1
|
Rusu B, Kukreja B, Wu T, Dan SJ, Feng MY, Kalish BT. Single-Nucleus Profiling Identifies Accelerated Oligodendrocyte Precursor Cell Senescence in a Mouse Model of Down Syndrome. eNeuro 2023; 10:ENEURO.0147-23.2023. [PMID: 37491366 PMCID: PMC10449487 DOI: 10.1523/eneuro.0147-23.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/06/2023] [Accepted: 07/10/2023] [Indexed: 07/27/2023] Open
Abstract
Down syndrome (DS), the most common genetic cause of intellectual disability, is associated with lifelong cognitive deficits. However, the mechanisms by which triplication of chromosome 21 genes drive neuroinflammation and cognitive dysfunction are poorly understood. Here, using the Ts65Dn mouse model of DS, we performed an integrated single-nucleus ATAC and RNA-sequencing (snATAC-seq and snRNA-seq) analysis of the adult cortex. We identified cell type-specific transcriptional and chromatin-associated changes in the Ts65Dn cortex, including regulators of neuroinflammation, transcription and translation, myelination, and mitochondrial function. We discovered enrichment of a senescence-associated transcriptional signature in Ts65Dn oligodendrocyte (OL) precursor cells (OPCs) and epigenetic changes consistent with a loss of heterochromatin. We found that senescence is restricted to a subset of OPCs concentrated in deep cortical layers. Treatment of Ts65Dn mice with a senescence-reducing flavonoid rescued cortical OPC proliferation, restored microglial homeostasis, and improved contextual fear memory. Together, these findings suggest that cortical OPC senescence may be an important driver of neuropathology in DS.
Collapse
Affiliation(s)
- Bianca Rusu
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
| | - Bharti Kukreja
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
| | - Taiyi Wu
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
| | - Sophie J Dan
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Department of Physiology, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario M5G 1A8, Canada
| | - Min Yi Feng
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
| | - Brian T Kalish
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario M5G 1A8, Canada
- Program in Neuroscience and Mental Health, SickKids Research Institute, Toronto, Ontario M5G 1L7, Canada
- Division of Neonatology, Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario M5G 1L7, Canada
| |
Collapse
|
2
|
The Oligodendrocyte Transcription Factor 2 OLIG2 regulates transcriptional repression during myelinogenesis in rodents. Nat Commun 2022; 13:1423. [PMID: 35301318 PMCID: PMC8931116 DOI: 10.1038/s41467-022-29068-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 02/25/2022] [Indexed: 12/13/2022] Open
Abstract
OLIG2 is a transcription factor that activates the expression of myelin-associated genes in the oligodendrocyte-lineage cells. However, the mechanisms of myelin gene inactivation are unclear. Here, we uncover a non-canonical function of OLIG2 in transcriptional repression to modulate myelinogenesis by functionally interacting with tri-methyltransferase SETDB1. Immunoprecipitation and chromatin-immunoprecipitation assays show that OLIG2 recruits SETDB1 for H3K9me3 modification on the Sox11 gene, which leads to the inhibition of Sox11 expression during the differentiation of oligodendrocytes progenitor cells (OPCs) into immature oligodendrocytes (iOLs). Tissue-specific depletion of Setdb1 in mice results in the hypomyelination during development and remyelination defects in the injured rodents. Knockdown of Sox11 by siRNA in rat primary OPCs or depletion of Sox11 in the oligodendrocyte lineage in mice could rescue the hypomyelination phenotype caused by the loss of OLIG2. In summary, our work demonstrates that the OLIG2-SETDB1 complex can mediate transcriptional repression in OPCs, affecting myelination. Transcription factors regulate gene programs during myelination. Here, the authors show that the Oligodendrocyte Transcription Factor 2 (OLIG2) regulates the differentiation of oligodendrocyte progenitor cells into immature oligodendrocytes via SETDB1 during myelination and remyelination in rodents.
Collapse
|
3
|
Wang P, Ma K, Yang L, Zhang G, Ye M, Wang S, Wei S, Chen Z, Gu J, Zhang L, Niu J, Tao S. Predicting signaling pathways regulating demyelination in a rat model of lithium-pilocarpine-induced acute epilepsy: A proteomics study. Int J Biol Macromol 2021; 193:1457-1470. [PMID: 34742844 DOI: 10.1016/j.ijbiomac.2021.10.209] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 10/26/2021] [Accepted: 10/27/2021] [Indexed: 11/18/2022]
Abstract
Demyelination is observed in animal models of intractable epilepsy (IE). Epileptogenesis damages the myelin sheath and dysregulates oligodendrocyte precursor cell (OPC) development. However, the molecular pathways regulating demyelination in epilepsy are unclear. Here, we predicted the molecular mechanisms regulating demyelination in a rat model of lithium-pilocarpine hydrochloride-induced epilepsy. We identified DGKA/Mboat2/Inpp5j and NOS/Keratin 28 as the main target molecules that regulate demyelination via glycerolipid and glycerophospholipid metabolism, phosphatidylinositol signaling, and estrogen signaling in demyelinated forebrain slice cultures (FSCs). In seizure-like FCSs, the actin cytoskeleton was regulated by Cnp and MBP via Pak4/Tmsb4x (also known as Tβ4) and Kif5c/Kntc1. Tβ4 possibly prevented OPC differentiation and maturation and inhibited MBP phosphorylation via the p38MAPK/ERK1/JNK1 pathway. The MAPK signaling pathway was more likely activated in seizure-like FCSs than in demyelinated FCSs. pMBP expression was decreased in the hippocampus of lithium-pilocarpine hydrochloride-induced acute epilepsy rats. The expression of remyelination-related factors was suppressed in the hippocampus and corpus callosum in lithium-pilocarpine hydrochloride-induced epilepsy rats. These findings suggest that the actin cytoskeleton, Tβ4, and MAPK signaling pathways regulate the decrease in pMBP in the hippocampus in a rat model of epilepsy. Our results indicate that regulating the actin cytoskeleton, Tβ4, and MAPK signaling pathways may facilitate the prevention of demyelination in IE.
Collapse
Affiliation(s)
- Peng Wang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan 750004, China.
| | - Kang Ma
- Department of Anatomy, Ningxia Medical University, Yinchuan 750004, China
| | - Lu Yang
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan 750004, China
| | - Guodong Zhang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, China
| | - Mengyi Ye
- Ningxia Medical University College of Traditional Chinese Medicine, Yinchuan 750004, Ningxia, China
| | - Siqi Wang
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, China
| | - Shuangshuang Wei
- School of Clinical Medicine, Ningxia Medical University, Yinchuan 750004, China
| | - Zhangping Chen
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan 750004, China
| | - Jinghai Gu
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan 750004, China
| | - Lianxiang Zhang
- Department of Anatomy, Ningxia Medical University, Yinchuan 750004, China
| | - Jianguo Niu
- Department of Anatomy, Ningxia Medical University, Yinchuan 750004, China.
| | - Sun Tao
- Ningxia Key Laboratory of Cerebrocranial Diseases, Ningxia Medical University, Yinchuan 750004, China; Department of Neurosurgery, General Hospital of Ningxia Medical University, 804 Shengli Street, Yinchuan 750004, Ningxia, China.
| |
Collapse
|
4
|
Li S, Guan H, Zhang Y, Li S, Li K, Hu S, Zuo E, Zhang C, Zhang X, Gong G, Wang R, Piao F. Bone marrow mesenchymal stem cells promote remyelination in spinal cord by driving oligodendrocyte progenitor cell differentiation via TNFα/RelB-Hes1 pathway: a rat model study of 2,5-hexanedione-induced neurotoxicity. Stem Cell Res Ther 2021; 12:436. [PMID: 34348774 PMCID: PMC8336089 DOI: 10.1186/s13287-021-02518-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 07/18/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND N-hexane, with its metabolite 2,5-hexanedine (HD), is an industrial hazardous material. Chronic hexane exposure causes segmental demyelination in the peripheral nerves, and high-dose intoxication may also affect central nervous system. Demyelinating conditions are difficult to treat and stem cell therapy using bone marrow mesenchymal stem cells (BMSCs) is a promising novel strategy. Our previous study found that BMSCs promoted motor function recovery in rats modeling hexane neurotoxicity. This work aimed to explore the underlying mechanisms and focused on the changes in spinal cord. METHODS Sprague Dawley rats were intoxicated with HD (400 mg/kg/day, i.p, for 5 weeks). A bolus of BMSCs (5 × 107 cells/kg) was injected via tail vein. Demyelination and remyelination of the spinal cord before and after BMSC treatment were examined microscopically. Cultured oligodendrocyte progenitor cells (OPCs) were incubated with HD ± BMSC-derived conditional medium (BMSC-CM). OPC differentiation was studied by immunostaining and morphometric analysis. The expressional changes of Hes1, a transcription factor negatively regulating OPC-differentiation, were studied. The upstream Notch1 and TNFα/RelB pathways were studied, and some key signaling molecules were measured. The correlation between neurotrophin NGF and TNFα was also investigated. Statistical significance was evaluated using one-way ANOVA and performed using SPSS 13.0. RESULTS The demyelinating damage by HD and remyelination by BMSCs were evidenced by electron microscopy, LFB staining and NG2/MBP immunohistochemistry. In vitro cultured OPCs showed more differentiation after incubation with BMSC-CM. Hes1 expression was found to be significantly increased by HD and decreased by BMSC or BMSC-CM. The change of Hes1 was found, however, independent of Notch1 activation, but dependent on TNFα/RelB signaling. HD was found to increase TNFα, RelB and Hes1 expression, and BMSCs were found to have the opposite effect. Addition of recombinant TNFα to OPCs or RelB overexpression similarly caused upregulation of Hes1 expression. The secretion of NGF by BMSC and activation of NGF receptor was found important for suppression of TNFα production in OPCs. CONCLUSIONS Our findings demonstrated that BMSCs promote remyelination in the spinal cord of HD-exposed rats via TNFα/RelB-Hes1 pathway, providing novel insights for evaluating and further exploring the therapeutical effect of BMSCs on demyelinating neurodegenerative disease.
Collapse
Affiliation(s)
- Shuangyue Li
- School of Public Health, Dalian Medical University, Dalian, Liaoning, 116044, People's Republic of China
| | - Huai Guan
- Department of Obstetrics and Gynecology, No. 967 Hospital of the Joint Logistics Support Force of the Chinese PLA, Dalian, People's Republic of China
| | - Yan Zhang
- School of Public Health, Dalian Medical University, Dalian, Liaoning, 116044, People's Republic of China.,Xunyi Center for Disease Control and Prevention, Xunyi, Shanxi, 711300, People's Republic of China
| | - Sheng Li
- Department of Biochemistry, Dalian Medical University, Dalian, Liaoning, 116044, People's Republic of China
| | - Kaixin Li
- School of Public Health, Dalian Medical University, Dalian, Liaoning, 116044, People's Republic of China.,Xian Center for Disease Control and Prevention, Xian, 710054, People's Republic of China
| | - Shuhai Hu
- College of Stomatology, Dalian Medical University, Dalian, Liaoning, 116044, People's Republic of China
| | - Enjun Zuo
- College of Stomatology, Dalian Medical University, Dalian, Liaoning, 116044, People's Republic of China
| | - Cong Zhang
- School of Public Health, Dalian Medical University, Dalian, Liaoning, 116044, People's Republic of China
| | - Xin Zhang
- Department of Clinical Nutrition, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People's Republic of China
| | - Guanyu Gong
- Integrative Laboratory, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China. .,Department of Medical Oncology, Affiliated Zhongshan Hospital of Dalian University, Dalian, People's Republic of China.
| | - Ruoyu Wang
- Integrative Laboratory, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China.
| | - Fengyuan Piao
- Integrative Laboratory, Affiliated Zhongshan Hospital of Dalian University, Dalian, 116001, People's Republic of China.
| |
Collapse
|
5
|
Fredrickx E, Colombo E, Canevazzi P, La Marca R, Pellegatta M, Dina G, Podini P, Nave KA, Quattrini A, Taveggia C. Ablation of neuronal ADAM17 impairs oligodendrocyte differentiation and myelination. Glia 2019; 68:1148-1164. [PMID: 31851405 DOI: 10.1002/glia.23765] [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] [Received: 02/08/2018] [Revised: 11/26/2019] [Accepted: 12/03/2019] [Indexed: 11/09/2022]
Abstract
Myelin, one of the most important adaptations of vertebrates, is essential to ensure efficient propagation of the electric impulse in the nervous system and to maintain neuronal integrity. In the central nervous system (CNS), the development of oligodendrocytes and the process of myelination are regulated by the coordinated action of several positive and negative cell-extrinsic factors. We and others previously showed that secretases regulate the activity of proteins essential for myelination. We now report that the neuronal α-secretase ADAM17 controls oligodendrocyte differentiation and myelin formation in the CNS. Ablation of Adam17 in neurons impairs in vivo and in vitro oligodendrocyte differentiation, delays myelin formation throughout development and results in hypomyelination. Furthermore, we show that this developmental defect is, in part, the result of altered Notch/Jagged 1 signaling. Surprisingly, in vivo conditional loss of Adam17 in immature oligodendrocytes has no effect on myelin formation. Collectively, our data indicate that the neuronal α-secretase ADAM17 is required for proper CNS myelination. Further, our studies confirm that secretases are important post-translational regulators of myelination although the mechanisms controlling CNS and peripheral nervous system (PNS) myelination are distinct.
Collapse
Affiliation(s)
- Evelien Fredrickx
- INSPE, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Colombo
- INSPE, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paolo Canevazzi
- INSPE, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Rosa La Marca
- INSPE, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marta Pellegatta
- INSPE, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giorgia Dina
- INSPE, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Paola Podini
- INSPE, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Klaus A Nave
- Department of Neurogenetics, Max Planck Institute of Experimental Medicine, Göttingen, Germany
| | - Angelo Quattrini
- INSPE, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Carla Taveggia
- INSPE, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
6
|
Falcão AM, van Bruggen D, Marques S, Meijer M, Jäkel S, Agirre E, Samudyata, Floriddia EM, Vanichkina DP, Ffrench-Constant C, Williams A, Guerreiro-Cacais AO, Castelo-Branco G. Disease-specific oligodendrocyte lineage cells arise in multiple sclerosis. Nat Med 2018; 24:1837-1844. [PMID: 30420755 PMCID: PMC6544508 DOI: 10.1038/s41591-018-0236-y] [Citation(s) in RCA: 368] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2018] [Accepted: 09/14/2018] [Indexed: 11/22/2022]
Abstract
Multiple sclerosis (MS) is characterized by an immune system attack targeting myelin, which is produced by oligodendrocytes (OLs). We performed single-cell transcriptomic analysis of OL lineage cells from the spinal cord of mice induced with experimental autoimmune encephalomyelitis (EAE), which mimics several aspects of MS. We found unique OLs and OL precursor cells (OPCs) in EAE and uncovered several genes specifically alternatively spliced in these cells. Surprisingly, EAE-specific OL lineage populations expressed genes involved in antigen processing and presentation via major histocompatibility complex class I and II (MHC-I and -II), and in immunoprotection, suggesting alternative functions of these cells in a disease context. Importantly, we found that disease-specific oligodendroglia are also present in human MS brains and that a substantial number of genes known to be susceptibility genes for MS, so far mainly associated with immune cells, are expressed in the OL lineage cells. Finally, we demonstrate that OPCs can phagocytose and that MHC-II-expressing OPCs can activate memory and effector CD4-positive T cells. Our results suggest that OLs and OPCs are not passive targets but instead active immunomodulators in MS. The disease-specific OL lineage cells, for which we identify several biomarkers, may represent novel direct targets for immunomodulatory therapeutic approaches in MS.
Collapse
Affiliation(s)
- Ana Mendanha Falcão
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, Stockholm, Sweden.
| | - David van Bruggen
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - Sueli Marques
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - Mandy Meijer
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - Sarah Jäkel
- MRC Centre for Regenerative Medicine and MS Society Edinburgh Centre, Edinburgh bioQuarter, University of Edinburgh, Edinburgh, UK
| | - Eneritz Agirre
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - Samudyata
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - Elisa M Floriddia
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, Stockholm, Sweden
| | - Darya P Vanichkina
- Gene and Stem Cell Therapy Program, Centenary Institute, University of Sydney, Camperdown, New South Wales , Australia
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Queensland, Australia
| | - Charles Ffrench-Constant
- MRC Centre for Regenerative Medicine and MS Society Edinburgh Centre, Edinburgh bioQuarter, University of Edinburgh, Edinburgh, UK
| | - Anna Williams
- MRC Centre for Regenerative Medicine and MS Society Edinburgh Centre, Edinburgh bioQuarter, University of Edinburgh, Edinburgh, UK
| | | | - Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Biomedicum, Stockholm, Sweden.
- Ming Wai Lau Centre for Reparative Medicine, Stockholm node, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
7
|
Santos AK, Vieira MS, Vasconcellos R, Goulart VAM, Kihara AH, Resende RR. Decoding cell signalling and regulation of oligodendrocyte differentiation. Semin Cell Dev Biol 2018; 95:54-73. [PMID: 29782926 DOI: 10.1016/j.semcdb.2018.05.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/15/2018] [Accepted: 05/17/2018] [Indexed: 12/20/2022]
Abstract
Oligodendrocytes are fundamental for the functioning of the nervous system; they participate in several cellular processes, including axonal myelination and metabolic maintenance for astrocytes and neurons. In the mammalian nervous system, they are produced through waves of proliferation and differentiation, which occur during embryogenesis. However, oligodendrocytes and their precursors continue to be generated during adulthood from specific niches of stem cells that were not recruited during development. Deficiencies in the formation and maturation of these cells can generate pathologies mainly related to myelination. Understanding the mechanisms involved in oligodendrocyte development, from the precursor to mature cell level, will allow inferring therapies and treatments for associated pathologies and disorders. Such mechanisms include cell signalling pathways that involve many growth factors, small metabolic molecules, non-coding RNAs, and transcription factors, as well as specific elements of the extracellular matrix, which act in a coordinated temporal and spatial manner according to a given stimulus. Deciphering those aspects will allow researchers to replicate them in vitro in a controlled environment and thus mimic oligodendrocyte maturation to understand the role of oligodendrocytes in myelination in pathologies and normal conditions. In this study, we review these aspects, based on the most recent in vivo and in vitro data on oligodendrocyte generation and differentiation.
Collapse
Affiliation(s)
- A K Santos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - M S Vieira
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - R Vasconcellos
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil
| | - V A M Goulart
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil
| | - A H Kihara
- Centro de Matemática, Computação e Cognição, Universidade Federal do ABC, São Bernardo do Campo, SP, Brazil
| | - R R Resende
- Departamento de Bioquímica e Imunologia, Instituto de Ciência Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, 31270-901 Belo Horizonte, MG, Brazil; Instituto Nanocell, Rua Santo Antônio, 420, 35500-041 Divinópolis, MG, Brazil.
| |
Collapse
|
8
|
Lentferink DH, Jongsma JM, Werkman I, Baron W. Grey matter OPCs are less mature and less sensitive to IFNγ than white matter OPCs: consequences for remyelination. Sci Rep 2018; 8:2113. [PMID: 29391408 PMCID: PMC5794790 DOI: 10.1038/s41598-018-19934-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 01/05/2018] [Indexed: 12/11/2022] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease characterized by the formation of demyelinated lesions in the central nervous system. At later stages of the disease repair in the form of remyelination often fails, which leads to axonal degeneration and neurological disability. For the regeneration of myelin, oligodendrocyte progenitor cells (OPCs) have to migrate, proliferate and differentiate into remyelinating oligodendrocytes. Remyelination occurs faster and is more extensive in grey matter (GM) lesions than in white matter (WM) lesions. Here, we examined differences in neonatal OPCs from GM (gmOPCs) and WM (wmOPCs), both intrinsically and in response to environmental (injury) signals. We show that gmOPCs are less mature than wmOPCs, both on morphological and on gene-expression level. Additionally, gmOPCs proliferate more and differentiate slower than wmOPCs. When exposed to astrocyte-secreted signals wmOPC, but not gmOPC, migration decreases. In addition, wmOPCs are more sensitive to the detrimental effects of IFNγ treatment on proliferation, differentiation, and process arborisation, which is potentiated by TNFα. Our results demonstrate that OPCs from GM and WM differ both intrinsically and in response to their environment, which may contribute to the difference in remyelination efficiency between GM and WM MS lesions.
Collapse
Affiliation(s)
- Dennis H Lentferink
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Jacomien M Jongsma
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Inge Werkman
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Wia Baron
- Department of Cell Biology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
9
|
Ichihara Y, Doi T, Ryu Y, Nagao M, Sawada Y, Ogata T. Oligodendrocyte Progenitor Cells Directly Utilize Lactate for Promoting Cell Cycling and Differentiation. J Cell Physiol 2016; 232:986-995. [PMID: 27861886 PMCID: PMC5299506 DOI: 10.1002/jcp.25690] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 11/11/2016] [Indexed: 12/17/2022]
Abstract
Oligodendrocyte progenitor cells (OPCs) undergo marked morphological changes to become mature oligodendrocytes, but the metabolic resources for this process have not been fully elucidated. Although lactate, a metabolic derivative of glycogen, has been reported to be consumed in oligodendrocytes as a metabolite, and to ameliorate hypomyelination induced by low glucose conditions, it is not clear about the direct contribution of lactate to cell cycling and differentiation of OPCs, and the source of lactate for remyelination. Therefore, we evaluated the effect of 1,4‐dideoxy‐1,4‐imino‐d‐arabinitol (DAB), an inhibitor of the glycogen catabolic enzyme glycogen phosphorylase, in a mouse cuprizone model. Cuprizone induced demyelination in the corpus callosum and remyelination occurred after cuprizone treatment ceased. This remyelination was inhibited by the administration of DAB. To further examine whether lactate affects proliferation or differentiation of OPCs, we cultured mouse primary OPC‐rich cells and analyzed the effect of lactate. Lactate rescued the slowed cell cycling induced by 0.4 mM glucose, as assessed by the BrdU‐positive cell ratio. Lactate also promoted OPC differentiation detected by monitoring the mature oligodendrocyte marker myelin basic protein, in the presence of both 36.6 mM and 0.4 mM glucose. Furthermore, these lactate‐mediated effects were suppressed by the reported monocarboxylate transporter inhibitor, α‐cyano‐4‐hydroxy‐cinnamate. These results suggest that lactate directly promotes the cell cycling rate and differentiation of OPCs, and that glycogen, one of the sources of lactate, contributes to remyelination in vivo. J. Cell. Physiol. 232: 986–995, 2017. © 2016 The Authors. Journal of Cellular Physiology Published by Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yoshinori Ichihara
- Department of Rehabilitation for Movement Functions, National Rehabilitation Center for Persons With Disabilities, Saitama, Japan
| | - Toru Doi
- Department of Rehabilitation for Movement Functions, National Rehabilitation Center for Persons With Disabilities, Saitama, Japan
| | - Youngjae Ryu
- Department of Rehabilitation for Movement Functions, National Rehabilitation Center for Persons With Disabilities, Saitama, Japan
| | - Motoshi Nagao
- Department of Rehabilitation for Movement Functions, National Rehabilitation Center for Persons With Disabilities, Saitama, Japan
| | - Yasuhiro Sawada
- Department of Rehabilitation for Movement Functions, National Rehabilitation Center for Persons With Disabilities, Saitama, Japan
| | - Toru Ogata
- Department of Rehabilitation for Movement Functions, National Rehabilitation Center for Persons With Disabilities, Saitama, Japan
| |
Collapse
|
10
|
Gopinath C, Law WD, Rodríguez-Molina JF, Prasad AB, Song L, Crawford GE, Mullikin JC, Svaren J, Antonellis A. Stringent comparative sequence analysis reveals SOX10 as a putative inhibitor of glial cell differentiation. BMC Genomics 2016; 17:887. [PMID: 27821050 PMCID: PMC5100263 DOI: 10.1186/s12864-016-3167-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2016] [Accepted: 10/18/2016] [Indexed: 01/22/2023] Open
Abstract
Background The transcription factor SOX10 is essential for all stages of Schwann cell development including myelination. SOX10 cooperates with other transcription factors to activate the expression of key myelin genes in Schwann cells and is therefore a context-dependent, pro-myelination transcription factor. As such, the identification of genes regulated by SOX10 will provide insight into Schwann cell biology and related diseases. While genome-wide studies have successfully revealed SOX10 target genes, these efforts mainly focused on myelinating stages of Schwann cell development. We propose that less-biased approaches will reveal novel functions of SOX10 outside of myelination. Results We developed a stringent, computational-based screen for genome-wide identification of SOX10 response elements. Experimental validation of a pilot set of predicted binding sites in multiple systems revealed that SOX10 directly regulates a previously unreported alternative promoter at SOX6, which encodes a transcription factor that inhibits glial cell differentiation. We further explored the utility of our computational approach by combining it with DNase-seq analysis in cultured Schwann cells and previously published SOX10 ChIP-seq data from rat sciatic nerve. Remarkably, this analysis enriched for genomic segments that map to loci involved in the negative regulation of gliogenesis including SOX5, SOX6, NOTCH1, HMGA2, HES1, MYCN, ID4, and ID2. Functional studies in Schwann cells revealed that: (1) all eight loci are expressed prior to myelination and down-regulated subsequent to myelination; (2) seven of the eight loci harbor validated SOX10 binding sites; and (3) seven of the eight loci are down-regulated upon repressing SOX10 function. Conclusions Our computational strategy revealed a putative novel function for SOX10 in Schwann cells, which suggests a model where SOX10 activates the expression of genes that inhibit myelination during non-myelinating stages of Schwann cell development. Importantly, the computational and functional datasets we present here will be valuable for the study of transcriptional regulation, SOX protein function, and glial cell biology. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-3167-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chetna Gopinath
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - William D Law
- Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - José F Rodríguez-Molina
- Cellular and Molecular Pathology Program, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Arjun B Prasad
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lingyun Song
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA
| | - Gregory E Crawford
- Center for Genomic and Computational Biology, Duke University Medical Center, Durham, NC, 27708, USA.,Department of Pediatrics, Duke University Medical Center, Durham, NC, 27708, USA
| | - James C Mullikin
- Genome Technology Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - John Svaren
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53706, USA.,Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | - Anthony Antonellis
- Cellular and Molecular Biology Program, University of Michigan Medical School, Ann Arbor, MI, 48109, USA. .,Department of Human Genetics, University of Michigan Medical School, Ann Arbor, MI, 48109, USA. .,Department of Neurology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
11
|
Dhanesh SB, Subashini C, James J. Hes1: the maestro in neurogenesis. Cell Mol Life Sci 2016; 73:4019-42. [PMID: 27233500 PMCID: PMC11108451 DOI: 10.1007/s00018-016-2277-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 05/12/2016] [Accepted: 05/12/2016] [Indexed: 10/21/2022]
Abstract
The process of neurogenesis is well orchestrated by the harmony of multiple cues in a spatiotemporal manner. In this review, we focus on how a dynamic gene, Hes1, is involved in neurogenesis with the view of its regulation and functional implications. Initially, we have reviewed the immense functional significance drawn by this maestro during neural development in a context-dependent manner. How this indispensable role of Hes1 in conferring the competency for neural differentiation partly relies on the direct/indirect mode of repression mediated by very specific structural and functional arms of this protein has also been outlined here. We also review the detailed molecular mechanisms behind the well-tuned oscillatory versus sustained expression of this antineurogenic bHLH repressor, which indeed makes it a master gene to implement the elusive task of neural progenitor propensity. Apart from the functional aspects of Hes1, we also discuss the molecular insights into the endogenous regulatory machinery that regulates its expression. Though Hes1 is a classical target of the Notch signaling pathway, we discuss here its differential expression at the molecular, cellular, and/or regional level. Moreover, we describe how its expression is fine-tuned by all possible ways of gene regulation such as epigenetic, transcriptional, post-transcriptional, post-translational, and environmental factors during vertebrate neurogenesis.
Collapse
Affiliation(s)
- Sivadasan Bindu Dhanesh
- Neuro Stem Cell Biology Laboratory, Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thycaud PO, Thiruvananthapuram, 695 014, Kerala, India
| | - Chandramohan Subashini
- Neuro Stem Cell Biology Laboratory, Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thycaud PO, Thiruvananthapuram, 695 014, Kerala, India
| | - Jackson James
- Neuro Stem Cell Biology Laboratory, Neurobiology Division, Rajiv Gandhi Centre for Biotechnology, Thycaud PO, Thiruvananthapuram, 695 014, Kerala, India.
| |
Collapse
|
12
|
Aaker JD, Elbaz B, Wu Y, Looney TJ, Zhang L, Lahn BT, Popko B. Transcriptional Fingerprint of Hypomyelination in Zfp191null and Shiverer (Mbpshi) Mice. ASN Neuro 2016; 8:8/5/1759091416670749. [PMID: 27683878 PMCID: PMC5046175 DOI: 10.1177/1759091416670749] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 08/22/2016] [Indexed: 12/20/2022] Open
Abstract
The transcriptional program that controls oligodendrocyte maturation and central nervous system (CNS) myelination has not been fully characterized. In this study, we use high-throughput RNA sequencing to analyze how the loss of a key transcription factor, zinc finger protein 191 (ZFP191), results in oligodendrocyte development abnormalities and CNS hypomyelination. Using a previously described mutant mouse that is deficient in ZFP191 protein expression (Zfp191null), we demonstrate that key transcripts are reduced in the whole brain as well as within oligodendrocyte lineage cells cultured in vitro. To determine whether the loss of myelin seen in Zfp191null mice contributes indirectly to these perturbations, we also examined the transcriptome of a well-characterized mouse model of hypomyelination, in which the myelin structural protein myelin basic protein (MBP) is deficient. Interestingly, Mbpshi (shiverer) mice had far fewer transcripts perturbed with the loss of myelin alone. This study demonstrates that the loss of ZFP191 disrupts expression of genes involved in oligodendrocyte maturation and myelination, largely independent from the loss of myelin. Nevertheless, hypomyelination in both mouse mutants results in the perturbation of lipid synthesis pathways, suggesting that oligodendrocytes have a feedback system that allows them to regulate myelin lipid synthesis depending on their myelinating state. The data presented are of potential clinical relevance as the human orthologs of the Zfp191 and MBP genes reside on a region of Chromosome 18 that is deleted in childhood leukodystrophies.
Collapse
Affiliation(s)
- Joshua D Aaker
- Department of Neurology, The University of Chicago Center for Peripheral Neuropathy, The University of Chicago, IL, USA
| | - Benayahu Elbaz
- Department of Neurology, The University of Chicago Center for Peripheral Neuropathy, The University of Chicago, IL, USA
| | - Yuwen Wu
- Department of Neurology, The University of Chicago Center for Peripheral Neuropathy, The University of Chicago, IL, USA
| | - Timothy J Looney
- Department of Human Genetics, The University of Chicago, IL, USA
| | - Li Zhang
- Department of Human Genetics, The University of Chicago, IL, USA
| | - Bruce T Lahn
- Department of Human Genetics, The University of Chicago, IL, USA
| | - Brian Popko
- Department of Neurology, The University of Chicago Center for Peripheral Neuropathy, The University of Chicago, IL, USA
| |
Collapse
|
13
|
Li J, Zhang L, Chu Y, Namaka M, Deng B, Kong J, Bi X. Astrocytes in Oligodendrocyte Lineage Development and White Matter Pathology. Front Cell Neurosci 2016; 10:119. [PMID: 27242432 PMCID: PMC4861901 DOI: 10.3389/fncel.2016.00119] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2016] [Accepted: 04/25/2016] [Indexed: 01/14/2023] Open
Abstract
White matter is primarily composed of myelin and myelinated axons. Structural and functional completeness of myelin is critical for the reliable and efficient transmission of information. White matter injury has been associated with the development of many demyelinating diseases. Despite a variety of scientific advances aimed at promoting re-myelination, their benefit has proven at best to be marginal. Research suggests that the failure of the re-myelination process may be the result of an unfavorable microenvironment. Astrocytes, are the most ample and diverse type of glial cells in central nervous system (CNS) which display multiple functions for the cells of the oligodendrocytes lineage. As such, much attention has recently been drawn to astrocyte function in terms of white matter myelin repair. They are different in white matter from those in gray matter in specific regards to development, morphology, location, protein expression and other supportive functions. During the process of demyelination and re-myelination, the functions of astrocytes are dynamic in that they are able to change functions in accordance to different time points, triggers or reactive pathways resulting in vastly different biologic effects. They have pivotal effects on oligodendrocytes and other cell types in the oligodendrocyte lineage by serving as an energy supplier, a participant of immunological and inflammatory functions, a source of trophic factors and iron and a sustainer of homeostasis. Astrocytic impairment has been shown to be directly linked to the development of neuromyelities optica (NMO). In addition, astroctyes have also been implicated in other white matter conditions such as psychiatric disorders and neurodegenerative diseases such as Alzheimer’s disease (AD), multiple sclerosis (MS) and amyotrophic lateral sclerosis (ALS). Inhibiting specifically detrimental signaling pathways in astrocytes while preserving their beneficial functions may be a promising approach for remyelination strategies. As such, the ability to manipulate astrocyte function represents a novel therapeutic approach that can repair the damaged myelin that is known to occur in a variety of white matter-related disorders.
Collapse
Affiliation(s)
- Jiasi Li
- Department of Neurology, Shanghai Changhai Hospital Shanghai, China
| | - Lei Zhang
- Department of Vascular Surgery, Shanghai Changhai Hospital Shanghai, China
| | - Yongxin Chu
- Department of Vascular Surgery, Affiliated Huai'an Hospital of Xuzhou Medical College Huai'an, China
| | - Michael Namaka
- Faculty of Health Sciences, College of Pharmacy and Medicine, University of Manitoba Winnipeg, MB, Canada
| | - Benqiang Deng
- Department of Neurology, Shanghai Changhai Hospital Shanghai, China
| | - Jiming Kong
- Department of Human Anatomy and Cell Science, University of Manitoba Winnipeg, MB, Canada
| | - Xiaoying Bi
- Department of Neurology, Shanghai Changhai Hospital Shanghai, China
| |
Collapse
|
14
|
Biamino E, Di Gregorio E, Belligni EF, Keller R, Riberi E, Gandione M, Calcia A, Mancini C, Giorgio E, Cavalieri S, Pappi P, Talarico F, Fea AM, De Rubeis S, Cirillo Silengo M, Ferrero GB, Brusco A. A novel 3q29 deletion associated with autism, intellectual disability, psychiatric disorders, and obesity. Am J Med Genet B Neuropsychiatr Genet 2016; 171B:290-9. [PMID: 26620927 DOI: 10.1002/ajmg.b.32406] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 11/12/2015] [Indexed: 12/22/2022]
Abstract
Copy number variation (CNV) has been associated with a variety of neuropsychiatric disorders, including intellectual disability/developmental delay (ID/DD), autism spectrum disorder (ASD), and schizophrenia (SCZ). Often, individuals carrying the same pathogenic CNV display high clinical variability. By array-CGH analysis, we identified a novel familial 3q29 deletion (1.36 Mb), centromeric to the 3q29 deletion region, which manifests with variable expressivity. The deletion was identified in a 3-year-old girl diagnosed with ID/DD and autism and segregated in six family members, all affected by severe psychiatric disorders including schizophrenia, major depression, anxiety disorder, and personality disorder. All individuals carrying the deletion were overweight or obese, and anomalies compatible with optic atrophy were observed in three out of four cases examined. Amongst the 10 genes encompassed by the deletion, the haploinsufficiency of Optic Atrophy 1 (OPA1), associated with autosomal dominant optic atrophy, is likely responsible for the ophthalmological anomalies. We hypothesize that the haploinsufficiency of ATPase type 13A4 (ATP13A4) and/or Hairy/Enhancer of Split Drosophila homolog 1 (HES1) contribute to the neuropsychiatric phenotype, while HES1 deletion might underlie the overweight/obesity. In conclusion, we propose a novel contiguous gene syndrome due to a proximal 3q29 deletion variably associated with autism, ID/DD, psychiatric traits and overweight/obesity.
Collapse
Affiliation(s)
- Elisa Biamino
- Department of Public Health and Pediatrics, University of Torino, Torino, Italy
| | - Eleonora Di Gregorio
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Torino, Italy
| | - Elga Fabia Belligni
- Department of Public Health and Pediatrics, University of Torino, Torino, Italy
| | | | - Evelise Riberi
- Department of Public Health and Pediatrics, University of Torino, Torino, Italy
| | - Marina Gandione
- Department of Neuropsychiatry, University of Torino, Torino, Italy
| | | | - Cecilia Mancini
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Elisa Giorgio
- Department of Medical Sciences, University of Torino, Torino, Italy
| | - Simona Cavalieri
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Torino, Italy.,Department of Medical Sciences, University of Torino, Torino, Italy
| | - Patrizia Pappi
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Torino, Italy
| | - Flavia Talarico
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Torino, Italy
| | - Antonio M Fea
- Department of Surgical Sciences, University of Torino, Torino, Italy
| | - Silvia De Rubeis
- Seaver Autism Center for Research and Treatment, Icahn School of Medicine at Mount Sinai, New York, New York.,Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, New York
| | | | | | - Alfredo Brusco
- Medical Genetics Unit, Città della Salute e della Scienza University Hospital, Torino, Italy.,Department of Medical Sciences, University of Torino, Torino, Italy
| |
Collapse
|
15
|
Küspert M, Wegner M. SomethiNG 2 talk about-Transcriptional regulation in embryonic and adult oligodendrocyte precursors. Brain Res 2015; 1638:167-182. [PMID: 26232072 DOI: 10.1016/j.brainres.2015.07.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/14/2015] [Accepted: 07/18/2015] [Indexed: 12/26/2022]
Abstract
Glial cells that express the chondroitin sulfate proteoglycan NG2 represent an inherently heterogeneous population. These so-called NG2-glia are present during development and in the adult CNS, where they are referred to as embryonic oligodendrocyte precursors and adult NG2-glia, respectively. They give rise to myelinating oligodendrocytes at all times of life. Over the years much has been learnt about the transcriptional network in embryonic oligodendrocyte precursors, and several transcription factors from the HLH, HMG-domain, zinc finger and homeodomain protein families have been identified as main constituents. Much less is known about the corresponding network in adult NG2-glia. Here we summarize and discuss current knowledge on functions of each of these transcription factor families in NG2-glia, and where possible compare transcriptional regulation in embryonic oligodendrocyte precursors and adult NG2-glia. This article is part of a Special Issue entitled SI:NG2-glia (Invited only).
Collapse
Affiliation(s)
- Melanie Küspert
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, Erlangen D-91054, Germany.
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, Erlangen D-91054, Germany.
| |
Collapse
|
16
|
Hernandez M, Casaccia P. Interplay between transcriptional control and chromatin regulation in the oligodendrocyte lineage. Glia 2015; 63:1357-75. [PMID: 25970296 DOI: 10.1002/glia.22818] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/26/2015] [Indexed: 12/21/2022]
Abstract
The recent years have been characterized by a surge of studies on the role of transcription factors and histone modifications in regulating the progression of progenitors into oligodendrocytes. This review summarizes this body of evidence and presents an integrated view of transcriptional networks and epigenetic regulators defining proliferating progenitors and their differentiation along the oligodendrocyte lineage. We suggest that transcription factors in proliferating progenitors have direct access to DNA, due to predominantly euchromatic nuclei. As progenitors differentiate, however, transcriptional competence is modulated by the formation of heterochromatin, which modifies the association of DNA with nucleosomal histones and renders the access of transcription factors dependent on the activity of epigenetic modulators. These concepts are delineated within the context of development, and the potential functional implications are discussed.
Collapse
Affiliation(s)
- Marylens Hernandez
- Department of Neuroscience, Friedman Brain Institute and Icahn School of Medicine at Mount Sinai, New York City, New York.,Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Patrizia Casaccia
- Department of Neuroscience, Friedman Brain Institute and Icahn School of Medicine at Mount Sinai, New York City, New York.,Department of Genomics and Multiscale Biology, Friedman Brain Institute and Icahn School of Medicine at Mount Sinai, New York City, New York
| |
Collapse
|
17
|
Havrda MC, Paolella BR, Ran C, Jering KS, Wray CM, Sullivan JM, Nailor A, Hitoshi Y, Israel MA. Id2 mediates oligodendrocyte precursor cell maturation arrest and is tumorigenic in a PDGF-rich microenvironment. Cancer Res 2014; 74:1822-32. [PMID: 24425046 DOI: 10.1158/0008-5472.can-13-1839] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Maturation defects occurring in adult tissue progenitor cells have the potential to contribute to tumor development; however, there is little experimental evidence implicating this cellular mechanism in the pathogenesis of solid tumors. Inhibitor of DNA-binding 2 (Id2) is a transcription factor known to regulate the proliferation and differentiation of primitive stem and progenitor cells. Id2 is derepressed in adult tissue neural stem cells (NSC) lacking the tumor suppressor Tp53 and modulates their proliferation. Constitutive expression of Id2 in differentiating NSCs resulted in maturation-resistant oligodendroglial precursor cells (OPC), a cell population implicated in the initiation of glioma. Mechanistically, Id2 overexpression was associated with inhibition of the Notch effector Hey1, a bHLH transcription factor that we here characterize as a direct transcriptional repressor of the oligodendroglial lineage determinant Olig2. Orthotopic inoculation of NSCs with enhanced Id2 expression into brains of mice engineered to express platelet-derived growth factor in the central nervous system resulted in glioma. These data implicate a mechanism of altered NSC differentiation in glioma development and characterize a novel mouse model that reflects key characteristics of the recently described proneural subtype of glioblastoma multiforme. Such findings support the emerging concept that the cellular and molecular characteristics of tumor cells are linked to the transformation of distinct subsets of adult tissue progenitors.
Collapse
Affiliation(s)
- Matthew C Havrda
- Authors' Affiliations: Norris Cotton Cancer Center; Departments of Genetics; Pediatrics; and Pharmacology and Toxicology, Geisel School of Medicine at Dartmouth, Lebanon, New Hampshire
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Nguyen GD, Gokhan S, Molero AE, Mehler MF. Selective roles of normal and mutant huntingtin in neural induction and early neurogenesis. PLoS One 2013; 8:e64368. [PMID: 23691206 PMCID: PMC3653864 DOI: 10.1371/journal.pone.0064368] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2013] [Accepted: 04/12/2013] [Indexed: 01/08/2023] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by abnormal polyglutamine expansion in the amino-terminal end of the huntingtin protein (Htt) and characterized by progressive striatal and cortical pathology. Previous reports have shown that Htt is essential for embryogenesis, and a recent study by our group revealed that the pathogenic form of Htt (mHtt) causes impairments in multiple stages of striatal development. In this study, we have examined whether HD-associated striatal developmental deficits are reflective of earlier maturational alterations occurring at the time of neurulation by assessing differential roles of Htt and mHtt during neural induction and early neurogenesis using an in vitro mouse embryonic stem cell (ESC) clonal assay system. We demonstrated that the loss of Htt in ESCs (KO ESCs) severely disrupts the specification of primitive and definitive neural stem cells (pNSCs, dNSCs, respectively) during the process of neural induction. In addition, clonally derived KO pNSCs and dNSCs displayed impaired proliferative potential, enhanced cell death and altered multi-lineage potential. Conversely, as observed in HD knock-in ESCs (Q111 ESCs), mHtt enhanced the number and size of pNSC clones, which exhibited enhanced proliferative potential and precocious neuronal differentiation. The transition from Q111 pNSCs to fibroblast growth factor 2 (FGF2)-responsive dNSCs was marked by potentiation in the number of dNSCs and altered proliferative potential. The multi-lineage potential of Q111 dNSCs was also enhanced with precocious neurogenesis and oligodendrocyte progenitor elaboration. The generation of Q111 epidermal growth factor (EGF)-responsive dNSCs was also compromised, whereas their multi-lineage potential was unaltered. These abnormalities in neural induction were associated with differential alterations in the expression profiles of Notch, Hes1 and Hes5. These cumulative observations indicate that Htt is required for multiple stages of neural induction, whereas mHtt enhances this process and promotes precocious neurogenesis and oligodendrocyte progenitor cell elaboration.
Collapse
Affiliation(s)
- Giang D. Nguyen
- Roslyn and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Solen Gokhan
- Roslyn and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Aldrin E. Molero
- Roslyn and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Mark F. Mehler
- Roslyn and Leslie Goldstein Laboratory for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Institute for Brain Disorders and Neural Regeneration, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Neurology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Psychiatry and Behavioral Sciences, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Rose F. Kennedy Center for Research on Intellectual and Developmental Disabilities, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Center for Epigenomics, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York, United States of America
- * E-mail:
| |
Collapse
|
19
|
Chou SJ, O'Leary DDM. Role for Lhx2 in corticogenesis through regulation of progenitor differentiation. Mol Cell Neurosci 2013; 56:1-9. [PMID: 23454273 DOI: 10.1016/j.mcn.2013.02.006] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Accepted: 02/19/2013] [Indexed: 01/04/2023] Open
Abstract
The neocortex represents the brain region that has undergone a major increase in its relative size during the course of mammalian evolution. The larger cortex results from a corresponding increase in progenitor cell number. The progenitors giving rise to neocortex are located in the ventricular zone of the dorsal telencephalon and highly express Lhx2, a LIM-homeodomain transcription factor. The neocortex fails to form in the Lhx2 constitutive knockout, indicating a role for Lhx2 in corticogenesis, but mid-embryonic lethality of the Lhx2 knockout requires the use of conditional strategies for further studies. Therefore, to explore Lhx2 function in neocortical progenitors, we generated mice with Lhx2 conditionally deleted from cortical progenitors at the onset of neurogenesis. We find that Lhx2 is critical for maintaining the proliferative state of neocortical progenitors during corticogenesis. In the conditional knockouts, the neocortex is formed but is significantly smaller than wild type. We find that deletion of Lhx2 leads to significantly decreased numbers of cortical progenitors and premature neuronal differentiation. A likely mechanism is indicated by our findings that Lhx2 is required for the expression of Hes1 in cortical progenitors, a key effector in the Notch signaling pathway that maintains the proliferative progenitor state. We conclude that Lhx2 regulates the balance between proliferation and differentiation in cortical progenitors and through this mechanism Lhx2 controls cortical size.
Collapse
Affiliation(s)
- Shen-Ju Chou
- Molecular Neurobiology Laboratory, The Salk Institute for Biological Studies, 10010 N. Torrey Pines Rd., La Jolla, CA 92037, USA.
| | | |
Collapse
|
20
|
Ueno T, Ito J, Hoshikawa S, Ohori Y, Fujiwara S, Yamamoto S, Ohtsuka T, Kageyama R, Akai M, Nakamura K, Ogata T. The identification of transcriptional targets of Ascl1 in oligodendrocyte development. Glia 2012; 60:1495-505. [PMID: 22714260 DOI: 10.1002/glia.22369] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2012] [Revised: 04/24/2012] [Accepted: 05/21/2012] [Indexed: 11/06/2022]
Abstract
The basic helix-loop-helix (bHLH) transcription factor Ascl1 plays crucial roles in both oligodendrocyte development and neuronal development; however, the molecular target of Ascl1 in oligodendrocyte progenitor cells (OPCs) remains elusive. To identify the downstream targets of Ascl1 in OPCs, we performed gene expression microarray analysis and identified Hes5 as a putative downstream target of Ascl1. In vivo analysis revealed that Ascl1 and Hes5 were coexpressed in early developmental oligodendrocytes in both the telencephalon and the ventral spinal cord. We also found that Hes5 expression was reduced in the OPCs of Ascl1 mutant mice. Furthermore, we demonstrated that Ascl1 directly binds to an E-box region within the Hes5 promoter and regulates Hes5 expression at the transcriptional level. Taken together, these in vivo and in vitro data suggest that Ascl1 induces Hes5 expression in a cell-autonomous manner. Considering the previously known function of Hes5 as a repressor of Ascl1, our data indicate that Hes5 is involved in the negative feedback regulation of Ascl1.
Collapse
Affiliation(s)
- Takaaki Ueno
- Department of Rehabilitation for the Movement Functions, Research Institute, National Rehabilitation Center for Persons with Disabilities, Saitama, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Weng Q, Chen Y, Wang H, Xu X, Yang B, He Q, Shou W, Chen Y, Higashi Y, van den Berghe V, Seuntjens E, Kernie SG, Bukshpun P, Sherr EH, Huylebroeck D, Lu QR. Dual-mode modulation of Smad signaling by Smad-interacting protein Sip1 is required for myelination in the central nervous system. Neuron 2012; 73:713-28. [PMID: 22365546 DOI: 10.1016/j.neuron.2011.12.021] [Citation(s) in RCA: 135] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2011] [Indexed: 01/17/2023]
Abstract
Myelination by oligodendrocytes in the central nervous system (CNS) is essential for proper brain function, yet the molecular determinants that control this process remain poorly understood. The basic helix-loop-helix transcription factors Olig1 and Olig2 promote myelination, whereas bone morphogenetic protein (BMP) and Wnt/β-catenin signaling inhibit myelination. Here we show that these opposing regulators of myelination are functionally linked by the Olig1/2 common target Smad-interacting protein-1 (Sip1). We demonstrate that Sip1 is an essential modulator of CNS myelination. Sip1 represses differentiation inhibitory signals by antagonizing BMP receptor-activated Smad activity while activating crucial oligodendrocyte-promoting factors. Importantly, a key Sip1-activated target, Smad7, is required for oligodendrocyte differentiation and partially rescues differentiation defects caused by Sip1 loss. Smad7 promotes myelination by blocking the BMP- and β-catenin-negative regulatory pathways. Thus, our findings reveal that Sip1-mediated antagonism of inhibitory signaling is critical for promoting CNS myelination and point to new mediators for myelin repair.
Collapse
Affiliation(s)
- Qinjie Weng
- Department of Developmental Biology and Kent Waldrep Foundation Center for Basic Neuroscience Research on Nerve Growth and Regeneration, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|