Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 184
Filter
Add more filters

Publication year range
1.
Cell ; 187(10): 2465-2484.e22, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38701782

ABSTRACT

Remyelination failure in diseases like multiple sclerosis (MS) was thought to involve suppressed maturation of oligodendrocyte precursors; however, oligodendrocytes are present in MS lesions yet lack myelin production. We found that oligodendrocytes in the lesions are epigenetically silenced. Developing a transgenic reporter labeling differentiated oligodendrocytes for phenotypic screening, we identified a small-molecule epigenetic-silencing-inhibitor (ESI1) that enhances myelin production and ensheathment. ESI1 promotes remyelination in animal models of demyelination and enables de novo myelinogenesis on regenerated CNS axons. ESI1 treatment lengthened myelin sheaths in human iPSC-derived organoids and augmented (re)myelination in aged mice while reversing age-related cognitive decline. Multi-omics revealed that ESI1 induces an active chromatin landscape that activates myelinogenic pathways and reprograms metabolism. Notably, ESI1 triggered nuclear condensate formation of master lipid-metabolic regulators SREBP1/2, concentrating transcriptional co-activators to drive lipid/cholesterol biosynthesis. Our study highlights the potential of targeting epigenetic silencing to enable CNS myelin regeneration in demyelinating diseases and aging.


Subject(s)
Epigenesis, Genetic , Myelin Sheath , Oligodendroglia , Remyelination , Animals , Myelin Sheath/metabolism , Humans , Mice , Remyelination/drug effects , Oligodendroglia/metabolism , Central Nervous System/metabolism , Mice, Inbred C57BL , Rejuvenation , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/drug effects , Sterol Regulatory Element Binding Protein 1/metabolism , Organoids/metabolism , Organoids/drug effects , Demyelinating Diseases/metabolism , Demyelinating Diseases/genetics , Cell Differentiation/drug effects , Small Molecule Libraries/pharmacology , Male , Regeneration/drug effects , Multiple Sclerosis/metabolism , Multiple Sclerosis/genetics , Multiple Sclerosis/drug therapy , Multiple Sclerosis/pathology
2.
Cell ; 152(1-2): 15-6, 2013 Jan 17.
Article in English | MEDLINE | ID: mdl-23332743

ABSTRACT

For intracellular parasites that strongly prefer infecting a particular cell type, systemic spread is challenging. Masaki et al. show that the leprosy bacterium cleverly expands its repertoire by driving dedifferentiation of Schwann cells, their preferred host, to cells with stem cell properties that can subsequently redifferentiate into mesodermal lineages.

3.
Glia ; 72(8): 1435-1450, 2024 08.
Article in English | MEDLINE | ID: mdl-38613395

ABSTRACT

The plant homeodomain finger protein Phf8 is a histone demethylase implicated by mutation in mice and humans in neural crest defects and neurodevelopmental disturbances. Considering its widespread expression in cell types of the central nervous system, we set out to determine the role of Phf8 in oligodendroglial cells to clarify whether oligodendroglial defects are a possible contributing factor to Phf8-dependent neurodevelopmental disorders. Using loss- and gain-of-function approaches in oligodendroglial cell lines and primary cell cultures, we show that Phf8 promotes the proliferation of rodent oligodendrocyte progenitor cells and impairs their differentiation to oligodendrocytes. Intriguingly, Phf8 has a strong positive impact on Olig2 expression by acting on several regulatory regions of the gene and changing their histone modification profile. Taking the influence of Olig2 levels on oligodendroglial proliferation and differentiation into account, Olig2 likely acts as an important downstream effector of Phf8 in these cells. In line with such an effector function, ectopic Olig2 expression in Phf8-deficient cells rescues the proliferation defect. Additionally, generation of human oligodendrocytes from induced pluripotent stem cells did not require PHF8 in a system that relies on forced expression of Olig2 during oligodendroglial induction. We conclude that Phf8 may impact nervous system development at least in part through its action in oligodendroglial cells.


Subject(s)
Cell Proliferation , Oligodendrocyte Transcription Factor 2 , Oligodendroglia , Transcription Factors , Oligodendroglia/metabolism , Oligodendrocyte Transcription Factor 2/metabolism , Animals , Humans , Transcription Factors/metabolism , Transcription Factors/genetics , Mice , Cell Proliferation/physiology , Cell Differentiation/physiology , Cells, Cultured , Histone Demethylases/metabolism , Histone Demethylases/genetics , Rats , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Induced Pluripotent Stem Cells/metabolism
4.
Glia ; 72(7): 1304-1318, 2024 07.
Article in English | MEDLINE | ID: mdl-38546197

ABSTRACT

Oligodendrocyte differentiation and myelination in the central nervous system are controlled and coordinated by a complex gene regulatory network that contains several transcription factors, including Zfp488 and Nkx2.2. Despite the proven role in oligodendrocyte differentiation little is known about the exact mode of Zfp488 and Nkx2.2 action, including their target genes. Here, we used overexpression of Zfp488 and Nkx2.2 in differentiating CG4 cells to identify aspects of the oligodendroglial expression profile that depend on these transcription factors. Although both transcription factors are primarily described as repressors, the detected changes argue for an additional function as activators. Among the genes activated by both Zfp488 and Nkx2.2 was the G protein-coupled receptor Gpr37 that is important during myelination. In agreement with a positive effect on Gpr37 expression, downregulation of the G protein-coupled receptor was observed in Zfp488- and in Nkx2.2-deficient oligodendrocytes in the mouse. We also identified several potential regulatory regions of the Gpr37 gene. Although Zfp488 and Nkx2.2 both bind to one of the regulatory regions downstream of the Gpr37 gene in vivo, none of the regulatory regions was activated by either transcription factor alone. Increased activation by Zfp488 or Nkx2.2 was only observed in the presence of Sox10, a transcription factor continuously present in oligodendroglial cells. Our results argue that both Zfp488 and Nkx2.2 also act as transcriptional activators during oligodendrocyte differentiation and cooperate with Sox10 to allow the expression of Gpr37 as a modulator of the myelination process.


Subject(s)
Cell Differentiation , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Oligodendroglia , Receptors, G-Protein-Coupled , SOXE Transcription Factors , Transcription Factors , Animals , Female , Male , Mice , Cell Differentiation/physiology , Homeodomain Proteins/metabolism , Homeodomain Proteins/genetics , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/genetics , SOXE Transcription Factors/metabolism , SOXE Transcription Factors/genetics , Transcription Factors/metabolism , Transcription Factors/genetics
5.
EMBO J ; 39(13): e104159, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32627520

ABSTRACT

γδ T cells with distinct properties develop in the embryonic and adult thymus and have been identified as critical players in a broad range of infections, antitumor surveillance, autoimmune diseases, and tissue homeostasis. Despite their potential value for immunotherapy, differentiation of γδ T cells in the thymus is incompletely understood. Here, we establish a high-resolution map of γδ T-cell differentiation from the fetal and adult thymus using single-cell RNA sequencing. We reveal novel sub-types of immature and mature γδ T cells and identify an unpolarized thymic population which is expanded in the blood and lymph nodes. Our detailed comparative analysis reveals remarkable similarities between the gene networks active during fetal and adult γδ T-cell differentiation. By performing a combined single-cell analysis of Sox13, Maf, and Rorc knockout mice, we demonstrate sequential activation of these factors during IL-17-producing γδ T-cell (γδT17) differentiation. These findings substantially expand our understanding of γδ T-cell ontogeny in fetal and adult life. Our experimental and computational strategy provides a blueprint for comparing immune cell differentiation across developmental stages.


Subject(s)
Cell Differentiation/immunology , Fetus/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Animals , Autoantigens/genetics , Autoantigens/immunology , Cell Differentiation/genetics , Mice , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Proto-Oncogene Proteins c-maf/genetics , Proto-Oncogene Proteins c-maf/immunology , Receptors, Antigen, T-Cell, gamma-delta/genetics , T-Lymphocytes/cytology
6.
Development ; 148(14)2021 07 15.
Article in English | MEDLINE | ID: mdl-34184026

ABSTRACT

Transcription factor 4 (TCF4) is a crucial regulator of neurodevelopment and has been linked to the pathogenesis of autism, intellectual disability and schizophrenia. As a class I bHLH transcription factor (TF), it is assumed that TCF4 exerts its neurodevelopmental functions through dimerization with proneural class II bHLH TFs. Here, we aim to identify TF partners of TCF4 in the control of interhemispheric connectivity formation. Using a new bioinformatic strategy integrating TF expression levels and regulon activities from single cell RNA-sequencing data, we find evidence that TCF4 interacts with non-bHLH TFs and modulates their transcriptional activity in Satb2+ intercortical projection neurons. Notably, this network comprises regulators linked to the pathogenesis of neurodevelopmental disorders, e.g. FOXG1, SOX11 and BRG1. In support of the functional interaction of TCF4 with non-bHLH TFs, we find that TCF4 and SOX11 biochemically interact and cooperatively control commissure formation in vivo, and regulate the transcription of genes implicated in this process. In addition to identifying new candidate interactors of TCF4 in neurodevelopment, this study illustrates how scRNA-Seq data can be leveraged to predict TF networks in neurodevelopmental processes.


Subject(s)
RNA, Small Cytoplasmic/metabolism , Single-Cell Analysis , Transcription Factor 4/genetics , Transcription Factor 4/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , DNA Helicases , Embryo, Mammalian , Forkhead Transcription Factors , Gene Expression Regulation, Developmental , Gene Regulatory Networks , Intellectual Disability , Matrix Attachment Region Binding Proteins , Mice , Mice, Knockout , Nerve Tissue Proteins , Neurons/physiology , Nuclear Proteins , Protein Interaction Domains and Motifs , RNA, Small Cytoplasmic/genetics , SOXC Transcription Factors , Schizophrenia/genetics , Schizophrenia/metabolism
7.
Nucleic Acids Res ; 50(4): 1951-1968, 2022 02 28.
Article in English | MEDLINE | ID: mdl-35137157

ABSTRACT

In oligodendrocytes of the vertebrate central nervous system a complex network of transcriptional regulators is required to ensure correct and timely myelination of neuronal axons. Here we identify Zfp276, the only mammalian ZAD-domain containing zinc finger protein, as a transcriptional regulator of oligodendrocyte differentiation and central myelination downstream of Sox10. In the central nervous system, Zfp276 is exclusively expressed in mature oligodendrocytes. Oligodendroglial deletion of Zfp276 led to strongly reduced expression of myelin genes in the early postnatal mouse spinal cord. Retroviral overexpression of Zfp276 in cultured oligodendrocyte precursor cells induced precocious expression of maturation markers and myelin genes, further supporting its role in oligodendroglial differentiation. On the molecular level, Zfp276 directly binds to and represses Sox10-dependent gene regulatory regions of immaturity factors and functionally interacts with the transcriptional repressor Zeb2 to enable fast transition of oligodendrocytes to the myelinating stage.


Subject(s)
Oligodendroglia , Spinal Cord/cytology , Transcription Factors , Animals , Cell Differentiation , Mice , Myelin Sheath/physiology , Neurogenesis , Oligodendroglia/cytology , Oligodendroglia/metabolism , Spinal Cord/metabolism , Stem Cells , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Nucleic Acids Res ; 50(20): 11509-11528, 2022 11 11.
Article in English | MEDLINE | ID: mdl-36318265

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.


Subject(s)
Kruppel-Like Transcription Factors , Myelin Sheath , Oligodendroglia , SOXE Transcription Factors , Animals , Mice , Cell Differentiation/genetics , Gene Expression , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Myelin Sheath/genetics , Myelin Sheath/metabolism , Oligodendroglia/metabolism , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism
9.
Int J Mol Sci ; 25(10)2024 May 11.
Article in English | MEDLINE | ID: mdl-38791273

ABSTRACT

The HMG-domain containing transcription factor Sox10 plays a crucial role in regulating Schwann cell survival and differentiation and is expressed throughout the entire Schwann cell lineage. While its importance in peripheral myelination is well established, little is known about its role in the early stages of Schwann cell development. In a search for direct target genes of Sox10 in Schwann cell precursors, the transcriptional co-repressor Tle4 was identified. At least two regions upstream of the Tle4 gene appear involved in mediating the Sox10-dependent activation. Once induced, Tle4 works in tandem with the bHLH transcriptional repressor Hes1 and exerts a dual inhibitory effect on Sox10 by preventing the Sox10 protein from transcriptionally activating maturation genes and by suppressing Sox10 expression through known enhancers of the gene. This mechanism establishes a regulatory barrier that prevents premature activation of factors involved in differentiation and myelin formation by Sox10 in immature Schwann cells. The identification of Tle4 as a critical downstream target of Sox10 sheds light on the gene regulatory network in the early phases of Schwann cell development. It unravels an elaborate regulatory circuitry that fine-tunes the timing and extent of Schwann cell differentiation and myelin gene expression.


Subject(s)
Cell Differentiation , DNA-Binding Proteins , SOXE Transcription Factors , Schwann Cells , Animals , Humans , Mice , Rats , Cell Differentiation/genetics , Co-Repressor Proteins/metabolism , Co-Repressor Proteins/genetics , Feedback, Physiological , Gene Expression Regulation , Myelin Sheath/metabolism , Schwann Cells/metabolism , Schwann Cells/cytology , SOXE Transcription Factors/metabolism , SOXE Transcription Factors/genetics , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics , DNA-Binding Proteins/metabolism
10.
Glia ; 71(8): 1890-1905, 2023 08.
Article in English | MEDLINE | ID: mdl-37017184

ABSTRACT

The high-mobility-group domain-containing transcription factor Sox9 confers glial competence to neuroepithelial precursors in the developing central nervous system and is an important determinant of astroglial and oligodendroglial specification. In oligodendroglial cells, it remains expressed in oligodendrocyte progenitor cells (OPCs) of the developing nervous system, but is shut off in differentiating oligodendrocytes as well as in OPCs that persist in the adult nervous system. To better understand the role of Sox9 in OPCs, we generated mouse models that allowed oligodendroglial expression of a Sox9 transgene during development or in the adult. With transgene expression beginning in the last trimester of pregnancy, the number of OPCs increased dramatically, followed by comparable gains in the number of pre-myelinating and myelinating oligodendrocytes as assessed by marker gene expression. This argues that Sox9 boosts oligodendrogenesis during ontogenetic development at all stages, including terminal oligodendrocyte differentiation. When Sox9 transgene expression started in the adult, many transgene-expressing OPCs failed to maintain their progenitor cell identity and instead converted into myelinating oligodendrocytes. As infrequent and inefficient differentiation of adult OPCs is one of the main obstacles to effective remyelination in demyelinating diseases such as Multiple Sclerosis, increased Sox9 levels in adult OPCs may substantially increase their remyelination capacity.


Subject(s)
Multiple Sclerosis , Oligodendroglia , Mice , Animals , Oligodendroglia/metabolism , Cell Differentiation/physiology , Neuroglia/metabolism , Multiple Sclerosis/metabolism , Stem Cells/metabolism , Myelin Sheath/metabolism
11.
Proc Natl Acad Sci U S A ; 117(45): 28422-28432, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33109720

ABSTRACT

The human cerebral cortex contains many cell types that likely underwent independent functional changes during evolution. However, cell-type-specific regulatory landscapes in the cortex remain largely unexplored. Here we report epigenomic and transcriptomic analyses of the two main cortical neuronal subtypes, glutamatergic projection neurons and GABAergic interneurons, in human, chimpanzee, and rhesus macaque. Using genome-wide profiling of the H3K27ac histone modification, we identify neuron-subtype-specific regulatory elements that previously went undetected in bulk brain tissue samples. Human-specific regulatory changes are uncovered in multiple genes, including those associated with language, autism spectrum disorder, and drug addiction. We observe preferential evolutionary divergence in neuron subtype-specific regulatory elements and show that a substantial fraction of pan-neuronal regulatory elements undergoes subtype-specific evolutionary changes. This study sheds light on the interplay between regulatory evolution and cell-type-dependent gene-expression programs, and provides a resource for further exploration of human brain evolution and function.


Subject(s)
Cerebral Cortex/metabolism , Evolution, Molecular , Neurons/metabolism , Animals , Autism Spectrum Disorder/genetics , Brain/metabolism , Epigenesis, Genetic , Epigenomics , Gene Expression , Histone Code , Humans , Interneurons/metabolism , Macaca mulatta/genetics , Pan troglodytes/genetics , Primates/genetics , Regulatory Elements, Transcriptional , Regulatory Sequences, Nucleic Acid , Transcriptome
12.
J Neurosci ; 41(43): 8876-8886, 2021 10 27.
Article in English | MEDLINE | ID: mdl-34503995

ABSTRACT

Cortical parvalbumin-expressing (Pvalb+) neurons provide robust inhibition to neighboring pyramidal neurons, crucial for the proper functioning of cortical networks. This class of inhibitory neurons undergoes extensive synaptic formation and maturation during the first weeks after birth and continue to dynamically maintain their synaptic output throughout adulthood. While several transcription factors, such as Nkx2-1, Lhx6, and Sox6, are known to be necessary for the differentiation of progenitors into Pvalb+ neurons, which transcriptional programs underlie the postnatal maturation and maintenance of Pvalb+ neurons' innervation and synaptic function remains largely unknown. Because Sox6 is continuously expressed in Pvalb+ neurons until adulthood, we used conditional knock-out strategies to investigate its putative role in the postnatal maturation and synaptic function of cortical Pvalb+ neurons in mice of both sexes. We found that early postnatal loss of Sox6 in Pvalb+ neurons leads to failure of synaptic bouton growth, whereas later removal in mature Pvalb+ neurons in the adult causes shrinkage of already established synaptic boutons. Paired recordings between Pvalb+ neurons and pyramidal neurons revealed reduced release probability and increased failure rate of Pvalb+ neurons' synaptic output. Furthermore, Pvalb+ neurons lacking Sox6 display reduced expression of full-length tropomyosin-receptor kinase B (TrkB), a key modulator of GABAergic transmission. Once re-expressed in neurons lacking Sox6, TrkB was sufficient to rescue the morphologic synaptic phenotype. Finally, we showed that Sox6 mRNA levels were increased by motor training. Our data thus suggest a constitutive role for Sox6 in the maintenance of synaptic output from Pvalb+ neurons into adulthood.SIGNIFICANCE STATEMENT Cortical parvalbumin-expressing (Pvalb+) inhibitory neurons provide robust inhibition to neighboring pyramidal neurons, crucial for the proper functioning of cortical networks. These inhibitory neurons undergo extensive synaptic formation and maturation during the first weeks after birth and continue to dynamically maintain their synaptic output throughout adulthood. However, it remains largely unknown which transcriptional programs underlie the postnatal maturation and maintenance of Pvalb+ neurons. Here, we show that the transcription factor Sox6 cell-autonomously regulates the synaptic maintenance and output of Pvalb+ neurons until adulthood, leaving unaffected other maturational features of this neuronal population.


Subject(s)
Cerebral Cortex/metabolism , Neurons/metabolism , Parvalbumins/biosynthesis , SOXD Transcription Factors/biosynthesis , Synapses/metabolism , Animals , Animals, Newborn , Cerebral Cortex/cytology , Female , Gene Knock-In Techniques , Male , Mice , Mice, Transgenic , Organ Culture Techniques , Parvalbumins/genetics , SOXD Transcription Factors/genetics , Synapses/genetics
13.
Glia ; 70(3): 522-535, 2022 03.
Article in English | MEDLINE | ID: mdl-34787332

ABSTRACT

Recently, oligodendrocytes (Ol) have been attributed potential immunomodulatory effects. Yet, the exact mode of interaction with pathogenic CNS infiltrating lymphocytes remains unclear. Here, we attempt to dissect mechanisms of Ol modulation during neuroinflammation and characterize the interaction of Ol with pathogenic T cells. RNA expression analysis revealed an upregulation of immune-modulatory genes and adhesion molecules (AMs), ICAM-1 and VCAM-1, in Ol when isolated from mice undergoing experimental autoimmune encephalomyelitis (EAE). To explore whether AMs are involved in the interaction of Ol with infiltrating T cells, we performed co-culture studies on mature Ol and Th1 cells. Live cell imaging analysis showed direct interaction between both cell types. Eighty percentage of Th1 cells created contacts with Ol that lasted longer than 15 min, which may be regarded as physiologically relevant. Exposure of Ol to Th1 cells or their supernatant resulted in a significant extension of Ol processes, and upregulation of AMs as well as other immunomodulatory genes. Our observations indicate that blocking of oligodendroglial ICAM-1 can reduce the number of Th1 cells initially contacting the Ol. These results suggest that AMs may play a role in the interaction between Ol and Th1 cells. We identified Ol interacting with CD4+ cells in vivo in spinal cord tissue of EAE diseased mice indicating that our in vitro findings are of interest to further scientific research in this field. Further characterization and understanding of Ol interaction with infiltrating cells may lead to new therapeutic strategies enhancing Ol protection and remyelination potential. Oligodendrocytes regulate immune modulatory genes and adhesion molecules during autoimmune neuroinflammation Oligodendrocytes interact with Th1 cells in vitro in a physiologically relevant manner Adhesion molecules may be involved in Ol-Th1 cell interaction.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Intercellular Adhesion Molecule-1/metabolism , Animals , Encephalomyelitis, Autoimmune, Experimental/pathology , Intercellular Adhesion Molecule-1/genetics , Mice , Mice, Inbred C57BL , Neuroinflammatory Diseases , Oligodendroglia/metabolism
14.
Nucleic Acids Res ; 48(3): 1254-1270, 2020 02 20.
Article in English | MEDLINE | ID: mdl-31828317

ABSTRACT

Oligodendrocytes generate myelin in the vertebrate central nervous system and thus ensure rapid propagation of neuronal activity. Their development is controlled by a network of transcription factors that function as determinants of cell identity or as temporally restricted stage-specific regulators. The continuously expressed Sox10 and Myrf, a factor induced during late development, are particularly important for terminal differentiation. How these factors function together mechanistically and influence each other, is not well understood. Here we show that Myrf not only cooperates with Sox10 during the induction of genes required for differentiation and myelin formation. Myrf also inhibits the activity of Sox10 on genes that are essential during earlier phases of oligodendroglial development. By characterization of the exact DNA-binding requirements of Myrf, we furthermore show that cooperative activation is a consequence of joint binding of Sox10 and Myrf to the same regulatory regions. In contrast, inhibition of Sox10-dependent gene activation occurs on genes that lack Myrf binding sites and likely involves physical interaction between Myrf and Sox10 followed by sequestration. These two opposite activities allow Myrf to redirect Sox10 from genes that it activates in oligodendrocyte precursor cells to genes that need to be induced during terminal differentiation.


Subject(s)
Cell Differentiation/genetics , Membrane Proteins/genetics , Oligodendroglia/metabolism , SOXE Transcription Factors/genetics , Transcription Factors/genetics , Animals , Central Nervous System/growth & development , Central Nervous System/metabolism , Embryonic Development/genetics , HEK293 Cells , Humans , Mice , Myelin Sheath/genetics , Neurogenesis/genetics , Rats
15.
Nucleic Acids Res ; 48(9): 4839-4857, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32266943

ABSTRACT

Development of oligodendrocytes and myelin formation in the vertebrate central nervous system is under control of several basic helix-loop-helix transcription factors such as Olig2, Ascl1, Hes5 and the Id proteins. The class I basic helix-loop-helix proteins Tcf3, Tcf4 and Tcf12 represent potential heterodimerization partners and functional modulators for all, but have not been investigated in oligodendrocytes so far. Using mouse mutants, organotypic slice and primary cell cultures we here show that Tcf4 is required in a cell-autonomous manner for proper terminal differentiation and myelination in vivo and ex vivo. Partial compensation is provided by the paralogous Tcf3, but not Tcf12. On the mechanistic level Tcf4 was identified as the preferred heterodimerization partner of the central regulator of oligodendrocyte development Olig2. Both genetic studies in the mouse as well as functional studies on enhancer regions of myelin genes confirmed the relevance of this physical interaction for oligodendrocyte differentiation. Considering that alterations in TCF4 are associated with syndromic and non-syndromic forms of intellectual disability, schizophrenia and autism in humans, our findings point to the possibility of an oligodendroglial contribution to these disorders.


Subject(s)
Oligodendrocyte Transcription Factor 2/genetics , Oligodendroglia/cytology , Transcription Factor 4/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cell Line, Tumor , Cells, Cultured , Dimerization , Female , Gene Deletion , HEK293 Cells , Humans , Male , Mice , Myelin Sheath/physiology , Oligodendroglia/metabolism , Rats, Wistar
16.
Nucleic Acids Res ; 48(16): 8959-8976, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32672815

ABSTRACT

Schwann cells are the nerve ensheathing cells of the peripheral nervous system. Absence, loss and malfunction of Schwann cells or their myelin sheaths lead to peripheral neuropathies such as Charcot-Marie-Tooth disease in humans. During Schwann cell development and myelination chromatin is dramatically modified. However, impact and functional relevance of these modifications are poorly understood. Here, we analyzed histone H2B monoubiquitination as one such chromatin modification by conditionally deleting the Rnf40 subunit of the responsible E3 ligase in mice. Rnf40-deficient Schwann cells were arrested immediately before myelination or generated abnormally thin, unstable myelin, resulting in a peripheral neuropathy characterized by hypomyelination and progressive axonal degeneration. By combining sequencing techniques with functional studies we show that H2B monoubiquitination does not influence global gene expression patterns, but instead ensures selective high expression of myelin and lipid biosynthesis genes and proper repression of immaturity genes. This requires the specific recruitment of the Rnf40-containing E3 ligase by Egr2, the central transcriptional regulator of peripheral myelination, to its target genes. Our study identifies histone ubiquitination as essential for Schwann cell myelination and unravels new disease-relevant links between chromatin modifications and transcription factors in the underlying regulatory network.


Subject(s)
Early Growth Response Protein 2/physiology , Hereditary Sensory and Motor Neuropathy/metabolism , Histones/metabolism , Peripheral Nervous System/metabolism , Schwann Cells/metabolism , Animals , Cell Line, Tumor , HEK293 Cells , Humans , Mice , Mice, Transgenic , Peripheral Nervous System/pathology , Rats , Schwann Cells/pathology , Ubiquitin-Protein Ligases/genetics , Ubiquitination
17.
Glia ; 69(6): 1464-1477, 2021 06.
Article in English | MEDLINE | ID: mdl-33566433

ABSTRACT

The transcription factor Sox10 is an essential regulator of genes that code for structural components of the myelin sheath and for lipid metabolic enzymes in both types of myelinating glia in the central and peripheral nervous systems. In an attempt to characterize additional Sox10 target genes in Schwann cells, we identified in this study a strong influence of Sox10 on the expression of genes associated with adhesion in the MSC80 Schwann cell line. These included the genes for Gliomedin, Neuronal cell adhesion molecule and Neurofascin that together constitute essential Schwann cell contributions to paranode and node of Ranvier. Using bioinformatics and molecular biology techniques we provide evidence that Sox10 directly activates these genes by binding to conserved regulatory regions. For activation, Sox10 cooperates with Krox20, a transcription factor previously identified as the central regulator of Schwann cell myelination. Both the activating function of Sox10 as well as its cooperation with Krox20 were confirmed in vivo. We conclude that the employment of Sox10 and Krox20 as regulators of structural myelin sheath components and genes associated with the node of Ranvier is one way of ensuring a biologically meaningful coordinated formation of both structures during peripheral myelination.


Subject(s)
Schwann Cells , Cell Line , Myelin Sheath , Neuroglia , Transcription Factors/genetics
18.
Nucleic Acids Res ; 47(12): 6208-6224, 2019 07 09.
Article in English | MEDLINE | ID: mdl-31081019

ABSTRACT

Differentiating oligodendrocytes generate myelin to ensure rapid saltatory conduction in the vertebrate central nervous system. Although oligodendroglial differentiation and myelination are accompanied by dramatic chromatin reorganizations, previously studied chromatin remodelers had only limited direct effects on the process. To study the functional significance of chromatin changes for myelination and identify relevant remodelers, we deleted Ep400, the central ATP-hydrolyzing subunit of the TIP60/EP400 complex, at defined times of mouse oligodendrocyte development. Whereas Ep400-deficient oligodendrocyte precursors develop normally, terminal differentiation and myelination are dramatically impaired. Mechanistically, Ep400 interacts with transcription factor Sox10, binds to regulatory regions of the Myrf gene and is required to induce this central transcriptional regulator of the myelination program. In addition to reduced and aberrant myelin formation, oligodendrocytes exhibit increased DNA damage and apoptosis so that numbers never reach wildtype levels during the short lifespan of Ep400-deficient mice. Ep400 deletion in already mature oligodendrocytes remains phenotypically inapparent arguing that Ep400 is dispensable for myelin maintenance. Given its essential function in myelin formation, modulation of Ep400 activity may be beneficial in conditions such as multiple sclerosis where this process is compromised.


Subject(s)
DNA Helicases/physiology , DNA-Binding Proteins/physiology , Myelin Sheath/physiology , Oligodendroglia/cytology , Spinal Cord/cytology , Animals , Brain/cytology , Brain/embryology , Brain/growth & development , Brain/metabolism , Cell Differentiation , Cell Survival , Cells, Cultured , DNA Helicases/genetics , DNA Helicases/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Regulatory Networks , HEK293 Cells , Humans , Mice, Transgenic , Myelin Sheath/ultrastructure , Oligodendroglia/metabolism , Rats , Rats, Wistar , Spinal Cord/embryology , Spinal Cord/growth & development , Spinal Cord/metabolism
19.
Glia ; 68(8): 1596-1603, 2020 08.
Article in English | MEDLINE | ID: mdl-31837180

ABSTRACT

Schwann cells develop from neural crest cells in an ordered series of events and give rise to myelinating and nonmyelinating subtypes. In their mature state, myelinating Schwann cells produce myelin sheaths that provide trophic support to axons and allow saltatory conduction in the vertebrate peripheral nervous system. Each step of Schwann cell development requires defined changes in chromatin structure that are catalyzed by chromatin remodeling complexes. Over the last years, all major types of chromatin remodeling complexes have been detected in Schwann cells and several have been functionally analyzed. SWI/SNF-type, CHD-type, and INO80/SWR-type chromatin remodelers in particular have been shown to interact with multiple cell-type specific transcription factors and histone modifiers and to be important regulators of Schwann cell development. As a result of different recruitment strategies, each chromatin remodeler targets defined genomic areas and impacts unique mechanisms at specific stages of Schwann cell development. Chromatin remodeling complexes undoubtedly constitute essential components of the Schwann cell regulatory network.


Subject(s)
Cell Differentiation/physiology , Chromatin Assembly and Disassembly/physiology , Myelin Sheath/metabolism , Schwann Cells/metabolism , Animals , Chromatin/metabolism , Humans , Neurogenesis/physiology , Transcription Factors/metabolism
20.
Glia ; 68(10): 2015-2027, 2020 10.
Article in English | MEDLINE | ID: mdl-32176386

ABSTRACT

Oligodendrocytes wrap and physically shield axons of the central nervous system with myelin sheaths, resulting in rapid signal transduction and accurate neuronal function. The complex oligodendroglial development from immature oligodendrocyte precursor cells (OPCs) to myelinating oligodendrocytes (OLs) is profoundly dependent on the activity of transcription factors of the Sox protein family. Target genes of the crucial regulator Sox10 have recently been expanded to microRNAs. Here, we report miR-204 as a novel transcriptional target of Sox10. Regulatory regions of miR-204 show responsiveness to and binding of Sox10 in reporter gene assays and electromobility shift assays. Once expressed, miR-204 inhibits OPC proliferation and facilitates differentiation into OLs in the presence of Sox10 as evident from overexpression in primary rat and mouse oligodendroglial cultures. Phenotypes are at least in part caused by miR-204-dependent repression of the pro-proliferative Ccnd2 and the differentiation inhibiting Sox4. These findings argue that the transcriptional activator Sox10 forces oligodendroglial cells to exit the cell cycle and start differentiation by gene inhibition via miR-204 induction.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/physiology , MicroRNAs/metabolism , Oligodendroglia/metabolism , Animals , Animals, Newborn , Cells, Cultured , HEK293 Cells , Humans , Mice , Mice, Inbred C3H , MicroRNAs/genetics , Rats
SELECTION OF CITATIONS
SEARCH DETAIL