H2AK119 deubiquitinase Bap1 (BRCA1-associated protein). We found that loss of Bap1 causes tomacula formation, decreased axon diameters and eventual loss of myelinated axons. The gene expression changes are accompanied by redistribution of H2AK119ub1 and H3K27me3 modifications to extragenic sites throughout the genome. BAP1 interacts with OGT in the PR-DUB complex, and our data suggest that the PR-DUB complex plays a multifunctional role in repression of the injury program. Overall, our results indicate Bap1 is required to restrict the spread of polycomb-associated histone modifications in Schwann cells and to promote myelin homeostasis in peripheral nerve.
Collapse
Affiliation(s)
- Phu Duong
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Raghu Ramesh
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Andrew Schneider
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Seongsik Won
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Aaron J Cooper
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - John Svaren
- Waisman Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department Of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
11
|
El-Bazzal L, Ghata A, Estève C, Gadacha J, Quintana P, Castro C, Roeckel-Trévisiol N, Lembo F, Lenfant N, Mégarbané A, Borg JP, Lévy N, Bartoli M, Poitelon Y, Roubertoux PL, Delague V, Bernard-Marissal N. Imbalance of NRG1-ERBB2/3 signalling underlies altered myelination in Charcot-Marie-Tooth disease 4H. Brain 2023; 146:1844-1858. [PMID: 36314052 PMCID: PMC10151191 DOI: 10.1093/brain/awac402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 08/30/2022] [Accepted: 10/02/2022] [Indexed: 11/12/2022] Open
Abstract
Charcot-Marie-Tooth (CMT) disease is one of the most common inherited neurological disorders, affecting either axons from the motor and/or sensory neurons or Schwann cells of the peripheral nervous system (PNS) and caused by more than 100 genes. We previously identified mutations in FGD4 as responsible for CMT4H, an autosomal recessive demyelinating form of CMT disease. FGD4 encodes FRABIN, a GDP/GTP nucleotide exchange factor, particularly for the small GTPase Cdc42. Remarkably, nerves from patients with CMT4H display excessive redundant myelin figures called outfoldings that arise from focal hypermyelination, suggesting that FRABIN could play a role in the control of PNS myelination. To gain insights into the role of FGD4/FRABIN in Schwann cell myelination, we generated a knockout mouse model (Fgd4SC-/-), with conditional ablation of Fgd4 in Schwann cells. We show that the specific deletion of FRABIN in Schwann cells leads to aberrant myelination in vitro, in dorsal root ganglia neuron/Schwann cell co-cultures, as well as in vivo, in distal sciatic nerves from Fgd4SC-/- mice. We observed that those myelination defects are related to an upregulation of some interactors of the NRG1 type III/ERBB2/3 signalling pathway, which is known to ensure a proper level of myelination in the PNS. Based on a yeast two-hybrid screen, we identified SNX3 as a new partner of FRABIN, which is involved in the regulation of endocytic trafficking. Interestingly, we showed that the loss of FRABIN impairs endocytic trafficking, which may contribute to the defective NRG1 type III/ERBB2/3 signalling and myelination. Using RNA-Seq, in vitro, we identified new potential effectors of the deregulated pathways, such as ERBIN, RAB11FIP2 and MAF, thereby providing cues to understand how FRABIN contributes to proper ERBB2 trafficking or even myelin membrane addition through cholesterol synthesis. Finally, we showed that the re-establishment of proper levels of the NRG1 type III/ERBB2/3 pathway using niacin treatment reduces myelin outfoldings in nerves of CMT4H mice. Overall, our work reveals a new role of FRABIN in the regulation of NRG1 type III/ERBB2/3 NRG1signalling and myelination and opens future therapeutic strategies based on the modulation of the NRG1 type III/ERBB2/3 pathway to reduce CMT4H pathology and more generally other demyelinating types of CMT disease.
Collapse
Affiliation(s)
- Lara El-Bazzal
- Aix Marseille Univ, INSERM, MMG, U 1251, Marseille, France
| | - Adeline Ghata
- Aix Marseille Univ, INSERM, MMG, U 1251, Marseille, France
| | | | - Jihane Gadacha
- Aix Marseille Univ, INSERM, MMG, U 1251, Marseille, France
| | | | | | | | - Frédérique Lembo
- Aix Marseille Univ, INSERM, CNRS, CRCM, Institut Paoli-Calmettes, Marseille, France
| | | | - André Mégarbané
- Department of Human Genetics, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| | - Jean-Paul Borg
- Aix Marseille Univ, INSERM, CNRS, CRCM, Institut Paoli-Calmettes, Marseille, France
| | - Nicolas Lévy
- Aix Marseille Univ, INSERM, MMG, U 1251, Marseille, France
| | - Marc Bartoli
- Aix Marseille Univ, INSERM, MMG, U 1251, Marseille, France
| | - Yannick Poitelon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, USA
| | | | | | | |
Collapse
|
12
|
Yang Z, Yu Z, Xiao B. Coordinated Regulation of Myelination by Growth Factor and Amino-acid Signaling Pathways. Neurosci Bull 2023; 39:453-465. [PMID: 36352321 PMCID: PMC10043148 DOI: 10.1007/s12264-022-00967-x] [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: 03/11/2022] [Accepted: 06/27/2022] [Indexed: 11/11/2022] Open
Abstract
Myelin-forming oligodendrocytes in the central nervous system (CNS) and Schwann cells in the peripheral nervous system (PNS) are essential for structural and functional homeostasis of nervous tissue. Albeit with certain similarities, the regulation of CNS and PNS myelination is executed differently. Recent advances highlight the coordinated regulation of oligodendrocyte myelination by amino-acid sensing and growth factor signaling pathways. In this review, we discuss novel insights into the understanding of differential regulation of oligodendrocyte and Schwann cell biology in CNS and PNS myelination, with particular focus on the roles of growth factor-stimulated RHEB-mTORC1 and GATOR2-mediated amino-acid sensing/signaling pathways. We also discuss recent progress on the metabolic regulation of oligodendrocytes and Schwann cells and the impact of their dysfunction on neuronal function and disease.
Collapse
Affiliation(s)
- Zhiwen Yang
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Zongyan Yu
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518000, China.
| | - Bo Xiao
- Shenzhen Key Laboratory of Gene Regulation and Systems Biology, Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, 518000, China.
| |
Collapse
|
13
|
Takenaka T, Ohnishi Y, Yamamoto M, Setoyama D, Kishima H. Glycolytic System in Axons Supplement Decreased ATP Levels after Axotomy of the Peripheral Nerve. eNeuro 2023; 10:ENEURO.0353-22.2023. [PMID: 36894321 PMCID: PMC10035771 DOI: 10.1523/eneuro.0353-22.2023] [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: 08/30/2022] [Revised: 02/04/2023] [Accepted: 02/25/2023] [Indexed: 03/11/2023] Open
Abstract
Wallerian degeneration (WD) occurs in the early stages of numerous neurologic disorders, and clarifying WD pathology is crucial for the advancement of neurologic therapies. ATP is acknowledged as one of the key pathologic substances in WD. The ATP-related pathologic pathways that regulate WD have been defined. The elevation of ATP levels in axon contributes to delay WD and protects axons. However, ATP is necessary for the active processes to proceed WD, given that WD is stringently managed by auto-destruction programs. But little is known about the bioenergetics during WD. In this study, we made sciatic nerve transection models for GO-ATeam2 knock-in rats and mice. We presented the spatiotemporal ATP distribution in the injured axons with in vivo ATP imaging systems, and investigated the metabolic source of ATP in the distal nerve stump. A gradual decrease in ATP levels was observed before the progression of WD. In addition, the glycolytic system and monocarboxylate transporters (MCTs) were activated in Schwann cells following axotomy. Interestingly, in axons, we found the activation of glycolytic system and the inactivation of the tricarboxylic acid (TCA) cycle. Glycolytic inhibitors, 2-deoxyglucose (2-DG) and MCT inhibitors, a-cyano-4-hydroxycinnamic acid (4-CIN) decreased ATP and enhanced WD progression, whereas mitochondrial pyruvate carrier (MPC) inhibitors (MSDC-0160) did not change. Finally, ethyl pyruvate (EP) increased ATP levels and delayed WD. Together, our findings suggest that glycolytic system, both in Schwann cells and axons, is the main source of maintaining ATP levels in the distal nerve stump.
Collapse
Affiliation(s)
- Tomofumi Takenaka
- Department of neurosurgery, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
- Department of Research Promotion and Management, National Cerebral and Cardiovascular Center, Osaka, 564-8565, Japan
| | - Yuichiro Ohnishi
- Department of Research Promotion and Management, National Cerebral and Cardiovascular Center, Osaka, 564-8565, Japan
- Department of Neurosurgery, Osaka Gyoumeikan Hospital, Osaka, 554-0012, Japan
| | - Masamichi Yamamoto
- Department of Research Promotion and Management, National Cerebral and Cardiovascular Center, Osaka, 564-8565, Japan
| | - Daiki Setoyama
- Department of Clinical Chemistry and Laboratory Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Haruhiko Kishima
- Department of neurosurgery, Graduate School of Medicine, Osaka University, Osaka, 565-0871, Japan
| |
Collapse
|
14
|
Physical exercise mediates a cortical FMRP-mTOR pathway to improve resilience against chronic stress in adolescent mice. Transl Psychiatry 2023; 13:16. [PMID: 36658152 PMCID: PMC9852236 DOI: 10.1038/s41398-023-02311-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 12/31/2022] [Accepted: 01/06/2023] [Indexed: 01/20/2023] Open
Abstract
Aerobic exercise effectively relieves anxiety disorders via modulating neurogenesis and neural activity. The molecular mechanism of exercise-mediated anxiolysis, however, remains incomplete. On a chronic restrain stress (CRS) model in adolescent mice, we showed that 14-day treadmill exercise profoundly maintained normal neural activity and axonal myelination in the medial prefrontal cortex (mPFC), in association with the prevention of anxiety-like behaviors. Further interrogation of molecular mechanisms revealed the activation of the mechanistic target of the rapamycin (mTOR) pathway within mPFC under exercise training. At the upstream of mTOR, exercise-mediated brain RNA methylation inhibited the expression of Fragile X mental retardation protein (FMRP) to activate the mTOR pathway. In summary, treadmill exercise modulates an FMRP-mTOR pathway to maintain cortical neural activity and axonal myelination, contributing to improved stress resilience. These results extended our understanding of the molecular substrate of exercise-mediated anxiolytic effect during adolescent period.
Collapse
|
15
|
Pérez-Villegas EM, Ruiz R, Bachiller S, Ventura F, Armengol JA, Rosa JL. The HERC proteins and the nervous system. Semin Cell Dev Biol 2022; 132:5-15. [PMID: 34848147 DOI: 10.1016/j.semcdb.2021.11.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/15/2021] [Accepted: 11/16/2021] [Indexed: 12/14/2022]
Abstract
The HERC protein family is one of three subfamilies of Homologous to E6AP C-terminus (HECT) E3 ubiquitin ligases. Six HERC genes have been described in humans, two of which encode Large HERC proteins -HERC1 and HERC2- with molecular weights above 520 kDa that are constitutively expressed in the brain. There is a large body of evidence that mutations in these Large HERC genes produce clinical syndromes in which key neurodevelopmental events are altered, resulting in intellectual disability and other neurological disorders like epileptic seizures, dementia and/or signs of autism. In line with these consequences in humans, two mice carrying mutations in the Large HERC genes have been studied quite intensely: the tambaleante mutant for Herc1 and the Herc2+/530 mutant for Herc2. In both these mutant mice there are clear signs that autophagy is dysregulated, eliciting cerebellar Purkinje cell death and impairing motor control. The tambaleante mouse was the first of these mice to appear and is the best studied, in which the Herc1 mutation elicits: (i) delayed neural transmission in the peripheral nervous system; (ii) impaired learning, memory and motor control; and (iii) altered presynaptic membrane dynamics. In this review, we discuss the information currently available on HERC proteins in the nervous system and their biological activity, the dysregulation of which could explain certain neurodevelopmental syndromes and/or neurodegenerative diseases.
Collapse
Affiliation(s)
- Eva M Pérez-Villegas
- Department of Physiology, Anatomy and Cell Biology, University Pablo de Olavide, Seville, Spain
| | - Rocío Ruiz
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, Seville, Spain
| | - Sara Bachiller
- Clinical Unit of Infectious Diseases, Microbiology and Preventive Medicine, Institute of Biomedicine of Sevilla, Virgen del Rocío University Hospital, CSIC, University of Sevilla, Sevilla, Spain
| | - Francesc Ventura
- Departament de Ciències Fisiològiques, IBIDELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Jose A Armengol
- Department of Physiology, Anatomy and Cell Biology, University Pablo de Olavide, Seville, Spain.
| | - Jose Luis Rosa
- Departament de Ciències Fisiològiques, IBIDELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
16
|
Okura A, Inoue K, Sakuma E, Takase H, Ueki T, Mase M. SGK1 in Schwann cells is a potential molecular switch involved in axonal and glial regeneration during peripheral nerve injury. Biochem Biophys Res Commun 2022; 607:158-165. [DOI: 10.1016/j.bbrc.2022.03.123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 03/23/2022] [Indexed: 12/29/2022]
|
17
|
Wang S, Wang Y, Zou S. A Glance at the Molecules That Regulate Oligodendrocyte Myelination. Curr Issues Mol Biol 2022; 44:2194-2216. [PMID: 35678678 PMCID: PMC9164040 DOI: 10.3390/cimb44050149] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/10/2022] [Accepted: 05/13/2022] [Indexed: 11/16/2022] Open
Abstract
Oligodendrocyte (OL) myelination is a critical process for the neuronal axon function in the central nervous system. After demyelination occurs because of pathophysiology, remyelination makes repairs similar to myelination. Proliferation and differentiation are the two main stages in OL myelination, and most factors commonly play converse roles in these two stages, except for a few factors and signaling pathways, such as OLIG2 (Oligodendrocyte transcription factor 2). Moreover, some OL maturation gene mutations induce hypomyelination or hypermyelination without an obvious function in proliferation and differentiation. Herein, three types of factors regulating myelination are reviewed in sequence.
Collapse
Affiliation(s)
- Shunqi Wang
- Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang 330031, China; (S.W.); (Y.W.)
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
| | - Yingxing Wang
- Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang 330031, China; (S.W.); (Y.W.)
| | - Suqi Zou
- Institute of Life Science & School of Life Sciences, Nanchang University, Nanchang 330031, China; (S.W.); (Y.W.)
- School of Basic Medical Sciences, Nanchang University, Nanchang 330031, China
- Correspondence:
| |
Collapse
|
18
|
Zhao Y, Liang Y, Xu Z, Liu J, Liu X, Ma J, Sun C, Yang Y. Exosomal miR-673-5p from fibroblasts promotes Schwann cell-mediated peripheral neuron myelination by targeting the TSC2/mTORC1/SREBP2 axis. J Biol Chem 2022; 298:101718. [PMID: 35151688 PMCID: PMC8908274 DOI: 10.1016/j.jbc.2022.101718] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Revised: 02/06/2022] [Accepted: 02/07/2022] [Indexed: 12/30/2022] Open
Abstract
Peripheral myelination is a complicated process, wherein Schwann cells (SCs) promote the formation of the myelin sheath around the axons of peripheral neurons. Fibroblasts are the second resident cells in the peripheral nerves; however, the precise function of fibroblasts in SC-mediated myelination has rarely been examined. Here, we show that exosomes derived from fibroblasts boost myelination-related gene expression in SCs. We used exosome sequencing, together with bioinformatic analysis, to demonstrate that exosomal microRNA miR-673-5p is capable of stimulating myelin gene expression in SCs. Subsequent functional studies revealed that miR-673-5p targets the regulator of mechanistic target of the rapamycin (mTOR) complex 1 (mTORC1) tuberous sclerosis complex 2 in SCs, leading to the activation of downstream signaling pathways including mTORC1 and sterol-regulatory element binding protein 2. In vivo experiments further confirmed that miR-673-5p activates the tuberous sclerosis complex 2/mTORC1/sterol-regulatory element binding protein 2 axis, thus promoting the synthesis of cholesterol and related lipids and subsequently accelerating myelin sheath maturation in peripheral nerves. Overall, our findings revealed exosome-mediated cross talk between fibroblasts and SCs that plays a pivotal role in peripheral myelination. We propose that exosomes derived from fibroblasts and miR-673-5p might be useful for promoting peripheral myelination in translational medicine.
Collapse
Affiliation(s)
- Yahong Zhao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education; Co-innovation Center of Neurogeneration; NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China.
| | - Yunyun Liang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education; Co-innovation Center of Neurogeneration; NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Zhixin Xu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education; Co-innovation Center of Neurogeneration; NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Jina Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education; Co-innovation Center of Neurogeneration; NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Xiaoyu Liu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education; Co-innovation Center of Neurogeneration; NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Jinyu Ma
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education; Co-innovation Center of Neurogeneration; NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China
| | - Cheng Sun
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education; Co-innovation Center of Neurogeneration; NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China.
| | - Yumin Yang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education; Co-innovation Center of Neurogeneration; NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, Nantong, China.
| |
Collapse
|
19
|
Analysis of Signal Transduction Pathways Downstream M2 Receptor Activation: Effects on Schwann Cell Migration and Morphology. Life (Basel) 2022; 12:life12020211. [PMID: 35207498 PMCID: PMC8875146 DOI: 10.3390/life12020211] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 01/25/2022] [Accepted: 01/27/2022] [Indexed: 01/14/2023] Open
Abstract
Background: Schwann cells (SCs) express cholinergic receptors, suggesting a role of cholinergic signaling in the control of SC proliferation, differentiation and/or myelination. Our previous studies largely demonstrated that the pharmacological activation of the M2 muscarinic receptor subtype caused an inhibition of cell proliferation and promoted the expression of pro-myelinating differentiation genes. In order to elucidate the molecular signaling activated downstream the M2 receptor activation, in the present study we investigated the signal transduction pathways activated by the M2 orthosteric agonist arecaidine propargyl ester (APE) in SCs. Methods: Using Western blot we analyzed some components of the noncanonical pathways involving β1-arrestin and PI3K/AKT/mTORC1 signaling. A wound healing assay was used to evaluate SC migration. Results: Our results demonstrated that M2 receptor activation negatively modulated the PI3K/Akt/mTORC1 axis, possibly through β1-arrestin downregulation. The involvement of the mTORC1 complex was also supported by the decreased expression of its specific target p-p70 S6KThr389. Then, we also analyzed the expression of p-AMPKαthr172, a negative regulator of myelination that resulted in reduced levels after M2 agonist treatment. The analysis of cell migration and morphology allowed us to demonstrate that M2 receptor activation caused an arrest of SC migration and modified cell morphology probably by the modulation of β1-arrestin/cofilin-1 and PKCα expression, respectively. Conclusions: The data obtained demonstrated that M2 receptor activation in addition to the canonical Gi protein-coupled pathway modulates noncanonical pathways involving the mTORC1 complex and other kinases whose activation may contribute to the inhibition of SC proliferation and migration and address SC differentiation.
Collapse
|
20
|
Terry BK, Park R, Cho SH, Crino PB, Kim S. Abnormal activation of Yap/Taz contributes to the pathogenesis of tuberous sclerosis complex. Hum Mol Genet 2022; 31:1979-1996. [PMID: 34999833 PMCID: PMC9239747 DOI: 10.1093/hmg/ddab374] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 10/31/2021] [Accepted: 12/27/2021] [Indexed: 01/09/2023] Open
Abstract
The multi-systemic genetic disorder tuberous sclerosis complex (TSC) impacts multiple neurodevelopmental processes including neuronal morphogenesis, neuronal migration, myelination and gliogenesis. These alterations contribute to the development of cerebral cortex abnormalities and malformations. Although TSC is caused by mTORC1 hyperactivation, cognitive and behavioral impairments are not improved through mTORC1 targeting, making the study of the downstream effectors of this complex important for understanding the mechanisms underlying TSC. As mTORC1 has been shown to promote the activity of the transcriptional co-activator Yap, we hypothesized that altered Yap/Taz signaling contributes to the pathogenesis of TSC. We first observed that the levels of Yap/Taz are increased in human cortical tuber samples and in embryonic cortices of Tsc2 conditional knockout (cKO) mice. Next, to determine how abnormal upregulation of Yap/Taz impacts the neuropathology of TSC, we deleted Yap/Taz in Tsc2 cKO mice. Importantly, Yap/Taz/Tsc2 triple conditional knockout (tcKO) animals show reduced cortical thickness and cortical neuron cell size, despite the persistence of high mTORC1 activity, suggesting that Yap/Taz play a downstream role in cytomegaly. Furthermore, Yap/Taz/Tsc2 tcKO significantly restored cortical and hippocampal lamination defects and reduced hippocampal heterotopia formation. Finally, the loss of Yap/Taz increased the distribution of myelin basic protein in Tsc2 cKO animals, consistent with an improvement in myelination. Overall, our results indicate that targeting Yap/Taz lessens the severity of neuropathology in a TSC animal model. This study is the first to implicate Yap/Taz as contributors to cortical pathogenesis in TSC and therefore as potential novel targets in the treatment of this disorder.
Collapse
Affiliation(s)
- Bethany K Terry
- Department of Neural Sciences, Lewis Katz School of Medicine, Shriners Hospitals Pediatrics Research Center, Temple University, Philadelphia, PA 19140, USA,Biomedical Sciences Graduate Program, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Raehee Park
- Department of Neural Sciences, Lewis Katz School of Medicine, Shriners Hospitals Pediatrics Research Center, Temple University, Philadelphia, PA 19140, USA
| | - Seo-Hee Cho
- Department of Medicine, Sidney Kimmel Medical College, Center for Translational Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Peter B Crino
- Department of Neurology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Seonhee Kim
- To whom correspondence should be addressed. Tel: 215-926-9360; Fax: 215-926-9325;
| |
Collapse
|
21
|
Abstract
Myelin is a key evolutionary specialization and adaptation of vertebrates formed by the plasma membrane of glial cells, which insulate axons in the nervous system. Myelination not only allows rapid and efficient transmission of electric impulses in the axon by decreasing capacitance and increasing resistance but also influences axonal metabolism and the plasticity of neural circuits. In this review, we will focus on Schwann cells, the glial cells which form myelin in the peripheral nervous system. Here, we will describe the main extrinsic and intrinsic signals inducing Schwann cell differentiation and myelination and how myelin biogenesis is achieved. Finally, we will also discuss how the study of human disorders in which molecules and pathways relevant for myelination are altered has enormously contributed to the current knowledge on myelin biology.
Collapse
Affiliation(s)
- Alessandra Bolino
- Human Inherited Neuropathies Unit, Institute of Experimental Neurology INSPE, Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy.
| |
Collapse
|
22
|
Della-Flora Nunes G, Wilson ER, Hurley E, He B, O'Malley BW, Poitelon Y, Wrabetz L, Feltri ML. Activation of mTORC1 and c-Jun by Prohibitin1 loss in Schwann cells may link mitochondrial dysfunction to demyelination. eLife 2021; 10:e66278. [PMID: 34519641 PMCID: PMC8478418 DOI: 10.7554/elife.66278] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 09/13/2021] [Indexed: 12/26/2022] Open
Abstract
Schwann cell (SC) mitochondria are quickly emerging as an important regulator of myelin maintenance in the peripheral nervous system (PNS). However, the mechanisms underlying demyelination in the context of mitochondrial dysfunction in the PNS are incompletely understood. We recently showed that conditional ablation of the mitochondrial protein Prohibitin 1 (PHB1) in SCs causes a severe and fast progressing demyelinating peripheral neuropathy in mice, but the mechanism that causes failure of myelin maintenance remained unknown. Here, we report that mTORC1 and c-Jun are continuously activated in the absence of Phb1, likely as part of the SC response to mitochondrial damage. Moreover, we demonstrate that these pathways are involved in the demyelination process, and that inhibition of mTORC1 using rapamycin partially rescues the demyelinating pathology. Therefore, we propose that mTORC1 and c-Jun may play a critical role as executioners of demyelination in the context of perturbations to SC mitochondria.
Collapse
Affiliation(s)
- Gustavo Della-Flora Nunes
- Hunter James Kelly Research Institute, University at BuffaloBuffaloUnited States
- Department of Biochemistry, University at BuffaloBuffaloUnited States
| | - Emma R Wilson
- Hunter James Kelly Research Institute, University at BuffaloBuffaloUnited States
- Department of Biochemistry, University at BuffaloBuffaloUnited States
| | - Edward Hurley
- Hunter James Kelly Research Institute, University at BuffaloBuffaloUnited States
| | - Bin He
- Immunobiology & Transplant Science Center and Department of Surgery, Houston Methodist HospitalHoustonUnited States
| | - Bert W O'Malley
- Department of Medicine and Molecular and Cellular Biology, Baylor College of MedicineHoustonUnited States
| | - Yannick Poitelon
- Department of Neuroscience and Experimental Therapeutics, Albany Medical CollegeAlbanyUnited States
| | - Lawrence Wrabetz
- Hunter James Kelly Research Institute, University at BuffaloBuffaloUnited States
- Department of Biochemistry, University at BuffaloBuffaloUnited States
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at BuffaloBuffaloUnited States
| | - M Laura Feltri
- Hunter James Kelly Research Institute, University at BuffaloBuffaloUnited States
- Department of Biochemistry, University at BuffaloBuffaloUnited States
- Department of Neurology, Jacobs School of Medicine and Biomedical Sciences, University at BuffaloBuffaloUnited States
| |
Collapse
|
23
|
Dysregulation of myelin synthesis and actomyosin function underlies aberrant myelin in CMT4B1 neuropathy. Proc Natl Acad Sci U S A 2021; 118:2009469118. [PMID: 33653949 DOI: 10.1073/pnas.2009469118] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Charcot-Marie-Tooth type 4B1 (CMT4B1) is a severe autosomal recessive demyelinating neuropathy with childhood onset, caused by loss-of-function mutations in the myotubularin-related 2 (MTMR2) gene. MTMR2 is a ubiquitously expressed catalytically active 3-phosphatase, which in vitro dephosphorylates the 3-phosphoinositides PtdIns3P and PtdIns(3,5)P 2, with a preference for PtdIns(3,5)P 2 A hallmark of CMT4B1 neuropathy are redundant loops of myelin in the nerve termed myelin outfoldings, which can be considered the consequence of altered growth of myelinated fibers during postnatal development. How MTMR2 loss and the resulting imbalance of 3'-phosphoinositides cause CMT4B1 is unknown. Here we show that MTMR2 by regulating PtdIns(3,5)P 2 levels coordinates mTORC1-dependent myelin synthesis and RhoA/myosin II-dependent cytoskeletal dynamics to promote myelin membrane expansion and longitudinal myelin growth. Consistent with this, pharmacological inhibition of PtdIns(3,5)P 2 synthesis or mTORC1/RhoA signaling ameliorates CMT4B1 phenotypes. Our data reveal a crucial role for MTMR2-regulated lipid turnover to titrate mTORC1 and RhoA signaling thereby controlling myelin growth.
Collapse
|
24
|
Gong QQ, Wang X, Dou ZL, Zhang KY, Liu XG, Gao JG, Sun XY. A novel mouse line with epididymal initial segment-specific expression of Cre recombinase driven by the endogenous Lcn9 promoter. PLoS One 2021; 16:e0254802. [PMID: 34310634 PMCID: PMC8312960 DOI: 10.1371/journal.pone.0254802] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 07/03/2021] [Indexed: 12/12/2022] Open
Abstract
Spermatozoa released from testes undergo a maturation process and acquire the capacity to fertilize ova through epididymal transit. The epididymis is divided into four regions, each with unique morphology, gene profile, luminal microenvironment and distinct function. To study the functions of relevant genes in the epididymal initial segment (IS), a novel IS-specific mouse model, Lcn9-Cre knock-in (KI) mouse line was generated via CRISPR/Cas9 technology. The TAG stop codon was replaced by a 2A-NLS-Cre cassette, resulting in the co-expression of Lcn9 and Cre recombinase. IS-specific Cre expression was first observed from postnatal day 17. Using the Rosa26tdTomato reporter mice, the Cre-mediated DNA recombination was detected exclusively in principal cells. The epididymal IS-specific Cre activity in vivo was further confirmed using Lcn9-Cre mice crossed with a mouse strain carrying Tsc1 floxed alleles (Tsc1flox/+). Cre expression did not affect either normal development or male fecundity. Different from any epididymis-specific Cre mice reported previously, the novel Lcn9-Cre mouse line can be used to introduce entire IS-specific conditional gene editing for gene functional study.
Collapse
Affiliation(s)
- Qian-qian Gong
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Xiao Wang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Zhi-lin Dou
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Ke-yi Zhang
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Xiang-guo Liu
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Jian-gang Gao
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
| | - Xiao-yang Sun
- Shandong Provincial Key Laboratory of Animal Cell and Developmental Biology, School of Life Sciences, Shandong University, Qingdao, China
- * E-mail:
| |
Collapse
|
25
|
Ishii A, Furusho M, Bansal R. Mek/ERK1/2-MAPK and PI3K/Akt/mTOR signaling plays both independent and cooperative roles in Schwann cell differentiation, myelination and dysmyelination. Glia 2021; 69:2429-2446. [PMID: 34157170 DOI: 10.1002/glia.24049] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 04/29/2021] [Accepted: 06/04/2021] [Indexed: 01/15/2023]
Abstract
Multiple signals are involved in the regulation of developmental myelination by Schwann cells and in the maintenance of a normal myelin homeostasis throughout adult life, preserving the integrity of the axons in the PNS. Recent studies suggest that Mek/ERK1/2-MAPK and PI3K/Akt/mTOR intracellular signaling pathways play important, often overlapping roles in the regulation of myelination in the PNS. In addition, hyperactivation of these signaling pathways in Schwann cells leads to a late onset of various pathological changes in the sciatic nerves. However, it remains poorly understood whether these pathways function independently or sequentially or converge using a common mechanism to facilitate Schwann cell differentiation and myelin growth during development and in causing pathological changes in the adult animals. To address these questions, we analyzed multiple genetically modified mice using simultaneous loss- and constitutive gain-of-function approaches. We found that during development, the Mek/ERK1/2-MAPK pathway plays a primary role in Schwann cell differentiation, distinct from mTOR. However, during active myelination, ERK1/2 is dependent on mTOR signaling to drive the growth of the myelin sheath and regulate its thickness. Finally, our data suggest that peripheral nerve pathology during adulthood caused by hyperactivation of Mek/ERK1/2-MAPK or PI3K is likely to be independent or dependent on mTOR-signaling in different contexts. Thus, this study highlights the complexities in the roles played by two major intracellular signaling pathways in Schwann cells that affect their differentiation, myelination, and later PNS pathology and predicts that potential therapeutic modulation of these pathways in PNS neuropathies could be a complex process.
Collapse
Affiliation(s)
- Akihiro Ishii
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Miki Furusho
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Rashmi Bansal
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| |
Collapse
|
26
|
Deficiency of Microglial Autophagy Increases the Density of Oligodendrocytes and Susceptibility to Severe Forms of Seizures. eNeuro 2021; 8:ENEURO.0183-20.2021. [PMID: 33472865 PMCID: PMC7890520 DOI: 10.1523/eneuro.0183-20.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 12/11/2020] [Accepted: 01/02/2021] [Indexed: 12/14/2022] Open
Abstract
Excessive activation of mTOR in microglia impairs CNS homeostasis and causes severe epilepsy. Autophagy constitutes an important part of mTOR signaling. The contribution of microglial autophagy to CNS homeostasis and epilepsy remains to be determined. Here, we report that ATG7KO mice deficient for autophagy in microglia display a marked increase of myelination markers, a higher density of mature oligodendrocytes (ODCs), and altered lengths of the nodes of Ranvier. Moreover, we found that deficiency of microglial autophagy (ATG7KO) leads to increased seizure susceptibility in three seizure models (pilocarpine, kainic acid, and amygdala kindling). We demonstrated that ATG7KO mice develop severe generalized seizures and display nearly 100% mortality to convulsions induced by pilocarpine and kainic acid. In the amygdala kindling model, we observed significant facilitation of contralateral propagation of seizures, a process underlying the development of generalized seizures. Taken together, our results reveal impaired microglial autophagy as a novel mechanism underlying altered homeostasis of ODCs and increased susceptibility to severe and fatal generalized seizures.
Collapse
|
27
|
Comprehensive Analysis of Age-related Changes in Lipid Metabolism and Myelin Sheath Formation in Sciatic Nerves. J Mol Neurosci 2021; 71:2310-2323. [PMID: 33492614 DOI: 10.1007/s12031-020-01768-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 11/30/2020] [Indexed: 02/06/2023]
Abstract
To investigate the molecular changes related to myelin formation and lipid metabolism in the sciatic nerve in Sprague Dawley (SD) rats during aging. Thirty-six healthy male SD rats were divided into five groups according to age: 1 week, 1 month, 6 months, 12 months, and 24 months. Sciatic nerves were collected from 1-month-old and 24-month-old SD rats (n = 3) to perform next-generation sequencing (NGS) and bioinformatics analysis. Specimens from each group were harvested and analyzed by qPCR, Western blotting, and transmission electron microscopy (TEM). Protein-protein interaction (PPI) networks of differentially expressed mRNAs (DEmRNAs) related to myelin and lipid metabolism were constructed. DEmRNAs in subnetworks were verified using qPCR. A total of 4580 DEmRNAs were found during aging. The top enriched GO biological processes were primarily clustered in cholesterol and lipid metabolism, including the cholesterol biosynthetic process (RF = 3.16), sterol biosynthetic process (RF = 3.03), cholesterol metabolic process (RF = 2.15), sterol metabolic process (RF = 2.11), fatty acid biosynthetic process (RF = 2.09), and lipid biosynthetic process (RF = 1.79). The mRNA levels of MBP, PMP22, and MPZ were downregulated during aging, while the protein expression of MBP showed an increasing trend. The TEM results showed thin myelin sheaths and an increased number of unmyelinated axons in the 1-week-old rats, and the sheaths became thickened with degenerated axons appearing in older animals. Forty PPI subnetworks related to lipid metabolism were constructed, including one primary subnetwork and two smaller subnetworks. The hub genes were mTOR in sub-network 1, Akt1 in sub-network 2, and SIRT1 in sub-network 3. No gene expression was found consistent with the sequencing results, while in the downregulated genes, AKT1, CEBPA, LIPE, LRP5, PHB, and Rara were significantly downregulated in 24-month-old rats. Lipid metabolism might play an important role in maintaining the structure and physiological function in sciatic nerves during aging and could be candidates for nerve aging research.
Collapse
|
28
|
Jung K, Kim HN, Jeon NL, Hyung S. Comparison of the Efficacy of Optogenetic Stimulation of Glia versus Neurons in Myelination. ACS Chem Neurosci 2020; 11:4280-4288. [PMID: 33269905 DOI: 10.1021/acschemneuro.0c00542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Increasing evidence demonstrates that optogenetics contributes to the regulation of brain behavior, cognition, and physiology, particularly during myelination, potentially allowing for the bidirectional modulation of specific cell lines with spatiotemporal accuracy. However, the type of cell to be targeted, namely, glia vs neurons, and the degree to which optogenetically induced cell activity can regulate myelination during the development of the peripheral nervous system (PNS) are still underexplored. Herein, we report the comparison of optogenetic stimulation (OS) of Schwann cells (SCs) and motor neurons (MNs) for activation of myelination in the PNS. Capitalizing on these optogenetic tools, we confirmed that the formation of the myelin sheath was initially promoted more by OS of calcium translocating channelrhodopsin (CatCh)-transfected SCs than by OS of transfected MNs at 7 days in vitro (DIV). Additionally, the level of myelination was substantially enhanced even until 14 DIV. Surprisingly, after OS of SCs, > 91.1% ± 5.9% of cells expressed myelin basic protein, while that of MNs was 67.8% ± 6.1%. The potent effect of OS of SCs was revealed by the increased thickness of the myelin sheath at 14 DIV. Thus, the OS of SCs could highly accelerate myelination, while the OS of MNs only somewhat promoted myelination, indicating a clear direction for the optogenetic application of unique cell types for initiating and promoting myelination. Together, our findings support the importance of precise cell type selection for use in optogenetics, which in turn can be broadly applied to overcome the limitations of optogenetics after injury.
Collapse
Affiliation(s)
- Kyuhwan Jung
- Yonsei Biomedical Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
- Department and Research Institute of Rehabilitation Medicine, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hong Nam Kim
- Center for BioMicrosystems, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Noo Li Jeon
- Department of Mechanical Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Advanced Machinery and Design, Seoul National University, Seoul 08826, Republic of Korea
| | - Sujin Hyung
- Department of Mechanical Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Bio-MAX Institute, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
29
|
A glycolytic shift in Schwann cells supports injured axons. Nat Neurosci 2020; 23:1215-1228. [PMID: 32807950 DOI: 10.1038/s41593-020-0689-4] [Citation(s) in RCA: 84] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 07/07/2020] [Indexed: 01/09/2023]
Abstract
Axon degeneration is a hallmark of many neurodegenerative disorders. The current assumption is that the decision of injured axons to degenerate is cell-autonomously regulated. Here we show that Schwann cells (SCs), the glia of the peripheral nervous system, protect injured axons by virtue of a dramatic glycolytic upregulation that arises in SCs as an inherent adaptation to axon injury. This glycolytic response, paired with enhanced axon-glia metabolic coupling, supports the survival of axons. The glycolytic shift in SCs is largely driven by the metabolic signaling hub, mammalian target of rapamycin complex 1, and the downstream transcription factors hypoxia-inducible factor 1-alpha and c-Myc, which together promote glycolytic gene expression. The manipulation of glial glycolytic activity through this pathway enabled us to accelerate or delay the degeneration of perturbed axons in acute and subacute rodent axon degeneration models. Thus, we demonstrate a non-cell-autonomous metabolic mechanism that controls the fate of injured axons.
Collapse
|
30
|
Kou Y, Yu F, Yuan Y, Niu S, Han N, Zhang Y, Yin X, Xu H, Jiang B. Effects of NP-1 on proliferation, migration, and apoptosis of Schwann cell line RSC96 through the NF-κB signaling pathway. Am J Transl Res 2020; 12:4127-4140. [PMID: 32913493 PMCID: PMC7476162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 07/03/2020] [Indexed: 06/11/2023]
Abstract
Peripheral nerve injury is a common refractory disease in the clinic that often leads to dysfunction of movement and sensation. Different from other tissue injuries, peripheral nerve injury needs a longer time for regeneration. Therefore, effective drug therapy is needed to promote nerve regeneration in the treatment of peripheral nerve injury. Our preliminary studies have shown that continuous intramuscular injection of NP-1 promotes the regeneration of injured sciatic nerve in rats, but the mechanisms were still unknown. Schwann cells are very important cells in the formation of myelin sheath of peripheral nerves and participate in the repair and regeneration of peripheral nerve injury. To further investigate the effect of NP-1 on rat Schwann cells and the underlying mechanism, different concentrations of NP-1 were used to treat rat Schwann cell line RSC96. Light microscopy, CCK-8 assay, cell scratch assay, and special cell staining were performed to investigate RSC96 cell aging and apoptosis. mRNA and protein expression of NF-κB signaling pathway-related factors were determined using qPCR and immunohistochemistry respectively. Light microscopy, CCK-8 assay, cell scratch assay, and special cell staining showed NP-1 could improve the ability of proliferation, immigration of Schwann cells. QPCR and immunohistochemistry showed NP-1 influenced the expression of multiple factors associated with nerve regeneration which NF-κB signaling pathway played a key role. The results show that NP-1 promoted the proliferation and migration of RSC96 cells and inhibited cell aging and apoptosis possibly through the NF-κB signaling pathway. These findings provide a potential target for clinical treatment of peripheral neuropathy and experimental data support.
Collapse
Affiliation(s)
- Yuhui Kou
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Peking UniversityBeijing, China
- Key Laboratory of Trauma and Neural Regeneration (Peking University), Ministry of EducationBeijing, China
| | - Fei Yu
- National & Local Joint Engineering Research Center of Orthopaedic Biomaterials, Department of Bone & Joint Surgery, Peking University Shenzhen HospitalShenzhen, China
| | - Yusong Yuan
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Peking UniversityBeijing, China
- Diabetic Foot Treatment Center, Peking University People’s Hospital, Peking UniversityBeijing, China
| | - Suping Niu
- Office of Academic Research, Peking University People’s Hospital, Peking UniversityBeijing, China
| | - Na Han
- Key Laboratory of Trauma and Neural Regeneration (Peking University), Ministry of EducationBeijing, China
- Office of Academic Research, Peking University People’s Hospital, Peking UniversityBeijing, China
| | - Yajun Zhang
- National Center for Trauma MedicineBeijing, China
| | - Xiaofeng Yin
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Peking UniversityBeijing, China
- Key Laboratory of Trauma and Neural Regeneration (Peking University), Ministry of EducationBeijing, China
| | - Hailin Xu
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Peking UniversityBeijing, China
- Diabetic Foot Treatment Center, Peking University People’s Hospital, Peking UniversityBeijing, China
| | - Baoguo Jiang
- Department of Trauma and Orthopedics, Peking University People’s Hospital, Peking UniversityBeijing, China
- Key Laboratory of Trauma and Neural Regeneration (Peking University), Ministry of EducationBeijing, China
- National Center for Trauma MedicineBeijing, China
| |
Collapse
|
31
|
Sawade L, Grandi F, Mignanelli M, Patiño-López G, Klinkert K, Langa-Vives F, Di Guardo R, Echard A, Bolino A, Haucke V. Rab35-regulated lipid turnover by myotubularins represses mTORC1 activity and controls myelin growth. Nat Commun 2020; 11:2835. [PMID: 32503983 PMCID: PMC7275063 DOI: 10.1038/s41467-020-16696-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 05/18/2020] [Indexed: 01/21/2023] Open
Abstract
Inherited peripheral neuropathies (IPNs) represent a broad group of disorders including Charcot-Marie-Tooth (CMT) neuropathies characterized by defects primarily arising in myelin, axons, or both. The molecular mechanisms by which mutations in nearly 100 identified IPN/CMT genes lead to neuropathies are poorly understood. Here we show that the Ras-related GTPase Rab35 controls myelin growth via complex formation with the myotubularin-related phosphatidylinositol (PI) 3-phosphatases MTMR13 and MTMR2, encoded by genes responsible for CMT-types 4B2 and B1 in humans, and found that it downregulates lipid-mediated mTORC1 activation, a pathway known to crucially regulate myelin biogenesis. Targeted disruption of Rab35 leads to hyperactivation of mTORC1 signaling caused by elevated levels of PI 3-phosphates and to focal hypermyelination in vivo. Pharmacological inhibition of phosphatidylinositol 3,5-bisphosphate synthesis or mTORC1 signaling ameliorates this phenotype. These findings reveal a crucial role for Rab35-regulated lipid turnover by myotubularins to repress mTORC1 activity and to control myelin growth. Charcot-Marie-Tooth (CMT) is an inherited peripheral neuropathy. Here, the authors show that Rab35 forms a complex with genes implicated in CMT, MTMR13 and MTMR2, which regulates myelin growth by controlling mTORC1 signaling through lipid turnover.
Collapse
Affiliation(s)
- Linda Sawade
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125, Berlin, Germany
| | - Federica Grandi
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Marianna Mignanelli
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy.,San Raffaele Vita-Salute University, Via Olgettina 60, 20132, Milan, Italy
| | - Genaro Patiño-López
- Laboratorio de Investigación en Inmunología y Proteómica, Hospital Infantil de México, Federico Gómez. C.P, 06720, Ciudad de México, México
| | - Kerstin Klinkert
- Membrane Traffic and Cell Division Lab, Institut Pasteur, UMR3691, CNRS, 25-28 rue du Dr Roux, F-75015, Paris, France.,Sorbonne Université, Collège doctoral, F-75005, Paris, France
| | - Francina Langa-Vives
- Centre d'Ingénierie Génétique Murine, Institut Pasteur, 25-28 rue du Dr Roux, F-75015, Paris, France
| | - Roberta Di Guardo
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy
| | - Arnaud Echard
- Membrane Traffic and Cell Division Lab, Institut Pasteur, UMR3691, CNRS, 25-28 rue du Dr Roux, F-75015, Paris, France
| | - Alessandra Bolino
- Institute of Experimental Neurology (InSpe), Division of Neuroscience, IRCCS Ospedale San Raffaele, Via Olgettina 60, 20132, Milan, Italy.
| | - Volker Haucke
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Strasse 10, 13125, Berlin, Germany. .,Freie Universität Berlin, Faculty of Biology, Chemistry and Pharmacy, 14195, Berlin, Germany. .,Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt- Universität zu Berlin, NeuroCure Cluster of Excellence, 10117, Berlin, Germany.
| |
Collapse
|
32
|
Silva VM, Gomes JA, Tenório LPG, de Omena Neta GC, da Costa Paixão K, Duarte AKF, da Silva GCB, Ferreira RJS, Koike BDV, de Sales Marques C, da Silva Miguel RD, de Queiroz AC, Pereira LX, de Carvalho Fraga CA. Schwann cell reprogramming and lung cancer progression: a meta-analysis of transcriptome data. Oncotarget 2019; 10:7288-7307. [PMID: 31921388 PMCID: PMC6944448 DOI: 10.18632/oncotarget.27204] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 07/29/2019] [Indexed: 11/25/2022] Open
Abstract
Schwann cells were identified in the tumor surrounding area prior to initiate the invasion process underlying connective tissue. These cells promote cancer invasion through direct contact, while paracrine signaling and matrix remodeling are not sufficient to proceed. Considering the intertwined structure of signaling, regulatory, and metabolic processes within a cell, we employed a genome-scale biomolecular network. Accordingly, a meta-analysis of Schwann cells associated transcriptomic datasets was performed, and the core information on differentially expressed genes (DEGs) was obtained by statistical analyses. Gene set over-representation analyses was performed on core DEGs to identify significantly functional and pathway enrichment analysis between Schwann cells and, lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC). DEGs were further integrated with genome-scale human biomolecular networks. miRNAs were proposed by the reconstruction of a transcriptional and post-transcriptional regulatory network. Moreover, microarray-based transcriptome profiling was performed, and the prognostic power of selected dedifferentiated Schwann cell biomolecules was predicted. We observed that pathways associated with Schwann cells dedifferentiation was overexpressed in lung cancer samples. However, genes associated with Schwann cells migration inhibition system were downregulated. Besides, miRNA targeting those pathways were also deregulated. In this study, we report valuable data for further experimental and clinical analysis, because the proposed biomolecules have significant potential as systems biomarkers for screening or for therapeutic purposes in perineural invasion of lung cancer.
Collapse
Affiliation(s)
| | - Jessica Alves Gomes
- Department of Medicine, Federal University of Alagoas, Campus Arapiraca, Brazil
| | | | | | | | | | | | | | - Bruna Del Vechio Koike
- Department of Medicine, Federal University of the São Francisco Valley, Petrolina, Brazil
| | | | | | | | | | | |
Collapse
|
33
|
Hackett AR, Strickland A, Milbrandt J. Disrupting insulin signaling in Schwann cells impairs myelination and induces a sensory neuropathy. Glia 2019; 68:963-978. [PMID: 31758725 DOI: 10.1002/glia.23755] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 10/28/2019] [Accepted: 11/05/2019] [Indexed: 12/14/2022]
Abstract
Although diabetic mice have been studied for decades, little is known about the cell type specific contributions to diabetic neuropathy (DN). Schwann cells (SCs) myelinate and provide trophic support to peripheral nervous system axons. Altered SC metabolism leads to myelin defects, which can be seen both in inherited and DNs. How SC metabolism is altered in DN is not fully understood, but it is clear that insulin resistance underlies impaired lipid metabolism in many cell types throughout the body via the phosphoinositide 3-kinase/protein kinase b (PKB)/mammalian target of rapamycin (PI3K/AKT/mTOR) pathway. Here, we created an insulin resistant SC by deleting both insulin receptor (INSR) and insulin-like growth factor receptor 1 (IGF1R), to determine the role of this signaling pathway in development and response to injury in order to understand SC defects in DN. We found that myelin is thinner throughout development and adulthood in INSR/IGF1R Schwann cell specific knock out mice. The nerves of these mutant mice had reduced expression of key genes that mediate fatty acid and cholesterol synthesis due to reduced mTOR-sterol regulatory element-binding protein signaling. In adulthood, these mice show sensory neuropathy phenotypes reminiscent of diabetic mice. Altogether, these data suggest that SCs may play an important role in DN and targeting their metabolism could lead to new therapies for DN.
Collapse
Affiliation(s)
- Amber R Hackett
- Department of Genetics, Washington University School of Medicine, Saint Louis, Missouri
| | - Amy Strickland
- Department of Genetics, Washington University School of Medicine, Saint Louis, Missouri
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine, Saint Louis, Missouri
| |
Collapse
|
34
|
Harty BL, Coelho F, Pease-Raissi SE, Mogha A, Ackerman SD, Herbert AL, Gereau RW, Golden JP, Lyons DA, Chan JR, Monk KR. Myelinating Schwann cells ensheath multiple axons in the absence of E3 ligase component Fbxw7. Nat Commun 2019; 10:2976. [PMID: 31278268 PMCID: PMC6611888 DOI: 10.1038/s41467-019-10881-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 06/05/2019] [Indexed: 01/12/2023] Open
Abstract
In the central nervous system (CNS), oligodendrocytes myelinate multiple axons; in the peripheral nervous system (PNS), Schwann cells (SCs) myelinate a single axon. Why are the myelinating potentials of these glia so fundamentally different? Here, we find that loss of Fbxw7, an E3 ubiquitin ligase component, enhances the myelinating potential of SCs. Fbxw7 mutant SCs make thicker myelin sheaths and sometimes appear to myelinate multiple axons in a fashion reminiscent of oligodendrocytes. Several Fbxw7 mutant phenotypes are due to dysregulation of mTOR; however, the remarkable ability of mutant SCs to ensheathe multiple axons is independent of mTOR signaling. This indicates distinct roles for Fbxw7 in SC biology including modes of axon interactions previously thought to fundamentally distinguish myelinating SCs from oligodendrocytes. Our data reveal unexpected plasticity in the myelinating potential of SCs, which may have important implications for our understanding of both PNS and CNS myelination and myelin repair. The authors find that deletion from Schwann cells of an E3 ubiquitin ligase component called Fbxw7 leads to a phenotype reminiscent of myelination in the central nervous system where a single oligodendrocyte ensheaths multiple axons.
Collapse
Affiliation(s)
- Breanne L Harty
- Thaden School, 410 SE Staggerwing Lane, Bentonville, AR, 72712, USA.,Department of Developmental Biology, Washington University School of Medicine, 660S. Euclid Ave., St. Louis, MO, 63110, USA.,Vollum Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, 97239, USA
| | - Fernanda Coelho
- Vollum Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, 97239, USA
| | - Sarah E Pease-Raissi
- Department of Neurology, Weill Institute for Neuroscience, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Amit Mogha
- Vollum Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, 97239, USA
| | - Sarah D Ackerman
- Department of Developmental Biology, Washington University School of Medicine, 660S. Euclid Ave., St. Louis, MO, 63110, USA.,Institute of Neuroscience, University of Oregon, 1440 Franklin Blvd., Eugene, OR, 97403, USA
| | - Amy L Herbert
- Department of Developmental Biology, Washington University School of Medicine, 660S. Euclid Ave., St. Louis, MO, 63110, USA.,Department of Developmental Biology, Stanford University, 279W. Campus Dr., Stanford, CA, 94305, USA
| | - Robert W Gereau
- Department of Anesthesiology, Washington University Pain Center, 660S. Euclid Ave., St. Louis, MO, 63110, USA
| | - Judith P Golden
- Department of Anesthesiology, Washington University Pain Center, 660S. Euclid Ave., St. Louis, MO, 63110, USA
| | - David A Lyons
- Centre for Brain Discovery Sciences, MS Society Centre for Translational Research, Euan MacDonald Centre for Motor Neurone Disease Research, University of Edinburgh, 49 Little France Crescent, Edinburgh, EH16 4SB, UK
| | - Jonah R Chan
- Department of Neurology, Weill Institute for Neuroscience, University of California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Kelly R Monk
- Department of Developmental Biology, Washington University School of Medicine, 660S. Euclid Ave., St. Louis, MO, 63110, USA. .,Vollum Institute, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd., Portland, OR, 97239, USA.
| |
Collapse
|
35
|
Radu AG, Torch S, Fauvelle F, Pernet-Gallay K, Lucas A, Blervaque R, Delmas V, Schlattner U, Lafanechère L, Hainaut P, Tricaud N, Pingault V, Bondurand N, Bardeesy N, Larue L, Thibert C, Billaud M. LKB1 specifies neural crest cell fates through pyruvate-alanine cycling. SCIENCE ADVANCES 2019; 5:eaau5106. [PMID: 31328154 PMCID: PMC6636984 DOI: 10.1126/sciadv.aau5106] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 06/10/2019] [Indexed: 05/08/2023]
Abstract
Metabolic processes underlying the development of the neural crest, an embryonic population of multipotent migratory cells, are poorly understood. Here, we report that conditional ablation of the Lkb1 tumor suppressor kinase in mouse neural crest stem cells led to intestinal pseudo-obstruction and hind limb paralysis. This phenotype originated from a postnatal degeneration of the enteric nervous ganglia and from a defective differentiation of Schwann cells. Metabolomic profiling revealed that pyruvate-alanine conversion is enhanced in the absence of Lkb1. Mechanistically, inhibition of alanine transaminases restored glial differentiation in an mTOR-dependent manner, while increased alanine level directly inhibited the glial commitment of neural crest cells. Treatment with the metabolic modulator AICAR suppressed mTOR signaling and prevented Schwann cell and enteric defects of Lkb1 mutant mice. These data uncover a link between pyruvate-alanine cycling and the specification of glial cell fate with potential implications in the understanding of the molecular pathogenesis of neural crest diseases.
Collapse
Affiliation(s)
- Anca G. Radu
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38000 Grenoble, France
| | - Sakina Torch
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38000 Grenoble, France
| | - Florence Fauvelle
- Univ. Grenoble Alpes, INSERM, U1216, Grenoble Institute of Neurosciences GIN, 38000 Grenoble, France
- Univ. Grenoble Alpes, INSERM, US17, MRI facility IRMaGe, 38000 Grenoble, France
| | - Karin Pernet-Gallay
- Univ. Grenoble Alpes, INSERM, U1216, Grenoble Institute of Neurosciences GIN, 38000 Grenoble, France
| | - Anthony Lucas
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38000 Grenoble, France
| | - Renaud Blervaque
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38000 Grenoble, France
| | - Véronique Delmas
- Institut Curie, Normal and Pathological Development of Melanocytes, CNRS UMR3347; INSERM U1021; Equipe Labellisée–Ligue Nationale Contre le Cancer, Orsay, France
| | - Uwe Schlattner
- Laboratory of Fundamental and Applied Bioenergetics, Univ Grenoble Alpes, 38185 Grenoble, France
- INSERM U1055, 38041 Grenoble France
| | - Laurence Lafanechère
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38000 Grenoble, France
| | - Pierre Hainaut
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38000 Grenoble, France
| | - Nicolas Tricaud
- INSERM U1051, Institut des Neurosciences de Montpellier (INM), Université de Montpellier, Montpellier, France
| | | | | | - Nabeel Bardeesy
- Cancer Center, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA
- Department of Medicine, Harvard Medical School, Boston, MA 02114, USA
| | - Lionel Larue
- Institut Curie, Normal and Pathological Development of Melanocytes, CNRS UMR3347; INSERM U1021; Equipe Labellisée–Ligue Nationale Contre le Cancer, Orsay, France
| | - Chantal Thibert
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38000 Grenoble, France
- Corresponding author. (M.B.); (C.T.)
| | - Marc Billaud
- Institute for Advanced Biosciences, INSERM U1209, CNRS UMR5309, Université Grenoble Alpes, 38000 Grenoble, France
- “Clinical and experimental model of lymphomagenesis” Univ Lyon, Université Claude Bernard Lyon1, INSERM 1052, CNRS 5286, Centre Léon Bérard, Centre de recherche en cancérologie de Lyon, Lyon France
- Corresponding author. (M.B.); (C.T.)
| |
Collapse
|
36
|
Balancing mTOR Signaling and Autophagy in the Treatment of Parkinson's Disease. Int J Mol Sci 2019; 20:ijms20030728. [PMID: 30744070 PMCID: PMC6387269 DOI: 10.3390/ijms20030728] [Citation(s) in RCA: 155] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 02/01/2019] [Accepted: 02/01/2019] [Indexed: 12/18/2022] Open
Abstract
The mammalian target of rapamycin (mTOR) signaling pathway plays a critical role in regulating cell growth, proliferation, and life span. mTOR signaling is a central regulator of autophagy by modulating multiple aspects of the autophagy process, such as initiation, process, and termination through controlling the activity of the unc51-like kinase 1 (ULK1) complex and vacuolar protein sorting 34 (VPS34) complex, and the intracellular distribution of TFEB/TFE3 and proto-lysosome tubule reformation. Parkinson’s disease (PD) is a serious, common neurodegenerative disease characterized by dopaminergic neuron loss in the substantia nigra pars compacta (SNpc) and the accumulation of Lewy bodies. An increasing amount of evidence indicates that mTOR and autophagy are critical for the pathogenesis of PD. In this review, we will summarize recent advances regarding the roles of mTOR and autophagy in PD pathogenesis and treatment. Further characterizing the dysregulation of mTOR pathway and the clinical translation of mTOR modulators in PD may offer exciting new avenues for future drug development.
Collapse
|
37
|
Beirowski B. The LKB1-AMPK and mTORC1 Metabolic Signaling Networks in Schwann Cells Control Axon Integrity and Myelination: Assembling and upholding nerves by metabolic signaling in Schwann cells. Bioessays 2018; 41:e1800075. [PMID: 30537168 DOI: 10.1002/bies.201800075] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 11/03/2018] [Indexed: 01/10/2023]
Abstract
The Liver kinase B1 with its downstream target AMP activated protein kinase (LKB1-AMPK), and the key nutrient sensor mammalian target of rapamycin complex 1 (mTORC1) form two signaling systems that coordinate metabolic and cellular activity with changes in the environment in order to preserve homeostasis. For example, nutritional fluctuations rapidly feed back on these signaling systems and thereby affect cell-specific functions. Recent studies have started to reveal important roles of these strategic metabolic regulators in Schwann cells for the trophic support and myelination of axons. Because aberrant intermediate metabolism along with mitochondrial dysfunction in Schwann cells is mechanistically linked to nerve abnormalities found in acquired and inherited peripheral neuropathies, manipulation of the LKB1-AMPK, and mTORC1 signaling hubs may be a worthwhile therapeutic target to mitigate nerve damage in disease. Here, recent advances in our understanding of LKB1-AMPK and mTORC1 functions in Schwann cells are covered, and future research areas for this key metabolic signaling network are proposed.
Collapse
Affiliation(s)
- Bogdan Beirowski
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14203, USA.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, 14214, USA
| |
Collapse
|
38
|
Yu R, Li Z, Liu S, Huwatibieke B, Li Y, Yin Y, Zhang W. Activation of mTORC1 signaling in gastric X/A-like cells induces spontaneous pancreatic fibrosis and derangement of glucose metabolism by reducing ghrelin production. EBioMedicine 2018; 36:304-315. [PMID: 30266297 PMCID: PMC6197745 DOI: 10.1016/j.ebiom.2018.09.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 09/15/2018] [Accepted: 09/15/2018] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Pancreatic fibrosis is a pathophysiological process associated with excessive deposition of extracellular matrix in pancreas, leading to reduced insulin secretion and derangement of glucose metabolism. X/A-like cells, a group of unique endocrine cells in gastric oxyntic mucosa, produce and secret ghrelin to influence energy balance. Whether gastric X/A-like cells affect pancreatic fibrosis and subsequent glucose homeostasis remains unclear. METHODS We established a Ghrl-cre transgene in which the cre enzyme is expressed in X/A-like cells under the control of ghrelin-promoter. TSC1flox/flox mice were bred with Ghrl-cre mice to generate Ghrl-TSC1-/- (TG) mice, within which mTORC1 signaling was activated in X/A-like cells. Pancreatic fibrosis and insulin secretion were analyzed in the TG mice. FINDINGS Activation of mTORC1 signaling by deletion of TSC1 gene in gastric X/A-like cells induced spontaneous pancreatic fibrosis. This alteration was associated with reduced insulin expression and secretion, as well as impaired glucose metabolism. Activation of mTORC1 signaling in gastric X/A-like cells reduced gastric and circulating ghrelin levels. Exogenous ghrelin reversed pancreatic fibrosis and glucose intolerance induced by activation of mTORC1 signaling in these cells. Rapamycin, an inhibitor of mTOR, reversed the decrease of ghrelin levels and pancreatic fibrosis. INTERPRETATION Activation of mTORC1 signaling in gastric X/A-like cells induces spontaneous pancreatic fibrosis and subsequently impairs glucose homeostasis via suppression of ghrelin.
Collapse
Affiliation(s)
- Ruili Yu
- School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Ziru Li
- School of Basic Medical Sciences, Peking University, Beijing 100191, China; Department of Surgery, University of Michigan Medical Center, Ann Arbor, MI 48109-0346, USA
| | - Shiying Liu
- School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | | | - Yin Li
- School of Basic Medical Sciences, Peking University, Beijing 100191, China
| | - Yue Yin
- School of Basic Medical Sciences, Peking University, Beijing 100191, China.
| | - Weizhen Zhang
- School of Basic Medical Sciences, Peking University, Beijing 100191, China.
| |
Collapse
|
39
|
Wu LY, Li M, Qu ML, Li X, Pi LH, Chen Z, Zhou SL, Yi XQ, Shi XJ, Wu J, Wang S. High glucose up-regulates Semaphorin 3A expression via the mTOR signaling pathway in keratinocytes: A potential mechanism and therapeutic target for diabetic small fiber neuropathy. Mol Cell Endocrinol 2018; 472:107-116. [PMID: 29203371 DOI: 10.1016/j.mce.2017.11.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 11/28/2017] [Accepted: 11/30/2017] [Indexed: 02/08/2023]
Abstract
Small fiber neuropathy (SFN) is a common complication in diabetes, and is characterized by decreased intraepidermal nerve fiber density (IENFD). Semaphorin 3A (Sema3A), which is produced by keratinocytes, has a chemorepulsive effect on intraepidermal nerve fibers. mTOR signaling can mediate local protein synthesis that is critical for growth of axons and dendrites. Therefore, this study aimed to investigate whether Sema3A is up-regulated in diabetic keratinocytes via the mTOR-mediated p70 S6K and 4E-BP1 signaling pathways, and furthermore whether it is involved in the pathogenesis of diabetic SFN. IENFD, expression of Sema3A, and mTOR signaling, were evaluated in the skin of diabetic patients with SFN as well as control subjects. Sema3A and mTOR signaling were also assessed in HaCaT cells which had been treated with high glucose (HG) or recombinant Sema3A (rSema3A) in the presence or absence of rapamycin. Small fiber dysfunction was evaluated by examining IENFD and using behavioral tests in control and streptozotocin-induced diabetic rats treated with or without rapamycin. We found that higher Sema3A expression and over-activation of mTOR signaling, was accompanied by reduced IENFD in the skin of diabetic patients compared with control subjects. The expression of Sema3A, and mTOR signaling were up-regulated in HaCaT cells incubated with HG or rSema3A, and this could be attenuated by rapamycin. Hyperalgesia, reduced IENFD, and up-regulated Sema3A and mTOR signaling were also detected in diabetic rats. These effects were ameliorated by rapamycin treatment. Our data indicate that HG up-regulates Sema3A expression by activating mTOR signaling in diabetic keratinocytes. This pathway may therefore play a critical role in diabetic SFN.
Collapse
Affiliation(s)
- Liang-Yan Wu
- Dept of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Mei Li
- Dept of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Min-Li Qu
- Dept of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Xin Li
- Dept of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China
| | - Lin-Hua Pi
- Dept of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Zi Chen
- Dept of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Shan-Lei Zhou
- Dept of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Xiao-Qing Yi
- Dept of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Xia-Jie Shi
- Dept of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China
| | - Jing Wu
- Dept of Endocrinology, Xiang-Ya Hospital, Central South University, Changsha, China.
| | - Shan Wang
- Dept of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, China.
| |
Collapse
|
40
|
Jiang M, Rao R, Wang J, Wang J, Xu L, Wu LM, Chan JR, Wang H, Lu QR. The TSC1-mTOR-PLK axis regulates the homeostatic switch from Schwann cell proliferation to myelination in a stage-specific manner. Glia 2018; 66:1947-1959. [PMID: 29722913 PMCID: PMC6185760 DOI: 10.1002/glia.23449] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 04/10/2018] [Accepted: 04/10/2018] [Indexed: 12/17/2022]
Abstract
Proper peripheral myelination depends upon the balance between Schwann cell proliferation and differentiation programs. The serine/threonine kinase mTOR integrates various environmental cues to serve as a central regulator of cell growth, metabolism, and function. We report here that tuberous sclerosis complex 1 (TSC1), a negative regulator of mTOR activity, establishes a stage-dependent program for Schwann cell lineage progression and myelination by controlling cell proliferation and myelin homeostasis. Tsc1 ablation in Schwann cell progenitors in mice resulted in activation of mTOR signaling, and caused over-proliferation of Schwann cells and blocked their differentiation, leading to hypomyelination. Transcriptome profiling analysis revealed that mTOR activation in Tsc1 mutants resulted in upregulation of a polo-like kinase (PLK)-dependent pathway and cell cycle regulators. Attenuation of mTOR or pharmacological inhibition of polo-like kinases partially rescued hypomyelination caused by Tsc1 loss in the developing peripheral nerves. In contrast, deletion of Tsc1 in mature Schwann cells led to redundant and overgrown myelin sheaths in adult mice. Together, our findings indicate stage-specific functions for the TSC1-mTOR-PLK signaling axis in controlling the transition from proliferation to differentiation and myelin homeostasis during Schwann cell development.
Collapse
Affiliation(s)
- Minqing Jiang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
- The Institute of Cognitive Neuroscience, East China Normal University, Shanghai, China
| | - Rohit Rao
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jincheng Wang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jiajia Wang
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Lingli Xu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Lai Man Wu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jonah R. Chan
- Department of Neurology and Programs in Biomedical and Neurosciences, University of California, San Francisco, CA 94158
| | - Huimin Wang
- The Institute of Cognitive Neuroscience, East China Normal University, Shanghai, China
| | - Q. Richard Lu
- Department of Pediatrics, Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
41
|
Shi Q, Saifetiarova J, Taylor AM, Bhat MA. mTORC1 Activation by Loss of Tsc1 in Myelinating Glia Causes Downregulation of Quaking and Neurofascin 155 Leading to Paranodal Domain Disorganization. Front Cell Neurosci 2018; 12:201. [PMID: 30050412 PMCID: PMC6052123 DOI: 10.3389/fncel.2018.00201] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 06/20/2018] [Indexed: 11/29/2022] Open
Abstract
Mutations in human tuberous sclerosis complex (TSC) genes TSC1 and TSC2 are the leading causes of developmental brain abnormalities and large tumors in other tissues. Murine Tsc1/2 have been shown to negatively regulate the mammalian target of rapamycin complex 1 (mTORC1) signaling pathway in most tissues, and this pathway has been shown to be essential for proper oligodendrocytes/Schwann cell differentiation and myelination. Here, we report that ablation of Tsc1 gene specifically in oligodendrocytes/Schwann cells activates mTORC1 signaling resulting in severe motor disabilities, weight loss, and early postnatal death. The mutant mice of either sex showed reduced myelination, disrupted paranodal domains in myelinated axons, and disorganized unmyelinated Remak bundles. mRNA and protein expression analyses revealed strong reduction in the RNA-binding protein Quaking (Qk) and the 155 kDa glial Neurofascin (NfascNF155). Re-introduction of exogenous Qk gene in Tsc1 mutant oligodendrocytes restored NfascNF155 protein levels indicating that Qk is required for the stabilization of NfascNF155 mRNA. Interestingly, injection of Rapamycin, a pharmacological mTORC1 inhibitor, to pregnant mothers increased the lifespan of the mutant offspring, restored myelination as well as the levels of Qk and NfascNF155, and consequently the organization of the paranodal domains. Together our studies show a critical role of mTORC1 signaling in the differentiation of myelinating glial cells and proper organization of axonal domains and provide insights into TSC-associated myelinated axon abnormalities.
Collapse
Affiliation(s)
| | | | | | - Manzoor A. Bhat
- Department of Cellular and Integrative Physiology, School of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, United States
| |
Collapse
|
42
|
Norrmén C, Figlia G, Pfistner P, Pereira JA, Bachofner S, Suter U. mTORC1 Is Transiently Reactivated in Injured Nerves to Promote c-Jun Elevation and Schwann Cell Dedifferentiation. J Neurosci 2018; 38:4811-4828. [PMID: 29695414 PMCID: PMC5956991 DOI: 10.1523/jneurosci.3619-17.2018] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 04/14/2018] [Accepted: 04/21/2018] [Indexed: 12/31/2022] Open
Abstract
Schwann cells (SCs) are endowed with a remarkable plasticity. When peripheral nerves are injured, SCs dedifferentiate and acquire new functions to coordinate nerve repair as so-called repair SCs. Subsequently, SCs redifferentiate to remyelinate regenerated axons. Given the similarities between SC dedifferentiation/redifferentiation in injured nerves and in demyelinating neuropathies, elucidating the signals involved in SC plasticity after nerve injury has potentially wider implications. c-Jun has emerged as a key transcription factor regulating SC dedifferentiation and the acquisition of repair SC features. However, the upstream pathways that control c-Jun activity after nerve injury are largely unknown. We report that the mTORC1 pathway is transiently but robustly reactivated in dedifferentiating SCs. By inducible genetic deletion of the functionally crucial mTORC1-subunit Raptor in mouse SCs (including male and female animals), we found that mTORC1 reactivation is necessary for proper myelin clearance, SC dedifferentiation, and consequently remyelination, without major alterations in the inflammatory response. In the absence of mTORC1 signaling, c-Jun failed to be upregulated correctly. Accordingly, a c-Jun binding motif was found to be enriched in promoters of genes with reduced expression in injured mutants. Furthermore, using cultured SCs, we found that mTORC1 is involved in c-Jun regulation by promoting its translation, possibly via the eIF4F-subunit eIF4A. These results provide evidence that proper c-Jun elevation after nerve injury involves also mTORC1-dependent post-transcriptional regulation to ensure timely dedifferentiation of SCs.SIGNIFICANCE STATEMENT A crucial evolutionary acquisition of vertebrates is the envelopment of axons in myelin sheaths produced by oligodendrocytes in the CNS and Schwann cells (SCs) in the PNS. When myelin is damaged, conduction of action potentials along axons slows down or is blocked, leading to debilitating diseases. Unlike oligodendrocytes, SCs have a high regenerative potential, granted by their remarkable plasticity. Thus, understanding the mechanisms underlying SC plasticity may uncover new therapeutic targets in nerve regeneration and demyelinating diseases. Our work reveals that reactivation of the mTORC1 pathway in SCs is essential for efficient SC dedifferentiation after nerve injury. Accordingly, modulating this signaling pathway might be of therapeutic relevance in peripheral nerve injury and other diseases.
Collapse
Affiliation(s)
- Camilla Norrmén
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology, ETH Zürich, Zürich CH-8093, Switzerland
| | - Gianluca Figlia
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology, ETH Zürich, Zürich CH-8093, Switzerland
| | - Patrick Pfistner
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology, ETH Zürich, Zürich CH-8093, Switzerland
| | - Jorge A Pereira
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology, ETH Zürich, Zürich CH-8093, Switzerland
| | - Sven Bachofner
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology, ETH Zürich, Zürich CH-8093, Switzerland
| | - Ueli Suter
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology, ETH Zürich, Zürich CH-8093, Switzerland
| |
Collapse
|
43
|
Sohn EJ, Park HT. MicroRNA Mediated Regulation of Schwann Cell Migration and Proliferation in Peripheral Nerve Injury. BIOMED RESEARCH INTERNATIONAL 2018; 2018:8198365. [PMID: 29854793 PMCID: PMC5952561 DOI: 10.1155/2018/8198365] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Accepted: 03/26/2018] [Indexed: 11/17/2022]
Abstract
Schwann cells (SCs) contribute to nerve repair following injury; however, the underlying molecular mechanism is poorly understood. MicroRNAs (miRNAs), which are short noncoding RNAs, have been shown to play a role in neuronal disease. In this work, we show that miRNAs regulate the peripheral nerve system by modulating the migration and proliferation of SCs. Thus, miRNAs expressed in peripheral nerves may provide a potential therapeutic target for peripheral nerve injury or repair.
Collapse
Affiliation(s)
- Eun Jung Sohn
- Peripheral Neuropathy Research Center, Department of Physiology, College of Medicine, Dong-A University, Busan, Republic of Korea
| | - Hwan Tae Park
- Peripheral Neuropathy Research Center, Department of Physiology, College of Medicine, Dong-A University, Busan, Republic of Korea
| |
Collapse
|
44
|
Bachiller S, Roca-Ceballos MA, García-Domínguez I, Pérez-Villegas EM, Martos-Carmona D, Pérez-Castro MÁ, Real LM, Rosa JL, Tabares L, Venero JL, Armengol JÁ, Carrión ÁM, Ruiz R. HERC1 Ubiquitin Ligase Is Required for Normal Axonal Myelination in the Peripheral Nervous System. Mol Neurobiol 2018; 55:8856-8868. [PMID: 29603094 DOI: 10.1007/s12035-018-1021-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 03/16/2018] [Indexed: 12/14/2022]
Abstract
A missense mutation in HERC1 provokes loss of cerebellar Purkinje cells, tremor, and unstable gait in tambaleante (tbl) mice. Recently, we have shown that before cerebellar degeneration takes place, the tbl mouse suffers from a reduction in the number of vesicles available for release at the neuromuscular junction (NMJ). The aim of the present work was to study to which extent the alteration in HERC1 may affect other cells in the nervous system and how this may influence the motor dysfunction observed in these mice. The functional analysis showed a consistent delay in the propagation of the action potential in mutant mice in comparison with control littermates. Morphological analyses of glial cells in motor axons revealed signs of compact myelin damage as tomacula and local hypermyelination foci. Moreover, we observed an alteration in non-myelinated terminal Schwann cells at the level of the NMJ. Additionally, we found a significant increment of phosphorylated Akt-2 in the sciatic nerve. Based on these findings, we propose a molecular model that could explain how mutated HERC1 in tbl mice affects the myelination process in the peripheral nervous system. Finally, since the myelin abnormalities found in tbl mice are histological hallmarks of neuropathic periphery diseases, tbl mutant mice could be considered as a new mouse model for this type of diseases.
Collapse
Affiliation(s)
- Sara Bachiller
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Seville, Spain
| | - María Angustias Roca-Ceballos
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Calle Profesor García González 2, 41012, Sevilla, Spain
| | - Irene García-Domínguez
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Calle Profesor García González 2, 41012, Sevilla, Spain
| | - Eva María Pérez-Villegas
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Seville, Spain
| | - David Martos-Carmona
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Seville, Spain
| | - Miguel Ángel Pérez-Castro
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Calle Profesor García González 2, 41012, Sevilla, Spain
| | - Luis Miguel Real
- Unit of Infectious Diseases and Microbiology, Valme University Hospital, Seville, Spain
| | - José Luis Rosa
- Departament de Ciències Fisiològiques II, IDIBELL, Campus de Bellvitge, Universitat de Barcelona, L'Hospitalet de Llobregat, E-08907, Barcelona, Spain
| | - Lucía Tabares
- Department of Medical Physiology and Biophysics, School of Medicine, University of Seville, Seville, Spain
| | - José Luis Venero
- Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Calle Profesor García González 2, 41012, Sevilla, Spain
| | - José Ángel Armengol
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Seville, Spain
| | - Ángel Manuel Carrión
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Seville, Spain
| | - Rocío Ruiz
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Seville, Spain. .,Department of Biochemistry and Molecular Biology, School of Pharmacy, University of Seville, and Instituto de Biomedicina de Sevilla-Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Calle Profesor García González 2, 41012, Sevilla, Spain.
| |
Collapse
|
45
|
Carlin D, Golden JP, Mogha A, Samineni VK, Monk KR, Gereau RW, Cavalli V. Deletion of Tsc2 in Nociceptors Reduces Target Innervation, Ion Channel Expression, and Sensitivity to Heat. eNeuro 2018; 5:ENEURO.0436-17.2018. [PMID: 29766046 PMCID: PMC5952427 DOI: 10.1523/eneuro.0436-17.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 04/13/2018] [Accepted: 04/16/2018] [Indexed: 01/10/2023] Open
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) is known to regulate cellular growth pathways, and its genetic activation is sufficient to enhance regenerative axon growth following injury to the central or peripheral nervous systems. However, excess mTORC1 activation may promote innervation defects, and mTORC1 activity mediates injury-induced hypersensitivity, reducing enthusiasm for the pathway as a therapeutic target. While mTORC1 activity is required for full expression of some pain modalities, the effects of pathway activation on nociceptor phenotypes and sensory behaviors are currently unknown. To address this, we genetically activated mTORC1 in mouse peripheral sensory neurons by conditional deletion of its negative regulator Tuberous Sclerosis Complex 2 (Tsc2). Consistent with the well-known role of mTORC1 in regulating cell size, soma size and axon diameter of C-nociceptors were increased in Tsc2-deleted mice. Glabrous skin and spinal cord innervation by C-fiber neurons were also disrupted. Transcriptional profiling of nociceptors enriched by fluorescence-associated cell sorting (FACS) revealed downregulation of multiple classes of ion channels as well as reduced expression of markers for peptidergic nociceptors in Tsc2-deleted mice. In addition to these changes in innervation and gene expression, Tsc2-deleted mice exhibited reduced noxious heat sensitivity and decreased injury-induced cold hypersensitivity, but normal baseline sensitivity to cold and mechanical stimuli. Together, these data show that excess mTORC1 activity in sensory neurons produces changes in gene expression, neuron morphology and sensory behavior.
Collapse
Affiliation(s)
- Dan Carlin
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110
| | - Judith P. Golden
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Amit Mogha
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110
| | - Vijay K. Samineni
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Kelly R. Monk
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110
| | - Robert W. Gereau
- Washington University Pain Center and Department of Anesthesiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Valeria Cavalli
- Department of Neuroscience, Hope Center for Neurological Disorders and Center of Regenerative Medicine, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
46
|
Wong KM, Beirowski B. Multiple lines of inhibitory feedback on AKT kinase in Schwann cells lacking TSC1/2 hint at distinct functions of mTORC1 and AKT in nerve development. Commun Integr Biol 2018; 11:e1433441. [PMID: 29497474 PMCID: PMC5824964 DOI: 10.1080/19420889.2018.1433441] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Revised: 01/02/2018] [Accepted: 01/19/2018] [Indexed: 11/29/2022] Open
Abstract
During nerve development, Schwann cells (SCs) build multilayered myelin sheaths around axons to accelerate nerve conduction. The mechanistic target of rapamycin complex 1 (mTORC1) downstream of PI3K/AKT signaling lately emerged as a central anabolic regulator of myelination. Using mutant mice with sustained mTORC1 hyperactivity in developing SCs we recently uncovered that mTORC1 impedes developmental myelination by promoting proliferation of immature SCs while antagonizing SC differentiation. In contrast, mTORC1 stimulates myelin production, rather than SC proliferation, in already differentiated SCs. Importantly, these diametrical mTORC1 functions were unmasked under settings of greatly suppressed AKT signaling. Here we demonstrate, inter alia, additional mechanisms of feedback inhibition of AKT by mTORC1, such as strikingly elevated PTEN levels in SCs with disruption of the mTORC1 inhibitory complex, TSC1/2. These data lead us to propose a model wherein mTORC1 and AKT have distinct roles in developing SCs that have to be precisely coordinated for normal myelinogenesis.
Collapse
Affiliation(s)
- Keit Men Wong
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Bogdan Beirowski
- Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA.,Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| |
Collapse
|
47
|
Figlia G, Gerber D, Suter U. Myelination and mTOR. Glia 2017; 66:693-707. [PMID: 29210103 PMCID: PMC5836902 DOI: 10.1002/glia.23273] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 11/08/2017] [Accepted: 11/17/2017] [Indexed: 02/06/2023]
Abstract
Myelinating cells surround axons to accelerate the propagation of action potentials, to support axonal health, and to refine neural circuits. Myelination is metabolically demanding and, consistent with this notion, mTORC1—a signaling hub coordinating cell metabolism—has been implicated as a key signal for myelination. Here, we will discuss metabolic aspects of myelination, illustrate the main metabolic processes regulated by mTORC1, and review advances on the role of mTORC1 in myelination of the central nervous system and the peripheral nervous system. Recent progress has revealed a complex role of mTORC1 in myelinating cells that includes, besides positive regulation of myelin growth, additional critical functions in the stages preceding active myelination. Based on the available evidence, we will also highlight potential nonoverlapping roles between mTORC1 and its known main upstream pathways PI3K‐Akt, Mek‐Erk1/2, and AMPK in myelinating cells. Finally, we will discuss signals that are already known or hypothesized to be responsible for the regulation of mTORC1 activity in myelinating cells. Myelination is metabolically demanding. The metabolic regulator mTORC1 controls differentiation of myelinating cells and promotes myelin
growth. mTORC1‐independent targets of the PI3K‐Akt and Mek‐Erk1/2 pathways may also be significant in myelination.
Collapse
Affiliation(s)
- Gianluca Figlia
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology, ETH Zürich, Zürich, CH 8093, Switzerland
| | - Daniel Gerber
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology, ETH Zürich, Zürich, CH 8093, Switzerland
| | - Ueli Suter
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology, ETH Zürich, Zürich, CH 8093, Switzerland
| |
Collapse
|
48
|
Figlia G, Norrmén C, Pereira JA, Gerber D, Suter U. Dual function of the PI3K-Akt-mTORC1 axis in myelination of the peripheral nervous system. eLife 2017; 6:e29241. [PMID: 28880149 PMCID: PMC5589416 DOI: 10.7554/elife.29241] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 07/31/2017] [Indexed: 01/24/2023] Open
Abstract
Myelination is a biosynthetically demanding process in which mTORC1, the gatekeeper of anabolism, occupies a privileged regulatory position. We have shown previously that loss of mTORC1 function in Schwann cells (SCs) hampers myelination. Here, we genetically disrupted key inhibitory components upstream of mTORC1, TSC1 or PTEN, in mouse SC development, adult homeostasis, and nerve injury. Surprisingly, the resulting mTORC1 hyperactivity led to markedly delayed onset of both developmental myelination and remyelination after injury. However, if mTORC1 was hyperactivated after myelination onset, radial hypermyelination was observed. At early developmental stages, physiologically high PI3K-Akt-mTORC1 signaling suppresses expression of Krox20 (Egr2), the master regulator of PNS myelination. This effect is mediated by S6K and contributes to control mechanisms that keep SCs in a not-fully differentiated state to ensure proper timing of myelination initiation. An ensuing decline in mTORC1 activity is crucial to allow myelination to start, while remaining mTORC1 activity drives myelin growth.
Collapse
Affiliation(s)
- Gianluca Figlia
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| | - Camilla Norrmén
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| | - Jorge A Pereira
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| | - Daniel Gerber
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| | - Ueli Suter
- Department of BiologyInstitute of Molecular Health Sciences, Swiss Federal Institute of TechnologyZürichSwitzerland
| |
Collapse
|
49
|
Affiliation(s)
- Bogdan Beirowski
- a Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo , Buffalo , NY , USA.,b Department of Biochemistry , Jacobs School of Medicine and Biomedical Sciences, University at Buffalo , Buffalo , NY , USA
| | - Keit Men Wong
- a Hunter James Kelly Research Institute, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo , Buffalo , NY , USA
| |
Collapse
|