1
|
Yang Y, Song R, Gao Y, Yu H, Wang S. Regulatory mechanisms and therapeutic potential of JAB1 in neurological development and disorders. Mol Med 2023; 29:80. [PMID: 37365502 DOI: 10.1186/s10020-023-00675-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/30/2023] [Indexed: 06/28/2023] Open
Abstract
c-Jun activation domain binding protein-1 (JAB1) is a multifunctional regulator that plays vital roles in diverse cellular processes. It regulates AP-1 transcriptional activity and also acts as the fifth component of the COP9 signalosome complex. While JAB1 is considered an oncoprotein that triggers tumor development, recent studies have shown that it also functions in neurological development and disorders. In this review, we summarize the general features of the JAB1 gene and protein, and present recent updates on the regulation of JAB1 expression. Moreover, we also highlight the functional roles and regulatory mechanisms of JAB1 in neurodevelopmental processes such as neuronal differentiation, synaptic morphogenesis, myelination, and hair cell development and in the pathogenesis of some neurological disorders such as Alzheimer's disease, multiple sclerosis, neuropathic pain, and peripheral nerve injury. Furthermore, current challenges and prospects are discussed, including updates on drug development targeting JAB1.
Collapse
Affiliation(s)
- Yu Yang
- Department of Psychiatry, Jining Medical University, Jianshe South Road No. 45, Jining, Shandong, China
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, Shandong, China
| | - Ruying Song
- Department of Psychiatry, Jining Medical University, Jianshe South Road No. 45, Jining, Shandong, China
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, Shandong, China
| | - Yiming Gao
- Department of Psychiatry, Jining Medical University, Jianshe South Road No. 45, Jining, Shandong, China
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, Shandong, China
| | - Hao Yu
- Department of Psychiatry, Jining Medical University, Jianshe South Road No. 45, Jining, Shandong, China.
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, Shandong, China.
| | - Shuai Wang
- Department of Psychiatry, Jining Medical University, Jianshe South Road No. 45, Jining, Shandong, China.
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Jining Medical University, Jining, Shandong, China.
| |
Collapse
|
2
|
Domingo-Muelas A, Morante-Redolat JM, Moncho-Amor V, Jordán-Pla A, Pérez-Villalba A, Carrillo-Barberà P, Belenguer G, Porlan E, Kirstein M, Bachs O, Ferrón SR, Lovell-Badge R, Fariñas I. The rates of adult neurogenesis and oligodendrogenesis are linked to cell cycle regulation through p27-dependent gene repression of SOX2. Cell Mol Life Sci 2023; 80:36. [PMID: 36627412 PMCID: PMC9832098 DOI: 10.1007/s00018-022-04676-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 12/15/2022] [Indexed: 01/12/2023]
Abstract
Cell differentiation involves profound changes in global gene expression that often has to occur in coordination with cell cycle exit. Because cyclin-dependent kinase inhibitor p27 reportedly regulates proliferation of neural progenitor cells in the subependymal neurogenic niche of the adult mouse brain, but can also have effects on gene expression, we decided to molecularly analyze its role in adult neurogenesis and oligodendrogenesis. At the cell level, we show that p27 restricts residual cyclin-dependent kinase activity after mitogen withdrawal to antagonize cycling, but it is not essential for cell cycle exit. By integrating genome-wide gene expression and chromatin accessibility data, we find that p27 is coincidentally necessary to repress many genes involved in the transit from multipotentiality to differentiation, including those coding for neural progenitor transcription factors SOX2, OLIG2 and ASCL1. Our data reveal both a direct association of p27 with regulatory sequences in the three genes and an additional hierarchical relationship where p27 repression of Sox2 leads to reduced levels of its downstream targets Olig2 and Ascl1. In vivo, p27 is also required for the regulation of the proper level of SOX2 necessary for neuroblasts and oligodendroglial progenitor cells to timely exit cell cycle in a lineage-dependent manner.
Collapse
Affiliation(s)
- Ana Domingo-Muelas
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departamento de Biología Celular Biología Funcional y Antropología Física, Universidad de Valencia, 46100, Burjassot, Spain
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universidad de Valencia, Valencia, Spain
- Department of Cell and Developmental Biology, Smilow Center for Translational Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jose Manuel Morante-Redolat
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departamento de Biología Celular Biología Funcional y Antropología Física, Universidad de Valencia, 46100, Burjassot, Spain
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universidad de Valencia, Valencia, Spain
| | - Verónica Moncho-Amor
- The Francis Crick Institute, London, NW1 1AT, UK
- IIS Biodonostia, 48013, Bilbao, Spain
| | - Antonio Jordán-Pla
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universidad de Valencia, Valencia, Spain
| | - Ana Pérez-Villalba
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departamento de Biología Celular Biología Funcional y Antropología Física, Universidad de Valencia, 46100, Burjassot, Spain
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universidad de Valencia, Valencia, Spain
| | - Pau Carrillo-Barberà
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departamento de Biología Celular Biología Funcional y Antropología Física, Universidad de Valencia, 46100, Burjassot, Spain
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universidad de Valencia, Valencia, Spain
- Institute for Research in Biomedicine, 6500, Bellinzona, Switzerland
| | - Germán Belenguer
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departamento de Biología Celular Biología Funcional y Antropología Física, Universidad de Valencia, 46100, Burjassot, Spain
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universidad de Valencia, Valencia, Spain
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307, Dresden, Germany
| | - Eva Porlan
- Departamento de Biología Molecular, Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Instituto de Investigación Hospital Universitario La Paz (IdiPAZ), Instituto de Salud Carlos III, Madrid, Spain
| | - Martina Kirstein
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departamento de Biología Celular Biología Funcional y Antropología Física, Universidad de Valencia, 46100, Burjassot, Spain
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universidad de Valencia, Valencia, Spain
| | - Oriol Bachs
- Department of Biomedical Sciences, University of Barcelona-IDIBAPS, CIBERONC, Barcelona, Spain
| | - Sacri R Ferrón
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Departamento de Biología Celular Biología Funcional y Antropología Física, Universidad de Valencia, 46100, Burjassot, Spain
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universidad de Valencia, Valencia, Spain
| | | | - Isabel Fariñas
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
- Departamento de Biología Celular Biología Funcional y Antropología Física, Universidad de Valencia, 46100, Burjassot, Spain.
- Instituto de Biotecnología y Biomedicina (BioTecMed), Universidad de Valencia, Valencia, Spain.
| |
Collapse
|
3
|
Dermitzakis I, Manthou ME, Meditskou S, Miliaras D, Kesidou E, Boziki M, Petratos S, Grigoriadis N, Theotokis P. Developmental Cues and Molecular Drivers in Myelinogenesis: Revisiting Early Life to Re-Evaluate the Integrity of CNS Myelin. Curr Issues Mol Biol 2022; 44:3208-3237. [PMID: 35877446 PMCID: PMC9324160 DOI: 10.3390/cimb44070222] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/14/2022] [Accepted: 07/17/2022] [Indexed: 02/07/2023] Open
Abstract
The mammalian central nervous system (CNS) coordinates its communication through saltatory conduction, facilitated by myelin-forming oligodendrocytes (OLs). Despite the fact that neurogenesis from stem cell niches has caught the majority of attention in recent years, oligodendrogenesis and, more specifically, the molecular underpinnings behind OL-dependent myelinogenesis, remain largely unknown. In this comprehensive review, we determine the developmental cues and molecular drivers which regulate normal myelination both at the prenatal and postnatal periods. We have indexed the individual stages of myelinogenesis sequentially; from the initiation of oligodendrocyte precursor cells, including migration and proliferation, to first contact with the axon that enlists positive and negative regulators for myelination, until the ultimate maintenance of the axon ensheathment and myelin growth. Here, we highlight multiple developmental pathways that are key to successful myelin formation and define the molecular pathways that can potentially be targets for pharmacological interventions in a variety of neurological disorders that exhibit demyelination.
Collapse
Affiliation(s)
- Iasonas Dermitzakis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Maria Eleni Manthou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Soultana Meditskou
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Dimosthenis Miliaras
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
| | - Evangelia Kesidou
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Marina Boziki
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC 3004, Australia;
| | - Nikolaos Grigoriadis
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
| | - Paschalis Theotokis
- Department of Histology-Embryology, School of Medicine, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (I.D.); (M.E.M.); (S.M.); (D.M.)
- Laboratory of Experimental Neurology and Neuroimmunology, Second Department of Neurology, AHEPA University Hospital, 54621 Thessaloniki, Greece; (E.K.); (M.B.); (N.G.)
- Correspondence:
| |
Collapse
|
4
|
Neumann DP, Goodall GJ, Gregory PA. The Quaking RNA-binding proteins as regulators of cell differentiation. WILEY INTERDISCIPLINARY REVIEWS. RNA 2022; 13:e1724. [PMID: 35298877 PMCID: PMC9786888 DOI: 10.1002/wrna.1724] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 12/30/2022]
Abstract
The RNA-binding protein Quaking (QKI) has emerged as a potent regulator of cellular differentiation in developmental and pathological processes. The QKI gene is itself alternatively spliced to produce three major isoforms, QKI-5, QKI-6, and QKI-7, that possess very distinct functions. Here, we highlight roles of the different QKI isoforms in neuronal, vascular, muscle, and monocyte cell differentiation, and during epithelial-mesenchymal transition in cancer progression. QKI isoforms control cell differentiation through regulating alternative splicing, mRNA stability and translation, with activities in gene transcription now also becoming evident. These diverse functions of the QKI isoforms contribute to their broad influences on RNA metabolism and cellular differentiation. This article is categorized under: RNA Interactions with Proteins and Other Molecules > Protein-RNA Interactions: Functional Implications RNA Processing > Splicing Regulation/Alternative Splicing RNA in Disease and Development > RNA in Development.
Collapse
Affiliation(s)
- Daniel P. Neumann
- Centre for Cancer BiologyUniversity of South Australia and SA PathologyAdelaideSouth Australia
| | - Gregory J. Goodall
- Centre for Cancer BiologyUniversity of South Australia and SA PathologyAdelaideSouth Australia,Faculty of Health and Medical SciencesThe University of AdelaideAdelaideSouth Australia
| | - Philip A. Gregory
- Centre for Cancer BiologyUniversity of South Australia and SA PathologyAdelaideSouth Australia,Faculty of Health and Medical SciencesThe University of AdelaideAdelaideSouth Australia
| |
Collapse
|
5
|
Grison A, Atanasoski S. Cyclins, Cyclin-Dependent Kinases, and Cyclin-Dependent Kinase Inhibitors in the Mouse Nervous System. Mol Neurobiol 2020; 57:3206-3218. [PMID: 32506380 DOI: 10.1007/s12035-020-01958-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Accepted: 05/26/2020] [Indexed: 12/11/2022]
Abstract
Development and normal physiology of the nervous system require proliferation and differentiation of stem and progenitor cells in a strictly controlled manner. The number of cells generated depends on the type of cell division, the cell cycle length, and the fraction of cells that exit the cell cycle to become quiescent or differentiate. The underlying processes are tightly controlled and modulated by cyclin-dependent kinases (Cdks) and their interactions with cyclins and Cdk inhibitors (CKIs). Studies performed in the nervous system with mouse models lacking individual Cdks, cyclins, and CKIs, or combinations thereof, have shown that many of these molecules control proliferation rates in a cell-type specific and time-dependent manner. In this review, we will provide an update on the in vivo studies on cyclins, Cdks, and CKIs in neuronal and glial tissue. The goal is to highlight their impact on proliferation processes during the development of the peripheral and central nervous system, including and comparing normal and pathological conditions in the adult.
Collapse
Affiliation(s)
- Alice Grison
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Suzana Atanasoski
- Department of Biomedicine, University of Basel, Basel, Switzerland. .,Faculty of Medicine, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
6
|
Craig GA, Yoo S, Du TY, Xiao J. Plasticity in oligodendrocyte lineage progression: An OPC puzzle on our nerves. Eur J Neurosci 2020; 54:5747-5761. [PMID: 32478920 DOI: 10.1111/ejn.14845] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 05/20/2020] [Accepted: 05/21/2020] [Indexed: 11/28/2022]
Abstract
Myelin deposition in the central nervous system has been shown to be responsive to experience, with sensory enrichment increasing myelination and sensory or social deprivation decreasing myelination. This process is referred to as "adaptive myelination" or "myelin plasticity" and signifies an essential component of new learning. However, whether these experience-driven adaptations are driven by (a) underlying changes in the generation of myelinating cells, (b) altered interactions between myelin sheath and axon, or (c) a combination of the above remains unclear. It has been suggested that myelination largely follows an "innate" and automatic programme, allowing for a predictable pattern of central nervous system myelin deposition over time. Adaptive myelination is thought to account for more nuanced alterations that do not dramatically shift this pattern, but ultimately drive functional responses. This makes the study of myelin plasticity particularly difficult, as it necessitates being able to clearly and specifically draw boundaries between the innate and adaptive programme. Thus, the field requires a holistic understanding of the remit of innate myelin development, prior to investigation of adaptive myelination. This review will collate literature regarding different aspects of oligodendrocyte and myelin development (namely, oligodendrocyte proliferation, differentiation, death and myelin sheath formation) in an innate context, before discussing how these parameters are proposed to change under adaptive conditions. It is the hope that this review will highlight the need for a comprehensive and integrated approach towards studying both innate and adaptive forms of myelination.
Collapse
Affiliation(s)
- Georgina A Craig
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Vic., Australia
| | - SangWon Yoo
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Vic., Australia
| | - Tian Y Du
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Vic., Australia
| | - Junhua Xiao
- Department of Anatomy and Neuroscience, School of Biomedical Sciences, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Parkville, Vic., Australia
| |
Collapse
|
7
|
Colwell CS, Ghiani CA. Potential Circadian Rhythms in Oligodendrocytes? Working Together Through Time. Neurochem Res 2019; 45:591-605. [PMID: 30906970 DOI: 10.1007/s11064-019-02778-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Accepted: 03/14/2019] [Indexed: 12/15/2022]
Abstract
Oligodendrocytes (OL) are the only myelinating cells of the central nervous system thus interferences, either environmental or genetic, with their maturation or function have devastating consequences. Albeit so far neglected, one of the less appreciated, nevertheless possible, regulators of OL maturation and function is the circadian cycle. Yet, disruptions in these rhythms are unfortunately becoming a common "disorder" in the today's world. The temporal patterning of behaviour and physiology is controlled by a circadian timing system based in the anterior hypothalamus. At the molecular level, circadian rhythms are generated by a transcriptional/translational feedback system that regulates transcription and has a major impact on cellular function(s). Fundamental cellular properties/functions in most cell types vary with the daily circadian cycle: OL are unlikely an exception! To be clear, the presence of circadian oscillators or the cell-specific function(s) of the circadian clock in OL has yet to be defined. Furthermore, we wish to entertain the idea of links between the "thin" evidence on OL intrinsic circadian rhythms and their interjection(s) at different stages of lineage progression as well as in supporting/regulating OL crucial function: myelination. Individuals with intellectual and developmental syndromes as well as neurodegenerative diseases present with a disrupted sleep/wake cycle; hence, we raise the possibility that these disturbances in timing can contribute to the loss of white matter observed in these disorders. Preclinical and clinical work in this area is needed for a better understanding of how circadian rhythms influence OL maturation and function(s), to aid the development of new therapeutic strategies and standards of care for these patients.
Collapse
Affiliation(s)
- Christopher S Colwell
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.,Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Cristina A Ghiani
- Department of Psychiatry and Biobehavioral Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA. .,Department of Pathology & Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA. .,Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
8
|
Abstract
Oligodendrocytes produce myelin, an insulating sheath required for the saltatory conduction of electrical impulses along axons. Oligodendrocyte loss results in demyelination, which leads to impaired neurological function in a broad array of diseases ranging from pediatric leukodystrophies and cerebral palsy, to multiple sclerosis and white matter stroke. Accordingly, replacing lost oligodendrocytes, whether by transplanting oligodendrocyte progenitor cells (OPCs) or by mobilizing endogenous progenitors, holds great promise as a therapeutic strategy for the diseases of central white matter. In this Primer, we describe the molecular events regulating oligodendrocyte development and how our understanding of this process has led to the establishment of methods for producing OPCs and oligodendrocytes from embryonic stem cells and induced pluripotent stem cells, as well as directly from somatic cells. In addition, we will discuss the safety of engrafted stem cell-derived OPCs, as well as approaches by which to modulate their differentiation and myelinogenesis in vivo following transplantation.
Collapse
Affiliation(s)
- Steven A Goldman
- Center for Translational Neuromedicine and the Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA Center for Basic and Translational Neuroscience, University of Copenhagen, Faculty of Health and Medical Sciences, Copenhagen 2200, Denmark Neuroscience Center, Rigshospitalet, Copenhagen 2100, Denmark
| | - Nicholas J Kuypers
- Center for Translational Neuromedicine and the Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
9
|
Moyon S, Liang J, Casaccia P. Epigenetics in NG2 glia cells. Brain Res 2016; 1638:183-198. [PMID: 26092401 PMCID: PMC4683112 DOI: 10.1016/j.brainres.2015.06.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 05/11/2015] [Accepted: 06/02/2015] [Indexed: 12/16/2022]
Abstract
The interplay of transcription and epigenetic marks is essential for oligodendrocyte progenitor cell (OPC) proliferation and differentiation during development. Here, we review the recent advances in this field and highlight mechanisms of transcriptional repression and activation involved in OPC proliferation, differentiation and plasticity. We also describe how dysregulation of these epigenetic events may affect demyelinating disorders, and consider potential ways to manipulate NG2 cell behavior through modulation of the epigenome. This article is part of a Special Issue entitled SI:NG2-glia(Invited only).
Collapse
Affiliation(s)
- Sarah Moyon
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jialiang Liang
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Patrizia Casaccia
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neurology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Corinne Goldsmith Dickinson Center for Multiple Sclerosis, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
10
|
Haines JD, Fulton DL, Richard S, Almazan G. p38 Mitogen-Activated Protein Kinase Pathway Regulates Genes during Proliferation and Differentiation in Oligodendrocytes. PLoS One 2015; 10:e0145843. [PMID: 26714323 PMCID: PMC4699908 DOI: 10.1371/journal.pone.0145843] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2010] [Accepted: 12/09/2015] [Indexed: 01/06/2023] Open
Abstract
We have previously shown that p38 mitogen-activated protein kinase (p38 MAPK) is important for oligodendrocyte (OLG) differentiation and myelination. However, the precise cellular mechanisms by which p38 regulates OLG differentiation remain largely unknown. To determine whether p38 functions in part through transcriptional events in regulating OLG identity, we performed microarray analysis on differentiating oligodendrocyte progenitors (OLPs) treated with a p38 inhibitor. Consistent with a role in OLG differentiation, pharmacological inhibition of p38 down-regulated the transcription of genes that are involved in myelin biogenesis, transcriptional control and cell cycle. Proliferation assays showed that OLPs treated with the p38 inhibitor retained a proliferative capacity which could be induced upon application of mitogens demonstrating that after two days of p38-inhibition OLGs remained poised to continue mitosis. Together, our results suggest that the p38 pathway regulates gene transcription which can coordinate OLG differentiation. Our microarray dataset will provide a useful resource for future studies investigating the molecular mechanisms by which p38 regulates oligodendrocyte differentiation and myelination.
Collapse
Affiliation(s)
- Jeffery D. Haines
- Department of Pharmacology and Therapeutics, McGill University, 3655 Sir William Osler Promenade, Montreal, Quebec, Canada, H3G 1Y6
| | - Debra L. Fulton
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, 3801 University St, Montreal, Quebec, Canada, H3A 2B4
| | - Stephane Richard
- Terry Fox Molecular Oncology Group, Bloomfield Center for Research on Aging, Lady Davis Institute for Medical Research, and Departments of Oncology and Medicine, McGill University, Montreal, Quebec, Canada, H3T 1E2
| | - Guillermina Almazan
- Department of Pharmacology and Therapeutics, McGill University, 3655 Sir William Osler Promenade, Montreal, Quebec, Canada, H3G 1Y6
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, 3801 University St, Montreal, Quebec, Canada, H3A 2B4
- * E-mail:
| |
Collapse
|
11
|
Myers KR, Liu G, Feng Y, Zheng JQ. Oligodendroglial defects during quakingviable cerebellar development. Dev Neurobiol 2015; 76:972-82. [PMID: 26645409 DOI: 10.1002/dneu.22369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Revised: 11/16/2015] [Accepted: 12/01/2015] [Indexed: 11/06/2022]
Abstract
The selective RNA-binding protein Quaking I (QKI) has previously been implicated in RNA localization and stabilization, alternative splicing, cell proliferation, and differentiation. The spontaneously-occurring quakingviable (qkv) mutant mouse exhibits a sharply attenuated level of QKI in myelin-producing cells, including oligodendrocytes (OL) because of the loss of an OL-specific promoter. The disruption of QKI in OLs results in severe hypomyelination of the central nervous system, but the underlying cellular mechanisms remain to be fully elucidated. In this study, we used the qkv mutant mouse as a model to study myelination defects in the cerebellum. We found that oligodendroglial development and myelination are adversely affected in the cerebellum of qkv mice. Specifically, we identified an increase in the total number of oligodendroglial precursor cells in qkv cerebella, a substantial portion of which migrated into the grey matter. Furthermore, these mislocalized oligodendroglial precursor cells retained their migratory morphology late into development. Interestingly, a number of these presumptive oligodendrocyte precursors were found at the Purkinje cell layer in qkv cerebella, resembling Bergman glia. These findings indicate that QKI is involved in multiple aspects of oligodendroglial development. QKI disruption can impact the cell fate of oligodendrocyte precursor cells, their migration and differentiation, and ultimately myelination in the cerebellum. © 2015 Wiley Periodicals, Inc. Develop Neurobiol 76: 972-982, 2016.
Collapse
Affiliation(s)
- Kenneth R Myers
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, 30322.,Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, Georgia, 30322
| | - Guanglu Liu
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia, 30322
| | - Yue Feng
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia, 30322
| | - James Q Zheng
- Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, 30322.,Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, Georgia, 30322.,Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, 30322
| |
Collapse
|
12
|
A Basal Tone of 2-Arachidonoylglycerol Contributes to Early Oligodendrocyte Progenitor Proliferation by Activating Phosphatidylinositol 3-Kinase (PI3K)/AKT and the Mammalian Target of Rapamycin (MTOR) Pathways. J Neuroimmune Pharmacol 2015; 10:309-17. [DOI: 10.1007/s11481-015-9609-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Accepted: 03/30/2015] [Indexed: 12/19/2022]
|
13
|
|
14
|
Liu B, Chen X, Wang ZQ, Tong WM. Nbn gene inactivation in the CNS of mouse inhibits the myelinating ability of the mature cortical oligodendrocytes. Glia 2014; 62:133-44. [PMID: 24272708 DOI: 10.1002/glia.22593] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 10/07/2013] [Accepted: 10/17/2013] [Indexed: 01/12/2023]
Abstract
Nijmegen Breakage Syndrome (NBS) is a recessive genetic disorder characterized by immunodeficiency, elevated sensitivity to ionizing radiation, chromosomal instability, microcephaly, and high predisposition to malignancies. Since the underlying molecular mechanisms of the NBS microcephaly are still obscure, thus our group previously inactivated the Nbn gene in the central nervous system (CNS) of mice by nestin-Cre targeting gene system, and generated Nbn(CNS-del) mice. Interestingly, the newborn Nbn(CNS-del) mice exhibit obvious microcephaly, which is accompanied by severe ataxia and balance deficiency. In this study presented here, we report that Nbn-deficiency induces the enhanced apoptosis of the mature oligodendrocytes at postnatal day 7, which further affects the myelination of the nerve fibers of cerebrum and corpus callosum.The distinct regulatory roles of Ataxia telangiectasia mutated (ATM) signaling and protein kinase B(Akt)/the mammalian target of Rapamycin (AKT/mTOR) signaling are responsible for the enhanced apoptosis of the Nbn-deficient oligodendrocytes. In addition, a series of transcriptional factors including histonedeacetylase (HDAC), zinc finger protein 191 (ZFP-191) and myelin sheath regulatory factor (MRF) play distinct roles in regulating the myelination of the Nbn-deficient oligodendrocytes. Based on these results, it concludes that ATM-Chk2-P53-P21 signaling pathway and the AKT/mTOR signaling pathway are both responsible for the enhanced apoptosis of the Nbn-deficient oligodendrocytes. HDAC, ZFP-191, and MRF are also involved in the pathogenesis of the hypomyelination of the Nbn-deficient oligodendrocytes.
Collapse
Affiliation(s)
- Bo Liu
- Department of Pathology, Center for Experimental Animal Research, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | |
Collapse
|
15
|
Porrello E, Rivellini C, Dina G, Triolo D, Del Carro U, Ungaro D, Panattoni M, Feltri ML, Wrabetz L, Pardi R, Quattrini A, Previtali SC. Jab1 regulates Schwann cell proliferation and axonal sorting through p27. ACTA ACUST UNITED AC 2013; 211:29-43. [PMID: 24344238 PMCID: PMC3892969 DOI: 10.1084/jem.20130720] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Jab1 constitutes a regulatory molecule that integrates laminin211 signals in Schwann cells to govern cell cycle, cell number, and differentiation. Axonal sorting is a crucial event in nerve formation and requires proper Schwann cell proliferation, differentiation, and contact with axons. Any defect in axonal sorting results in dysmyelinating peripheral neuropathies. Evidence from mouse models shows that axonal sorting is regulated by laminin211– and, possibly, neuregulin 1 (Nrg1)–derived signals. However, how these signals are integrated in Schwann cells is largely unknown. We now report that the nuclear Jun activation domain–binding protein 1 (Jab1) may transduce laminin211 signals to regulate Schwann cell number and differentiation during axonal sorting. Mice with inactivation of Jab1 in Schwann cells develop a dysmyelinating neuropathy with axonal sorting defects. Loss of Jab1 increases p27 levels in Schwann cells, which causes defective cell cycle progression and aberrant differentiation. Genetic down-regulation of p27 levels in Jab1-null mice restores Schwann cell number, differentiation, and axonal sorting and rescues the dysmyelinating neuropathy. Thus, Jab1 constitutes a regulatory molecule that integrates laminin211 signals in Schwann cells to govern cell cycle, cell number, and differentiation. Finally, Jab1 may constitute a key molecule in the pathogenesis of dysmyelinating neuropathies.
Collapse
Affiliation(s)
- Emanuela Porrello
- Institute of Experimental Neurology (INSPE), Division of Neuroscience; 2 Department of Neurology; and 3 Division of Immunology, Transplantation, and Infectious Disease; San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Liu B, Chen X, Wang ZQ, Tong WM. DNA damage and oxidative injury are associated with hypomyelination in the corpus callosum of newborn Nbn(CNS-del) mice. J Neurosci Res 2013; 92:254-66. [PMID: 24272991 DOI: 10.1002/jnr.23313] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 09/10/2013] [Accepted: 09/18/2013] [Indexed: 01/16/2023]
Abstract
Nijmegen breakage syndrome (NBS), caused by mutation of the Nbn gene, is a recessive genetic disorder characterized by immunodeficiency, elevated sensitivity to ionizing radiation, chromosomal instability, microcephaly, and high predisposition to malignancies. To explore the underlying molecular mechanisms of NBS microcephaly, Frappart et al. previously inactivated Nbn gene in the central nervous system (CNS) of mice by the nestin-Cre targeting gene system and generated Nbn(CNS-del) mice. Here we first report that Nbn gene inactivation induces the defective proliferation and enhanced apoptosis of the oligodendrocyte precursor cells (OPCs), contributing to the severe hypomyelination of the nerve fibers of the corpus callosum. Under conditions of DNA damage and oxidative stress, the distinct regulatory roles of ATM-Chk2 signaling and AKT/mTOR signaling are responsible for the defective proliferation and enhanced apoptosis of the Nbn-deficient OPCs. In addition, specific HDAC isoforms may play distinctive roles in regulating the myelination of the Nbn-deficient OPCs. However, brain-derived neurotrophic factor and nerve growth factor stimulation attenuates the oxidative stress and thereby increases the proliferation of the Nbn-deficient OPCs, which is accompanied by upregulation of the AKT/mTOR/P70S6K signaling pathway. Taken together, these findings demonstrate that DNA damage and oxidative stress resulting from Nbn gene inactivation are associated with hypomyelination of the nerve fibers of corpus callosum.
Collapse
Affiliation(s)
- B Liu
- Department of Pathology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | |
Collapse
|
17
|
De Paula ML, Cui QL, Hossain S, Antel J, Almazan G. The PTEN inhibitor bisperoxovanadium enhances myelination by amplifying IGF-1 signaling in rat and human oligodendrocyte progenitors. Glia 2013; 62:64-77. [DOI: 10.1002/glia.22584] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Revised: 09/17/2013] [Accepted: 09/18/2013] [Indexed: 11/07/2022]
Affiliation(s)
- Marcio L. De Paula
- Department of Pharmacology and Therapeutics; McGill University, Montreal; Quebec Canada
- Integrated Program in Neuroscience; McGill University, Montreal; Quebec Canada
| | - Qiao-Ling Cui
- Neuroimmunology Unit, Montreal Neurological Institute; McGill University, Montreal; Quebec Canada
| | - Shireen Hossain
- Department of Pharmacology and Therapeutics; McGill University, Montreal; Quebec Canada
| | - Jack Antel
- Integrated Program in Neuroscience; McGill University, Montreal; Quebec Canada
- Neuroimmunology Unit, Montreal Neurological Institute; McGill University, Montreal; Quebec Canada
| | - Guillermina Almazan
- Department of Pharmacology and Therapeutics; McGill University, Montreal; Quebec Canada
- Integrated Program in Neuroscience; McGill University, Montreal; Quebec Canada
| |
Collapse
|
18
|
Cheffer A, Tárnok A, Ulrich H. Cell Cycle Regulation During Neurogenesis in the Embryonic and Adult Brain. Stem Cell Rev Rep 2013; 9:794-805. [DOI: 10.1007/s12015-013-9460-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
19
|
Plemel JR, Manesh SB, Sparling JS, Tetzlaff W. Myelin inhibits oligodendroglial maturation and regulates oligodendrocytic transcription factor expression. Glia 2013; 61:1471-87. [PMID: 23839973 DOI: 10.1002/glia.22535] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Revised: 04/20/2013] [Accepted: 04/26/2013] [Indexed: 12/12/2022]
Abstract
Myelin loss is a hallmark of multiple sclerosis (MS) and promoting central nervous system myelin repair has become a major therapeutic target. Despite the presence of oligodendrocytes precursors cells (OPCs) in chronic lesions of MS, remyelination often fails. The mechanism underlying this failure of remyelination remains unknown, but it is hypothesized that environmental cues act to inhibit the maturation/differentiation of oligodendroglia, preventing remyelination. The rate of CNS remyelination is correlated to the speed of phagocytosis of myelin debris, which is present following demyelination and trauma. Thus, myelin debris could inhibit CNS remyelination. Here, we demonstrate that OPCs cultured on myelin were robustly inhibited in their maturation, as characterized by the decreased expression of immature and mature oligodendrocytes markers, the impaired production of myelin gene products, as well as their stalled morphological complexity relative to OPCs cultured on a control substrate. OPCs in contact with myelin stopped proliferating and decreased the expression of OPC markers to a comparable degree as cells grown on a control substrate. The expression of two transcription factors known to prevent OPC differentiation and maturation were increased in cells that were in contact with myelin: inhibitor of differentiation family (ID) members 2 and 4. Overexpression of ID2 and ID4 in OPCs was previously reported to decrease the percentage of cells expressing mature oligodendrocyte markers. However, knockdown of ID2 and/or ID4 in OPCs did not increase oligodendroglial maturation on or off of myelin, suggesting that contact with myelin regulates additional regulatory elements.
Collapse
Affiliation(s)
- Jason R Plemel
- ICORD (International Collaboration on Repair Discoveries), Blusson Spinal Cord Centre, Vancouver, British Columbia, Canada
| | | | | | | |
Collapse
|
20
|
Huang H, Zhao XF, Zheng K, Qiu M. Regulation of the timing of oligodendrocyte differentiation: mechanisms and perspectives. Neurosci Bull 2013; 29:155-64. [PMID: 23456566 DOI: 10.1007/s12264-013-1314-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 12/08/2012] [Indexed: 12/19/2022] Open
Abstract
Axonal myelination is an essential process for normal functioning of the vertebrate central nervous system. Proper formation of myelin sheaths around axons depends on the timely differentiation of oligodendrocytes. This differentiation occurs on a predictable schedule both in culture and during development. However, the timing mechanisms for oligodendrocyte differentiation during normal development have not been fully uncovered. Recent studies have identified a large number of regulatory factors, including cell-intrinsic factors and extracellular signals, that could control the timing of oligodendrocyte differentiation. Here we provide a mechanistic and critical review of the timing control of oligodendrocyte differentiation.
Collapse
Affiliation(s)
- Hao Huang
- Institute of Developmental and Regenerative Biology, College of Life Sciences, Hangzhou Normal University, Hangzhou 310018, China
| | | | | | | |
Collapse
|
21
|
Beukelaers P, Vandenbosch R, Caron N, Nguyen L, Moonen G, Malgrange B. Cycling or not cycling: cell cycle regulatory molecules and adult neurogenesis. Cell Mol Life Sci 2012; 69:1493-503. [PMID: 22068613 PMCID: PMC11114951 DOI: 10.1007/s00018-011-0880-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 10/10/2011] [Accepted: 10/27/2011] [Indexed: 12/11/2022]
Abstract
The adult brain most probably reaches its highest degree of plasticity with the lifelong generation and integration of new neurons in the hippocampus and olfactory system. Neural precursor cells (NPCs) residing both in the subgranular zone of the dentate gyrus and in the subventricular zone of the lateral ventricles continuously generate neurons that populate the dentate gyrus and the olfactory bulb, respectively. The regulation of NPC proliferation in the adult brain has been widely investigated in the past few years. Yet, the intrinsic cell cycle machinery underlying NPC proliferation remains largely unexplored. In this review, we discuss the cell cycle components that are involved in the regulation of NPC proliferation in both neurogenic areas of the adult brain.
Collapse
Affiliation(s)
- Pierre Beukelaers
- GIGA- Neurosciences, University of Liège, Avenue de l’Hôpital, 1 Bâtiment C.H.U B36, +1, 4000 Liège, Belgium
| | - Renaud Vandenbosch
- GIGA- Neurosciences, University of Liège, Avenue de l’Hôpital, 1 Bâtiment C.H.U B36, +1, 4000 Liège, Belgium
- Present Address: Department of Cellular and Molecular Medicine, University of Ottawa, 451 Smyth Road, Ottawa, ON K1H 8M5 Canada
| | - Nicolas Caron
- GIGA- Neurosciences, University of Liège, Avenue de l’Hôpital, 1 Bâtiment C.H.U B36, +1, 4000 Liège, Belgium
| | - Laurent Nguyen
- GIGA- Neurosciences, University of Liège, Avenue de l’Hôpital, 1 Bâtiment C.H.U B36, +1, 4000 Liège, Belgium
| | - Gustave Moonen
- GIGA- Neurosciences, University of Liège, Avenue de l’Hôpital, 1 Bâtiment C.H.U B36, +1, 4000 Liège, Belgium
- Department of Neurology, C.H.U. Sart Tilman, B35, 4000 Liège, Belgium
| | - Brigitte Malgrange
- GIGA- Neurosciences, University of Liège, Avenue de l’Hôpital, 1 Bâtiment C.H.U B36, +1, 4000 Liège, Belgium
| |
Collapse
|
22
|
Wang PS, Wang J, Zheng Y, Pallen CJ. Loss of protein-tyrosine phosphatase α (PTPα) increases proliferation and delays maturation of oligodendrocyte progenitor cells. J Biol Chem 2012; 287:12529-40. [PMID: 22354965 DOI: 10.1074/jbc.m111.312769] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Tightly controlled termination of proliferation determines when oligodendrocyte progenitor cells (OPCs) can initiate differentiation and mature into myelin-forming cells. Protein-tyrosine phosphatase α (PTPα) promotes OPC differentiation, but its role in proliferation is unknown. Here we report that loss of PTPα enhanced in vitro proliferation and survival and decreased cell cycle exit and growth factor dependence of OPCs but not neural stem/progenitor cells. PTPα(-/-) mice have more oligodendrocyte lineage cells in embryonic forebrain and delayed OPC maturation. On the molecular level, PTPα-deficient mouse OPCs and rat CG4 cells have decreased Fyn and increased Ras, Cdc42, Rac1, and Rho activities, and reduced expression of the Cdk inhibitor p27Kip1. Moreover, Fyn was required to suppress Ras and Rho and for p27Kip1 accumulation, and Rho inhibition in PTPα-deficient cells restored expression of p27Kip1. We propose that PTPα-Fyn signaling negatively regulates OPC proliferation by down-regulating Ras and Rho, leading to p27Kip1 accumulation and cell cycle exit. Thus, PTPα acts in OPCs to limit self-renewal and facilitate differentiation.
Collapse
Affiliation(s)
- Pei-Shan Wang
- Department of Pathology, University of British Columbia, Vancouver, British Columbia V5Z 4H4, Canada
| | | | | | | |
Collapse
|
23
|
Ouyang Y, Song Y, Lu B. dp53 Restrains ectopic neural stem cell formation in the Drosophila brain in a non-apoptotic mechanism involving Archipelago and cyclin E. PLoS One 2011; 6:e28098. [PMID: 22140513 PMCID: PMC3225381 DOI: 10.1371/journal.pone.0028098] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Accepted: 11/01/2011] [Indexed: 02/06/2023] Open
Abstract
Accumulating evidence suggests that tumor-initiating stem cells or cancer stem cells (CSCs) possibly originating from normal stem cells may be the root cause of certain malignancies. How stem cell homeostasis is impaired in tumor tissues is not well understood, although certain tumor suppressors have been implicated. In this study, we use the Drosophila neural stem cells (NSCs) called neuroblasts as a model to study this process. Loss-of-function of Numb, a key cell fate determinant with well-conserved mammalian counterparts, leads to the formation of ectopic neuroblasts and a tumor phenotype in the larval brain. Overexpression of the Drosophila tumor suppressor p53 (dp53) was able to suppress ectopic neuroblast formation caused by numb loss-of-function. This occurred in a non-apoptotic manner and was independent of Dacapo, the fly counterpart of the well-characterized mammalian p53 target p21 involved in cellular senescence. The observation that dp53 affected Edu incorporation into neuroblasts led us to test the hypothesis that dp53 acts through regulation of factors involved in cell cycle progression. Our results show that the inhibitory effect of dp53 on ectopic neuroblast formation was mediated largely through its regulation of Cyclin E (Cyc E). Overexpression of Cyc E was able to abrogate dp53's ability to rescue numb loss-of-function phenotypes. Increasing Cyc E levels by attenuating Archipelago (Ago), a recently identified transcriptional target of dp53 and a negative regulator of Cyc E, had similar effects. Conversely, reducing Cyc E activity by overexpressing Ago blocked ectopic neuroblast formation in numb mutant. Our results reveal an intimate connection between cell cycle progression and NSC self-renewal vs. differentiation control, and indicate that p53-mediated regulation of ectopic NSC self-renewal through the Ago/Cyc E axis becomes particularly important when NSC homeostasis is perturbed as in numb loss-of-function condition. This has important clinical implications.
Collapse
Affiliation(s)
- Yingshi Ouyang
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Yan Song
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
| | - Bingwei Lu
- Department of Pathology, Stanford University School of Medicine, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
24
|
Katsel P, Tan W, Abazyan B, Davis KL, Ross C, Pletnikov MV, Haroutunian V. Expression of mutant human DISC1 in mice supports abnormalities in differentiation of oligodendrocytes. Schizophr Res 2011; 130:238-49. [PMID: 21605958 PMCID: PMC3139741 DOI: 10.1016/j.schres.2011.04.021] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2011] [Revised: 04/18/2011] [Accepted: 04/20/2011] [Indexed: 01/15/2023]
Abstract
Abnormalities in oligodendrocyte (OLG) differentiation and OLG gene expression deficit have been described in schizophrenia (SZ). Recent studies revealed a critical requirement for Disrupted-in-Schizophrenia 1 (DISC1) in neural development. Transgenic mice with forebrain restricted expression of mutant human DISC1 (ΔhDISC1) are characterized by neuroanatomical and behavioral abnormalities reminiscent of some features of SZ. We sought to determine whether the expression of ΔhDISC1 may influence the development of OLGs in this mouse model. OLG- and cell cycle-associated gene and protein expression were characterized in the forebrain of ΔhDISC1 mice during different stages of neurodevelopment (E15 and P1 days) and in adulthood. The results suggest that the expression of ΔhDISC1 exerts a significant influence on oligodendrocyte differentiation and function, evidenced by premature OLG differentiation and increased proliferation of their progenitors. Additional findings showed that neuregulin 1 and its receptors may be contributing factors to the observed upregulation of OLG genes. Thus, OLG function may be perturbed by mutant hDISC1 in a model system that provides new avenues for studying aspects of the pathogenesis of SZ.
Collapse
Affiliation(s)
- Pavel Katsel
- Department of Psychiatry, The Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029-6575, USA.
| | | | | | | | | | | | | |
Collapse
|
25
|
|
26
|
The oligodendrocyte-specific G protein-coupled receptor GPR17 is a cell-intrinsic timer of myelination. Nat Neurosci 2009; 12:1398-406. [PMID: 19838178 PMCID: PMC2783566 DOI: 10.1038/nn.2410] [Citation(s) in RCA: 254] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2009] [Accepted: 08/04/2009] [Indexed: 01/06/2023]
Abstract
The bHLH transcription factor Olig1 promotes oligodendrocyte maturation and is required for myelin repair. In this report, we characterize an Olig1-regulated G-protein coupled receptor GPR17 whose function is to oppose the action of Olig1. GPR17 is restricted to oligodendrocyte lineage cells but downregulated during the peak period of myelination and in adulthood. Transgenic mice with sustained GPR17 expression in oligodendrocytes exhibit stereotypic features of myelinating disorders in the CNS. GPR17 overexpression inhibits oligodendrocyte differentiation and maturation both in vivo and in vitro. Conversely, GPR17 knockout mice display early onset of oligodendrocyte myelination. The opposing action of GPR17 on oligodendrocyte maturation reflects, at least partially, upregulation and nuclear translocation of the potent oligodendrocyte differentiation inhibitors ID2/4. Collectively, these findings suggest that GPR17 orchestrates the transition between immature and myelinating oligodendrocytes via an ID protein-mediated negative regulation, and may serve as a potential therapeutic target for CNS myelin repair.
Collapse
|
27
|
|
28
|
Narayanan SP, Flores AI, Wang F, Macklin WB. Akt signals through the mammalian target of rapamycin pathway to regulate CNS myelination. J Neurosci 2009; 29:6860-70. [PMID: 19474313 PMCID: PMC2757755 DOI: 10.1523/jneurosci.0232-09.2009] [Citation(s) in RCA: 270] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Revised: 03/23/2009] [Accepted: 04/13/2009] [Indexed: 02/08/2023] Open
Abstract
Mammalian target of rapamycin (mTOR), a well known Akt substrate, regulates multiple cellular functions including cell growth and protein synthesis. The current study identifies a novel role of the Akt/mTOR pathway as a regulator of CNS myelination. Previously, we showed that overexpressing constitutively active Akt in oligodendrocytes in a transgenic mouse model induces enhanced CNS myelination, with no changes in the proliferation or survival of oligodendrocyte progenitor or mature cells. The present study focused on the signaling mechanisms regulating this hypermyelination induced by Akt. Activation of mTOR and its downstream substrates (p70S6 kinase and S6 ribosomal protein) was observed in Akt-overexpressing oligodendrocytes. When mTOR signaling was inhibited chronically in vivo with rapamycin starting at 6 weeks of age, the observed hypermyelination was reduced to approximately the amount of myelin seen in wild-type mice. mTOR inhibition had little impact on wild-type myelination between 6 and 12 weeks of age, suggesting that, in normal adults, myelination is relatively complete and is no longer regulated by mTOR signaling. However, when mTOR was chronically inhibited in young adult wild-type mice, myelination was reduced. These results suggest that, during active myelination, the major Akt signal regulating CNS myelination is the mTOR pathway.
Collapse
Affiliation(s)
| | - Ana I. Flores
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Feng Wang
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| | - Wendy B. Macklin
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195
| |
Collapse
|
29
|
Akundi RS, Rivkees SA. Hypoxia alters cell cycle regulatory protein expression and induces premature maturation of oligodendrocyte precursor cells. PLoS One 2009; 4:e4739. [PMID: 19270736 PMCID: PMC2649433 DOI: 10.1371/journal.pone.0004739] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2008] [Accepted: 02/08/2009] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Periventricular white matter injury (PWMI) is a common form of brain injury sustained by preterm infants. A major factor that predisposes to PWMI is hypoxia. Because oligodendrocytes (OLs) are responsible for myelination of axons, abnormal OL development or function may affect brain myelination. At present our understanding of the influences of hypoxia on OL development is limited. To examine isolated effects of hypoxia on OLs, we examined the influences of hypoxia on OL development in vitro. METHODOLOGY/FINDINGS Cultures of oligodendrocyte precursor cells (OPCs) were prepared from mixed glial cultures and were 99% pure. OPCs were maintained at 21% O(2) or hypoxia (1% or 4% O(2)) for up to 7 days. We observed that 1% O(2) lead to an increase in the proportion of myelin basic protein (MBP)-positive OLs after 1 week in culture, and a decrease in the proportion of platelet-derived growth factor receptor alpha (PDGFRalpha)-positive cells suggesting premature OL maturation. Increased expression of the cell cycle regulatory proteins p27(Kip1) and phospho-cdc2, which play a role in OL differentiation, was seen as well. CONCLUSIONS These results show that hypoxia interferes with the normal process of OL differentiation by inducing premature OPC maturation.
Collapse
Affiliation(s)
- Ravi Shankar Akundi
- Yale Child Health Research Center, Section of Developmental Biology & Endocrinology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Scott A. Rivkees
- Yale Child Health Research Center, Section of Developmental Biology & Endocrinology, Yale University School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
30
|
Bockbrader K, Feng Y. Essential function, sophisticated regulation and pathological impact of the selective RNA-binding protein QKI in CNS myelin development. FUTURE NEUROLOGY 2008; 3:655-668. [PMID: 19727426 DOI: 10.2217/14796708.3.6.655] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The selective RNA-binding protein QKI play a key role in advancing oligodendrocyte-dependent myelination, which is essential for the function and development of the CNS. The emerging evidence that QKI abnormalities are associated with schizophrenia and may underlie myelin impairment in this devastating disease has greatly increased interest in understanding the function of QKI. Despite the discovery of the biochemical basis for QKI-RNA interaction, a comprehensive model is currently missing regarding how QKI regulates its mRNA ligands to promote normal myelinogenesis and how deficiency of the QKI pathway is involved in the pathogenesis of human diseases that affect CNS myelin. In this review, we will focus on the role of QKI in regulating distinct mRNA targets at critical developmental steps to promote oligodendrocyte differentiation and myelin formation. In addition, we will discuss molecular mechanisms that control QKI expression and activity during normal myelinogenesis as well as the pathological impact of QKI deficiency in dysmyelination mutant animals and in human myelin disorders.
Collapse
Affiliation(s)
- Katrina Bockbrader
- Department of Pharmacology, Emory University School of Medicine, 1510 Clifton Road, Atlanta, GA 30322, USA, Tel.: +1 404 727 0351, ,
| | | |
Collapse
|
31
|
Sato T, Torashima T, Sugihara K, Hirai H, Asano M, Yoshioka K. The scaffold protein JSAP1 regulates proliferation and differentiation of cerebellar granule cell precursors by modulating JNK signaling. Mol Cell Neurosci 2008; 39:569-78. [DOI: 10.1016/j.mcn.2008.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2008] [Revised: 07/30/2008] [Accepted: 08/10/2008] [Indexed: 12/11/2022] Open
|
32
|
Misumi S, Kim TS, Jung CG, Masuda T, Urakawa S, Isobe Y, Furuyama F, Nishino H, Hida H. Enhanced neurogenesis from neural progenitor cells with G1/S-phase cell cycle arrest is mediated by transforming growth factor beta1. Eur J Neurosci 2008; 28:1049-59. [PMID: 18783370 DOI: 10.1111/j.1460-9568.2008.06420.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
We have previously demonstrated that a G1/S-phase cell cycle blocker, deferoxamine (DFO), increased the number of new neurons from rat neurosphere cultures, which correlated with prolonged expression of cyclin-dependent kinase (cdk) inhibitor p27(kip1) [H. J. Kim et al. (2006)Brain Research, 1092, 1-15]. The present study focuses on neuronal differentiation mechanisms following treatment of neural stem/progenitor cells (NPCs) with a G1/S-phase cell cycle blocker. The addition of DFO (0.5 mm) or aphidicolin (Aph) (1.5 microm) to neurospheres for 8 h, followed by 3 days of differentiation, resulted in an increased number of neurons and neurite outgrowth. DFO induced enhanced expression of transforming growth factor (TGF)-beta1 and cdk5 at 24 h after differentiation, whereas Aph only increased TGF-beta1 expression. DFO-induced neurogenesis and neurite outgrowth were attenuated by administration of a cdk5 inhibitor, roscovitine, suggesting that the neurogenic mechanisms differ between DFO and Aph. TGF-beta1 (10 ng/mL) did not increase neurite outgrowth but rather the number of beta-tubulin III-positive cells, which was accompanied by enhanced p27(kip1) mRNA expression. In addition, TGF-beta receptor type II expression was observed in nestin-positive NPCs. Results indicated that DFO-induced TGF-beta1 signaling activated smad3 translocation from the cytoplasm to the nucleus. In contrast, TGF-beta1 signaling inhibition, via a TGF-beta receptor type I inhibitor (SB-505124), resulted in decreased DFO-induced neurogenesis, in conjunction with decreased p27(kip1) protein expression and smad3 translocation to the nucleus. These results suggest that cell cycle arrest during G1/S-phase induces TGF-beta1 expression. This, in turn, prompts enhanced neuronal differentiation via smad3 translocation to the nucleus and subsequent p27(kip1) activation in NPCs.
Collapse
Affiliation(s)
- Sachiyo Misumi
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Mizuho-ku, Nagoya 467-8601, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Post-transcriptional regulation of myelin formation. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2008; 1779:486-94. [PMID: 18590840 DOI: 10.1016/j.bbagrm.2008.06.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2008] [Revised: 05/15/2008] [Accepted: 06/03/2008] [Indexed: 12/21/2022]
Abstract
Myelin is a specialized structure of the nervous system that both enhances electrical conductance and protects neurons from degeneration. In the central nervous system, extensively polarized oligodendrocytes form myelin by wrapping cellular processes in a spiral pattern around neuronal axons. Myelin formation requires the oligodendrocyte to regulate gene expression in response to changes in its extracellular environment. Because these changes occur at a distance from the cell body, post-transcriptional control of gene expression allows the cell to fine-tune its response. Here, we review the RNA-binding proteins that control myelin formation in the brain, highlighting the molecular mechanisms by which they control gene expression and drawing parallels from studies in other cell types.
Collapse
|
34
|
Shen S, Casaccia-Bonnefil P. Post-translational modifications of nucleosomal histones in oligodendrocyte lineage cells in development and disease. J Mol Neurosci 2008; 35:13-22. [PMID: 17999198 PMCID: PMC2323904 DOI: 10.1007/s12031-007-9014-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The role of epigenetics in modulating gene expression in the development of organs and tissues and in disease states is becoming increasingly evident. Epigenetics refers to the several mechanisms modulating inheritable changes in gene expression that are independent of modifications of the primary DNA sequence and include post-translational modifications of nucleosomal histones, changes in DNA methylation, and the role of microRNA. This review focuses on the epigenetic regulation of gene expression in oligodendroglial lineage cells. The biological effects that post-translational modifications of critical residues in the N-terminal tails of nucleosomal histones have on oligodendroglial cells are reviewed, and the implications for disease and repair are critically discussed.
Collapse
Affiliation(s)
- Siming Shen
- Department Neuroscience and Cell Biology, Robert Wood Johnson Medical School, R-304 Research Tower 675 Hoes Lane, Piscataway, NJ 08854, USA
| | | |
Collapse
|
35
|
Convergence and divergence in the etiology of myelin impairment in psychiatric disorders and drug addiction. Neurochem Res 2008; 33:1940-9. [PMID: 18404371 DOI: 10.1007/s11064-008-9693-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2008] [Accepted: 03/28/2008] [Indexed: 12/30/2022]
Abstract
Impairment of oligodendroglia (OL)-dependent myelination in the central nervous system (CNS) is a remarkable parallel recently identified in major psychiatric disorders and chronic drug abuse. Neuroimaging and neuropathological studies revealed myelin defects and microarray-profiling analysis demonstrated aberrant expression of myelin-related genes in schizophrenia (SZ), bipolar disorder (BD), major depressive disorder (MDD) and cocaine addiction. However, the etiology underlying myelin impairment in these clinically distinct subjects remains elusive. This article reviews myelin impairment in line with dopaminergic dysfunction, a prime neuropathophysiological trait shared in psychiatric disorders and drug abuse, as well as the genetic and epigenetic alterations associated with these diseases. The current findings support the hypothesis that aberrant dopamine (DA) action on OLs is a common pathologic mechanism for myelin impairment in the aforementioned mental morbidities, whereas inherited genetic variations that specifically affect OL development and myelinogenesis may further increase myelin vulnerability in psychiatric disorders. Importantly, OL defect is not only a pathological consequence but also a causative factor for dopaminergic dysfunction. Hence, myelin impairment is a key factor in the pathogenic loop of psychiatric diseases and drug addiction.
Collapse
|
36
|
Jablonska B, Aguirre A, Vandenbosch R, Belachew S, Berthet C, Kaldis P, Gallo V. Cdk2 is critical for proliferation and self-renewal of neural progenitor cells in the adult subventricular zone. ACTA ACUST UNITED AC 2008; 179:1231-45. [PMID: 18086919 PMCID: PMC2140044 DOI: 10.1083/jcb.200702031] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
We investigated the function of cyclin-dependent kinase 2 (Cdk2) in neural progenitor cells during postnatal development. Chondroitin sulfate proteoglycan (NG2)–expressing progenitor cells of the subventricular zone (SVZ) show no significant difference in density and proliferation between Cdk2−/− and wild-type mice at perinatal ages and are reduced only in adult Cdk2−/− mice. Adult Cdk2−/− SVZ cells in culture display decreased self-renewal capacity and enhanced differentiation. Compensatory mechanisms in perinatal Cdk2−/− SVZ cells, which persist until postnatal day 15, involve increased Cdk4 expression that results in retinoblastoma protein inactivation. A subsequent decline in Cdk4 activity to wild-type levels in postnatal day 28 Cdk2−/− cells coincides with lower NG2+ proliferation and self-renewal capacity similar to adult levels. Cdk4 silencing in perinatal Cdk2−/− SVZ cells abolishes Cdk4 up-regulation and reduces cell proliferation and self- renewal to adult levels. Conversely, Cdk4 overexpression in adult SVZ cells restores proliferative capacity to wild-type levels. Thus, although Cdk2 is functionally redundant in perinatal SVZ, it is important for adult progenitor cell proliferation and self-renewal through age-dependent regulation of Cdk4.
Collapse
Affiliation(s)
- Beata Jablonska
- Center for Neuroscience Research, Children's National Medical Center, Washington, DC 20010, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Liu Y, Yeh N, Zhu XH, Leversha M, Cordon-Cardo C, Ghossein R, Singh B, Holland E, Koff A. Somatic cell type specific gene transfer reveals a tumor-promoting function for p21(Waf1/Cip1). EMBO J 2007; 26:4683-93. [PMID: 17948060 PMCID: PMC2048756 DOI: 10.1038/sj.emboj.7601886] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2007] [Accepted: 09/19/2007] [Indexed: 02/08/2023] Open
Abstract
How proteins participate in tumorigenesis can be obscured by their multifunctional nature. For example, depending on the cellular context, the cdk inhibitors can affect cell proliferation, cell motility, apoptosis, receptor tyrosine kinase signaling, and transcription. Thus, to determine how a protein contributes to tumorigenesis, we need to evaluate which functions are required in the developing tumor. Here we demonstrate that the RCAS/TvA system, originally developed to introduce oncogenes into somatic cells of mice, can be adapted to allow us to define the contribution that different functional domains make to tumor development. Studying the development of growth-factor-induced oligodendroglioma, we identified a critical role for the Cy elements in p21, and we showed that cyclin D1T286A, which accumulates in the nucleus of p21-deficient cells and binds to cdk4, could bypass the requirement for p21 during tumor development. These genetic results suggest that p21 acts through the cyclin D1–cdk4 complex to support tumor growth, and establish the utility of using a somatic cell modeling system for defining the contribution proteins make to tumor development.
Collapse
Affiliation(s)
- Yuhui Liu
- Department of Molecular Biology, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
He Y, Dupree J, Wang J, Sandoval J, Li J, Liu H, Shi Y, Nave KA, Casaccia-Bonnefil P. The transcription factor Yin Yang 1 is essential for oligodendrocyte progenitor differentiation. Neuron 2007; 55:217-30. [PMID: 17640524 PMCID: PMC2034312 DOI: 10.1016/j.neuron.2007.06.029] [Citation(s) in RCA: 214] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2006] [Revised: 05/24/2007] [Accepted: 06/21/2007] [Indexed: 11/17/2022]
Abstract
The progression of progenitors to oligodendrocytes requires proliferative arrest and the activation of a transcriptional program of differentiation. While regulation of cell cycle exit has been extensively characterized, the molecular mechanisms responsible for the initiation of differentiation remain ill-defined. Here, we identify the transcription factor Yin Yang 1 (YY1) as a critical regulator of oligodendrocyte progenitor differentiation. Conditional ablation of yy1 in the oligodendrocyte lineage in vivo induces a phenotype characterized by defective myelination, ataxia, and tremor. At the cellular level, lack of yy1 arrests differentiation of oligodendrocyte progenitors after they exit from the cell cycle. At the molecular level, YY1 acts as a lineage-specific repressor of transcriptional inhibitors of myelin gene expression (Tcf4 and Id4), by recruiting histone deacetylase-1 to their promoters during oligodendrocyte differentiation. Thus, we identify YY1 as an essential component of the transcriptional network regulating the transition of oligodendrocyte progenitors from cell cycle exit to differentiation.
Collapse
Affiliation(s)
- Ye He
- Department of Neuroscience and Cell Biology, R. Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Yeh N, Miller JP, Gaur T, Capellini TD, Nikolich-Zugich J, de la Hoz C, Selleri L, Bromage TG, van Wijnen AJ, Stein GS, Lian JB, Vidal A, Koff A. Cooperation between p27 and p107 during endochondral ossification suggests a genetic pathway controlled by p27 and p130. Mol Cell Biol 2007; 27:5161-71. [PMID: 17502351 PMCID: PMC1951950 DOI: 10.1128/mcb.02431-06] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2006] [Revised: 02/21/2007] [Accepted: 05/02/2007] [Indexed: 02/05/2023] Open
Abstract
Pocket proteins and cyclin-dependent kinase (CDK) inhibitors negatively regulate cell proliferation and can promote differentiation. However, which members of these gene families, which cell type they interact in, and what they do to promote differentiation in that cell type during mouse development are largely unknown. To identify the cell types in which p107 and p27 interact, we generated compound mutant mice. These mice were null for p107 and had a deletion in p27 that prevented its binding to cyclin-CDK complexes. Although a fraction of these animals survived into adulthood and looked similar to single p27 mutant mice, a larger number of animals died at birth or within a few weeks thereafter. These animals displayed defects in chondrocyte maturation and endochondral bone formation. Proliferation of chondrocytes was increased, and ectopic ossification was observed. Uncommitted mouse embryo fibroblasts could be induced into the chondrocytic lineage ex vivo, but these cells failed to mature normally. These results demonstrate that p27 carries out overlapping functions with p107 in controlling cell cycle exit during chondrocyte maturation. The phenotypic similarities between p107(-/-) p27(D51/D51) and p107(-/-) p130(-/-) mice and the cells derived from them suggest that p27 and p130 act in an analogous pathway during chondrocyte maturation.
Collapse
Affiliation(s)
- Nancy Yeh
- Sloan-Kettering Institute, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Chen Y, Tian D, Ku L, Osterhout DJ, Feng Y. The selective RNA-binding protein quaking I (QKI) is necessary and sufficient for promoting oligodendroglia differentiation. J Biol Chem 2007; 282:23553-60. [PMID: 17575274 DOI: 10.1074/jbc.m702045200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Quaking I (QKI) is a selective RNA-binding protein essential for myelination of the central nervous system. Three QKI isoforms with distinct C termini and subcellular localization, namely QKI-5, QKI-6, and QKI-7, are expressed in oligodendroglia progenitor cells (OPCs) prior to the initiation of myelin formation and implicated in promoting oligodendrocyte lineage development. However, the functional requirement for each QKI isoform and the mechanisms by which QKI isoforms govern OPC development still remain elusive. We report here that exogenous expression of each QKI isoform is sufficient to enhance differentiation of OPCs with different efficiency, which is abolished by a point mutation that abrogates the RNA binding activity of QKI. Reciprocally, small interfering RNA-mediated QKI knockdown blocks OPC differentiation, which can be partly rescued by QKI-5 and QKI-6 but not by QKI-7, indicating the differential requirement of QKI isoform function in advancing OPC differentiation. Furthermore, we found that abrogation of OPC differentiation, as a result of QKI deficiency, is not due to altered proliferation capacity or cell cycle progression. These results indicate that QKI isoforms are necessary and sufficient for promoting OPC development, which must involve direct influence of QKI on differentiation/maturation of OPCs independent of cell cycle exit, likely via regulating the expression of the target mRNAs of QKI that support OPC differentiation.
Collapse
Affiliation(s)
- Yuntao Chen
- Department of Pharmacology, Emory University School of Medicine, Atlanta, Georgia 30322, USA
| | | | | | | | | |
Collapse
|
41
|
Du Y, Lercher LD, Zhou R, Dreyfus CF. Mitogen-activated protein kinase pathway mediates effects of brain-derived neurotrophic factor on differentiation of basal forebrain oligodendrocytes. J Neurosci Res 2007; 84:1692-702. [PMID: 17044032 DOI: 10.1002/jnr.21080] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Previous studies indicate that brain-derived neurotrophic factor (BDNF), through the mediation of the trkB receptor, modulates the expression of differentiated traits in basal forebrain (BF) oligodendrocytes (OLGs). Specifically, BDNF up-regulates the expression of myelin basic protein (MBP), proteolipid protein (PLP), and myelin associated glycoprotein (MAG; Du et al. [2006] Mol. Cell. Neurosci. 31:366-375). However, the signaling cascades mediating the effects of BDNF have not been defined. The current study employs biochemical and molecular biological approaches to examine the involvements of the mitogen-activated protein kinase (MAPK) pathway, the phosphatidylinositol-3 kinase (PI3K) pathway, and the phospholipase C-gamma (PLC-gamma) pathway. Our results indicate that, in BF OLGs, BDNF activates the MAPK pathway and the PLC-gamma pathway but not the PI3K-Akt signaling cascade. By using specific inhibitors and mutated dominant negative or constitutively active forms of MAPK kinase, we demonstrate that the MAPK pathway is mediating the effects of BDNF on expression of differentiated traits in BF OLGs.
Collapse
Affiliation(s)
- Yangzhou Du
- Department of Neuroscience and Cell Biology, UMDNJ/Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | | | | | |
Collapse
|
42
|
Nguyen L, Borgs L, Vandenbosch R, Mangin JM, Beukelaers P, Moonen G, Gallo V, Malgrange B, Belachew S. The Yin and Yang of cell cycle progression and differentiation in the oligodendroglial lineage. ACTA ACUST UNITED AC 2006; 12:85-96. [PMID: 16807909 DOI: 10.1002/mrdd.20103] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
In white matter disorders such as leukodystrophies (LD), periventricular leucomalacia (PVL), or multiple sclerosis (MS), the hypomyelination or the remyelination failure by oligodendrocyte progenitor cells involves errors in the sequence of events that normally occur during development when progenitors proliferate, migrate through the white matter, contact the axon, and differentiate into myelin-forming oligodendrocytes. Multiple mechanisms underlie the eventual progressive deterioration that typifies the natural history of developmental demyelination in LD and PVL and of adult-onset demyelination in MS. Over the past few years, pathophysiological studies have mostly focused on seeking abnormalities that impede oligodendroglial maturation at the level of migration, myelination, and survival. In contrast, there has been a strikingly lower interest for early proliferative and differentiation events that are likely to be equally critical for white matter development and myelin repair. This review highlights the Yin and Yang principles of interactions between intrinsic factors that coordinately regulate progenitor cell division and the onset of differentiation, i.e. the initial steps of oligodendrocyte lineage progression that are obviously crucial in health and diseases.
Collapse
Affiliation(s)
- Laurent Nguyen
- Developmental Neurobiology Unit, Center for Cellular and Molecular Neuroscience, University of Liège, C.H.U. Sart Tilman, B36, 4000 Liège, Belgium
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Francis JS, Olariu A, McPhee SW, Leone P. Novel role for aspartoacylase in regulation of BDNF and timing of postnatal oligodendrogenesis. J Neurosci Res 2006; 84:151-69. [PMID: 16634055 DOI: 10.1002/jnr.20866] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neuronal growth factors are thought to exert a significant degree of control over postnatal oligodendrogenesis, but mechanisms by which these factors coordinateoligodendrocyte development with the maturation of neural networks are poorly characterized. We present here a developmental analysis of aspartoacylase (Aspa)-null tremor rats and show a potential role for this hydrolytic enzyme in the regulation of a postnatal neurotrophic stimulus that impacts on early stages of oligodendrocyte differentiation. Abnormally high levels of brain-derived neurotrophic factor (BDNF) expression in the Aspa-null Tremor brain are associated with dysregulated oligodendrogenesis at a stage in development normally characterized by high levels of Aspa expression. BDNF promotes the survival of proliferating cells during the early stages of oligodendrocyte maturation in vitro, but seems to compromise the ability of these cells to populate the cortex in vivo. Aspartoacylase activity in oligodendrocytes is shown to provide for the negative regulation of BDNF in neurons, thereby determining the availability of a developmental stimulus via a mechanism that links oligodendroglial differentiation with neuronal maturation.
Collapse
Affiliation(s)
- Jeremy S Francis
- Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey,Camden, NJ, USA
| | | | | | | |
Collapse
|
44
|
Paez PM, Garcia CI, Soto EF, Pasquini JM. Apotransferrin decreases the response of oligodendrocyte progenitors to PDGF and inhibits the progression of the cell cycle. Neurochem Int 2006; 49:359-71. [PMID: 16621163 DOI: 10.1016/j.neuint.2006.02.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2005] [Revised: 02/13/2006] [Accepted: 02/20/2006] [Indexed: 10/24/2022]
Abstract
In the CNS, transferrin (Tf) is expressed by the oligodendroglial cells (OLGcs) and is essential for their development. We have previously shown that apotransferrin (aTf) accelerates maturation of OLGcs in vivo as well as in vitro. The mechanisms involved in this action appear to be complex and have not been completely elucidated. The aim of this study was to investigate if Tf participates in the regulation of the cell cycle of oligodendroglial progenitor cells (OPcs). Primary cultures of OPcs were treated with aTf and/or with different combinations of mitogenic factors. Cell cycle progression was studied by BrdU incorporation, flow cytometry and by the expression of cell cycle regulatory proteins. Apotransferrin decreased the number of BrdU+ cells, increasing the cell cycle time and decreasing the number of cells in S phase. The cell cycle inhibitors p27kip1, p21cip1 and p53 were increased, and in agreement with these results, the activity of the complexes involved in G1-S progression (cyclin D/CDK4, cyclin E/CDK2), was dramatically decreased. Apotransferrin also inhibited the mitogenic effects of PDGF and PDGF/IGF on OPcs, but did not affect their proliferation rate in the presence of bFGF, bFGF/PDGF or bFGF/IGF. Our results indicate that inhibition of the progression of the cell cycle of OPcs by aTf, even in the presence of PDGF, leads to an early beginning of the differentiation program, evaluated by different maturation markers (O4, GC and MBP) and by morphological criteria. The modulation by aTf of the response of OPcs to PDGF supports the idea that this glycoprotein might act as a key regulator of the OLGc lineage progression.
Collapse
Affiliation(s)
- P M Paez
- Instituto de Química y Fisicoquímica Biológica (IQUIFIB), UBA-CONICET, and Departamento de Química Biológica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Junin 956, Buenos Aires C1113AAD, Argentina
| | | | | | | |
Collapse
|
45
|
Kim HJ, Hida H, Jung CG, Miura Y, Nishino H. Treatment with deferoxamine increases neurons from neural stem/progenitor cells. Brain Res 2006; 1092:1-15. [PMID: 16697980 DOI: 10.1016/j.brainres.2006.02.046] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2004] [Revised: 11/24/2005] [Accepted: 02/11/2006] [Indexed: 01/05/2023]
Abstract
Neural transplantation is a promising approach for treating neurodegenerative disease. Neural stem/progenitor cells (NPCs) are self-renewing and multipotent and thus are good candidates for donor cells when they have been clearly defined to differentiate into neurons. As neuronal differentiation follows cell cycle exit, we investigated whether neuron production from NPCs is increased by treatment with cell cycle blockers. NPCs from E12.5 rat ventral mesencephalon were cultured as neurospheres in DMEM/F12 medium containing N2 supplements and bFGF. Treatment of NPCs with deferoxamine, a G1/S phase blocker, increased the number of beta-tubulin III-positive cells after differentiation, concomitant with increases of MAP2 mRNA and protein, and a decrease of GFAP protein. Further, an increase in beta-tubulin III/BrdU double-positive cells and a decrease in GFAP/BrdU double-positive cells were confirmed. In real-time PCR, the expressions of p21(cip1), p27(kip1) and p57(kip2) mRNAs remained unaltered for 8 h after treatment with deferoxamine but were significantly elevated after 1 day. Deferoxamine specifically enhanced the elevation of p27(kip1) mRNA at 1-2 days and the accumulation of p27(kip1) protein at 3 days, along with the activation of neuroD promoter and the elevation of neuroD mRNA. Transfection of p27(kip1) into NPCs induced activation of neuroD promoter and increase of number of beta-tubulin III-positive cells. These data suggest that pretreatment with deferoxamine increases the number of neurons from NPCs related to prolonged p27(kip1) elevation and activation of the neuroD signaling pathway. In this way, regulation of the cell cycle should be a useful first step in engineering NPCs for neural transplantation.
Collapse
Affiliation(s)
- Hye-Jung Kim
- Department of Neurophysiology and Brain Science, Nagoya City University Graduate School of Medical Sciences, Mizuho-ku, Nagoya 467-8601, Japan
| | | | | | | | | |
Collapse
|
46
|
Pirianov G, Jesurasa A, Mehmet H. Developmentally regulated changes in c-Jun N-terminal kinase signalling determine the apoptotic response of oligodendrocyte lineage cells. Cell Death Differ 2006; 13:531-3. [PMID: 16322755 DOI: 10.1038/sj.cdd.4401805] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
47
|
Chew LJ, King WC, Kennedy A, Gallo V. Interferon-gamma inhibits cell cycle exit in differentiating oligodendrocyte progenitor cells. Glia 2005; 52:127-43. [PMID: 15920731 DOI: 10.1002/glia.20232] [Citation(s) in RCA: 89] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The developmental processes of the oligodendrocyte progenitor cell (OPC) lineage that are targeted by interferon-gamma (IFN-gamma) were studied in primary rat OPC cultures. Under conditions of thyroid hormone-mediated oligodendrocyte differentiation, IFN-gamma produced a dose-dependent apoptotic response in OPCs. The lowest dose tested (15 ng/ml or 75 U/ml) was nonapoptotic, but activated detectable STAT1 DNA-binding. At this dose, IFN-gamma reduced the percentage of mature O1+ cells and increased the percentage of immature A2B5+ OPCs. This was observed without significant change in total cell number and cytotoxicity, and was accompanied by an increase in BrdU-labeled A2B5+ and O4+ cells. FACS analysis confirmed a lack of apoptotic sub-G1 cells and revealed a greater percentage of S- and G2/M-phase OPCs with IFN-gamma treatment. Dual immunostaining with Ki-67 and Olig2 showed a smaller percentage of Olig2+ cells in G0 phase in IFN-gamma-treated OPCs, indicating loss of G1 control. Instead, increased levels and phosphorylation of the checkpoint protein p34cdc2 by IFN- suggested increased partial arrest in G2. IFN-gamma not only sustained expression of PCNA and the G1-S regulators retinoblastoma protein, cyclin D1, cyclin E, and cdk2, but also decreased p27 levels. In addition to changes in cell proliferation and differentiation, IFN-gamma attenuated myelin basic protein (MBP) expression significantly, which was associated with decreased expression of both MBP and Sox10 RNAs. These findings indicate that IFN-gamma not only maintains cell cycle activity that could predispose OPCs to apoptosis, but also overrides G1-G0 signals leading to thyroid hormone-mediated terminal differentiation and myelin gene expression.
Collapse
Affiliation(s)
- Li-Jin Chew
- Center for Neuroscience Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA.
| | | | | | | |
Collapse
|
48
|
Bansal R, Marin-Husstege M, Bryant M, Casaccia-Bonnefil P. S-phase entry of oligodendrocyte lineage cells is associated with increased levels of p21Cip1. J Neurosci Res 2005; 80:360-8. [PMID: 15789403 DOI: 10.1002/jnr.20454] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The mechanisms regulating the number of myelinating cells in the central nervous system are crucial for both normal development and repair in pathological conditions. Among relevant growth factors involved in this process, fibroblast growth factor-2 (FGF2) induces oligodendrocyte progenitors (OLPs) to proliferate and stimulates mature oligodendrocytes (OLs) to reenter the S-phase of the cell cycle. S-phase entry is modulated by the formation of complexes between cyclins and cyclin-dependent kinases (CDKs), on one hand, and by their interactions with cell cycle inhibitors (e.g., p18INK, p27Kip1, p21Cip1), on the other. Although the roles of cyclin E/CDK2 complexes and the inhibitor p27Kip1 have been extensively investigated relative to proliferation and differentiation in the OL lineage, less is known about the regulation of the formation of cyclin D1/CDK4 complexes and the role of p21Cip1 in these events. In this study, we show that the FGF2-mediated increase in bromodeoxyuridine (BrdU) incorporation into OL progenitors and mature OLs occurs concomitantly with increase in the levels of p21Cip1 and the formation of p21Cip1/cyclin D1/CDK4 ternary complexes. These complexes are functionally active is indicated by the ensuing FGF2-dependent hyperphosphorylation of the downstream target Rb. In untreated mature OLs that do not incorporate BrdU, the levels of p21Cip1 are low, and the level of the inhibitor p18INK is high. Furthermore, p18INK sequesters CDK2 into binary complexes, precluding the formation of p21Cip1/cyclin D1/CDK4 ternary complexes in these cells. Therefore, we propose that p21Cip1 is acting as a positive regulator, rather than an inhibitor, of cell cycle entry by favoring the assembly of active cyclin D1/CDK4 complexes.
Collapse
Affiliation(s)
- Rashmi Bansal
- Department of Neuroscience, University of Connecticut Medical School, Farmington, Connecticut 06030, USA.
| | | | | | | |
Collapse
|
49
|
Bryja V, Cajánek L, Pacherník J, Hall AC, Horváth V, Dvorák P, Hampl A. Abnormal development of mouse embryoid bodies lacking p27Kip1 cell cycle regulator. Stem Cells 2005; 23:965-74. [PMID: 15941856 DOI: 10.1634/stemcells.2004-0174] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cultures of three-dimensional aggregates of embryonic stem cells (ESCs) called embryoid bodies (EBs) provide a valuable system for analyzing molecular mechanisms that regulate differentiation of this unique cell type. Cyclin-dependent kinase inhibitor p27Kip1 (p27) becomes elevated during the differentiation of mouse ESCs (mESCs). In this study, various aspects of differentiation of EBs produced from normal and p27-deficient mESCs were analyzed to address the biological significance of this elevation. It was found that EBs lacking p27 grew significantly bigger, but this was not accompanied by detect-able abnormalities in the activities of cyclin-dependent kinases (CDKs). In most EB cells, downregulation of activating cyclins rather than upregulation of inhibiting p27 is probably responsible for lowering the activity of their CDKs. Abnormalities in the development of specific cell lineages were also observed in p27-deficient EBs. These included elimination of cells positive for cytokeratin endo-A (TROMA-I) and increased proliferation and formation of cavities originating from cells positive for Lewis-X. Our data also suggest that although two different pools of Lewis-X-expressing cells, cluster forming (ESC-like) and cavity forming (neural progenitors), normally exist in EBs, the absence of p27 leads to the enhancement of only the neural pool. No failure was found when the neurogenic capacity of p27-deficient mESCs was tested using various protein markers. Together, our data point to a dual role of p27 in mESCs, with one role being in the regulation of proliferation and the other role in establishing some other aspects of a differentiated phenotype.
Collapse
Affiliation(s)
- Vítezslav Bryja
- Laboratory of Molecular Embryology, Mendel University Brno, Zemedelská1, 61300 Brno, Czech Republic
| | | | | | | | | | | | | |
Collapse
|
50
|
Shen S, Li J, Casaccia-Bonnefil P. Histone modifications affect timing of oligodendrocyte progenitor differentiation in the developing rat brain. ACTA ACUST UNITED AC 2005; 169:577-89. [PMID: 15897262 PMCID: PMC2171688 DOI: 10.1083/jcb.200412101] [Citation(s) in RCA: 248] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Timely differentiation of progenitor cells is critical for development. In this study we asked whether global epigenetic mechanisms regulate timing of progenitor cell differentiation into myelin-forming oligodendrocytes in vivo. Histone deacetylation was essential during a specific temporal window of development and was dependent on the enzymatic activity of histone deacetylases, whose expression was detected in the developing corpus callosum. During the first 10 postnatal days, administration of valproic acid (VPA), the specific inhibitor for histone deacetylase activity, resulted in significant hypomyelination with delayed expression of late differentiation markers and retained expression of progenitor markers. Differentiation resumed in VPA-injected rats if a recovery period was allowed. Administration of VPA after myelination onset had no effect on myelin gene expression and was consistent with changes of nucleosomal histones from reversible deacetylation to more stable methylation and chromatin compaction. Together, these data identify global modifications of nucleosomal histones critical for timing of oligodendrocyte differentiation and myelination in the developing corpus callosum.
Collapse
Affiliation(s)
- Siming Shen
- Department of Neuroscience and Cell Biology, R. Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | | | |
Collapse
|