Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 139(2): 366-79, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19837037

ABSTRACT

Current opinion holds that pigment cells, melanocytes, are derived from neural crest cells produced at the dorsal neural tube and that migrate under the epidermis to populate all parts of the skin. Here, we identify growing nerves projecting throughout the body as a stem/progenitor niche containing Schwann cell precursors (SCPs) from which large numbers of skin melanocytes originate. SCPs arise as a result of lack of neuronal specification by Hmx1 homeobox gene function in the neural crest ventral migratory pathway. Schwann cell and melanocyte development share signaling molecules with both the glial and melanocyte cell fates intimately linked to nerve contact and regulated in an opposing manner by Neuregulin and soluble signals including insulin-like growth factor and platelet-derived growth factor. These results reveal SCPs as a cellular origin of melanocytes, and have broad implications on the molecular mechanisms regulating skin pigmentation during development, in health and pigmentation disorders.


Subject(s)
Melanocytes/cytology , Schwann Cells/cytology , Skin/innervation , Animals , Cell Differentiation , Cell Movement , Homeodomain Proteins , Mice , Neuroglia , Receptor, ErbB-3/metabolism , Stem Cells/cytology , Transcription Factors/metabolism
2.
Hum Mol Genet ; 29(8): 1253-1273, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32129442

ABSTRACT

Some mutations affecting dynamin 2 (DNM2) can cause dominantly inherited Charcot-Marie-Tooth (CMT) neuropathy. Here, we describe the analysis of mice carrying the DNM2 K562E mutation which has been associated with dominant-intermediate CMT type B (CMTDIB). Contrary to our expectations, heterozygous DNM2 K562E mutant mice did not develop definitive signs of an axonal or demyelinating neuropathy. Rather, we found a primary myopathy-like phenotype in these mice. A likely interpretation of these results is that the lack of a neuropathy in this mouse model has allowed the unmasking of a primary myopathy due to the DNM2 K562E mutation which might be overshadowed by the neuropathy in humans. Consequently, we hypothesize that a primary myopathy may also contribute to the disease mechanism in some CMTDIB patients. We propose that these findings should be considered in the evaluation of patients, the determination of the underlying disease processes and the development of tailored potential treatment strategies.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Dynamin II/deficiency , Muscular Diseases/genetics , Myopathies, Structural, Congenital/genetics , Animals , Axons/metabolism , Axons/pathology , Charcot-Marie-Tooth Disease/pathology , Dynamin II/genetics , Heterozygote , Humans , Mice , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Muscular Diseases/pathology , Mutation/genetics , Myopathies, Structural, Congenital/pathology , Phenotype
3.
Nature ; 513(7519): 551-4, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25079316

ABSTRACT

Mesenchymal stem cells occupy niches in stromal tissues where they provide sources of cells for specialized mesenchymal derivatives during growth and repair. The origins of mesenchymal stem cells have been the subject of considerable discussion, and current consensus holds that perivascular cells form mesenchymal stem cells in most tissues. The continuously growing mouse incisor tooth offers an excellent model to address the origin of mesenchymal stem cells. These stem cells dwell in a niche at the tooth apex where they produce a variety of differentiated derivatives. Cells constituting the tooth are mostly derived from two embryonic sources: neural crest ectomesenchyme and ectodermal epithelium. It has been thought for decades that the dental mesenchymal stem cells giving rise to pulp cells and odontoblasts derive from neural crest cells after their migration in the early head and formation of ectomesenchymal tissue. Here we show that a significant population of mesenchymal stem cells during development, self-renewal and repair of a tooth are derived from peripheral nerve-associated glia. Glial cells generate multipotent mesenchymal stem cells that produce pulp cells and odontoblasts. By combining a clonal colour-coding technique with tracing of peripheral glia, we provide new insights into the dynamics of tooth organogenesis and growth.


Subject(s)
Cell Differentiation , Cell Lineage , Incisor/cytology , Mesenchymal Stem Cells/cytology , Neuroglia/cytology , Animals , Cell Tracking , Clone Cells/cytology , Dental Pulp/cytology , Female , Incisor/embryology , Male , Mice , Models, Biological , Neural Crest/cytology , Odontoblasts/cytology , Regeneration , Schwann Cells/cytology
4.
J Neurosci ; 38(20): 4811-4828, 2018 05 16.
Article in English | MEDLINE | ID: mdl-29695414

ABSTRACT

Schwann cells (SCs) are endowed with a remarkable plasticity. When peripheral nerves are injured, SCs dedifferentiate and acquire new functions to coordinate nerve repair as so-called repair SCs. Subsequently, SCs redifferentiate to remyelinate regenerated axons. Given the similarities between SC dedifferentiation/redifferentiation in injured nerves and in demyelinating neuropathies, elucidating the signals involved in SC plasticity after nerve injury has potentially wider implications. c-Jun has emerged as a key transcription factor regulating SC dedifferentiation and the acquisition of repair SC features. However, the upstream pathways that control c-Jun activity after nerve injury are largely unknown. We report that the mTORC1 pathway is transiently but robustly reactivated in dedifferentiating SCs. By inducible genetic deletion of the functionally crucial mTORC1-subunit Raptor in mouse SCs (including male and female animals), we found that mTORC1 reactivation is necessary for proper myelin clearance, SC dedifferentiation, and consequently remyelination, without major alterations in the inflammatory response. In the absence of mTORC1 signaling, c-Jun failed to be upregulated correctly. Accordingly, a c-Jun binding motif was found to be enriched in promoters of genes with reduced expression in injured mutants. Furthermore, using cultured SCs, we found that mTORC1 is involved in c-Jun regulation by promoting its translation, possibly via the eIF4F-subunit eIF4A. These results provide evidence that proper c-Jun elevation after nerve injury involves also mTORC1-dependent post-transcriptional regulation to ensure timely dedifferentiation of SCs.SIGNIFICANCE STATEMENT A crucial evolutionary acquisition of vertebrates is the envelopment of axons in myelin sheaths produced by oligodendrocytes in the CNS and Schwann cells (SCs) in the PNS. When myelin is damaged, conduction of action potentials along axons slows down or is blocked, leading to debilitating diseases. Unlike oligodendrocytes, SCs have a high regenerative potential, granted by their remarkable plasticity. Thus, understanding the mechanisms underlying SC plasticity may uncover new therapeutic targets in nerve regeneration and demyelinating diseases. Our work reveals that reactivation of the mTORC1 pathway in SCs is essential for efficient SC dedifferentiation after nerve injury. Accordingly, modulating this signaling pathway might be of therapeutic relevance in peripheral nerve injury and other diseases.


Subject(s)
Cell Dedifferentiation , Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/metabolism , Proto-Oncogene Proteins c-jun/biosynthesis , Schwann Cells , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Activation, Metabolic/genetics , Activation, Metabolic/physiology , Animals , Eukaryotic Initiation Factor-4F/genetics , Female , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Knockout , Mutation/genetics , Myelin Sheath/metabolism , Proto-Oncogene Proteins c-jun/genetics , Rats , Rats, Sprague-Dawley , Regulatory-Associated Protein of mTOR/genetics , Signal Transduction/physiology
5.
Nature ; 493(7431): 226-30, 2013 Jan 10.
Article in English | MEDLINE | ID: mdl-23201681

ABSTRACT

Mechanisms controlling the proliferative activity of neural stem and progenitor cells (NSPCs) have a pivotal role to ensure life-long neurogenesis in the mammalian brain. How metabolic programs are coupled with NSPC activity remains unknown. Here we show that fatty acid synthase (Fasn), the key enzyme of de novo lipogenesis, is highly active in adult NSPCs and that conditional deletion of Fasn in mouse NSPCs impairs adult neurogenesis. The rate of de novo lipid synthesis and subsequent proliferation of NSPCs is regulated by Spot14, a gene previously implicated in lipid metabolism, that we found to be selectively expressed in low proliferating adult NSPCs. Spot14 reduces the availability of malonyl-CoA, which is an essential substrate for Fasn to fuel lipogenesis. Thus, we identify here a functional coupling between the regulation of lipid metabolism and adult NSPC proliferation.


Subject(s)
Adult Stem Cells/metabolism , Fatty Acid Synthases/metabolism , Lipogenesis , Neural Stem Cells/metabolism , Adult Stem Cells/cytology , Animals , Cell Proliferation , Dentate Gyrus/metabolism , Fatty Acid Synthases/deficiency , Fatty Acid Synthases/genetics , Gene Expression Profiling , Hippocampus/cytology , Hippocampus/metabolism , Malonyl Coenzyme A/metabolism , Mice , Mice, Transgenic , Neural Stem Cells/cytology , Neurogenesis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Glia ; 66(4): 693-707, 2018 04.
Article in English | MEDLINE | ID: mdl-29210103

ABSTRACT

Myelinating cells surround axons to accelerate the propagation of action potentials, to support axonal health, and to refine neural circuits. Myelination is metabolically demanding and, consistent with this notion, mTORC1-a signaling hub coordinating cell metabolism-has been implicated as a key signal for myelination. Here, we will discuss metabolic aspects of myelination, illustrate the main metabolic processes regulated by mTORC1, and review advances on the role of mTORC1 in myelination of the central nervous system and the peripheral nervous system. Recent progress has revealed a complex role of mTORC1 in myelinating cells that includes, besides positive regulation of myelin growth, additional critical functions in the stages preceding active myelination. Based on the available evidence, we will also highlight potential nonoverlapping roles between mTORC1 and its known main upstream pathways PI3K-Akt, Mek-Erk1/2, and AMPK in myelinating cells. Finally, we will discuss signals that are already known or hypothesized to be responsible for the regulation of mTORC1 activity in myelinating cells.


Subject(s)
Myelin Sheath/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Humans
7.
Glia ; 66(12): 2632-2644, 2018 12.
Article in English | MEDLINE | ID: mdl-30295958

ABSTRACT

Proper function of the nervous system depends on myelination. In peripheral nerves, Schwann cells (SCs) myelinate axons and the miRNA biogenesis pathway is required for developmental myelination and myelin maintenance. However, regulatory roles of this pathway at different stages of myelination are only partially understood. We addressed the requirement of the core miRNA biogenesis pathway components Dgcr8, Drosha, and Dicer in developing and adult SCs using mouse mutants with a comparative genetics and transcriptomics approach. We found that the microprocessor components Dgcr8 and Drosha are crucial for axonal radial sorting and to establish correct SC numbers upon myelination. Transcriptome analyses revealed a requirement of the microprocessor to prevent aberrantly increased expression of injury-response genes. Those genes are predicted targets of abundant miRNAs in sciatic nerves (SNs) during developmental myelination. In agreement, Dgcr8 and Dicer are required for proper maintenance of the myelinated SC state, where abundant miRNAs in adult SNs are predicted to target injury-response genes. We conclude that the miRNA biogenesis pathway in SCs is crucial for preventing inappropriate activity of injury-response genes in developing and adult SCs.


Subject(s)
Gene Expression Regulation, Developmental/physiology , MicroRNAs/metabolism , Schwann Cells/pathology , Sciatic Neuropathy/pathology , Sciatic Neuropathy/prevention & control , Signal Transduction/physiology , Animals , Animals, Newborn , Connexins/genetics , Connexins/metabolism , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/genetics , Microscopy, Electron , Myelin Sheath/pathology , Myelin Sheath/ultrastructure , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribonuclease III/genetics , Ribonuclease III/metabolism , Schwann Cells/metabolism , Schwann Cells/ultrastructure , Transcription Factors/metabolism , Gap Junction beta-1 Protein
8.
PLoS Biol ; 13(9): e1002258, 2015.
Article in English | MEDLINE | ID: mdl-26406915

ABSTRACT

The pathogenesis of peripheral neuropathies in adults is linked to maintenance mechanisms that are not well understood. Here, we elucidate a novel critical maintenance mechanism for Schwann cell (SC)-axon interaction. Using mouse genetics, ablation of the transcriptional regulators histone deacetylases 1 and 2 (HDAC1/2) in adult SCs severely affected paranodal and nodal integrity and led to demyelination/remyelination. Expression levels of the HDAC1/2 target gene myelin protein zero (P0) were reduced by half, accompanied by altered localization and stability of neurofascin (NFasc)155, NFasc186, and loss of Caspr and septate-like junctions. We identify P0 as a novel binding partner of NFasc155 and NFasc186, both in vivo and by in vitro adhesion assay. Furthermore, we demonstrate that HDAC1/2-dependent P0 expression is crucial for the maintenance of paranodal/nodal integrity and axonal function through interaction of P0 with neurofascins. In addition, we show that the latter mechanism is impaired by some P0 mutations that lead to late onset Charcot-Marie-Tooth disease.


Subject(s)
Cell Adhesion Molecules/metabolism , Charcot-Marie-Tooth Disease/genetics , Myelin P0 Protein/genetics , Myelin Sheath/physiology , Nerve Growth Factors/metabolism , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Charcot-Marie-Tooth Disease/enzymology , Gene Knockout Techniques , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Humans , Mice
9.
Nature ; 485(7399): 517-21, 2012 Apr 29.
Article in English | MEDLINE | ID: mdl-22622581

ABSTRACT

Oligodendrocytes, the myelin-forming glial cells of the central nervous system, maintain long-term axonal integrity. However, the underlying support mechanisms are not understood. Here we identify a metabolic component of axon-glia interactions by generating conditional Cox10 (protoheme IX farnesyltransferase) mutant mice, in which oligodendrocytes and Schwann cells fail to assemble stable mitochondrial cytochrome c oxidase (COX, also known as mitochondrial complex IV). In the peripheral nervous system, Cox10 conditional mutants exhibit severe neuropathy with dysmyelination, abnormal Remak bundles, muscle atrophy and paralysis. Notably, perturbing mitochondrial respiration did not cause glial cell death. In the adult central nervous system, we found no signs of demyelination, axonal degeneration or secondary inflammation. Unlike cultured oligodendrocytes, which are sensitive to COX inhibitors, post-myelination oligodendrocytes survive well in the absence of COX activity. More importantly, by in vivo magnetic resonance spectroscopy, brain lactate concentrations in mutants were increased compared with controls, but were detectable only in mice exposed to volatile anaesthetics. This indicates that aerobic glycolysis products derived from oligodendrocytes are rapidly metabolized within white matter tracts. Because myelinated axons can use lactate when energy-deprived, our findings suggest a model in which axon-glia metabolic coupling serves a physiological function.


Subject(s)
Axons/physiology , Glycolysis , Myelin Sheath/metabolism , Oligodendroglia/metabolism , Action Potentials , Alkyl and Aryl Transferases/deficiency , Alkyl and Aryl Transferases/genetics , Alkyl and Aryl Transferases/metabolism , Animals , Brain/cytology , Brain/metabolism , Cell Respiration , Cell Survival , Demyelinating Diseases/enzymology , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Electron Transport Complex IV/antagonists & inhibitors , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Lactic Acid/metabolism , Magnetic Resonance Spectroscopy , Membrane Proteins/deficiency , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mitochondria/enzymology , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/pathology , Mutant Proteins/genetics , Mutant Proteins/metabolism , Oligodendroglia/cytology , Oligodendroglia/drug effects , Oligodendroglia/enzymology , Protons , Schwann Cells/enzymology , Schwann Cells/metabolism , Time Factors
10.
J Neurosci ; 36(49): 12351-12367, 2016 12 07.
Article in English | MEDLINE | ID: mdl-27927955

ABSTRACT

Schwann cells (SCs) are essential for proper peripheral nerve development and repair, although the mechanisms regulating these processes are incompletely understood. We previously showed that the adhesion G protein-coupled receptor Gpr126/Adgrg6 is essential for SC development and myelination. Interestingly, the expression of Gpr126 is maintained in adult SCs, suggestive of a function in the mature nerve. We therefore investigated the role of Gpr126 in nerve repair by studying an inducible SC-specific Gpr126 knock-out mouse model. Here, we show that remyelination is severely delayed after nerve-crush injury. Moreover, we also observe noncell-autonomous defects in macrophage recruitment and axon regeneration in injured nerves following loss of Gpr126 in SCs. This work demonstrates that Gpr126 has critical SC-autonomous and SC-nonautonomous functions in remyelination and peripheral nerve repair. SIGNIFICANCE STATEMENT: Lack of robust remyelination represents one of the major barriers to recovery of neurological functions in disease or following injury in many disorders of the nervous system. Here we show that the adhesion class G protein-coupled receptor (GPCR) Gpr126/Adgrg6 is required for remyelination, macrophage recruitment, and axon regeneration following nerve injury. At least 30% of all approved drugs target GPCRs; thus, Gpr126 represents an attractive potential target to stimulate repair in myelin disease or following nerve injury.


Subject(s)
Peripheral Nerve Injuries/genetics , Peripheral Nerve Injuries/pathology , Receptors, G-Protein-Coupled/genetics , Schwann Cells/pathology , Animals , Axons , Mice , Mice, Knockout , Muscle, Skeletal/innervation , Muscle, Skeletal/pathology , Myelin Sheath , Nerve Crush , Nerve Regeneration , Neutrophil Infiltration , Sciatic Nerve/injuries
11.
Development ; 141(7): 1553-61, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24598164

ABSTRACT

Myelination allows rapid saltatory propagation of action potentials along the axon and is an essential prerequisite for the normal functioning of the nervous system. During peripheral nervous system (PNS) development, myelin-forming Schwann cells (SCs) generate radial lamellipodia to sort and ensheath axons. This process requires controlled cytoskeletal remodeling, and we show that SC lamellipodia formation depends on the function of profilin 1 (Pfn1), an actin-binding protein involved in microfilament polymerization. Pfn1 is inhibited upon phosphorylation by ROCK, a downstream effector of the integrin linked kinase pathway. Thus, a dramatic reduction of radial lamellipodia formation is observed in SCs lacking integrin-linked kinase or treated with the Rho/ROCK activator lysophosphatidic acid. Knocking down Pfn1 expression by lentiviral-mediated shRNA delivery impairs SC lamellipodia formation in vitro, suggesting a direct role for this protein in PNS myelination. Indeed, SC-specific gene ablation of Pfn1 in mice led to profound radial sorting and myelination defects, confirming a central role for this protein in PNS development. Our data identify Pfn1 as a key effector of the integrin linked kinase/Rho/ROCK pathway. This pathway, acting in parallel with integrin ß1/LCK/Rac1 and their effectors critically regulates SC lamellipodia formation, radial sorting and myelination during peripheral nervous system maturation.


Subject(s)
Myelin Sheath/physiology , Peripheral Nerves/physiology , Peripheral Nervous System/physiology , Profilins/physiology , Animals , Axonal Transport/genetics , Cells, Cultured , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenesis/genetics , Neuropeptides/physiology , Pseudopodia/genetics , Schwann Cells/physiology , rac1 GTP-Binding Protein/physiology
12.
J Neurosci ; 34(15): 5222-32, 2014 Apr 09.
Article in English | MEDLINE | ID: mdl-24719101

ABSTRACT

Controlling neural stem and progenitor cell (NSPC) proliferation is critical to maintain neurogenesis in the mammalian brain throughout life. However, it remains poorly understood how niche-derived cues such as ß1-integrin-mediated signaling are translated into NSPC-intrinsic molecular changes to regulate NSPC activity. Here we show that genetic deletion of integrin-linked kinase (ILK) increases NSPC proliferation through PINCH1/2-dependent enhancement of c-Jun N-terminal protein kinase activity in both neurogenic regions of the adult mouse brain. This effect downstream of ILK signaling is mediated through loss of Ras suppressor unit-1 (RSU-1), as virus-based reconstitution of RSU-1 expression rescued the ILK-dependent effects on NSPC proliferation. Thus, we here identified an intracellular signaling cascade linking extrinsic integrin-mediated signaling to NSPC proliferation and characterized a novel mechanism that regulates NSPC activity in the adult mammalian brain.


Subject(s)
Brain/metabolism , Cell Proliferation , Neural Stem Cells/metabolism , Protein Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Brain/growth & development , Cells, Cultured , Female , Gene Deletion , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , MAP Kinase Signaling System , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Neural Stem Cells/physiology , Neurogenesis , Protein Serine-Threonine Kinases/genetics , Stem Cell Niche , Transcription Factors/genetics , Transcription Factors/metabolism
13.
J Neurosci ; 34(25): 8432-48, 2014 Jun 18.
Article in English | MEDLINE | ID: mdl-24948799

ABSTRACT

The mammalian target of rapamycin (mTOR) pathway integrates multiple signals and regulates crucial cell functions via the molecular complexes mTORC1 and mTORC2. These complexes are functionally dependent on their raptor (mTORC1) or rictor (mTORC2) subunits. mTOR has been associated with oligodendrocyte differentiation and myelination downstream of the PI3K/Akt pathway, but the functional contributions of individual complexes are largely unknown. We show, by oligodendrocyte-specific genetic deletion of Rptor and/or Rictor in the mouse, that CNS myelination is mainly dependent on mTORC1 function, with minor mTORC2 contributions. Myelin-associated lipogenesis and protein gene regulation are strongly reliant on mTORC1. We found that also oligodendrocyte-specific overactivation of mTORC1, via ablation of tuberous sclerosis complex 1 (TSC1), causes hypomyelination characterized by downregulation of Akt signaling and lipogenic pathways. Our data demonstrate that a delicately balanced regulation of mTORC1 activation and action in oligodendrocytes is essential for CNS myelination, which has practical overtones for understanding CNS myelin disorders.


Subject(s)
Multiprotein Complexes/metabolism , Nerve Fibers, Myelinated/metabolism , Oligodendroglia/metabolism , Spinal Cord/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Central Nervous System/metabolism , Central Nervous System/pathology , Female , Male , Mechanistic Target of Rapamycin Complex 1 , Mice , Mice, Knockout , Mice, Transgenic , Nerve Fibers, Myelinated/pathology , Oligodendroglia/pathology , Spinal Cord/pathology
14.
J Neurosci ; 34(17): 6112-22, 2014 Apr 23.
Article in English | MEDLINE | ID: mdl-24760871

ABSTRACT

Schwann cells, the myelinating glia of the peripheral nervous system (PNS), originate from multipotent neural crest cells that also give rise to other cells, including neurons, melanocytes, chondrocytes, and smooth muscle cells. The transcription factor Sox10 is required for peripheral glia specification. However, all neural crest cells express Sox10 and the mechanisms directing neural crest cells into a specific lineage are poorly understood. We show here that histone deacetylases 1 and 2 (HDAC1/2) are essential for the specification of neural crest cells into Schwann cell precursors and satellite glia, which express the early determinants of their lineage myelin protein zero (P0) and/or fatty acid binding protein 7 (Fabp7). In neural crest cells, HDAC1/2 induced expression of the transcription factor Pax3 by binding and activating the Pax3 promoter. In turn, Pax3 was required to maintain high Sox10 levels and to trigger expression of Fabp7. In addition, HDAC1/2 were bound to the P0 promoter and activated P0 transcription. Consistently, in vivo genetic deletion of HDAC1/2 in mouse neural crest cells led to strongly decreased Sox10 expression, no detectable Pax3, virtually no satellite glia, and no Schwann cell precursors in dorsal root ganglia and peripheral nerves. Similarly, in vivo ablation of Pax3 in the mouse neural crest resulted in strongly reduced expression of Sox10 and Fabp7. Therefore, by controlling the expression of Pax3 and the concerted action of Pax3 and Sox10 on their target genes, HDAC1/2 direct the specification of neural crest cells into peripheral glia.


Subject(s)
Cell Differentiation/physiology , Histone Deacetylase 1/metabolism , Histone Deacetylase 2/metabolism , Neural Crest/metabolism , Neural Stem Cells/metabolism , Oligodendroglia/metabolism , Schwann Cells/metabolism , Animals , Gene Expression Regulation, Developmental , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Mice , Neural Crest/cytology , Neural Stem Cells/cytology , Oligodendroglia/cytology , PAX3 Transcription Factor , Paired Box Transcription Factors/genetics , Paired Box Transcription Factors/metabolism , SOXE Transcription Factors/genetics , SOXE Transcription Factors/metabolism , Schwann Cells/cytology
15.
Glia ; 63(4): 699-717, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25530205

ABSTRACT

Oligodendrocytes are the myelinating cells of the central nervous system. Multiple markers are available to analyze the populations of oligodendroglial cells and their precursors during development and in pathological conditions. However, the behavior of oligodendrocytes remains poorly characterized in vivo, especially at the level of individual cells. Studying this aspect has been impaired so far by the lack of suitable methods for visualizing single oligodendrocytes, their processes, and their interactions during myelination. Here, we have used multicolor labeling technology to single-out simultaneously many individual oligodendrocytes in the postnatal mouse optic nerve. This method is based on Brainbow, a transgenic system for stochastic expression of multiple fluorescent protein genes through Cre-lox recombination, previously used for visualizing axons and neurons. We used tamoxifen-inducible recombination in myelinating cells of Brainbow transgenic mice to obtain multicolor labeling of oligodendrocytes. We show that the palette of colors expressed by labeled oligodendrocytes is tamoxifen dependent, with the highest doses producing the densest and most colorful labeling. At low doses of tamoxifen, the morphology of single or small clusters of fluorescent oligodendrocytes can be studied during postnatal development and in adult. Internodes are labeled to their extremities, revealing nodes of Ranvier. The new mouse model presented here opens new possibilities to explore the organization and development of the oligodendrocyte network with single-cell resolution.


Subject(s)
Luminescent Proteins/metabolism , Nerve Fibers, Myelinated/metabolism , Oligodendroglia/cytology , Optic Nerve/cytology , Staining and Labeling/methods , Animals , Fluorescent Antibody Technique/methods , Luminescent Proteins/genetics , Mice , Mice, Transgenic , Oligodendroglia/metabolism , Recombination, Genetic , Stochastic Processes , Tamoxifen/administration & dosage , Transgenes
16.
Hum Mol Genet ; 22(21): 4417-29, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23813975

ABSTRACT

The ubiquitously expressed large GTPase Dynamin 2 (DNM2) plays a critical role in the regulation of intracellular membrane trafficking through its crucial function in membrane fission, particularly in endocytosis. Autosomal-dominant mutations in DNM2 cause tissue-specific human disorders. Different sets of DNM2 mutations are linked to dominant intermediate Charcot-Marie-Tooth neuropathy type B, a motor and sensory neuropathy affecting primarily peripheral nerves, or autosomal-dominant centronuclear myopathy (CNM) presenting with primary damage in skeletal muscles. To understand the underlying disease mechanisms, it is imperative to determine to which degree the primary affected cell types require DNM2. Thus, we used cell type-specific gene ablation to examine the consequences of DNM2 loss in skeletal muscle cells, the major relevant cell type involved in CNM. We found that DNM2 function in skeletal muscle is required for proper mouse development. Skeletal muscle-specific loss of DNM2 causes a reduction in muscle mass and in the numbers of muscle fibers, altered muscle fiber size distributions, irregular neuromuscular junctions (NMJs) and isolated degenerating intramuscular peripheral nerve fibers. Intriguingly, a lack of muscle-expressed DNM2 triggers an increase of lipid droplets (LDs) and mitochondrial defects. We conclude that loss of DNM2 function in skeletal muscles initiates a chain of harmful parallel and serial events, involving dysregulation of LDs and mitochondrial defects within altered muscle fibers, defective NMJs and peripheral nerve degeneration. These findings provide the essential basis for further studies on DNM2 function and malfunction in skeletal muscles in health and disease, potentially including metabolic diseases such as diabetes.


Subject(s)
Charcot-Marie-Tooth Disease/physiopathology , Dynamin II/deficiency , Dynamin II/physiology , Lipid Metabolism , Mitochondria/metabolism , Muscle, Skeletal/physiology , Myopathies, Structural, Congenital/physiopathology , Neuromuscular Junction/physiology , Peripheral Nerves/physiology , Animals , Charcot-Marie-Tooth Disease/genetics , Dynamin II/genetics , Dynamin II/metabolism , Humans , Mice , Mice, Transgenic , Muscle, Skeletal/growth & development , Muscle, Skeletal/metabolism , Mutation , Myopathies, Structural, Congenital/genetics , Neuromuscular Junction/metabolism , Organ Specificity , Peripheral Nerves/metabolism
17.
Development ; 139(12): 2107-17, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22573620

ABSTRACT

Wnt/ß-catenin signaling controls multiple steps of neural crest development, ranging from neural crest induction, lineage decisions, to differentiation. In mice, conditional ß-catenin inactivation in premigratory neural crest cells abolishes both sensory neuron and melanocyte formation. Intriguingly, the generation of melanocytes is also prevented by activation of ß-catenin in the premigratory neural crest, which promotes sensory neurogenesis at the expense of other neural crest derivatives. This raises the question of how Wnt/ß-catenin signaling regulates the formation of distinct lineages from the neural crest. Using various Cre lines to conditionally activate ß-catenin in neural crest cells at different developmental stages, we show that neural crest cell fate decisions in vivo are subject to temporal control by Wnt/ß-catenin. Unlike in premigratory neural crest, ß-catenin activation in migratory neural crest cells promotes the formation of ectopic melanoblasts, while the production of most other lineages is suppressed. Ectopic melanoblasts emerge at sites of neural crest target structures and in many tissues usually devoid of neural crest-derived cells. ß-catenin activation at later stages in glial progenitors or in melanoblasts does not lead to surplus melanoblasts, indicating a narrow time window of Wnt/ß-catenin responsiveness during neural crest cell migration. Thus, neural crest cells appear to be multipotent in vivo both before and after emigration from the neural tube but adapt their response to extracellular signals in a temporally controlled manner.


Subject(s)
Cell Lineage , Neural Crest/cytology , Wnt Signaling Pathway , Animals , Attachment Sites, Microbiological , Biomarkers/metabolism , Body Patterning , Cell Movement , Doublecortin Domain Proteins , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/metabolism , Immunohistochemistry , Integrases/metabolism , Melanocytes/cytology , Melanocytes/metabolism , Mice , Microphthalmia-Associated Transcription Factor/metabolism , Microtubule-Associated Proteins/metabolism , Neural Crest/metabolism , Neurons/cytology , Neurons/metabolism , Neuropeptides/metabolism , SOXE Transcription Factors/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Time Factors , beta Catenin/metabolism
18.
Development ; 139(2): 397-410, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22186729

ABSTRACT

The cellular origin and molecular mechanisms regulating pigmentation of head and neck are largely unknown. Melanocyte specification is controlled by the transcriptional activity of Mitf, but no general logic has emerged to explain how Mitf and progenitor transcriptional activities consolidate melanocyte and progenitor cell fates. We show that cranial melanocytes arise from at least two different cellular sources: initially from nerve-associated Schwann cell precursors (SCPs) and later from a cellular source that is independent of nerves. Unlike the midbrain-hindbrain cluster from which melanoblasts arise independently of nerves, a large center of melanocytes in and around cranial nerves IX-X is derived from SCPs, as shown by genetic cell-lineage tracing and analysis of ErbB3-null mutant mice. Conditional gain- and loss-of-function experiments show genetically that cell fates in the neural crest involve both the SRY transcription factor Sox2 and Mitf, which consolidate an SCP progenitor or melanocyte fate by cross-regulatory interactions. A gradual downregulation of Sox2 in progenitors during development permits the differentiation of both neural crest- and SCP-derived progenitors into melanocytes, and an initial small pool of nerve-associated melanoblasts expands in number and disperses under the control of endothelin receptor B (Ednrb) and Wnt5a signaling.


Subject(s)
Gene Expression Regulation, Developmental/physiology , Melanocytes/cytology , Microphthalmia-Associated Transcription Factor/metabolism , Neural Crest/embryology , Pigmentation/physiology , SOXB1 Transcription Factors/metabolism , Animals , Chromatin Immunoprecipitation , Embryo, Mammalian/embryology , Imaging, Three-Dimensional , Immunohistochemistry , In Situ Hybridization , Melanocytes/metabolism , Mice , Neural Crest/metabolism , Plasmids/genetics , RNA, Small Interfering/genetics , Receptors, Endothelin/metabolism , Schwann Cells/cytology , Signal Transduction/physiology , Wnt Proteins/metabolism , Wnt-5a Protein
19.
Ann Neurol ; 75(2): 255-65, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24339129

ABSTRACT

OBJECTIVE: The peripheral myelin protein-22 (PMP22) gene is associated with the most common types of inherited neuropathies, including hereditary neuropathy with liability to pressure palsies (HNPP) caused by PMP22 deficiency. However, the function of PMP22 has yet to be defined. Our previous study has shown that PMP22 deficiency causes an impaired propagation of nerve action potentials in the absence of demyelination. In the present study, we tested an alternative mechanism relating to myelin permeability. METHODS: Utilizing Pmp22(+) (/) (-) mice as a model of HNPP, we evaluated myelin junctions and their permeability using morphological, electrophysiological, and biochemical approaches. RESULTS: We show disruption of multiple types of cell junction complexes in peripheral nerve, resulting in increased permeability of myelin and impaired action potential propagation. We further demonstrate that PMP22 interacts with immunoglobulin domain-containing proteins known to regulate tight/adherens junctions and/or transmembrane adhesions, including junctional adhesion molecule-C (JAM-C) and myelin-associated glycoprotein (MAG). Deletion of Jam-c or Mag in mice recapitulates pathology in HNPP. INTERPRETATION: Our study reveals a novel mechanism by which PMP22 deficiency affects nerve conduction not through removal of myelin, but through disruption of myelin junctions.


Subject(s)
Arthrogryposis/genetics , Arthrogryposis/metabolism , Hereditary Sensory and Motor Neuropathy/genetics , Hereditary Sensory and Motor Neuropathy/metabolism , Myelin Proteins/deficiency , Myelin Sheath/metabolism , Tight Junctions/pathology , Action Potentials/drug effects , Action Potentials/genetics , Age Factors , Animals , Axons/drug effects , Axons/metabolism , Axons/pathology , Disease Models, Animal , Gene Expression Regulation/genetics , HEK293 Cells , Humans , Junctional Adhesion Molecules/metabolism , Mice , Mice, Transgenic , Mutation/genetics , Neural Conduction/drug effects , Neural Conduction/genetics , Peripheral Nerves/metabolism , Peripheral Nerves/pathology , Potassium/pharmacology , Tight Junction Proteins/metabolism , Tight Junctions/genetics , Tight Junctions/metabolism
20.
EMBO Rep ; 14(6): 545-52, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23628762

ABSTRACT

Mitochondria and peroxisomes can be fragmented by the process of fission. The fission machineries of both organelles share a set of proteins. GDAP1 is a tail-anchored protein of mitochondria and induces mitochondrial fragmentation. Mutations in GDAP1 lead to Charcot-Marie-Tooth disease (CMT), an inherited peripheral neuropathy, and affect mitochondrial dynamics. Here, we show that GDAP1 is also targeted to peroxisomes mediated by the import receptor Pex19. Knockdown of GDAP1 leads to peroxisomal elongation that can be rescued by re-expressing GDAP1 and by missense mutated forms found in CMT patients. GDAP1-induced peroxisomal fission is dependent on the integrity of its hydrophobic domain 1, and on Drp1 and Mff, as is mitochondrial fission. Thus, GDAP1 regulates mitochondrial and peroxisomal fission by a similar mechanism. However, our results reveal also a more critical role of the amino-terminal GDAP1 domains, carrying most CMT-causing mutations, in the regulation of mitochondrial compared to peroxisomal fission.


Subject(s)
Mitochondrial Dynamics , Mutation, Missense , Nerve Tissue Proteins/genetics , Peroxisomes/physiology , Animals , COS Cells , Cell Shape , Charcot-Marie-Tooth Disease/genetics , Chlorocebus aethiops , Dynamins , GTP Phosphohydrolases/metabolism , HEK293 Cells , Hippocampus/pathology , Humans , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Mitochondrial Membranes/metabolism , Mitochondrial Proteins/metabolism , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Neurons/physiology , Primary Cell Culture , Protein Structure, Tertiary , Protein Transport
SELECTION OF CITATIONS
SEARCH DETAIL