1
|
Zhu H, Hu E, Guo X, Yuan Z, Jiang H, Zhang W, Tang T, Wang Y, Li T. Promoting remyelination in central nervous system diseases: Potentials and prospects of natural products and herbal medicine. Pharmacol Res 2024; 210:107533. [PMID: 39617281 DOI: 10.1016/j.phrs.2024.107533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/22/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024]
Abstract
Myelin damage is frequently associated with central nervous system (CNS) diseases and is a critical factor influencing neurological function and disease prognosis. Nevertheless, the majority of current treatments for the CNS concentrate on gray matter injury and repair strategies, while clinical interventions specifically targeting myelin repair remain unavailable. In recent years, natural products and herbal medicine have achieved considerable progress in the domain of myelin repair, given their remarkable curative effect and low toxic side effects, demonstrating significant therapeutic potential. In this review, we present a rather comprehensive account of the mechanisms underlying myelin formation, injury, and repair, with a particular emphasis on the interactions between oligodendrocytes and other glial cells. Furthermore, we summarize the natural products and herbal medicine currently employed in remyelination along with their mechanisms of action, highlighting the potential and challenges of certain natural compounds to enhance myelin repair. This review aims to facilitate the expedited development of innovative therapeutics derived from natural products and herbal medicine and furnish novel insights into myelin repair in the CNS.
Collapse
Affiliation(s)
- Haonan Zhu
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - En Hu
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Xin Guo
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Zhiqiang Yuan
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Haoying Jiang
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Wei Zhang
- The College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
| | - Tao Tang
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Yang Wang
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Teng Li
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China.
| |
Collapse
|
2
|
Fischer I, Shohat S, Leichtmann-Bardoogo Y, Nayak R, Wiener G, Rosh I, Shemen A, Tripathi U, Rokach M, Bar E, Hussein Y, Castro AC, Chen G, Soffer A, Schokoroy-Trangle S, Elad-Sfadia G, Assaf Y, Schroeder A, Monteiro P, Stern S, Maoz BM, Barak B. Shank3 mutation impairs glutamate signaling and myelination in ASD mouse model and human iPSC-derived OPCs. SCIENCE ADVANCES 2024; 10:eadl4573. [PMID: 39392881 PMCID: PMC11468907 DOI: 10.1126/sciadv.adl4573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 09/06/2024] [Indexed: 10/13/2024]
Abstract
Autism spectrum disorder (ASD) is characterized by social and neurocognitive impairments, with mutations of the SHANK3 gene being prominent in patients with monogenic ASD. Using the InsG3680 mouse model with a Shank3 mutation seen in humans, we revealed an unknown role for Shank3 in postsynaptic oligodendrocyte (OL) features, similar to its role in neurons. This was shown by impaired molecular and physiological glutamatergic traits of InsG3680-derived primary OL cultures. In vivo, InsG3680 mice exhibit significant reductions in the expression of key myelination-related transcripts and proteins, along with deficits in myelin ultrastructure, white matter, axonal conductivity, and motor skills. Last, we observed significant impairments, with clinical relevance, in induced pluripotent stem cell-derived OLs from a patient with the InsG3680 mutation. Together, our study provides insight into Shank3's role in OLs and reveals a mechanism of the crucial connection of myelination to ASD pathology.
Collapse
Affiliation(s)
- Inbar Fischer
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Sophie Shohat
- The School of Psychological Sciences, Faculty of Social Sciences, Tel Aviv University, Tel Aviv, Israel
- The School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yael Leichtmann-Bardoogo
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Ritu Nayak
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Gal Wiener
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Idan Rosh
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Aviram Shemen
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Utkarsh Tripathi
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - May Rokach
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Ela Bar
- The School of Psychological Sciences, Faculty of Social Sciences, Tel Aviv University, Tel Aviv, Israel
- The School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yara Hussein
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Ana Carolina Castro
- Department of Biomedicine–Experimental Biology Unit, Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Gal Chen
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion, Haifa, Israel
- The Interdisciplinary Program for Biotechnology, Technion, Haifa, Israel
| | - Adi Soffer
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
| | - Sari Schokoroy-Trangle
- The School of Psychological Sciences, Faculty of Social Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Galit Elad-Sfadia
- The School of Psychological Sciences, Faculty of Social Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Yaniv Assaf
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- The School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- The Strauss Center for Neuroimaging, Tel Aviv University, Tel Aviv, Israel
| | - Avi Schroeder
- The Louis Family Laboratory for Targeted Drug Delivery and Personalized Medicine Technologies, Department of Chemical Engineering, Technion, Haifa, Israel
| | - Patricia Monteiro
- Department of Biomedicine–Experimental Biology Unit, Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Shani Stern
- Sagol Department of Neurobiology, Faculty of Natural Sciences, University of Haifa, Haifa, Israel
| | - Ben M. Maoz
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- Department of Biomedical Engineering, Faculty of Engineering, Tel Aviv University, Tel Aviv, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv, Israel
- Sagol Center for Regenerative Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Boaz Barak
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
- The School of Psychological Sciences, Faculty of Social Sciences, Tel Aviv University, Tel Aviv, Israel
- The School of Neurobiology, Biochemistry and Biophysics, Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
3
|
Yang L, Peng J, Zhang L, Zhang F, Wu J, Zhang X, Pang J, Jiang Y. Advanced Diffusion Tensor Imaging in White Matter Injury After Subarachnoid Hemorrhage. World Neurosurg 2024; 189:77-88. [PMID: 38789033 DOI: 10.1016/j.wneu.2024.05.107] [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: 04/23/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024]
Abstract
Subarachnoid hemorrhage (SAH) is recognized as an especially severe stroke variant, notorious for its high mortality and long-term disability rates, in addition to a range of both immediate and enduring neurologic impacts. Over half of the SAH survivors experience varying degrees of neurologic disorders, with many enduring chronic neuropsychiatric conditions. Due to the limitations of traditional imaging techniques in depicting subtle changes within brain tissues posthemorrhage, the accurate detection and diagnosis of white matter (WM) injuries are complicated. Against this backdrop, diffusion tensor imaging (DTI) has emerged as a promising biomarker for structural imaging, renowned for its enhanced sensitivity in identifying axonal damage. This capability positions DTI as an invaluable tool for forming precise and expedient prognoses for SAH survivors. This study synthesizes an assessment of DTI for the diagnosis and prognosis of neurologic dysfunctions in patients with SAH, emphasizing the notable changes observed in DTI metrics and their association with potential pathophysiological processes. Despite challenges associated with scanning technology differences and data processing, DTI demonstrates significant clinical potential for early diagnosis of cognitive impairments following SAH and monitoring therapeutic effects. Future research requires the development of highly standardized imaging paradigms to enhance diagnostic accuracy and devise targeted therapeutic strategies for SAH patients. In sum, DTI technology not only augments our understanding of the impact of SAH but also may offer new avenues for improving patient prognoses.
Collapse
Affiliation(s)
- Lei Yang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China; Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jianhua Peng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China; Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China; Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lifang Zhang
- Institute of Brain Science, Southwest Medical University, Luzhou, China; Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Fan Zhang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China; Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jinpeng Wu
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China; Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xianhui Zhang
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jinwei Pang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yong Jiang
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China; Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China; Institute of Brain Science, Southwest Medical University, Luzhou, China; Sichuan Clinical Research Center for Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China.
| |
Collapse
|
4
|
Parmeggiani B, Signori MF, Cecatto C, Frusciante MR, Marcuzzo MB, Souza DG, Ribeiro RT, Seminotti B, Gomes de Souza DO, Ribeiro CAJ, Wajner M, Leipnitz G. Glycine disrupts myelin, glutamatergic neurotransmission, and redox homeostasis in a neonatal model for non ketotic hyperglycinemia. Biochimie 2024; 219:21-32. [PMID: 37541567 DOI: 10.1016/j.biochi.2023.07.022] [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: 04/10/2023] [Revised: 06/26/2023] [Accepted: 07/30/2023] [Indexed: 08/06/2023]
Abstract
Non ketotic hyperglycinemia (NKH) is an inborn error of glycine metabolism caused by mutations in the genes encoding glycine cleavage system proteins. Classic NKH has a neonatal onset, and patients present with severe neurodegeneration. Although glycine accumulation has been implicated in NKH pathophysiology, the exact mechanisms underlying the neurological damage and white matter alterations remain unclear. We investigated the effects of glycine in the brain of neonatal rats and MO3.13 oligodendroglial cells. Glycine decreased myelin basic protein (MBP) and myelin-associated glycoprotein (MAG) in the corpus callosum and striatum of rats on post-natal day (PND) 15. Glycine also reduced neuroglycan 2 (NG2) and N-methyl-d-aspartate receptor subunit 1 (NR1) in the cerebral cortex and striatum on PND15. Moreover, glycine reduced striatal glutamate aspartate transporter 1 (GLAST) content and neuronal nucleus (NeuN), and increased glial fibrillary acidic protein (GFAP) on PND15. Glycine also increased DCFH oxidation and malondialdehyde levels and decreased GSH concentrations in the cerebral cortex and striatum on PND6, but not on PND15. Glycine further reduced viability but did not alter DCFH oxidation and GSH levels in MO3.13 cells after 48- and 72-h incubation. These data indicate that impairment of myelin structure and glutamatergic system and induction of oxidative stress are involved in the neuropathophysiology of NKH.
Collapse
Affiliation(s)
- Belisa Parmeggiani
- Programa de Pós-Graduação em Ciências Biológicas - Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| | - Marian Flores Signori
- Programa de Pós-Graduação em Ciências Biológicas - Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Cristiane Cecatto
- Programa de Pós-Graduação em Ciências Biológicas - Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Marina Rocha Frusciante
- Programa de Pós-Graduação em Ciências Biológicas - Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Manuela Bianchin Marcuzzo
- Programa de Pós-Graduação em Ciências Biológicas - Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Débora Guerini Souza
- Programa de Pós-Graduação em Ciências Biológicas - Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Rafael Teixeira Ribeiro
- Programa de Pós-Graduação em Ciências Biológicas - Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Bianca Seminotti
- Programa de Pós-Graduação em Ciências Biológicas - Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Diogo Onofre Gomes de Souza
- Programa de Pós-Graduação em Ciências Biológicas - Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - César Augusto João Ribeiro
- Natural and Humanities Sciences Center, Universidade Federal do ABC, São Bernardo do Campo, SP, 09606-070, Brazil
| | - Moacir Wajner
- Programa de Pós-Graduação em Ciências Biológicas - Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil; Serviço de Genética Médica do Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Guilhian Leipnitz
- Programa de Pós-Graduação em Ciências Biológicas - Bioquímica, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
5
|
Zhou Y, Zhang J. Neuronal activity and remyelination: new insights into the molecular mechanisms and therapeutic advancements. Front Cell Dev Biol 2023; 11:1221890. [PMID: 37564376 PMCID: PMC10410458 DOI: 10.3389/fcell.2023.1221890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 07/18/2023] [Indexed: 08/12/2023] Open
Abstract
This article reviews the role of neuronal activity in myelin regeneration and the related neural signaling pathways. The article points out that neuronal activity can stimulate the formation and regeneration of myelin, significantly improve its conduction speed and neural signal processing ability, maintain axonal integrity, and support axonal nutrition. However, myelin damage is common in various clinical diseases such as multiple sclerosis, stroke, dementia, and schizophrenia. Although myelin regeneration exists in these diseases, it is often incomplete and cannot promote functional recovery. Therefore, seeking other ways to improve myelin regeneration in clinical trials in recent years is of great significance. Research has shown that controlling neuronal excitability may become a new intervention method for the clinical treatment of demyelinating diseases. The article discusses the latest research progress of neuronal activity on myelin regeneration, including direct or indirect stimulation methods, and the related neural signaling pathways, including glutamatergic, GABAergic, cholinergic, histaminergic, purinergic and voltage-gated ion channel signaling pathways, revealing that seeking treatment strategies to promote myelin regeneration through precise regulation of neuronal activity has broad prospects.
Collapse
Affiliation(s)
| | - Jing Zhang
- Department of Pharmacy, School of Medicine, Sir Run Run Shaw Hospital, Zhejiang University, Hangzhou, China
| |
Collapse
|
6
|
Temmermand R, Barrett JE, Fontana ACK. Glutamatergic systems in neuropathic pain and emerging non-opioid therapies. Pharmacol Res 2022; 185:106492. [PMID: 36228868 PMCID: PMC10413816 DOI: 10.1016/j.phrs.2022.106492] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/05/2022] [Accepted: 10/06/2022] [Indexed: 01/14/2023]
Abstract
Neuropathic pain, a disease of the somatosensory nervous system, afflicts many individuals and adequate management with current pharmacotherapies remains elusive. The glutamatergic system of neurons, receptors and transporters are intimately involved in pain but, to date, there have been few drugs developed that therapeutically modulate this system. Glutamate transporters, or excitatory amino acid transporters (EAATs), remove excess glutamate around pain transmitting neurons to decrease nociception suggesting that the modulation of glutamate transporters may represent a novel approach to the treatment of pain. This review highlights and summarizes (1) the physiology of the glutamatergic system in neuropathic pain, (2) the preclinical evidence for dysregulation of glutamate transport in animal pain models, and (3) emerging novel therapies that modulate glutamate transporters. Successful drug discovery requires continuous focus on basic and translational methods to fully elucidate the etiologies of this disease to enable the development of targeted therapies. Increasing the efficacy of astrocytic EAATs may serve as a new way to successfully treat those suffering from this devastating disease.
Collapse
Affiliation(s)
- Rhea Temmermand
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - James E Barrett
- Center for Substance Abuse Research, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Andréia C K Fontana
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA 19102, USA.
| |
Collapse
|
7
|
Wang Y, Zhu X, Wang K, Cai Y, Liu C, Pan J, Sun J, Liu T, Huang Y, Li Y, Lu Y. Cell Metabolomics Study on Synergistic anti-Hepatocellular Carcinoma Effect of Aidi Injection Combined with Doxorubicin. Biomed Chromatogr 2022; 36:e5451. [PMID: 35848595 DOI: 10.1002/bmc.5451] [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: 04/06/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 11/06/2022]
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer worldwide and the second most common cause of cancer deaths. This study aimed to explore the inhibitory effect and mechanism of Aidi injection (ADI) combined with doxorubicin (DOX) in HCC treatment. The drug concentrations in combined threapy was determined by investigating the effect of various concentrations of ADI and DOX on the viability of H22 cells. The combination index (CI) was also calculated. A metabolomic strategy based on ultrahigh performance liquid chromatography tandem mass spectrometry (UHPLC-MS/MS) platform was established to analyze the metabolites. As a result, the CI values were less than 1, indicating that the combination of ADI and DOX exerted a synergistic effect on HCC treatment. The combination of 40‰ ADI and 1 μmol/L DOX had the strongest inhibitory effect and was used for subsequent metabolomic analysis. A total of 19 metabolic markers were obtained in metabolomic analysis, including amino acids (L-glutamic acid, L-arginine, and L-tyrosine), organic acids (succinic acid and citric acid), adenosine, and hypoxanthine , etc. Compared with the treatment using DOX or ADI alone, the combined therapy further regulated the levels of metabolic markers in HCC, which may be the reason for the synergistic effect. Seven metabolic pathways were significantly enriched, including phenylalanine, tyrosine and tryptophan biosynthesis, D-glutamine and D-glutamate metabolism, alanine, aspartate and glutamate metabolism, phenylalanine metabolism, arginine biosynthesis, tricarboxylic acid (TCA) cycle, and purine metabolism. These findings demonstrated that ADI combined with DOX can effectively inhibit the viability of H22 cells, which may exert a synergistic anti-tumor effect by balancing the metabolism of amino acids and energy-related substances.
Collapse
Affiliation(s)
- Yanli Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China.,School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Xiaoqing Zhu
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Kailiang Wang
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Ying Cai
- School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Chunhua Liu
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, China
| | - Jie Pan
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, China
| | - Jia Sun
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China
| | - Ting Liu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China
| | - Yong Huang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China
| | - Yongjun Li
- Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Medical University, Guiyang, China.,School of Pharmacy, Guizhou Medical University, Guiyang, China
| | - Yuan Lu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Provincial Key Laboratory of Pharmaceutics, Guizhou Medical University, Guiyang, China
| |
Collapse
|
8
|
Sonkodi B, Hegedűs Á, Kopper B, Berkes I. Significantly Delayed Medium-Latency Response of the Stretch Reflex in Delayed-Onset Muscle Soreness of the Quadriceps Femoris Muscles Is Indicative of Sensory Neuronal Microdamage. J Funct Morphol Kinesiol 2022; 7:jfmk7020043. [PMID: 35736014 PMCID: PMC9224667 DOI: 10.3390/jfmk7020043] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/24/2022] [Accepted: 05/26/2022] [Indexed: 11/16/2022] Open
Abstract
Unaccustomed or strenuous eccentric exercise is known to cause delayed-onset muscle soreness. A recent hypothesis postulated that mechano-energetic microinjury of the primary afferent sensory neuron terminals in the muscle spindles, namely a transient Piezo2 channelopathy, could be the critical cause of delayed-onset muscle soreness in the form of a bi-phasic non-contact injury mechanism. This theory includes that this microlesion could delay the medium-latency response of the stretch reflex. Our aim with this study was to investigate this hypothesis. According to our knowledge, no study has examined the effect of delayed-onset muscle soreness on the medium-latency response of the stretch reflex. Our findings demonstrated that a significant delay in the medium-latency stretch reflex could be observed right after a multi-stage fitness test in the quadriceps femoris muscles of Hungarian professional handball players who consequently experienced delayed-onset muscle soreness. The long-latency stretch reflex and most likely short-latency stretch reflex were unaffected by delayed-onset muscle soreness in our study, which is in line with earlier findings. We translate these findings as indicative of proprioceptive Type Ia terminal microdamage in the muscle spindle in line with the aforementioned new acute non-contact compression axonopathy theory of delayed-onset muscles soreness.
Collapse
Affiliation(s)
- Balázs Sonkodi
- Department of Health Sciences and Sport Medicine, Hungarian University of Sport Science, 1123 Budapest, Hungary;
- Correspondence:
| | - Ádám Hegedűs
- Faculty of Kinesiology, Hungarian University of Sport Science, 1123 Budapest, Hungary; (Á.H.); (B.K.)
| | - Bence Kopper
- Faculty of Kinesiology, Hungarian University of Sport Science, 1123 Budapest, Hungary; (Á.H.); (B.K.)
| | - István Berkes
- Department of Health Sciences and Sport Medicine, Hungarian University of Sport Science, 1123 Budapest, Hungary;
| |
Collapse
|
9
|
Turan F, Yilmaz Ö, Schünemann L, Lindenberg TT, Kalanithy JC, Harder A, Ahmadi S, Duman T, MacDonald RB, Winter D, Liu C, Odermatt B. Effect of modulating glutamate signaling on myelinating oligodendrocytes and their development-A study in the zebrafish model. J Neurosci Res 2021; 99:2774-2792. [PMID: 34520578 DOI: 10.1002/jnr.24940] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 07/12/2021] [Accepted: 07/21/2021] [Indexed: 01/02/2023]
Abstract
Myelination is crucial for the development and maintenance of axonal integrity, especially fast axonal action potential conduction. There is increasing evidence that glutamate signaling and release through neuronal activity modulates the myelination process. In this study, we examine the effect of manipulating glutamate signaling on myelination of oligodendrocyte (OL) lineage cells and their development in zebrafish (zf). We use the "intensity-based glutamate-sensing fluorescent reporter" (iGluSnFR) in the zf model (both sexes) to address the hypothesis that glutamate is implicated in regulation of myelinating OLs. Our results show that glial iGluSnFR expression significantly reduces OL lineage cell number and the expression of myelin markers in larvae (zfl) and adult brains. The specific glutamate receptor agonist, L-AP4, rescues this iGluSnFR effect by significantly increasing the expression of the myelin-related genes, plp1b and mbpa, and enhances myelination in L-AP4-injected zfl compared to controls. Furthermore, we demonstrate that degrading glutamate using Glutamat-Pyruvate Transaminase (GPT) or the blockade of glutamate reuptake by L-trans-pyrrolidine-2,4-dicarboxylate (PDC) significantly decreases myelin-related genes and drastically declines myelination in brain ventricle-injected zfl. Moreover, we found that myelin-specific ClaudinK (CldnK) and 36K protein expression is significantly decreased in iGluSnFR-expressing zfl and adult brains compared to controls. Taken together, this study confirms that glutamate signaling is directly required for the preservation of myelinating OLs and for the myelination process itself. These findings further suggest that glutamate signaling may provide novel targets to therapeutically boost remyelination in several demyelinating diseases of the CNS.
Collapse
Affiliation(s)
- Funda Turan
- Medical Faculty, Institute of Neuroanatomy, University of Bonn, Bonn, Germany.,Faculty of Science, Biology Department, Ankara University, Ankara, Turkey
| | - Öznur Yilmaz
- Medical Faculty, Institute of Anatomy and Cell-Biology, University of Bonn, Bonn, Germany
| | - Lena Schünemann
- Medical Faculty, Institute of Anatomy and Cell-Biology, University of Bonn, Bonn, Germany
| | - Tobias T Lindenberg
- Medical Faculty, Institute of Neuroanatomy, University of Bonn, Bonn, Germany
| | - Jeshurun C Kalanithy
- Medical Faculty, Institute of Anatomy and Cell-Biology, University of Bonn, Bonn, Germany
| | - Alexander Harder
- Institute of Physical and Theoretical Chemistry, University of Bonn, Bonn, Germany
| | - Shiva Ahmadi
- Medical Faculty, Institute for Biochemistry and Molecular Biology (IBMB), University of Bonn, Bonn, Germany
| | - Türker Duman
- Faculty of Science, Biology Department, Ankara University, Ankara, Turkey
| | - Ryan B MacDonald
- Institute of Ophthalmology, University College London, London, UK
| | - Dominic Winter
- Medical Faculty, Institute for Biochemistry and Molecular Biology (IBMB), University of Bonn, Bonn, Germany
| | - Changsheng Liu
- Medical Faculty, Institute of Anatomy and Cell-Biology, University of Bonn, Bonn, Germany
| | - Benjamin Odermatt
- Medical Faculty, Institute of Neuroanatomy, University of Bonn, Bonn, Germany.,Medical Faculty, Institute of Anatomy and Cell-Biology, University of Bonn, Bonn, Germany
| |
Collapse
|
10
|
Monje M, Káradóttir RT. The bright and the dark side of myelin plasticity: Neuron-glial interactions in health and disease. Semin Cell Dev Biol 2021; 116:10-15. [PMID: 33293232 PMCID: PMC8178421 DOI: 10.1016/j.semcdb.2020.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 11/24/2020] [Accepted: 11/26/2020] [Indexed: 12/16/2022]
Abstract
Neuron-glial interactions shape neural circuit establishment, refinement and function. One of the key neuron-glial interactions takes place between axons and oligodendroglial precursor cells. Interactions between neurons and oligodendrocyte precursor cells (OPCs) promote OPC proliferation, generation of new oligodendrocytes and myelination, shaping myelin development and ongoing adaptive myelin plasticity in the brain. Communication between neurons and OPCs can be broadly divided into paracrine and synaptic mechanisms. Following the Nobel mini-symposium "The Dark Side of the Brain" in late 2019 at the Karolinska Institutet, this mini-review will focus on the bright and dark sides of neuron-glial interactions and discuss paracrine and synaptic interactions between neurons and OPCs and their malignant counterparts.
Collapse
Affiliation(s)
- Michelle Monje
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
| | - Ragnhildur Thóra Káradóttir
- Wellcome - Medical Research Council Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, UK; Department of Physiology, BioMedical Center, Faculty of Medicine, University of Iceland, Reykjavik, Iceland.
| |
Collapse
|
11
|
Kirdajova D, Valihrach L, Valny M, Kriska J, Krocianova D, Benesova S, Abaffy P, Zucha D, Klassen R, Kolenicova D, Honsa P, Kubista M, Anderova M. Transient astrocyte-like NG2 glia subpopulation emerges solely following permanent brain ischemia. Glia 2021; 69:2658-2681. [PMID: 34314531 PMCID: PMC9292252 DOI: 10.1002/glia.24064] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 12/13/2022]
Abstract
NG2 glia display wide proliferation and differentiation potential under physiological and pathological conditions. Here, we examined these two features following different types of brain disorders such as focal cerebral ischemia (FCI), cortical stab wound (SW), and demyelination (DEMY) in 3‐month‐old mice, in which NG2 glia are labeled by tdTomato under the Cspg4 promoter. To compare NG2 glia expression profiles following different CNS injuries, we employed single‐cell RT‐qPCR and self‐organizing Kohonen map analysis of tdTomato‐positive cells isolated from the uninjured cortex/corpus callosum and those after specific injury. Such approach enabled us to distinguish two main cell populations (NG2 glia, oligodendrocytes), each of them comprising four distinct subpopulations. The gene expression profiling revealed that a subpopulation of NG2 glia expressing GFAP, a marker of reactive astrocytes, is only present transiently after FCI. However, following less severe injuries, namely the SW and DEMY, subpopulations mirroring different stages of oligodendrocyte maturation markedly prevail. Such injury‐dependent incidence of distinct subpopulations was also confirmed by immunohistochemistry. To characterize this unique subpopulation of transient astrocyte‐like NG2 glia, we used single‐cell RNA‐sequencing analysis and to disclose their basic membrane properties, the patch‐clamp technique was employed. Overall, we have proved that astrocyte‐like NG2 glia are a specific subpopulation of NG2 glia emerging transiently only following FCI. These cells, located in the postischemic glial scar, are active in the cell cycle and display a current pattern similar to that identified in cortical astrocytes. Astrocyte‐like NG2 glia may represent important players in glial scar formation and repair processes, following ischemia.
Collapse
Affiliation(s)
- Denisa Kirdajova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic.,Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Lukas Valihrach
- Laboratory of Gene Expression, Institute of Biotechnology CAS, BIOCEV, Vestec, Czech Republic
| | - Martin Valny
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Jan Kriska
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Daniela Krocianova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Sarka Benesova
- Laboratory of Gene Expression, Institute of Biotechnology CAS, BIOCEV, Vestec, Czech Republic.,Faculty of Chemical Technology, Laboratory of Informatics and Chemistry, University of Chemistry and Technology, Prague, Czech Republic
| | - Pavel Abaffy
- Laboratory of Gene Expression, Institute of Biotechnology CAS, BIOCEV, Vestec, Czech Republic
| | - Daniel Zucha
- Laboratory of Gene Expression, Institute of Biotechnology CAS, BIOCEV, Vestec, Czech Republic.,Faculty of Science, Charles University, Prague, Czech Republic
| | - Ruslan Klassen
- Laboratory of Gene Expression, Institute of Biotechnology CAS, BIOCEV, Vestec, Czech Republic
| | - Denisa Kolenicova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic.,Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Pavel Honsa
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic
| | - Mikael Kubista
- Laboratory of Gene Expression, Institute of Biotechnology CAS, BIOCEV, Vestec, Czech Republic
| | - Miroslava Anderova
- Department of Cellular Neurophysiology, Institute of Experimental Medicine, Czech Academy of Sciences, Prague, Czech Republic.,Second Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
12
|
Ru X, Gao L, Zhou J, Li Q, Zuo S, Chen Y, Liu Z, Feng H. Secondary White Matter Injury and Therapeutic Targets After Subarachnoid Hemorrhage. Front Neurol 2021; 12:659740. [PMID: 34335439 PMCID: PMC8319471 DOI: 10.3389/fneur.2021.659740] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/11/2021] [Indexed: 01/19/2023] Open
Abstract
Aneurysmal subarachnoid hemorrhage (SAH) is one of the special stroke subtypes with high mortality and mobility. Although the mortality of SAH has decreased by 50% over the past two decades due to advances in neurosurgery and management of neurocritical care, more than 70% of survivors suffer from varying degrees of neurological deficits and cognitive impairments, leaving a heavy burden on individuals, families, and the society. Recent studies have shown that white matter is vulnerable to SAH, and white matter injuries may be one of the causes of long-term neurological deficits caused by SAH. Attention has recently focused on the pivotal role of white matter injury in the pathophysiological processes after SAH, mainly related to mechanical damage caused by increased intracerebral pressure and the metabolic damage induced by blood degradation and hypoxia. In the present review, we sought to summarize the pathophysiology processes and mechanisms of white matter injury after SAH, with a view to providing new strategies for the prevention and treatment of long-term cognitive dysfunction after SAH.
Collapse
Affiliation(s)
- Xufang Ru
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Ling Gao
- Department of General Practice, Audio-Visual Education Center, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jiru Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qiang Li
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shilun Zuo
- Department of Neurology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yujie Chen
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Zhi Liu
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hua Feng
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.,Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
13
|
Kohnke S, Buller S, Nuzzaci D, Ridley K, Lam B, Pivonkova H, Bentsen MA, Alonge KM, Zhao C, Tadross J, Holmqvist S, Shimizu T, Hathaway H, Li H, Macklin W, Schwartz MW, Richardson WD, Yeo GSH, Franklin RJM, Karadottir RT, Rowitch DH, Blouet C. Nutritional regulation of oligodendrocyte differentiation regulates perineuronal net remodeling in the median eminence. Cell Rep 2021; 36:109362. [PMID: 34260928 PMCID: PMC8293628 DOI: 10.1016/j.celrep.2021.109362] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 04/26/2021] [Accepted: 06/17/2021] [Indexed: 12/13/2022] Open
Abstract
The mediobasal hypothalamus (MBH; arcuate nucleus of the hypothalamus [ARH] and median eminence [ME]) is a key nutrient sensing site for the production of the complex homeostatic feedback responses required for the maintenance of energy balance. Here, we show that refeeding after an overnight fast rapidly triggers proliferation and differentiation of oligodendrocyte progenitors, leading to the production of new oligodendrocytes in the ME specifically. During this nutritional paradigm, ME perineuronal nets (PNNs), emerging regulators of ARH metabolic functions, are rapidly remodeled, and this process requires myelin regulatory factor (Myrf) in oligodendrocyte progenitors. In genetically obese ob/ob mice, nutritional regulations of ME oligodendrocyte differentiation and PNN remodeling are blunted, and enzymatic digestion of local PNN increases food intake and weight gain. We conclude that MBH PNNs are required for the maintenance of energy balance in lean mice and are remodeled in the adult ME by the nutritional control of oligodendrocyte differentiation.
Collapse
Affiliation(s)
- Sara Kohnke
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Sophie Buller
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Danae Nuzzaci
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Katherine Ridley
- Department of Paediatrics and Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Brian Lam
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Helena Pivonkova
- Wellcome-MRC Cambridge Stem Cell Institute, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Marie A Bentsen
- University of Washington Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Kimberly M Alonge
- University of Washington Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Chao Zhao
- Wellcome-MRC Cambridge Stem Cell Institute, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - John Tadross
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Staffan Holmqvist
- Department of Paediatrics and Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Takahiro Shimizu
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Hannah Hathaway
- Department of Cell & Developmental Biology and Program in Neuroscience, University of Colorado School of Medicine, Aurora, CO, USA
| | - Huiliang Li
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Wendy Macklin
- Department of Cell & Developmental Biology and Program in Neuroscience, University of Colorado School of Medicine, Aurora, CO, USA
| | - Michael W Schwartz
- University of Washington Medicine Diabetes Institute, Department of Medicine, University of Washington, Seattle, WA, USA
| | - William D Richardson
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Giles S H Yeo
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Robin J M Franklin
- Wellcome-MRC Cambridge Stem Cell Institute, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Ragnhildur T Karadottir
- Wellcome-MRC Cambridge Stem Cell Institute, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - David H Rowitch
- Department of Paediatrics and Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK; Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Clemence Blouet
- MRC Metabolic Diseases Unit, University of Cambridge Metabolic Research Laboratories, WT-MRC Institute of Metabolic Science, University of Cambridge, Cambridge CB2 0QQ, UK.
| |
Collapse
|
14
|
Ion Channels as New Attractive Targets to Improve Re-Myelination Processes in the Brain. Int J Mol Sci 2021; 22:ijms22147277. [PMID: 34298893 PMCID: PMC8305962 DOI: 10.3390/ijms22147277] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/20/2022] Open
Abstract
Multiple sclerosis (MS) is the most demyelinating disease of the central nervous system (CNS) characterized by neuroinflammation. Oligodendrocyte progenitor cells (OPCs) are cycling cells in the developing and adult CNS that, under demyelinating conditions, migrate to the site of lesions and differentiate into mature oligodendrocytes to remyelinate damaged axons. However, this process fails during disease chronicization due to impaired OPC differentiation. Moreover, OPCs are crucial players in neuro-glial communication as they receive synaptic inputs from neurons and express ion channels and neurotransmitter/neuromodulator receptors that control their maturation. Ion channels are recognized as attractive therapeutic targets, and indeed ligand-gated and voltage-gated channels can both be found among the top five pharmaceutical target groups of FDA-approved agents. Their modulation ameliorates some of the symptoms of MS and improves the outcome of related animal models. However, the exact mechanism of action of ion-channel targeting compounds is often still unclear due to the wide expression of these channels on neurons, glia, and infiltrating immune cells. The present review summarizes recent findings in the field to get further insights into physio-pathophysiological processes and possible therapeutic mechanisms of drug actions.
Collapse
|
15
|
Aguado T, Huerga-Gómez A, Sánchez-de la Torre A, Resel E, Chara JC, Matute C, Mato S, Galve-Roperh I, Guzman M, Palazuelos J. Δ 9 -Tetrahydrocannabinol promotes functional remyelination in the mouse brain. Br J Pharmacol 2021; 178:4176-4192. [PMID: 34216154 DOI: 10.1111/bph.15608] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/07/2021] [Accepted: 06/18/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND PURPOSE Research on demyelinating disorders aims to find novel molecules that are able to induce oligodendrocyte precursor cell differentiation to promote central nervous system remyelination and functional recovery. Δ9 -Tetrahydrocannabinol (THC), the most prominent active constituent of the hemp plant Cannabis sativa, confers neuroprotection in animal models of demyelination. However, the possible effect of THC on myelin repair has never been studied. EXPERIMENTAL APPROACH By using oligodendroglia-specific reporter mouse lines in combination with two models of toxin-induced demyelination, we analysed the effect of THC on the processes of oligodendrocyte regeneration and functional remyelination. KEY RESULTS We show that THC administration enhanced oligodendrocyte regeneration, white matter remyelination and motor function recovery. THC also promoted axonal remyelination in organotypic cerebellar cultures. THC remyelinating action relied on the induction of oligodendrocyte precursor differentiation upon cell cycle exit and via CB1 cannabinoid receptor activation. CONCLUSIONS AND IMPLICATIONS Overall, our study identifies THC administration as a promising pharmacological strategy aimed to promote functional CNS remyelination in demyelinating disorders.
Collapse
Affiliation(s)
- Tania Aguado
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación en Neuroquímica (IUIN), Complutense University, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Alba Huerga-Gómez
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación en Neuroquímica (IUIN), Complutense University, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Aníbal Sánchez-de la Torre
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación en Neuroquímica (IUIN), Complutense University, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Eva Resel
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación en Neuroquímica (IUIN), Complutense University, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Juan Carlos Chara
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Neurosciences, University of the Basque Country UPV/EHU and Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Carlos Matute
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Neurosciences, University of the Basque Country UPV/EHU and Achucarro Basque Center for Neuroscience, Leioa, Spain.,Biocruces, Barakaldo, Spain
| | - Susana Mato
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Department of Neurosciences, University of the Basque Country UPV/EHU and Achucarro Basque Center for Neuroscience, Leioa, Spain.,Biocruces, Barakaldo, Spain
| | - Ismael Galve-Roperh
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación en Neuroquímica (IUIN), Complutense University, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Manuel Guzman
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación en Neuroquímica (IUIN), Complutense University, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Javier Palazuelos
- Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain.,Department of Biochemistry and Molecular Biology, Instituto Universitario de Investigación en Neuroquímica (IUIN), Complutense University, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| |
Collapse
|
16
|
Roth LM, Akay-Espinoza C, Grinspan JB, Jordan-Sciutto KL. HIV-induced neuroinflammation inhibits oligodendrocyte maturation via glutamate-dependent activation of the PERK arm of the integrated stress response. Glia 2021; 69:2252-2271. [PMID: 34058792 DOI: 10.1002/glia.24033] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 12/15/2022]
Abstract
Despite combined antiretroviral therapy (cART), HIV-associated neurocognitive disorder (HAND) affects 30-50% of HIV-positive patients. Importantly, persistent white matter pathologies, specifically corpus callosum thinning and disruption of white matter microstructures observed in patients with HAND despite viral control through cART, raise the possibility that HIV infection in the setting of suboptimal cART may perturb oligodendrocyte (OL) maturation, function and/or survival, influencing HAND persistence in the cART era. To examine the effect of HIV infection on OL maturation, we used supernatants of primary human monocyte-derived macrophages infected with HIV (HIV/MDMs) to treat primary cultures of rat oligodendrocyte precursor cells (OPCs) during their differentiation to mature OLs. Using immunostaining for lineage-specific markers, we found that HIV/MDMs significantly inhibited OPC maturation. Based on our previous studies, we examined the potential role of several signaling pathways, including ionotropic glutamate receptors and the integrated stress response (ISR), and found that AMPA receptors (AMPAR)/kainic acid (KA) receptors (KARs) mediated the HIV/MDMs-induced defect in OL maturation. We also found that the treatment of OPC cultures with glutamate or AMPAR/KAR agonists phenocopied this effect. Blocking ISR activation, specifically the PERK arm of the ISR, protected OPCs from HIV/MDMs-mediated inhibition of OL maturation. Further, while glutamate, AMPA, and KA activated the ISR, inhibition of AMPAR/KAR activation prevented ISR induction in OPCs and rescued OL maturation. Collectively, these data identify glutamate signaling via ISR activation as a potential therapeutic pathway to ameliorate white matter pathologies in HAND and highlight the need for further investigation of their contribution to cognitive impairment.
Collapse
Affiliation(s)
- Lindsay M Roth
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.,Department of Neurology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Cagla Akay-Espinoza
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Judith B Grinspan
- Department of Neurology, The Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Kelly L Jordan-Sciutto
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
17
|
GluA2 overexpression in oligodendrocyte progenitors promotes postinjury oligodendrocyte regeneration. Cell Rep 2021; 35:109147. [PMID: 34010640 PMCID: PMC8185898 DOI: 10.1016/j.celrep.2021.109147] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 01/28/2020] [Accepted: 04/27/2021] [Indexed: 01/18/2023] Open
Abstract
Oligodendrocyte precursor cells (OPCs) are essential for developmental myelination and oligodendrocyte regeneration after CNS injury. These progenitors express calcium-permeable AMPA receptors (AMPARs) and form direct synapses with neurons throughout the CNS, but the roles of this signaling are unclear. To enable selective alteration of the properties of AMPARs in oligodendroglia, we generate mice that allow cell-specific overexpression of EGFP-GluA2 in vivo. In healthy conditions, OPC-specific GluA2 overexpression significantly increase their proliferation in an age-dependent manner but did not alter their rate of differentiation into oligodendrocytes. In contrast, after demyelinating brain injury in neonates or adults, higher GluA2 levels promote both OPC proliferation and oligodendrocyte regeneration, but do not prevent injury-induced initial cell loss. These findings indicate that AMPAR GluA2 content regulates the proliferative and regenerative behavior of adult OPCs, serving as a putative target for better myelin repair.
Collapse
|
18
|
Schumacher T, Röntgen M, Maak S. Congenital Splay Leg Syndrome in Piglets-Current Knowledge and a New Approach to Etiology. Front Vet Sci 2021; 8:609883. [PMID: 33718467 PMCID: PMC7952305 DOI: 10.3389/fvets.2021.609883] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/25/2021] [Indexed: 11/13/2022] Open
Abstract
The porcine congenital splay leg syndrome (PCS), even though being of transient nature, is still one of the most important causes for piglet losses due to its high incidence and mortality. Although, described decades ago, the pathogenetic mechanism is still elusive. Numerous, mostly descriptive studies characterized the syndrome at clinical, histological and cellular levels but resulted in a highly diverse picture of the syndrome. Broad variability in phenotypical expression and, in case of proper care, the rapid recovery of affected animals complicated a systematical analysis of the underlying pathogenesis. Although, several environmental factors were discussed as potential causes of PCS, most of the evidence points to a hereditary basis of PCS. Nevertheless, only few of the suggested candidate genes from transcriptome and mapping analyses, like F-box protein 32 (FBXO32), could be confirmed so far. Only recently, a genome wide association study revealed genomic regions on five porcine chromosomes and named a number of potential candidate genes, among them homer scaffold protein 1 (HOMER1). This new candidate-a cellular scaffold protein-plays a role in a plethora of cellular signaling cascades, and is not only involved in skeletal muscle differentiation but also critical for muscular function. In this review, we critically elucidate the current state of knowledge in the field and evaluate current achievements in the identification of the pathogenetic mechanism for the syndrome.
Collapse
Affiliation(s)
| | | | - Steffen Maak
- Institute of Muscle Biology and Growth, Leibniz Institute for Farm Animal Biology, Dummerstorf, Germany
| |
Collapse
|
19
|
Sonkodi B. Delayed Onset Muscle Soreness (DOMS): The Repeated Bout Effect and Chemotherapy-Induced Axonopathy May Help Explain the Dying-Back Mechanism in Amyotrophic Lateral Sclerosis and Other Neurodegenerative Diseases. Brain Sci 2021; 11:brainsci11010108. [PMID: 33467407 PMCID: PMC7830646 DOI: 10.3390/brainsci11010108] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/07/2021] [Accepted: 01/13/2021] [Indexed: 12/12/2022] Open
Abstract
Delayed onset muscle soreness (DOMS) is hypothesized to be caused by glutamate excitotoxicity-induced acute compression axonopathy of the sensory afferents in the muscle spindle. Degeneration of the same sensory afferents is implicated in the disease onset and progression of amyotrophic lateral sclerosis (ALS). A series of “silent” acute compression proprioceptive axonopathies with underlying genetic/environmental factors, damaging eccentric contractions and the non-resolving neuroinflammatory process of aging could lead to ALS disease progression. Since the sensory terminals in the muscle spindle could not regenerate from the micro-damage in ALS, unlike in DOMS, the induced protective microcircuits and their long-term functional plasticity (the equivalent of the repeated bout effect in DOMS) will be dysfunctional. The acute stress invoking osteocalcin, bradykinin, COX1, COX2, GDNF, PGE2, NGF, glutamate and N-methyl-D-aspartate (NMDA) receptors are suggested to be the critical signalers of this theory. The repeated bout effect of DOMS and the dysfunctional microcircuits in ALS are suggested to involve several dimensions of memory and learning, like pain memory, inflammation, working and episodic memory. The spatial encoding of these memory dimensions is compromised in ALS due to blunt position sense from the degenerating proprioceptive axon terminals of the affected muscle spindles. Dysfunctional microcircuits progressively and irreversibly interfere with postural control, with motor command and locomotor circuits, deplete the neuroenergetic system, and ultimately interfere with life-sustaining central pattern generators in ALS. The activated NMDA receptor is suggested to serve the “gate control” function in DOMS and ALS in line with the gate control theory of pain. Circumvention of muscle spindle-loading could be a choice of exercise therapy in muscle spindle-affected neurodegenerative diseases.
Collapse
Affiliation(s)
- Balázs Sonkodi
- Department of Health Sciences and Sport Medicine, University of Physical Education, Alkotas u. 44, H-1123 Budapest, Hungary
| |
Collapse
|
20
|
Bonetto G, Kamen Y, Evans KA, Káradóttir RT. Unraveling Myelin Plasticity. Front Cell Neurosci 2020; 14:156. [PMID: 32595455 PMCID: PMC7301701 DOI: 10.3389/fncel.2020.00156] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Accepted: 05/11/2020] [Indexed: 12/24/2022] Open
Abstract
Plasticity in the central nervous system (CNS) allows for responses to changing environmental signals. While the majority of studies on brain plasticity focus on neuronal synapses, myelin plasticity has now begun to emerge as a potential modulator of neuronal networks. Oligodendrocytes (OLs) produce myelin, which provides fast signal transmission, allows for synchronization of neuronal inputs, and helps to maintain neuronal function. Thus, myelination is also thought to be involved in learning. OLs differentiate from oligodendrocyte precursor cells (OPCs), which are distributed throughout the adult brain, and myelination continues into late adulthood. This process is orchestrated by numerous cellular and molecular signals, such as axonal diameter, growth factors, extracellular signaling molecules, and neuronal activity. However, the relative importance of, and cooperation between, these signaling pathways is currently unknown. In this review, we focus on the current knowledge about myelin plasticity in the CNS. We discuss new insights into the link between this type of plasticity, learning and behavior, as well as mechanistic aspects of myelin formation that may underlie myelin plasticity, highlighting OPC diversity in the CNS.
Collapse
Affiliation(s)
- Giulia Bonetto
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Yasmine Kamen
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Kimberley Anne Evans
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Ragnhildur Thóra Káradóttir
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom.,Department of Physiology, Biomedical Centre, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| |
Collapse
|
21
|
Suárez-Pozos E, Thomason EJ, Fuss B. Glutamate Transporters: Expression and Function in Oligodendrocytes. Neurochem Res 2020; 45:551-560. [PMID: 30628017 PMCID: PMC6616022 DOI: 10.1007/s11064-018-02708-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 12/19/2018] [Accepted: 12/22/2018] [Indexed: 12/14/2022]
Abstract
Glutamate, the main excitatory neurotransmitter of the vertebrate central nervous system (CNS), is well known as a regulator of neuronal plasticity and neurodevelopment. Such glutamate function is thought to be mediated primarily by signaling through glutamate receptors. Thus, it requires a tight regulation of extracellular glutamate levels and a fine-tuned homeostasis that, when dysregulated, has been associated with a wide range of central pathologies including neuropsychiatric, neurodevelopmental, and neurodegenerative disorders. In the mammalian CNS, extracellular glutamate levels are controlled by a family of sodium-dependent glutamate transporters belonging to the solute carrier family 1 (SLC1) that are also referred to as excitatory amino acid transporters (EAATs). The presumed main function of EAATs has been best described in the context of synaptic transmission where EAATs expressed by astrocytes and neurons effectively regulate extracellular glutamate levels so that synapses can function independently. There is, however, increasing evidence that EAATs are expressed by cells other than astrocytes and neurons, and that they exhibit functions beyond glutamate clearance. In this review, we will focus on the expression and functions of EAATs in the myelinating cells of the CNS, oligodendrocytes. More specifically, we will discuss potential roles of oligodendrocyte-expressed EAATs in contributing to extracellular glutamate homeostasis, and in regulating oligodendrocyte maturation and CNS myelination by exerting signaling functions that have traditionally been associated with glutamate receptors. In addition, we will provide some examples for how dysregulation of oligodendrocyte-expressed EAATs may be involved in the pathophysiology of neurologic diseases.
Collapse
Affiliation(s)
- Edna Suárez-Pozos
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Box 980709, Richmond, VA, 23298, USA
| | - Elizabeth J Thomason
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Box 980709, Richmond, VA, 23298, USA
| | - Babette Fuss
- Department of Anatomy and Neurobiology, Virginia Commonwealth University School of Medicine, Box 980709, Richmond, VA, 23298, USA.
| |
Collapse
|
22
|
Extrinsic Factors Driving Oligodendrocyte Lineage Cell Progression in CNS Development and Injury. Neurochem Res 2020; 45:630-642. [PMID: 31997102 PMCID: PMC7058689 DOI: 10.1007/s11064-020-02967-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 12/19/2019] [Accepted: 12/21/2019] [Indexed: 12/15/2022]
Abstract
Oligodendrocytes (OLs) generate myelin membranes for the rapid propagation of electrical signals along axons in the central nervous system (CNS) and provide metabolites to support axonal integrity and function. Differentiation of OLs from oligodendrocyte progenitor cells (OPCs) is orchestrated by a multitude of intrinsic and extrinsic factors in the CNS. Disruption of this process, or OL loss in the developing or adult brain, as observed in various neurological conditions including hypoxia/ischemia, stroke, and demyelination, results in axonal dystrophy, neuronal dysfunction, and severe neurological impairments. While much is known regarding the intrinsic regulatory signals required for OL lineage cell progression in development, studies from pathological conditions highlight the importance of the CNS environment and external signals in regulating OL genesis and maturation. Here, we review the recent findings in OL biology in the context of the CNS physiological and pathological conditions, focusing on extrinsic factors that facilitate OL development and regeneration.
Collapse
|
23
|
Abstract
In the peripheral nervous system, the vast majority of axons are accommodated within the fibre bundles that constitute the peripheral nerves. Axons within the nerves are in close contact with myelinating glia, the Schwann cells that are ideally placed to respond to, and possibly shape, axonal activity. The mechanisms of intercellular communication in the peripheral nerves may involve direct contact between the cells, as well as signalling via diffusible substances. Neurotransmitter glutamate has been proposed as a candidate extracellular molecule mediating the cross-talk between cells in the peripheral nerves. Two types of experimental findings support this idea: first, glutamate has been detected in the nerves and can be released upon electrical or chemical stimulation of the nerves; second, axons and Schwann cells in the peripheral nerves express glutamate receptors. Yet, the studies providing direct experimental evidence that intercellular glutamatergic signalling takes place in the peripheral nerves during physiological or pathological conditions are largely missing. Remarkably, in the central nervous system, axons and myelinating glia are involved in glutamatergic signalling. This signalling occurs via different mechanisms, the most intriguing of which is fast synaptic communication between axons and oligodendrocyte precursor cells. Glutamate receptors and/or synaptic axon-glia signalling are involved in regulation of proliferation, migration, and differentiation of oligodendrocyte precursor cells, survival of oligodendrocytes, and re-myelination of axons after damage. Does synaptic signalling exist between axons and Schwann cells in the peripheral nerves? What is the functional role of glutamate receptors in the peripheral nerves? Is activation of glutamate receptors in the nerves beneficial or harmful during diseases? In this review, we summarise the limited information regarding glutamate release and glutamate receptors in the peripheral nerves and speculate about possible mechanisms of glutamatergic signalling in the nerves. We highlight the necessity of further research on this topic because it should help to understand the mechanisms of peripheral nervous system development and nerve regeneration during diseases.
Collapse
Affiliation(s)
- Ting-Jiun Chen
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Centre, Washington, DC, USA
| | - Maria Kukley
- Group of Neuron Glia Interaction, University of Tübingen; Research Institute of Ophthalmology, Tübingen University Hospital, Tübingen, Germany
| |
Collapse
|
24
|
Zhan Y, Xia J, Wang X. Effects of glutamate-related drugs on anxiety and compulsive behavior in rats with obsessive-compulsive disorder. Int J Neurosci 2019; 130:551-560. [PMID: 31680595 DOI: 10.1080/00207454.2019.1684276] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Yuhua Zhan
- Department of Psychiatry, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jing Xia
- Department of Psychiatry, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xumei Wang
- Department of Psychiatry, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
25
|
Pukos N, Goodus MT, Sahinkaya FR, McTigue DM. Myelin status and oligodendrocyte lineage cells over time after spinal cord injury: What do we know and what still needs to be unwrapped? Glia 2019; 67:2178-2202. [PMID: 31444938 DOI: 10.1002/glia.23702] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 01/04/2023]
Abstract
Spinal cord injury (SCI) affects over 17,000 individuals in the United States per year, resulting in sudden motor, sensory and autonomic impairments below the level of injury. These deficits may be due at least in part to the loss of oligodendrocytes and demyelination of spared axons as it leads to slowed or blocked conduction through the lesion site. It has long been accepted that progenitor cells form new oligodendrocytes after SCI, resulting in the acute formation of new myelin on demyelinated axons. However, the chronicity of demyelination and the functional significance of remyelination remain contentious. Here we review work examining demyelination and remyelination after SCI as well as the current understanding of oligodendrocyte lineage cell responses to spinal trauma, including the surprisingly long-lasting response of NG2+ oligodendrocyte progenitor cells (OPCs) to proliferate and differentiate into new myelinating oligodendrocytes for months after SCI. OPCs are highly sensitive to microenvironmental changes, and therefore respond to the ever-changing post-SCI milieu, including influx of blood, monocytes and neutrophils; activation of microglia and macrophages; changes in cytokines, chemokines and growth factors such as ciliary neurotrophic factor and fibroblast growth factor-2; glutamate excitotoxicity; and axon degeneration and sprouting. We discuss how these changes relate to spontaneous oligodendrogenesis and remyelination, the evidence for and against demyelination being an important clinical problem and if remyelination contributes to motor recovery.
Collapse
Affiliation(s)
- Nicole Pukos
- Neuroscience Graduate Program, Ohio State University, Columbus, Ohio.,Belford Center for Spinal Cord Injury, Ohio State University, Columbus, Ohio
| | - Matthew T Goodus
- Belford Center for Spinal Cord Injury, Ohio State University, Columbus, Ohio.,Department of Neuroscience, Wexner Medical Center, Ohio State University, Columbus, Ohio
| | - Fatma R Sahinkaya
- Neuroscience Graduate Program, Ohio State University, Columbus, Ohio
| | - Dana M McTigue
- Belford Center for Spinal Cord Injury, Ohio State University, Columbus, Ohio.,Department of Neuroscience, Wexner Medical Center, Ohio State University, Columbus, Ohio
| |
Collapse
|
26
|
Habermacher C, Angulo MC, Benamer N. Glutamate versus GABA in neuron-oligodendroglia communication. Glia 2019; 67:2092-2106. [PMID: 30957306 DOI: 10.1002/glia.23618] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 02/28/2019] [Accepted: 03/19/2019] [Indexed: 12/20/2022]
Abstract
In the central nervous system (CNS), myelin sheaths around axons are formed by glial cells named oligodendrocytes (OLs). In turn, OLs are generated by oligodendrocyte precursor cells (OPCs) during postnatal development and in adults, according to a process that depends on the proliferation and differentiation of these progenitors. The maturation of OL lineage cells as well as myelination by OLs are complex and highly regulated processes in the CNS. OPCs and OLs express an array of receptors for neurotransmitters, in particular for the two main CNS neurotransmitters glutamate and GABA, and are therefore endowed with the capacity to respond to neuronal activity. Initial studies in cell cultures demonstrated that both glutamate and GABA signaling mechanisms play important roles in OL lineage cell development and function. However, much remains to be learned about the communication of glutamatergic and GABAergic neurons with oligodendroglia in vivo. This review focuses on recent major advances in our understanding of the neuron-oligodendroglia communication mediated by glutamate and GABA in the CNS, and highlights the present controversies in the field. We discuss the expression, activation modes and potential roles of synaptic and extrasynaptic receptors along OL lineage progression. We review the properties of OPC synaptic connectivity with presynaptic glutamatergic and GABAergic neurons in the brain and consider the implication of glutamate and GABA signaling in activity-driven adaptive myelination.
Collapse
Affiliation(s)
- Chloé Habermacher
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France.,Université Paris Descartes, Paris, France
| | - María C Angulo
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France.,Université Paris Descartes, Paris, France
| | - Najate Benamer
- Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Paris, France.,Université Paris Descartes, Paris, France
| |
Collapse
|
27
|
de Faria O, Gonsalvez DG, Nicholson M, Xiao J. Activity-dependent central nervous system myelination throughout life. J Neurochem 2019; 148:447-461. [PMID: 30225984 PMCID: PMC6587454 DOI: 10.1111/jnc.14592] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 09/07/2018] [Indexed: 01/03/2023]
Abstract
Myelin, the multilayered membrane surrounding many axons in the nervous system, increases the speed by which electrical signals travel along axons and facilitates neuronal communication between distant regions of the nervous system. However, how neuronal signals influence the myelinating process in the CNS is still largely unclear. Recent studies have significantly advanced this understanding, identifying important roles for neuronal activity in controlling oligodendrocyte development and their capacity of producing myelin in both developing and mature CNS. Here, we review these recent advances, and discuss potential mechanisms underpinning activity-dependent myelination and how remyelination may be stimulated via manipulating axonal activity, raising new questions for future research.
Collapse
Affiliation(s)
- Omar de Faria
- Wellcome Trust MRC Stem Cell Institute & Department of Veterinary MedicineUniversity of CambridgeCambridgeUK
| | - David G. Gonsalvez
- Department of Anatomy and NeuroscienceFaculty of MedicineDentistry and Health SciencesUniversity of MelbourneParkvilleVictoriaAustralia
| | - Madeline Nicholson
- Department of Anatomy and NeuroscienceFaculty of MedicineDentistry and Health SciencesUniversity of MelbourneParkvilleVictoriaAustralia
| | - Junhua Xiao
- Department of Anatomy and NeuroscienceFaculty of MedicineDentistry and Health SciencesUniversity of MelbourneParkvilleVictoriaAustralia
| |
Collapse
|
28
|
Sosunov A, Olabarria M, Goldman JE. Alexander disease: an astrocytopathy that produces a leukodystrophy. Brain Pathol 2019; 28:388-398. [PMID: 29740945 DOI: 10.1111/bpa.12601] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 03/02/2018] [Indexed: 02/02/2023] Open
Abstract
Alexander Disease (AxD) is a degenerative disorder caused by mutations in the GFAP gene, which encodes the major intermediate filament of astrocytes. As other cells in the CNS do not express GFAP, AxD is a primary astrocyte disease. Astrocytes acquire a large number of pathological features, including changes in morphology, the loss or diminution of a number of critical astrocyte functions and the activation of cell stress and inflammatory pathways. AxD is also characterized by white matter degeneration, a pathology that has led it to be included in the "leukodystrophies." Furthermore, variable degrees of neuronal loss take place. Thus, the astrocyte pathology triggers alterations in other cell types. Here, we will review the neuropathology of AxD and discuss how a disease of astrocytes can lead to severe pathologies in non-astrocytic cells. Our knowledge of the pathophysiology of AxD will also lead to a better understanding of how astrocytes interact with other CNS cells and how astrocytes in the gliosis that accompanies many neurological disorders can damage the function and survival of other cells.
Collapse
Affiliation(s)
| | - Markel Olabarria
- Departments of Pathology and Cell Biology, Columbia University, New York, NY
| | - James E Goldman
- Departments of Pathology and Cell Biology, Columbia University, New York, NY
| |
Collapse
|
29
|
Spitzer SO, Sitnikov S, Kamen Y, Evans KA, Kronenberg-Versteeg D, Dietmann S, de Faria O, Agathou S, Káradóttir RT. Oligodendrocyte Progenitor Cells Become Regionally Diverse and Heterogeneous with Age. Neuron 2019; 101:459-471.e5. [PMID: 30654924 PMCID: PMC6372724 DOI: 10.1016/j.neuron.2018.12.020] [Citation(s) in RCA: 237] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Revised: 10/12/2018] [Accepted: 12/12/2018] [Indexed: 01/12/2023]
Abstract
Oligodendrocyte progenitor cells (OPCs), which differentiate into myelinating oligodendrocytes during CNS development, are the main proliferative cells in the adult brain. OPCs are conventionally considered a homogeneous population, particularly with respect to their electrophysiological properties, but this has been debated. We show, by using single-cell electrophysiological recordings, that OPCs start out as a homogeneous population but become functionally heterogeneous, varying both within and between brain regions and with age. These electrophysiological changes in OPCs correlate with the differentiation potential of OPCs; thus, they may underlie the differentiational differences in OPCs between regions and, likewise, differentiation failure with age. Oligodendrocyte progenitor cells (OPCs) acquire ion channels with age OPCs become functionally heterogeneous both between brain regions and with age NMDARs disappear in non-myelinating regions but remain in actively myelinating areas Heterogeneity in OPC ion channel density indicates different functional states
Collapse
Affiliation(s)
- Sonia Olivia Spitzer
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Sergey Sitnikov
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Yasmine Kamen
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Kimberley Anne Evans
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Deborah Kronenberg-Versteeg
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Sabine Dietmann
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Omar de Faria
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Sylvia Agathou
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Ragnhildur Thóra Káradóttir
- Wellcome - Medical Research Council Cambridge Stem Cell Institute and Department of Veterinary Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
30
|
Agathou S, Káradóttir RT. Whole-Cell Patch Clamp Recordings from Oligodendrocyte Lineage Cells in Brain Slices. Methods Mol Biol 2019; 1936:141-168. [PMID: 30820898 DOI: 10.1007/978-1-4939-9072-6_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The whole-cell configuration of the patch-clamp technique is widely used to study electrically active cells and passive membrane properties, as well as the properties and pharmacology of ion channels, neurotransmitter receptors, and electrogenic transporters, in almost any cell type. In the brain, in addition to neurons, oligodendrocyte precursor cells (OPCs) that give rise to myelinating oligodendrocytes (OLs) are also excitable. Electrophysiological techniques provide the main tool for the thorough investigation of the electrogenic capacity of such cell types. Although there are many published protocols for whole-cell recordings, there are very few that touch upon the electrophysiological characteristics of oligodendrocyte lineage cells. Here we provide a detailed methodology for how to acquire and analyze whole-cell recordings from excitable cells, with a focus on oligodendrocyte lineage cells. We provide a protocol on how to successfully identify OPCs and OLs in brain slices, either with the use of transgenic animal models or through morphological and electrophysiological profiling. The method described can also be easily adopted for whole-cell recordings from oligodendrocyte lineage cells in vitro.
Collapse
Affiliation(s)
- Sylvia Agathou
- Department of Veterinary Medicine, Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Ragnhildur Thóra Káradóttir
- Department of Veterinary Medicine, Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
31
|
Chorghay Z, Káradóttir RT, Ruthazer ES. White Matter Plasticity Keeps the Brain in Tune: Axons Conduct While Glia Wrap. Front Cell Neurosci 2018; 12:428. [PMID: 30519159 PMCID: PMC6251003 DOI: 10.3389/fncel.2018.00428] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2018] [Accepted: 10/30/2018] [Indexed: 12/28/2022] Open
Abstract
Precise timing of neuronal inputs is crucial for brain circuit function and development, where it contributes critically to experience-dependent plasticity. Myelination therefore provides an important adaptation mechanism for vertebrate circuits. Despite its importance to circuit activity, the interplay between neuronal activity and myelination has yet to be fully elucidated. In recent years, significant attention has been devoted to uncovering and explaining the phenomenon of white matter (WM) plasticity. Here, we summarize some of the critical evidence for modulation of the WM by neuronal activity, ranging from human diffusion tensor imaging (DTI) studies to experiments in animal models. These experiments reveal activity-dependent changes in the differentiation and proliferation of the oligodendrocyte lineage, and in the critical properties of the myelin sheaths. We discuss the implications of such changes for synaptic function and plasticity, and present the underlying mechanisms of neuron–glia communication, with a focus on glutamatergic signaling and the axomyelinic synapse. Finally, we examine evidence that myelin plasticity may be subject to critical periods. Taken together, the present review aims to provide insights into myelination in the context of brain circuit formation and function, emphasizing the bidirectional interplay between neurons and myelinating glial cells to better inform future investigations of nervous system plasticity.
Collapse
Affiliation(s)
- Zahraa Chorghay
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Ragnhildur Thóra Káradóttir
- Department of Veterinary Medicine, Wellcome Trust - Medical Research Council Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Edward S Ruthazer
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
32
|
Ilyasov AA, Milligan CE, Pharr EP, Howlett AC. The Endocannabinoid System and Oligodendrocytes in Health and Disease. Front Neurosci 2018; 12:733. [PMID: 30416422 PMCID: PMC6214135 DOI: 10.3389/fnins.2018.00733] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 09/24/2018] [Indexed: 12/22/2022] Open
Abstract
Cannabinoid-based interventions are being explored for central nervous system (CNS) pathologies such as neurodegeneration, demyelination, epilepsy, stroke, and trauma. As these disease states involve dysregulation of myelin integrity and/or remyelination, it is important to consider effects of the endocannabinoid system on oligodendrocytes and their precursors. In this review, we examine research reports on the effects of the endocannabinoid system (ECS) components on oligodendrocytes and their precursors, with a focus on therapeutic implications. Cannabinoid ligands and modulators of the endocannabinoid system promote cell signaling in oligodendrocyte precursor survival, proliferation, migration and differentiation, and mature oligodendrocyte survival and myelination. Agonist stimulation of oligodendrocyte precursor cells (OPCs) at both CB1 and CB2 receptors counter apoptotic processes via Akt/PI3K, and promote proliferation via Akt/mTOR and ERK pathways. CB1 receptors in radial glia promote proliferation and conversion to progenitors fated to become oligodendroglia, whereas CB2 receptors promote OPC migration in neonatal development. OPCs produce 2-arachidonoylglycerol (2-AG), stimulating cannabinoid receptor-mediated ERK pathways responsible for differentiation to arborized, myelin basic protein (MBP)-producing oligodendrocytes. In cell culture models of excitotoxicity, increased reactive oxygen species, and depolarization-dependent calcium influx, CB1 agonists improved viability of oligodendrocytes. In transient and permanent middle cerebral artery occlusion models of anoxic stroke, WIN55212-2 increased OPC proliferation and maturation to oligodendroglia, thereby reducing cerebral tissue damage. In several models of rodent encephalomyelitis, chronic treatment with cannabinoid agonists ameliorated the damage by promoting OPC survival and oligodendrocyte function. Pharmacotherapeutic strategies based upon ECS and oligodendrocyte production and survival should be considered.
Collapse
Affiliation(s)
- Alexander A Ilyasov
- Graduate Program in Neuroscience, Wake Forest School of Medicine, Winston Salem, NC, United States.,Department of Physiology and Pharmacology and Center for Research on Substance Use and Addiction, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Carolanne E Milligan
- Graduate Program in Neuroscience, Wake Forest School of Medicine, Winston Salem, NC, United States.,Department of Neurobiology and Anatomy, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Emily P Pharr
- Graduate Program in Neuroscience, Wake Forest School of Medicine, Winston Salem, NC, United States.,Department of Neurology and Comprehensive Multiple Sclerosis Center, Wake Forest School of Medicine, Winston-Salem, NC, United States
| | - Allyn C Howlett
- Graduate Program in Neuroscience, Wake Forest School of Medicine, Winston Salem, NC, United States.,Department of Physiology and Pharmacology and Center for Research on Substance Use and Addiction, Wake Forest School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
33
|
Almeida RG. The Rules of Attraction in Central Nervous System Myelination. Front Cell Neurosci 2018; 12:367. [PMID: 30374292 PMCID: PMC6196289 DOI: 10.3389/fncel.2018.00367] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 09/27/2018] [Indexed: 12/12/2022] Open
Abstract
The wrapping of myelin around axons is crucial for the development and function of the central nervous system (CNS) of vertebrates, greatly regulating the conduction of action potentials. Oligodendrocytes, the myelinating glia of the CNS, have an intrinsic tendency to wrap myelin around any permissive structure in vitro, but in vivo, myelin is targeted with remarkable specificity only to certain axons. Despite the importance of myelination, the mechanisms by which oligodendrocytes navigate a complex milieu that includes many types of cells and their cellular projections and select only certain axons for myelination remains incompletely understood. In this Mini-review, I highlight recent studies that shed light on the molecular and cellular rules governing CNS myelin targeting.
Collapse
Affiliation(s)
- Rafael Góis Almeida
- Centre for Discovery Brain Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
34
|
Bastian C, Quinn J, Tripathi A, Aquila D, McCray A, Dutta R, Baltan S, Brunet S. CK2 inhibition confers functional protection to young and aging axons against ischemia by differentially regulating the CDK5 and AKT signaling pathways. Neurobiol Dis 2018; 126:47-61. [PMID: 29944965 DOI: 10.1016/j.nbd.2018.05.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 05/08/2018] [Accepted: 05/21/2018] [Indexed: 12/25/2022] Open
Abstract
White matter (WM) is injured in most strokes, which contributes to functional deficits during recovery. Casein kinase 2 (CK2) is a protein kinase that is expressed in brain, including WM. To assess the impact of CK2 inhibition on axon recovery following oxygen glucose deprivation (OGD), mouse optic nerves (MONs), which are pure WM tracts, were subjected to OGD with or without the selective CK2 inhibitor CX-4945. CX-4945 application preserved axon function during OGD and promoted axon function recovery when applied before or after OGD. This protective effect of CK2 inhibition correlated with preservation of oligodendrocytes and conservation of axon structure and axonal mitochondria. To investigate the pertinent downstream signaling pathways, siRNA targeting the CK2α subunit identified CDK5 and AKT as downstream molecules. Consequently, MK-2206 and roscovitine, which are selective AKT and CDK5 inhibitors, respectively, protected young and aging WM function only when applied before OGD. However, a novel pan-AKT allosteric inhibitor, ARQ-092, which targets both the inactive and active conformations of AKT, conferred protection to young and aging axons when applied before or after OGD. These results suggest that AKT and CDK5 signaling contribute to the WM functional protection conferred by CK2 inhibition during ischemia, while inhibition of activated AKT signaling plays the primary role in post-ischemic protection conferred by CK2 inhibition in WM independent of age. CK2 inhibitors are currently being used in clinical trials for cancer patients; therefore, our results will provide rationale for repurposing these drugs as therapeutic options for stroke patients by adding novel targets.
Collapse
Affiliation(s)
- Chinthasagar Bastian
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - John Quinn
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Ajai Tripathi
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Danielle Aquila
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Andrew McCray
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Ranjan Dutta
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Selva Baltan
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America.
| | - Sylvain Brunet
- Departments of Neurosciences, Cleveland Clinic, Cleveland, OH 44195, United States of America.
| |
Collapse
|
35
|
Li T, Wang L, Ma T, Wang S, Niu J, Li H, Xiao L. Dynamic Calcium Release From Endoplasmic Reticulum Mediated by Ryanodine Receptor 3 Is Crucial for Oligodendroglial Differentiation. Front Mol Neurosci 2018; 11:162. [PMID: 29867353 PMCID: PMC5968115 DOI: 10.3389/fnmol.2018.00162] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 04/30/2018] [Indexed: 12/15/2022] Open
Abstract
Increased intracellular Ca2+ in oligodendrocyte progenitor cells (OPCs) is important to initiate their differentiation, but the intracellular Ca2+ channel involved in this process remains unclear. As a Ca2+-induced Ca2+ release (CICR) channel that mediates endoplasmic reticulum (ER) Ca2+ release, the role of ryanodine receptors (RyRs) in oligodendroglial development is unexplored. In the present study, we observed that among the three mammalian isoforms, oligodendroglial lineage cells selectively expressed RyR3. Strong RyR3-positive signal was distributed all over the cytoplasm and processes in OPCs and/or immature OLs (imOLs), whereas it gradually decreased and was located mainly around the perinuclear region in mature oligodendrocytes (OLs). In addition, RyR3-mediated intracellular Ca2+ waves following caffeine stimulation were correlated with the expression pattern of RyR3, in which high flat Ca2+ fluctuations and oscillatory Ca2+ waves were more frequently recorded in OPCs and/or imOLs than in OLs. Through further functional exploration, we demonstrated that pretreatment with the RyR antagonist ryanodine could neutralize the increase in intracellular Ca2+ induced by OPC differentiation and reduce the number of mature OLs. Moreover, gene-level knockdown of RyR3 by lentivirus in OPCs resulted in inhibition of OPC differentiation. Taken together, our results provide new insight into the crucial role of RyR3-mediated ER Ca2+ release in the regulation of OPC differentiation and/or myelination.
Collapse
Affiliation(s)
- Tao Li
- Chongqing Key Laboratory of Neurobiology, Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| | - Lingyun Wang
- Chongqing Key Laboratory of Neurobiology, Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| | - Teng Ma
- Chongqing Key Laboratory of Neurobiology, Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| | - Shouyu Wang
- Chongqing Key Laboratory of Neurobiology, Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| | - Jianqin Niu
- Chongqing Key Laboratory of Neurobiology, Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| | - Hongli Li
- Chongqing Key Laboratory of Neurobiology, Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| | - Lan Xiao
- Chongqing Key Laboratory of Neurobiology, Department of Histology and Embryology, Third Military Medical University, Chongqing, China
| |
Collapse
|
36
|
Káradóttir RT, Kuo CT. Neuronal Activity-Dependent Control of Postnatal Neurogenesis and Gliogenesis. Annu Rev Neurosci 2018; 41:139-161. [PMID: 29618286 DOI: 10.1146/annurev-neuro-072116-031054] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The addition of new neurons and oligodendroglia in the postnatal and adult mammalian brain presents distinct forms of gray and white matter plasticity. Substantial effort has been devoted to understanding the cellular and molecular mechanisms controlling postnatal neurogenesis and gliogenesis, revealing important parallels to principles governing the embryonic stages. While during central nervous system development, scripted temporal and spatial patterns of neural and glial progenitor proliferation and differentiation are necessary to create the nervous system architecture, it remains unclear what driving forces maintain and sustain postnatal neural stem cell (NSC) and oligodendrocyte progenitor cell (OPC) production of new neurons and glia. In recent years, neuronal activity has been identified as an important modulator of these processes. Using the distinct properties of neurotransmitter ionotropic and metabotropic channels to signal downstream cellular events, NSCs and OPCs share common features in their readout of neuronal activity patterns. Here we review the current evidence for neuronal activity-dependent control of NSC/OPC proliferation and differentiation in the postnatal brain, highlight some potential mechanisms used by the two progenitor populations, and discuss future studies that might advance these research areas further.
Collapse
Affiliation(s)
- Ragnhildur T Káradóttir
- Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, United Kingdom; .,Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, United Kingdom
| | - Chay T Kuo
- Departments of Cell Biology and Neurobiology, Duke University School of Medicine, Durham, North Carolina 27710, USA; .,Preston Robert Tisch Brain Tumor Center, Duke University Medical Center, Durham, North Carolina 27710, USA.,Institute for Brain Sciences, Duke University, Durham, North Carolina 27708, USA
| |
Collapse
|
37
|
Doyle S, Hansen DB, Vella J, Bond P, Harper G, Zammit C, Valentino M, Fern R. Vesicular glutamate release from central axons contributes to myelin damage. Nat Commun 2018. [PMID: 29531223 PMCID: PMC5847599 DOI: 10.1038/s41467-018-03427-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The axon myelin sheath is prone to injury associated with N-methyl-d-aspartate (NMDA)-type glutamate receptor activation but the source of glutamate in this context is unknown. Myelin damage results in permanent action potential loss and severe functional deficit in the white matter of the CNS, for example in ischemic stroke. Here, we show that in rats and mice, ischemic conditions trigger activation of myelinic NMDA receptors incorporating GluN2C/D subunits following release of axonal vesicular glutamate into the peri-axonal space under the myelin sheath. Glial sources of glutamate such as reverse transport did not contribute significantly to this phenomenon. We demonstrate selective myelin uptake and retention of a GluN2C/D NMDA receptor negative allosteric modulator that shields myelin from ischemic injury. The findings potentially support a rational approach toward a low-impact prophylactic therapy to protect patients at risk of stroke and other forms of excitotoxic injury. Neuronal activity can lead to vesicular release of glutamate. Here the authors demonstrate that vesicular release of glutamate occurs in axons during ischemic conditions, and that an allosteric modulator of GluN2C/D is protective in models of ischemic injury.
Collapse
Affiliation(s)
- Sean Doyle
- University of Plymouth, Plymouth, PL6 8BY, UK
| | | | | | - Peter Bond
- University of Plymouth, Plymouth, PL6 8BY, UK
| | | | | | | | - Robert Fern
- University of Plymouth, Plymouth, PL6 8BY, UK.
| |
Collapse
|
38
|
Valny M, Honsa P, Waloschkova E, Matuskova H, Kriska J, Kirdajova D, Androvic P, Valihrach L, Kubista M, Anderova M. A single-cell analysis reveals multiple roles of oligodendroglial lineage cells during post-ischemic regeneration. Glia 2018; 66:1068-1081. [DOI: 10.1002/glia.23301] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Revised: 01/07/2018] [Accepted: 01/16/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Martin Valny
- Department of Cellular Neurophysiology; Institute of Experimental Medicine, Academy of Sciences of the Czech Republic; Prague Czech Republic
- 2nd Faculty of Medicine; Charles University; Prague Czech Republic
| | - Pavel Honsa
- Department of Cellular Neurophysiology; Institute of Experimental Medicine, Academy of Sciences of the Czech Republic; Prague Czech Republic
| | - Eliska Waloschkova
- Department of Cellular Neurophysiology; Institute of Experimental Medicine, Academy of Sciences of the Czech Republic; Prague Czech Republic
| | - Hana Matuskova
- Department of Cellular Neurophysiology; Institute of Experimental Medicine, Academy of Sciences of the Czech Republic; Prague Czech Republic
| | - Jan Kriska
- Department of Cellular Neurophysiology; Institute of Experimental Medicine, Academy of Sciences of the Czech Republic; Prague Czech Republic
- 2nd Faculty of Medicine; Charles University; Prague Czech Republic
| | - Denisa Kirdajova
- Department of Cellular Neurophysiology; Institute of Experimental Medicine, Academy of Sciences of the Czech Republic; Prague Czech Republic
- 2nd Faculty of Medicine; Charles University; Prague Czech Republic
| | - Peter Androvic
- Laboratory of Gene Expression; Institute of Biotechnology, Academy of Sciences of the Czech Republic; Prague Czech Republic
| | - Lukas Valihrach
- Laboratory of Gene Expression; Institute of Biotechnology, Academy of Sciences of the Czech Republic; Prague Czech Republic
| | - Mikael Kubista
- Laboratory of Gene Expression; Institute of Biotechnology, Academy of Sciences of the Czech Republic; Prague Czech Republic
| | - Miroslava Anderova
- Department of Cellular Neurophysiology; Institute of Experimental Medicine, Academy of Sciences of the Czech Republic; Prague Czech Republic
- 2nd Faculty of Medicine; Charles University; Prague Czech Republic
| |
Collapse
|
39
|
Thurm A, Powell EM, Neul JL, Wagner A, Zwaigenbaum L. Loss of skills and onset patterns in neurodevelopmental disorders: Understanding the neurobiological mechanisms. Autism Res 2018; 11:212-222. [PMID: 29226600 PMCID: PMC5825269 DOI: 10.1002/aur.1903] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/19/2017] [Accepted: 11/25/2017] [Indexed: 12/15/2022]
Abstract
Patterns of onset in Autism Spectrum Disorder, including a pattern that includes loss of previously acquired skills, have been identified since the first reports of the disorder. However, attempts to study such "regression" have been limited to clinical studies, that until recently mostly involved retrospective reports. The current report reflects discussion that occurred at an NIMH convened meeting in 2016 with the purpose of bridging clinical autism research with basic and translational work in this area. This summary describes the state of the field with respect to clinical studies, describing gaps in knowledge based on limited methods and prospective data collected. Biological mechanisms that have been shown to account for regression early in development in specific conditions are discussed, as well as potential mechanisms that have not yet been explored. Suggestions include use of model systems during the developmental period and cutting-edge methods, including non-invasive imaging that may afford opportunities for a better understanding of the neurobiological pathways that result in loss of previously-attained skills. Autism Res 2018, 11: 212-222. © 2017 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY Loss of previously acquired skills, or regression, has been reported in Autism Spectrum Disorder since Kanner's reports in the 1950's. The current report reflects discussion from an NIMH convened meeting in 2016 with the purpose of bridging clinical autism research with basic and translational work in this area. This summary describes the state of the field regarding clinical studies and suggests use of model systems during the developmental period and cutting-edge methods, for a better understanding of the neurobiological pathways that result in loss of previously-attained skills.
Collapse
Affiliation(s)
- Audrey Thurm
- Office of the Clinical Director, National Institute of Mental Health, National Institute of Health, Bethesda, Maryland, USA
| | - Elizabeth M. Powell
- Division of Neuroscience and Behavior, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Jeffrey L. Neul
- Vanderbilt Kennedy Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ann Wagner
- Division of Translational Research, National Institute of Mental Health, National Institute of Health, Bethesda, Maryland, USA
| | - Lonnie Zwaigenbaum
- Autism Research Center, Glenrose Rehabilitation Hospital, Edmonton, Alberta, Canada
| |
Collapse
|
40
|
Jackson J, Bianco G, Rosa AO, Cowan K, Bond P, Anichtchik O, Fern R. White matter tauopathy: Transient functional loss and novel myelin remodeling. Glia 2018; 66:813-827. [PMID: 29315804 DOI: 10.1002/glia.23286] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/08/2017] [Accepted: 12/11/2017] [Indexed: 01/03/2023]
Abstract
Early white matter (WM) changes are common in dementia and may contribute to functional decline. We here examine this phenomenon in an induced dementia model for the first time. We report a novel and selective form of myelin injury as the first manifestation of tauopathy in the adult central nervous system. Myelin pathology rapidly followed the induction of a P301 tau mutation associated with fronto-temporal dementia in humans (rTG4510 line). Damage involved focal disruption of the ad-axonal myelin lamella and internal oligodendrocyte tongue process, followed by myelin remodeling with features of re-myelination that included myelin thinning and internodal shortening. The evolution of the re-myelinated phenotype was complete in the molecular layer of the dentate gyrus after 1 month and in the optic nerve (ON) after 9 months of transgene induction and proceeded in the absence of actual demyelination, reactive glial changes or inflammatory response. The initial rapid myelin pathology was associated with loss of WM function and performance decline in a novel recognition test and both these effects largely reversed during the myelin re-modeling phase. The initial phase of myelin injury was accompanied by disruption of the vesicle population present in the axoplasm of hippocampal and ON axons. Axoplasmic vesicle release is significant for the regulation of myelin plasticity and disruption of this pathway may underlie the myelin damage and remodeling evoked by tauopathy. WM dysfunction early in tauopathy will disorder neural circuits, the current findings suggest this event may make a significant contribution to early clinical deficit in dementia.
Collapse
Affiliation(s)
| | | | - Angelo O Rosa
- Plymouth Electron Microscopy Centre, University of Plymouth, Plymouth, United Kingdom
| | - Katrina Cowan
- Peninsula School of Medicine and Dentistry, University of Plymouth, PUPSMD, Plymouth, United Kingdom
| | - Peter Bond
- Plymouth Electron Microscopy Centre, University of Plymouth, Plymouth, United Kingdom
| | - Oleg Anichtchik
- Peninsula School of Medicine and Dentistry, University of Plymouth, PUPSMD, Plymouth, United Kingdom
| | - Robert Fern
- Peninsula School of Medicine and Dentistry, University of Plymouth, PUPSMD, Plymouth, United Kingdom
| |
Collapse
|
41
|
Novgorodov SA, Voltin JR, Gooz MA, Li L, Lemasters JJ, Gudz TI. Acid sphingomyelinase promotes mitochondrial dysfunction due to glutamate-induced regulated necrosis. J Lipid Res 2017; 59:312-329. [PMID: 29282302 DOI: 10.1194/jlr.m080374] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 12/05/2017] [Indexed: 12/11/2022] Open
Abstract
Inhibiting the glutamate/cystine antiporter system xc-, a key antioxidant defense machinery in the CNS, could trigger a novel form of regulated necrotic cell death, ferroptosis. The underlying mechanisms of system xc--dependent cell demise were elucidated using primary oligodendrocytes (OLs) treated with glutamate to block system xc- function. Pharmacological analysis revealed ferroptosis as a major contributing factor to glutamate-initiated OL death. A sphingolipid profile showed elevations of ceramide species and sphingosine that were preventable by inhibiting of an acid sphingomyelinase (ASM) activity. OL survival was enhanced by both downregulating ASM expression and blocking ASM activity. Glutamate-induced ASM activation seems to involve posttranscriptional mechanisms and was associated with a decreased GSH level. Further investigation of the mechanisms of OL response to glutamate revealed enhanced reactive oxygen species production, augmented lipid peroxidation, and opening of the mitochondrial permeability transition pore that were attenuated by hindering ASM. Of note, knocking down sirtuin 3, a deacetylase governing the mitochondrial antioxidant system, reduced OL survival. The data highlight the importance of the mitochondrial compartment in regulated necrotic cell death and accentuate the novel role of ASM in disturbing mitochondrial functions during OL response to glutamate toxicity, which is essential for pathobiology in stroke and traumatic brain injury.
Collapse
Affiliation(s)
- Sergei A Novgorodov
- Departments of Neuroscience Medical University of South Carolina, Charleston, SC 29425
| | - Joshua R Voltin
- Departments of Neuroscience Medical University of South Carolina, Charleston, SC 29425
| | - Monika A Gooz
- Departments of Drug Discovery, Medical University of South Carolina, Charleston, SC 29425
| | - Li Li
- Departments of Drug Discovery, Medical University of South Carolina, Charleston, SC 29425
| | - John J Lemasters
- Departments of Drug Discovery, Medical University of South Carolina, Charleston, SC 29425
| | - Tatyana I Gudz
- Departments of Neuroscience Medical University of South Carolina, Charleston, SC 29425 .,Ralph H. Johnson Veterans Affairs Medical Center, Charleston, SC 29401
| |
Collapse
|
42
|
Saab AS, Nave KA. Myelin dynamics: protecting and shaping neuronal functions. Curr Opin Neurobiol 2017; 47:104-112. [PMID: 29065345 DOI: 10.1016/j.conb.2017.09.013] [Citation(s) in RCA: 148] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 09/11/2017] [Accepted: 09/21/2017] [Indexed: 12/28/2022]
Abstract
Myelinating glial cells are well-known to insulate axons and to speed up action potential propagation. Through adjustments in the axonal coverage with myelin, myelin sheath thickness and possibly nodal/internode length oligodendrocytes are involved in fine-tuning the brain's computational power throughout life. Be it motor skill learning or social behaviors in higher vertebrates, proper myelination is critical in shaping brain functions. Neurons rely on their myelinating partners not only for setting conduction speed, but also for regulating the ionic environment and fueling their energy demands with metabolites. Also, long-term axonal integrity and neuronal survival are maintained by oligodendrocytes and loss of this well-coordinated axon-glial interplay contributes to neuropsychiatric diseases. Better insight into how myelination and oligodendrocyte functions are constantly fine-tuned in the adult CNS, which includes sensing of neuronal activity and adjusting glial metabolic support, will be critical for understanding higher brain functions and cognitive decline associated with myelin abnormalities in the aging brain.
Collapse
Affiliation(s)
- Aiman S Saab
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland; Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland.
| | - Klaus-Armin Nave
- Max Planck Institute of Experimental Medicine, Department of Neurogenetics, Göttingen, Germany.
| |
Collapse
|
43
|
Glutamate Activates AMPA Receptor Conductance in the Developing Schwann Cells of the Mammalian Peripheral Nerves. J Neurosci 2017; 37:11818-11834. [PMID: 29089441 DOI: 10.1523/jneurosci.1168-17.2017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 09/01/2017] [Accepted: 10/02/2017] [Indexed: 11/21/2022] Open
Abstract
Schwann cells (SCs) are myelinating cells of the PNS. Although SCs are known to express different channels and receptors on their surface, little is known about the activation and function of these proteins. Ionotropic glutamate receptors are thought to play an essential role during development of SC lineage and during peripheral nerve injury, so we sought to study their functional properties. We established a novel preparation of living peripheral nerve slices with preserved cellular architecture and used a patch-clamp technique to study AMPA-receptor (AMPAR)-mediated currents in SCs for the first time. We found that the majority of SCs in the nerves dissected from embryonic and neonatal mice of both sexes respond to the application of glutamate with inward current mediated by Ca2+-permeable AMPARs. Using stationary fluctuation analysis (SFA), we demonstrate that single-channel conductance of AMPARs in SCs is 8-11 pS, which is comparable to that in neurons. We further show that, when SCs become myelinating, they downregulate functional AMPARs. This study is the first to demonstrate AMPAR-mediated conductance in SCs of vertebrates, to investigate elementary properties of AMPARs in these cells, and to provide detailed electrophysiological and morphological characterization of SCs at different stages of development.SIGNIFICANCE STATEMENT We provide several important conceptual and technical advances in research on the PNS. We pioneer the first description of AMPA receptor (AMPAR)-mediated currents in the PNS glia of vertebrates and provide new insights into the properties of AMPAR channels in peripheral glia; for example, their Ca2+ permeability and single-channel conductance. We describe for the first time the electrophysiological and morphological properties of Schwann cells (SCs) at different stages of development and show that functional AMPARs are expressed only in developing, not mature, SCs. Finally, we introduce a preparation of peripheral nerve slices for patch-clamp recordings. This preparation opens new possibilities for studying the physiology of SCs in animal models and in surgical human samples.
Collapse
|
44
|
de Faria O, Pama EAC, Evans K, Luzhynskaya A, Káradóttir RT. Neuroglial interactions underpinning myelin plasticity. Dev Neurobiol 2017; 78:93-107. [PMID: 28941015 DOI: 10.1002/dneu.22539] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 08/11/2017] [Accepted: 09/14/2017] [Indexed: 11/07/2022]
Abstract
The CNS is extremely responsive to an ever-changing environment. Studies of neural circuit plasticity focus almost exclusively on functional and structural changes of neuronal synapses. In recent years, however, myelin plasticity has emerged as a potential modulator of neuronal networks. Myelination of previously unmyelinated axons and changes in the structure of myelin on already-myelinated axons (similar to changes in internode number and length or myelin thickness or geometry of the nodal area) can in theory have significant effects on the function of neuronal networks. In this article, the authors review the current evidence for myelin changes occurring in the adult CNS, highlight some potential underlying mechanisms of how neuronal activity may regulate myelin changes, and explore the similarities between neuronal and myelin plasticity. © 2017 Wiley Periodicals, Inc. Develop Neurobiol 78: 93-107, 2018.
Collapse
Affiliation(s)
- Omar de Faria
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Ewa Anastazia Claudia Pama
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Kimberley Evans
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Aryna Luzhynskaya
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Ragnhildur Thóra Káradóttir
- Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute & Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
45
|
H 2O 2 attenuates IGF-1R tyrosine phosphorylation and its survival signaling properties in neuronal cells via NR2B containing NMDA receptor. Oncotarget 2017; 8:65313-65328. [PMID: 29029433 PMCID: PMC5630333 DOI: 10.18632/oncotarget.18625] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Accepted: 05/01/2017] [Indexed: 11/25/2022] Open
Abstract
Impairment of insulin-like growth factor I (IGF-I) signaling plays an important role in the development of neurodegeneration. In the present study, we investigated the effect of H2O2 on the survival signaling of IGF-1 and its underlying mechanisms in human neuronal cells SH-SY5Y. Our results showed that IGF-1 promoted cell survival and stimulated phosphorylation of IGF-1R as well as its downstream targets like AKT and ERK1/2 in these cells. Meanwhile, these effects of IGF-1 were abolished by H2O2 at 200μM concentration which did not cause any significant toxicity to cells itself in our experiments. Moreover, studies using various glutamate receptor subtype antagonists displayed that N-methyl-D -aspartate (NMDA) receptor antagonist dizocilpine maleate (MK-801) blocked the effects of H2O2, whereas other glutamate receptor subtype antagonists, such as non-NMDA receptor antagonist 6,7-dinitroquinoxaline-2,3-dione (DNQX), metabolic glutamate receptor antagonists LY341495 and CPCCOEt, had no effect. Further studies revealed that NR2B-containing NMDARs are responsible for these effects as its effects were blocked by pharmacological inhibitor Ro25-698 or specific siRNA for NR2B, but not NR2A. Finally, our data also showed that Ca2+ influx contributes to the effects of H2O2. Similar results were obtained in primary cultured cortical neurons. Taken together, the results from the present study suggested that H2O2 attenuated IGF-1R tyrosine phosphorylation and its survival signaling properties via NR2B containing NMDA receptors and Ca2+ influx in SH-SY5Y cells. Therefore, NMDAR antagonists, especially NR2B-selective ones, combined with IGF-1 may serve as an alternative therapeutic agent for oxidative stress related neurodegenerative disease.
Collapse
|
46
|
Fern R. The Leukocentric Theory of Neurological Disorder: A Manifesto. Neurochem Res 2017; 42:2666-2672. [DOI: 10.1007/s11064-017-2279-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/12/2017] [Accepted: 04/21/2017] [Indexed: 01/26/2023]
|
47
|
Zhang Z, Zhang Y, Liu C, Zhao M, Yang Y, Wu H, Zhang H, Lin H, Zheng L, Jiang H. Serum Metabolomic Profiling Identifies Characterization of Non-Obstructive Azoospermic Men. Int J Mol Sci 2017; 18:ijms18020238. [PMID: 28125052 PMCID: PMC5343775 DOI: 10.3390/ijms18020238] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Revised: 01/10/2017] [Accepted: 01/12/2017] [Indexed: 12/22/2022] Open
Abstract
Male infertility is considered a common health problem, and non-obstructive azoospermia with unclear pathogenesis is one of the most challenging tasks for clinicians. The objective of this study was to investigate the differential serum metabolic pattern in non-obstructive azoospermic men and to determine potential biomarkers related to spermatogenic dysfunction. Serum samples from patients with non-obstructive azoospermia (n = 22) and healthy controls (n = 31) were examined using high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). Serum metabolomic profiling could differentiate non-obstructive azoospermic patients from healthy control subjects. A total of 24 metabolites were screened and identified as potential markers, many of which are involved in energy production, oxidative stress and cell apoptosis in spermatogenesis. Moreover, the results showed that various metabolic pathways, including d-glutamine and d-glutamate metabolism, taurine and hypotaurine metabolism, pyruvate metabolism, the citrate cycle and alanine, aspartate and glutamate metabolism, were disrupted in patients with non-obstructive azoospermia. Our results indicated that the serum metabolic disorders may contribute to the etiology of non-obstructive azoospermia. This study suggested that serum metabolomics could identify unique metabolic patterns of non-obstructive azoospermia and provide novel insights into the pathogenesis underlying male infertility.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Urology, Peking University Third Hospital, Beijing 100191, China.
| | - Yingwei Zhang
- Department of Nephrology, General Hospital of Jinan Military, Jinan 250000, China.
| | - Changjie Liu
- The Institute of Cardiovascular Sciences, the Institute of Systems Biomedicine, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University Health Science Center, Beijing 100191, China.
| | - Mingming Zhao
- The Institute of Cardiovascular Sciences, the Institute of Systems Biomedicine, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University Health Science Center, Beijing 100191, China.
| | - Yuzhuo Yang
- Department of Urology, Peking University Third Hospital, Beijing 100191, China.
| | - Han Wu
- Department of Urology, Peking University Third Hospital, Beijing 100191, China.
| | - Hongliang Zhang
- Department of Human Sperm Bank, Peking University Third Hospital, Beijing 100191, China.
| | - Haocheng Lin
- Department of Urology, Peking University Third Hospital, Beijing 100191, China.
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences, the Institute of Systems Biomedicine, School of Basic Medical Sciences, and Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Peking University Health Science Center, Beijing 100191, China.
| | - Hui Jiang
- Department of Urology, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
48
|
Mogha A, D'Rozario M, Monk KR. G Protein-Coupled Receptors in Myelinating Glia. Trends Pharmacol Sci 2016; 37:977-987. [PMID: 27670389 DOI: 10.1016/j.tips.2016.09.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Revised: 09/02/2016] [Accepted: 09/06/2016] [Indexed: 12/21/2022]
Abstract
The G protein-coupled receptor (GPCR) superfamily represents the largest class of functionally selective drug targets for disease modulation and therapy. GPCRs have been studied in great detail in central nervous system (CNS) neurons, but these important molecules have been relatively understudied in glia. In recent years, however, exciting new roles for GPCRs in glial cell biology have emerged. We focus here on the key roles of GPCRs in a specialized subset of glia, myelinating glia. We highlight recent work firmly establishing GPCRs as regulators of myelinating glial cell development and myelin repair. These advances expand our understanding of myelinating glial cell biology and underscore the utility of targeting GPCRs to promote myelin repair in human disease.
Collapse
Affiliation(s)
- Amit Mogha
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mitchell D'Rozario
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Kelly R Monk
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
49
|
Huang JK, Káradóttir RT. Oligodendrocytes in health and disease. Neuropharmacology 2016; 110:537-538. [PMID: 27553122 DOI: 10.1016/j.neuropharm.2016.06.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/20/2016] [Indexed: 11/27/2022]
Affiliation(s)
- Jeffrey K Huang
- Dept. of Biology, Georgetown University, 37th and O St., NW, Washington, DC 20057, USA.
| | - Ragnhildur T Káradóttir
- Wellcome Trust - MRC Stem Cell Institute, University of Cambridge, Tennis Court Road, Cambridge CB2 1QR, UK.
| |
Collapse
|