1
|
Zhang Y, Li D, Cai Y, Zou R, Zhang Y, Deng X, Wang Y, Tang T, Ma Y, Wu F, Xie Y. Astrocyte allocation during brain development is controlled by Tcf4-mediated fate restriction. EMBO J 2024; 43:5114-5140. [PMID: 39300210 DOI: 10.1038/s44318-024-00218-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 08/07/2024] [Accepted: 08/09/2024] [Indexed: 09/22/2024] Open
Abstract
Astrocytes in the brain exhibit regional heterogeneity contributing to regional circuits involved in higher-order brain functions, yet the mechanisms controlling their distribution remain unclear. Here, we show that the precise allocation of astrocytes to specific brain regions during development is achieved through transcription factor 4 (Tcf4)-mediated fate restriction based on their embryonic origin. Loss of Tcf4 in ventral telencephalic neural progenitor cells alters the fate of oligodendrocyte precursor cells to transient intermediate astrocyte precursor cells, resulting in mislocalized astrocytes in the dorsal neocortex. These ectopic astrocytes engage with neocortical neurons and acquire features reminiscent of dorsal neocortical astrocytes. Furthermore, Tcf4 functions as a suppressor of astrocyte fate during the differentiation of oligodendrocyte precursor cells derived from the ventral telencephalon, thereby restricting the fate to the oligodendrocyte lineage in the dorsal neocortex. Together, our findings highlight a previously unappreciated role for Tcf4 in regulating astrocyte allocation, offering additional insights into the mechanisms underlying neurodevelopmental disorders linked to Tcf4 mutations.
Collapse
Affiliation(s)
- Yandong Zhang
- Department of Anesthesia, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Dan Li
- Department of Anesthesia, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yuqun Cai
- Department of Anesthesia, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Rui Zou
- Department of Anesthesia, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yilan Zhang
- Department of Anesthesia, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Xin Deng
- Department of Anesthesia, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yafei Wang
- Department of Anesthesia, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Tianxiang Tang
- Department of Anesthesia, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yuanyuan Ma
- Department of Anesthesia, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Feizhen Wu
- Laboratory of Epi-Informatics, Intelligent Medicine Institute of Fudan University, Shanghai, 200032, China
| | - Yunli Xie
- Department of Anesthesia, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, and Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
2
|
Teng XY, Hu P, Zhang CM, Zhang QX, Yang G, Zang YY, Liu ZX, Chen G, Shi YS. OPALIN is an LGI1 receptor promoting oligodendrocyte differentiation. Proc Natl Acad Sci U S A 2024; 121:e2403652121. [PMID: 39083419 PMCID: PMC11317624 DOI: 10.1073/pnas.2403652121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 07/02/2024] [Indexed: 08/02/2024] Open
Abstract
Leucine-rich glioma-inactivated protein 1 (LGI1), a secretory protein in the brain, plays a critical role in myelination; dysfunction of this protein leads to hypomyelination and white matter abnormalities (WMAs). Here, we hypothesized that LGI1 may regulate myelination through binding to an unidentified receptor on the membrane of oligodendrocytes (OLs). To search for this hypothetic receptor, we analyzed LGI1 binding proteins through LGI1-3 × FLAG affinity chromatography with mouse brain lysates followed by mass spectrometry. An OL-specific membrane protein, the oligodendrocytic myelin paranodal and inner loop protein (OPALIN), was identified. Conditional knockout (cKO) of OPALIN in the OL lineage caused hypomyelination and WMAs, phenocopying LGI1 deficiency in mice. Biochemical analysis revealed the downregulation of Sox10 and Olig2, transcription factors critical for OL differentiation, further confirming the impaired OL maturation in Opalin cKO mice. Moreover, virus-mediated re-expression of OPALIN successfully restored myelination in Opalin cKO mice. In contrast, re-expression of LGI1-unbound OPALIN_K23A/D26A failed to reverse the hypomyelination phenotype. In conclusion, our study demonstrated that OPALIN on the OL membrane serves as an LGI1 receptor, highlighting the importance of the LGI1/OPALIN complex in orchestrating OL differentiation and myelination.
Collapse
Affiliation(s)
- Xiao-Yu Teng
- Guangdong Institute of Intelligence Science and Technology, 519031Hengqin, Zhuhai, China
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210032Nanjing, China
| | - Ping Hu
- Department of Prenatal Diagnosis, State Key Laboratory of Reproductive Medicine, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, 210004Nanjing, China
| | - Cai-Ming Zhang
- Department of Thoracic Surgery, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, 510315Guangzhou, China
| | - Qin-Xin Zhang
- Department of Prenatal Diagnosis, State Key Laboratory of Reproductive Medicine, Women’s Hospital of Nanjing Medical University, Nanjing Women and Children’s Healthcare Hospital, 210004Nanjing, China
| | - Guolin Yang
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210032Nanjing, China
| | - Yan-Yu Zang
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210032Nanjing, China
| | - Zhi-Xiong Liu
- Guangdong Institute of Intelligence Science and Technology, 519031Hengqin, Zhuhai, China
| | - Guiquan Chen
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210032Nanjing, China
| | - Yun Stone Shi
- Guangdong Institute of Intelligence Science and Technology, 519031Hengqin, Zhuhai, China
- Ministry of Education Key Laboratory of Model Animal for Disease Study, Model Animal Research Center, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, 210032Nanjing, China
| |
Collapse
|
3
|
Isasi E, Wajner M, Duarte JA, Olivera-Bravo S. Cerebral White Matter Alterations Associated With Oligodendrocyte Vulnerability in Organic Acidurias: Insights in Glutaric Aciduria Type I. Neurotox Res 2024; 42:33. [PMID: 38963434 DOI: 10.1007/s12640-024-00710-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 04/27/2024] [Accepted: 06/14/2024] [Indexed: 07/05/2024]
Abstract
The white matter is an important constituent of the central nervous system, containing axons, oligodendrocytes, and its progenitor cells, astrocytes, and microglial cells. Oligodendrocytes are central for myelin synthesis, the insulating envelope that protects axons and allows normal neural conduction. Both, oligodendrocytes and myelin, are highly vulnerable to toxic factors in many neurodevelopmental and neurodegenerative disorders associated with disturbances of myelination. Here we review the main alterations in oligodendrocytes and myelin observed in some organic acidurias/acidemias, which correspond to inherited neurometabolic disorders biochemically characterized by accumulation of potentially neurotoxic organic acids and their derivatives. The yet incompletely understood mechanisms underlying the high vulnerability of OLs and/or myelin in glutaric acidemia type I, the most prototypical cerebral organic aciduria, are particularly discussed.
Collapse
Affiliation(s)
- Eugenia Isasi
- Laboratorio de Neurobiología Celular y Molecular, Unidad Académica de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Departamento de Neurobiología y Neuropatología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
| | - Moacir Wajner
- Department of Biochemistry, Instituto de Ciencias Básicas da Saude, Universidade Federal de Río Grande do Sul, Porto Alegre, Brazil
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Juliana Avila Duarte
- Departamento de Medicina Interna, Serviço de Radiología, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Silvia Olivera-Bravo
- Departamento de Neurobiología y Neuropatología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay.
| |
Collapse
|
4
|
Yugami M, Hayakawa-Yano Y, Ogasawara T, Yokoyama K, Furukawa T, Hara H, Hashikami K, Tsuji I, Takebayashi H, Araki S, Okano H, Yano M. Sbp2l contributes to oligodendrocyte maturation through translational control in Tcf7l2 signaling. iScience 2023; 26:108451. [PMID: 38213786 PMCID: PMC10783607 DOI: 10.1016/j.isci.2023.108451] [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: 05/24/2023] [Revised: 10/09/2023] [Accepted: 11/10/2023] [Indexed: 01/13/2024] Open
Abstract
Oligodendrocytes (OLs) are the myelin-forming cells in the CNS that support neurons through the insulating sheath of axons. This unique feature and developmental processes are achieved by extrinsic and intrinsic gene expression programs, where RNA-binding proteins can contribute to dynamic and fine-tuned post-transcriptional regulation. Here, we identified SECIS-binding protein 2-like (Sbp2l), which is specifically expressed in OLs by integrated transcriptomics. Histological analysis revealed that Sbp2l is a molecular marker of OL maturation. Sbp2l knockdown (KD) led to suppression of matured OL markers, but not a typical selenoprotein, Gpx4. Transcriptome analysis demonstrated that Sbp2l KD decreased cholesterol-biosynthesis-related genes regulated by Tcf7l2 transcription factor. Indeed, we confirmed the downregulation of Tcf7l2 protein without changing its mRNA in Sbp2l KD OPCs. Furthermore, Sbp2l KO mice showed the decrease of Tcf7l2 protein and deficiency of OL maturation. These results suggest that Sbp2l contributes to OL maturation by translational control of Tcf7l2.
Collapse
Affiliation(s)
- Masato Yugami
- Research, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Yoshika Hayakawa-Yano
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, 1-757, Asahimachidori, Chuo-ku, Niigata, Niigata 951-8510, Japan
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takahisa Ogasawara
- Research, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Kazumasa Yokoyama
- Research, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Takako Furukawa
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, 1-757, Asahimachidori, Chuo-ku, Niigata, Niigata 951-8510, Japan
| | - Hiroe Hara
- Research, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Kentaro Hashikami
- Research, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Isamu Tsuji
- Research, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, 1-757, Asahimachidori, Chuo-ku, Niigata, Niigata 951-8510, Japan
| | - Shinsuke Araki
- Research, Takeda Pharmaceutical Company Limited, 26-1 Muraoka-higashi 2-chome, Fujisawa, Kanagawa 251-8555, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Masato Yano
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, 1-757, Asahimachidori, Chuo-ku, Niigata, Niigata 951-8510, Japan
- Department of Physiology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| |
Collapse
|
5
|
Hu HJ, Fan DF, Ye ZH, Sun Q. Effects of hyperbaric oxygen on Notch signaling pathway after severe carbon monoxide poisoning in mice. Med Gas Res 2023; 13:23-28. [PMID: 35946219 PMCID: PMC9480357 DOI: 10.4103/2045-9912.344971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/20/2021] [Accepted: 03/27/2021] [Indexed: 11/04/2022] Open
Abstract
Demyelination of the cerebral white matter is the most common pathological change after carbon monoxide (CO) poisoning. Notch signaling, the mechanism underlying the differentiation of astrocytes and oligodendrocytes, is critical to remyelination of the white matter after brain lesion. The purpose of this work was to determine the effects of hyperbaric oxygen (HBO) on Notch signaling pathway after CO poisoning for the explanation of the protective effects of HBO on CO-poisoning-related cerebral white matter demyelination. The male C57 BL/6 mice with severe CO poisoning were treated by HBO. And HBO therapy shortened the escape latency and improved the body mass after CO poisoning. HBO therapy also significantly suppressed protein and mRNA levels of Notch1 and Hes5 after CO poisoning. Our findings suggested that HBO could suppress the activation of Notch signaling pathway after CO poisoning, which is the mechanism underlying the neuroprotection of HBO on demyelination after severe CO poisoning.
Collapse
Affiliation(s)
- Hui-Jun Hu
- Department of Hyperbaric Medicine, the Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Dan-Feng Fan
- Department of Hyperbaric Medicine, the Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhou-Heng Ye
- Department of Special Operations Medicine, the Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Qiang Sun
- Department of Hyperbaric Medicine, the Sixth Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
6
|
Huang H, He W, Tang T, Qiu M. Immunological Markers for Central Nervous System Glia. Neurosci Bull 2022; 39:379-392. [PMID: 36028641 PMCID: PMC10043115 DOI: 10.1007/s12264-022-00938-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/09/2022] [Indexed: 10/15/2022] Open
Abstract
Glial cells in the central nervous system (CNS) are composed of oligodendrocytes, astrocytes and microglia. They contribute more than half of the total cells of the CNS, and are essential for neural development and functioning. Studies on the fate specification, differentiation, and functional diversification of glial cells mainly rely on the proper use of cell- or stage-specific molecular markers. However, as cellular markers often exhibit different specificity and sensitivity, careful consideration must be given prior to their application to avoid possible confusion. Here, we provide an updated overview of a list of well-established immunological markers for the labeling of central glia, and discuss the cell-type specificity and stage dependency of their expression.
Collapse
Affiliation(s)
- Hao Huang
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, China.
| | - Wanjun He
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, China
| | - Tao Tang
- Department of Anatomy, Cell Biology and Physiology Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Mengsheng Qiu
- Zhejiang Key Laboratory of Organ Development and Regeneration, Institute of Life Sciences, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 311121, China.
| |
Collapse
|
7
|
Ngo C, Kothary R. MicroRNAs in oligodendrocyte development and remyelination. J Neurochem 2022; 162:310-321. [PMID: 35536759 DOI: 10.1111/jnc.15618] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Revised: 03/14/2022] [Accepted: 04/20/2022] [Indexed: 11/28/2022]
Abstract
Oligodendrocytes are the glial cells responsible for the formation of myelin around axons of the central nervous system (CNS). Myelin is an insulating layer that allows electrical impulses to transmit quickly and efficiently along neurons. If myelin is damaged, as in chronic demyelinating disorders such as multiple sclerosis (MS), these impulses slow down. Remyelination by oligodendrocytes is often ineffective in MS, in part because of the failure of oligodendrocyte precursor cells (OPCs) to differentiate into mature, myelinating oligodendrocytes. The process of oligodendrocyte differentiation is tightly controlled by several regulatory networks involving transcription factors, intracellular signaling pathways, and extrinsic cues. Understanding the factors that regulate oligodendrocyte development is essential for the discovery of new therapeutic strategies capable of enhancing remyelination. Over the past decade, microRNAs (miRNAs) have emerged as key regulators of oligodendrocyte development, exerting effects on cell specification, proliferation, differentiation, and myelination. This article will review the role of miRNAs on oligodendrocyte biology and discuss their potential as promising therapeutic tools for remyelination.
Collapse
Affiliation(s)
- Clarissa Ngo
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Program in Biomedical Sciences, Faculty of Science, University of Ottawa, Ottawa, Ontario, Canada
| | - Rashmi Kothary
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada.,Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario, Canada.,Centre for Neuromuscular Disease, University of Ottawa, Ottawa, Ontario, Canada.,Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, and Department of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
8
|
Matrine inhibits the Wnt3a/β-catenin/TCF7L2 signaling pathway in experimental autoimmune encephalomyelitis. J Neuroimmunol 2022; 367:577876. [DOI: 10.1016/j.jneuroim.2022.577876] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/03/2022] [Accepted: 04/19/2022] [Indexed: 02/07/2023]
|
9
|
Manzari-Tavakoli A, Babajani A, Farjoo MH, Hajinasrollah M, Bahrami S, Niknejad H. The Cross-Talks Among Bone Morphogenetic Protein (BMP) Signaling and Other Prominent Pathways Involved in Neural Differentiation. Front Mol Neurosci 2022; 15:827275. [PMID: 35370542 PMCID: PMC8965007 DOI: 10.3389/fnmol.2022.827275] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/14/2022] [Indexed: 11/21/2022] Open
Abstract
The bone morphogenetic proteins (BMPs) are a group of potent morphogens which are critical for the patterning, development, and function of the central nervous system. The appropriate function of the BMP pathway depends on its interaction with other signaling pathways involved in neural differentiation, leading to synergistic or antagonistic effects and ultimately favorable biological outcomes. These opposite or cooperative effects are observed when BMP interacts with fibroblast growth factor (FGF), cytokines, Notch, Sonic Hedgehog (Shh), and Wnt pathways to regulate the impact of BMP-induced signaling in neural differentiation. Herein, we review the cross-talk between BMP signaling and the prominent signaling pathways involved in neural differentiation, emphasizing the underlying basic molecular mechanisms regarding the process of neural differentiation. Knowing these cross-talks can help us to develop new approaches in regenerative medicine and stem cell based therapy. Recently, cell therapy has received significant attention as a promising treatment for traumatic or neurodegenerative diseases. Therefore, it is important to know the signaling pathways involved in stem cell differentiation toward neural cells. Our better insight into the cross-talk of signaling pathways during neural development would improve neural differentiation within in vitro tissue engineering approaches and pre-clinical practices and develop futuristic therapeutic strategies for patients with neurological disease.
Collapse
Affiliation(s)
- Asma Manzari-Tavakoli
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Rayan Center for Neuroscience & Behavior, Department of Biology, Faculty of Science, Ferdowsi University, Mashhad, Iran
| | - Amirhesam Babajani
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Hadi Farjoo
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mostafa Hajinasrollah
- Department of Stem Cells and Developmental Biology, Cell Sciences Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Soheyl Bahrami
- Ludwig Boltzmann Institute for Experimental and Clinical Traumatology in AUVA Research Center, Vienna, Austria
| | - Hassan Niknejad
- Department of Pharmacology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- *Correspondence: Hassan Niknejad
| |
Collapse
|
10
|
Koch K, Bartmann K, Hartmann J, Kapr J, Klose J, Kuchovská E, Pahl M, Schlüppmann K, Zühr E, Fritsche E. Scientific Validation of Human Neurosphere Assays for Developmental Neurotoxicity Evaluation. FRONTIERS IN TOXICOLOGY 2022; 4:816370. [PMID: 35295221 PMCID: PMC8915868 DOI: 10.3389/ftox.2022.816370] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 01/21/2022] [Indexed: 01/06/2023] Open
Abstract
There is a call for a paradigm shift in developmental neurotoxicity (DNT) evaluation, which demands the implementation of faster, more cost-efficient, and human-relevant test systems than current in vivo guideline studies. Under the umbrella of the Organisation for Economic Co-operation and Development (OECD), a guidance document is currently being prepared that instructs on the regulatory use of a DNT in vitro battery (DNT IVB) for fit-for-purpose applications. One crucial issue for OECD application of methods is validation, which for new approach methods (NAMs) requires novel approaches. Here, mechanistic information previously identified in vivo, as well as reported neurodevelopmental adversities in response to disturbances on the cellular and tissue level, are of central importance. In this study, we scientifically validate the Neurosphere Assay, which is based on human primary neural progenitor cells (hNPCs) and an integral part of the DNT IVB. It assesses neurodevelopmental key events (KEs) like NPC proliferation (NPC1ab), radial glia cell migration (NPC2a), neuronal differentiation (NPC3), neurite outgrowth (NPC4), oligodendrocyte differentiation (NPC5), and thyroid hormone-dependent oligodendrocyte maturation (NPC6). In addition, we extend our work from the hNPCs to human induced pluripotent stem cell-derived NPCs (hiNPCs) for the NPC proliferation (iNPC1ab) and radial glia assays (iNPC2a). The validation process we report for the endpoints studied with the Neurosphere Assays is based on 1) describing the relevance of the respective endpoints for brain development, 2) the confirmation of the cell type-specific morphologies observed in vitro, 3) expressions of cell type-specific markers consistent with those morphologies, 4) appropriate anticipated responses to physiological pertinent signaling stimuli and 5) alterations in specific in vitro endpoints upon challenges with confirmed DNT compounds. With these strong mechanistic underpinnings, we posit that the Neurosphere Assay as an integral part of the DNT in vitro screening battery is well poised for DNT evaluation for regulatory purposes.
Collapse
Affiliation(s)
- Katharina Koch
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Kristina Bartmann
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Julia Hartmann
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Julia Kapr
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Jördis Klose
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Eliška Kuchovská
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Melanie Pahl
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Kevin Schlüppmann
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Etta Zühr
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
| | - Ellen Fritsche
- IUF—Leibniz Research Institute for Environmental Medicine, Duesseldorf, Germany
- Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany
- *Correspondence: Ellen Fritsche,
| |
Collapse
|
11
|
Ren Q, Rao Y. The exit of axons and glial membrane from the developing Drosophila retina requires integrins. Mol Brain 2022; 15:2. [PMID: 34980203 PMCID: PMC8722191 DOI: 10.1186/s13041-021-00888-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/19/2021] [Indexed: 11/11/2022] Open
Abstract
Coordinated development of neurons and glia is essential for the establishment of neuronal circuits during embryonic development. In the developing Drosophila visual system, photoreceptor (R cell) axons and wrapping glial (WG) membrane extend from the eye disc through the optic stalk into the optic lobe. Extensive studies have identified a number of genes that control the establishment of R-cell axonal projection pattern in the optic lobe. The molecular mechanisms directing the exit of R-cell axons and WG membrane from the eye disc, however, remain unknown. In this study, we show that integrins are required in R cells for the extension of R-cell axons and WG membrane from the eye disc into the optic stalk. Knockdown of integrins in R cells but not WG caused the stalling of both R-cell axons and WG membrane in the eye disc. Interfering with the function of Rhea (i.e. the Drosophila ortholog of vertebrate talin and a key player of integrin-mediated adhesion), caused an identical stalling phenotype. These results support a key role for integrins on R-cell axons in directing R-cell axons and WG membrane to exit the eye disc.
Collapse
Affiliation(s)
- Qian Ren
- McGill Centre for Research in Neuroscience, McGill University Health Centre, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada.,Integrated Program in Neuroscience, McGill University Health Centre, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada
| | - Yong Rao
- McGill Centre for Research in Neuroscience, McGill University Health Centre, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada. .,Department of Neurology and Neurosurgery, McGill University Health Centre, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada. .,Integrated Program in Neuroscience, McGill University Health Centre, 1650 Cedar Avenue, Montreal, QC, H3G 1A4, Canada. .,Centre for Research in Neuroscience, McGill University Health Centre, Room L7-136, 1650 Cedar Avenue, Montreal, QC, Canada.
| |
Collapse
|
12
|
Hu N, Zou L. Multiple functions of Hes genes in the proliferation and differentiation of neural stem cells. Ann Anat 2021; 239:151848. [PMID: 34715307 DOI: 10.1016/j.aanat.2021.151848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 07/24/2021] [Accepted: 10/18/2021] [Indexed: 12/20/2022]
Abstract
The HES proteins (hairy and Enhancer of split (E(spl)) homologs) are basic helix-loop-helix (bHLH) transcription factors that regulate the proliferation and differentiation of stem cells. Family members HES1, 3, and 5 are all critical regulators of nervous system development. The Hes genes exhibit oscillatory expression levels, and this dynamic expression allows for the complex regulation of numerous downstream genes such as Ascl1, Neurog2, Olig2 involved in the differentiation of specific cell types. In addition, HES proteins act as hubs for the molecule crosstalk among Notch, Wnt, and other signaling pathways that regulate nervous system development.
Collapse
Affiliation(s)
- Nan Hu
- Department of Human Anatomy, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China
| | - Linqing Zou
- Department of Human Anatomy, Jiangsu Key Laboratory of Neuroregeneration, Nantong University, Nantong, Jiangsu 226001, China.
| |
Collapse
|
13
|
Zhao H, Gao XY, Wu XJ, Zhang YB, Wang XF. The Shh/Gli1 signaling pathway regulates regeneration via transcription factor Olig1 expression after focal cerebral ischemia in rats. Neurol Res 2021; 44:318-330. [PMID: 34592910 DOI: 10.1080/01616412.2021.1981106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
OBJECTIVE Ischemic stroke is a major cause of death in the global population, with a high disability and mortality rate. Lack of regenerative ability is considered to be the fundamental cause. This study aims to determine the effect of Shh pathway, which mediates regenerative signaling in response to CNS injury, on myelin repair and Olig1 expression in focal ischemic lesions in the rat. METHODS A model of middle cerebral artery occlusion (MCAO) was established using the intraluminal suture method where the middle cerebral artery (MCA) was restricted for 120 min. Cyclopamine, a specific inhibitor of Shh, or saline was administered 12 h after MCAO surgery and lasted for 7 days. After MCA occlusion, male Sprague-Dawley rats were randomly allocated to cyclopamine- or saline-treated groups. A group of no-injection animals after MCAO were used as controls. The Shh signaling pathway, myelinogenesis-related factor MBP and Olig1 were testedby immunohistochemistry and RT-PCR assay. RESULTS The levels of Shh and its component Gli1 were elevated from 1 d up to 14 d following ischemia, indicating that the Shh-Gli1 axis was broadly reactivated. Treatment with cyclopamine can partially block the Shh signaling pathway, prevent myelin repair, and decrease the Olig1 expression following ischemic stroke. CONCLUSION That blockade of Shh signaling concurrently with the creation of a lesion aggravated ischemic myelin damage, probably via its downstream effects on Olig1 transcription. Shh plays a contributory role during regeneration in the CNS, thereby providing promising new therapeutic strategies to assist in recovery from ischemic stroke.
Collapse
Affiliation(s)
- Hong Zhao
- Department of Neurology, Dalian Municipal Central Hospital, Dalian
| | - Xiao-Yu Gao
- Department of Neurology, Yuhuangding Hospital, Yantai
| | - Xiao-Jun Wu
- Department of Neurology, Anshan Hospital, the First Affiliated Hospital of China Medical University, Anshan
| | - Yong-Bo Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing
| | | |
Collapse
|
14
|
Guo F, Zhang YF, Liu K, Huang X, Li RX, Wang SY, Wang F, Xiao L, Mei F, Li T. Chronic Exposure to Alcohol Inhibits New Myelin Generation in Adult Mouse Brain. Front Cell Neurosci 2021; 15:732602. [PMID: 34512271 PMCID: PMC8429601 DOI: 10.3389/fncel.2021.732602] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Accepted: 08/10/2021] [Indexed: 12/01/2022] Open
Abstract
Chronic alcohol consumption causes cognitive impairments accompanying with white matter atrophy. Recent evidence has shown that myelin dynamics remain active and are important for brain functions in adulthood. For example, new myelin generation is required for learning and memory functions. However, it remains undetermined whether alcohol exposure can alter myelin dynamics in adulthood. In this study, we examine the effect of chronic alcohol exposure on myelin dynamics by using genetic approaches to label newly generated myelin (NG2-CreERt; mT/mG). Our results indicated that alcohol exposure (either 5% or 10% in drinking water) for 3 weeks remarkably reduced mGFP + /NG2- new myelin and mGFP + /CC1 + new oligodendrocytes in the prefrontal cortex and corpus callosum of 6-month-old NG2-CreERt; mT/mG mice as compared to controls without changing the mGFP + /NG2 + oligodendrocyte precursor cells (OPCs) density, suggesting that alcohol exposure may inhibit oligodendrocyte differentiation. In support with these findings, the alcohol exposure did not significantly alter apoptotic cell number or overall MBP expression in the brains. Further, the alcohol exposure decreased the histone deacetylase1 (HDAC1) expression in mGFP + /NG2 + OPCs, implying epigenetic mechanisms were involved in the arrested OPC differentiation. Together, our results indicate that chronic exposure to alcohol can inhibit myelinogenesis in the adult mouse brain and that may contribute to alcohol-related cognitive impairments.
Collapse
Affiliation(s)
- Feng Guo
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China.,The First Camp of Cadet Brigade, School of Basic Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yi-Fan Zhang
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China.,The First Camp of Cadet Brigade, School of Basic Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Kun Liu
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xu Huang
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Rui-Xue Li
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shu-Yue Wang
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fei Wang
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lan Xiao
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Feng Mei
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China
| | - Tao Li
- Brain and Intelligence Research Key Laboratory of Chongqing Education Commission, Department of Histology and Embryology, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
15
|
Wang W, Cho H, Kim D, Park Y, Moon JH, Lim SJ, Yoon SM, McCane M, Aicher SA, Kim S, Emery B, Lee JW, Lee S, Park Y, Lee SK. PRC2 Acts as a Critical Timer That Drives Oligodendrocyte Fate over Astrocyte Identity by Repressing the Notch Pathway. Cell Rep 2021; 32:108147. [PMID: 32937136 PMCID: PMC8070886 DOI: 10.1016/j.celrep.2020.108147] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 03/13/2020] [Accepted: 08/22/2020] [Indexed: 11/30/2022] Open
Abstract
PRC2 creates the repressive mark histone H3 Lys27 trimethylation. Although PRC2 is involved in various biological processes, its role in glial development remains ambiguous. Here, we show that PRC2 is required for oligodendrocyte (OL) differentiation and myelination, but not for OL precursor formation. PRC2-deficient OL lineage cells differentiate into OL precursors, but they fail to trigger the molecular program for myelination, highlighting that PRC2 is essential for directing the differentiation timing of OL precursors. PRC2 null OL lineage cells aberrantly induce Notch pathway genes and acquire astrocytic features. The repression of the Notch pathway restores the myelination program and inhibits abnormal astrocytic differentiation in the PRC2-deficient OL lineage, indicating that Notch is a major target of PRC2. Altogether, our studies propose a specific action of PRC2 as a novel gatekeeper that determines the glial fate choice and the timing of OL lineage progression and myelination by impinging on the Notch pathway. Wang et al. show that the polycomb repressive complex PRC2 is required for the differentiation of oligodendrocyte precursors to myelinating oligodendrocytes. They further show that PRC2 promotes oligodendrocyte differentiation and inhibits erroneous astrocytic fate by repressing the Notch pathway.
Collapse
Affiliation(s)
- Wenxian Wang
- Department of Biological Sciences, College of Arts and Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Hyeyoung Cho
- Computational Biology Program, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Dongkyeong Kim
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Younjung Park
- Department of Biological Sciences, College of Arts and Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Ji Hwan Moon
- Department of Biological Sciences, College of Arts and Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Su Jeong Lim
- Department of Bioinformatics and Life Science, Soongsil University, Seoul, Korea
| | - Sung Min Yoon
- Department of Bioinformatics and Life Science, Soongsil University, Seoul, Korea
| | - Michael McCane
- Jungers Center for Neurosciences Research, Department of Neurology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Sue A Aicher
- Department of Physiology and Pharmacology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Sangsoo Kim
- Department of Bioinformatics and Life Science, Soongsil University, Seoul, Korea
| | - Ben Emery
- Jungers Center for Neurosciences Research, Department of Neurology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA
| | - Jae W Lee
- Department of Biological Sciences, College of Arts and Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA
| | - Seunghee Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08826, Korea
| | - Yungki Park
- Hunter James Kelly Research Institute, Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Soo-Kyung Lee
- Department of Biological Sciences, College of Arts and Sciences, University at Buffalo, The State University of New York, Buffalo, NY 14260, USA.
| |
Collapse
|
16
|
Multiple sclerosis risk gene Mertk is required for microglial activation and subsequent remyelination. Cell Rep 2021; 34:108835. [PMID: 33691116 DOI: 10.1016/j.celrep.2021.108835] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/23/2020] [Accepted: 02/17/2021] [Indexed: 12/26/2022] Open
Abstract
In multiple sclerosis (MS) and other neurological diseases, the failure to repair demyelinated lesions contributes to axonal damage and clinical disability. Here, we provide evidence that Mertk, a gene highly expressed by microglia that alters MS risk, is required for efficient remyelination. Compared to wild-type (WT) mice, Mertk-knockout (KO) mice show impaired clearance of myelin debris and remyelination following demyelination. Using single-cell RNA sequencing, we characterize Mertk-influenced responses to cuprizone-mediated demyelination and remyelination across different cell types. Mertk-KO brains show an attenuated microglial response to demyelination but an elevated proportion of interferon (IFN)-responsive microglia. In addition, we identify a transcriptionally distinct subtype of surviving oligodendrocytes specific to demyelinated lesions. The inhibitory effect of myelin debris on remyelination is mediated in part by IFNγ, which further impedes microglial clearance of myelin debris and inhibits oligodendrocyte differentiation. Together, our work establishes a role for Mertk in microglia activation, phagocytosis, and migration during remyelination.
Collapse
|
17
|
Scott K, O'Rourke R, Gillen A, Appel B. Prdm8 regulates pMN progenitor specification for motor neuron and oligodendrocyte fates by modulating the Shh signaling response. Development 2020; 147:dev191023. [PMID: 32680935 PMCID: PMC7473643 DOI: 10.1242/dev.191023] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/13/2020] [Indexed: 12/13/2022]
Abstract
Spinal cord pMN progenitors sequentially produce motor neurons and oligodendrocyte precursor cells (OPCs). Some OPCs differentiate rapidly as myelinating oligodendrocytes, whereas others remain into adulthood. How pMN progenitors switch from producing motor neurons to OPCs with distinct fates is poorly understood. pMN progenitors express prdm8, which encodes a transcriptional repressor, during motor neuron and OPC formation. To determine whether prdm8 controls pMN cell fate specification, we used zebrafish as a model system to investigate prdm8 function. Our analysis revealed that prdm8 mutant embryos have fewer motor neurons resulting from a premature switch from motor neuron to OPC production. Additionally, prdm8 mutant larvae have excess oligodendrocytes and a concomitant deficit of OPCs. Notably, pMN cells of mutant embryos have elevated Shh signaling, coincident with the motor neuron to OPC switch. Inhibition of Shh signaling restored the number of motor neurons to normal but did not rescue the proportion of oligodendrocytes. These data suggest that Prdm8 regulates the motor neuron-OPC switch by controlling the level of Shh activity in pMN progenitors, and also regulates the allocation of oligodendrocyte lineage cell fates.This article has an associated 'The people behind the papers' interview.
Collapse
Affiliation(s)
- Kayt Scott
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado 40045, USA
| | - Rebecca O'Rourke
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado 40045, USA
| | - Austin Gillen
- RNA Bioscience Initiative, University of Colorado School of Medicine, Aurora, Colorado 40045, USA
- Division of Hematology, University of Colorado School of Medicine, Aurora, Colorado 40045, USA
| | - Bruce Appel
- Department of Pediatrics, Section of Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado 40045, USA
| |
Collapse
|
18
|
Janowska J, Gargas J, Ziemka-Nalecz M, Zalewska T, Sypecka J. Oligodendrocyte Response to Pathophysiological Conditions Triggered by Episode of Perinatal Hypoxia-Ischemia: Role of IGF-1 Secretion by Glial Cells. Mol Neurobiol 2020; 57:4250-4268. [PMID: 32691304 PMCID: PMC7467917 DOI: 10.1007/s12035-020-02015-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 07/08/2020] [Indexed: 12/11/2022]
Abstract
Differentiation of oligodendrocyte progenitors towards myelinating cells is influenced by a plethora of exogenous instructive signals. Insulin-like growth factor 1 (IGF-1) is one of the major factors regulating cell survival, proliferation, and maturation. Recently, there is an ever growing recognition concerning the role of autocrine/paracrine IGF-1 signaling in brain development and metabolism. Since oligodendrocyte functioning is altered after the neonatal hypoxic-ischemic (HI) insult, a question arises if the injury exerts any influence on the IGF-1 secreted by neural cells and how possibly the change in IGF-1 concentration affects oligodendrocyte growth. To quantify the secretory activity of neonatal glial cells, the step-wise approach by sequentially using the in vivo, ex vivo, and in vitro models of perinatal asphyxia was applied. A comparison of the results of in vivo and ex vivo studies allowed evaluating the role of autocrine/paracrine IGF-1 signaling. Accordingly, astroglia were indicated to be the main local source of IGF-1 in the developing brain, and the factor secretion was shown to be significantly upregulated during the first 24 h after the hypoxic-ischemic insult. And conversely, the IGF-1 amounts released by oligodendrocytes and microglia significantly decreased. A morphometric examination of oligodendrocyte differentiation by means of the Sholl analysis showed that the treatment with low IGF-1 doses markedly improved the branching of oligodendroglial cell processes and, in this way, promoted their differentiation. The changes in the IGF-1 amounts in the nervous tissue after HI might contribute to the resulting white matter disorders, observed in newborn children who experienced perinatal asphyxia. Pharmacological modulation of IGF-1 secretion by neural cells could be reasonable solution in studies aimed at searching for therapies alleviating the consequences of perinatal asphyxia.
Collapse
Affiliation(s)
- Justyna Janowska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5, A. Pawinskiego Str., 02-106, Warsaw, Poland
| | - Justyna Gargas
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5, A. Pawinskiego Str., 02-106, Warsaw, Poland
| | - Malgorzata Ziemka-Nalecz
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5, A. Pawinskiego Str., 02-106, Warsaw, Poland
| | - Teresa Zalewska
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5, A. Pawinskiego Str., 02-106, Warsaw, Poland
| | - Joanna Sypecka
- NeuroRepair Department, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5, A. Pawinskiego Str., 02-106, Warsaw, Poland.
| |
Collapse
|
19
|
White matter injury and neurodevelopmental disabilities: A cross-disease (dis)connection. Prog Neurobiol 2020; 193:101845. [PMID: 32505757 DOI: 10.1016/j.pneurobio.2020.101845] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 05/19/2020] [Accepted: 06/01/2020] [Indexed: 12/13/2022]
Abstract
White matter (WM) injury, once known primarily in preterm newborns, is emerging in its non-focal (diffused), non-necrotic form as a critical component of subtle brain injuries in many early-life diseases like prematurity, intrauterine growth restriction, congenital heart defects, and hypoxic-ischemic encephalopathy. While advances in medical techniques have reduced the number of severe outcomes, the incidence of tardive impairments in complex cognitive functions or psychopathology remains high, with lifelong detrimental effects. The importance of WM in coordinating neuronal assemblies firing and neural groups synchronizing within multiple frequency bands through myelination, even mild alterations in WM structure, may interfere with the cognitive performance that increasing social and learning demands would exploit tardively during children growth. This phenomenon may contribute to explaining longitudinally the high incidence of late-appearing impairments that affect children with a history of perinatal insults. Furthermore, WM abnormalities have been highlighted in several neuropsychiatric disorders, such as autism and schizophrenia. In this review, we gather and organize evidence on how diffused WM injuries contribute to neurodevelopmental disorders through different perinatal diseases and insults. An insight into a possible common, cross-disease, mechanism, neuroimaging and monitoring, biomarkers, and neuroprotective strategies will also be presented.
Collapse
|
20
|
Xiao G, Du J, Wu H, Ge X, Xu X, Yang A, Zhu Y, Hu X, Zheng K, Zhu Q, Qiu M. Differential Inhibition of Sox10 Functions by Notch-Hes Pathway. Cell Mol Neurobiol 2020; 40:653-662. [PMID: 31782037 DOI: 10.1007/s10571-019-00764-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 11/20/2019] [Indexed: 12/13/2022]
Abstract
In the developing central nervous system, the terminal differentiation of oligodendrocytes (OLs) is regulated by both extrinsic and intrinsic factors. Recent studies have suggested that the Notch-Hes signaling pathway influences the maturation of oligodendrocytes in culture and during development. However, the specific Notch receptors and their downstream effectors Hes genes that are involved in oligodendrocyte maturation have not been investigated systematically. In this study, we showed that Notch1 and Notch3 are expressed in oligodendrocyte precursor cells (OPCs) during gliogenesis, and Hes5 is the major Notch downstream transcription factor that is transiently expressed in OPCs. Overexpression of Notch intracellular domain (NICD) and Hes5 proteins in embryonic chicken spinal cord suppressed both the endogenous and Sox10-induced Mbp gene expression. Unexpectedly, overexpression of NICD/Hes5 did not inhibit Sox10 induction of Olig2 expression and Myrf induced Mbp expression, suggesting the differential inhibitory effects of NICD/Hes5 signaling on Sox10 activation of myelin-related genes and early progenitor genes.
Collapse
Affiliation(s)
- Guanxiu Xiao
- College of Life Sciences, Zhejiang University, Hangzhou, 310036, China
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 310036, China
| | - Junqing Du
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 310036, China
| | - Huihui Wu
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 310036, China
| | - Xinqi Ge
- College of Life Sciences, Zhejiang University, Hangzhou, 310036, China
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 310036, China
| | - Xiaofeng Xu
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 310036, China
| | - Aifen Yang
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 310036, China
| | - Ying Zhu
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, 40292, USA
| | - Xuemei Hu
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, 40292, USA
| | - Kang Zheng
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 310036, China
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, 40292, USA
| | - Qiang Zhu
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, 40292, USA
| | - Mengsheng Qiu
- College of Life Sciences, Zhejiang University, Hangzhou, 310036, China.
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, 310036, China.
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, KY, 40292, USA.
| |
Collapse
|
21
|
Lecca D, Raffaele S, Abbracchio MP, Fumagalli M. Regulation and signaling of the GPR17 receptor in oligodendroglial cells. Glia 2020; 68:1957-1967. [PMID: 32086854 DOI: 10.1002/glia.23807] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/10/2020] [Accepted: 02/12/2020] [Indexed: 12/14/2022]
Abstract
Remyelination, namely, the formation of new myelin sheaths around denuded axons, counteracts axonal degeneration and restores neuronal function. Considerable advances have been made in understanding this regenerative process that often fails in diseases like multiple sclerosis, leaving axons demyelinated and vulnerable to damage, thus contributing to disease progression. The identification of the membrane receptor GPR17 on a subset of oligodendrocyte precursor cells (OPCs), which mediate remyelination in the adult central nervous system (CNS), has led to a huge amount of evidence that validated this receptor as a new attractive target for remyelinating therapies. Here, we summarize the role of GPR17 in OPC function, myelination and remyelination, describing its atypical pharmacology, its downstream signaling, and the genetic and epigenetic factors modulating its activity. We also highlight crucial insights into GPR17 pathophysiology coming from the demonstration that oligodendrocyte injury, associated with inflammation in chronic neurodegenerative conditions, is invariably characterized by abnormal and persistent GPR17 upregulation, which, in turn, is accompanied by a block of OPCs at immature premyelinating stages. Finally, we discuss the current literature in light of the potential exploitment of GPR17 as a therapeutic target to promote remyelination.
Collapse
Affiliation(s)
- Davide Lecca
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Stefano Raffaele
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Maria P Abbracchio
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| | - Marta Fumagalli
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
22
|
Zhang C, Huang H, Chen Z, Zhang Z, Lu W, Qiu M. The transcription factor NKX2-2 regulates oligodendrocyte differentiation through domain-specific interactions with transcriptional corepressors. J Biol Chem 2020; 295:1879-1888. [PMID: 31932307 DOI: 10.1074/jbc.ra119.011163] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/30/2019] [Indexed: 01/21/2023] Open
Abstract
The homeodomain protein NK2 homeobox 2 (NKX2-2) is a transcription factor that plays a critical role in the control of cell fate specification and differentiation in many tissues. In the developing central nervous system, this developmentally important transcription factor functions as a transcriptional repressor that governs oligodendrocyte (OL) differentiation and myelin gene expression, but the roles of various NKX2-2 structural domains in this process are unclear. In this study, using in situ hybridization, immunofluorescence, and coimmunoprecipitation, we determined the structural domains that mediate the repressive functions of murine NKX2-2 and identified the transcriptional corepressors that interact with it in OL cells. Through in ovo electroporation in embryonic chicken spinal cords, we demonstrate that the N-terminal Tinman domain and C-terminal domain synergistically promote OL differentiation by recruiting distinct transcriptional corepressors, including enhancer of split Groucho 3 (GRG3), histone deacetylase 1 (HDAC1), and DNA methyltransferase 3 α (DNMT3A). We also observed that the NK2-specific domain suppresses the function of the C-terminal domain in OL differentiation. These findings delineate the distinct NKX2-2 domains and their roles in OL differentiation and suggest that NKX2-2 regulates differentiation by repressing gene expression via multiple cofactors and molecular mechanisms.
Collapse
Affiliation(s)
- Chengfu Zhang
- College of Life Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China; Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, Hangzhou Normal University, 16 Xuelin Street, Hangzhou 311121, China
| | - Hao Huang
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, Hangzhou Normal University, 16 Xuelin Street, Hangzhou 311121, China
| | - Zhen Chen
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, Hangzhou Normal University, 16 Xuelin Street, Hangzhou 311121, China
| | - Zunyi Zhang
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, Hangzhou Normal University, 16 Xuelin Street, Hangzhou 311121, China
| | - Wenwen Lu
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, Hangzhou Normal University, 16 Xuelin Street, Hangzhou 311121, China
| | - Mengsheng Qiu
- College of Life Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058, China; Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, Hangzhou Normal University, 16 Xuelin Street, Hangzhou 311121, China.
| |
Collapse
|
23
|
Liu Z, Chen Y, Rao Y. An RNAi screen for secreted factors and cell-surface players in coordinating neuron and glia development in Drosophila. Mol Brain 2020; 13:1. [PMID: 31900209 PMCID: PMC6942347 DOI: 10.1186/s13041-019-0541-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 12/19/2019] [Indexed: 11/10/2022] Open
Abstract
The establishment of the functional nervous system requires coordinated development of neurons and glia in the embryo. Our understanding of underlying molecular and cellular mechanisms, however, remains limited. The developing Drosophila visual system is an excellent model for understanding the developmental control of the nervous system. By performing a systematic transgenic RNAi screen, we investigated the requirements of secreted proteins and cell-surface receptors for the development of photoreceptor neurons (R cells) and wrapping glia (WG) in the Drosophila visual system. From the screen, we identified seven genes whose knockdown disrupted the development of R cells and/or WG, including amalgam (ama), domeless (dome), epidermal growth factor receptor (EGFR), kuzbanian (kuz), N-Cadherin (CadN), neuroglian (nrg), and shotgun (shg). Cell-type-specific analysis revealed that ama is required in the developing eye disc for promoting cell proliferation and differentiation, which is essential for the migration of glia in the optic stalk. Our results also suggest that nrg functions in both eye disc and WG for coordinating R-cell and WG development.
Collapse
Affiliation(s)
- Zhengya Liu
- Centre for Research in Neuroscience, McGill University Health Centre, Room L7-136, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A4, Canada.,Integrated Program in Neuroscience, McGill University Health Centre, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A4, Canada
| | - Yixu Chen
- Centre for Research in Neuroscience, McGill University Health Centre, Room L7-136, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A4, Canada.,Department of Neurology and Neurosurgery, McGill University Health Centre, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A4, Canada
| | - Yong Rao
- Centre for Research in Neuroscience, McGill University Health Centre, Room L7-136, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A4, Canada. .,Integrated Program in Neuroscience, McGill University Health Centre, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A4, Canada. .,Department of Neurology and Neurosurgery, McGill University Health Centre, 1650 Cedar Avenue, Montreal, Quebec, H3G 1A4, Canada.
| |
Collapse
|
24
|
Zhao H, Gao XY, Liu ZH, Lin JW, Wang SP, Wang DX, Zhang YB. Effects of the transcription factor Olig1 on the differentiation and remyelination of oligodendrocyte precursor cells after focal cerebral ischemia in rats. Mol Med Rep 2019; 20:4603-4611. [PMID: 31702031 PMCID: PMC6797933 DOI: 10.3892/mmr.2019.10713] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2019] [Accepted: 07/22/2019] [Indexed: 12/28/2022] Open
Abstract
The differentiation and maturation of oligodendrocyte precursor cells (OPCs) is important for remyelination in the central nervous system. Nevertheless, this process is often limited and incomplete in ischemic injury. Oligodendrocyte transcription factor 1 (Olig1) is important for the maturation of OPCs and the repair of demyelinated lesions. However, how Olig1 modulates the development of OPCs or the remyelination associated with ischemic injury remains unclear. The present study aimed to examine alterations in OPCs, and the expression of myelin and Olig1, at different time-points after focal cerebral ischemia using immunohistochemistry and western blot techniques to elucidate the role of Olig1 in the maturation of OPCs and remyelination. The present results showed that the expression of Olig1 significantly decreased at 1 day after middle cerebral artery occlusion (MCAO) and returned to normal levels from day 3 to 28. Additionally, Olig1 was found to translocate into the nucleus following ischemia in the brain. The number of OPCs in the ischemic striatum significantly declined at days 1 and 3 following MCAO, and increased at days 7, 14 and 28 compared with the control. The expression of myelin basic protein, a marker of mature oligodendrocytes and myelin, gradually decreased from day 1 to 7 after ischemia and recovered at day 14 and 28; however, the levels were lower than those in the control group. The present results indicated that the restored normal level of Olig1 following ischemia may play an important role in the maturation of OPCs through its translocation into the nucleus, where it may promote the growth and development of myelin under pathological conditions. However, this endogenous recovery mechanism fails to fully repair the demyelinated lesion. The data of the present study may help clinicians understand the expression pattern of Olig1 and its potential role in endogenous remyelination after ischemia, which may have implications for the treatment of diseases that lead to demyelination.
Collapse
Affiliation(s)
- Hong Zhao
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Xiao-Yu Gao
- Department of Neurology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong 264000, P.R. China
| | - Zan-Hua Liu
- Department of Neurology, Nanjing Gaochun People's Hospital, Nanjing, Jiangsu 211300, P.R. China
| | - Jian-Wen Lin
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - Su-Ping Wang
- Department of Neurology, Dalian Municipal Central Hospital, Dalian, Liaoning 116033, P.R. China
| | - De-Xin Wang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| | - Yong-Bo Zhang
- Department of Neurology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, P.R. China
| |
Collapse
|
25
|
Li S, Zheng J, Chai L, Lin M, Zeng R, Lu J, Bian J. Rapid and Efficient Differentiation of Rodent Neural Stem Cells into Oligodendrocyte Progenitor Cells. Dev Neurosci 2019; 41:79-93. [DOI: 10.1159/000499364] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 03/04/2019] [Indexed: 11/19/2022] Open
Abstract
Oligodendrocyte progenitor cells (OPCs) may have beneficial effects in cell replacement therapy of neurodegenerative disease owing to their unique capability to differentiate into myelinogenic oligodendrocytes (OLs) in response to extrinsic signals. Therefore, it is of significance to establish an effective differentiation methodology to generate highly pure OPCs and OLs from some easily accessible stem cell sources. To achieve this goal, in this study, we present a rapid and efficient protocol for oligodendroglial lineage differentiation from mouse neural stem cells (NSCs), rat NSCs, or mouse embryonic stem cell-derived neuroepithelial stem cells. In a defined culture medium containing Smoothened Agonist, basic fibroblast growth factor, and platelet-derived growth factor-AA, OPCs could be generated from the above stem cells over a time course of 4–6 days, achieving a cell purity as high as ∼90%. In particular, these derived OPCs showed high expandability and could further differentiate into myelin basic protein-positive OLs within 3 days or alternatively into glial fibrillary acidic protein-positive astrocytes within 7 days. Furthermore, transplantation of rodent NSC-derived OPCs into injured spinal cord indicated that it is a feasible strategy to treat spinal cord injury. Our results suggest a differentiation strategy for robust production of OPCs and OLs from rodent stem cells, which could provide an abundant OPC source for spinal cord injury.
Collapse
|
26
|
Ge X, Xiao G, Huang H, Du J, Tao Y, Yang A, Wu H, Zhang Z, Qiu M. Stage-dependent regulation of oligodendrocyte development and enhancement of myelin repair by dominant negative Master-mind 1 protein. Glia 2019; 67:1654-1666. [PMID: 31038233 DOI: 10.1002/glia.23633] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/21/2019] [Accepted: 04/10/2019] [Indexed: 11/07/2022]
Abstract
Notch signaling has been implicated in the inhibition of oligodendrocyte differentiation and myelin gene expression during early development. However, inactivation of a particular Notch or Hes gene only produces a mild phenotype in oligodendrocyte development possibly due to the functional redundancies among closely related family members. To uncover the full role of Notch signaling in myelin development and regeneration, we generated the Sox10rtTA/+ ; TetO-dnMAML1 double transgenic mice in which expression of dominant negative Master-mind 1 (dnMAML1) gene can be selectively induced in oligodendrocyte precursor cells (OPCs) for complete blockade of Notch signaling. It is found that dnMAML1 expression leads to robust precocious OL differentiation and premature axonal myelination in the spinal cord, possibly by upregulating Nkx2.2 and downregulating Pdgfra expression. Unexpectedly, at late embryonic stages, dnMAML1 expression dramatically increased the number of OPCs, indicating a stage-dependent effect of Notch signaling on OPC proliferation. In addition, dnMAML1 also significantly enhances axonal remyelination following chemical-induced demyelination, providing a promising therapeutic target for lesion repair in demyelinating disease.
Collapse
Affiliation(s)
- Xinqi Ge
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Guanxiu Xiao
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Hao Huang
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Junqing Du
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Yanmei Tao
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Aifen Yang
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Huihui Wu
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Zunyi Zhang
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
| | - Mengsheng Qiu
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Institute of Life Sciences, Key Laboratory of Organ Development and Regeneration of Zhejiang Province, College of Life and Environmental Sciences, Hangzhou Normal University, Hangzhou, China
- Department of Anatomical Sciences and Neurobiology, School of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
27
|
Tulina NM, Brown AG, Barila GO, Elovitz MA. The Absence of TLR4 Prevents Fetal Brain Injury in the Setting of Intrauterine Inflammation. Reprod Sci 2018; 26:1082-1093. [PMID: 30463495 DOI: 10.1177/1933719118805859] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Exposure to intrauterine inflammation during pregnancy is linked to brain injury and neurobehavioral disorders in affected children. Innate immunity, specifically Toll-like receptor (TLR) signaling pathways are present throughout the reproductive tract as well as in the placenta, fetal membranes, and fetus. The TLR pathways are mechanistically involved in host responses to foreign pathogens and may lead to brain injury associated with prenatal inflammation. OBJECTIVE We aimed to determine whether the activation of the TLR4 signaling pathway, in the mother and fetus, is critical to fetal brain injury in the setting of intrauterine inflammation. METHODS A mini-laparotomy was performed on time pregnant C57B6 mice and 2 knockout mouse strains lacking the function of the Tlr4 and Myd88 genes on embryonic day 15. Intrauterine injections of Escherichia coli lipopolysaccharide or saline were administered as described previously. Dams were killed 6 hours postsurgery, and placental, amniotic fluid, and fetal brain tissue were collected. To assess brain injury, quantitative polymerase chain reaction (qPCR) analysis was performed on multiple components of the NOTCH signaling pathway, including Hes genes. Interleukin (IL) IL6, IL1β, and CCL5 expression was assessed using qPCR and enzyme-linked immunosorbent assay. RESULTS Using an established mouse model of intrauterine inflammation, we demonstrate that the abrogation of TLR4 signaling eliminates the cytokine response in mother and fetus and prevents brain injury associated with increased expression of transcriptional effectors of the NOTCH signaling pathway, Hes1 and Hes5. CONCLUSIONS These data show that the activation of the TLR4 signaling pathway is necessary for the development of fetal brain injury in response to intrauterine inflammation.
Collapse
Affiliation(s)
- Natalia M Tulina
- 1 Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy G Brown
- 1 Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Guillermo O Barila
- 1 Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michal A Elovitz
- 1 Department of Obstetrics and Gynecology, Maternal and Child Health Research Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
28
|
Daynac M, Chouchane M, Collins HY, Murphy NE, Andor N, Niu J, Fancy SPJ, Stallcup WB, Petritsch CK. Lgl1 controls NG2 endocytic pathway to regulate oligodendrocyte differentiation and asymmetric cell division and gliomagenesis. Nat Commun 2018; 9:2862. [PMID: 30131568 PMCID: PMC6104045 DOI: 10.1038/s41467-018-05099-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2017] [Accepted: 06/13/2018] [Indexed: 12/29/2022] Open
Abstract
Oligodendrocyte progenitor cells (OPC) undergo asymmetric cell division (ACD) to generate one OPC and one differentiating oligodendrocyte (OL) progeny. Loss of pro-mitotic proteoglycan and OPC marker NG2 in the OL progeny is the earliest immunophenotypic change of unknown mechanism that indicates differentiation commitment. Here, we report that expression of the mouse homolog of Drosophila tumor suppressor Lethal giant larvae 1 (Lgl1) is induced during OL differentiation. Lgl1 conditional knockout OPC progeny retain NG2 and show reduced OL differentiation, while undergoing more symmetric self-renewing divisions at the expense of asymmetric divisions. Moreover, Lgl1 and hemizygous Ink4a/Arf knockouts in OPC synergistically induce gliomagenesis. Time lapse and total internal reflection microscopy reveals a critical role for Lgl1 in NG2 endocytic routing and links aberrant NG2 recycling to failed differentiation. These data establish Lgl1 as a suppressor of gliomagenesis and positive regulator of asymmetric division and differentiation in the healthy and demyelinated murine brain.
Collapse
Affiliation(s)
- Mathieu Daynac
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Malek Chouchane
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Hannah Y Collins
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Neurobiology, Stanford University, Stanford, CA, 94305, USA
| | - Nicole E Murphy
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Noemi Andor
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
- Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Jianqin Niu
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94158, USA
| | - Stephen P J Fancy
- Department of Pediatrics, University of California San Francisco, San Francisco, CA, 94158, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, 94158, USA
| | - William B Stallcup
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA 92037, USA
| | - Claudia K Petritsch
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA, 94143, USA.
- Brain Tumor Center, University of California San Francisco, San Francisco, CA, 94158, USA.
- Helen Diller Comprehensive Cancer Center, University of California San Francisco, San Francisco, CA, 94158, USA.
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA, 94143, USA.
| |
Collapse
|
29
|
Klocke C, Allen JL, Sobolewski M, Blum JL, Zelikoff JT, Cory-Slechta DA. Exposure to fine and ultrafine particulate matter during gestation alters postnatal oligodendrocyte maturation, proliferation capacity, and myelination. Neurotoxicology 2018; 65:196-206. [PMID: 29079486 PMCID: PMC5857223 DOI: 10.1016/j.neuro.2017.10.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 10/23/2017] [Accepted: 10/23/2017] [Indexed: 12/12/2022]
Abstract
Accumulating studies indicate that the brain is a direct target of air pollution exposure during the fetal period. We have previously demonstrated that exposure to concentrated ambient particles (CAPs) during gestation produces ventriculomegaly, periventricular hypermyelination, and enlargement of the corpus callosum (CC) during postnatal development in mice. This study aimed to further characterize the cellular basis of the observed hypermyelination and determine if this outcome, among other effects, persisted as the brain matured. Analysis of CC-1+ mature oligodendrocytes in the CC at postnatal days (PNDs) 11-15 suggest a premature maturational shift in number and proportion of total cells in prenatally CAPs-exposed males and females, with no overall change in total CC cellularity. The overall number of Olig2+ lineage cells in the CC was not affected in either sex at the same postnatal timepoint. Assessment of myelin status at early brain maturity (PNDs 57-61) revealed persistent hypermyelination in CAPs-exposed animals of both sexes. In addition, ventriculomegaly was persistent in CAPs-treated females, with possible amelioration of ventriculomegaly in CAPs-exposed males. When oligodendrocyte precursor cell (OPC) pool status was analyzed at PNDs 57-61, there were significant CAPs-induced alterations in cycling Ki67+/Olig2+ cell number and proportion of total cells in the female CC. Total CC cellularity was slightly elevated in CAPs-exposed males at PNDs 57-61. Overall, these data support a growing body of evidence that demonstrate the vulnerability of the developing brain to environmental insults such as ambient particulate matter. The sensitivity of oligodendrocytes and myelin, in particular, to such an insult warrants further investigation into the mechanistic underpinnings of OPC and myelin disruption by constituent air pollutants.
Collapse
Affiliation(s)
- Carolyn Klocke
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, NY, USA.
| | - Joshua L Allen
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, NY, USA
| | - Marissa Sobolewski
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, NY, USA
| | - Jason L Blum
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, NY, USA
| | - Judith T Zelikoff
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, NY, USA
| | - Deborah A Cory-Slechta
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, NY, USA
| |
Collapse
|
30
|
Sil S, Periyasamy P, Thangaraj A, Chivero ET, Buch S. PDGF/PDGFR axis in the neural systems. Mol Aspects Med 2018; 62:63-74. [PMID: 29409855 DOI: 10.1016/j.mam.2018.01.006] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 12/08/2017] [Accepted: 01/22/2018] [Indexed: 12/14/2022]
Abstract
Platelet-derived growth factors (PDGFs) and their receptors (PDGFRs) are expressed in several cell types including the brain cells such as neuronal progenitors, neurons, astrocytes, and oligodendrocytes. Emerging evidence shows that PDGF-mediated signaling regulates diverse functions in the central nervous system (CNS) such as neurogenesis, cell survival, synaptogenesis, modulation of ligand-gated ion channels, and development of specific types of neurons. Interestingly, PDGF/PDFGR signaling can elicit paradoxical roles in the CNS, depending on the cell type and the activation stimuli and is implicated in the pathogenesis of various neurodegenerative diseases. This review summarizes the role of PDGFs/PDGFRs in several neurodegenerative diseases such as Alzheimer disease, Parkinson disease, amyotrophic lateral sclerosis, brain cancer, cerebral ischemia, HIV-1 and drug abuse. Understanding PDGF/PDGFR signaling may lead to novel approaches for the future development of therapeutic strategies for combating CNS pathologies.
Collapse
Affiliation(s)
- Susmita Sil
- Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Palsamy Periyasamy
- Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Annadurai Thangaraj
- Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Ernest T Chivero
- Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, 985880 Nebraska Medical Center, University of Nebraska Medical Center, Omaha, NE 68198, USA.
| |
Collapse
|
31
|
Myt1L Promotes Differentiation of Oligodendrocyte Precursor Cells and is Necessary for Remyelination After Lysolecithin-Induced Demyelination. Neurosci Bull 2018; 34:247-260. [PMID: 29397565 DOI: 10.1007/s12264-018-0207-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Accepted: 12/11/2017] [Indexed: 12/12/2022] Open
Abstract
The differentiation and maturation of oligodendrocyte precursor cells (OPCs) is essential for myelination and remyelination in the CNS. The failure of OPCs to achieve terminal differentiation in demyelinating lesions often results in unsuccessful remyelination in a variety of human demyelinating diseases. However, the molecular mechanisms controlling OPC differentiation under pathological conditions remain largely unknown. Myt1L (myelin transcription factor 1-like), mainly expressed in neurons, has been associated with intellectual disability, schizophrenia, and depression. In the present study, we found that Myt1L was expressed in oligodendrocyte lineage cells during myelination and remyelination. The expression level of Myt1L in neuron/glia antigen 2-positive (NG2+) OPCs was significantly higher than that in mature CC1+ oligodendrocytes. In primary cultured OPCs, overexpression of Myt1L promoted, while knockdown inhibited OPC differentiation. Moreover, Myt1L was potently involved in promoting remyelination after lysolecithin-induced demyelination in vivo. ChIP assays showed that Myt1L bound to the promoter of Olig1 and transcriptionally regulated Olig1 expression. Taken together, our findings demonstrate that Myt1L is an essential regulator of OPC differentiation, thereby supporting Myt1L as a potential therapeutic target for demyelinating diseases.
Collapse
|
32
|
Sox2 Is Essential for Oligodendroglial Proliferation and Differentiation during Postnatal Brain Myelination and CNS Remyelination. J Neurosci 2018; 38:1802-1820. [PMID: 29335358 DOI: 10.1523/jneurosci.1291-17.2018] [Citation(s) in RCA: 70] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 12/11/2017] [Accepted: 01/08/2018] [Indexed: 11/21/2022] Open
Abstract
In the CNS, myelination and remyelination depend on the successful progression and maturation of oligodendroglial lineage cells, including proliferation and differentiation of oligodendroglial progenitor cells (OPCs). Previous studies have reported that Sox2 transiently regulates oligodendrocyte (OL) differentiation in the embryonic and perinatal spinal cord and appears dispensable for myelination in the postnatal spinal cord. However, the role of Sox2 in OL development in the brain has yet to be defined. We now report that Sox2 is an essential positive regulator of developmental myelination in the postnatal murine brain of both sexes. Stage-specific paradigms of genetic disruption demonstrated that Sox2 regulated brain myelination by coordinating upstream OPC population supply and downstream OL differentiation. Transcriptomic analyses further supported a crucial role of Sox2 in brain developmental myelination. Consistently, oligodendroglial Sox2-deficient mice developed severe tremors and ataxia, typical phenotypes indicative of hypomyelination, and displayed severe impairment of motor function and prominent deficits of brain OL differentiation and myelination persisting into the later CNS developmental stages. We also found that Sox2 was required for efficient OPC proliferation and expansion and OL regeneration during remyelination in the adult brain and spinal cord. Together, our genetic evidence reveals an essential role of Sox2 in brain myelination and CNS remyelination, and suggests that manipulation of Sox2 and/or Sox2-mediated downstream pathways may be therapeutic in promoting CNS myelin repair.SIGNIFICANCE STATEMENT Promoting myelin formation and repair has translational significance in treating myelin-related neurological disorders, such as periventricular leukomalacia and multiple sclerosis in which brain developmental myelin formation and myelin repair are severely affected, respectively. In this report, analyses of a series of genetic conditional knock-out systems targeting different oligodendrocyte stages reveal a previously unappreciated role of Sox2 in coordinating upstream proliferation and downstream differentiation of oligodendroglial lineage cells in the mouse brain during developmental myelination and CNS remyelination. Our study points to the potential of manipulating Sox2 and its downstream pathways to promote oligodendrocyte regeneration and CNS myelin repair.
Collapse
|
33
|
van Tilborg E, de Theije CGM, van Hal M, Wagenaar N, de Vries LS, Benders MJ, Rowitch DH, Nijboer CH. Origin and dynamics of oligodendrocytes in the developing brain: Implications for perinatal white matter injury. Glia 2017; 66:221-238. [PMID: 29134703 PMCID: PMC5765410 DOI: 10.1002/glia.23256] [Citation(s) in RCA: 172] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 10/17/2017] [Accepted: 10/20/2017] [Indexed: 12/11/2022]
Abstract
Infants born prematurely are at high risk to develop white matter injury (WMI), due to exposure to hypoxic and/or inflammatory insults. Such perinatal insults negatively impact the maturation of oligodendrocytes (OLs), thereby causing deficits in myelination. To elucidate the precise pathophysiology underlying perinatal WMI, it is essential to fully understand the cellular mechanisms contributing to healthy/normal white matter development. OLs are responsible for myelination of axons. During brain development, OLs are generally derived from neuroepithelial zones, where neural stem cells committed to the OL lineage differentiate into OL precursor cells (OPCs). OPCs, in turn, develop into premyelinating OLs and finally mature into myelinating OLs. Recent studies revealed that OPCs develop in multiple waves and form potentially heterogeneous populations. Furthermore, it has been shown that myelination is a dynamic and plastic process with an excess of OPCs being generated and then abolished if not integrated into neural circuits. Myelination patterns between rodents and humans show high spatial and temporal similarity. Therefore, experimental studies on OL biology may provide novel insights into the pathophysiology of WMI in the preterm infant and offers new perspectives on potential treatments for these patients.
Collapse
Affiliation(s)
- Erik van Tilborg
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Caroline G M de Theije
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Maurik van Hal
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Nienke Wagenaar
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Linda S de Vries
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - Manon J Benders
- Department of Neonatology, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| | - David H Rowitch
- Department of Pediatrics, Eli and Edythe Broad Center for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, San Francisco, California.,Department of Paediatrics, Wellcome Trust-MRC Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Cora H Nijboer
- Laboratory of Neuroimmunology and Developmental Origins of Disease, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
34
|
Jin XR, Chen XS, Xiao L. MeCP2 Deficiency in Neuroglia: New Progress in the Pathogenesis of Rett Syndrome. Front Mol Neurosci 2017; 10:316. [PMID: 29046627 PMCID: PMC5632713 DOI: 10.3389/fnmol.2017.00316] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2017] [Accepted: 09/19/2017] [Indexed: 01/24/2023] Open
Abstract
Rett syndrome (RTT) is an X-linked neurodevelopmental disease predominantly caused by mutations of the methyl-CpG-binding protein 2 (MeCP2) gene. Generally, RTT has been attributed to neuron-centric dysfunction. However, increasing evidence has shown that glial abnormalities are also involved in the pathogenesis of RTT. Mice that are MeCP2-null specifically in glial cells showed similar behavioral and/or neuronal abnormalities as those found in MeCP2-null mice, a mouse model of RTT. MeCP2 deficiency in astrocytes impacts the expression of glial intermediate filament proteins such as fibrillary acidic protein (GFAP) and S100 and induces neuron toxicity by disturbing glutamate metabolism or enhancing microtubule instability. MeCP2 deficiency in oligodendrocytes (OLs) results in down-regulation of myelin gene expression and impacts myelination. While MeCP2-deficient microglia cells fail in response to environmental stimuli, release excessive glutamate, and aggravate impairment of the neuronal circuit. In this review, we mainly focus on the progress in determining the role of MeCP2 in glial cells involved in RTT, which may provide further insight into a therapeutic intervention for RTT.
Collapse
Affiliation(s)
- Xu-Rui Jin
- Department of Histology and Embryology, Faculty of Basic Medicine, Collaborative Program for Brain Research, Third Military Medical University, Chongqing, China.,The Cadet Brigade of Clinic Medicine, Third Military Medical University, Chongqing, China
| | - Xing-Shu Chen
- Department of Histology and Embryology, Faculty of Basic Medicine, Collaborative Program for Brain Research, Third Military Medical University, Chongqing, China
| | - Lan Xiao
- Department of Histology and Embryology, Faculty of Basic Medicine, Collaborative Program for Brain Research, Third Military Medical University, Chongqing, China
| |
Collapse
|
35
|
Liu C, Hu Q, Jing J, Zhang Y, Jin J, Zhang L, Mu L, Liu Y, Sun B, Zhang T, Kong Q, Wang G, Wang D, Zhang Y, Liu X, Zhao W, Wang J, Feng T, Li H. Regulator of G protein signaling 5 (RGS5) inhibits sonic hedgehog function in mouse cortical neurons. Mol Cell Neurosci 2017; 83:65-73. [PMID: 28684360 DOI: 10.1016/j.mcn.2017.06.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Revised: 02/21/2017] [Accepted: 06/20/2017] [Indexed: 12/12/2022] Open
Abstract
Regulator of G protein signaling 5 (RGS5) acts as a GTPase-activating protein (GAP) for the Gαi subunit and negatively regulates G protein-coupled receptor signaling. However, its presence and function in postmitotic differentiated primary neurons remains largely uncharacterized. During neural development, sonic hedgehog (Shh) signaling is involved in cell signaling pathways via Gαi activity. In particular, Shh signaling is essential for embryonic neural tube patterning, which has been implicated in neuronal polarization involving neurite outgrowth. Here, we examined whether RGS5 regulates Shh signaling in neurons. RGS5 transcripts were found to be expressed in cortical neurons and their expression gradually declined in a time-dependent manner in culture system. When an adenovirus expressing RGS5 was introduced into an in vitro cell culture model of cortical neurons, RGS5 overexpression significantly reduced neurite outgrowth and FM4-64 uptake, while cAMP-PKA signaling was also affected. These findings suggest that RGS5 inhibits Shh function during neurite outgrowth and the presynaptic terminals of primary cortical neurons mature via modulation of cAMP.
Collapse
Affiliation(s)
- Chuanliang Liu
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China; Vocational College Daxing'an Mountains, Jiagedaqi District, Heilongjiang 165000, China
| | - Qiongqiong Hu
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Jia Jing
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Yun Zhang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Jing Jin
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Liulei Zhang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Lili Mu
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Yumei Liu
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Bo Sun
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Tongshuai Zhang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Qingfei Kong
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Guangyou Wang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Dandan Wang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Yao Zhang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Xijun Liu
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Wei Zhao
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China
| | - Jinghua Wang
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China.
| | - Tao Feng
- Department of Neurology, The Nangang Branch of Heilongjiang Provincial Hospital, Harbin, Heilongjiang 150001, China.
| | - Hulun Li
- Department of Neurobiology, Harbin Medical University, Heilongjiang Provincial Key Laboratory of Neurobiology, Harbin, Heilongjiang 150086, China; Key Laboratories of Education Ministry for Myocardial Ischemia Mechanism and Treatment, Harbin, Heilongjiang 150086, China
| |
Collapse
|
36
|
Weng C, Ding M, Fan S, Cao Q, Lu Z. Transcription factor 7 like 2 promotes oligodendrocyte differentiation and remyelination. Mol Med Rep 2017; 16:1864-1870. [PMID: 28656232 PMCID: PMC5562062 DOI: 10.3892/mmr.2017.6843] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Accepted: 03/23/2017] [Indexed: 11/06/2022] Open
Abstract
Transcription factor 7 like 2 (TCF7L2, also termed TCF4), is a Wnt effector induced transiently in the oligodendroglial lineage. The current well accepted hypothesis is that TCF7L2 inhibits oligodendrocyte differentiation and remyelination through canonical Wnt/β‑catenin signaling. However, recent studies indicated that TCF7L2 activity is required during oligodendrocyte differentiation and remyelination. In order to clarify this, in situ hybridization, immunofluorescence and western blot analysis using in vivo TCF7L2 conditional knockout mice, were performed and it was found that TCF7L2 promotes oligodendrocyte differentiation during myelin formation and remyelination. Furthermore, it was established that TCF7L2 does not affect oligodendrocyte precursor cells during remyelination. These data are of important clinical significance to develop novel therapeutic targets to overcome multiple sclerosis and other demyelinating diseases.
Collapse
Affiliation(s)
- Chao Weng
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Man Ding
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Shanghua Fan
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Qian Cao
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Zuneng Lu
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
37
|
Roese-Koerner B, Stappert L, Brüstle O. Notch/Hes signaling and miR-9 engage in complex feedback interactions controlling neural progenitor cell proliferation and differentiation. NEUROGENESIS 2017; 4:e1313647. [PMID: 28573150 PMCID: PMC5443189 DOI: 10.1080/23262133.2017.1313647] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 11/27/2016] [Accepted: 01/18/2017] [Indexed: 02/04/2023]
Abstract
Canonical Notch signaling has diverse functions during nervous system development and is critical for neural progenitor self-renewal, timing of differentiation and specification of various cell fates. A key feature of Notch-mediated self-renewal is its fluctuating activity within the neural progenitor cell population and the oscillatory expression pattern of the Notch effector Hes1 and its target genes. A negative feedback loop between Hes1 and neurogenic microRNA miR-9 was found to be part of this oscillatory clock. In a recent study we discovered that miR-9 expression is further modulated by direct binding of the Notch intracellular domain/RBPj transcriptional complex to the miR-9_2 promoter. In turn, miR-9 not only targets Hes1 but also Notch2 to attenuate Notch signaling and promote neuronal differentiation. Here, we discuss how the two interwoven feedback loops may provide an additional fail-save mechanism to control proliferation and differentiation within the neural progenitor cell population. Furthermore, we explore potential implications of miR-9-mediated regulation of Notch/Hes1 signaling with regard to neural progenitor homeostasis, patterning, timing of differentiation and tumor formation.
Collapse
Affiliation(s)
- Beate Roese-Koerner
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn Medical Faculty, Bonn, Germany
| | - Laura Stappert
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn Medical Faculty, Bonn, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, LIFE & BRAIN Center, University of Bonn Medical Faculty, Bonn, Germany
| |
Collapse
|
38
|
Zhang Z, Li Z, Deng W, He Q, Wang Q, Shi W, Chen Q, Yang W, Spector M, Gong A, Yu J, Xu X. Ectoderm mesenchymal stem cells promote differentiation and maturation of oligodendrocyte precursor cells. Biochem Biophys Res Commun 2016; 480:727-733. [PMID: 27983986 DOI: 10.1016/j.bbrc.2016.10.115] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2016] [Accepted: 10/26/2016] [Indexed: 11/24/2022]
Abstract
Many neurological diseases are closely associated with demyelination caused by pathological changes of oligodendrocytes. Although intrinsic remyelination occurs after injury, the regeneration efficiency of myelinating oligodendrocytes remains to be improved. Herein, we reported an initiative finding of employing a valuable cell source, namely neural crest-derived ectoderm mesenchymal stem cells (EMSCs), for promoting oligodendrocyte differentiation and maturation by co-culturing oligodendrocyte precursor cells (OPCs) with the EMSCs. The results demonstrated that the OPCs/EMSCs co-culture could remarkably increase the number and length of oligodendrocyte processes in comparison with the mono-cultured OPCs and non-contact OPCs/EMSCs transwell culture. Furthermore, the inhibition experiments revealed that the EMSCs-produced soluble factor Sonic hedgehog, gap junction protein connexin 43 and extracellular matrix molecule laminin accounted for the promoted OPC differentiation since inhibiting the function of anyone of the three proteins led to substantial retraction of processes and detachment of oligodendrocytes. Altogether, OPCs/EMSCs co-culture system could be a paradigmatic approach for promoting differentiation and maturation of oligodendrocytes, and EMSCs will be a promising cell source for the treatment of neurological diseases caused by oligodendrocyte death and demyelination.
Collapse
Affiliation(s)
- Zhijian Zhang
- School of Medicine, Jiangsu University, Zhenjiang, 212001, PR China; Center for Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, 212001, PR China
| | - Zhengnan Li
- School of Medicine, Jiangsu University, Zhenjiang, 212001, PR China; Center for Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, 212001, PR China
| | - Wenwen Deng
- Center for Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, 212001, PR China; Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, 212001, PR China
| | - Qinghua He
- Center for Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, 212001, PR China; Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, 212001, PR China
| | - Qiang Wang
- Center for Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, 212001, PR China; Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, 212001, PR China
| | - Wentao Shi
- School of Medicine, Jiangsu University, Zhenjiang, 212001, PR China
| | - Qian Chen
- School of Medicine, Jiangsu University, Zhenjiang, 212001, PR China
| | - Wenjing Yang
- School of Medicine, Jiangsu University, Zhenjiang, 212001, PR China
| | - Myron Spector
- Department of Orthopedic Surgery, Harvard Medical School, Brigham and Women's Hospital, 75 Francis St, Boston, MA, 02115, USA
| | - Aihua Gong
- School of Medicine, Jiangsu University, Zhenjiang, 212001, PR China; Center for Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, 212001, PR China
| | - Jiangnan Yu
- Center for Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, 212001, PR China; Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, 212001, PR China
| | - Ximing Xu
- Center for Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, 212001, PR China; Department of Pharmaceutics, School of Pharmacy, Jiangsu University, Zhenjiang, 212001, PR China.
| |
Collapse
|
39
|
A neuronal PI(3,4,5)P 3-dependent program of oligodendrocyte precursor recruitment and myelination. Nat Neurosci 2016; 20:10-15. [PMID: 27775720 DOI: 10.1038/nn.4425] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/26/2016] [Indexed: 12/16/2022]
Abstract
The molecular trigger of CNS myelination is unknown. By targeting Pten in cerebellar granule cells and activating the AKT1-mTOR pathway, we increased the caliber of normally unmyelinated axons and the expression of numerous genes encoding regulatory proteins. This led to the expansion of genetically wild-type oligodendrocyte progenitor cells, oligodendrocyte differentiation and de novo myelination of parallel fibers. Thus, a neuronal program dependent on the phosphoinositide PI(3,4,5)P3 is sufficient to trigger all steps of myelination.
Collapse
|
40
|
Naruse M, Ishizaki Y, Ikenaka K, Tanaka A, Hitoshi S. Origin of oligodendrocytes in mammalian forebrains: a revised perspective. J Physiol Sci 2016; 67:63-70. [PMID: 27573166 PMCID: PMC5368213 DOI: 10.1007/s12576-016-0479-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 08/16/2016] [Indexed: 12/11/2022]
Abstract
Oligodendrocyte precursor cells (OPCs) appear in the late embryonic brain, mature into oligodendrocytes (OLs), and form myelin in the postnatal brain. It has been proposed that early born OPCs derived from the ventral forebrain are eliminated postnatally and late-born OLs predominate in the adult mouse cortex. However, the temporal and regional niche for cortical OL generation, which persists throughout life in adult mammals, remains to be determined. Our recent study provides new insight into self-renewing and multipotent neural stem cells (NSCs). Our results, together with previous studies, suggest that NSCs at the dorsoventral boundary are uniquely specialized to produce myelin-forming OLs in the cortex during a restricted temporal window. These findings may help identify transcription factors or gene expression patterns which confer neural precursors with the characteristic ability of dorsoventral boundary NSCs to differentiate into OLs, and facilitate the development of new strategies for regenerative medicine of the damaged brain.
Collapse
Affiliation(s)
- Masae Naruse
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Yasuki Ishizaki
- Department of Molecular and Cellular Neurobiology, Gunma University Graduate School of Medicine, Maebashi, Japan
| | - Kazuhiro Ikenaka
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan
| | - Aoi Tanaka
- Department of Integrative Physiology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Seiji Hitoshi
- Division of Neurobiology and Bioinformatics, National Institute for Physiological Sciences, Okazaki, Japan.
- Department of Integrative Physiology, Shiga University of Medical Science, Otsu, Shiga, Japan.
| |
Collapse
|
41
|
Zhang L, He X, Liu L, Jiang M, Zhao C, Wang H, He D, Zheng T, Zhou X, Hassan A, Ma Z, Xin M, Sun Z, Lazar MA, Goldman SA, Olson EN, Lu QR. Hdac3 Interaction with p300 Histone Acetyltransferase Regulates the Oligodendrocyte and Astrocyte Lineage Fate Switch. Dev Cell 2016; 36:316-30. [PMID: 26859354 DOI: 10.1016/j.devcel.2016.01.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 01/01/2016] [Accepted: 01/05/2016] [Indexed: 12/31/2022]
Abstract
Establishment and maintenance of CNS glial cell identity ensures proper brain development and function, yet the epigenetic mechanisms underlying glial fate control remain poorly understood. Here, we show that the histone deacetylase Hdac3 controls oligodendrocyte-specification gene Olig2 expression and functions as a molecular switch for oligodendrocyte and astrocyte lineage determination. Hdac3 ablation leads to a significant increase of astrocytes with a concomitant loss of oligodendrocytes. Lineage tracing indicates that the ectopic astrocytes originate from oligodendrocyte progenitors. Genome-wide occupancy analysis reveals that Hdac3 interacts with p300 to activate oligodendroglial lineage-specific genes, while suppressing astroglial differentiation genes including NFIA. Furthermore, we find that Hdac3 modulates the acetylation state of Stat3 and competes with Stat3 for p300 binding to antagonize astrogliogenesis. Thus, our data suggest that Hdac3 cooperates with p300 to prime and maintain oligodendrocyte identity while inhibiting NFIA and Stat3-mediated astrogliogenesis, and thereby regulates phenotypic commitment at the point of oligodendrocyte-astrocytic fate decision.
Collapse
Affiliation(s)
- Liguo Zhang
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Xuelian He
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Lei Liu
- Department of Pediatrics, West China Second Hospital, Sichuan University, Chengdu 610041, China
| | - Minqing Jiang
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Chuntao Zhao
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Haibo Wang
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Danyang He
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Molecular Biology and Integrated Biology Program, University of Texas Southwestern Medical Center, Dallas, TX 75239, USA
| | - Tao Zheng
- Department of Pediatrics, West China Second Hospital, Sichuan University, Chengdu 610041, China
| | - Xianyao Zhou
- Department of Pediatrics, West China Second Hospital, Sichuan University, Chengdu 610041, China
| | - Aishlin Hassan
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Zhixing Ma
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Mei Xin
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Zheng Sun
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mitchell A Lazar
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Steven A Goldman
- Center for Translational Neuromedicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA
| | - Eric N Olson
- Department of Molecular Biology and Integrated Biology Program, University of Texas Southwestern Medical Center, Dallas, TX 75239, USA
| | - Q Richard Lu
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Key Laboratory of Birth Defects, Children's Hospital of Fudan University, Shanghai 201102, China.
| |
Collapse
|
42
|
Tong M, Andreani T, Krotow A, Gundogan F, de la Monte SM. Potential Contributions of the Tobacco Nicotine-Derived Nitrosamine Ketone to White Matter Molecular Pathology in Fetal Alcohol Spectrum Disorder. ACTA ACUST UNITED AC 2016; 3. [PMID: 28868525 PMCID: PMC5575815 DOI: 10.15436/2377-1348.16.729] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Background Fetal alcohol spectrum disorder (FASD) is associated with long-term
deficits in cognitive and motor functions. Previous studies linked
neurodevelopmental abnormalities to increased oxidative stress and white
matter hypotrophy. However, similar effects occur with low-dose nitrosamine
exposures, alcohol abuse correlates with cigarette smoking, and tobacco
smoke contains tobacco-specific nitrosamines, including NNK. Hypothesis Tobacco smoke exposure is a co-factor in FASD. Design Long Evans rat pups were i.p. administered ethanol (2 g/kg) on
postnatal days (P) 2, 4, 6 and/or NNK (2 mg/kg) on P3, P5, and P7 to
simulate third trimester human exposures. Oligodendroglial
myelin-associated, neuroglial, and relevant transcription factor mRNA
transcripts were measured using targeted PCR arrays. Results Ethanol and NNK differentially altered the expression of immature and
mature oligodendroglial, neuronal and astrocytic structural and
plasticity-associated, and various transcription factor genes. NNK’s
effects were broader and more pronounced than ethanol’s, and
additive or synergistic effects of dual exposures impacted expression of all
four categories of genes investigated. Conclusion Developmental exposures to alcohol and NNK (via tobacco smoke)
contribute to sustained abnormalities in brain white matter structure and
function via distinct but overlapping alterations in the expression of genes
that regulate oligodendrocyte survival, maturation and function, neuroglial
structural integrity, and synaptic plasticity. The results support the
hypothesis that smoking may contribute to brain abnormalities associated
with FASD.
Collapse
Affiliation(s)
- Ming Tong
- Department of Medicine, Division of Gastroenterology, and the Liver Research Center Rhode Island Hospital, Providence, RI.,Warren Alpert Medical School of Brown University, Providence, RI
| | - Tomas Andreani
- Department of Medicine, Division of Gastroenterology, and the Liver Research Center Rhode Island Hospital, Providence, RI
| | | | - Fusun Gundogan
- Warren Alpert Medical School of Brown University, Providence, RI.,Department of Pathology, Women and Infants Hospital of Rhode Island, Providence, RI
| | - Suzanne M de la Monte
- Department of Medicine, Division of Gastroenterology, and the Liver Research Center Rhode Island Hospital, Providence, RI.,Warren Alpert Medical School of Brown University, Providence, RI.,Pathobiology Graduate Program, Brown University, Providence, RI.,Departments of Pathology and Neurology, and the Division of Neuropathology, Rhode Island Hospital, Providence, RI
| |
Collapse
|
43
|
The Activators of Cyclin-Dependent Kinase 5 p35 and p39 Are Essential for Oligodendrocyte Maturation, Process Formation, and Myelination. J Neurosci 2016; 36:3024-37. [PMID: 26961956 DOI: 10.1523/jneurosci.2250-15.2016] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The regulation of oligodendrocyte development and myelin formation in the CNS is poorly defined. Multiple signals influence the rate and extent of CNS myelination, including the noncanonical cyclin-dependent kinase 5 (Cdk5) whose functions are regulated by its activators p35 and p39. Here we show that selective loss of either p35 or p39 perturbed specific aspects of oligodendrocyte development, whereas loss of both p35 and p39 completely inhibited the development of mature oligodendrocytes and myelination. In the absence of p35, oligodendrocyte differentiation was delayed, process outgrowth was truncated in vitro, and the patterning and extent of myelination were perturbed in the CNS of p35(-/-) mice. In the absence of p39, oligodendrocyte maturation was transiently affected both in vitro and in vivo. However, loss of both p35 and p39 in oligodendrocyte lineage cells completely inhibited oligodendrocyte progenitor cell differentiation and myelination both in vitro and after transplantation into shiverer slice cultures. Loss of p35 and p39 had a more profound effect on oligodendrocyte development than simply the loss of Cdk5 and could not be rescued by Cdk5 overexpression. These data suggest p35 and p39 have specific and overlapping roles in oligodendrocyte development, some of which may be independent of Cdk5 activation.
Collapse
|
44
|
Regulation of PERK-eIF2α signalling by tuberous sclerosis complex-1 controls homoeostasis and survival of myelinating oligodendrocytes. Nat Commun 2016; 7:12185. [PMID: 27416896 PMCID: PMC4947172 DOI: 10.1038/ncomms12185] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2015] [Accepted: 06/08/2016] [Indexed: 12/20/2022] Open
Abstract
Tuberous sclerosis complex-1 or 2 (TSC1/2) mutations cause white matter abnormalities, including myelin deficits in the CNS; however, underlying mechanisms are not fully understood. TSC1/2 negatively regulate the function of mTOR, which is required for oligodendrocyte differentiation. Here we report that, unexpectedly, constitutive activation of mTOR signalling by Tsc1 deletion in the oligodendrocyte lineage results in severe myelination defects and oligodendrocyte cell death in mice, despite an initial increase of oligodendrocyte precursors during early development. Expression profiling analysis reveals that Tsc1 ablation induces prominent endoplasmic reticulum (ER) stress responses by activating a PERK–eIF2α signalling axis and Fas–JNK apoptotic pathways. Enhancement of the phospho-eIF2α adaptation pathway by inhibition of Gadd34-PP1 phosphatase with guanabenz protects oligodendrocytes and partially rescues myelination defects in Tsc1 mutants. Thus, TSC1-mTOR signalling acts as an important checkpoint for maintaining oligodendrocyte homoeostasis, pointing to a previously uncharacterized ER stress mechanism that contributes to hypomyelination in tuberous sclerosis. The molecular mechanisms regulating myelination are only partially understood. Here authors show that Tsc1 ablation in oligodendrocyte lineage activates ER stress and apoptotic programs in mice, and that enhancing PERK-eIF2α signalling partially rescues the myelination defects in Tsc1 mutants.
Collapse
|
45
|
Goldstein EZ, Church JS, Hesp ZC, Popovich PG, McTigue DM. A silver lining of neuroinflammation: Beneficial effects on myelination. Exp Neurol 2016; 283:550-9. [PMID: 27151600 DOI: 10.1016/j.expneurol.2016.05.001] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 04/27/2016] [Accepted: 05/01/2016] [Indexed: 12/19/2022]
Abstract
Myelin accelerates action potential conduction velocity and provides essential energy support for axons. Unfortunately, myelin and myelinating cells are often vulnerable to injury or disease, resulting in myelin damage, which in turn can lead to axon dysfunction, overt pathology and neurological impairment. Inflammation is a common component of trauma and disease in both the CNS and PNS and therefore an active inflammatory response is often considered deleterious to myelin health. While inflammation can certainly damage myelin, inflammatory processes also can positively affect oligodendrocyte lineage progression, myelin debris clearance, oligodendrocyte metabolism and myelin repair. In the periphery, inflammatory cascades can also augment myelin repair, including processes initiated by infiltrating immune cells as well as by local Schwann cells. In this review, various aspects of inflammation beneficial to myelin repair are discussed and should be considered when designing or implementing anti-inflammatory therapies for CNS and PNS injury involving myelinating cells.
Collapse
Affiliation(s)
- Evan Z Goldstein
- Neuroscience Graduate Program, Wexner Medical Center, The Ohio State University, United States; Center for Brain and Spinal Cord Repair, Wexner Medical Center, The Ohio State University, United States
| | - Jamie S Church
- Neuroscience Graduate Program, Wexner Medical Center, The Ohio State University, United States; Center for Brain and Spinal Cord Repair, Wexner Medical Center, The Ohio State University, United States
| | - Zoe C Hesp
- Neuroscience Graduate Program, Wexner Medical Center, The Ohio State University, United States; Center for Brain and Spinal Cord Repair, Wexner Medical Center, The Ohio State University, United States
| | - Phillip G Popovich
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, United States; Center for Brain and Spinal Cord Repair, Wexner Medical Center, The Ohio State University, United States
| | - Dana M McTigue
- Department of Neuroscience, Wexner Medical Center, The Ohio State University, United States; Center for Brain and Spinal Cord Repair, Wexner Medical Center, The Ohio State University, United States.
| |
Collapse
|
46
|
Zhou S, Ding F, Gu X. Non-coding RNAs as Emerging Regulators of Neural Injury Responses and Regeneration. Neurosci Bull 2016; 32:253-64. [PMID: 27037691 DOI: 10.1007/s12264-016-0028-7] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 03/14/2016] [Indexed: 02/06/2023] Open
Abstract
Non-coding RNAs (ncRNAs) are a large cluster of RNAs that do not encode proteins, but have multiple functions in diverse cellular processes. Mounting evidence indicates the involvement of ncRNAs in the physiology and pathophysiology of the central and peripheral nervous systems. It has been shown that numerous ncRNAs, especially microRNAs and long non-coding RNAs, are differentially expressed after insults such as acquired brain injury, spinal cord injury, and peripheral nerve injury. These ncRNAs affect neuronal survival, neurite regrowth, and glial phenotype primarily by targeting specific mRNAs, resulting in translation repression or degradation of the mRNAs. An increasing number of studies have investigated the regulatory roles of microRNAs and long non-coding RNAs in neural injury and regeneration, and thus a new research field is emerging. In this review, we highlight current progress in the field in an attempt to provide further insight into post-transcriptional changes occurring after neural injury, and to facilitate the potential use of ncRNAs for improving neural regeneration. We also suggest potential directions for future studies.
Collapse
Affiliation(s)
- Songlin Zhou
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Fei Ding
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China
| | - Xiaosong Gu
- Jiangsu Key Laboratory of Neuroregeneration, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001, China.
| |
Collapse
|
47
|
Dual regulatory switch through interactions of Tcf7l2/Tcf4 with stage-specific partners propels oligodendroglial maturation. Nat Commun 2016; 7:10883. [PMID: 26955760 PMCID: PMC4786870 DOI: 10.1038/ncomms10883] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 01/28/2016] [Indexed: 01/04/2023] Open
Abstract
Constitutive activation of Wnt/β-catenin inhibits oligodendrocyte myelination. Tcf7l2/Tcf4, a β-catenin transcriptional partner, is required for oligodendrocyte differentiation. How Tcf7l2 modifies β-catenin signalling and controls myelination remains elusive. Here we define a stage-specific Tcf7l2-regulated transcriptional circuitry in initiating and sustaining oligodendrocyte differentiation. Multistage genome occupancy analyses reveal that Tcf7l2 serially cooperates with distinct co-regulators to control oligodendrocyte lineage progression. At the differentiation onset, Tcf7l2 interacts with a transcriptional co-repressor Kaiso/Zbtb33 to block β-catenin signalling. During oligodendrocyte maturation, Tcf7l2 recruits and cooperates with Sox10 to promote myelination. In that context, Tcf7l2 directly activates cholesterol biosynthesis genes and cholesterol supplementation partially rescues oligodendrocyte differentiation defects in Tcf712 mutants. Together, we identify stage-specific co-regulators Kaiso and Sox10 that sequentially interact with Tcf7l2 to coordinate the switch at the transitions of differentiation initiation and maturation during oligodendrocyte development, and point to a previously unrecognized role of Tcf7l2 in control of cholesterol biosynthesis for CNS myelinogenesis.
Collapse
|
48
|
Abstract
In the nervous system, axons transmit information in the form of electrical impulses over long distances. The speed of impulse conduction is enhanced by myelin, a lipid-rich membrane that wraps around axons. Myelin also is required for the long-term health of axons by providing metabolic support. Accordingly, myelin deficiencies are implicated in a wide range of neurodevelopmental and neuropsychiatric disorders, intellectual disabilities, and neurodegenerative conditions. Central nervous system myelin is formed by glial cells called oligodendrocytes. During development, oligodendrocyte precursor cells migrate from their origins to their target axons, extend long membrane processes that wrap axons, and produce the proteins and lipids that provide myelin membrane with its unique characteristics. Myelination is a dynamic process that involves intricate interactions between multiple cell types. Therefore, an in vivo myelination model, such as the zebrafish, which allows for live observation of cell dynamics and cell-to-cell interactions, is well suited for investigating oligodendrocyte development. Zebrafish offer several advantages to investigating myelination, including the use of transgenic reporter lines, live imaging, forward genetic screens, chemical screens, and reverse genetic approaches. This chapter will describe how these tools and approaches have provided new insights into the regulatory mechanisms that guide myelination.
Collapse
Affiliation(s)
- E S Mathews
- University of Colorado School of Medicine, Aurora, CO, United States
| | - B Appel
- University of Colorado School of Medicine, Aurora, CO, United States
| |
Collapse
|
49
|
Dionne N, Dib S, Finsen B, Denarier E, Kuhlmann T, Drouin R, Kokoeva M, Hudson TJ, Siminovitch K, Friedman HC, Peterson AC. Functional organization of anMbpenhancer exposes striking transcriptional regulatory diversity within myelinating glia. Glia 2015; 64:175-94. [DOI: 10.1002/glia.22923] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/04/2015] [Accepted: 09/09/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Nancy Dionne
- Laboratory of Developmental Biology; Ludmer Research and Training Building, McGill University; Montreal Quebec Canada
| | - Samar Dib
- Laboratory of Developmental Biology; Ludmer Research and Training Building, McGill University; Montreal Quebec Canada
| | - Bente Finsen
- Department of Neurobiology Research; Institute of Molecular Medicine, University of Southern Denmark; Odense Denmark
| | - Eric Denarier
- Institut National De La Santé Et De La Recherche Médicale, U836-GIN iRTSV-GPC; Site Santé La Tronche, BP170 Grenoble Cedex 9 France
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital, Münster; Pottkamp 2 Münster Germany
| | - Régen Drouin
- Division of Genetics, Department of Pediatrics, Faculty of Medicine and Health Sciences; Université De Sherbrooke; Sherbrooke Quebec Canada
| | - Maia Kokoeva
- Department of Medicine; McGill University/MUHC Research Institute; Montreal Quebec Canada
| | - Thomas J. Hudson
- Ontario Institute for Cancer Research, MaRS Centre; South Tower Toronto Ontario Canada
| | - Kathy Siminovitch
- Department of Medicine; University of Toronto, Samuel Lunenfeld and Toronto General Research Institutes; Toronto Ontario Canada
- Department of Immunology and Molecular Genetics; University of Toronto; Toronto Ontario Canada
| | - Hana C Friedman
- Laboratory of Developmental Biology; Ludmer Research and Training Building, McGill University; Montreal Quebec Canada
| | - Alan C. Peterson
- Laboratory of Developmental Biology; Ludmer Research and Training Building, McGill University; Montreal Quebec Canada
| |
Collapse
|
50
|
Involvement of MeCP2 in Regulation of Myelin-Related Gene Expression in Cultured Rat Oligodendrocytes. J Mol Neurosci 2015; 57:176-84. [PMID: 26140854 DOI: 10.1007/s12031-015-0597-3] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 06/02/2015] [Indexed: 01/08/2023]
Abstract
Methyl CpG binding protein 2 (MeCP2) is a multifunctional protein which binds to methylated CpG, mutation of which cause a neurodevelopmental disorder, Rett syndrome. MeCP2 can function as both transcriptional activator and repressor of target gene. MeCP2 regulate gene expression in both neuron and glial cells in central nervous system (CNS). Oligodendrocytes, the myelinating cells of CNS, are required for normal functioning of neurons and are regulated by several transcription factors during their differentiation. In current study, we focused on the role of MeCP2 as transcription regulator of myelin genes in cultured rat oligodendrocytes. We have observed expression of MeCP2 at all stages of oligodendrocyte development. MeCP2 knockdown in cultured oligodendrocytes by small interference RNA (siRNA) has shown increase in myelin genes (myelin basic protein (MBP), proteolipid protein (PLP), myelin oligodendrocyte glycoprotein (MOG), and myelin-associated oligodendrocyte basic protein (MOBP)), neurotrophin (brain-derived neurotrophic factor (BDNF)), and transcriptional regulator (YY1) transcripts level, which are involved in regulation of oligodendrocyte differentiation and myelination. Further, we also found that protein levels of MBP, PLP, DM-20, and BDNF also significantly upregulated in MeCP2 knockdown oligodendrocytes. Our study suggests that the MeCP2 acts as a negative regulator of myelin protein expression.
Collapse
|