Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
1.
Glia ; 72(8): 1518-1540, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38794866

ABSTRACT

In the central nervous system, the formation of myelin by oligodendrocytes (OLs) relies on the switch from the polymerization of the actin cytoskeleton to its depolymerization. The molecular mechanisms that trigger this switch have yet to be elucidated. Here, we identified P21-activated kinase 1 (PAK1) as a major regulator of actin depolymerization in OLs. Our results demonstrate that PAK1 accumulates in OLs in a kinase-inhibited form, triggering actin disassembly and, consequently, myelin membrane expansion. Remarkably, proteomic analysis of PAK1 binding partners enabled the identification of NF2/Merlin as its endogenous inhibitor. Our findings indicate that Nf2 knockdown in OLs results in PAK1 activation, actin polymerization, and a reduction in OL myelin membrane expansion. This effect is rescued by treatment with a PAK1 inhibitor. We also provide evidence that the specific Pak1 loss-of-function in oligodendroglia stimulates the thickening of myelin sheaths in vivo. Overall, our data indicate that the antagonistic actions of PAK1 and NF2/Merlin on the actin cytoskeleton of the OLs are critical for proper myelin formation. These findings have broad mechanistic and therapeutic implications in demyelinating diseases and neurodevelopmental disorders.


Subject(s)
Myelin Sheath , Oligodendroglia , p21-Activated Kinases , p21-Activated Kinases/metabolism , Oligodendroglia/metabolism , Animals , Myelin Sheath/metabolism , Neurofibromin 2/metabolism , Neurofibromin 2/genetics , Rats , Actins/metabolism , Cells, Cultured , Mice , Mice, Inbred C57BL , Actin Cytoskeleton/metabolism
2.
Proc Natl Acad Sci U S A ; 115(35): E8246-E8255, 2018 08 28.
Article in English | MEDLINE | ID: mdl-30108144

ABSTRACT

Oligodendrocyte precursor cells (OPCs) constitute the main proliferative cells in the adult brain, and deregulation of OPC proliferation-differentiation balance results in either glioma formation or defective adaptive (re)myelination. OPC differentiation requires significant genetic reprogramming, implicating chromatin remodeling. Mounting evidence indicates that chromatin remodelers play important roles during normal development and their mutations are associated with neurodevelopmental defects, with CHD7 haploinsuficiency being the cause of CHARGE syndrome and CHD8 being one of the strongest autism spectrum disorder (ASD) high-risk-associated genes. Herein, we report on uncharacterized functions of the chromatin remodelers Chd7 and Chd8 in OPCs. Their OPC-chromatin binding profile, combined with transcriptome and chromatin accessibility analyses of Chd7-deleted OPCs, demonstrates that Chd7 protects nonproliferative OPCs from apoptosis by chromatin closing and transcriptional repression of p53 Furthermore, Chd7 controls OPC differentiation through chromatin opening and transcriptional activation of key regulators, including Sox10, Nkx2.2, and Gpr17 However, Chd7 is dispensable for oligodendrocyte stage progression, consistent with Chd8 compensatory function, as suggested by their common chromatin-binding profiles and genetic interaction. Finally, CHD7 and CHD8 bind in OPCs to a majority of ASD risk-associated genes, suggesting an implication of oligodendrocyte lineage cells in ASD neurological defects. Our results thus offer new avenues to understand and modulate the CHD7 and CHD8 functions in normal development and disease.


Subject(s)
Chromatin Assembly and Disassembly , DNA-Binding Proteins/metabolism , Oligodendroglia/metabolism , Stem Cells/metabolism , Animals , Autism Spectrum Disorder/genetics , Autism Spectrum Disorder/metabolism , Autism Spectrum Disorder/pathology , CHARGE Syndrome/genetics , CHARGE Syndrome/metabolism , CHARGE Syndrome/pathology , Cell Survival , DNA-Binding Proteins/genetics , Homeobox Protein Nkx-2.2 , Homeodomain Proteins , Mice , Mice, Knockout , Nuclear Proteins , Oligodendroglia/pathology , Stem Cells/pathology , Transcription Factors
3.
J Neurosci ; 32(48): 17172-85, 2012 Nov 28.
Article in English | MEDLINE | ID: mdl-23197710

ABSTRACT

Oligodendrocytes are the myelin-forming cells of the vertebrate CNS. Little is known about the molecular control of region-specific oligodendrocyte development. Here, we show that oligodendrogenesis in the mouse rostral hindbrain, which is organized in a metameric series of rhombomere-derived (rd) territories, follows a rhombomere-specific pattern, with extensive production of oligodendrocytes in the pontine territory (r4d) and delayed and reduced oligodendrocyte production in the prepontine region (r2d, r3d). We demonstrate that segmental organization of oligodendrocytes is controlled by Hox genes, namely Hoxa2 and Hoxb2. Specifically, Hoxa2 loss of function induced a dorsoventral enlargement of the Olig2/Nkx2.2-expressing oligodendrocyte progenitor domain, whereas conditional Hoxa2 overexpression in the Olig2(+) domain inhibited oligodendrogenesis throughout the brain. In contrast, Hoxb2 deletion resulted in a reduction of the pontine oligodendrogenic domain. Compound Hoxa2(-/-)/Hoxb2(-/-) mutant mice displayed the phenotype of Hoxb2(-/-) mutants in territories coexpressing Hoxa2 and Hoxb2 (rd3, rd4), indicating that Hoxb2 antagonizes Hoxa2 during rostral hindbrain oligodendrogenesis. This study provides the first in vivo evidence that Hox genes determine oligodendrocyte regional identity in the mammalian brain.


Subject(s)
Cell Differentiation/genetics , Homeodomain Proteins/genetics , Oligodendroglia/metabolism , Rhombencephalon/metabolism , Transcription Factors/genetics , Animals , Body Patterning/genetics , Cell Proliferation , Gene Expression Regulation, Developmental , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/metabolism , Mice , Mice, Knockout , Myelin Sheath/genetics , Myelin Sheath/metabolism , Rhombencephalon/embryology , Transcription Factors/metabolism
4.
Elife ; 112022 10 10.
Article in English | MEDLINE | ID: mdl-36214451

ABSTRACT

The differentiation of oligodendroglia from oligodendrocyte precursor cells (OPCs) to complex and extensive myelinating oligodendrocytes (OLs) is a multistep process that involves large-scale morphological changes with significant strain on the cytoskeleton. While key chromatin and transcriptional regulators of differentiation have been identified, their target genes responsible for the morphological changes occurring during OL myelination are still largely unknown. Here, we show that the regulator of focal adhesion, Tensin3 (Tns3), is a direct target gene of Olig2, Chd7, and Chd8, transcriptional regulators of OL differentiation. Tns3 is transiently upregulated and localized to cell processes of immature OLs, together with integrin-ß1, a key mediator of survival at this transient stage. Constitutive <i>Tns3</i> loss of function leads to reduced viability in mouse and humans, with surviving knockout mice still expressing Tns3 in oligodendroglia. Acute deletion of <i>Tns3</i> in vivo, either in postnatal neural stem cells (NSCs) or in OPCs, leads to a twofold reduction in OL numbers. We find that the transient upregulation of Tns3 is required to protect differentiating OPCs and immature OLs from cell death by preventing the upregulation of p53, a key regulator of apoptosis. Altogether, our findings reveal a specific time window during which transcriptional upregulation of Tns3 in immature OLs is required for OL differentiation likely by mediating integrin-ß1 survival signaling to the actin cytoskeleton as OL undergo the large morphological changes required for their terminal differentiation.


Subject(s)
Focal Adhesions , Tumor Suppressor Protein p53 , Humans , Animals , Mice , Focal Adhesions/metabolism , Tumor Suppressor Protein p53/genetics , Oligodendroglia/metabolism , Cell Differentiation/genetics , Mice, Knockout , Transcription Factors/metabolism , Chromatin/metabolism , Integrins/metabolism
5.
Nat Neurosci ; 19(5): 678-689, 2016 05.
Article in English | MEDLINE | ID: mdl-26928066

ABSTRACT

Mutations in CHD7, encoding ATP-dependent chromodomain helicase DNA-binding protein 7, in CHARGE syndrome lead to multiple congenital anomalies, including craniofacial malformations, neurological dysfunction and growth delay. Mechanisms underlying the CNS phenotypes remain poorly understood. We found that Chd7 is a direct transcriptional target of oligodendrogenesis-promoting factors Olig2 and Smarca4/Brg1 and is required for proper onset of CNS myelination and remyelination. Genome-occupancy analyses in mice, coupled with transcriptome profiling, revealed that Chd7 interacted with Sox10 and targeted the enhancers of key myelinogenic genes. These analyses identified previously unknown Chd7 targets, including bone formation regulators Osterix (also known as Sp7) and Creb3l2, which are also critical for oligodendrocyte maturation. Thus, Chd7 coordinates with Sox10 to regulate the initiation of myelinogenesis and acts as a molecular nexus of regulatory networks that account for the development of a seemingly diverse array of lineages, including oligodendrocytes and osteoblasts, pointing to previously uncharacterized Chd7 functions in white matter pathogenesis in CHARGE syndrome.


Subject(s)
CHARGE Syndrome/physiopathology , DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental/physiology , Myelin Sheath/physiology , Neurogenesis/physiology , SOXE Transcription Factors/physiology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Gene Expression Profiling , Mice , Mice, Knockout , Mutation , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Oligodendrocyte Transcription Factor 2 , Oligodendroglia/metabolism , Oligodendroglia/physiology , SOXE Transcription Factors/metabolism , Sp7 Transcription Factor , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL