Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 53
Filter
1.
Development ; 146(18)2019 09 16.
Article in English | MEDLINE | ID: mdl-31488565

ABSTRACT

Multiple members of the tumour necrosis factor superfamily (TNFSF) regulate the growth and branching of neural processes late in development, when neurons are establishing and refining connections. Here, we present the first evidence that a TNFSF member acts much earlier in development, when axons are growing to their targets. CD40L transiently enhanced axon growth from embryonic mouse DRG neurons cultured at this early stage. Early spinal nerves of embryos lacking the CD40L receptor (Cd40-/- mice) were significantly shorter in vivo than those of Cd40+/+ littermates. CD40L was synthesized in early DRG targets and was co-expressed with CD40 in early DRG neurons. Whereas CD40L enhanced early axon growth independently of neurotrophins, disruption of a CD40L/CD40 autocrine loop impaired early neurotrophin-promoted axon growth. In marked contrast to the widespread regulation of axon and dendrite growth by CD40L reverse signalling later in development, CD40-Fc, which activates reverse signalling, had no effect on early sensory axon growth. These results suggest that CD40 forward signalling is a novel physiological regulator of early axon growth that acts by target-derived and autocrine mechanisms.


Subject(s)
Axons/metabolism , CD40 Antigens/metabolism , Sensory Receptor Cells/metabolism , Signal Transduction , Animals , Autocrine Communication , CD40 Ligand/genetics , CD40 Ligand/metabolism , Cell Survival , Embryo, Mammalian/metabolism , Ganglia, Spinal/metabolism , Gene Deletion , Gene Expression Regulation, Developmental , Mice, Inbred C57BL , Nerve Growth Factors/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sensory Receptor Cells/cytology , Spinal Nerves/metabolism
2.
Cell Mol Life Sci ; 78(3): 1065-1083, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32506167

ABSTRACT

CD40-activated CD40L reverse signaling is a major physiological regulator of axon and dendrite growth from developing hippocampal pyramidal neurons. Here we have studied how CD40L-mediated reverse signaling promotes the growth of these processes. Cultures of hippocampal pyramidal neurons were established from Cd40-/- mouse embryos to eliminate endogenous CD40/CD40L signaling, and CD40L reverse signaling was stimulated by a CD40-Fc chimera. CD40L reverse signaling increased phosphorylation and hence activation of proteins in the PKC, ERK, and JNK signaling pathways. Pharmacological activators and inhibitors of these pathways revealed that whereas activation of JNK inhibited growth, activation of PKC and ERK1/ERK2 enhanced growth. Experiments using combinations of pharmacological reagents revealed that these signaling pathways regulate growth by functioning as an interconnected and interdependent network rather than acting in a simple linear sequence. Immunoprecipitation studies suggested that stimulation of CD40L reverse signaling generated a receptor complex comprising CD40L, PKCß, and the Syk tyrosine kinase. Our studies have begun to elucidate the molecular network and interactions that promote axon and dendrite growth from developing hippocampal neurons following activation of CD40L reverse signaling.


Subject(s)
Axons/metabolism , CD40 Ligand/metabolism , Dendrites/physiology , Signal Transduction , Animals , Butadienes/pharmacology , CD40 Antigens/deficiency , CD40 Antigens/genetics , Cells, Cultured , Dendrites/drug effects , Hippocampus/cytology , Hippocampus/metabolism , JNK Mitogen-Activated Protein Kinases/chemistry , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/chemistry , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/chemistry , Mitogen-Activated Protein Kinase 3/metabolism , Nitriles/pharmacology , Phosphorylation , Protein Interaction Domains and Motifs , Protein Kinase C/metabolism , Signal Transduction/drug effects , Syk Kinase/metabolism
3.
Development ; 145(22)2018 11 19.
Article in English | MEDLINE | ID: mdl-30337376

ABSTRACT

TWE-PRIL is a naturally occurring fusion protein of components of two TNF superfamily members: the extracellular domain of APRIL; and the intracellular and transmembrane domains of TWEAK with no known function. Here, we show that April-/- mice (which lack APRIL and TWE-PRIL) exhibited overgrowth of sympathetic fibres in vivo, and sympathetic neurons cultured from these mice had significantly longer axons than neurons cultured from wild-type littermates. Enhanced axon growth from sympathetic neurons cultured from April-/- mice was prevented by expressing full-length TWE-PRIL in these neurons but not by treating them with soluble APRIL. Soluble APRIL, however, enhanced axon growth from the sympathetic neurons of wild-type mice. siRNA knockdown of TWE-PRIL but not siRNA knockdown of APRIL alone also enhanced axon growth from wild-type sympathetic neurons. Our work reveals the first and physiologically relevant role for TWE-PRIL and suggests that it mediates reverse signalling.


Subject(s)
Axons/metabolism , Signal Transduction , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Animals , B-Cell Maturation Antigen/genetics , B-Cell Maturation Antigen/metabolism , Cells, Cultured , Cytokine TWEAK/genetics , Cytokine TWEAK/metabolism , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Knockdown Techniques , Mice , Models, Biological , Nerve Growth Factor/pharmacology , Phenotype , RNA, Small Interfering/metabolism , Solubility , Superior Cervical Ganglion/metabolism , Sympathetic Nervous System/growth & development , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics
4.
Development ; 140(10): 2108-17, 2013 May.
Article in English | MEDLINE | ID: mdl-23633509

ABSTRACT

The somatosensory and sympathetic innervation of the vertebrate head is derived principally from the neurons of trigeminal and superior cervical ganglia (SCG), respectively. During development, the survival of both populations of neurons and the terminal growth and branching of their axons in the tissues they innervate is regulated by the supply of nerve growth factor (NGF) produced by these tissues. NGF is derived by proteolytic cleavage of a large precursor protein, proNGF, which is recognised to possess distinctive biological functions. Here, we show that proNGF promotes profuse neurite growth and branching from cultured postnatal mouse SCG neurons. In marked contrast, proNGF does not promote the growth of trigeminal neurites. Studies using compartment cultures demonstrated that proNGF acts locally on SCG neurites to promote growth. The neurite growth-promoting effect of proNGF is not observed in SCG neurons cultured from p75(NTR)-deficient mice, and proNGF does not phosphorylate the NGF receptor tyrosine kinase TrkA. These findings suggest that proNGF selectively promotes the growth of neurites from a subset of NGF-responsive neurons by a p75(NTR)-dependent mechanism during postnatal development when the axons of these neurons are ramifying within their targets in vivo.


Subject(s)
Nerve Growth Factor/metabolism , Neurites/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Cells, Cultured , Gene Expression Regulation, Developmental , Mice , Nerve Growth Factors/metabolism , Neurons/cytology , Neurons/metabolism , RNA, Small Interfering/metabolism , Receptor, trkA/metabolism , Signal Transduction , Time Factors , Trigeminal Nerve/metabolism
5.
Development ; 140(23): 4751-62, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24173804

ABSTRACT

Dendrite size and morphology are key determinants of the functional properties of neurons. Here, we show that growth differentiation factor 5 (GDF5), a member of the bone morphogenetic protein (BMP) subclass of the transforming growth factor ß superfamily with a well-characterised role in limb morphogenesis, is a key regulator of the growth and elaboration of pyramidal cell dendrites in the developing hippocampus. Pyramidal cells co-express GDF5 and its preferred receptors, BMP receptor 1B and BMP receptor 2, during development. In culture, GDF5 substantially increased dendrite, but not axon, elongation from these neurons by a mechanism that depends on activation of SMADs 1/5/8 and upregulation of the transcription factor HES5. In vivo, the apical and basal dendritic arbours of pyramidal cells throughout the hippocampus were markedly stunted in both homozygous and heterozygous Gdf5 null mutants, indicating that dendrite size and complexity are exquisitely sensitive to the level of endogenous GDF5 synthesis.


Subject(s)
Dendrites/metabolism , Growth Differentiation Factor 5/metabolism , Hippocampus/metabolism , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Morphogenetic Protein Receptors, Type I/metabolism , Bone Morphogenetic Protein Receptors, Type II/metabolism , Cells, Cultured , Enzyme Activation , Gene Expression Regulation, Developmental , Growth Differentiation Factor 5/biosynthesis , Growth Differentiation Factor 5/genetics , Hippocampus/embryology , Hippocampus/growth & development , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pyramidal Cells/metabolism , RNA Interference , RNA, Small Interfering , Repressor Proteins/metabolism , Signal Transduction/genetics , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Smad8 Protein/metabolism , Up-Regulation
6.
Mol Cell Neurosci ; 59: 24-36, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24444792

ABSTRACT

APRIL (A Proliferation-Inducing Ligand, TNFSF13) is a member of the tumor necrosis factor superfamily that regulates lymphocyte survival and activation and has been implicated in tumorigenesis and autoimmune diseases. Here we report the expression and first known activity of APRIL in the nervous system. APRIL and one of its receptors, BCMA (B-Cell Maturation Antigen, TNFRSF17), are expressed by hippocampal pyramidal cells of fetal and postnatal mice. In culture, these neurons secreted APRIL, and function-blocking antibodies to either APRIL or BCMA reduced axonal elongation. Recombinant APRIL enhanced axonal elongation, but did not influence dendrite elongation. The effect of APRIL on axon elongation was inhibited by anti-BCMA and the expression of a signaling-defective BCMA mutant in these neurons, suggesting that the axon growth-promoting effect of APRIL is mediated by BCMA. APRIL promoted phosphorylation and activation of ERK1, ERK2 and Akt and serine phosphorylation and inactivation of GSK-3ß in cultured hippocampal pyramidal cells. Inhibition of MEK1/MEK2 (activators of ERK1/ERK2), PI3-kinase (activator of Akt) or Akt inhibited the axon growth-promoting action of APRIL, as did pharmacological activation of GSK-3ß and the expression of a constitutively active form of GSK-3ß. These findings suggest that APRIL promotes axon elongation by a mechanism that depends both on ERK signaling and PI3-kinase/Akt/GSK-3ß signaling.


Subject(s)
Axons/metabolism , Hippocampus/metabolism , Neurogenesis , Tumor Necrosis Factor Ligand Superfamily Member 13/metabolism , Animals , B-Cell Maturation Antigen/metabolism , Cells, Cultured , Dendrites/metabolism , Glycogen Synthase Kinase 3/metabolism , Glycogen Synthase Kinase 3 beta , Hippocampus/cytology , Hippocampus/growth & development , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 2/antagonists & inhibitors , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Pyramidal Cells/cytology , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Signal Transduction , Tumor Necrosis Factor Ligand Superfamily Member 13/genetics
7.
J Neurosci ; 33(12): 5399-410, 2013 Mar 20.
Article in English | MEDLINE | ID: mdl-23516305

ABSTRACT

Axonal branches of the trigeminal ganglion (TG) display characteristic growth and arborization patterns during development. Subsets of TG neurons express different receptors for growth factors, but these are unlikely to explain the unique patterns of axonal arborizations. Intrinsic modulators may restrict or enhance cellular responses to specific ligands and thereby contribute to the development of axon growth patterns. Protein tyrosine phosphatase receptor type O (PTPRO), which is required for Eph receptor-dependent retinotectal development in chick and for development of subsets of trunk sensory neurons in mouse, may be such an intrinsic modulator of TG neuron development. PTPRO is expressed mainly in TrkB-expressing (TrkB(+)) and Ret(+) mechanoreceptors within the TG during embryogenesis. In PTPRO mutant mice, subsets of TG neurons grow longer and more elaborate axonal branches. Cultured PTPRO(-/-) TG neurons display enhanced axonal outgrowth and branching in response to BDNF and GDNF compared with control neurons, indicating that PTPRO negatively controls the activity of BDNF/TrkB and GDNF/Ret signaling. Mouse PTPRO fails to regulate Eph signaling in retinocollicular development and in hindlimb motor axon guidance, suggesting that chick and mouse PTPRO have different substrate specificities. PTPRO has evolved to fine tune growth factor signaling in a cell-type-specific manner and to thereby increase the diversity of signaling output of a limited number of receptor tyrosine kinases to control the branch morphology of developing sensory neurons. The regulation of Eph receptor-mediated developmental processes by protein tyrosine phosphatases has diverged between chick and mouse.


Subject(s)
Axons/physiology , Membrane Glycoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-ret/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 3/metabolism , Trigeminal Ganglion/cytology , Trigeminal Ganglion/metabolism , Animals , Animals, Newborn , Cells, Cultured , Female , Green Fluorescent Proteins/genetics , HEK293 Cells , HeLa Cells , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Motor Neurons/cytology , Motor Neurons/metabolism , Pregnancy , Receptor, EphA1/metabolism , Receptor, trkA/metabolism , Receptor, trkC/metabolism , Signal Transduction/physiology , Trigeminal Ganglion/embryology , Trigeminal Nerve/cytology , Trigeminal Nerve/embryology , Trigeminal Nerve/metabolism
8.
Development ; 136(20): 3405-12, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19762427

ABSTRACT

For a given cell type, particular extracellular signals generate characteristic patterns of activity in intracellular signalling networks that lead to distinctive cell-type specific responses. Here, we report the first known occurrence of a developmental switch in the intracellular signalling network required for an identical cellular response to the same extracellular signal in the same cell type. We show that although NF-kappaB signalling is required for BDNF-promoted neurite growth from both foetal and postnatal mouse sensory neurons, there is a developmental switch between these stages in the NF-kappaB activation mechanism and the phosphorylation status of the p65 NF-kappaB subunit required for neurite growth. Shortly before birth, BDNF activates NF-kappaB by an atypical mechanism that involves tyrosine phosphorylation of IkappaBalpha by Src family kinases, and dephosphorylates p65 at serine 536. Immediately after birth, BDNF-independent constitutive activation of NF-kappaB signalling by serine phosphorylation of IkappaBalpha and constitutive dephosphorylation of p65 at serine 536 are required for BDNF-promoted neurite growth. This abrupt developmental switch in NF-kappaB signalling in a highly differentiated cell type illustrates an unsuspected plasticity in signalling networks in the generation of identical cellular responses to the same extracellular signal.


Subject(s)
Gene Expression Regulation, Developmental , NF-kappa B/metabolism , Neurites/metabolism , Signal Transduction , Animals , Brain-Derived Neurotrophic Factor/metabolism , Cells, Cultured , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Mice , Phosphorylation , Phosphoserine/metabolism , Phosphotyrosine/metabolism , src-Family Kinases/metabolism
9.
Nat Neurosci ; 11(2): 135-42, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18176559

ABSTRACT

Nerve growth factor (NGF) has an important role in regulating sympathetic neuron survival and target field innervation during development. Here we show that glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR), a member of the TNF superfamily, and its ligand (GITRL) are co-expressed in mouse sympathetic neurons when their axons are innervating their targets under the influence of target-derived NGF. In culture, GITRL enhanced NGF-promoted neurite growth from neonatal sympathetic neurons, and preventing GITR-GITRL interaction in these neurons or knocking down GITR inhibited NGF-promoted neurite growth without affecting neuronal survival. Tnfrsf18(-/-) (Gitr) neonates have reduced sympathetic innervation density in vivo compared with Gitr(+/+) littermates. GITR activation is required for the phosphorylation of extracellular signal-regulated kinases 1 and 2 by NGF that is necessary for neurite growth. Our results reveal a previously unknown signaling loop in developing sympathetic neurons that is crucial for NGF-dependent axon growth and target innervation.


Subject(s)
Axons/drug effects , Glucocorticoid-Induced TNFR-Related Protein/physiology , Nerve Growth Factor/pharmacology , Neurons/cytology , Signal Transduction/physiology , Tumor Necrosis Factors/metabolism , Age Factors , Analysis of Variance , Animals , Animals, Newborn , Cells, Cultured , Embryo, Mammalian , Fluoresceins/metabolism , Glucocorticoid-Induced TNFR-Related Protein/deficiency , Luminescent Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/drug effects , Phosphorylation/drug effects , Superior Cervical Ganglion/cytology , Tumor Necrosis Factors/deficiency
10.
Nat Neurosci ; 11(3): 285-91, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18223649

ABSTRACT

The extracellular calcium-sensing receptor (CaSR) monitors the systemic, extracellular, free ionized-calcium level ([Ca(2+)](o)) in organs involved in systemic [Ca(2+)](o) homeostasis. However, CaSR is also expressed in the nervous system, where its role is unknown. We found large amounts of CaSR in perinatal mouse sympathetic neurons when their axons were innervating and branching extensively in their targets. Manipulating CaSR function in these neurons by varying [Ca(2+)](o), using CaSR agonists and antagonists, or expressing a dominant-negative CaSR markedly affected neurite growth in vitro. Sympathetic neurons lacking CaSR had smaller neurite arbors in vitro, and sympathetic innervation density was reduced in CaSR-deficient mice in vivo. Hippocampal pyramidal neurons, which also express CaSR, had smaller dendrites when transfected with dominant-negative CaSR in postnatal organotypic cultures. Our findings reveal a crucial role for CaSR in regulating the growth of neural processes in the peripheral and central nervous systems.


Subject(s)
Axons/metabolism , Calcium Signaling/genetics , Dendrites/metabolism , Ganglia, Sympathetic/embryology , Ganglia, Sympathetic/growth & development , Receptors, Calcium-Sensing/metabolism , Animals , Axons/ultrastructure , Calcium Signaling/drug effects , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Shape/genetics , Cells, Cultured , Dendrites/ultrastructure , Fluorescent Dyes , Ganglia, Sympathetic/cytology , Growth Cones/metabolism , Growth Cones/ultrastructure , Hippocampus/cytology , Hippocampus/embryology , Hippocampus/metabolism , Luminescent Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurites/metabolism , Neurites/ultrastructure , Organ Culture Techniques , Pyramidal Cells/cytology , Pyramidal Cells/metabolism , Receptors, Calcium-Sensing/drug effects , Receptors, Calcium-Sensing/genetics
11.
J Neurosci ; 30(17): 6094-105, 2010 Apr 28.
Article in English | MEDLINE | ID: mdl-20427667

ABSTRACT

FLICE-inhibitory protein (FLIP) is an endogenous inhibitor of the signaling pathway triggered by the activation of death receptors. Here, we reveal a novel biological function for the long form of FLIP (FLIP-L) in neuronal differentiation, which can be dissociated from its antiapoptotic role. We show that FLIP-L is expressed in different regions of the mouse embryonic nervous system. Immunohistochemistry of mouse brain sections at different stages reveals that, in neurons, FLIP is expressed early during the embryonic neuronal development (embryonic day 16) and decreases at later stages (postnatal days 5-15), when its expression is essentially detected in glial cells. FLIP-L overexpression significantly enhances neurotrophin-induced neurite outgrowth in motoneurons, superior cervical ganglion neurons, and PC12 cells. Conversely, the downregulation of FLIP-L protein levels by specific RNA interference significantly reduces neurite outgrowth, even in the presence of the appropriate neurotrophin stimulus. Moreover, NGF-dependent activation of two main intracellular pathways involved in the regulation of neurite outgrowth, extracellular signal-regulated kinases (ERKs) and nuclear factor kappaB (NF-kappaB), is impaired when endogenous FLIP-L is downregulated, although TrkA remains activated. Finally, we demonstrate that FLIP-L interacts with TrkA, and not with p75(NTR), in an NGF-dependent manner, and endogenous FLIP-L interacts with TrkB in whole-brain lysates from embryonic day 15 mice embryos. Altogether, we uncover a new role for FLIP-L as an unexpected critical player in neurotrophin-induced mitogen-activated protein kinase/ERK- and NF-kappaB-mediated control of neurite growth in developing neurons.


Subject(s)
Brain/physiology , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Nerve Growth Factors/metabolism , Neurites/physiology , Neurogenesis/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Brain/embryology , Brain/growth & development , Cell Death/physiology , Cell Differentiation/physiology , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Motor Neurons/physiology , NF-kappa B/metabolism , Nerve Tissue Proteins , Neuroglia/metabolism , PC12 Cells , Rats , Receptor, trkA/metabolism , Receptor, trkB/metabolism , Receptors, Growth Factor , Receptors, Nerve Growth Factor/metabolism , Superior Cervical Ganglion/embryology , Superior Cervical Ganglion/growth & development , Superior Cervical Ganglion/physiology
12.
Cells ; 10(4)2021 04 07.
Article in English | MEDLINE | ID: mdl-33917019

ABSTRACT

CD40-activated CD40L-mediated reverse signalling is a major physiological regulator of neurite growth from excitatory and inhibitory neurons in the developing central nervous system (CNS). Whereas in excitatory pyramidal neurons, CD40L reverse signalling promotes the growth and elaboration of dendrites and axons, in inhibitory GABAergic striatal medium spiny neurons (MSNs), it restricts neurite growth and branching. In pyramidal neurons, we previously reported that CD40L reverse signalling activates an interconnected and interdependent signalling network involving protein kinase C (PKC), extracellular regulated kinases 1 and 2 (ERK1/2), and c-Jun N-terminal kinase (JNK) signalling pathways that regulates dendrite and axon growth. Here, we have studied whether these signalling pathways also influence neurite growth from striatal inhibitory MSNs. To unequivocally activate CD40L reverse signalling, we treated MSN cultures from CD40-deficient mice with CD40-Fc. Here, we report that activation of CD40L reverse signalling in these cultures also increased the phosphorylation of PKC, ERK1/2, and JNK. Using pharmacological activators and inhibitors of these signalling pathways singularly and in combination, we have shown that, as in pyramidal neurons, these signalling pathways work in an interconnected and interdependent network to regulate the neurite growth, but their functions, relationships, and interdependencies are different from those observed in pyramidal neurons. Furthermore, immunoprecipitation studies showed that stimulation of CD40L reverse signalling recruits the catalytic fragment of Syk tyrosine kinase, but in contrast to pyramidal neurons, PKC does not participate in this recruitment. Our findings show that distinctive networks of three signalling pathways mediate the opposite effects of CD40L reverse signalling on neurite growth in excitatory and inhibitory neurons.


Subject(s)
CD40 Antigens/metabolism , CD40 Ligand/metabolism , Neurites/metabolism , Signal Transduction , Animals , CD40 Antigens/deficiency , Cell Proliferation , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , GABAergic Neurons/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mice, Inbred C57BL , Models, Biological , Phosphorylation , Protein Interaction Maps , Protein Kinase C/metabolism , Syk Kinase/metabolism
13.
Dev Neurobiol ; 81(2): 139-148, 2021 03.
Article in English | MEDLINE | ID: mdl-33369884

ABSTRACT

Recent work has shown that neuregulin-4 (NRG4) is a physiological regulator of the growth of sympathetic axons and CNS dendrites in the developing nervous system. Here, we have investigated whether NRG4 plays a role in sensory axon growth and the establishment of cutaneous sensory innervation. Imaging early nerve fibers in the well-characterized cutaneous trigeminal territory, the brachial plexus, and thorax revealed very marked and highly significant decreases in nerve fiber length and branching density in Nrg4-/- embryos compared with Nrg4+/+ littermates. NRG4 promoted neurotrophin-independent sensory axon growth from correspondingly early trigeminal ganglion and DRG neurons in culture but not from enteroceptive nodose ganglion neurons. High levels of Nrg4 mRNA were detected in cutaneous tissues but not in sensory ganglia. Our findings suggest that NRG4 is an important target-derived factor that participates in the establishment of early cutaneous sensory innervation.


Subject(s)
Nerve Growth Factors , Neuregulins/physiology , Axons/physiology , Neuregulins/chemistry , Neuregulins/metabolism , Neurons/physiology , Neurons, Afferent/physiology
14.
eNeuro ; 8(1)2021.
Article in English | MEDLINE | ID: mdl-33495243

ABSTRACT

The regulation of neuronal soma size is essential for appropriate brain circuit function and its dysregulation is associated with several neurodevelopmental disorders. A defect in the dendritic growth and elaboration of motor neocortical pyramidal neurons in neonates lacking neuregulin-4 (NRG4) has previously been reported. In this study, we investigated whether the loss of NRG4 causes further morphologic defects that are specific to these neurons. We analyzed the soma size of pyramidal neurons of layer (L)2/3 and L5 of the motor cortex and a subpopulation of multipolar interneurons in this neocortical region in Nrg4+/+ and Nrg4-/- mice. There were significant decreases in pyramidal neuron soma size in Nrg4-/- mice compared with Nrg4+/+ littermates at all stages studied [postnatal day (P)10, P30, and P60]. The reduction was especially marked at P10 and in L5 pyramidal neurons. Soma size was not significantly different for multipolar interneurons at any age. This in vivo phenotype was replicated in pyramidal neurons cultured from Nrg4-/- mice and was rescued by NRG treatment. Analysis of a public single-cell RNA sequencing repository revealed discrete Nrg4 and Erbb4 expression in subpopulations of L5 pyramidal neurons, suggesting that the observed defects were due in part to loss of autocrine Nrg4/ErbB4 signaling. The pyramidal phenotype in the motor cortex of Nrg4-/- mice was associated with a lack of Rotarod test improvement in P60 mice, suggesting that absence of NRG4 causes alterations in motor performance.


Subject(s)
Motor Cortex , Neuregulins/genetics , Neurons/cytology , Pyramidal Cells/cytology , Animals , Mice , Mice, Knockout , Motor Cortex/metabolism
15.
J Neurosci ; 29(6): 1599-607, 2009 Feb 11.
Article in English | MEDLINE | ID: mdl-19211867

ABSTRACT

In a PCR screen to identify novel cytokine candidates involved in neuronal development, we identified transcripts for the tumor necrosis factor superfamily member 14 (TNFSF14), generally known as LIGHT (lymphotoxin-related inducible ligand that competes for glycoprotein D binding to herpesvirus entry mediator on T cells), together with its receptors, lymphotoxin-beta receptor (LTbetaR) and TNF family receptor herpesvirus entry mediator (HVEM), in the experimentally tractable sensory neurons of the mouse nodose ganglion. Immunocytochemistry revealed coexpression of LIGHT and its receptors in all nodose ganglion neurons in neonates. Enhancing LIGHT signaling in these neurons by overexpressing LIGHT inhibited BDNF-promoted neurite growth during a narrow window of development in the immediate perinatal period without affecting neuronal survival. Overexpressing a LIGHT mutant that selectively activates HVEM, but not one that selectively activates LTbetaR, also inhibited BDNF-promoted growth, suggesting that neurite growth inhibition is mediated via HVEM. Blocking HVEM signaling by a function-blocking anti-HVEM antibody significantly enhanced neurite growth from nodose neurons grown both with and without BDNF. Likewise, neurons from LIGHT-deficient neonates exhibited significantly greater neurite growth than neurons from wild-type littermates in both the presence and absence of BDNF. LIGHT overexpression significantly inhibited NF-kappaB activity, while preventing LIGHT-induced NF-kappaB inhibition by overexpressing the p65 and p50 NF-kappaB subunits prevented LIGHT-mediated growth inhibition. Together, these findings show that LIGHT/HVEM signaling negatively regulates neurite growth from developing sensory neurons via NF-kappaB inhibition.


Subject(s)
Multigene Family , Neurites/physiology , Neurogenesis/physiology , Sensory Receptor Cells/physiology , Tumor Necrosis Factor Ligand Superfamily Member 14/physiology , Animals , Animals, Newborn , Cells, Cultured , Down-Regulation , Growth Inhibitors/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Multigene Family/physiology , Tumor Necrosis Factor Ligand Superfamily Member 14/biosynthesis , Tumor Necrosis Factors/biosynthesis , Tumor Necrosis Factors/physiology
16.
J Cell Biol ; 171(2): 337-48, 2005 Oct 24.
Article in English | MEDLINE | ID: mdl-16247031

ABSTRACT

Hepatocyte growth factor (HGF)/Met signaling controls cell migration, growth and differentiation in several embryonic organs and is implicated in human cancer. The physiologic mechanisms that attenuate Met signaling are not well understood. Here we report a mechanism by which mitogen-inducible gene 6 (Mig6; also called Gene 33 and receptor-associated late transducer) negatively regulates HGF/Met-induced cell migration. The effect is observed by Mig6 overexpression and is reversed by Mig6 small interfering RNA knock-down experiments; this indicates that endogenous Mig6 is part of a mechanism that inhibits Met signaling. Mig6 functions in cells of hepatic origin and in neurons, which suggests a role for Mig6 in different cell lineages. Mechanistically, Mig6 requires an intact Cdc42/Rac interactive binding site to exert its inhibitory action, which suggests that Mig6 acts, at least in part, distally from Met, possibly by inhibiting Rho-like GTPases. Because Mig6 also is induced by HGF stimulation, our results suggest that Mig6 is part of a negative feedback loop that attenuates Met functions in different contexts and cell types.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Movement/drug effects , Hepatocyte Growth Factor/antagonists & inhibitors , Neurites/drug effects , Adaptor Proteins, Signal Transducing/genetics , Animals , Cell Line , Cell Movement/physiology , Gene Expression Regulation , Hepatocyte Growth Factor/metabolism , Hepatocyte Growth Factor/pharmacology , Intracellular Signaling Peptides and Proteins , Mice , Neurites/metabolism , Protein Conformation , RNA, Messenger/genetics , Signal Transduction/physiology , cdc42 GTP-Binding Protein/genetics , cdc42 GTP-Binding Protein/metabolism , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism
17.
EMBO Rep ; 9(10): 1041-7, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18724272

ABSTRACT

Expression of the basic helix-loop-helix transcription factor HAND2 begins early in sympathetic neuron development and is essential for the differentiation of noradrenergic neurons. Here, we show that the expression of HAND2 and related HAND1 are maintained in sympathetic neurons throughout fetal and postnatal development when these neurons depend on target-derived nerve growth factor (NGF) for survival. Short interfering RNA knockdown of endogenous HAND2 and, to a lesser extent, HAND1 in neonatal sympathetic neurons cultured with NGF, reduced the expression of the NGF receptor tyrosine kinase TrkA (tropomyosin-related kinase A), as well as neuronal survival. Chromatin immunoprecipitation analysis showed that NGF promotes HAND2 binding to the TrkA minimal enhancer and that transfection of sympathetic neurons with a TrkA expression plasmid rescued the neurons from HAND knockdown. These findings show that HAND transcription factors have a crucial function in sustaining the survival of neonatal sympathetic neurons with NGF by a feed-forward loop that maintains the expression of TrkA.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Neurons/cytology , Superior Cervical Ganglion/metabolism , Animals , Animals, Newborn , Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Survival/physiology , Cells, Cultured , Mice , Nerve Growth Factors/physiology , Neurons/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/physiology , Superior Cervical Ganglion/cytology , Superior Cervical Ganglion/embryology
18.
Mol Cell Neurosci ; 41(2): 175-85, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19285136

ABSTRACT

Macrophage stimulating protein (MSP) is a pleiotropic growth factor that signals via the RON receptor tyrosine kinase. Here we demonstrate that MSP increases the proportion of cultured adult mouse DRG neurons displaying discernable neuritic processes and promotes the elongation and branching of these processes in a dose dependent manner. RON expression in adult DRG is largely restricted to nerve growth factor (NGF)-responsive nociceptive neurons, and MSP mimics the effects of NGF by increasing the expression of several mRNAs that encode functionally important proteins that are characteristically expressed by this neuronal sub-population. MSP mRNA is expressed at high levels in the peripheral target fields of DRG somatic afferents, but is undetectable in DRG, spinal cord or freshly dissected sciatic nerve. These results suggest that MSP is a peripheral target-derived neurotrophic factor for NGF-responsive adult DRG neurons.


Subject(s)
Ganglia, Spinal/cytology , Hepatocyte Growth Factor/pharmacology , Nerve Growth Factors/metabolism , Nociceptors/drug effects , Nociceptors/physiology , Proto-Oncogene Proteins/pharmacology , Animals , Mice , Neurites/metabolism , Neurites/ultrastructure , Nociceptors/cytology , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, trkA/metabolism , Receptors, Opioid, mu/genetics , Receptors, Opioid, mu/metabolism , TRPA1 Cation Channel , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism , Transient Receptor Potential Channels/genetics , Transient Receptor Potential Channels/metabolism
19.
Front Cell Dev Biol ; 8: 254, 2020.
Article in English | MEDLINE | ID: mdl-32411702

ABSTRACT

CD40-activated CD40L reverse signaling is a major physiological regulator of neural process growth from many kinds of developing neurons. Here we have investigated whether CD40L-reverse signaling also influences dendrite spine number and morphology in striatal medium spiny neurons (MSNs). Golgi preparations revealed no differences in the spine density, but because the dendrite arbors of MSNs were larger and branched in Cd40 -/- mice, the total number of spines was greater in Cd40 -/- mice. We also detected more mature spines compared with wild-type littermates. Western blot revealed that MSN cultures from Cd40 -/- mice had significantly less PSD-95 and there were changes in RhoA/B/C and Cdc42. Immunocytochemistry revealed that PSD-95 was clustered in spines in Cd40 -/- neurons compared with more diffuse labeling in Cd40 +/+ neurons. Activation of CD40L-reverse signaling with CD40-Fc prevented the changes observed in Cd40 -/- cultures. Our findings suggest that CD40L-reverse signaling influences dendrite spine morphology and related protein expression and distribution.

20.
Dev Neurobiol ; 80(3-4): 126-131, 2020 03.
Article in English | MEDLINE | ID: mdl-32289872

ABSTRACT

Axon growth rate from different populations of sensory neurons is correlated with the distance they have to grow to reach their targets in development: neurons with more distant targets extend axons at intrinsically faster rates. With growth of the embryo, later-born neurons within each population have further to extend their axons to reach their targets than early-born neurons. Here we examined whether the axon growth rate is related to birth date by studying the axon growth from neurons that differentiate in vitro from precursor cells isolated throughout the period of neurogenesis. We first showed that neurons that differentiated in vitro from different precursor cell populations exhibited differences in axon growth rate related to in vivo target distance. We then examined the axon growth rate from neurons that differentiate from the same precursor population at different stages throughout the period of neurogenesis. We studied the epibranchial placode precursors that give rise to nodose ganglion neurons in the chicken embryo. We observed a highly significant, threefold difference in axon growth rate from neurons that differentiate from precursor cells cultured early and late during the period of neurogenesis. Our findings suggest that intrinsic differences in axon growth rate are correlated with the neuronal birth date.


Subject(s)
Axons/physiology , Embryonic Development/physiology , Neurogenesis/physiology , Nodose Ganglion/growth & development , Sensory Receptor Cells/physiology , Animals , Cells, Cultured , Chick Embryo , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL