1
|
Abasi M, Kianmehr A, Variji A, Sangali P, Mahrooz A. microRNAs as molecular tools for brain health: Neuroprotective potential in neurodegenerative disorders. Neuroscience 2025; 574:83-103. [PMID: 40210196 DOI: 10.1016/j.neuroscience.2025.04.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2024] [Revised: 03/09/2025] [Accepted: 04/05/2025] [Indexed: 04/12/2025]
Abstract
As research on microRNAs (miRNAs) advances, it is becoming increasingly clear that these small molecules play crucial roles in the central nervous system (CNS). They are involved in various essential neuronal functions, with specific miRNAs preferentially expressed in different cell types within the nervous system. Notably, certain miRNAs are found at higher levels in the brain and spinal cord compared to other tissues, suggesting they may have specialized functions in the CNS. miRNAs associated with long-term neurodegenerative changes could serve as valuable tools for early treatment decisions and disease monitoring. The significance of miRNAs such as miR-320, miR-146 and miR-29 in the early diagnosis of neurodegenerative disorders becomes evident, especially considering that many neurological and physical symptoms manifest only after substantial degeneration of specific neurons. Interestingly, serum miRNA levels such as miR-92 and miR-486 may correlate with various MRI parameters in multiple sclerosis. Targeting miRNAs using antisense strategies, such as antisense miR-146 and miR-485, may provide advantages over targeting mRNAs, as a single anti-miRNA can regulate multiple disease-related genes. In the future, anti-miRNA-based therapeutic approaches could be integrated into the clinical management of neurological diseases. Certain miRNAs, including miR-223, miR-106, miR-181, and miR-146, contribute to the pathogenesis of various neurodegenerative diseases and thus warrant greater attention. This knowledge could pave the way for the identification of new diagnostic, prognostic, and theranostic biomarkers, and potentially guiding the development of RNA-based therapeutic strategies. This review highlights recent research on the roles of miRNAs in the nervous system, particularly their protective functions in neurodegenerative disorders.
Collapse
Affiliation(s)
- Mozhgan Abasi
- Department of Tissue Engineering and Applied Cell Sciences, Faculty of Advanced Technologies in Medicine, Mazandaran University of Medical Sciences, Sari, Iran; Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Anvarsadat Kianmehr
- Medical Cellular and Molecular Research Center, Golestan University of Medical Sciences, Gorgan, Iran
| | - Athena Variji
- Adelaide Medical School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, Australia
| | - Parisa Sangali
- Department of Clinical Biochemistry and Medical Genetics, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Abdolkarim Mahrooz
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
2
|
Shidlovskii YV, Ulianova YA, Shaposhnikov AV, Kolesnik VV, Pravednikova AE, Stepanov NG, Chetverina D, Saccone G, Lebedeva LA, Chmykhalo VK, Giordano E. Subunits Med12 and Med13 of Mediator Cooperate with Subunits SAYP and Bap170 of SWI/SNF in Active Transcription in Drosophila. Int J Mol Sci 2024; 25:12781. [PMID: 39684492 DOI: 10.3390/ijms252312781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/21/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
SAYP and Bap170, subunits of the SWI/SNF remodeling complex, have the ability to support enhancer-dependent transcription when artificially recruited to the promoter on a transgene. We found that the phenomenon critically depends on two subunits of the Mediator kinase module, Med12 and Med13 but does not require the two other subunits of the module (Cdk8 and CycC) or other subunits of the core part of the complex. A cooperation of the above proteins in active transcription was also observed at endogenous loci, but the contribution of the subunits to the activity of a particular gene differed in different loci. The factors SAYP/Bap170 and Med12/Med13 did not form sufficiently stable interactions in the extract, and their cooperation was apparently local at regulatory elements, the presence of SAYP and Bap170 in a locus being necessary for stable recruitment of Med12 and Med13 to the locus. In addition to the above factors, the Nelf-A protein was found to participate in the process. The cooperation of the factors, independent of enzymatic activities of the complexes they are part of, appears to be a novel mechanism that maintains promoter activity and may be used in many loci of the genome. Extended intrinsically disordered regions of the factors were assumed to sustain the mechanism.
Collapse
Affiliation(s)
- Yulii V Shidlovskii
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Department of Biology and General Genetics, Sechenov University, 119992 Moscow, Russia
| | - Yulia A Ulianova
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Alexander V Shaposhnikov
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Valeria V Kolesnik
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Anna E Pravednikova
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Nikita G Stepanov
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Department of Biology and General Genetics, Sechenov University, 119992 Moscow, Russia
| | - Darya Chetverina
- Group of Epigenetics, Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov St., 119334 Moscow, Russia
| | - Giuseppe Saccone
- Department of Biology, University of Naples "Federico II", 80126 Naples, Italy
| | - Lyubov A Lebedeva
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Victor K Chmykhalo
- Laboratory of Gene Expression Regulation in Development, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Ennio Giordano
- Department of Biology, University of Naples "Federico II", 80126 Naples, Italy
| |
Collapse
|
3
|
Ilchuk LA, Kubekina MV, Okulova YD, Silaeva YY, Tatarskiy VV, Filatov MA, Bruter AV. Genetically Engineered Mice Unveil In Vivo Roles of the Mediator Complex. Int J Mol Sci 2023; 24:ijms24119330. [PMID: 37298278 DOI: 10.3390/ijms24119330] [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: 04/28/2023] [Revised: 05/16/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
The Mediator complex is a multi-subunit protein complex which plays a significant role in the regulation of eukaryotic gene transcription. It provides a platform for the interaction of transcriptional factors and RNA polymerase II, thus coupling external and internal stimuli with transcriptional programs. Molecular mechanisms underlying Mediator functioning are intensively studied, although most often using simple models such as tumor cell lines and yeast. Transgenic mouse models are required to study the role of Mediator components in physiological processes, disease, and development. As constitutive knockouts of most of the Mediator protein coding genes are embryonically lethal, conditional knockouts and corresponding activator strains are needed for these studies. Recently, they have become more easily available with the development of modern genetic engineering techniques. Here, we review existing mouse models for studying the Mediator, and data obtained in corresponding experiments.
Collapse
Affiliation(s)
- Leonid A Ilchuk
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Marina V Kubekina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Yulia D Okulova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Yulia Yu Silaeva
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia
| | - Victor V Tatarskiy
- Institute of Gene Biology, Russian Academy of Sciences, 34/5 Vavilov Street, 119334 Moscow, Russia
| | - Maxim A Filatov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
| | - Alexandra V Bruter
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Gene Biology, Russian Academy of Sciences, 119334 Moscow, Russia
- Federal State Budgetary Institution "N.N. Blokhin National Medical Research Center of Oncology", Ministry of Health of the Russian Federation, Kashirskoe Sh. 24, 115478 Moscow, Russia
| |
Collapse
|
4
|
Samtani G, Kim S, Michaud D, Hillhouse AE, Szule JA, Konganti K, Li J. Brain region dependent molecular signatures and myelin repair following chronic demyelination. Front Cell Neurosci 2023; 17:1169786. [PMID: 37180951 PMCID: PMC10171432 DOI: 10.3389/fncel.2023.1169786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 04/03/2023] [Indexed: 05/16/2023] Open
Abstract
Multiple sclerosis (MS) is the most prevalent demyelinating disease of the central nervous system, characterized by myelin destruction, axonal degeneration, and progressive loss of neurological functions. Remyelination is considered an axonal protection strategy and may enable functional recovery, but the mechanisms of myelin repair, especially after chronic demyelination, remain poorly understood. Here, we used the cuprizone demyelination mouse model to investigate spatiotemporal characteristics of acute and chronic de- and remyelination and motor functional recovery following chronic demyelination. Extensive remyelination occurred after both the acute and chronic insults, but with less robust glial responses and slower myelin recovery in the chronic phase. Axonal damage was found at the ultrastructural level in the chronically demyelinated corpus callosum and in remyelinated axons in the somatosensory cortex. Unexpectedly, we observed the development of functional motor deficits after chronic remyelination. RNA sequencing of isolated brain regions revealed significantly altered transcripts across the corpus callosum, cortex and hippocampus. Pathway analysis identified selective upregulation of extracellular matrix/collagen pathways and synaptic signaling in the chronically de/remyelinating white matter. Our study demonstrates regional differences of intrinsic reparative mechanisms after a chronic demyelinating insult and suggests a potential link between long-term motor function alterations and continued axonal damage during chronic remyelination. Moreover, the transcriptome dataset of three brain regions and over an extended de/remyelination period provides a valuable platform for a better understanding of the mechanisms of myelin repair as well as the identification of potential targets for effective remyelination and neuroprotection for progressive MS.
Collapse
Affiliation(s)
- Grace Samtani
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, United States
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, United States
| | - Sunja Kim
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, United States
| | - Danielle Michaud
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, United States
| | - Andrew E. Hillhouse
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, TX, United States
| | - Joseph A. Szule
- Department of Veterinary Pathobiology, Texas A&M University, College Station, TX, United States
| | - Kranti Konganti
- Texas A&M Institute for Genome Sciences and Society, Texas A&M University, College Station, TX, United States
| | - Jianrong Li
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, United States
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX, United States
| |
Collapse
|
5
|
Bernhardt C, Sock E, Fröb F, Hillgärtner S, Nemer M, Wegner M. KLF9 and KLF13 transcription factors boost myelin gene expression in oligodendrocytes as partners of SOX10 and MYRF. Nucleic Acids Res 2022; 50:11509-11528. [PMID: 36318265 PMCID: PMC9723594 DOI: 10.1093/nar/gkac953] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 10/06/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
Differentiated oligodendrocytes produce myelin and thereby ensure rapid nerve impulse conduction and efficient information processing in the vertebrate central nervous system. The Krüppel-like transcription factor KLF9 enhances oligodendrocyte differentiation in culture, but appears dispensable in vivo. Its mode of action and role within the oligodendroglial gene regulatory network are unclear. Here we show that KLF9 shares its expression in differentiating oligodendrocytes with the closely related KLF13 protein. Both KLF9 and KLF13 bind to regulatory regions of genes that are important for oligodendrocyte differentiation and equally recognized by the central differentiation promoting transcription factors SOX10 and MYRF. KLF9 and KLF13 physically interact and synergistically activate oligodendrocyte-specific regulatory regions with SOX10 and MYRF. Similar to KLF9, KLF13 promotes differentiation and myelination in primary oligodendroglial cultures. Oligodendrocyte differentiation is also altered in KLF13-deficient mice as demonstrated by a transiently reduced myelin gene expression during the first postnatal week. Considering mouse phenotypes, the similarities in expression pattern and genomic binding and the behaviour in functional assays, KLF9 and KLF13 are important and largely redundant components of the gene regulatory network in charge of oligodendrocyte differentiation and myelination.
Collapse
Affiliation(s)
- Celine Bernhardt
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Elisabeth Sock
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Franziska Fröb
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Simone Hillgärtner
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Mona Nemer
- Molecular Genetics and Cardiac Regeneration Laboratory, Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, Canada
| | - Michael Wegner
- To whom correspondence should be addressed. Tel: +49 9131 85 24620; Fax: +49 9131 85 22484;
| |
Collapse
|
6
|
Comparative role of SOX10 gene in the gliogenesis of central, peripheral, and enteric nervous systems. Differentiation 2022; 128:13-25. [DOI: 10.1016/j.diff.2022.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 09/10/2022] [Accepted: 09/19/2022] [Indexed: 11/17/2022]
|
7
|
Venigalla S, Straub J, Idigo O, Rinderle C, Stephens JM, Newman JJ. MED12 Regulates Human Adipose-Derived Stem Cell Adipogenesis and Mediator Kinase Subunit Expression in Murine Adipose Depots. Stem Cells Dev 2022; 31:119-131. [PMID: 35018809 PMCID: PMC9206493 DOI: 10.1089/scd.2021.0302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The mediator kinase module plays a critical role in the regulation of transcription during metabolic processes. Here we demonstrate that in human adipose-derived stem cells (hASCs), kinase module subunits have distinct mRNA and protein expression profiles during different stages of adipogenesis. In addition, siRNA-mediated loss of MED12 results in decreased adipogenesis as evident through decreased lipid accumulation and decreased expression of PPARγ, a master regulator of adipogenesis. Moreover, the decrease in adipogenesis and reduced PPARγ expression are observed only during the early stages of MED12 knockdown. At later stages, knockdown of MED12 did not have any significant effects on adipogenesis or PPARγ expression. We also observed that MED12 was present in a protein complex with PPARγ and C/EBPα during all stages of adipogenesis in hASCs. In 3T3-L1 preadipocytes and adipocytes, MED12 is present in protein complexes with PPARγ1, C/EBPα, and STAT5A. CDK8, another member of the kinase module, was only found to interact with C/EBPα. We found that the expression of all kinase module subunits decreased in inguinal, gonadal, and retroperitoneal white adipose tissue (WAT) depots in the fed state after an overnight fast, whereas the expression of kinase module subunits remained consistent in mesenteric WAT (mWAT) and brown adipose tissue. These data demonstrate that the kinase module undergoes physiologic regulation during fasting and feeding in specific mouse adipose tissue depots, and that MED12 likely plays a specific role in initiating and maintaining adipogenesis.
Collapse
Affiliation(s)
- Sree Venigalla
- School of Biological Sciences, Louisiana Tech University, Ruston, Louisiana, USA
| | - Joseph Straub
- School of Biological Sciences, Louisiana Tech University, Ruston, Louisiana, USA
| | - Onyekachi Idigo
- School of Biological Sciences, Louisiana Tech University, Ruston, Louisiana, USA
| | - Caroline Rinderle
- School of Biological Sciences, Louisiana Tech University, Ruston, Louisiana, USA
| | | | - Jamie J. Newman
- School of Biological Sciences, Louisiana Tech University, Ruston, Louisiana, USA.,Address correspondence to: Dr. Jamie J. Newman, School of Biological Sciences, Louisiana Tech University, Ruston, LA 71272, USA
| |
Collapse
|
8
|
Sock E, Wegner M. Using the lineage determinants Olig2 and Sox10 to explore transcriptional regulation of oligodendrocyte development. Dev Neurobiol 2021; 81:892-901. [PMID: 34480425 DOI: 10.1002/dneu.22849] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/30/2021] [Accepted: 08/31/2021] [Indexed: 02/02/2023]
Abstract
The transcription factors Olig2 and Sox10 jointly define oligodendroglial identity. Because of their continuous presence during development and in the differentiated state they shape the oligodendroglial regulatory network at all times. In this review, we exploit their eminent role and omnipresence to elaborate the central principles that organize the gene regulatory network in oligodendrocytes in such a way that it preserves its identity, but at the same time allows defined and stimulus-dependent changes that result in an ordered lineage progression, differentiation, and myelination. For this purpose, we outline the multiple functional and physical interactions and intricate cross-regulatory relationships with other transcription factors, such as Hes5, Id, and SoxD proteins, in oligodendrocyte precursors and Tcf7l2, Sip1, Nkx2.2, Zfp24, and Myrf during differentiation and myelination, and interpret them in the context of the regulatory network.
Collapse
Affiliation(s)
- Elisabeth Sock
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Wegner
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
9
|
Huang TW, Iyer AA, Manalo JM, Woo J, Bosquez Huerta NA, McGovern MM, Schrewe H, Pereira FA, Groves AK, Ohlemiller KK, Deneen B. Glial-Specific Deletion of Med12 Results in Rapid Hearing Loss via Degradation of the Stria Vascularis. J Neurosci 2021; 41:7171-7181. [PMID: 34253626 PMCID: PMC8387121 DOI: 10.1523/jneurosci.0070-21.2021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 06/11/2021] [Accepted: 07/05/2021] [Indexed: 11/21/2022] Open
Abstract
Mediator protein complex subunit 12 (Med12) is a core component of the basal transcriptional apparatus and plays a critical role in the development of many tissues. Mutations in Med12 are associated with X-linked intellectual disability syndromes and hearing loss; however, its role in nervous system function remains undefined. Here, we show that temporal conditional deletion of Med12 in astrocytes in the adult CNS results in region-specific alterations in astrocyte morphology. Surprisingly, behavioral studies revealed rapid hearing loss after adult deletion of Med12 that was confirmed by a complete abrogation of auditory brainstem responses. Cellular analysis of the cochlea revealed degeneration of the stria vascularis, in conjunction with disorganization of basal cells adjacent to the spiral ligament and downregulation of key cell adhesion proteins. Physiologic analysis revealed early changes in endocochlear potential, consistent with strial-specific defects. Together, our studies reveal that Med12 regulates auditory function in the adult by preserving the structural integrity of the stria vascularis.SIGNIFICANCE STATEMENT Mutations in Mediator protein complex subunit 12 (Med12) are associated with X-linked intellectual disability syndromes and hearing loss. Using temporal-conditional genetic approaches in CNS glia, we found that loss of Med12 results in severe hearing loss in adult animals through rapid degeneration of the stria vascularis. Our study describes the first animal model that recapitulates hearing loss identified in Med12-related disorders and provides a new system in which to examine the underlying cellular and molecular mechanisms of Med12 function in the adult nervous system.
Collapse
Affiliation(s)
- Teng-Wei Huang
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas 77030
| | - Amrita A Iyer
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
- Program in Genetics & Genomics, Baylor College of Medicine, Houston, Texas 77030
| | - Jeanne M Manalo
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas 77030
| | - Junsung Woo
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas 77030
| | - Navish A Bosquez Huerta
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas 77030
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030
| | - Melissa M McGovern
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
| | - Heinrich Schrewe
- Department of Developmental Genetics, Max Planck Institute for Molecular Genetics, 14195 Berlin, Germany
| | - Fredrick A Pereira
- Huffington Center on Aging, Baylor College of Medicine, Houston, Texas 77030
- Department of Otolaryngology, Baylor College of Medicine, Houston, Texas 77030
| | - Andrew K Groves
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030
- Program in Genetics & Genomics, Baylor College of Medicine, Houston, Texas 77030
| | - Kevin K Ohlemiller
- Department of Otolaryngolgy, Central Institute for the Deaf, Fay and Carl Simons Center for Biology of Hearing and Deafness, Washington University in St. Louis, St. Louis, Missouri 63110
| | - Benjamin Deneen
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, Texas 77030
- Department of Neuroscience, Baylor College of Medicine, Houston, Texas 77030
- Program in Developmental Biology, Baylor College of Medicine, Houston, TX 77030
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas 77030
| |
Collapse
|
10
|
Chen X, Lin B, Luo M, Chu W, Li P, Liu H, Xi Z, Fan R. Identifying circRNA- and lncRNA-associated-ceRNA networks in the hippocampi of rats exposed to PM 2.5 using RNA-seq analysis. Genomics 2020; 113:193-204. [PMID: 33338629 DOI: 10.1016/j.ygeno.2020.12.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 12/04/2020] [Accepted: 12/13/2020] [Indexed: 11/16/2022]
Abstract
Non-coding RNAs appear to be involved in the regulation of the nervous system. However, no competing endogenous RNA (ceRNA) network related to PM2.5 damage in the hippocampal function has yet been constructed. Herein, we used whole-transcriptome sequencing technology to systematically study the ceRNA network in rat hippocampi after PM2.5 exposure. We identified 100 circRNAs, 67 lncRNAs, 28 miRNAs, and 539 mRNAs and constructed the most comprehensive ceRNA network to date, to our knowledge. Gene Ontology and KEGG analyses showed that the network molecules are involved in synapses, neural projections, and neural development and involve signal pathways such as the synaptic vesicle cycle. Finally, the expression of the differentially expressed RNAs confirmed by quantitative real-time PCR was consistent with the sequencing data. This study systematically dissected the ceRNA atlas related to cognitive memory function in the hippocampal tissue of PM2.5-exposed rats for the first time, to our knowledge, and promotes the development of potential new treatments for cognitive impairment.
Collapse
Affiliation(s)
- Xuewei Chen
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Bencheng Lin
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Mingzhu Luo
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Chengdu University of Traditional Chinese Medicine, Chengdu 611137, Sichuan, China
| | - Wenbin Chu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China
| | - Ping Li
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Huanliang Liu
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Zhuge Xi
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China.
| | - Rong Fan
- Tianjin Institute of Environmental and Operational Medicine, Tianjin 300050, China; Central laboratory, Tianjin Xiqing Hospital, Tianjin 300380, China.
| |
Collapse
|
11
|
Straub J, Venigalla S, Newman JJ. Mediator's Kinase Module: A Modular Regulator of Cell Fate. Stem Cells Dev 2020; 29:1535-1551. [PMID: 33161841 DOI: 10.1089/scd.2020.0164] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Selective gene expression is crucial in maintaining the self-renewing and multipotent properties of stem cells. Mediator is a large, evolutionarily conserved, multi-subunit protein complex that modulates gene expression by relaying signals from cell type-specific transcription factors to RNA polymerase II. In humans, this complex consists of 30 subunits arranged in four modules. One critical module of the Mediator complex is the kinase module consisting of four subunits: MED12, MED13, CDK8, and CCNC. The kinase module exists in variable association with the 26-subunit Mediator core and affects transcription through phosphorylation of transcription factors and by controlling Mediator structure and function. Many studies have shown the kinase module to be a key player in the maintenance of stem cells that is distinct from a general role in transcription. Genetic studies have revealed that dysregulation of this kinase subunit contributes to the development of many human diseases. In this review, we discuss the importance of the Mediator kinase module by examining how this module functions with the more recently identified transcriptional super-enhancers, how changes in the kinase module and its activity can lead to the development of human disease, and the role of this unique module in directing and maintaining cell state. As we look to use stem cells to understand human development and treat human disease through both cell-based therapies and tissue engineering, we need to remain aware of the on-going research and address critical gaps in knowledge related to the molecular mechanisms that control cell fate.
Collapse
Affiliation(s)
- Joseph Straub
- School of Biological Sciences, Louisiana Tech University, Ruston, Louisiana, USA
| | - Sree Venigalla
- School of Biological Sciences, Louisiana Tech University, Ruston, Louisiana, USA
| | - Jamie J Newman
- School of Biological Sciences, Louisiana Tech University, Ruston, Louisiana, USA
| |
Collapse
|
12
|
SOX10-regulated promoter use defines isoform-specific gene expression in Schwann cells. BMC Genomics 2020; 21:549. [PMID: 32770939 PMCID: PMC7430845 DOI: 10.1186/s12864-020-06963-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/29/2020] [Indexed: 01/12/2023] Open
Abstract
Background Multicellular organisms adopt various strategies to tailor gene expression to cellular contexts including the employment of multiple promoters (and the associated transcription start sites (TSSs)) at a single locus that encodes distinct gene isoforms. Schwann cells—the myelinating cells of the peripheral nervous system (PNS)—exhibit a specialized gene expression profile directed by the transcription factor SOX10, which is essential for PNS myelination. SOX10 regulates promoter elements associated with unique TSSs and gene isoforms at several target loci, implicating SOX10-mediated, isoform-specific gene expression in Schwann cell function. Here, we report on genome-wide efforts to identify SOX10-regulated promoters and TSSs in Schwann cells to prioritize genes and isoforms for further study. Results We performed global TSS analyses and mined previously reported ChIP-seq datasets to assess the activity of SOX10-bound promoters in three models: (i) an adult mammalian nerve; (ii) differentiating primary Schwann cells, and (iii) cultured Schwann cells with ablated SOX10 function. We explored specific characteristics of SOX10-dependent TSSs, which provides confidence in defining them as SOX10 targets. Finally, we performed functional studies to validate our findings at four previously unreported SOX10 target loci: ARPC1A, CHN2, DDR1, and GAS7. These findings suggest roles for the associated SOX10-regulated gene products in PNS myelination. Conclusions In sum, we provide comprehensive computational and functional assessments of SOX10-regulated TSS use in Schwann cells. The data presented in this study will stimulate functional studies on the specific mRNA and protein isoforms that SOX10 regulates, which will improve our understanding of myelination in the peripheral nerve.
Collapse
|
13
|
Bagheri H, Friedman H, Siminovitch KA, Peterson AC. Transcriptional regulators of the Golli/myelin basic protein locus integrate additive and stealth activities. PLoS Genet 2020; 16:e1008752. [PMID: 32790717 PMCID: PMC7446974 DOI: 10.1371/journal.pgen.1008752] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 08/25/2020] [Accepted: 07/01/2020] [Indexed: 11/19/2022] Open
Abstract
Myelin is composed of plasma membrane spirally wrapped around axons and compacted into dense sheaths by myelin-associated proteins. Myelin is elaborated by neuroepithelial derived oligodendrocytes in the central nervous system (CNS) and by neural crest derived Schwann cells in the peripheral nervous system (PNS). While some myelin proteins accumulate in only one lineage, myelin basic protein (Mbp) is expressed in both. Overlapping the Mbp gene is Golli, a transcriptional unit that is expressed widely both within and beyond the nervous system. A super-enhancer domain within the Golli/Mbp locus contains multiple enhancers shown previously to drive reporter construct expression specifically in oligodendrocytes or Schwann cells. In order to determine the contribution of each enhancer to the Golli/Mbp expression program, and to reveal if functional interactions occur among them, we derived mouse lines in which they were deleted, either singly or in different combinations, and relative mRNA accumulation was measured at key stages of early development and at maturity. Although super-enhancers have been shown previously to facilitate interaction among their component enhancers, the enhancers investigated here demonstrated largely additive relationships. However, enhancers demonstrating autonomous activity strictly in one lineage, when missing, were found to significantly reduce output in the other, thus revealing cryptic "stealth" activity. Further, in the absence of a key oligodendrocyte enhancer, Golli accumulation was markedly and uniformly attenuated in all cell types investigated. Our observations suggest a model in which enhancer-mediated DNA-looping and potential super-enhancer properties underlie Golli/Mbp regulatory organization.
Collapse
Affiliation(s)
- Hooman Bagheri
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Hana Friedman
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Katherine A. Siminovitch
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Mount Sinai Hospital, Lunenfeld-Tanenbaum and Toronto General Hospital Research Institutes, Toronto, Ontario, Canada
| | - Alan C. Peterson
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, Quebec, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
14
|
Parras C, Marie C, Zhao C, Lu QR. Chromatin remodelers in oligodendroglia. Glia 2020; 68:1604-1618. [PMID: 32460418 DOI: 10.1002/glia.23837] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 04/09/2020] [Accepted: 04/16/2020] [Indexed: 01/09/2023]
Abstract
Oligodendrocytes, the myelinating cells in the vertebrate central nervous system, produce myelin sheaths to enable saltatory propagation of action potentials. The process of oligodendrocyte myelination entails a stepwise progression from precursor specification to differentiation, which is coordinated by a series of transcriptional and chromatin remodeling events. ATP-dependent chromatin remodeling enzymes, which utilize ATP as an energy source to control chromatin dynamics and regulate the accessibility of chromatin to transcriptional regulators, are critical for oligodendrocyte lineage development and regeneration. In this review, we focus on the latest insights into the spatial and temporal specificity of chromatin remodelers during oligodendrocyte development, myelinogenesis, and regeneration. We will also bring together various plausible mechanisms by which lineage specific transcriptional regulators coordinate with chromatin remodeling factors for programming genomic landscapes to specifically modulate these different processes during developmental myelination and remyelination upon injury.
Collapse
Affiliation(s)
- Carlos Parras
- Sorbonne Université, UPMC University Paris 06, Inserm U1127, CNRS UMR 7225, GH Pitié-Salpêtrière, Institut du Cerveau et de la Moelle Épinière, ICM, Paris, France
| | - Corentine Marie
- Sorbonne Université, UPMC University Paris 06, Inserm U1127, CNRS UMR 7225, GH Pitié-Salpêtrière, Institut du Cerveau et de la Moelle Épinière, ICM, Paris, France
| | - Chuntao Zhao
- Department of Pediatrics, Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Qing Richard Lu
- Department of Pediatrics, Brain Tumor Center, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
15
|
Fufa TD, Baxter LL, Wedel JC, Gildea DE, Loftus SK, Pavan WJ. MEK inhibition remodels the active chromatin landscape and induces SOX10 genomic recruitment in BRAF(V600E) mutant melanoma cells. Epigenetics Chromatin 2019; 12:50. [PMID: 31399133 PMCID: PMC6688322 DOI: 10.1186/s13072-019-0297-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 07/28/2019] [Indexed: 01/03/2023] Open
Abstract
Background The MAPK/ERK signaling pathway is an essential regulator of numerous cell processes that are crucial for normal development as well as cancer progression. While much is known regarding MAPK/ERK signal conveyance from the cell membrane to the nucleus, the transcriptional and epigenetic mechanisms that govern gene expression downstream of MAPK signaling are not fully elucidated. Results This study employed an integrated epigenome analysis approach to interrogate the effects of MAPK/ERK pathway inhibition on the global transcriptome, the active chromatin landscape, and protein–DNA interactions in 501mel melanoma cells. Treatment of these cells with the small-molecule MEK inhibitor AZD6244 induces hyperpigmentation, widespread gene expression changes including alteration of genes linked to pigmentation, and extensive epigenomic reprogramming of transcriptionally distinct regulatory regions associated with the active chromatin mark H3K27ac. Regulatory regions with differentially acetylated H3K27ac regions following AZD6244 treatment are enriched in transcription factor binding motifs of ETV/ETS and ATF family members as well as the lineage-determining factors MITF and SOX10. H3K27ac-dense enhancer clusters known as super-enhancers show similar transcription factor motif enrichment, and furthermore, these super-enhancers are associated with genes encoding MITF, SOX10, and ETV/ETS proteins. Along with genome-wide resetting of the active enhancer landscape, MEK inhibition also results in widespread SOX10 recruitment throughout the genome, including increased SOX10 binding density at H3K27ac-marked enhancers. Importantly, these MEK inhibitor-responsive enhancers marked by H3K27ac and occupied by SOX10 are located near melanocyte lineage-specific and pigmentation genes and overlap numerous human SNPs associated with pigmentation and melanoma phenotypes, highlighting the variants located within these regions for prioritization in future studies. Conclusions These results reveal the epigenetic reprogramming underlying the re-activation of melanocyte pigmentation and developmental transcriptional programs in 501mel cells in response to MEK inhibition and suggest extensive involvement of a MEK-SOX10 axis in the regulation of these processes. The dynamic chromatin changes identified here provide a rich genomic resource for further analyses of the molecular mechanisms governing the MAPK pathway in pigmentation- and melanocyte-associated diseases. Electronic supplementary material The online version of this article (10.1186/s13072-019-0297-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Temesgen D Fufa
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA.,Ophthalmic Genetics and Visual Function Branch, National Eye Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Laura L Baxter
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Julia C Wedel
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Derek E Gildea
- Computational and Statistical Genomics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | | | - Stacie K Loftus
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - William J Pavan
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
16
|
Miao YL, Gambini A, Zhang Y, Padilla-Banks E, Jefferson WN, Bernhardt ML, Huang W, Li L, Williams CJ. Mediator complex component MED13 regulates zygotic genome activation and is required for postimplantation development in the mouse. Biol Reprod 2019; 98:449-464. [PMID: 29325037 DOI: 10.1093/biolre/ioy004] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 01/08/2018] [Indexed: 12/21/2022] Open
Abstract
Understanding factors that regulate zygotic genome activation (ZGA) is critical for determining how cells are reprogrammed to become totipotent or pluripotent. There is limited information regarding how this process occurs physiologically in early mammalian embryos. Here, we identify a mediator complex subunit, MED13, as translated during mouse oocyte maturation and transcribed early from the zygotic genome. Knockdown and conditional knockout approaches demonstrate that MED13 is essential for ZGA in the mouse, in part by regulating expression of the embryo-specific chromatin remodeling complex, esBAF. The role of MED13 in ZGA is mediated in part by interactions with E2F transcription factors. In addition to MED13, its paralog, MED13L, is required for successful preimplantation embryo development. MED13L partially compensates for loss of MED13 function in preimplantation knockout embryos, but postimplantation development is not rescued by MED13L. Our data demonstrate an essential role for MED13 in supporting chromatin reprogramming and directed transcription of essential genes during ZGA.
Collapse
Affiliation(s)
- Yi-Liang Miao
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA.,Key Laboratory of Agricultural Animal Genetics, Breeding, and Reproduction, Ministry of Education College of Animal Science and Technology, Huazhong Agricultural University, China
| | - Andrés Gambini
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Yingpei Zhang
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Elizabeth Padilla-Banks
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Wendy N Jefferson
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Miranda L Bernhardt
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Weichun Huang
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Leping Li
- Biostatistics and Computational Biology Branch, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| | - Carmen J Williams
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina, USA
| |
Collapse
|
17
|
Sock E, Wegner M. Transcriptional control of myelination and remyelination. Glia 2019; 67:2153-2165. [PMID: 31038810 DOI: 10.1002/glia.23636] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Revised: 04/01/2019] [Accepted: 04/11/2019] [Indexed: 12/11/2022]
Abstract
Myelination is an evolutionary recent differentiation program that has been independently acquired in vertebrates by Schwann cells in the peripheral nervous system and oligodendrocytes in the central nervous system. Therefore, it is not surprising that regulating transcription factors differ substantially between both cell types. However, overall principles are similar as transcriptional control in Schwann cells and oligodendrocytes combines lineage determining and stage-specific factors in complex regulatory networks. Myelination does not only occur during development, but also as remyelination in the adult. In line with the different conditions during developmental myelination and remyelination and the distinctive properties of Schwann cells and oligodendrocytes, transcriptional regulation of remyelination exhibits unique features and differs between the two cell types. This review gives an overview of the current state in the field.
Collapse
Affiliation(s)
- Elisabeth Sock
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
18
|
Dannappel MV, Sooraj D, Loh JJ, Firestein R. Molecular and in vivo Functions of the CDK8 and CDK19 Kinase Modules. Front Cell Dev Biol 2019; 6:171. [PMID: 30693281 PMCID: PMC6340071 DOI: 10.3389/fcell.2018.00171] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/06/2018] [Indexed: 12/21/2022] Open
Abstract
CDK8 and its paralog, CDK19, collectively termed ‘Mediator Kinase,’ are cyclin-dependent kinases that have been implicated as key rheostats in cellular homeostasis and developmental programming. CDK8 and CDK19 are incorporated, in a mutually exclusive manner, as part of a 4-protein complex called the Mediator kinase module. This module reversibly associates with the Mediator, a 26 subunit protein complex that regulates RNA Polymerase II mediated gene expression. As part of this complex, the Mediator kinases have been implicated in diverse process such as developmental signaling, metabolic homeostasis and in innate immunity. In recent years, dysregulation of Mediator kinase module proteins, including CDK8/19, has been implicated in the development of different human diseases, and in particular cancer. This has led to intense efforts to understand how CDK8/19 regulate diverse biological outputs and develop Mediator kinase inhibitors that can be exploited therapeutically. Herein, we review both context and function of the Mediator kinases at a molecular, cellular and animal level. In so doing, we illuminate emerging concepts underpinning Mediator kinase biology and highlight certain aspects that remain unsolved.
Collapse
Affiliation(s)
- Marius Volker Dannappel
- Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Dhanya Sooraj
- Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| | - Jia Jian Loh
- Hudson Institute of Medical Research, Clayton, VIC, Australia.,Faculty of Science, School of Biological Sciences, Monash University, Clayton, VIC, Australia
| | - Ron Firestein
- Hudson Institute of Medical Research, Clayton, VIC, Australia.,Department of Molecular and Translational Sciences, Monash University, Clayton, VIC, Australia
| |
Collapse
|
19
|
Ulanska-Poutanen J, Mieczkowski J, Zhao C, Konarzewska K, Kaza B, Pohl HB, Bugajski L, Kaminska B, Franklin RJ, Zawadzka M. Injury-induced perivascular niche supports alternative differentiation of adult rodent CNS progenitor cells. eLife 2018; 7:30325. [PMID: 30222103 PMCID: PMC6141235 DOI: 10.7554/elife.30325] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Accepted: 09/03/2018] [Indexed: 01/06/2023] Open
Abstract
Following CNS demyelination, oligodendrocyte progenitor cells (OPCs) are able to differentiate into either remyelinating oligodendrocytes (OLs) or remyelinating Schwann cells (SCs). However, the signals that determine which type of remyelinating cell is generated and the underlying mechanisms involved have not been identified. Here, we show that distinctive microenvironments created in discrete niches within demyelinated white matter determine fate decisions of adult OPCs. By comparative transcriptome profiling we demonstrate that an ectopic, injury-induced perivascular niche is enriched with secreted ligands of the BMP and Wnt signalling pathways, produced by activated OPCs and endothelium, whereas reactive astrocyte within non-vascular area express the dual BMP/Wnt antagonist Sostdc1. The balance of BMP/Wnt signalling network is instructive for OPCs to undertake fate decision shortly after their activation: disruption of the OPCs homeostasis during demyelination results in BMP4 upregulation, which, in the absence of Socstdc1, favours SCs differentiation.
Collapse
Affiliation(s)
- Justyna Ulanska-Poutanen
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Jakub Mieczkowski
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Chao Zhao
- Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom.,Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Katarzyna Konarzewska
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Beata Kaza
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Hartmut Bf Pohl
- Department of Biology, Institute of Molecular Health Sciences, Zurich, Switzerland
| | - Lukasz Bugajski
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Bozena Kaminska
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Robin Jm Franklin
- Wellcome Trust - Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom.,Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - Malgorzata Zawadzka
- Laboratory of Molecular Neurobiology, Neurobiology Center, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
20
|
Beadle EP, Straub JA, Bunnell BA, Newman JJ. MED31 involved in regulating self-renewal and adipogenesis of human mesenchymal stem cells. Mol Biol Rep 2018; 45:1545-1550. [PMID: 30006772 DOI: 10.1007/s11033-018-4241-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Accepted: 07/06/2018] [Indexed: 01/14/2023]
Abstract
Regulation of gene expression is critical for the maintenance of cell state and homeostasis. Aberrant regulation of genes can lead to unwanted cell proliferation or misdirected differentiation. Here we investigate the role of MED31, a highly conserved subunit of the Mediator complex, to determine the role this subunit plays in the maintenance of human mesenchymal stem cell (hMSC) state. Using siRNA-mediated knockdown of MED31 we demonstrate a decrease in self-renewal based on cell assays and monitoring of gene expression. In addition, in the absence of MED31, hMSCs also displayed a reduction in adipogenesis as evidenced by diminished lipid vesicle formation and expression of specific adipogenic markers. These data present evidence for a significant role for MED31 in maintaining adult stem cell homeostasis, thereby introducing potential novel targets for future investigation and use in better understanding stem cell behavior and adipogenesis.
Collapse
Affiliation(s)
- Erik P Beadle
- School of Biological Sciences, Louisiana Tech University, Ruston, LA, USA
| | - Joseph A Straub
- School of Biological Sciences, Louisiana Tech University, Ruston, LA, USA
| | - Bruce A Bunnell
- Center for Stem Cell Research and Regenerative Medicine, Tulane University School of Medicine, New Orleans, LA, USA
- Departments of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
- Division of Regenerative Medicine, Tulane National Primate Research Center, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jamie J Newman
- School of Biological Sciences, Louisiana Tech University, Ruston, LA, USA.
| |
Collapse
|
21
|
Putlyaev EV, Ibragimov AN, Lebedeva LA, Georgiev PG, Shidlovskii YV. Structure and Functions of the Mediator Complex. BIOCHEMISTRY (MOSCOW) 2018; 83:423-436. [PMID: 29626929 DOI: 10.1134/s0006297918040132] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Mediator is a key factor in the regulation of expression of RNA polymerase II-transcribed genes. Recent studies have shown that Mediator acts as a coordinator of transcription activation and participates in maintaining chromatin architecture in the cell nucleus. In this review, we present current concepts on the structure and functions of Mediator.
Collapse
Affiliation(s)
- E V Putlyaev
- Institute of Gene Biology, Russian Academy of Sciences, Moscow, 119334, Russia.
| | | | | | | | | |
Collapse
|
22
|
Uncovering the Role of Sox2 in Oligodendroglia. J Neurosci 2018; 38:4460-4461. [PMID: 29743345 DOI: 10.1523/jneurosci.0556-18.2018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/05/2018] [Accepted: 04/12/2018] [Indexed: 01/20/2023] Open
|
23
|
García-León JA, Kumar M, Boon R, Chau D, One J, Wolfs E, Eggermont K, Berckmans P, Gunhanlar N, de Vrij F, Lendemeijer B, Pavie B, Corthout N, Kushner SA, Dávila JC, Lambrichts I, Hu WS, Verfaillie CM. SOX10 Single Transcription Factor-Based Fast and Efficient Generation of Oligodendrocytes from Human Pluripotent Stem Cells. Stem Cell Reports 2018; 10:655-672. [PMID: 29337119 PMCID: PMC5830935 DOI: 10.1016/j.stemcr.2017.12.014] [Citation(s) in RCA: 80] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 12/15/2017] [Accepted: 12/15/2017] [Indexed: 02/06/2023] Open
Abstract
Scarce access to primary samples and lack of efficient protocols to generate oligodendrocytes (OLs) from human pluripotent stem cells (hPSCs) are hampering our understanding of OL biology and the development of novel therapies. Here, we demonstrate that overexpression of the transcription factor SOX10 is sufficient to generate surface antigen O4-positive (O4+) and myelin basic protein-positive OLs from hPSCs in only 22 days, including from patients with multiple sclerosis or amyotrophic lateral sclerosis. The SOX10-induced O4+ population resembles primary human OLs at the transcriptome level and can myelinate neurons in vivo. Using in vitro OL-neuron co-cultures, myelination of neurons by OLs can also be demonstrated, which can be adapted to a high-throughput screening format to test the response of pro-myelinating drugs. In conclusion, we provide an approach to generate OLs in a very rapid and efficient manner, which can be used for disease modeling, drug discovery efforts, and potentially for therapeutic OL transplantation. SOX10 is sufficient to generate myelinating human OLs from hPSCs in only 22 days SOX10-induced OLs resemble primary human OLs at the transcriptome level The methodology allows efficient generation of OLs from MS and ALS patients OL-neuron co-cultures respond to myelinating drugs in a high-throughput setting
Collapse
Affiliation(s)
- Juan Antonio García-León
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Herestraat 49, Onderwijs en Navorsing 4, Box 804, 3000 Leuven, Belgium.
| | - Manoj Kumar
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Herestraat 49, Onderwijs en Navorsing 4, Box 804, 3000 Leuven, Belgium
| | - Ruben Boon
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Herestraat 49, Onderwijs en Navorsing 4, Box 804, 3000 Leuven, Belgium
| | - David Chau
- Department of Chemical Engineering and Materials Science, University of Minnesota, 421 Washington Avenue Southeast, Minneapolis, MN 55455, USA
| | - Jennifer One
- Department of Chemical Engineering and Materials Science, University of Minnesota, 421 Washington Avenue Southeast, Minneapolis, MN 55455, USA
| | - Esther Wolfs
- Laboratory of Morphology, Biomedical Research Institute (BIOMED), Hasselt University, Agoralaan, Building C, 3590 Diepenbeek, Belgium
| | - Kristel Eggermont
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Herestraat 49, Onderwijs en Navorsing 4, Box 804, 3000 Leuven, Belgium
| | - Pieter Berckmans
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Herestraat 49, Onderwijs en Navorsing 4, Box 804, 3000 Leuven, Belgium
| | - Nilhan Gunhanlar
- Department of Psychiatry, Erasmus MC, 3015 CE Rotterdam, the Netherlands
| | - Femke de Vrij
- Department of Psychiatry, Erasmus MC, 3015 CE Rotterdam, the Netherlands
| | - Bas Lendemeijer
- Department of Psychiatry, Erasmus MC, 3015 CE Rotterdam, the Netherlands
| | - Benjamin Pavie
- VIB Center for Brain and Disease Research, VIB-Leuven, 3000 Leuven, Belgium; VIB Bio Imaging Core, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Nikky Corthout
- VIB Center for Brain and Disease Research, VIB-Leuven, 3000 Leuven, Belgium; VIB Bio Imaging Core, KU Leuven, Herestraat 49, 3000 Leuven, Belgium
| | - Steven A Kushner
- Department of Psychiatry, Erasmus MC, 3015 CE Rotterdam, the Netherlands
| | - José Carlos Dávila
- Department of Cell Biology, Genetics and Physiology, Faculty of Sciences, Research Biomedical Institute of Málaga (IBIMA), University of Málaga, Campus de Teatinos s/n, 29010 Málaga, Spain; Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Ivo Lambrichts
- Laboratory of Morphology, Biomedical Research Institute (BIOMED), Hasselt University, Agoralaan, Building C, 3590 Diepenbeek, Belgium
| | - Wei-Shou Hu
- Department of Chemical Engineering and Materials Science, University of Minnesota, 421 Washington Avenue Southeast, Minneapolis, MN 55455, USA
| | - Catherine M Verfaillie
- Department of Development and Regeneration, Stem Cell Biology and Embryology, KU Leuven Stem Cell Institute, Herestraat 49, Onderwijs en Navorsing 4, Box 804, 3000 Leuven, Belgium.
| |
Collapse
|
24
|
Abstract
The journey of Schwann cells from their origin in the neural crest to their ensheathment and myelination of peripheral nerves is a remarkable one. Their apparent static function in enabling saltatory conduction of mature nerve is not only vital for long-term health of peripheral nerve but also belies an innate capacity of terminally differentiated Schwann cells to radically alter their differentiation status in the face of nerve injury. The transition from migrating neural crest cells to nerve ensheathment, and then myelination of large diameter axons has been characterized extensively and several of the transcriptional networks have been identified. However, transcription factors must also modify chromatin structure during Schwann cell maturation and this review will focus on chromatin modification machinery that is involved in promoting the transition to, and maintenance of, myelinating Schwann cells. In addition, Schwann cells are known to play important regenerative roles after peripheral nerve injury, and information on epigenomic reprogramming of the Schwann cell genome has emerged. Characterization of epigenomic requirements for myelin maintenance and Schwann cell responses to injury will be vital in understanding how the various Schwann cell functions can be optimized to maintain and repair peripheral nerve function.
Collapse
Affiliation(s)
- Ki H Ma
- 1 Waisman Center, University of Wisconsin-Madison, Madison, WI, USA
| | - John Svaren
- 1 Waisman Center, University of Wisconsin-Madison, Madison, WI, USA.,2 Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
25
|
Glia-specific enhancers and chromatin structure regulate NFIA expression and glioma tumorigenesis. Nat Neurosci 2017; 20:1520-1528. [PMID: 28892058 DOI: 10.1038/nn.4638] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 08/03/2017] [Indexed: 12/18/2022]
Abstract
Long-range enhancer interactions critically regulate gene expression, yet little is known about how their coordinated activities contribute to CNS development or how this may, in turn, relate to disease states. By examining the regulation of the transcription factor NFIA in the developing spinal cord, we identified long-range enhancers that recapitulate NFIA expression across glial and neuronal lineages in vivo. Complementary genetic studies found that Sox9-Brn2 and Isl1-Lhx3 regulate enhancer activity and NFIA expression in glial and neuronal populations. Chromatin conformation analysis revealed that these enhancers and transcription factors form distinct architectures within these lineages in the spinal cord. In glioma models, the glia-specific architecture is present in tumors, and these enhancers are required for NFIA expression and contribute to glioma formation. By delineating three-dimensional mechanisms of gene expression regulation, our studies identify lineage-specific chromatin architectures and associated enhancers that regulate cell fate and tumorigenesis in the CNS.
Collapse
|
26
|
Chen L, Jin Y, Yang X, Liu Z, Wang Y, Wang G, Qi Z, Shen Z. Fat tissue, a potential Schwann cell reservoir: isolation and identification of adipose-derived Schwann cells. Am J Transl Res 2017; 9:2579-2594. [PMID: 28560006 PMCID: PMC5446538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 02/06/2017] [Indexed: 06/07/2023]
Abstract
Schwann cells can be used to promote peripheral nerve repair. However, it is challenging to obtain abundant autologous Schwann cells without sacrificing nerve integrity. In this study, we isolated Schwann cells from murine inguinal adipose tissue and identified the cell phenotype and function in vivo and in vitro. Through H&E and immunofluorescence staining, we detected tiny nerve fibers and Schwann cells in adipose tissue. We evaluated the phenotype of spindle-shaped cells (Schwann cell-like cells, SCLCs) isolated from adipose tissue using immunofluorescence staining and real-time RT-PCR. The results showed that SCLCs expressed specific Schwann cell markers. Analysis of conditioned SCLC culture media showed that, similar to Schwann cells isolated from sciatic nerves, SCLCs secreted NGF and BDNF. SCLCs were harvested from CAG-EGFP transgenic mice and combined with silicone nerve conduits to repair sciatic nerve defects in wild-type mice. Six months post-surgery, we found EGFP-positive SCLCs forming myelin sheaths in the same way as sciatic nerve-derived Schwann cells. This research indicates the existence of Schwann cells in adipose tissue and identifies the spindle-shaped cells isolated from adipose tissue as Schwann cells using in vitro and in vivo evaluations. Thus, SCLCs might be promising seed cells for peripheral nerve tissue engineering.
Collapse
Affiliation(s)
- Lulu Chen
- Department No. 16 of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, PR China
- Department of Plastic and Reconstructive Surgery, Shanghai 1 People’s Hospital, Shanghai Jiao Tong University School of MedicineShanghai, PR China
| | - Yuqing Jin
- Department of Plastic and Reconstructive Surgery, Shanghai 1 People’s Hospital, Shanghai Jiao Tong University School of MedicineShanghai, PR China
| | - Xiaonan Yang
- Department No. 16 of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, PR China
| | - Zhangyin Liu
- Department of Plastic and Reconstructive Surgery, Shanghai 1 People’s Hospital, Shanghai Jiao Tong University School of MedicineShanghai, PR China
| | - Yang Wang
- Department of Plastic and Reconstructive Surgery, Shanghai 1 People’s Hospital, Shanghai Jiao Tong University School of MedicineShanghai, PR China
| | - Gangyang Wang
- Department of Plastic and Reconstructive Surgery, Shanghai 1 People’s Hospital, Shanghai Jiao Tong University School of MedicineShanghai, PR China
| | - Zuoliang Qi
- Department No. 16 of Plastic Surgery Hospital, Chinese Academy of Medical Sciences & Peking Union Medical CollegeBeijing, PR China
| | - Zunli Shen
- Department of Plastic and Reconstructive Surgery, Shanghai 1 People’s Hospital, Shanghai Jiao Tong University School of MedicineShanghai, PR China
| |
Collapse
|
27
|
Grove M, Kim H, Santerre M, Krupka AJ, Han SB, Zhai J, Cho JY, Park R, Harris M, Kim S, Sawaya BE, Kang SH, Barbe MF, Cho SH, Lemay MA, Son YJ. YAP/TAZ initiate and maintain Schwann cell myelination. eLife 2017; 6:e20982. [PMID: 28124973 PMCID: PMC5287714 DOI: 10.7554/elife.20982] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 01/22/2017] [Indexed: 12/12/2022] Open
Abstract
Nuclear exclusion of the transcriptional regulators and potent oncoproteins, YAP/TAZ, is considered necessary for adult tissue homeostasis. Here we show that nuclear YAP/TAZ are essential regulators of peripheral nerve development and myelin maintenance. To proliferate, developing Schwann cells (SCs) require YAP/TAZ to enter S-phase and, without them, fail to generate sufficient SCs for timely axon sorting. To differentiate, SCs require YAP/TAZ to upregulate Krox20 and, without them, completely fail to myelinate, resulting in severe peripheral neuropathy. Remarkably, in adulthood, nuclear YAP/TAZ are selectively expressed by myelinating SCs, and conditional ablation results in severe peripheral demyelination and mouse death. YAP/TAZ regulate both developmental and adult myelination by driving TEAD1 to activate Krox20. Therefore, YAP/TAZ are crucial for SCs to myelinate developing nerve and to maintain myelinated nerve in adulthood. Our study also provides a new insight into the role of nuclear YAP/TAZ in homeostatic maintenance of an adult tissue.
Collapse
Affiliation(s)
- Matthew Grove
- Shriners Hospitals Pediatric Research Center, Center for Neural Repair, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Hyukmin Kim
- Shriners Hospitals Pediatric Research Center, Center for Neural Repair, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Maryline Santerre
- FELS Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Alexander J Krupka
- Department of Bioengineering, Temple University, Philadelphia, United States
| | - Seung Baek Han
- Shriners Hospitals Pediatric Research Center, Center for Neural Repair, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Jinbin Zhai
- Shriners Hospitals Pediatric Research Center, Center for Neural Repair, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Jennifer Y Cho
- Shriners Hospitals Pediatric Research Center, Center for Neural Repair, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Raehee Park
- Shriners Hospitals Pediatric Research Center, Center for Neural Repair, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Michele Harris
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Seonhee Kim
- Shriners Hospitals Pediatric Research Center, Center for Neural Repair, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Bassel E Sawaya
- FELS Institute for Cancer Research and Molecular Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Shin H Kang
- Shriners Hospitals Pediatric Research Center, Center for Neural Repair, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Mary F Barbe
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Seo-Hee Cho
- Shriners Hospitals Pediatric Research Center, Center for Neural Repair, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| | - Michel A Lemay
- Department of Bioengineering, Temple University, Philadelphia, United States
| | - Young-Jin Son
- Shriners Hospitals Pediatric Research Center, Center for Neural Repair, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, United States
| |
Collapse
|
28
|
Mittal P, Wang X, Rajkovic A. The Role of Mediator Complex Subunit 12 in Leiomyoma Biology. CURRENT GENETIC MEDICINE REPORTS 2016. [DOI: 10.1007/s40142-016-0106-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
29
|
Abstract
Myelin is a lipid-rich sheath formed by the spiral wrapping of specialized glial cells around axon segments. Myelinating glia allow for rapid transmission of nerve impulses and metabolic support of axons, and the absence of or disruption to myelin results in debilitating motor, cognitive, and emotional deficits in humans. Because myelin is a jawed vertebrate innovation, zebrafish are one of the simplest vertebrate model systems to study the genetics and development of myelinating glia. The morphogenetic cellular movements and genetic program that drive myelination are conserved between zebrafish and mammals, and myelin develops rapidly in zebrafish larvae, within 3-5days postfertilization. Myelin ultrastructure can be visualized in the zebrafish from larval to adult stages via transmission electron microscopy, and the dynamic development of myelinating glial cells may be observed in vivo via transgenic reporter lines in zebrafish larvae. Zebrafish are amenable to genetic and pharmacological screens, and screens for myelinating glial phenotypes have revealed both genes and drugs that promote myelin development, many of which are conserved in mammalian glia. Recently, zebrafish have been employed as a model to understand the complex dynamics of myelinating glia during development and regeneration. In this chapter, we describe these key methodologies and recent insights into mechanisms that regulate myelination using the zebrafish model.
Collapse
Affiliation(s)
- M D'Rozario
- Washington University School of Medicine, St. Louis, MO, United States
| | - K R Monk
- Washington University School of Medicine, St. Louis, MO, United States; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO, United States
| | | |
Collapse
|
30
|
Weider M, Wegner M. SoxE factors: Transcriptional regulators of neural differentiation and nervous system development. Semin Cell Dev Biol 2016; 63:35-42. [PMID: 27552919 DOI: 10.1016/j.semcdb.2016.08.013] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 08/16/2016] [Accepted: 08/18/2016] [Indexed: 12/20/2022]
Abstract
Sox8, Sox9 and Sox10 represent the three vertebrate members of the SoxE subclass of high-mobility-group domain containing Sox transcription factors. They play important roles in the peripheral and central nervous systems as regulators of stemness, specification, survival, lineage progression, glial differentiation and homeostasis. Functions are frequently overlapping, but sometimes antagonistic. SoxE proteins dynamically interact with transcriptional regulators, chromatin changing complexes and components of the transcriptional machinery. By establishing regulatory circuits with other transcription factors and microRNAs, SoxE proteins perform divergent functions in several cell lineages of the vertebrate nervous system, and at different developmental stages in the same cell lineage. The underlying molecular mechanisms are the topic of this review.
Collapse
Affiliation(s)
- Matthias Weider
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany.
| |
Collapse
|
31
|
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
|
32
|
Lopez-Anido C, Sun G, Koenning M, Srinivasan R, Hung HA, Emery B, Keles S, Svaren J. Differential Sox10 genomic occupancy in myelinating glia. Glia 2015; 63:1897-1914. [PMID: 25974668 PMCID: PMC4644515 DOI: 10.1002/glia.22855] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 04/22/2015] [Indexed: 11/11/2022]
Abstract
Myelin is formed by specialized myelinating glia: oligodendrocytes and Schwann cells in the central and peripheral nervous systems, respectively. While there are distinct developmental aspects and regulatory pathways in these two cell types, myelination in both systems requires the transcriptional activator Sox10. Sox10 interacts with cell type-specific transcription factors at some loci to induce myelin gene expression, but it is largely unknown how Sox10 transcriptional networks globally compare between oligodendrocytes and Schwann cells. We used in vivo ChIP-Seq analysis of spinal cord and peripheral nerve (sciatic nerve) to identify unique and shared Sox10 binding sites and assess their correlation with active enhancers and transcriptional profiles in oligodendrocytes and Schwann cells. Sox10 binding sites overlap with active enhancers and critical cell type-specific regulators of myelination, such as Olig2 and Myrf in oligodendrocytes, and Egr2/Krox20 in Schwann cells. Sox10 sites also associate with genes critical for myelination in both oligodendrocytes and Schwann cells and are found within super-enhancers previously defined in brain. In Schwann cells, Sox10 sites contain binding motifs of putative partners in the Sp/Klf, Tead, and nuclear receptor protein families. Specifically, siRNA analysis of nuclear receptors Nr2f1 and Nr2f2 revealed downregulation of myelin genes Mbp and Ndrg1 in primary Schwann cells. Our analysis highlights different mechanisms that establish cell type-specific genomic occupancy of Sox10, which reflects the unique characteristics of oligodendrocyte and Schwann cell differentiation. GLIA 2015;63:1897-1914.
Collapse
Affiliation(s)
- Camila Lopez-Anido
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Comparative Biomedical Sciences Graduate Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Guannan Sun
- Department of Biostatistics & Medical Informatics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Matthias Koenning
- Department of Anatomy and Neuroscience and the Centre for Neuroscience Research, University of Melbourne, Melbourne, Australia
| | - Rajini Srinivasan
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Holly A. Hung
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Ben Emery
- Department of Anatomy and Neuroscience and the Centre for Neuroscience Research, University of Melbourne, Melbourne, Australia
| | - Sunduz Keles
- Cellular and Molecular Pathology Graduate Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - John Svaren
- Waisman Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
33
|
Dionne N, Dib S, Finsen B, Denarier E, Kuhlmann T, Drouin R, Kokoeva M, Hudson TJ, Siminovitch K, Friedman HC, Peterson AC. Functional organization of anMbpenhancer exposes striking transcriptional regulatory diversity within myelinating glia. Glia 2015; 64:175-94. [DOI: 10.1002/glia.22923] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2015] [Revised: 09/04/2015] [Accepted: 09/09/2015] [Indexed: 11/11/2022]
Affiliation(s)
- Nancy Dionne
- Laboratory of Developmental Biology; Ludmer Research and Training Building, McGill University; Montreal Quebec Canada
| | - Samar Dib
- Laboratory of Developmental Biology; Ludmer Research and Training Building, McGill University; Montreal Quebec Canada
| | - Bente Finsen
- Department of Neurobiology Research; Institute of Molecular Medicine, University of Southern Denmark; Odense Denmark
| | - Eric Denarier
- Institut National De La Santé Et De La Recherche Médicale, U836-GIN iRTSV-GPC; Site Santé La Tronche, BP170 Grenoble Cedex 9 France
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital, Münster; Pottkamp 2 Münster Germany
| | - Régen Drouin
- Division of Genetics, Department of Pediatrics, Faculty of Medicine and Health Sciences; Université De Sherbrooke; Sherbrooke Quebec Canada
| | - Maia Kokoeva
- Department of Medicine; McGill University/MUHC Research Institute; Montreal Quebec Canada
| | - Thomas J. Hudson
- Ontario Institute for Cancer Research, MaRS Centre; South Tower Toronto Ontario Canada
| | - Kathy Siminovitch
- Department of Medicine; University of Toronto, Samuel Lunenfeld and Toronto General Research Institutes; Toronto Ontario Canada
- Department of Immunology and Molecular Genetics; University of Toronto; Toronto Ontario Canada
| | - Hana C Friedman
- Laboratory of Developmental Biology; Ludmer Research and Training Building, McGill University; Montreal Quebec Canada
| | - Alan C. Peterson
- Laboratory of Developmental Biology; Ludmer Research and Training Building, McGill University; Montreal Quebec Canada
| |
Collapse
|
34
|
Stolt CC, Wegner M. Schwann cells and their transcriptional network: Evolution of key regulators of peripheral myelination. Brain Res 2015; 1641:101-110. [PMID: 26423937 DOI: 10.1016/j.brainres.2015.09.025] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 09/18/2015] [Accepted: 09/20/2015] [Indexed: 11/29/2022]
Abstract
As derivatives of the neural crest, Schwann cells represent a vertebrate invention. Their development and differentiation is under control of a newly constructed, vertebrate-specific regulatory network that contains Sox10, Oct6 and Krox20 as cornerstones and central regulators of peripheral myelination. In this review, we discuss the function and relationship of these transcription factors among each other and in the context of their regulatory network, and present ideas of how neofunctionalization may have helped to recruit them to their novel task in Schwann cells. This article is part of a Special Issue entitled SI: Myelin Evolution.
Collapse
Affiliation(s)
- C Claus Stolt
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany.
| |
Collapse
|
35
|
Küspert M, Wegner M. SomethiNG 2 talk about-Transcriptional regulation in embryonic and adult oligodendrocyte precursors. Brain Res 2015; 1638:167-182. [PMID: 26232072 DOI: 10.1016/j.brainres.2015.07.024] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Revised: 07/14/2015] [Accepted: 07/18/2015] [Indexed: 12/26/2022]
Abstract
Glial cells that express the chondroitin sulfate proteoglycan NG2 represent an inherently heterogeneous population. These so-called NG2-glia are present during development and in the adult CNS, where they are referred to as embryonic oligodendrocyte precursors and adult NG2-glia, respectively. They give rise to myelinating oligodendrocytes at all times of life. Over the years much has been learnt about the transcriptional network in embryonic oligodendrocyte precursors, and several transcription factors from the HLH, HMG-domain, zinc finger and homeodomain protein families have been identified as main constituents. Much less is known about the corresponding network in adult NG2-glia. Here we summarize and discuss current knowledge on functions of each of these transcription factor families in NG2-glia, and where possible compare transcriptional regulation in embryonic oligodendrocyte precursors and adult NG2-glia. This article is part of a Special Issue entitled SI:NG2-glia (Invited only).
Collapse
Affiliation(s)
- Melanie Küspert
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, Erlangen D-91054, Germany.
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, Erlangen D-91054, Germany.
| |
Collapse
|
36
|
Clark AD, Oldenbroek M, Boyer TG. Mediator kinase module and human tumorigenesis. Crit Rev Biochem Mol Biol 2015; 50:393-426. [PMID: 26182352 DOI: 10.3109/10409238.2015.1064854] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mediator is a conserved multi-subunit signal processor through which regulatory informatiosn conveyed by gene-specific transcription factors is transduced to RNA Polymerase II (Pol II). In humans, MED13, MED12, CDK8 and Cyclin C (CycC) comprise a four-subunit "kinase" module that exists in variable association with a 26-subunit Mediator core. Genetic and biochemical studies have established the Mediator kinase module as a major ingress of developmental and oncogenic signaling through Mediator, and much of its function in signal-dependent gene regulation derives from its resident CDK8 kinase activity. For example, CDK8-targeted substrate phosphorylation impacts transcription factor half-life, Pol II activity and chromatin chemistry and functional status. Recent structural and biochemical studies have revealed a precise network of physical and functional subunit interactions required for proper kinase module activity. Accordingly, pathologic change in this activity through altered expression or mutation of constituent kinase module subunits can have profound consequences for altered signaling and tumor formation. Herein, we review the structural organization, biological function and oncogenic potential of the Mediator kinase module. We focus principally on tumor-associated alterations in kinase module subunits for which mechanistic relationships as opposed to strictly correlative associations are established. These considerations point to an emerging picture of the Mediator kinase module as an oncogenic unit, one in which pathogenic activation/deactivation through component change drives tumor formation through perturbation of signal-dependent gene regulation. It follows that therapeutic strategies to combat CDK8-driven tumors will involve targeted modulation of CDK8 activity or pharmacologic manipulation of dysregulated CDK8-dependent signaling pathways.
Collapse
Affiliation(s)
- Alison D Clark
- a Department of Molecular Medicine , Institute of Biotechnology, University of Texas Health Science Center at San Antonio , San Antonio , TX , USA
| | - Marieke Oldenbroek
- a Department of Molecular Medicine , Institute of Biotechnology, University of Texas Health Science Center at San Antonio , San Antonio , TX , USA
| | - Thomas G Boyer
- a Department of Molecular Medicine , Institute of Biotechnology, University of Texas Health Science Center at San Antonio , San Antonio , TX , USA
| |
Collapse
|
37
|
Abstract
Myelinated nerve fibers are essential for the rapid propagation of action potentials by saltatory conduction. They form as the result of reciprocal interactions between axons and Schwann cells. Extrinsic signals from the axon, and the extracellular matrix, drive Schwann cells to adopt a myelinating fate, whereas myelination reorganizes the axon for its role in conduction and is essential for its integrity. Here, we review our current understanding of the development, molecular organization, and function of myelinating Schwann cells. Recent findings into the extrinsic signals that drive Schwann cell myelination, their cognate receptors, and the downstream intracellular signaling pathways they activate will be described. Together, these studies provide important new insights into how these pathways converge to activate the transcriptional cascade of myelination and remodel the actin cytoskeleton that is critical for morphogenesis of the myelin sheath.
Collapse
Affiliation(s)
- James L Salzer
- Department of Neuroscience and Physiology, New York University Neuroscience Institute, New York University School of Medicine, New York, New York 10016
| |
Collapse
|
38
|
Hernandez M, Casaccia P. Interplay between transcriptional control and chromatin regulation in the oligodendrocyte lineage. Glia 2015; 63:1357-75. [PMID: 25970296 DOI: 10.1002/glia.22818] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 02/26/2015] [Indexed: 12/21/2022]
Abstract
The recent years have been characterized by a surge of studies on the role of transcription factors and histone modifications in regulating the progression of progenitors into oligodendrocytes. This review summarizes this body of evidence and presents an integrated view of transcriptional networks and epigenetic regulators defining proliferating progenitors and their differentiation along the oligodendrocyte lineage. We suggest that transcription factors in proliferating progenitors have direct access to DNA, due to predominantly euchromatic nuclei. As progenitors differentiate, however, transcriptional competence is modulated by the formation of heterochromatin, which modifies the association of DNA with nucleosomal histones and renders the access of transcription factors dependent on the activity of epigenetic modulators. These concepts are delineated within the context of development, and the potential functional implications are discussed.
Collapse
Affiliation(s)
- Marylens Hernandez
- Department of Neuroscience, Friedman Brain Institute and Icahn School of Medicine at Mount Sinai, New York City, New York.,Graduate School of Biological Sciences, Icahn School of Medicine at Mount Sinai, New York City, New York
| | - Patrizia Casaccia
- Department of Neuroscience, Friedman Brain Institute and Icahn School of Medicine at Mount Sinai, New York City, New York.,Department of Genomics and Multiscale Biology, Friedman Brain Institute and Icahn School of Medicine at Mount Sinai, New York City, New York
| |
Collapse
|
39
|
Hunter JM, Kiefer J, Balak CD, Jooma S, Ahearn ME, Hall JG, Baumbach-Reardon L. Review of X-linked syndromes with arthrogryposis or early contractures-aid to diagnosis and pathway identification. Am J Med Genet A 2015; 167A:931-73. [DOI: 10.1002/ajmg.a.36934] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Accepted: 12/05/2014] [Indexed: 02/03/2023]
Affiliation(s)
- Jesse M. Hunter
- Integrated Functional Cancer Genomics; Translational Genomics Research Institute; Phoenix Arizona
| | - Jeff Kiefer
- Knowledge Mining; Translational Genomics Research Institute; Phoenix Arizona
| | - Christopher D. Balak
- Integrated Functional Cancer Genomics; Translational Genomics Research Institute; Phoenix Arizona
| | - Sonya Jooma
- Integrated Functional Cancer Genomics; Translational Genomics Research Institute; Phoenix Arizona
| | - Mary Ellen Ahearn
- Integrated Functional Cancer Genomics; Translational Genomics Research Institute; Phoenix Arizona
| | - Judith G. Hall
- Departments of Medical Genetics and Pediatrics; University of British Columbia and BC Children's Hospital Vancouver; British Columbia Canada
| | - Lisa Baumbach-Reardon
- Integrated Functional Cancer Genomics; Translational Genomics Research Institute; Phoenix Arizona
| |
Collapse
|
40
|
Arter J, Wegner M. Transcription factors Sox10 and Sox2 functionally interact with positive transcription elongation factor b in Schwann cells. J Neurochem 2015; 132:384-93. [PMID: 25524031 DOI: 10.1111/jnc.13013] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Revised: 12/10/2014] [Accepted: 12/12/2014] [Indexed: 12/29/2022]
Abstract
Sox proteins are mechanistically versatile regulators with established relevance to different developmental processes and crucial impact on chromatin structure, DNA conformation, and transcriptional initiation. Here, we show that Sox2 and Sox10, two Sox proteins important for Schwann cell development, also have the capability to activate transcriptional elongation in a Schwann cell line by recruiting the positive transcription elongation factor b. Recruitment is mediated by physical interaction between the carboxyterminal transactivation domains of the two Sox proteins and the Cyclin T1 subunit of positive transcription elongation factor b, with interaction interfaces for the two Sox proteins being mapped to adjacent regions of the central part of Cyclin T1. Supporting the relevance of this interaction to Schwann cell development, transcription of myelin genes appears regulated at the level of elongation. Our results thus add a new facet to the activity of Sox proteins and expand the functional repertoire of this important group of developmental regulators. Sox transcription factors are important regulators of nervous system development. While they are known to regulate transcription by recruiting and stabilizing the RNA polymerase II preinitiation complex directly or with help of the Mediator complex, this study provides evidence that Sox10 and Sox2 additionally influence transcription in glial cells at the elongation stage by recruiting P-TEFb. Cdk9, cyclin-dependent kinase 9; P-TEFb, positive transcription elongation factor b; Pol II, RNA polymerase II; Sox, Sox2 or Sox10 protein.
Collapse
Affiliation(s)
- Juliane Arter
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | |
Collapse
|
41
|
Svaren J. MicroRNA and transcriptional crosstalk in myelinating glia. Neurochem Int 2014; 77:50-7. [PMID: 24979526 PMCID: PMC4177339 DOI: 10.1016/j.neuint.2014.06.010] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Revised: 06/10/2014] [Accepted: 06/17/2014] [Indexed: 12/21/2022]
Abstract
Several recent studies have addressed the important role of microRNA in regulation of differentiation of myelinating glia. While Schwann cells and oligodendrocytes in the peripheral and central nervous systems, respectively, exhibit significant morphological and regulatory differences, some aspects of transcriptional and microRNA regulation are shared between these two cell types. This review focuses on the intersection of microRNAs with transcriptional regulation in Schwann cell and oligodendrocyte differentiation. In particular, several microRNAs have been shown to modulate expression of critical transcription factors, and in turn, the regulation of microRNA expression is enmeshed within transcriptional networks that coordinate both coding gene and noncoding RNA profiles of myelinating cells. These hubs of regulation control both myelin gene expression as well as the cell cycle transitions of Schwann cells and oligodendrocytes as they terminally differentiate. In addition, some studies have begin to highlight the combinatorial effects of different microRNAs that establish the narrow range of gene regulation required for efficient and stable myelin formation. Overall, the integration of microRNA and transcriptional aspects will help elucidate mechanistic control of the myelination process.
Collapse
Affiliation(s)
- John Svaren
- Department of Comparative Biosciences and Waisman Center, University of Wisconsin-Madison, 1500 Highland Ave., Madison, WI 53705, USA.
| |
Collapse
|
42
|
Reiprich S, Wegner M. Sox2: A multitasking networker. NEUROGENESIS 2014; 1:e962391. [PMID: 27502481 PMCID: PMC4973596 DOI: 10.4161/23262125.2014.962391] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 09/02/2014] [Indexed: 01/15/2023]
Abstract
The transcription factor Sox2 is best known as a pluripotency factor in stem and precursor cells and its expression generally correlates with an undifferentiated state. Proposed modes of action include those as classical transcription factor and pre-patterning factor with influence on histone modifications and chromatin structure. Recently, we provided the first detailed analysis of Sox2 expression and function during development of oligodendrocytes, the myelin-forming cells of the CNS. Surprisingly, we found evidence for a role of Sox2 as differentiation factor and found it to act through modulation of microRNA levels. Thus, we add new facets to the functional repertoire of Sox2 and throw light on the networking activity of this multitasking developmental regulator.
Collapse
Affiliation(s)
- Simone Reiprich
- Institut für Biochemie; Emil-Fischer-Zentrum; Friedrich-Alexander-Universität Erlangen-Nürnberg ; Erlangen, Germany
| | - Michael Wegner
- Institut für Biochemie; Emil-Fischer-Zentrum; Friedrich-Alexander-Universität Erlangen-Nürnberg ; Erlangen, Germany
| |
Collapse
|
43
|
Mitew S, Hay C, Peckham H, Xiao J, Koenning M, Emery B. Mechanisms regulating the development of oligodendrocytes and central nervous system myelin. Neuroscience 2014; 276:29-47. [DOI: 10.1016/j.neuroscience.2013.11.029] [Citation(s) in RCA: 154] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 11/13/2013] [Accepted: 11/14/2013] [Indexed: 12/29/2022]
|
44
|
Reiprich S, Wegner M. From CNS stem cells to neurons and glia: Sox for everyone. Cell Tissue Res 2014; 359:111-24. [PMID: 24894327 DOI: 10.1007/s00441-014-1909-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 05/05/2014] [Indexed: 12/17/2022]
Abstract
Neuroepithelial precursor cells of the vertebrate central nervous system either self-renew or differentiate into neurons, oligodendrocytes or astrocytes under the influence of a gene regulatory network that consists in transcription factors, epigenetic modifiers and microRNAs. Sox transcription factors are central to this regulatory network, especially members of the SoxB, SoxC, SoxD, SoxE and SoxF groups. These Sox proteins are widely expressed in neuroepithelial precursor cells and in newly specified, differentiating and mature neurons, oligodendrocytes and astrocytes and influence their identity, survival and development. They exert their effect predominantly at the transcriptional level but also have substantial impact on expression at the epigenetic and posttranscriptional levels with some Sox proteins acting as pioneer factors, recruiting chromatin-modifying and -remodelling complexes or influencing microRNA expression. They interact with a large variety of other transcription factors and influence the expression of regulatory molecules and effector genes in a cell-type-specific and temporally controlled manner. As versatile regulators with context-dependent functions, they are not only indispensable for central nervous system development but might also be instrumental for the development of reprogramming and cell conversion strategies for replacement therapies and for assisted regeneration after injury or degeneration-induced cell loss in the central nervous system.
Collapse
Affiliation(s)
- Simone Reiprich
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Fahrstrasse 17, 91054, Erlangen, Germany,
| | | |
Collapse
|
45
|
Yin JW, Wang G. The Mediator complex: a master coordinator of transcription and cell lineage development. Development 2014; 141:977-87. [PMID: 24550107 DOI: 10.1242/dev.098392] [Citation(s) in RCA: 146] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mediator is a multiprotein complex that is required for gene transcription by RNA polymerase II. Multiple subunits of the complex show specificity in relaying information from signals and transcription factors to the RNA polymerase II machinery, thus enabling control of the expression of specific genes. Recent studies have also provided novel mechanistic insights into the roles of Mediator in epigenetic regulation, transcriptional elongation, termination, mRNA processing, noncoding RNA activation and super enhancer formation. Based on these specific roles in gene regulation, Mediator has emerged as a master coordinator of development and cell lineage determination. Here, we describe the most recent advances in understanding the mechanisms of Mediator function, with an emphasis on its role during development and disease.
Collapse
Affiliation(s)
- Jing-wen Yin
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | | |
Collapse
|
46
|
Kuuluvainen E, Hakala H, Havula E, Sahal Estimé M, Rämet M, Hietakangas V, Mäkelä TP. Cyclin-dependent kinase 8 module expression profiling reveals requirement of mediator subunits 12 and 13 for transcription of Serpent-dependent innate immunity genes in Drosophila. J Biol Chem 2014; 289:16252-61. [PMID: 24778181 DOI: 10.1074/jbc.m113.541904] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The Cdk8 (cyclin-dependent kinase 8) module of Mediator integrates regulatory cues from transcription factors to RNA polymerase II. It consists of four subunits where Med12 and Med13 link Cdk8 and cyclin C (CycC) to core Mediator. Here we have investigated the contributions of the Cdk8 module subunits to transcriptional regulation using RNA interference in Drosophila cells. Genome-wide expression profiling demonstrated separation of Cdk8-CycC and Med12-Med13 profiles. However, transcriptional regulation by Cdk8-CycC was dependent on Med12-Med13. This observation also revealed that Cdk8-CycC and Med12-Med13 often have opposite transcriptional effects. Interestingly, Med12 and Med13 profiles overlapped significantly with that of the GATA factor Serpent. Accordingly, mutational analyses indicated that GATA sites are required for Med12-Med13 regulation of Serpent-dependent genes. Med12 and Med13 were also found to be required for Serpent-activated innate immunity genes in defense to bacterial infection. The results reveal a novel role for the Cdk8 module in Serpent-dependent transcription and innate immunity.
Collapse
Affiliation(s)
- Emilia Kuuluvainen
- From the Institute of Biotechnology, University of Helsinki, P. O. Box 56, 00014 Helsinki
| | - Heini Hakala
- From the Institute of Biotechnology, University of Helsinki, P. O. Box 56, 00014 Helsinki
| | - Essi Havula
- From the Institute of Biotechnology, University of Helsinki, P. O. Box 56, 00014 Helsinki, the Department of Biosciences, University of Helsinki, P. O. Box 65, 00014 Helsinki
| | - Michelle Sahal Estimé
- From the Institute of Biotechnology, University of Helsinki, P. O. Box 56, 00014 Helsinki
| | - Mika Rämet
- the Institute of Biomedical Technology, and BioMediTech, University of Tampere, 33014 Tampere, the Department of Pediatrics, Tampere University Hospital, 22521 Tampere, the Department of Pediatrics, Institute of Clinical Medicine, and Medical Research Center Oulu, University of Oulu, 90014 Oulu, and the Department of Children and Adolescents, Oulu University Hospital, 90029 Oulu, Finland
| | - Ville Hietakangas
- From the Institute of Biotechnology, University of Helsinki, P. O. Box 56, 00014 Helsinki, the Department of Biosciences, University of Helsinki, P. O. Box 65, 00014 Helsinki
| | - Tomi P Mäkelä
- From the Institute of Biotechnology, University of Helsinki, P. O. Box 56, 00014 Helsinki,
| |
Collapse
|
47
|
Hoffmann SA, Hos D, Küspert M, Lang RA, Lovell-Badge R, Wegner M, Reiprich S. Stem cell factor Sox2 and its close relative Sox3 have differentiation functions in oligodendrocytes. Development 2014; 141:39-50. [PMID: 24257626 PMCID: PMC3865748 DOI: 10.1242/dev.098418] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Accepted: 09/29/2013] [Indexed: 11/20/2022]
Abstract
Neural precursor cells of the ventricular zone give rise to all neurons and glia of the central nervous system and rely for maintenance of their precursor characteristics on the closely related SoxB1 transcription factors Sox1, Sox2 and Sox3. We show in mouse spinal cord that, whereas SoxB1 proteins are usually downregulated upon neuronal specification, they continue to be expressed in glial precursors. In the oligodendrocyte lineage, Sox2 and Sox3 remain present into the early phases of terminal differentiation. Surprisingly, their deletion does not alter precursor characteristics but interferes with proper differentiation. Although a direct influence on myelin gene expression may be part of their function, we provide evidence for another mode of action. SoxB1 proteins promote oligodendrocyte differentiation in part by negatively controlling miR145 and thereby preventing this microRNA from inhibiting several pro-differentiation factors. This study presents one of the few cases in which SoxB1 proteins, including the stem cell factor Sox2, are associated with differentiation rather than precursor functions.
Collapse
Affiliation(s)
- Stephanie A. Hoffmann
- Institut für Biochemie, Emil-Fischer-Zentrum, Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Deniz Hos
- Institut für Biochemie, Emil-Fischer-Zentrum, Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Melanie Küspert
- Institut für Biochemie, Emil-Fischer-Zentrum, Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Richard A. Lang
- Division of Pediatric Ophthalmology, Cincinnati Children’s Hospital Research Foundation, Cincinnati, OH 45229-3026, USA
| | | | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
| | - Simone Reiprich
- Institut für Biochemie, Emil-Fischer-Zentrum, Universität Erlangen-Nürnberg, D-91054 Erlangen, Germany
| |
Collapse
|
48
|
Stamova BS, Tian Y, Nordahl CW, Shen MD, Rogers S, Amaral DG, Sharp FR. Evidence for differential alternative splicing in blood of young boys with autism spectrum disorders. Mol Autism 2013; 4:30. [PMID: 24007566 PMCID: PMC3846739 DOI: 10.1186/2040-2392-4-30] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 08/06/2013] [Indexed: 12/22/2022] Open
Abstract
Background Since RNA expression differences have been reported in autism spectrum disorder (ASD) for blood and brain, and differential alternative splicing (DAS) has been reported in ASD brains, we determined if there was DAS in blood mRNA of ASD subjects compared to typically developing (TD) controls, as well as in ASD subgroups related to cerebral volume. Methods RNA from blood was processed on whole genome exon arrays for 2-4–year-old ASD and TD boys. An ANCOVA with age and batch as covariates was used to predict DAS for ALL ASD (n=30), ASD with normal total cerebral volumes (NTCV), and ASD with large total cerebral volumes (LTCV) compared to TD controls (n=20). Results A total of 53 genes were predicted to have DAS for ALL ASD versus TD, 169 genes for ASD_NTCV versus TD, 1 gene for ASD_LTCV versus TD, and 27 genes for ASD_LTCV versus ASD_NTCV. These differences were significant at P <0.05 after false discovery rate corrections for multiple comparisons (FDR <5% false positives). A number of the genes predicted to have DAS in ASD are known to regulate DAS (SFPQ, SRPK1, SRSF11, SRSF2IP, FUS, LSM14A). In addition, a number of genes with predicted DAS are involved in pathways implicated in previous ASD studies, such as ROS monocyte/macrophage, Natural Killer Cell, mTOR, and NGF signaling. The only pathways significant after multiple comparison corrections (FDR <0.05) were the Nrf2-mediated reactive oxygen species (ROS) oxidative response (superoxide dismutase 2, catalase, peroxiredoxin 1, PIK3C3, DNAJC17, microsomal glutathione S-transferase 3) and superoxide radical degradation (SOD2, CAT). Conclusions These data support differences in alternative splicing of mRNA in blood of ASD subjects compared to TD controls that differ related to head size. The findings are preliminary, need to be replicated in independent cohorts, and predicted alternative splicing differences need to be confirmed using direct analytical methods.
Collapse
Affiliation(s)
- Boryana S Stamova
- MIND Institute, University of California at Davis, Sacramento, CA 95817, USA.
| | | | | | | | | | | | | |
Collapse
|
49
|
Zuchero JB, Barres BA. Intrinsic and extrinsic control of oligodendrocyte development. Curr Opin Neurobiol 2013; 23:914-20. [PMID: 23831087 DOI: 10.1016/j.conb.2013.06.005] [Citation(s) in RCA: 128] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2013] [Accepted: 06/11/2013] [Indexed: 11/24/2022]
Abstract
Oligodendrocytes (OLs) are the myelinating glia of the central nervous system. Myelin is essential for the rapid propagation of action potentials as well as for metabolic support of axons, and its loss in demyelinating diseases like multiple sclerosis has profound pathological consequences. The many steps in the development of OLs - from the specification of oligodendrocyte precursor cells (OPCs) during embryonic development to their differentiation into OLs that myelinate axons - are under tight regulation. Here we discuss recent advances in understanding how these steps of OL development are controlled intrinsically by transcription factors and chromatin remodeling and extrinsically by signaling molecules and neuronal activity. We also discuss how knowledge of these pathways is now allowing us to take steps toward generating patient-specific OPCs for disease modeling and myelin repair.
Collapse
Affiliation(s)
- J Bradley Zuchero
- Department of Neurobiology, Stanford University, Fairchild Building D205, 299 Campus Drive West, Stanford, CA 94305, United States.
| | | |
Collapse
|