Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Elife ; 122023 Dec 14.
Article in English | MEDLINE | ID: mdl-38095361

ABSTRACT

In addition to their roles in protecting nerves and increasing conduction velocity, peripheral glia plays key functions in blood vessel development by secreting molecules governing arteries alignment and maturation with nerves. Here, we show in mice that a specific, nerve-attached cell population, derived from boundary caps (BCs), constitutes a major source of mural cells for the developing skin vasculature. Using Cre-based reporter cell tracing and single-cell transcriptomics, we show that BC derivatives migrate into the skin along the nerves, detach from them, and differentiate into pericytes and vascular smooth muscle cells. Genetic ablation of this population affects the organization of the skin vascular network. Our results reveal the heterogeneity and extended potential of the BC population in mice, which gives rise to mural cells, in addition to previously described neurons, Schwann cells, and melanocytes. Finally, our results suggest that mural specification of BC derivatives takes place before their migration along nerves to the mouse skin.


Subject(s)
Neural Crest , Neural Tube , Mice , Animals , Neural Crest/physiology , Neuroglia , Schwann Cells , Skin , Cell Differentiation/physiology
2.
Cancer Discov ; 9(1): 130-147, 2019 01.
Article in English | MEDLINE | ID: mdl-30348676

ABSTRACT

Patients carrying an inactive NF1 allele develop tumors of Schwann cell origin called neurofibromas (NF). Genetically engineered mouse models have significantly enriched our understanding of plexiform forms of NFs (pNF). However, this has not been the case for cutaneous neurofibromas (cNF), observed in all NF1 patients, as no previous model recapitulates their development. Here, we show that conditional Nf1 inactivation in Prss56-positive boundary cap cells leads to bona fide pNFs and cNFs. This work identifies subepidermal glia as a likely candidate for the cellular origin of cNFs and provides insights on disease mechanisms, revealing a long, multistep pathologic process in which inflammation-related signals play a pivotal role. This new mouse model is an important asset for future clinical and therapeutic investigations of NF1-associated neurofibromas. SIGNIFICANCE: Patients affected by NF1 develop numerous cNFs. We present a mouse model that faithfully recapitulates cNFs, identify a candidate cell type at their origin, analyze the steps involved in their formation, and show that their development is dramatically accelerated by skin injury. These findings have important clinical/therapeutic implications.This article is highlighted in the In This Issue feature, p. 1.


Subject(s)
Neurofibroma/metabolism , Neurofibromatosis 1/metabolism , Neurofibromin 1/genetics , Schwann Cells/metabolism , Skin Neoplasms/metabolism , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Knockout , Mutation , Neurofibroma/etiology , Neurofibroma/genetics , Neurofibroma/physiopathology , Neurofibromatosis 1/complications , Neurofibromatosis 1/genetics , Neurofibromatosis 1/physiopathology , Schwann Cells/physiology , Skin Neoplasms/etiology , Skin Neoplasms/genetics , Skin Neoplasms/physiopathology
3.
Brain Struct Funct ; 221(9): 4411-4427, 2016 12.
Article in English | MEDLINE | ID: mdl-26701169

ABSTRACT

Adult neurogenesis in the mammalian brain is restricted to specific regions, such as the dentate gyrus (DG) in the hippocampus and the subventricular zone (SVZ) in the walls of the lateral ventricles. Here, we used a mouse line carrying a knock-in of Cre recombinase in the Prss56 gene, in combination with two Cre-inducible fluorescent reporters (Rosa26 mTmG and Rosa26 tdTom ), to perform genetic tracing of Prss56-expressing cells in the adult brain. We found reporter-positive cells in three neurogenic niches: the DG, the SVZ and the hypothalamus ventricular zone. In the prospective DG, Prss56 is expressed during embryogenesis in a subpopulation of radial glia. The pattern of migration and differentiation of reporter-positive cells during development recapitulates the successive steps of DG neurogenesis, including the formation of a subpopulation of adult neural stem cells (NSC). In the SVZ, Prss56 is expressed postnatally in a subpopulation of adult NSC mainly localized in the medial-ventral region of the lateral wall. This subpopulation preferentially gives rise to deep granule and Calbindin-positive periglomerular interneurons in the olfactory bulb. Finally, Prss56 is also expressed in a subpopulation of α2-tanycytes, which are potential adult NSCs of the hypothalamus ventricular zone. Our observations suggest that some α2-tanycytes translocate their soma into the parenchyma and may give rise to a novel cell type in this territory. Overall, this study establishes the Prss56 Cre line as an efficient and promising new tool to study multiple aspects of adult neurogenesis in the mouse.


Subject(s)
Adult Stem Cells/physiology , Brain/physiology , Neural Stem Cells/physiology , Neurogenesis , Serine Proteases/metabolism , Adult Stem Cells/metabolism , Animals , Brain/embryology , Brain/metabolism , Cell Differentiation , Cell Movement , Dentate Gyrus/embryology , Dentate Gyrus/metabolism , Dentate Gyrus/physiology , Ependymoglial Cells/metabolism , Ependymoglial Cells/physiology , Interneurons/metabolism , Interneurons/physiology , Lateral Ventricles/embryology , Lateral Ventricles/metabolism , Lateral Ventricles/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neural Stem Cells/metabolism , Serine Proteases/physiology
4.
Stem Cell Reports ; 5(2): 278-90, 2015 Aug 11.
Article in English | MEDLINE | ID: mdl-26212662

ABSTRACT

While neurogenic stem cells have been identified in rodent and human skin, their manipulation and further characterization are hampered by a lack of specific markers. Here, we perform genetic tracing of the progeny of boundary cap (BC) cells, a neural-crest-derived cell population localized at peripheral nerve entry/exit points. We show that BC derivatives migrate along peripheral nerves to reach the skin, where they give rise to terminal glia associated with dermal nerve endings. Dermal BC derivatives also include cells that self-renew in sphere culture and have broad in vitro differentiation potential. Upon transplantation into adult mouse dorsal root ganglia, skin BC derivatives efficiently differentiate into various types of mature sensory neurons. Together, this work establishes the embryonic origin, pathway of migration, and in vivo neurogenic potential of a major component of skin stem-like cells. It provides genetic tools to study and manipulate this population of high interest for medical applications.


Subject(s)
Neural Stem Cells/cytology , Neurogenesis , Neuroglia/cytology , Skin/cytology , Animals , Cell Lineage , Cell Movement , Cells, Cultured , Mice , Mice, Inbred C57BL , Neural Stem Cells/physiology , Sensory Receptor Cells/cytology
5.
Subcell Biochem ; 58: 61-94, 2012.
Article in English | MEDLINE | ID: mdl-22403074

ABSTRACT

The physiological effects of many extracellular neurotransmitters, hormones, growth factors, and other stimuli are mediated by receptor-promoted activation of phospholipase C (PLC) and consequential activation of inositol lipid signaling pathways. These signaling responses include the classically described conversion of phosphatidylinositol(4,5)P(2) to the Ca(2+)-mobilizing second messenger inositol(1,4,5)P(3) and the protein kinase C-activating second messenger diacylglycerol as well as alterations in membrane association or activity of many proteins that harbor phosphoinositide binding domains. The 13 mammalian PLCs elaborate a minimal catalytic core typified by PLC-d to confer multiple modes of regulation of lipase activity. PLC-b isozymes are activated by Gaq- and Gbg-subunits of heterotrimeric G proteins, and activation of PLC-g isozymes occurs through phosphorylation promoted by receptor and non-receptor tyrosine kinases. PLC-e and certain members of the PLC-b and PLC-g subclasses of isozymes are activated by direct binding of small G proteins of the Ras, Rho, and Rac subfamilies of GTPases. Recent high resolution three dimensional structures together with biochemical studies have illustrated that the X/Y linker region of the catalytic core mediates autoinhibition of most if not all PLC isozymes. Activation occurs as a consequence of removal of this autoinhibition.


Subject(s)
Eukaryotic Cells/enzymology , Gene Expression Regulation , Heterotrimeric GTP-Binding Proteins/metabolism , Second Messenger Systems , Type C Phospholipases/metabolism , Animals , Diglycerides/metabolism , Enzyme Activation , Eukaryotic Cells/cytology , Heterotrimeric GTP-Binding Proteins/genetics , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Isoenzymes/chemistry , Isoenzymes/classification , Isoenzymes/genetics , Isoenzymes/metabolism , Models, Molecular , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphorylation , Protein Structure, Secondary , Protein Structure, Tertiary , Type C Phospholipases/chemistry , Type C Phospholipases/classification , Type C Phospholipases/genetics
6.
J Biol Chem ; 285(46): 35836-47, 2010 Nov 12.
Article in English | MEDLINE | ID: mdl-20807769

ABSTRACT

The lipase activity of most phospholipases C (PLCs) is basally repressed by a highly degenerate and mostly disordered X/Y linker inserted within the catalytic domain. Release of this auto-inhibition is driven by electrostatic repulsion between the plasma membrane and the electronegative X/Y linker. In contrast, PLC-γ isozymes (PLC-γ1 and -γ2) are structurally distinct from other PLCs because multiple domains are present in their X/Y linker. Moreover, although many tyrosine kinases directly phosphorylate PLC-γ isozymes to enhance their lipase activity, the underlying molecular mechanism of this activation remains unclear. Here we define the mechanism for the unique regulation of PLC-γ isozymes by their X/Y linker. Specifically, we identify the C-terminal SH2 domain within the X/Y linker as the critical determinant for auto-inhibition. Tyrosine phosphorylation of the X/Y linker mediates high affinity intramolecular interaction with the C-terminal SH2 domain that is coupled to a large conformational rearrangement and release of auto-inhibition. Consequently, PLC-γ isozymes link phosphorylation to phospholipase activation by elaborating upon primordial regulatory mechanisms found in other PLCs.


Subject(s)
Amino Acid Motifs/genetics , Mutation , Phospholipase C gamma/genetics , Phospholipase C gamma/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , Blotting, Western , Cells, Cultured , Circular Dichroism , Enzyme Activation , Enzyme Assays , HEK293 Cells , Humans , Molecular Sequence Data , Phospholipase C gamma/chemistry , Phosphorylation , Protein Binding , Protein Conformation , Rats , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Tyrosine/genetics , Tyrosine/metabolism , src Homology Domains/genetics
7.
Cell ; 138(5): 990-1004, 2009 Sep 04.
Article in English | MEDLINE | ID: mdl-19737524

ABSTRACT

During brain development, proper neuronal migration and morphogenesis is critical for the establishment of functional neural circuits. Here we report that srGAP2 negatively regulates neuronal migration and induces neurite outgrowth and branching through the ability of its F-BAR domain to induce filopodia-like membrane protrusions resembling those induced by I-BAR domains in vivo and in vitro. Previous work has suggested that in nonneuronal cells filopodia dynamics decrease the rate of cell migration and the persistence of leading edge protrusions. srGAP2 knockdown reduces leading process branching and increases the rate of neuronal migration in vivo. Overexpression of srGAP2 or its F-BAR domain has the opposite effects, increasing leading process branching and decreasing migration. These results suggest that F-BAR domains are functionally diverse and highlight the functional importance of proteins directly regulating membrane deformation for proper neuronal migration and morphogenesis.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Neurogenesis , Neurons/cytology , Animals , Cell Movement , Cerebral Cortex/cytology , Cerebral Cortex/embryology , GTPase-Activating Proteins , Mice , Pseudopodia/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...