Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Cell Stem Cell ; 29(4): 528-544.e9, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35276096

ABSTRACT

The autonomic nervous system is a master regulator of homeostatic processes and stress responses. Sympathetic noradrenergic nerve fibers decrease bone mass, but the role of cholinergic signaling in bone has remained largely unknown. Here, we describe that early postnatally, a subset of sympathetic nerve fibers undergoes an interleukin-6 (IL-6)-induced cholinergic switch upon contacting the bone. A neurotrophic dependency mediated through GDNF-family receptor-α2 (GFRα2) and its ligand, neurturin (NRTN), is established between sympathetic cholinergic fibers and bone-embedded osteocytes, which require cholinergic innervation for their survival and connectivity. Bone-lining osteoprogenitors amplify and propagate cholinergic signals in the bone marrow (BM). Moderate exercise augments trabecular bone partly through an IL-6-dependent expansion of sympathetic cholinergic nerve fibers. Consequently, loss of cholinergic skeletal innervation reduces osteocyte survival and function, causing osteopenia and impaired skeletal adaptation to moderate exercise. These results uncover a cholinergic neuro-osteocyte interface that regulates skeletogenesis and skeletal turnover through bone-anabolic effects.


Subject(s)
Interleukin-6 , Osteogenesis , Cholinergic Agents , Cholinergic Fibers , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology
2.
Cell Tissue Res ; 378(3): 441-456, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31302761

ABSTRACT

Artemin is a neurotrophic factor that plays a crucial role in the regulation of neural development and regeneration and has also been implicated in the pathogenesis of inflammatory pain. The receptor for artemin, GFRα3, is expressed by sympathetic and nociceptive sensory neurons, including some that innervate the bone marrow, but it is unclear if it is also expressed in other cell types in the bone marrow. Our goal in the present study was to characterise the expression of GFRα3 in nonneuronal cells in the bone marrow. Immunohistochemical studies revealed that GFRα3-expressing cells in the bone marrow are spatially associated with blood vessels and are in intimate contact with nerve fibres. We used various combinations of markers to distinguish different cell types and found that the GFRα3-expressing cells expressed markers of nonmyelinating Schwann cells (e.g. GFAP, p75NTR, nestin). Analysis of bone marrow sections of Wnt1-reporter mice also demonstrated that they originate from the neural crest. Further characterisation using flow cytometry revealed that GFRα3 is expressed in a population of CD51+Sca1-PDGFRα- cells, reinforcing the notion that they are neural crest-derived, nonmyelinating Schwann cells. In conclusion, there is a close association between peripheral nerve terminals and a population of nonneuronal cells that express GFRα3 in the bone marrow. The nonneuronal cells have characteristics consistent with a neural crest-derived, nonmyelinating Schwann cell phenotype. Our findings provide a better understanding of the expression pattern of GFRα3 in the bone marrow microenvironment.


Subject(s)
Bone Marrow Cells/metabolism , Bone Marrow/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Nerve Tissue Proteins/metabolism , Schwann Cells/metabolism , Animals , Bone Marrow Cells/cytology , Mice , Mice, Inbred C57BL , Schwann Cells/cytology
3.
Biochem Biophys Res Commun ; 510(3): 339-344, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30722993

ABSTRACT

Neuroblastoma is a childhood tumor, and high-stage neuroblastoma has a poor prognosis. The regulatory mechanisms for neuroblastoma progression are poorly understood. In present study, we found that GDNF family receptor alpha 2 (GFRA2) was upregulated in neuroblastoma cells and tissues, and its overexpression promoted neuroblastoma cell proliferation, as revealed using colony formation, soft agar growth, and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assays Tumor suppressor phosphatase and tensin homolog (PTEN) is an inhibitor of the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/AKT serine/threonine kinase (AKT) pathway that interacts with GFRA2. A luciferase activity assay showed GFRA2 inhibits the transcriptional activity of the forkhead box O (FOXO) family proteins, which suggested that GFRA2 activated the PI3K/AKT pathway. Inhibition of the PI3K/AKT pathway in GFRA2 overexpressing cells decreased cell proliferation, confirming that GFRA2 promoted neuroblastoma cell proliferation by activating the PI3K/AKT pathway. In summary, cell proliferation via the GFRA2-PTEN-PI3K/AKT axis may represent new target to develop treatments for neuroblastoma.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Neuroblastoma/metabolism , PTEN Phosphohydrolase/metabolism , Cell Line, Tumor , Cell Proliferation , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Humans , Neuroblastoma/enzymology , Neuroblastoma/genetics , Neuroblastoma/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Up-Regulation
4.
Blood ; 133(3): 224-236, 2019 01 17.
Article in English | MEDLINE | ID: mdl-30361261

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) and leukocytes circulate between the bone marrow (BM) and peripheral blood following circadian oscillations. Autonomic sympathetic noradrenergic signals have been shown to regulate HSPC and leukocyte trafficking, but the role of the cholinergic branch has remained unexplored. We have investigated the role of the cholinergic nervous system in the regulation of day/night traffic of HSPCs and leukocytes in mice. We show here that the autonomic cholinergic nervous system (including parasympathetic and sympathetic) dually regulates daily migration of HSPCs and leukocytes. At night, central parasympathetic cholinergic signals dampen sympathetic noradrenergic tone and decrease BM egress of HSPCs and leukocytes. However, during the daytime, derepressed sympathetic noradrenergic activity causes predominant BM egress of HSPCs and leukocytes via ß3-adrenergic receptor. This egress is locally supported by light-triggered sympathetic cholinergic activity, which inhibits BM vascular cell adhesion and homing. In summary, central (parasympathetic) and local (sympathetic) cholinergic signals regulate day/night oscillations of circulating HSPCs and leukocytes. This study shows how both branches of the autonomic nervous system cooperate to orchestrate daily traffic of HSPCs and leukocytes.


Subject(s)
Cell Movement , Cholinergic Agents/pharmacology , Circadian Rhythm , Hematopoietic Stem Cells/physiology , Leukocytes/physiology , Parasympathetic Nervous System/physiology , Sympathetic Nervous System/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Cell Adhesion , Cells, Cultured , Chemotaxis , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Female , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Leukocytes/cytology , Leukocytes/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Adrenergic, beta-2 , Receptors, Adrenergic, beta-3/physiology , Receptors, G-Protein-Coupled/physiology
5.
Cell Stem Cell ; 24(1): 79-92.e6, 2019 01 03.
Article in English | MEDLINE | ID: mdl-30581080

ABSTRACT

In many tissues, homeostasis is maintained by physical contact between stem cells and an anatomically defined niche. However, how stem cell homeostasis is achieved in environments where cells are motile and dispersed among their progeny remains unknown. Using murine spermatogenesis as a model, we find that spermatogenic stem cell density is tightly regulated by the supply of fibroblast growth factors (FGFs) from lymphatic endothelial cells. We propose that stem cell homeostasis is achieved through competition for a limited supply of FGFs. We show that the quantitative dependence of stem cell density on FGF dosage, the biased localization of stem cells toward FGF sources, and stem cell dynamics during regeneration following injury can all be predicted and explained within the framework of a minimal theoretical model based on "mitogen competition." We propose that this model provides a generic and robust mechanism to support stem cell homeostasis in open, or facultative, niche environments.


Subject(s)
Fibroblast Growth Factor 5/physiology , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Homeostasis , Mitogens/pharmacology , Spermatogenesis , Spermatozoa/cytology , Stem Cells/cytology , Animals , Cell Differentiation , Cell Self Renewal , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Spermatozoa/physiology , Stem Cells/drug effects , Stem Cells/physiology
6.
PLoS One ; 12(2): e0171416, 2017.
Article in English | MEDLINE | ID: mdl-28152033

ABSTRACT

BACKGROUND: Glial cell line-derived neurotrophic factor (GDNF) is a potent neurotrophic factor known to promote the survival and maintenance of neurons not only in the developing but also in the adult enteric nervous system. As diverticular disease (DD) is associated with reduced myenteric neurons, alterations of the GDNF system were studied in asymptomatic diverticulosis (diverticulosis) and DD. METHODS: Morphometric analysis for quantifying myenteric ganglia and neurons were assessed in colonic full-thickness sections of patients with diverticulosis and controls. Samples of tunica muscularis (TM) and laser-microdissected myenteric ganglia from patients with diverticulosis, DD and controls were analyzed for mRNA expression levels of GDNF, GFRA1, and RET by RT-qPCR. Myenteric protein expression of both receptors was quantified by fluorescence-immunohistochemistry of patients with diverticulosis, DD, and controls. RESULTS: Although no myenteric morphometric alterations were found in patients with diverticulosis, GDNF, GFRA1 and RET mRNA expression was down-regulated in the TM of patients with diverticulosis as well as DD. Furthermore GFRA1 and RET myenteric plexus mRNA expression of patients with diverticulosis and DD was down-regulated, whereas GDNF remained unaltered. Myenteric immunoreactivity of the receptors GFRα1 and RET was decreased in both asymptomatic diverticulosis and DD patients. CONCLUSION: Our data provide evidence for an impaired GDNF system at gene and protein level not only in DD but also during early stages of diverticula formation. Thus, the results strengthen the idea of a disturbed GDNF-responsiveness as contributive factor for a primary enteric neuropathy involved in the pathogenesis and disturbed intestinal motility observed in DD.


Subject(s)
Diverticulum/physiopathology , Glial Cell Line-Derived Neurotrophic Factor/physiology , Aged , Case-Control Studies , Colon/innervation , Colon/pathology , Diverticulum/pathology , Fluorescent Antibody Technique , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Humans , Laser Capture Microdissection , Male , Myenteric Plexus/pathology , Proto-Oncogene Proteins c-ret/physiology , Real-Time Polymerase Chain Reaction
7.
Development ; 143(22): 4224-4235, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27707798

ABSTRACT

The formation of synaptic connections during nervous system development requires the precise control of dendrite growth and synapse formation. Although glial cell line-derived neurotrophic factor (GDNF) and its receptor GFRα1 are expressed in the forebrain, the role of this system in the hippocampus remains unclear. Here, we investigated the consequences of GFRα1 deficiency for the development of hippocampal connections. Analysis of conditional Gfra1 knockout mice shows a reduction in dendritic length and complexity, as well as a decrease in postsynaptic density specializations and in the synaptic localization of postsynaptic proteins in hippocampal neurons. Gain- and loss-of-function assays demonstrate that the GDNF-GFRα1 complex promotes dendritic growth and postsynaptic differentiation in cultured hippocampal neurons. Finally, in vitro assays revealed that GDNF-GFRα1-induced dendrite growth and spine formation are mediated by NCAM signaling. Taken together, our results indicate that the GDNF-GFRα1 complex is essential for proper hippocampal circuit development.


Subject(s)
Dendrites/physiology , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Glial Cell Line-Derived Neurotrophic Factor/physiology , Hippocampus/growth & development , Neural Cell Adhesion Molecules/physiology , Neurogenesis/genetics , Neuronal Plasticity/genetics , Animals , Cell Differentiation/genetics , Cells, Cultured , Embryo, Mammalian , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Mice , Mice, Knockout , Multiprotein Complexes/physiology , Nerve Net/growth & development , Nerve Net/metabolism , Neurons/physiology , Protein Binding , Rats , Rats, Wistar
8.
J Physiol ; 591(8): 2175-88, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23339174

ABSTRACT

Efferent signals from the vagus nerve are thought to mediate both basal and meal-induced gastric acid secretion, and provide trophic support of the mucosa. However, the underlying mechanisms are incompletely understood. Neurturin, signalling via glial cell line-derived neurotrophic factor (GDNF)-family receptor α2 (GFRα2), is essential for parasympathetic innervation of many target tissues but its role in gastric innervation is unknown. Here we show that most nerve fibres in wild-type mouse gastric mucosa, including all positive for gastrin-releasing peptide, are cholinergic. GFRα2-deficient (KO) mice lacked virtually all cholinergic nerve fibres and associated glial cells in the gastric (oxyntic and pyloric) mucosa but not in the smooth muscle, consistent with the selective expression of neurturin mRNA in the gastric mucosa. 2-Deoxyglucose and hexamethonium failed to affect acid secretion in the GFRα2-KO mice indicating the lack of functional innervation in gastric mucosa. Interestingly, basal and maximal histamine-induced acid secretion did not differ between wild-type and GFRα2-KO mice. Moreover, circulating gastrin levels in both fasted and fed animals, thickness of gastric mucosa, and density of parietal and different endocrine cells were similar. Carbachol-stimulated acid secretion was higher in GFRα2-KO mice, while atropine reduced basal secretion similarly in both genotypes. We conclude that cholinergic innervation of gastric mucosa depends on neurturin-GFRα2 signalling but is dispensable for gastrin secretion and for basal and maximal acid output. Basal acid secretion in the KO mice appears to be, at least partly, facilitated by constitutive activity of muscarinic receptors.


Subject(s)
Gastric Acid/metabolism , Gastric Mucosa/innervation , Gastrins/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Animals , Cholinergic Neurons/physiology , Female , Gastrins/blood , Hydrogen-Ion Concentration , Male , Mice , Mice, Knockout , Neuroglia/physiology , Neurturin/physiology
9.
J Dent Res ; 90(10): 1240-5, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21828353

ABSTRACT

This study investigated the effects of glial cell line-derived neurotrophic factor (GDNF) on dental pulp cells (DPCs). Cultures of DPCs expressed GDNF as well as its receptors, GFRα1 and RET. Addition of recombinant GDNF to cultures in serum-containing medium did not significantly affect DPC growth; however, GDNF dose-dependently increased viable cell number under serum-free culture conditions. Live/dead, lactate dehydrogenase (LDH), and caspases-3/-7 assays demonstrated that cell death occurred under serum-free conditions, and that GDNF significantly reduced the number of dead cells by inhibiting apoptotic cell death. GDNF also stimulated cell proliferation in serum-free conditions, as assessed by the BrdU incorporation assay. The effect of GDNF was abolished in the presence of inhibitors to GFRα1 and RET suggesting receptor-mediated events. This study also demonstrated that GDNF counteracted TNFα-induced DPC cytotoxicity, suggesting that GDNF may be cytoprotective under disease conditions. In conclusion, our findings indicate that GDNF promotes cell survival and proliferation of DPCs and suggest that GDNF may play a multifunctional role in the regulation of dental pulp homeostasis.


Subject(s)
Apoptosis , Dental Pulp/cytology , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Glial Cell Line-Derived Neurotrophic Factor/physiology , Proto-Oncogene Proteins c-ret/physiology , Animals , Cell Proliferation , Cell Survival , Cells, Cultured , Culture Media, Serum-Free , Dental Pulp/metabolism , Glial Cell Line-Derived Neurotrophic Factor/biosynthesis , Glial Cell Line-Derived Neurotrophic Factor Receptors/biosynthesis , Humans , Male , Proto-Oncogene Proteins c-ret/biosynthesis , Rats , Rats, Wistar , Tumor Necrosis Factor-alpha/antagonists & inhibitors
11.
Development ; 138(10): 2015-24, 2011 May.
Article in English | MEDLINE | ID: mdl-21490065

ABSTRACT

An appropriate organisation of muscles is crucial for their function, yet it is not known how functionally related muscles are coordinated with each other during development. In this study, we show that the development of a subset of functionally related head muscles in the zebrafish is regulated by Ret tyrosine kinase signalling. Three genes in the Ret pathway (gfra3, artemin2 and ret) are required specifically for the development of muscles attaching to the opercular bone (gill cover), but not other adjacent muscles. In animals lacking Ret or Gfra3 function, myogenic gene expression is reduced in forming opercular muscles, but not in non-opercular muscles derived from the same muscle anlagen. These animals have a normal skeleton with small or missing opercular muscles and tightly closed mouths. Myogenic defects correlate with a highly restricted expression of artn2, gfra3 and ret in mesenchymal cells in and around the forming opercular muscles. ret(+) cells become restricted to the forming opercular muscles and a loss of Ret signalling results in reductions of only these, but not adjacent, muscles, revealing a specific role of Ret in a subset of head muscles. We propose that Ret signalling regulates myogenesis in head muscles in a modular manner and that this is achieved by restricting Ret function to a subset of muscle precursors.


Subject(s)
Muscle Development/physiology , Proto-Oncogene Proteins c-ret/physiology , Zebrafish Proteins/physiology , Zebrafish/embryology , Zebrafish/physiology , Animals , Animals, Genetically Modified , Base Sequence , Biological Evolution , DNA Primers/genetics , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Glial Cell Line-Derived Neurotrophic Factor Receptors/deficiency , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Head , Muscle Development/genetics , Muscle, Skeletal/embryology , Mutation , Phenotype , Proto-Oncogene Proteins c-ret/deficiency , Proto-Oncogene Proteins c-ret/genetics , Signal Transduction , Zebrafish/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
12.
Curr Biol ; 21(1): R30-2, 2011 Jan 11.
Article in English | MEDLINE | ID: mdl-21215934

ABSTRACT

The pathfinding of motor axons is an important model system for understanding binary axon guidance decisions. Recent work has shown that GDNF attracts motor neuron growth cones, and interacts synergistically with ephrinAs on growth cone directionality.


Subject(s)
Axons/physiology , Ephrins/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Motor Neurons/cytology , Animals , Chickens , Gene Expression Regulation/physiology , Mice , Motor Neurons/physiology
13.
J Neurochem ; 115(3): 759-70, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20807316

ABSTRACT

Malignant gliomas are highly invasive neuroepithelial tumors where the tendency to invade and migrate away from the primary tumor mass is thought to be a leading cause of tumor recurrence and treatment failures. Autocrine signals produced by secreted factors that signal through receptors on the tumor are known to contribute to the invasiveness. Glial cell line-derived neurotrophic factor and GDNF family receptor alpha 1 (GFRα1) are over-expressed in human gliomas. We have previously reported that human gliomas express high levels of GFRα1b, an alternatively spliced isoform of GFRα1. However, the functional significance of GFRα1b in glioma behaviors is currently unknown. In this study, we have designed isoform-specific small-interfering RNA to knockdown the highly homologous GFRα1a or GFRα1b isoform efficiently in malignant C6 glioma cells. Unexpectedly, the knockdown of GFRα1b but not GFRα1a induced cell elongation and inhibited C6 cell migration and invasion in vitro. In addition, GFRα1b was found to regulate the expression of RhoA small GTPase, which was required for migration of C6 cells. The decreases in RhoA expression and cell migration after GFRα1b knockdown were attenuated by small-interfering RNA -resistant GFRα1b but not GFRα1a, further demonstrating the specific role of GFRα1b in glioma migration. Interestingly, the knockdown of NCAM but not receptor tyrosine kinase Ret resulted in the reduction of RhoA expression and C6 cell migration. Taken together, these unanticipated results indicate that GFRα1b is involved in glioma migration through glial cell line-derived neurotrophic factor -GFRα1b-NCAM signaling complex and modulation of RhoA expression.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Glioma/pathology , rhoA GTP-Binding Protein/biosynthesis , Animals , Blotting, Western , Cell Division/physiology , Cell Movement/genetics , Cell Movement/physiology , Fluorescent Antibody Technique , Glial Cell Line-Derived Neurotrophic Factor , Glial Cell Line-Derived Neurotrophic Factor Receptors/chemistry , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Isomerism , Microscopy, Confocal , Neoplasm Invasiveness/pathology , Neural Cell Adhesion Molecules/biosynthesis , Neural Cell Adhesion Molecules/genetics , RNA, Small Interfering/genetics , Rats , Reverse Transcriptase Polymerase Chain Reaction , Transfection
14.
Front Horm Res ; 38: 127-138, 2010.
Article in English | MEDLINE | ID: mdl-20616503

ABSTRACT

The RET receptor is a tyrosine kinase receptor implicated in kidney and neural development. In the adenopituitary RET and the co-receptor GFRa1 are expressed exclusively in the somatotrophs secreting GH. RET is implicated in a clever pathway to maintain at physiological levels the number of somatotrophs and the GH production. Thus, in absence of its ligand GDNF, RET induces apoptosis through massive expression of Pit-1 leading to p53 accumulation. In the presence of the ligand GDNF, RET activates its tyrosine kinase and promotes survival at the expense of reducing Pit-1 expression and downregulating GH. Recent data suggest that RET can also have a second role in pituitary plasticity through a second co-receptor GFRa2.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Glial Cell Line-Derived Neurotrophic Factors/physiology , Pituitary Gland/physiology , Proto-Oncogene Proteins c-ret/physiology , Animals , Humans , Promoter Regions, Genetic , Signal Transduction , Transcription Factor Pit-1/physiology , Tumor Suppressor Protein p53/physiology
15.
Development ; 136(24): 4213-24, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19934016

ABSTRACT

Ureteric bud (UB) emergence from the Wolffian duct (WD), the initiating step in metanephric kidney morphogenesis, is dependent on GDNF; however, GDNF by itself is generally insufficient to induce robust budding of the isolated WD in culture. Thus, additional factors, presumably peptides or polypeptide growth factors, might be involved. Microarray data from in vivo budding and non-budding conditions were analyzed using non-negative matrix factorization followed by gene ontology filtering and network analysis to identify sets of genes that are highly regulated during budding. These included the GDNF co-receptors GFRalpha1 and RET, as well as neuropeptide Y (NPY). By using ANOVA with pattern matching, NPY was also found to correlate most significantly to the budded condition with a high degree of connectedness to genes with developmental roles. Exogenous NPY [as well as its homolog, peptide YY (PYY)] augmented GDNF-dependent budding in the isolated WD culture; conversely, inhibition of NPY signaling or perturbation of NPY expression inhibited budding, confirming that NPY facilitates this process. NPY was also found to reverse the decreased budding, the downregulation of RET expression, the mislocalization of GFRalpha1, and the inhibition of AKT phosphorylation that resulted from the addition of BMP4 to the isolated WD cultures, suggesting that NPY acts through the budding pathway and is reciprocally regulated by GDNF and BMP4. Thus, the outgrowth of the UB from the WD might result from a combination of the upregulation of the GDNF receptors together with genes that support GDNF signaling in a feed-forward loop and/or counteraction of the inhibitory pathway regulated by BMP4.


Subject(s)
Neuropeptide Y/physiology , Wolffian Ducts/physiology , Animals , Bone Morphogenetic Protein 4/physiology , Cells, Cultured , Female , Gene Expression Regulation, Developmental , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Morphogenesis , Oligonucleotide Array Sequence Analysis , Proto-Oncogene Proteins c-ret/physiology , Rats , Rats, Sprague-Dawley , Signal Transduction , Up-Regulation , Wolffian Ducts/embryology
16.
Glia ; 56(13): 1428-37, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18551627

ABSTRACT

The phenotypic development of satellite cells in mouse sympathetic ganglia was examined by localizing the transcription factors, Sox10 and Phox2b, the neuronal marker, tyrosine hydroxylase (TH), and brain-derived fatty acid binding protein (B-FABP), which identifies glial precursors and mature glia. In E10.5 mice, most cells in the sympathetic chain expressed both Sox10 and Phox2b, with a minority of cells expressing Sox10 only or Phox2b only. In E11.5 mice, the majority of cells expressed Sox10 only or Phox2b only. B-FABP was colocalized with Sox10 in satellite glial precursors, which were located on the periphery of the ganglion. There was no overlap between B-FABP and Phox2b or B-FABP and TH. During subsequent development, the number of B-FABP+ cells increased and they became more common deep within the ganglion. In E12.5 and E18.5 mice, there was no overlap between Sox10 and Phox2b, and 98% of Sox10 cells were also B-FABP+. Satellite glial precursors in E11.5-E15.5 mice also expressed the GDNF-binding molecule, GFRalpha1. B-FABP immunoreactive cells did not express Ret or NCAM, two potential signaling molecules for GDNF/GFRalpha1. In E12.5 and E18.5 mice lacking GFRalpha1 or GDNF, the development of B-FABP immunoreactive satellite cells was normal, and hence neither GDNF or GFRalpha1 are essential for the development of satellite glia in sympathetic ganglia.


Subject(s)
Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/growth & development , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Glial Cell Line-Derived Neurotrophic Factor/physiology , Neuroglia/cytology , Neuroglia/physiology , Animals , Female , Glial Cell Line-Derived Neurotrophic Factor/deficiency , Glial Cell Line-Derived Neurotrophic Factor/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/deficiency , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pregnancy
17.
Dev Biol ; 307(1): 14-28, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17509555

ABSTRACT

During chick liver development, the liver bud arises from the foregut, invaginates into the septum transversum, and elongates along and envelops the ductus venosus. However, the mechanism of liver bud migration is only poorly understood. Here, we demonstrate that a GDNF family ligand involved in neuronal outgrowth and migration, neurturin (NRTN), and its receptor, GFRalpha2, are essential for liver bud migration. In the chick embryo, we found that GFRalpha2 was expressed in the liver bud and that NRTN was expressed in the endothelial cells of the ductus venosus. Inhibition of GFRalpha2 signaling suppressed liver bud elongation along the ductus venous without affecting cell proliferation and apoptosis. Moreover, ectopic expression of NRTN perturbed the directional migration along the ductus venosus, leading to splitting or ectopic branching of the liver. We showed that liver buds selectively migrated toward an NRTN-soaked bead in vitro. These data represent a new model for liver bud migration: NRTN secreted from endothelial cells functions as a chemoattractant to direct the migration of the GFRalpha2-expressing liver bud in early liver development.


Subject(s)
Cell Movement , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Liver/embryology , Neurturin/physiology , Animals , Blood Circulation , Chemotactic Factors/physiology , Chick Embryo , Endothelial Cells , Liver/cytology , Liver/growth & development , Morphogenesis , Organogenesis , Signal Transduction
18.
J Neurosci ; 27(21): 5603-14, 2007 May 23.
Article in English | MEDLINE | ID: mdl-17522305

ABSTRACT

The glial cell line-derived neurotrophic factor (GDNF) and neurturin (NTN) belong to a structurally related family of neurotrophic factors. NTN exerts its effect through a multicomponent receptor system consisting of the GDNF family receptor alpha2 (GFR alpha2), RET, and/or NCAM (neural cell adhesion molecule). GFR alpha2 is alternatively spliced into at least three isoforms (GFR alpha2a, GFR alpha2b, and GFR alpha2c). It is currently unknown whether these isoforms share similar functional and biochemical properties. Using highly specific and sensitive quantitative real-time PCR, these isoforms were found to be expressed at comparable levels in various regions of the human brain. When stimulated with GDNF and NTN, both GFR alpha2a and GFR alpha2c, but not GFR alpha2b, promoted neurite outgrowth in transfected Neuro2A cells. These isoforms showed ligand selectivity in MAPK (mitogen-activated protein kinase) [ERK1/2 (extracellular signal-regulated kinase 1/2)] and Akt signaling. In addition, the GFR alpha2 isoforms regulated different early-response genes when stimulated with GDNF or NTN. In coexpression studies, GFR alpha2b was found to inhibit ligand-induced neurite outgrowth by GFR alpha2a and GFR alpha2c. Stimulation of GFR alpha2b also inhibited the neurite outgrowth induced by GFR alpha1a, another member of the GFR alpha. Furthermore, activation of GFR alpha2b inhibited neurite outgrowth induced by retinoic acid and activated RhoA. Together, these data suggest a novel paradigm for the regulation of growth factor signaling and neurite outgrowth via an inhibitory splice variant of the receptor. Thus, depending on the expressions of specific GFR alpha2 receptor spliced isoforms, GDNF and NTN may promote or inhibit neurite outgrowth through the multicomponent receptor complex.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Glial Cell Line-Derived Neurotrophic Factor/physiology , Neurites/physiology , Neurturin/physiology , rhoA GTP-Binding Protein/metabolism , Alternative Splicing/physiology , Animals , Cell Line , Cell Proliferation , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Growth Inhibitors/physiology , Humans , Mice , Protein Isoforms/genetics , Protein Isoforms/physiology
19.
Nat Neurosci ; 10(3): 293-300, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17310246

ABSTRACT

The establishment of synaptic connections requires precise alignment of pre- and postsynaptic terminals. The glial cell line-derived neurotrophic factor (GDNF) receptor GFRalpha1 is enriched at pre- and postsynaptic compartments in hippocampal neurons, suggesting that it has a function in synapse formation. GDNF triggered trans-homophilic binding between GFRalpha1 molecules and cell adhesion between GFRalpha1-expressing cells. This represents the first example of a cell-cell interaction being mediated by a ligand-induced cell adhesion molecule (LICAM). In the presence of GDNF, ectopic GFRalpha1 induced localized presynaptic differentiation in hippocampal neurons, as visualized by clustering of vesicular proteins and neurotransmitter transporters, and by activity-dependent vesicle recycling. Presynaptic differentiation induced by GDNF was markedly reduced in neurons lacking GFRalpha1. Gdnf mutant mice showed reduced synaptic localization of presynaptic proteins and a marked decrease in the density of presynaptic puncta, indicating a role for GDNF signaling in hippocampal synaptogenesis in vivo. We propose that GFRalpha1 functions as a LICAM to establish precise synaptic contacts and induce presynaptic differentiation.


Subject(s)
Cell Adhesion/physiology , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Glial Cell Line-Derived Neurotrophic Factor/physiology , Neurons/cytology , Synapses/physiology , Analysis of Variance , Animals , Brain/cytology , Cell Differentiation/physiology , Cell Membrane/metabolism , Cells, Cultured , Embryo, Mammalian , Gene Expression , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Ligands , Mice , Mice, Knockout , Nerve Growth Factors/metabolism , Neural Cell Adhesion Molecules/deficiency , Proto-Oncogene Proteins c-ret/deficiency , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction/methods
20.
Trends Pharmacol Sci ; 28(2): 68-74, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17218019

ABSTRACT

Glial-cell-line-derived neurotrophic factor (GDNF) family ligands (GFLs), which consist of GDNF, neurturin, artemin and persephin, regulate the development and maintenance of the nervous system. GDNF protects and repairs dopamine-containing neurons, which degenerate in Parkinson's disease, and motoneurons, which die in amyotrophic lateral sclerosis. GDNF and neurturin have shown promise in clinical trials of Parkinson's disease, and artemin is currently undergoing clinical trials for chronic pain treatment. However, the delivery of GFLs into the brain through invasive approaches such as neurosurgery, viral vectors or by the use of encapsulated cells is associated with multiple obstacles. The development of small molecules that specifically activate GFL receptors and that can be applied systemically would overcome most of these problems. The unique nature of the GFL receptors, recent progress in elucidation of the 3D structures of GFLs and GFL-receptor complexes and the use of high-throughput screening have resulted in the development of the first small molecules that mimic the effects of the different GFLs.


Subject(s)
Glial Cell Line-Derived Neurotrophic Factor Receptors/drug effects , Glial Cell Line-Derived Neurotrophic Factor Receptors/physiology , Glial Cell Line-Derived Neurotrophic Factor/physiology , Nervous System Diseases/drug therapy , Nervous System Diseases/physiopathology , Animals , Chronic Disease , Humans , Molecular Mimicry , Nerve Growth Factors/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...