Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Glia ; 2024 Apr 13.
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.

2.
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
3.
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
4.
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
5.
Glia ; 68(5): 932-946, 2020 05.
Article in English | MEDLINE | ID: mdl-31724774

ABSTRACT

The high-mobility-group (HMG)-domain protein Sox9 is one of few transcription factors implicated in gliogenesis in the vertebrate central nervous system. To further study the role of Sox9 in early spinal cord development, we generated a mouse that allows expression of Sox9 in a temporally and spatially controlled manner. Using this mouse, we show that premature Sox9 expression in neural precursor cells disrupted the neuroepithelium of the ventricular zone. Sox9 also compromised development and survival of neuronal precursors and neurons. Additionally, we observed in these mice substantial increases in oligodendroglial and astroglial cells. Reversing the normal order of appearance of essential transcriptional regulators during oligodendrogenesis, Sox10 preceded Olig2. Our study reinforces the notion that Sox9 has a strong gliogenic activity. It also argues that Sox9 expression has to be tightly controlled to prevent negative effects on early spinal cord structure and neuronal development.


Subject(s)
Astrocytes/metabolism , Oligodendroglia/metabolism , SOX9 Transcription Factor/metabolism , Spinal Cord/metabolism , Animals , Cell Line, Tumor , Mice , Mice, Transgenic , Oligodendrocyte Transcription Factor 2/genetics , Oligodendrocyte Transcription Factor 2/metabolism , SOX9 Transcription Factor/genetics , Spinal Cord/growth & development
6.
Nat Commun ; 10(1): 2361, 2019 05 29.
Article in English | MEDLINE | ID: mdl-31142747

ABSTRACT

Schwann cells ensure efficient nerve impulse conduction in the peripheral nervous system. Their development is accompanied by defined chromatin changes, including variant histone deposition and redistribution. To study the importance of variant histones for Schwann cell development, we altered their genomic distribution by conditionally deleting Ep400, the central subunit of the Tip60/Ep400 complex. Ep400 absence causes peripheral neuropathy in mice, characterized by terminal differentiation defects in myelinating and non-myelinating Schwann cells and immune cell activation. Variant histone H2A.Z is differently distributed throughout the genome and remains at promoters of Tfap2a, Pax3 and other transcriptional regulator genes with transient function at earlier developmental stages. Tfap2a deletion in Ep400-deficient Schwann cells causes a partial rescue arguing that continued expression of early regulators mediates the phenotypic defects. Our results show that proper genomic distribution of variant histones is essential for Schwann cell differentiation, and assign importance to Ep400-containing chromatin remodelers in the process.


Subject(s)
Histones/metabolism , Peripheral Nervous System Diseases/genetics , Schwann Cells/metabolism , Sciatic Nerve/metabolism , Transcription Factors/genetics , Animals , Chromatin Assembly and Disassembly , DNA Helicases , DNA-Binding Proteins , Gene Expression Regulation, Developmental , Mice , Mice, Transgenic , PAX3 Transcription Factor/genetics , PAX3 Transcription Factor/metabolism , Peripheral Nervous System Diseases/metabolism , Peripheral Nervous System Diseases/pathology , Promoter Regions, Genetic , Sciatic Nerve/pathology , Transcription Factor AP-2/genetics , Transcription Factor AP-2/metabolism
8.
Nat Commun ; 9(1): 899, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29500351

ABSTRACT

Oligodendrocytes produce myelin for rapid transmission and saltatory conduction of action potentials in the vertebrate central nervous system. Activation of the myelination program requires several transcription factors including Sox10, Olig2, and Nkx2.2. Functional interactions among them are poorly understood and important components of the regulatory network are still unknown. Here, we identify Nfat proteins as Sox10 targets and regulators of oligodendroglial differentiation in rodents and humans. Overall levels and nuclear fraction increase during differentiation. Inhibition of Nfat activity impedes oligodendrocyte differentiation in vitro and in vivo. On a molecular level, Nfat proteins cooperate with Sox10 to relieve reciprocal repression of Olig2 and Nkx2.2 as precondition for oligodendroglial differentiation and myelination. As Nfat activity depends on calcium-dependent activation of calcineurin signaling, regulatory network and oligodendroglial differentiation become sensitive to calcium signals. NFAT proteins are also detected in human oligodendrocytes, downregulated in active multiple sclerosis lesions and thus likely relevant in demyelinating disease.


Subject(s)
Calcineurin/metabolism , Cell Differentiation , Myelin Sheath/metabolism , NFATC Transcription Factors/metabolism , Oligodendroglia/cytology , Oligodendroglia/metabolism , Signal Transduction , Animals , Conserved Sequence , Evolution, Molecular , Gene Expression Regulation , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/metabolism , Humans , Mice , Nuclear Proteins , Oligodendrocyte Transcription Factor 2/metabolism , Rats , SOXE Transcription Factors/metabolism , Transcription Factors/metabolism , Zebrafish Proteins
9.
PLoS Genet ; 11(2): e1005008, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25680202

ABSTRACT

Oligodendrocytes are the myelinating glia of the central nervous system and ensure rapid saltatory conduction. Shortage or loss of these cells leads to severe malfunctions as observed in human leukodystrophies and multiple sclerosis, and their replenishment by reprogramming or cell conversion strategies is an important research aim. Using a transgenic approach we increased levels of the transcription factor Sox10 throughout the mouse embryo and thereby prompted Fabp7-positive glial cells in dorsal root ganglia of the peripheral nervous system to convert into cells with oligodendrocyte characteristics including myelin gene expression. These rarely studied and poorly characterized satellite glia did not go through a classic oligodendrocyte precursor cell stage. Instead, Sox10 directly induced key elements of the regulatory network of differentiating oligodendrocytes, including Olig2, Olig1, Nkx2.2 and Myrf. An upstream enhancer mediated the direct induction of the Olig2 gene. Unlike Sox10, Olig2 was not capable of generating oligodendrocyte-like cells in dorsal root ganglia. Our findings provide proof-of-concept that Sox10 can convert conducive cells into oligodendrocyte-like cells in vivo and delineates options for future therapeutic strategies.


Subject(s)
Cell Differentiation/genetics , Central Nervous System/metabolism , Multiple Sclerosis/genetics , SOXE Transcription Factors/genetics , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Central Nervous System/pathology , Embryo, Mammalian , Fatty Acid-Binding Protein 7 , Fatty Acid-Binding Proteins/genetics , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Humans , Mice , Multiple Sclerosis/pathology , Nerve Tissue Proteins/genetics , Neuroglia , Nuclear Proteins , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/metabolism , SOXE Transcription Factors/metabolism , Spinal Cord/metabolism , Spinal Cord/pathology , Transcription Factors/genetics , Zebrafish Proteins
10.
Dev Cell ; 23(1): 193-201, 2012 Jul 17.
Article in English | MEDLINE | ID: mdl-22814607

ABSTRACT

Schwann cells produce myelin sheaths and thereby permit rapid saltatory conductance in the vertebrate peripheral nervous system. Their stepwise differentiation from neural crest cells is driven by a defined set of transcription factors. How this is linked to chromatin changes is not well understood. Here we show that the glial transcription factor Sox10 functions in Schwann cells by recruiting Brg1-containing chromatin-remodeling complexes via Baf60a to regulatory regions of Oct6 and Krox20 genes. It thereby stimulates expression of these transcriptional regulators that then cooperate with Sox10 to convert immature into myelinating Schwann cells. The functional interaction between Sox10 and Brg1 is evident from gain- and loss-of-function studies, similar neuropathies in the corresponding mouse mutants, and an aggravated neuropathy in compound mutants. Our results demonstrate that the transcription factor-mediated recruitment of the chromatin-remodeling machinery to specific genomic loci is an essential driving force for Schwann cell differentiation and myelination.


Subject(s)
Cell Differentiation/physiology , Chromatin Assembly and Disassembly/physiology , DNA Helicases/physiology , Myelin Sheath/physiology , Nuclear Proteins/physiology , Schwann Cells/cytology , Schwann Cells/metabolism , Transcription Factors/physiology , Animals , Cell Line, Tumor , Chick Embryo , Chickens , DNA Helicases/genetics , HEK293 Cells , Humans , Mice , Mice, Transgenic , Myelin Sheath/ultrastructure , Nuclear Proteins/genetics , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , SOXE Transcription Factors/physiology , Transcription Factors/genetics
11.
Nucleic Acids Res ; 36(17): 5427-40, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18703590

ABSTRACT

The transcription factor Sox5 has previously been shown in chicken to be expressed in early neural crest cells and neural crest-derived peripheral glia. Here, we show in mouse that Sox5 expression also continues after neural crest specification in the melanocyte lineage. Despite its continued expression, Sox5 has little impact on melanocyte development on its own as generation of melanoblasts and melanocytes is unaltered in Sox5-deficient mice. Loss of Sox5, however, partially rescued the strongly reduced melanoblast generation and marker gene expression in Sox10 heterozygous mice arguing that Sox5 functions in the melanocyte lineage by modulating Sox10 activity. This modulatory activity involved Sox5 binding and recruitment of CtBP2 and HDAC1 to the regulatory regions of melanocytic Sox10 target genes and direct inhibition of Sox10-dependent promoter activation. Both binding site competition and recruitment of corepressors thus help Sox5 to modulate the activity of Sox10 in the melanocyte lineage.


Subject(s)
DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Gene Expression Regulation , High Mobility Group Proteins/antagonists & inhibitors , Melanocytes/metabolism , Nuclear Proteins/metabolism , Transcription Factors/antagonists & inhibitors , Animals , Cell Line, Tumor , DNA-Binding Proteins/genetics , Embryo, Mammalian/metabolism , Heterozygote , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Mice , Mice, Inbred C3H , Mice, Knockout , Nuclear Proteins/genetics , Promoter Regions, Genetic , Repressor Proteins/metabolism , Response Elements , SOXD Transcription Factors , SOXE Transcription Factors , Transcription Factors/genetics , Transcription Factors/metabolism
12.
Dev Cell ; 11(5): 697-709, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17084361

ABSTRACT

The myelin-forming oligodendrocytes are an excellent model to study transcriptional regulation of specification events, lineage progression, and terminal differentiation in the central nervous system. Here, we show that the group D Sox transcription factors Sox5 and Sox6 jointly and cell-autonomously regulate several stages of oligodendrocyte development in the mouse spinal cord. They repress specification and terminal differentiation and influence migration patterns. As a consequence, oligodendrocyte precursors and terminally differentiating oligodendrocytes appear precociously in spinal cords deficient for both Sox proteins. Sox5 and Sox6 have opposite functions than the group E Sox proteins Sox9 and Sox10, which promote oligodendrocyte specification and terminal differentiation. Both genetic as well as molecular evidence suggests that Sox5 and Sox6 directly interfere with the function of group E Sox proteins. Our studies reveal a complex regulatory network between different groups of Sox proteins that is essential for proper progression of oligodendrocyte development.


Subject(s)
DNA-Binding Proteins/physiology , High Mobility Group Proteins/physiology , Nuclear Proteins/physiology , Oligodendroglia/physiology , Spinal Cord/cytology , Transcription Factors/physiology , Animals , Cell Differentiation , Cell Lineage , DNA-Binding Proteins/genetics , Gene Expression Regulation, Developmental , High Mobility Group Proteins/genetics , Mice , Mice, Knockout , Mutation , Myelin Sheath/metabolism , Nuclear Proteins/genetics , Oligodendroglia/cytology , SOX9 Transcription Factor , SOXD Transcription Factors , SOXE Transcription Factors , Spinal Cord/embryology , Spinal Cord/metabolism , Transcription Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...