Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
PLoS One ; 10(12): e0144185, 2015.
Article in English | MEDLINE | ID: mdl-26692198

ABSTRACT

Elevated plasma triglycerides are associated with increased susceptibility to heart disease and stroke, but the mechanisms behind this relationship are unclear. A clearer understanding of gene products which influence plasma triglycerides might help identify new therapeutic targets for these diseases. The Endothelial Cell Surface expressed Chemotaxis and apoptosis Regulator (ECSCR) was initially studied as an endothelial cell marker, but has recently been identified in white adipocytes, the primary storage cell type for triglycerides. Here we confirm ECSCR expression in white adipocytes and show that Ecscr knockout mice show elevated fasting plasma triglycerides. At a cellular level, cultured 3T3-L1 adipocytes silenced for Ecscr show a blunted Akt phosphorylation response. Additionally we show that the phosphatase and tensin homology containing (PTEN) lipid phosphatase association with ECSCR is increased by insulin stimulation. These data suggest a scenario by which ECSCR contributes to control of white adipocyte lipolysis. In this scenario, white adipocytes lacking Ecscr display elevated PTEN activity, thereby reducing AKT activation and impairing insulin-mediated suppression of lipolysis. Collectively, these results suggest that ECSCR plays a critical function in regulating lipolysis in white adipose tissue.


Subject(s)
Adipocytes, White/metabolism , Apoptosis Regulatory Proteins/metabolism , Lipolysis/physiology , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , 3T3-L1 Cells , Adipocytes, White/cytology , Animals , Apoptosis Regulatory Proteins/genetics , Membrane Proteins , Mice , Mice, Knockout , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins c-akt/genetics , Triglycerides/genetics , Triglycerides/metabolism
2.
PLoS One ; 8(10): e76484, 2013.
Article in English | MEDLINE | ID: mdl-24098510

ABSTRACT

The extracellular matrix plays a critical role in neural crest (NC) cell migration. In this study, we characterize the contribution of the novel GPI-linked matrix metalloproteinase (MMP) zebrafish mmp17b. Mmp17b is expressed post-gastrulation in the developing NC. Morpholino inactivation of mmp17b function, or chemical inhibition of MMP activity results in aberrant NC cell migration with minimal change in NC proliferation or apoptosis. Intriguingly, a GPI anchored protein with metalloproteinase inhibitor properties, Reversion-inducing-Cysteine-rich protein with Kazal motifs (RECK), which has previously been implicated in NC development, is expressed in close apposition to NC cells expressing mmp17b, raising the possibility that these two gene products interact. Consistent with this possibility, embryos silenced for mmp17b show defective development of the dorsal root ganglia (DRG), a crest-derived structure affected in RECK mutant fish sensory deprived (sdp). Taken together, this study has identified the first pair of MMP, and their putative MMP inhibitor RECK that functions together in NC cell migration.


Subject(s)
Cell Movement/genetics , Matrix Metalloproteinase 17/genetics , Matrix Metalloproteinase 17/metabolism , Neural Crest/cytology , Neural Crest/metabolism , Amino Acid Sequence , Animals , Body Patterning/genetics , Embryonic Development/genetics , Enzyme Activation , GPI-Linked Proteins/genetics , GPI-Linked Proteins/metabolism , Gene Expression Profiling , Matrix Metalloproteinase 17/chemistry , Molecular Sequence Data , Organ Specificity/genetics , Sequence Alignment , Zebrafish
3.
J Biol Chem ; 288(15): 10265-74, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23393131

ABSTRACT

The endothelial cell-specific chemotaxis receptor (ECSCR) is a cell-surface protein selectively expressed by endothelial cells (ECs), with roles in EC migration, apoptosis and proliferation. Our previous study (Verma, A., Bhattacharya, R., Remadevi, I., Li, K., Pramanik, K., Samant, G. V., Horswill, M., Chun, C. Z., Zhao, B., Wang, E., Miao, R. Q., Mukhopadhyay, D., Ramchandran, R., and Wilkinson, G. A. (2010) Blood 115, 4614-4622) showed that loss of ECSCR in primary ECs reduced tyrosine phosphorylation of vascular endothelial growth factor (VEGF) receptor 2/kinase insert domain receptor (KDR) but not VEGF receptor 1/FLT1. Here, we show that ECSCR biochemically associates with KDR but not FLT1 and that the predicted ECSCR cytoplasmic and transmembrane regions can each confer association with KDR. Stimulation with VEGF165 rapidly and transiently increases ECSCR-KDR complex formation, a process blocked by the KDR tyrosine kinase inhibitor compound SU5416 or inhibitors of endosomal acidification. Triple labeling experiments show VEGF-stimulated KDR(+)/ECSCR(+) intracellular co-localization. Silencing of ECSCR disrupts VEGF-induced KDR activation and AKT and ERK phosphorylation and impairs VEGF-stimulated KDR degradation. In zebrafish, ecscr interacts with kdrl during intersomitic vessel sprouting. Human placenta and infantile hemangioma samples highly express ECSCR protein, suggesting a role for ECSCR-KDR interaction in these tissues.


Subject(s)
Human Umbilical Vein Endothelial Cells/metabolism , Membrane Proteins/metabolism , Proteolysis , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Animals, Genetically Modified , Apoptosis Regulatory Proteins , Cell Line , Endosomes/genetics , Endosomes/metabolism , Enzyme Activation/drug effects , Enzyme Activation/physiology , Extracellular Signal-Regulated MAP Kinases/genetics , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Hemangioma/genetics , Hemangioma/metabolism , Hemangioma/pathology , Human Umbilical Vein Endothelial Cells/cytology , Humans , Indoles/pharmacology , Male , Membrane Proteins/genetics , Placenta/metabolism , Placenta/pathology , Pregnancy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt , Pyrroles/pharmacology , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/metabolism , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/genetics , Zebrafish
4.
J Biol Chem ; 286(35): 30740-30747, 2011 Sep 02.
Article in English | MEDLINE | ID: mdl-21730073

ABSTRACT

Despite their importance as members of the Roundabout (Robo) family in the control of axonal and vascular patterning, the transcriptional regulation of these genes is poorly understood. In this study, we show that members of the Sry-related high mobility box (Sox) transcription factor family as being transcriptional regulators of roundabout4 (robo4), a Robo gene family member that participates in sprouting angiogenesis in vivo, in zebrafish. Double whole mount in situ hybridization analysis in zebrafish embryos revealed co-localization of the vascular relevant Sox factors sox7 or sox18 mRNA with robo4 transcripts in developing intersomitic vessels. A 3-kb human ROBO4 promoter element was able to drive reporter expression in zebrafish to recapitulate the endogenous temporal intersomitic vessel expression pattern of robo4. EMSA analysis confirmed binding of Sox18 to a canonical Sox binding site (from -1170 bp to -1176 bp) in the ROBO4 promoter (3 kb), and mutation analysis indicated that this site was partially responsible for ROBO4 promoter activity in ECs. A combination of gain- and loss-of-function analysis identified Sox7 and Sox18 co-regulation of robo4 but not fli1a transcripts in zebrafish. Finally, Sox-mediated robo4 transcriptional regulation is conserved across evolution. These studies imply Sox-mediated transcriptional regulation of Robo4 in the developing embryonic vasculature.


Subject(s)
Gene Expression Regulation, Developmental , Receptors, Cell Surface/biosynthesis , Zebrafish Proteins/biosynthesis , Animals , Cell Movement , DNA Mutational Analysis , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Mice , Mutation , Neovascularization, Pathologic , Oligonucleotide Array Sequence Analysis , Promoter Regions, Genetic , Receptors, Cell Surface/physiology , SOXF Transcription Factors/metabolism , Transcription, Genetic , Zebrafish , Zebrafish Proteins/physiology
5.
Blood ; 116(24): 5423-33, 2010 Dec 09.
Article in English | MEDLINE | ID: mdl-20813898

ABSTRACT

Our previous work has shown that axon guidance gene family Nogo-B and its receptor (NgBR) are essential for chemotaxis and morphogenesis of endothelial cells in vitro. To investigate NogoB-NgBR function in vivo, we cloned the zebrafish ortholog of both genes and studied loss of function in vivo using morpholino antisense technology. Zebrafish ortholog of Nogo-B is expressed in somite while expression of zebrafish NgBR is localized in intersomitic vessel (ISV) and axial dorsal aorta during embryonic development. NgBR or Nogo-B knockdown embryos show defects in ISV sprouting in the zebrafish trunk. Mechanistically, we found that NgBR knockdown not only abolished its ligand Nogo-B-stimulated endothelial cell migration but also reduced the vascular endothelial growth factor (VEGF)-stimulated phosphorylation of Akt and vascular endothelial growth factor-induced chemotaxis and morphogenesis of human umbilical vein endothelial cells. Further, constitutively activated Akt (myristoylated [myr]Akt) or human NgBR can rescue the NgBR knockdown umbilical vein endothelial cell migration defects in vitro or NgBR morpholino-caused ISV defects in vivo. These data place Akt at the downstream of NgBR in both Nogo-B- and VEGF-coordinated sprouting of ISVs. In summary, this study identifies the in vivo functional role for Nogo-B and its receptor (NgBR) in angiogenesis in zebrafish.


Subject(s)
Neovascularization, Physiologic , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Cell Surface/physiology , Zebrafish Proteins/physiology , Animals , Chemotaxis , Embryonic Development , Endothelial Cells/cytology , Endothelial Cells/physiology , Endothelium, Vascular/cytology , Humans , Phosphorylation , RNA, Antisense/pharmacology , Receptors, Cell Surface/genetics , Vascular Endothelial Growth Factor A/physiology , Zebrafish , Zebrafish Proteins/genetics
6.
Nature ; 465(7297): 487-91, 2010 May 27.
Article in English | MEDLINE | ID: mdl-20445540

ABSTRACT

The formation and guidance of specialized endothelial tip cells is essential for both developmental and pathological angiogenesis. Notch-1 signalling regulates the generation of tip cells, which respond to gradients of vascular endothelial growth factor (VEGF-A). The molecular cues and signalling pathways that control the guidance of tip cells are poorly understood. Bidirectional signalling by Eph receptors and ephrin ligands represents one of the most important guidance cues involved in axon path finding. Here we show that ephrin-B2 reverse signalling involving PDZ interactions regulates endothelial tip cell guidance to control angiogenic sprouting and branching in physiological and pathological angiogenesis. In vivo, ephrin-B2 PDZ-signalling-deficient mice (ephrin-B2DeltaV) exhibit a reduced number of tip cells with fewer filopodial extensions at the vascular front in the mouse retina. In pathological settings, impaired PDZ signalling decreases tumour vascularization and growth. Mechanistically, we show that ephrin-B2 controls VEGF receptor (VEGFR)-2 internalization and signalling. Importantly, internalization of VEGFR2 is necessary for activation and downstream signalling of the receptor and is required for VEGF-induced tip cell filopodial extension. Together, our results suggest that ephrin-B2 at the tip cell filopodia regulates the proper spatial activation of VEGFR2 endocytosis and signalling to direct filopodial extension. Blocking ephrin-B2 reverse signalling may be an attractive alternative or combinatorial anti-angiogenic therapy strategy to disrupt VEGFR2 function in tumour angiogenesis.


Subject(s)
Astrocytoma/blood supply , Astrocytoma/metabolism , Ephrin-B2/metabolism , Neovascularization, Pathologic , Vascular Endothelial Growth Factor Receptor-2/metabolism , Animals , Astrocytoma/pathology , Brain/blood supply , Cells, Cultured , Endocytosis , Endothelial Cells/cytology , Endothelial Cells/metabolism , Ephrin-B2/deficiency , Ephrin-B2/genetics , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Neovascularization, Physiologic , Pseudopodia/metabolism , Retina , Retinal Vessels/cytology , Retinal Vessels/physiology , Signal Transduction
7.
Blood ; 115(22): 4614-22, 2010 Jun 03.
Article in English | MEDLINE | ID: mdl-20086248

ABSTRACT

Endothelial cell-specific chemotaxis receptor (ECSCR) is a cell surface protein expressed by blood endothelial cells with roles in endothelial cell migration and signal transduction. We investigated the function of ecscr in the development of the zebrafish vasculature. Zebrafish ecscr is expressed in angioblasts and in axial vessels during angioblast migration and vasculogenesis. Morpholino-directed ecscr knockdown resulted in defective angioblast migration in the posterior lateral plate mesoderm, a process known to depend on vascular endothelial-derived growth factor (VEGF). In cultured cells, transfected ECSCR localized to actin-rich membrane protrusions, colocalizing with kinase insert domain protein receptor (KDR)/VEGF receptor 2 in these regions. ECSCR-silenced cells show reduced VEGF-induced phosphorylation of KDR but not of FMS-like tyrosine kinase 1 (FLT1)/VEGF receptor 1. Finally, chemical inhibition of VEGF receptor activity in zebrafish resulted in angioblast deficiencies that partially overlap with those seen in ecscr morphants. We propose that ecscr promotes migration of zebrafish angioblasts by enhancing endothelial kdr sensitivity to VEGF.


Subject(s)
Blood Vessels/embryology , Blood Vessels/metabolism , Chemotaxis/physiology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Zebrafish/metabolism , Animals , Apoptosis , Apoptosis Regulatory Proteins , Base Sequence , Cell Movement , Cell Proliferation , Cells, Cultured , DNA Primers/genetics , Endothelial Cells/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transfection , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors , Vascular Endothelial Growth Factor Receptor-2/genetics , Zebrafish/genetics , Zebrafish Proteins/genetics
8.
Blood ; 115(1): 133-9, 2010 Jan 07.
Article in English | MEDLINE | ID: mdl-19880500

ABSTRACT

Recently, messenger RNAs in eukaryotes have shown to associate with antisense (AS) transcript partners that are often referred to as long noncoding RNAs (lncRNAs) whose function is largely unknown. Here, we have identified a natural AS transcript for tyrosine kinase containing immunoglobulin and epidermal growth factor homology domain-1 (tie-1), tie-1AS lncRNA in zebrafish, mouse, and humans. In embryonic zebrafish, tie-1AS lncRNA transcript is expressed temporally and spatially in vivo with its native target, the tie-1 coding transcript and in additional locations (ear and brain). The tie-1AS lncRNA selectively binds tie-1 mRNA in vivo and regulates tie-1 transcript levels, resulting in specific defects in endothelial cell contact junctions in vivo and in vitro. The ratio of tie-1 versus tie-1AS lncRNA is altered in human vascular anomaly samples. These results directly implicate noncoding RNA-mediated transcriptional regulation of gene expression as a fundamental control mechanism for physiologic processes, such as vascular development.


Subject(s)
Genetic Loci/genetics , RNA, Antisense/genetics , RNA, Untranslated/genetics , Zebrafish Proteins/metabolism , Zebrafish/genetics , Animals , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium/drug effects , Endothelium/metabolism , Endothelium/pathology , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Intercellular Junctions/drug effects , Intercellular Junctions/metabolism , Intercellular Junctions/pathology , Mice , Neovascularization, Physiologic/drug effects , Phenotype , RNA, Antisense/metabolism , RNA, Untranslated/metabolism , Receptor, TIE-1/genetics , Receptor, TIE-1/metabolism , Species Specificity , Vascular Diseases/genetics , Vascular Diseases/pathology , Vascular Endothelial Growth Factor A/pharmacology , Zebrafish Proteins/genetics
9.
Dev Dyn ; 237(8): 2220-34, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18651661

ABSTRACT

Alveoli are formed in the lung by the insertion of secondary tissue folds, termed septa, which are subsequently remodeled to form the mature alveolar wall. Secondary septation requires interplay between three cell types: endothelial cells forming capillaries, contractile interstitial myofibroblasts, and epithelial cells. Here, we report that postnatal lung alveolization critically requires ephrinB2, a ligand for Eph receptor tyrosine kinases expressed by the microvasculature. Mice homozygous for the hypomorphic knockin allele ephrinB2DeltaV/DeltaV, encoding mutant ephrinB2 with a disrupted C-terminal PDZ interaction motif, show severe postnatal lung defects including an almost complete absence of lung alveoli and abnormal and disorganized elastic matrix. Lung alveolar formation is not sensitive to loss of ephrinB2 cytoplasmic tyrosine phosphorylation sites. Postnatal day 1 mutant lungs show extracellular matrix alterations without differences in proportions of major distal cell populations. We conclude that lung alveolar formation relies on endothelial ephrinB2 function.


Subject(s)
Ephrin-B2/genetics , Ephrin-B2/metabolism , Gene Expression Regulation, Developmental/physiology , Pulmonary Alveoli/growth & development , Pulmonary Alveoli/physiology , Actins/metabolism , Animals , Elasticity , Elastin/metabolism , Ephrin-B3/genetics , Ephrin-B3/metabolism , Extracellular Matrix/physiology , Extracellular Matrix/ultrastructure , Fibrillins , Ligands , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Microfilament Proteins/metabolism , Microscopy, Electron , Pulmonary Alveoli/abnormalities , Receptors, Eph Family/genetics , Receptors, Eph Family/metabolism , Respiratory Mucosa/abnormalities , Respiratory Mucosa/growth & development , Respiratory Mucosa/physiology
10.
J Neurosci ; 27(42): 11279-88, 2007 Oct 17.
Article in English | MEDLINE | ID: mdl-17942722

ABSTRACT

Long-lasting changes in synaptic function are thought to be the cellular basis for learning and memory and for activity-dependent plasticity during development. Long-term potentiation (LTP) and long-term depression (LTD) are two opposing forms of synaptic plasticity that help fine tune neural connections and possibly serve to store information in the brain. Eph receptor tyrosine kinases and their transmembrane ligands, the ephrinBs, have essential roles in certain forms of synaptic plasticity. At the CA3-CA1 hippocampal synapse, EphB2 and EphA4 receptors are critically involved in long-term plasticity independent of their cytoplasmic domains, suggesting that ephrinBs are the active signaling partners. In cell-based assays, ephrinB reverse signaling was previously shown to involve phosphotyrosine-dependent and postsynaptic density-95/Discs large/zona occludens-1 (PDZ) domain interaction-dependent pathways. Which reverse signaling mode is required at hippocampal synapses is unknown. To address this question, we used knock-in mice expressing mutant isoforms of ephrinB2 that are deficient in specific aspects of reverse signaling. Our analysis revealed that tyrosine phosphorylation sites in ephrinB2 are required to mediate normal hippocampal LTP, but not for LTD. Conversely, ephrinB2 lacking the C-terminal PDZ interaction site, but competent to undergo tyrosine phosphorylation, cannot mediate either form of long-term plasticity. Our results provide the first evidence for phosphotyrosine-dependent ephrinB reverse signaling in a neuronal network and for differential ephrinB2 reverse signaling in two forms of synaptic plasticity.


Subject(s)
Hippocampus/metabolism , Long-Term Potentiation/physiology , Long-Term Synaptic Depression/physiology , Receptor, EphB2/metabolism , Tyrosine/metabolism , Animals , Binding Sites/physiology , Cells, Cultured , HeLa Cells , Humans , Mice , Mice, Knockout , Neuronal Plasticity/physiology , Phosphorylation , Receptor, EphB2/genetics , Receptor, EphB2/physiology , Tyrosine/genetics
11.
Genes Dev ; 19(3): 397-410, 2005 Feb 01.
Article in English | MEDLINE | ID: mdl-15687262

ABSTRACT

The transmembrane ligand ephrinB2 and its cognate Eph receptor tyrosine kinases are important regulators of embryonic blood vascular morphogenesis. However, the molecular mechanisms required for ephrinB2 transduced cellular signaling in vivo have not been characterized. To address this question, we generated two sets of knock-in mice: ephrinB2DeltaV mice expressed ephrinB2 lacking the C-terminal PDZ interaction site, and ephrinB2(5F) mice expressed ephrinB2 in which the five conserved tyrosine residues were replaced by phenylalanine to disrupt phosphotyrosine-dependent signaling events. Our analysis revealed that the homozygous mutant mice survived the requirement of ephrinB2 in embryonic blood vascular remodeling. However, ephrinB2DeltaV/DeltaV mice exhibited major lymphatic defects, including a failure to remodel their primary lymphatic capillary plexus into a hierarchical vessel network, hyperplasia, and lack of luminal valve formation. Unexpectedly, ephrinB2(5F/5F) mice displayed only a mild lymphatic phenotype. Our studies define ephrinB2 as an essential regulator of lymphatic development and indicate that interactions with PDZ domain effectors are required to mediate its functions.


Subject(s)
Ephrin-B2/metabolism , Lymphatic Vessels/metabolism , Amino Acid Sequence , Animals , Binding Sites , Ephrin-B2/genetics , Lymphatic Vessels/pathology , Mice , Mice, Transgenic , Molecular Sequence Data , Mutation , Phosphorylation , Protein Structure, Tertiary , Receptor, EphB4/genetics , Receptor, EphB4/metabolism , Skin/pathology , Tyrosine/genetics , Tyrosine/metabolism
12.
Pediatr Res ; 57(4): 537-44, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15718372

ABSTRACT

Vascular malformations cause discomfort and pain in children and are often associated with skeletal hypertrophy. Their molecular basis is poorly understood. Ephrin ligands and Eph receptor tyrosine kinases are involved in embryonic vascular development. In mice, some ephrin/Eph family members show a complementary expression pattern in blood vessels, with ephrinB2 being expressed on arterial and EphB4 on venous endothelium. Targeted deletions of the genes reveal their essential roles for conduit vessel development in mice, suggesting similar functions during human vascular development and deregulation in vascular malformations. Here, we have defined the expression patterns of human ephrinB2, EphB4, and EphB2 in normal vessels of neonates (i.e. umbilici) and adults and compared them with those in congenital venous malformations. In adults, normal vessels of the skin, muscle, and legs express ephrinB2 and EphB2 on arterial endothelial cells (ECs), whereas EphB4 is found in arteries and veins. In the umbilicus, EphB2 is a specific marker of arterial ECs, whereas ephrinB2 is additionally expressed in venous ECs, suggesting an arterial function of the veins. In venous malformations, the expression of EphB4 is not altered, but both ephrinB2 and EphB2 are ectopically expressed in venous ECs. This may reflect a nonphysiologic arterialization of malformed veins. Our study shows that the arterial markers ephrin B2 and EphB2 are expressed in a subset of veins, and it remains to be studied whether this is cause or consequence of an altered vascular identity.


Subject(s)
Arteriovenous Malformations/metabolism , Blood Vessels/metabolism , Ephrin-B2/metabolism , Receptor, EphB2/metabolism , Receptor, EphB4/metabolism , Umbilical Cord/blood supply , Adult , Animals , Arteriovenous Malformations/pathology , Biomarkers , Blood Vessels/abnormalities , Child , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Humans , Immunohistochemistry , Infant , Infant, Newborn , Mice , Umbilical Cord/anatomy & histology
13.
Development ; 129(15): 3739-50, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12117822

ABSTRACT

This study demonstrates that innervation dependent on two different neurotrophin tyrosine kinase (trk) receptors can form the same types of sensory endings (Merkel endings) in the same target (Merkel cells of vibrissa follicles). Some endings transiently express trkA during their initial development, whereas others express trkC throughout their development. Consequently, elimination of kinase domains of either trkA or trkC each result in a partial loss of Merkel endings, whereas absence of kinase domains of both receptors results in a total loss. At the onset of Merkel ending development, at least one kinase-lacking trkC isoform is transiently expressed on all the follicle cells, while neurotrophin 3 is transiently expressed only in the cells at the middle third of the follicle where the Merkel endings and cells develop. This transient non-neuronal expression of truncated trkC is essential for development of any Merkel endings, whereas some Merkel endings and cells still begin to develop in the absence of neurotrophin 3. Therefore, truncated trkC plays a more important role in the development of this innervation than kinase forms of trkA or trkC or of NT3, the only known ligand for trkC receptors.


Subject(s)
Merkel Cells/physiology , Receptor, trkA/metabolism , Receptor, trkC/metabolism , Animals , Embryo, Mammalian/anatomy & histology , Embryo, Mammalian/physiology , Female , Genes, Reporter , In Situ Hybridization , Male , Merkel Cells/cytology , Mice , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/metabolism , Models, Biological , Plant Proteins/genetics , Plant Proteins/metabolism , Pregnancy , Protein Isoforms , Rats , Rats, Sprague-Dawley , Receptor, Nerve Growth Factor , Receptor, trkA/genetics , Receptor, trkC/genetics , Receptors, Nerve Growth Factor/metabolism , Recombinant Fusion Proteins , Vibrissae/cytology , Vibrissae/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL