Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Cell Biol ; 183(5): 893-908, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19047464

ABSTRACT

Localization of presynaptic components to synaptic sites is critical for hippocampal synapse formation. Cell adhesion-regulated signaling is important for synaptic development and function, but little is known about differentiation of the presynaptic compartment. In this study, we describe a pathway that promotes presynaptic development involving p120catenin (p120ctn), the cytoplasmic tyrosine kinase Fer, the protein phosphatase SHP-2, and beta-catenin. Presynaptic Fer depletion prevents localization of active zone constituents and synaptic vesicles and inhibits excitatory synapse formation and synaptic transmission. Depletion of p120ctn or SHP-2 similarly disrupts synaptic vesicle localization with active SHP-2, restoring synapse formation in the absence of Fer. Fer or SHP-2 depletion results in elevated tyrosine phosphorylation of beta-catenin. beta-Catenin overexpression restores normal synaptic vesicle localization in the absence of Fer or SHP-2. Our results indicate that a presynaptic signaling pathway through p120ctn, Fer, SHP-2, and beta-catenin promotes excitatory synapse development and function.


Subject(s)
Cell Adhesion Molecules/metabolism , Hippocampus/enzymology , Neurons/enzymology , Phosphoproteins/metabolism , Presynaptic Terminals/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein-Tyrosine Kinases/metabolism , Synaptic Transmission , beta Catenin/metabolism , Animals , Axons/enzymology , Catenins , Cell Adhesion Molecules/genetics , Cells, Cultured , Cytoplasm/enzymology , Excitatory Postsynaptic Potentials , Hippocampus/embryology , Phosphoproteins/genetics , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Protein-Tyrosine Kinases/genetics , RNA Interference , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Time Factors , Transfection , beta Catenin/genetics , rhoA GTP-Binding Protein/metabolism , Delta Catenin
2.
Am J Hum Genet ; 83(6): 703-13, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19012874

ABSTRACT

Cell-adhesion molecules play critical roles in brain development, as well as maintaining synaptic structure, function, and plasticity. Here we have found the disruption of two genes encoding putative cell-adhesion molecules, CDH15 (cadherin superfamily) and KIRREL3 (immunoglobulin superfamily), by a chromosomal translocation t(11;16) in a female patient with intellectual disability (ID). We screened coding regions of these two genes in a cohort of patients with ID and controls and identified four nonsynonymous CDH15 variants and three nonsynonymous KIRREL3 variants that appear rare and unique to ID. These variations altered highly conserved residues and were absent in more than 600 unrelated patients with ID and 800 control individuals. Furthermore, in vivo expression studies showed that three of the CDH15 variations adversely altered its ability to mediate cell-cell adhesion. We also show that in neuronal cells, human KIRREL3 colocalizes and interacts with the synaptic scaffolding protein, CASK, recently implicated in X-linked brain malformation and ID. Taken together, our data suggest that alterations in CDH15 and KIRREL3, either alone or in combination with other factors, could play a role in phenotypic expression of ID in some patients.


Subject(s)
Cadherins/genetics , Carrier Proteins/genetics , Cell Adhesion Molecules, Neuronal/genetics , Genetic Variation , Intellectual Disability/genetics , Membrane Proteins/genetics , Cadherins/chemistry , Cadherins/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Case-Control Studies , Cell Adhesion , Cell Adhesion Molecules, Neuronal/chemistry , Cell Adhesion Molecules, Neuronal/metabolism , Chromosomes, Human, Pair 11 , Chromosomes, Human, Pair 16 , Cohort Studies , Female , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Middle Aged , Models, Biological , Protein Structure, Tertiary , Translocation, Genetic
3.
Nat Cell Biol ; 9(8): 883-92, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17618275

ABSTRACT

Binding of the secreted axon guidance cue Slit to its Robo receptor results in inactivation of the neural, calcium-dependent cell-cell adhesion molecule N-cadherin, providing a rapid epigenetic mechanism for integrating guidance and adhesion information. This requires the formation of a multimolecular complex containing Robo, Abl tyrosine kinase and N-cadherin. Here we show that on binding of Slit to Robo, the adaptor protein Cables is recruited to Robo-associated Abl and forms a multimeric complex by binding directly to N-cadherin-associated beta-catenin. Complex formation results in Abl-mediated phosphorylation of beta-catenin on tyrosine 489, leading to a decrease in its affinity for N-cadherin, loss of N-cadherin function, and targeting of phospho-Y489-beta-catenin to the nucleus. Nuclear beta-catenin combines with the transcription factor Tcf/Lef and activates transcription. Thus, Slit-induced formation of the Robo-N-cadherin complex results in a rapid loss of cadherin-mediated adhesion and has more lasting effects on gene transcription.


Subject(s)
Cadherins/metabolism , Carrier Proteins/metabolism , Cell Adhesion/physiology , Cyclins/metabolism , Drosophila Proteins/metabolism , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Receptors, Immunologic/metabolism , beta Catenin/metabolism , Animals , Cadherins/genetics , Carrier Proteins/genetics , Cell Nucleus/metabolism , Cells, Cultured , Chick Embryo , Cyclin-Dependent Kinase 5/genetics , Cyclin-Dependent Kinase 5/metabolism , Cyclins/genetics , Drosophila Proteins/genetics , Drosophila melanogaster , Humans , Multiprotein Complexes , Nerve Tissue Proteins/genetics , Phosphoproteins/genetics , Phosphoproteins/metabolism , Protein Binding , Protein Subunits/genetics , Protein Subunits/metabolism , Proto-Oncogene Proteins c-abl/genetics , Receptors, Immunologic/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retina/cytology , Transcription, Genetic , beta Catenin/genetics , Roundabout Proteins
4.
J Cell Biol ; 177(4): 707-16, 2007 May 21.
Article in English | MEDLINE | ID: mdl-17502419

ABSTRACT

Point mutations in the cytoplasmic domain of myelin protein zero (P0; the major myelin protein in the peripheral nervous system) that alter a protein kinase Calpha (PKCalpha) substrate motif (198HRSTK201) or alter serines 199 and/or 204 eliminate P0-mediated adhesion. Mutation in the PKCalpha substrate motif (R198S) also causes a form of inherited peripheral neuropathy (Charcot Marie Tooth disease [CMT] 1B), indicating that PKCalpha-mediated phosphorylation of P0 is important for myelination. We have now identified a 65-kD adaptor protein that links P0 with the receptor for activated C kinase 1 (RACK1). The interaction of p65 with P0 maps to residues 179-197 within the cytoplasmic tail of P0. Mutations or deletions that abolish p65 binding reduce P0 phosphorylation and adhesion, which can be rescued by the substitution of serines 199 and 204 with glutamic acid. A mutation in the p65-binding sequence G184R occurs in two families with CMT, and mutation of this residue results in the loss of both p65 binding and adhesion function.


Subject(s)
GTP-Binding Proteins/physiology , Myelin P0 Protein/metabolism , Neoplasm Proteins/physiology , Protein Kinase C-alpha/physiology , Receptors, Cell Surface/physiology , Adaptor Proteins, Signal Transducing/physiology , Amino Acid Sequence , Animals , Humans , L Cells , Mice , Molecular Sequence Data , Myelin P0 Protein/physiology , Phosphorylation , Rats , Receptors for Activated C Kinase , Two-Hybrid System Techniques
5.
Mol Cell Neurosci ; 32(3): 230-41, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16757177

ABSTRACT

The architecture of dendritic arbors is a defining characteristic of neurons and is established through a sequential but overlapping series of events involving process outgrowth and branching, stabilization of the global pattern, and synapse formation. To investigate the roles of cadherins and beta1-integrins in maintaining the global architecture of the arbor, we used membrane permeable peptides and transfection with dominant-negative constructs to disrupt adhesion molecule function in intact chick neural retina at a stage when the architecture of the ganglion cell (RGC) arbor is established but synapse formation is just beginning. Inactivation of beta1-integrins induces rapid dendrite retraction, with loss of dynamic terminal filopodia followed by resorption of major branches. Disruption of N-cadherin-beta-catenin interactions has no effect; however, dendrites do retract following perturbation of the juxtamembrane region of N-cadherin, which disrupts N-cadherin-mediated adhesion and initiates a beta1-integrin inactivating signal. Thus, developing RGC dendritic arbors are stabilized by beta1-integrin-dependent processes.


Subject(s)
Cell Differentiation/physiology , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Integrin beta1/physiology , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Amino Acid Sequence , Animals , Cells, Cultured , Chick Embryo , Dendrites/genetics , Dendrites/metabolism , Dendritic Spines/genetics , Integrin beta1/genetics , Molecular Sequence Data , Tissue Culture Techniques
6.
J Cell Sci ; 119(Pt 7): 1233-43, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16522684

ABSTRACT

Here, we define the mechanism through which protein tyrosine phosphatase 1B (PTP1B) is targeted to cell-matrix adhesion sites. Green fluorescent protein (GFP)-labeled PTP1B bearing the substrate-trapping mutation D181A was found in punctate structures in lamellae. The puncta co-localized with focal adhesion kinase (FAK) and Src, and defined the distal tips of cell-matrix adhesion sites identified with paxillin and vinculin. PTP1B is largely associated with the external face of the endoplasmic reticulum (ER) and the puncta develop from ER projections over cell-matrix adhesion sites, a process dependent on microtubules. Deletion of the ER-targeting sequence resulted in cytosolic localization and altered the distribution of PTP1B at cell-matrix foci, whereas mutations disrupting interactions with Src homology 3 (SH3) domains, and the insulin and cadherin receptors had no effect. PTP1B recognizes substrates within forming adhesion foci as revealed by its preferential association with paxillin as opposed to zyxin-containing foci. Our results suggest that PTP1B targets to immature cell-matrix foci in newly forming lamellae by dynamic extensions of the ER and contributes to the maturation of these sites.


Subject(s)
Cell-Matrix Junctions/metabolism , Endoplasmic Reticulum/metabolism , Gene Targeting , Protein Tyrosine Phosphatases/metabolism , Animals , Blotting, Western , Cells, Cultured , Fibroblasts/metabolism , Fluorescent Antibody Technique, Direct , Gene Deletion , Mice , Mice, Knockout , Microscopy, Video , Paxillin/metabolism , Precipitin Tests , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/genetics , Substrate Specificity
7.
Curr Opin Cell Biol ; 17(5): 459-65, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16099633

ABSTRACT

The formation of stable cell-cell adhesions by type I cadherins depends on the association of their cytoplasmic domain with beta-catenin, and of beta-catenin with alpha-catenin. The binding of beta-catenin to these partners is regulated by phosphorylation of at least three critical tyrosine residues. Each of these residues is targeted by one or more specific kinases: Y142 by Fyn, Fer and cMet; Y489 by Abl; and Y654 by Src and the epidermal growth factor receptor. Developmental and physiological signals have been identified that initiate the specific phosphorylation and dephosphorylation of these residues, regulating cadherin function during neurite outgrowth, permeability of airway epithelium and synapse remodeling, and possibly initiating epithelial cell migration during development and metastasis.


Subject(s)
Cadherins/metabolism , Cell Adhesion/physiology , Tyrosine/metabolism , beta Catenin/metabolism , Animals , Cadherins/pharmacology , Cadherins/physiology , Cell Adhesion/drug effects , Models, Molecular , Phosphorylation , Protein Structure, Tertiary , Protein Tyrosine Phosphatases/metabolism , Transcription Factors/metabolism , Transcriptional Activation/physiology , Tyrosine/pharmacology
8.
J Cell Sci ; 117(Pt 15): 3207-19, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15226396

ABSTRACT

The function of Type 1, classic cadherins depends on their association with the actin cytoskeleton, a connection mediated by alpha- and beta-catenin. The phosphorylation state of beta-catenin is crucial for its association with cadherin and thus the association of cadherin with the cytoskeleton. We now show that the phosphorylation of beta-catenin is regulated by the combined activities of the tyrosine kinase Fer and the tyrosine phosphatase PTP1B. Fer phosphorylates PTP1B at tyrosine 152, regulating its binding to cadherin and the continuous dephosphorylation of beta-catenin at tyrosine 654. Fer interacts with cadherin indirectly, through p120ctn. We have mapped the interaction domains of Fer and p120ctn and peptides corresponding to these sequences release Fer from p120ctn in vitro and in live cells, resulting in loss of cadherin-associated PTP1B, an increase in the pool of tyrosine phosphorylated beta-catenin and loss of cadherin adhesion function. The effect of the peptides is lost when a beta-catenin mutant with a substitution at tyrosine 654 is introduced into cells. Thus, Fer phosphorylates PTP1B at tyrosine 152 enabling it to bind to the cytoplasmic domain of cadherin, where it maintains beta-catenin in a dephosphorylated state. Cultured fibroblasts from mouse embryos targeted with a kinase-inactivating ferD743R mutation have lost cadherin-associated PTP1B and beta-catenin, as well as localization of cadherin and beta-catenin in areas of cell-cell contacts. Expression of wild-type Fer or culture in epidermal growth factor restores the cadherin complex and localization at cell-cell contacts.


Subject(s)
Cadherins/metabolism , Cytoskeletal Proteins/metabolism , Proto-Oncogene Proteins/physiology , Trans-Activators/metabolism , Animals , Antennapedia Homeodomain Protein , Blotting, Western , Brain/embryology , Catenins , Cell Adhesion , Cell Adhesion Molecules/metabolism , Cell Communication , Cell Membrane/metabolism , Cytoskeleton/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Fibroblasts/metabolism , Glutathione Transferase/metabolism , Homeodomain Proteins/metabolism , Immunohistochemistry , Immunoprecipitation , Mice , Mutation , Neurons/metabolism , Nuclear Proteins/metabolism , Peptides/chemistry , Phenotype , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/metabolism , Protein-Tyrosine Kinases , Proto-Oncogene Proteins/metabolism , Retina/embryology , Time Factors , Transcription Factors/metabolism , Tyrosine/chemistry , Tyrosine/metabolism , beta Catenin , Delta Catenin
9.
Brain ; 127(Pt 2): 371-84, 2004 Feb.
Article in English | MEDLINE | ID: mdl-14711881

ABSTRACT

Myelin protein zero (MPZ) is a member of the immunoglobulin gene superfamily with single extracellular, transmembrane and cytoplasmic domains. Homotypic interactions between extracellular domains of MPZ adhere adjacent myelin wraps to each other. MPZ is also necessary for myelin compaction since mice which lack MPZ develop severe dysmyelinating neuropathies in which compaction is dramatically disrupted. MPZ mutations in humans cause the inherited demyelinating neuropathy CMT1B. Some mutations cause the severe neuropathies of infancy designated as Dejerine-Sottas disease, while others cause a 'classical' Charcot-Marie-Tooth (CMT) disease Type 1B (CMT1B) phenotype with normal early milestones but development of disability during the first two decades of life. Still other mutations cause a neuropathy that presents in adults, with normal nerve conduction velocities, designated as a 'CMT2' form of CMT1B. To correlate the phenotype of patients with MPZ mutations with their genotype, we identified and evaluated 13 patients from 12 different families with eight different MPZ mutations. In addition, we re-analysed the clinical data from 64 cases of CMT1B from the literature. Contrary to our expectations, we found that most patients presented with either an early onset neuropathy with signs and symptoms prior to the onset of walking or a late onset neuropathy with signs and symptoms at around age 40 years. Only occasional patients presented with a 'classical' CMT phenotype. Correlation of specific MPZ mutations with their phenotypes demonstrated that addition of either a charged amino acid or altering a cysteine residue in the extracellular domain caused a severe early onset neuropathy. Severe neuropathy was also caused by truncation of the cytoplasmic domain or alteration of an evolutionarily conserved amino acid. Taken together, these data suggest that early onset neuropathy is caused by MPZ mutations that significantly disrupt the tertiary structure of MPZ and thus interfere with MPZ-mediated adhesion and myelin compaction. In contrast, late onset neuropathy is caused by mutations that more subtly alter myelin structure and which probably disrupt Schwann cell-axonal interactions.


Subject(s)
Demyelinating Diseases/genetics , Hereditary Sensory and Motor Neuropathy/genetics , Mutation , Myelin P0 Protein/genetics , Adult , Age of Onset , Aged , Amino Acid Sequence , Child, Preschool , Demyelinating Diseases/physiopathology , Female , Hereditary Sensory and Motor Neuropathy/physiopathology , Humans , Male , Middle Aged , Molecular Sequence Data , Mutation, Missense , Neural Conduction , Open Reading Frames/genetics , Phenotype , Protein Structure, Secondary/genetics
10.
11.
J Biol Chem ; 277(51): 49989-97, 2002 Dec 20.
Article in English | MEDLINE | ID: mdl-12377785

ABSTRACT

The nonreceptor tyrosine phosphatase PTP1B associates with the cytoplasmic domain of N-cadherin and may regulate cadherin function through dephosphorylation of beta-catenin. We have now identified the domain on N-cadherin to which PTP1B binds and characterized the effect of perturbing this domain on cadherin function. Deletion constructs lacking amino acids 872-891 fail to bind PTP1B. This domain partially overlaps with the beta-catenin binding domain. To further define the relationship of these two sites, we used peptides to compete in vitro binding. A peptide representing the most NH(2)-terminal 8 amino acids of the PTP1B binding site, the region of overlap with the beta-catenin target, effectively competes for binding of beta-catenin but is much less effective in competing PTP1B, whereas two peptides representing the remaining 12 amino acids have no effect on beta-catenin binding but effectively compete for PTP1B binding. Introduction into embryonic chick retina cells of a cell-permeable peptide mimicking the 8 most COOH-terminal amino acids in the PTP1B target domain, the region most distant from the beta-catenin target site, prevents binding of PTP1B, increases the pool of free, tyrosine-phosphorylated beta-catenin, and results in loss of N-cadherin function. N-cadherin lacking this same region of the PTP1B target site does not associate with PTP1B or beta-catenin and is not efficiently expressed at the cell surface of transfected L cells. Thus, interaction of PTP1B with N-cadherin is essential for its association with beta-catenin, stable expression at the cell surface, and consequently, cadherin function.


Subject(s)
Cadherins/physiology , Cytoskeletal Proteins/metabolism , Protein Tyrosine Phosphatases/physiology , Trans-Activators/metabolism , Amino Acid Sequence , Animals , Binding Sites , Binding, Competitive , Cadherins/metabolism , Cell Membrane/metabolism , Chick Embryo , Cytoplasm/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Gene Deletion , Glutathione Transferase/metabolism , Green Fluorescent Proteins , Immunoblotting , Luminescent Proteins/metabolism , Microscopy, Confocal , Molecular Sequence Data , Peptides/chemistry , Phosphorylation , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Protein Tyrosine Phosphatases/metabolism , Recombinant Fusion Proteins/metabolism , Retina/embryology , Serine/metabolism , Time Factors , Transfection , Tyrosine/metabolism , beta Catenin
12.
Nat Cell Biol ; 4(10): 798-805, 2002 Oct.
Article in English | MEDLINE | ID: mdl-12360290

ABSTRACT

The formation of axon trajectories requires integration of local adhesive interactions with directional information from attractive and repulsive cues. Here, we show that these two types of information are functionally integrated; activation of the transmembrane receptor Roundabout (Robo) by its ligand, the secreted repulsive guidance cue Slit, inactivates N-cadherin-mediated adhesion. Loss of N-cadherin-mediated adhesion is accompanied by tyrosine phosphorylation of beta-catenin and its loss from the N-cadherin complex, concomitant with the formation of a supramolecular complex containing Robo, Abelson (Abl) kinase and N-cadherin. Local formation of such a receptor complex is an ideal mechanism to steer the growth cone while still allowing adhesion and growth in other directions.


Subject(s)
Cadherins/metabolism , Cell Adhesion/physiology , Cell Membrane/metabolism , Central Nervous System/metabolism , Growth Cones/metabolism , Receptors, Immunologic/metabolism , Animals , Cadherins/genetics , Cell Communication/physiology , Cell Differentiation/physiology , Cells, Cultured , Central Nervous System/embryology , Chick Embryo , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/metabolism , Fibroblasts , Glycoproteins/deficiency , Glycoproteins/genetics , Macromolecular Substances , Mice , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Peptide Fragments , Protein Structure, Tertiary/physiology , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-abl/metabolism , Receptors, Immunologic/genetics , Retina , Trans-Activators/genetics , Trans-Activators/metabolism , beta Catenin , Roundabout Proteins
13.
Dev Dyn ; 225(1): 1-13, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12203715

ABSTRACT

Previously, we demonstrated that chick embryos treated with antisense oligonucleotides against a striated muscle-specific Xin exhibit abnormal cardiac morphogenesis (Wang et al. [1999] Development 126:1281-1294); therefore, we surmised a role for Xin in cardiac development. Herein, we examine the developmental expression of Xin through immunofluorescent staining of whole-mount mouse embryos and frozen heart sections. Xin expression is first observed within the heart tube of embryonic day 8.0 (E8.0) mice, exhibiting a peripheral localization within the cardiomyocytes. Colocalization of Xin with both beta-catenin and N-cadherin is observed throughout embryogenesis and into adulthood. Additionally, Xin is found associated with beta-catenin within the N-cadherin complex in embryonic chick hearts by coimmunoprecipitation. Xin is detected earlier than vinculin in the developing heart and colocalizes with vinculin at the intercalated disc but not at the sarcolemma within embryonic and postnatal hearts. At E10.0, Xin is also detected in the developing somites and later in the myotendon junction of skeletal muscle but not within the costameric regions of muscle. In cultured C2C12 myotubes, the Xin protein is found in many speckled and filamentous structures, coincident with tropomyosin in the stress fibers. Additionally, Xin is enriched in the regions of cell-cell contacts. These data demonstrate that Xin is one of the components at the adherens junction of cardiac muscle, and its counterpart in skeletal muscle, the myotendon junction. Furthermore, temporal and spatial expressions of Xin in relation to intercalated disc proteins and thin filament proteins suggest roles for Xin in the formation of cell-cell contacts and possibly in myofibrillogenesis.


Subject(s)
DNA-Binding Proteins/biosynthesis , Heart/embryology , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Myocardium/metabolism , Nuclear Proteins/biosynthesis , Animals , Blotting, Western , Cadherins/metabolism , Cell Nucleus/metabolism , Chick Embryo , Cytoskeletal Proteins/metabolism , Mice , Microscopy, Fluorescence , Precipitin Tests , Protein Binding , Time Factors , Trans-Activators/metabolism , Transfection , beta Catenin
14.
Dev Dyn ; 224(1): 18-29, 2002 May.
Article in English | MEDLINE | ID: mdl-11984870

ABSTRACT

The classic cadherins are a group of calcium dependent, homophilic cell-cell adhesion molecules that drive morphogenetic rearrangements and maintain the integrity of cell groups through the formation of adherens junctions. The formation and maintenance of cadherin-mediated adhesions is a multistep process and mechanisms have evolved to regulate each step. This suggests that functional state switching plays an important role in development. Among the many challenges ahead is to determine the developmental role that functional state switching plays in tissue morphogenesis and to define the roles of each of the several regulatory interactions that participate in switching. One correlate of the loss of cadherin-mediated adhesion, the "turn-off" of cadherin function, is the exit, or "drop-out" of cells from neural and epithelial layers and their conversion to a motile phenotype. We suggest that epithelial mesenchymal conversions may be initiated by signaling pathways that result in the loss of cadherin function. Tyrosine phosphorylation of beta-catenin is one such mechanism. Enhanced phosphorylation of tyrosine residues on beta-catenin is almost invariably associated with loss of the cadherin-actin connection concomitant with loss of adhesive function. There are several tyrosine kinases and phosphatases that have been shown to have the potential to alter the phosphorylation state of beta-catenin and thus the function of cadherins. Our laboratory has focused on the role of the nonreceptor tyrosine phosphatase PTP1B in regulating the phosphorylation of beta-catenin on tyrosine residues. Our data suggest that PTP1B is crucial for maintenance of N-cadherin-mediated adhesions in embryonic neural retina cells. By using an L-cell model system constitutively expressing N-cadherin, we have worked out many of the molecular interactions essential for this regulatory interaction. Extracellular cues that bias this critical regulatory interaction toward increased phosphorylation of beta-catenin may be a critical component of many developmental events.


Subject(s)
Cadherins/metabolism , Protein Tyrosine Phosphatases/metabolism , Amino Acid Sequence , Animals , Cadherins/chemistry , Cadherins/genetics , Cytoskeletal Proteins/metabolism , Humans , Intercellular Junctions/physiology , Mesoderm/physiology , Models, Biological , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Trans-Activators/metabolism , beta Catenin
SELECTION OF CITATIONS
SEARCH DETAIL
...