Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 9(1): 11945, 2019 08 16.
Article in English | MEDLINE | ID: mdl-31420572

ABSTRACT

The signalling output of many transmembrane receptors that mediate cell-cell communication is restricted by the endosomal sorting complex required for transport (ESCRT), but the impact of this machinery on Eph tyrosine kinase receptor function is unknown. We identified the ESCRT-associated adaptor protein HD-PTP as part of an EphB2 proximity-dependent biotin identification (BioID) interactome, and confirmed this association using co-immunoprecipitation. HD-PTP loss attenuates the ephrin-B2:EphB2 signalling-induced collapse of cultured cells and axonal growth cones, and results in aberrant guidance of chick spinal motor neuron axons in vivo. HD-PTP depletion abrogates ephrin-B2-induced EphB2 clustering, and EphB2 and Src family kinase activation. HD-PTP loss also accelerates ligand-induced EphB2 degradation, contrasting the effects of HD-PTP loss on the relay of signals from other cell surface receptors. Our results link Eph function to the ESCRT machinery and demonstrate a role for HD-PTP in the earliest steps of ephrin-B:EphB signalling, as well as in obstructing premature receptor depletion.


Subject(s)
Axons/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomes/metabolism , Ephrin-B2/genetics , Motor Neurons/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/genetics , Receptor, EphB2/genetics , Animals , Axons/ultrastructure , Chick Embryo , Endosomal Sorting Complexes Required for Transport/genetics , Endosomes/ultrastructure , Ephrin-B2/metabolism , Gene Expression Regulation , HEK293 Cells , HeLa Cells , Humans , Motor Neurons/ultrastructure , Primary Cell Culture , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , Proteolysis , Receptor, EphB2/metabolism , Signal Transduction , Spinal Cord/metabolism , Spinal Cord/ultrastructure , src-Family Kinases/genetics , src-Family Kinases/metabolism
2.
Nat Commun ; 9(1): 4470, 2018 10 26.
Article in English | MEDLINE | ID: mdl-30367035

ABSTRACT

Myoblast fusion is tightly regulated during development and regeneration of muscle fibers. BAI3 is a receptor that orchestrates myoblast fusion via Elmo/Dock1 signaling, but the mechanisms regulating its activity remain elusive. Here we report that mice lacking BAI3 display small muscle fibers and inefficient muscle regeneration after cardiotoxin-induced injury. We describe two proteins that repress or activate BAI3 in muscle progenitors. We find that the secreted C1q-like1-4 proteins repress fusion by specifically interacting with BAI3. Using a proteomic approach, we identify Stabilin-2 as a protein that interacts with BAI3 and stimulates its fusion promoting activity. We demonstrate that Stabilin-2 activates the GPCR activity of BAI3. The resulting activated heterotrimeric G-proteins contribute to the initial recruitment of Elmo proteins to the membrane, which are then stabilized on BAI3 through a direct interaction. Collectively, our results demonstrate that the activity of BAI3 is spatiotemporally regulated by C1qL4 and Stabilin-2 during myoblast fusion.


Subject(s)
Cell Adhesion Molecules, Neuronal/metabolism , Complement C1q/metabolism , Membrane Proteins/metabolism , Muscle Fibers, Skeletal/physiology , Myoblasts, Skeletal/physiology , Nerve Tissue Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Regeneration/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Fusion , Cell Membrane/metabolism , Cells, Cultured , Complement C1q/genetics , Cytoskeletal Proteins/metabolism , Gene Expression , Gene Silencing , Membrane Proteins/deficiency , Mice , Mice, Knockout , Models, Biological , Muscle Development/physiology , Muscle Fibers, Skeletal/cytology , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/metabolism , Nerve Tissue Proteins/deficiency , Signal Transduction
3.
J Biol Chem ; 292(29): 12178-12191, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28600358

ABSTRACT

Developmental angiogenesis and the maintenance of the blood-brain barrier involve endothelial cell adhesion, which is linked to cytoskeletal dynamics. GPR124 (also known as TEM5/ADGRA2) is an adhesion G protein-coupled receptor family member that plays a pivotal role in brain angiogenesis and in ensuring a tight blood-brain barrier. However, the signaling properties of GPR124 remain poorly defined. Here, we show that ectopic expression of GPR124 promotes cell adhesion, additive to extracellular matrix-dependent effect, coupled with filopodia and lamellipodia formation and an enrichment of a pool of the G protein-coupled receptor at actin-rich cellular protrusions containing VASP, a filopodial marker. Accordingly, GPR124-expressing cells also displayed increased activation of both Rac and Cdc42 GTPases. Mechanistically, we uncover novel direct interactions between endogenous GPR124 and the Rho guanine nucleotide exchange factors Elmo/Dock and intersectin (ITSN). Small fragments of either Elmo or ITSN1 that bind GPR124 blocked GPR124-induced cell adhesion. In addition, Gßγ interacts with the C-terminal tail of GPR124 and promotes the formation of a GPR124-Elmo complex. Furthermore, GPR124 also promotes the activation of the Elmo-Dock complex, as measured by Elmo phosphorylation on a conserved C-terminal tyrosine residue. Interestingly, Elmo and ITSN1 also interact with each other independently of their GPR124-recognition regions. Moreover, endogenous phospho-Elmo and ITSN1 co-localize with GPR124 at lamellipodia of adhering endothelial cells, where GPR124 expression contributes to polarity acquisition during wound healing. Collectively, our results indicate that GPR124 promotes cell adhesion via Elmo-Dock and ITSN. This constitutes a previously unrecognized complex formed of atypical and conventional Rho guanine nucleotide exchange factors for Rac and Cdc42 that is putatively involved in GPR124-dependent angiogenic responses.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Endothelium, Vascular/metabolism , Protein Processing, Post-Translational , Receptors, G-Protein-Coupled/metabolism , rac GTP-Binding Proteins/metabolism , Adaptor Proteins, Signal Transducing/chemistry , Adaptor Proteins, Vesicular Transport/chemistry , Animals , COS Cells , Cell Adhesion , Cells, Cultured , Chlorocebus aethiops , Endothelium, Vascular/cytology , HEK293 Cells , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Phosphorylation , Protein Interaction Domains and Motifs , Protein Multimerization , Protein Transport , Pseudopodia/metabolism , RNA Interference , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , rac GTP-Binding Proteins/chemistry
4.
Proc Natl Acad Sci U S A ; 111(10): 3745-50, 2014 Mar 11.
Article in English | MEDLINE | ID: mdl-24567399

ABSTRACT

Muscle fibers form as a result of myoblast fusion, yet the cell surface receptors regulating this process are unknown in vertebrates. In Drosophila, myoblast fusion involves the activation of the Rac pathway by the guanine nucleotide exchange factor Myoblast City and its scaffolding protein ELMO, downstream of cell-surface cell-adhesion receptors. We previously showed that the mammalian ortholog of Myoblast City, DOCK1, functions in an evolutionarily conserved manner to promote myoblast fusion in mice. In search for regulators of myoblast fusion, we identified the G-protein coupled receptor brain-specific angiogenesis inhibitor (BAI3) as a cell surface protein that interacts with ELMO. In cultured cells, BAI3 or ELMO1/2 loss of function severely impaired myoblast fusion without affecting differentiation and cannot be rescued by reexpression of BAI3 mutants deficient in ELMO binding. The related BAI protein family member, BAI1, is functionally distinct from BAI3, because it cannot rescue the myoblast fusion defects caused by the loss of BAI3 function. Finally, embryonic muscle precursor expression of a BAI3 mutant unable to bind ELMO was sufficient to block myoblast fusion in vivo. Collectively, our findings provide a role for BAI3 in the relay of extracellular fusion signals to their intracellular effectors, identifying it as an essential transmembrane protein for embryonic vertebrate myoblast fusion.


Subject(s)
Muscle Fibers, Skeletal/physiology , Myoblasts/physiology , Nerve Tissue Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Cell Differentiation/physiology , Cell Fusion , Cell Line , Chick Embryo , Electroporation , Fluorescent Antibody Technique , Immunohistochemistry , In Situ Hybridization , Membrane Proteins , Mice , Muscle Fibers, Skeletal/cytology , Two-Hybrid System Techniques
SELECTION OF CITATIONS
SEARCH DETAIL
...