Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
1.
Br J Pharmacol ; 171(23): 5195-208, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24597515

ABSTRACT

BACKGROUND AND PURPOSE: The Eph receptor tyrosine kinases and their ephrin ligands are key players in tumorigenesis and many reports have correlated changes in their expression with a poor clinical prognosis in many solid tumours. Agents targeting the Eph-ephrin system might emerge as new tools useful for the inhibition of different components of cancer progression. Even if different classes of small molecules targeting Eph-ephrin interactions have been reported, their use is hampered by poor chemical stability and low potency. Stable and potent ligands are crucial to achieve robust pharmacological performance. EXPERIMENTAL APPROACH: UniPR129 (the L-homo-Trp conjugate of lithocholic acid) was designed by means of computational methods, synthetized and tested for its ability to inhibit the interaction between the EphA2 receptor and the ephrin-A1 ligand in an elisa binding study. The ability of UniPR129 to disrupt EphA2-ephrin-A1 interaction was functionally evaluated in a prostate adenocarcinoma cell line and its anti-angiogenic effect was tested in vitro using cultures of HUVECs. KEY RESULTS: UniPR129 disrupted EphA2-ephrin-A1 interaction with Ki = 370 nM in an elisa binding assay and with low micromolar potency in cellular functional assays, including inhibition of EphA2 activation, inhibition of PC3 cell rounding and disruption of in vitro angiogenesis, without cytotoxic effects. CONCLUSIONS AND IMPLICATIONS: The discovery of UniPR129 represents not only a major advance in potency compared with the existing Eph-ephrin antagonists but also an improvement in terms of cytotoxicity, making this molecule a useful pharmacological tool and a promising lead compound.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Ephrin-A1/antagonists & inhibitors , Lithocholic Acid/analogs & derivatives , Receptor, EphA2/antagonists & inhibitors , Tryptophan/analogs & derivatives , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Ephrin-A1/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Humans , Lithocholic Acid/pharmacology , Molecular Docking Simulation , Neovascularization, Physiologic/drug effects , Receptor, EphA2/metabolism , Receptor, EphB4/metabolism , Receptors, Vascular Endothelial Growth Factor/metabolism , Tryptophan/pharmacology
2.
Dev Biol ; 247(1): 89-101, 2002 Jul 01.
Article in English | MEDLINE | ID: mdl-12074554

ABSTRACT

Motor neurons in the ventral neural tube project axons specifically to their target muscles in the periphery. Although many of the transcription factors that specify motor neuron cell fates have been characterized, less is understood about the mechanisms that guide motor axons to their correct targets. We show that ectopic expression of EphA4 receptor tyrosine kinase alters the trajectories of a specific population of motor axons in the avian hindlimb. Most motor neurons in the medial portion of the lateral motor column (LMC) extend their axons aberrantly in the dorsal nerve trunk at the level of the crural plexus, in the presence of ectopic EphA4. This misrouting of motor axons is not accompanied by alterations in motor neuron identity, settling patterns in the neural tube, or the fasciculation of spinal nerves. However, ectopic EphA4 axons do make errors in pathway selection during sorting in the plexus at the base of the hindlimb. These results suggest that EphA4 in motor neurons acts as a population-specific guidance cue to control the dorsal trajectory of their axons in the hindlimb.


Subject(s)
Fetal Proteins/physiology , Hindlimb/physiology , Motor Neurons/physiology , Receptor Protein-Tyrosine Kinases/physiology , Animals , Axons/physiology , Cell Differentiation/physiology , Chick Embryo , Electroporation , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/physiology , Hindlimb/cytology , Hindlimb/embryology , Morphogenesis , Motor Neurons/cytology , Nerve Tissue Proteins/physiology , Receptor, EphA4
3.
Development ; 128(23): 4669-80, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11731448

ABSTRACT

Limb muscles derive from muscle precursor cells that lie initially in the lateral portion of the somitic dermomyotome and subsequently migrate to their target limb regions, where muscle-specific gene transcription is initiated. Although several molecules that control the generation and delamination of muscle precursor cells have been identified, little is known about the mechanisms that guide muscle precursor cell migration in the limb. We have examined the distribution of members of the Eph family during muscle precursor cell development. The EphA4 receptor tyrosine kinase and its ligand, ephrin-A5, are expressed by muscle precursor cells and forelimb mesoderm in unique spatiotemporal patterns during the period when muscle precursors delaminate from the dermomyotome and migrate into the limb. To test the function of EphA4/ephrin-A5 interactions in muscle precursor migration, we used targeted in ovo electroporation to express ephrin-A5 ectopically specifically in the presumptive limb mesoderm. In the presence of ectopic ephrin-A5, Pax7-positive muscle precursor cells are significantly reduced in number in the proximal limb, compared with controls, and congregate abnormally near the lateral dermomyotome. In stripe assays, isolated muscle precursor cells avoid substrate-bound ephrin-A5 and this avoidance is abolished by addition of soluble ephrin-A5. These data suggest that ephrin-A5 normally restricts migrating, EphA4-positive muscle precursor cells to their appropriate territories in the forelimb, disallowing entry into abnormal embryonic regions.


Subject(s)
Fetal Proteins/metabolism , Membrane Proteins/metabolism , Muscle, Skeletal/embryology , Muscle, Skeletal/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Cell Movement/physiology , Chick Embryo , Electroporation , Ephrin-A5 , Fetal Proteins/genetics , Forelimb , Gene Expression Regulation, Developmental , Green Fluorescent Proteins , Ligands , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Membrane Proteins/genetics , Mesoderm/cytology , Mesoderm/metabolism , Microscopy, Confocal , Muscle, Skeletal/cytology , Plasmids/administration & dosage , Plasmids/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor, EphA4 , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Stem Cells/cytology , Stem Cells/metabolism
4.
Neuron ; 31(6): 1001-13, 2001 Sep 27.
Article in English | MEDLINE | ID: mdl-11580899

ABSTRACT

We previously reported that the cell surface proteoglycan syndecan-2 can induce dendritic spine formation in hippocampal neurons. We demonstrate here that the EphB2 receptor tyrosine kinase phosphorylates syndecan-2 and that this phosphorylation event is crucial for syndecan-2 clustering and spine formation. Syndecan-2 is tyrosine phosphorylated and forms a complex with EphB2 in mouse brain. Dominant-negative inhibition of endogenous EphB receptor activities blocks clustering of endogenous syndecan-2 and normal spine formation in cultured hippocampal neurons. This is the first evidence that Eph receptors play a physiological role in dendritic spine morphogenesis. Our observations suggest that spine morphogenesis is triggered by the activation of Eph receptors, which causes tyrosine phosphorylation of target molecules, such as syndecan-2, in presumptive spines.


Subject(s)
Dendrites/metabolism , Excitatory Postsynaptic Potentials/physiology , Hippocampus/cytology , Membrane Glycoproteins/metabolism , Nerve Tissue Proteins/metabolism , Protein Processing, Post-Translational , Proteoglycans/metabolism , Receptor Protein-Tyrosine Kinases/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Dendrites/ultrastructure , Mice , Morphogenesis , Mutagenesis, Site-Directed , Neuronal Plasticity/physiology , Phosphorylation , Phosphotyrosine/analysis , Rats , Receptor, EphB2 , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Syndecan-2 , Transfection
5.
Oncogene ; 20(30): 3995-4006, 2001 Jul 05.
Article in English | MEDLINE | ID: mdl-11494128

ABSTRACT

The Eph family of receptor tyrosine kinases and the Abl family of non-receptor tyrosine kinases have both been implicated in tissue morphogenesis. They regulate the organization of the actin cytoskeleton in the developing nervous system and participate in signaling pathways involved in axon growth. Both Eph receptors and Abl are localized in the neuronal growth cone, suggesting that they play a role in axon pathfinding. Two-hybrid screens identified regions of Abl and Arg that bind to the EphB2 and EphA4 receptors, suggesting a novel signaling connection involving the two kinase families. The association of full-length Abl and Arg with EphB2 was confirmed by co-immunoprecipitation and found to involve several distinct protein interactions. The SH2 domains of Abl and Arg bind to tyrosine-phosphorylated motifs in the juxtamembrane region of EphB2. A second, phosphorylation-independent interaction with EphB2 involves non-conserved sequences in the C-terminal tails of Abl and Arg. A third interaction between Abl and EphB2 is probably mediated by an intermediary protein because it requires tyrosine phosphorylation of EphB2, but not the binding sites for the Abl SH2 domain. The connection between EphB2 and Abl/Arg appears to be reciprocal. Activated EphB2 causes tyrosine phosphorylation of Abl and Arg, and vice versa. Interestingly, treatment of COS cells and B35 neuronal-like cells with ephrin-B1 to activate endogenous EphB2 decreased the kinase activity of endogenous Abl. These data are consistent with the opposite effects that Eph receptors and Abl have on neurite ougrowth and suggest that Eph receptors and Abl family kinases have shared signaling activities.


Subject(s)
Protein-Tyrosine Kinases/physiology , Proto-Oncogene Proteins c-abl/physiology , Receptor Protein-Tyrosine Kinases/physiology , Signal Transduction/physiology , Actins/metabolism , Animals , Brain/embryology , Brain Chemistry , COS Cells , Cells, Cultured , Chick Embryo , Chlorocebus aethiops , Cytoskeleton/metabolism , Eye Proteins/analysis , Humans , Kidney/cytology , Macromolecular Substances , Mice , Nerve Tissue Proteins/analysis , Neurons/cytology , Neurons/metabolism , Phosphorylation , Protein Binding , Protein Processing, Post-Translational , Protein-Tyrosine Kinases/chemistry , Proto-Oncogene Proteins c-abl/chemistry , Receptor Protein-Tyrosine Kinases/chemistry , Receptor, EphB2 , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/physiology , Retina/chemistry , Retina/embryology , Saccharomyces cerevisiae/genetics , Two-Hybrid System Techniques , src Homology Domains
6.
J Biol Chem ; 276(42): 38940-8, 2001 Oct 19.
Article in English | MEDLINE | ID: mdl-11466320

ABSTRACT

EphB2 is a receptor tyrosine kinase of the Eph family and ephrin-B1 is one of its transmembrane ligands. In the embryo, EphB2 and ephrin-B1 participate in neuronal axon guidance, neural crest cell migration, the formation of blood vessels, and the development of facial structures and the inner ear. Interestingly, EphB2 and ephrin-B1 can both signal through their cytoplasmic domains and become tyrosine-phosphorylated when bound to each other. Tyrosine phosphorylation regulates EphB2 signaling and likely also ephrin-B1 signaling. Embryonic retina is a tissue that highly expresses both ephrin-B1 and EphB2. Although the expression patterns of EphB2 and ephrin-B1 in the retina are different, they partially overlap, and both proteins are substantially tyrosine-phosphorylated. To understand the role of ephrin-B1 phosphorylation, we have identified three tyrosines of ephrin-B1 as in vivo phosphorylation sites in transfected 293 cells stimulated with soluble EphB2 by using mass spectrometry and site-directed mutagenesis. These tyrosines are also physiologically phosphorylated in the embryonic retina, although the extent of phosphorylation at each site may differ. Furthermore, many of the tyrosines of EphB2 previously identified as phosphorylation sites in 293 cells (Kalo, M. S., and Pasquale, E. B. (1999) Biochemistry 38, 14396-14408) are also phosphorylated in retinal tissue. Our data underline the complexity of ephrin-Eph bidirectional signaling by implicating many tyrosine phosphorylation sites of the ligand-receptor complex.


Subject(s)
Membrane Proteins/metabolism , Tyrosine/metabolism , Amino Acid Sequence , Binding Sites , Cell Movement , Cells, Cultured , Cytoplasm/metabolism , Ephrin-B1 , Ephrin-B2 , Humans , Immunoblotting , Ligands , Mass Spectrometry , Membrane Proteins/biosynthesis , Molecular Sequence Data , Mutagenesis, Site-Directed , Phosphorylation , Precipitin Tests , Protein Binding , Retina/embryology , Signal Transduction , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Time Factors , Transfection
7.
Dev Biol ; 230(1): 74-88, 2001 Feb 01.
Article in English | MEDLINE | ID: mdl-11161563

ABSTRACT

In the embryonic visual system, EphA receptors are expressed on both temporal and nasal retinal ganglion cell axons. Only the temporal axons, however, are sensitive to the low concentrations of ephrin-A ligands found in the anterior optic tectum. The poor responsiveness of nasal axons to ephrin-A ligands, which allows them to traverse the anterior tectum and reach their targets in the posterior tectum, has been attributed to constitutive activation of the EphA4 receptor expressed in these axons. EphA4 is highly expressed throughout the retina, but is preferentially phosphorylated on tyrosine (activated) in nasal retina. In a screen for EphA4 ligands expressed in chicken embryonic retina, we have identified a novel ephrin, ephrin-A6. Like ephrin-A5, ephrin-A6 has high affinity for EphA4 and activates this receptor in cultured retinal cells. In the embryonic day 8 (E8) chicken visual system, ephrin-A6 is predominantly expressed in the nasal retina and ephrin-A5 in the posterior tectum. Thus, ephrin-A6 has the properties of a ligand that activates the EphA4 receptor in nasal retinal cells. Ephrin-A6 binds with high affinity to several other EphA receptors as well and causes growth cone collapse in retinal explants, demonstrating that it can elicit biological responses in retinal neurons. Ephrin-A6 expression is high at E6 and E8, when retinal axons grow to their tectal targets, and gradually declines at later developmental stages. The asymmetric distribution of ephrin-A6 in retinal cells, and the time course of its expression, suggest that this new ephrin plays a role in the establishment of visual system topography.


Subject(s)
Fetal Proteins/metabolism , Membrane Proteins/metabolism , Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Retinal Ganglion Cells/metabolism , Superior Colliculi/embryology , Superior Colliculi/metabolism , Amino Acid Sequence , Animals , Base Sequence , Chick Embryo , Cloning, Molecular , DNA Primers/genetics , DNA, Complementary/genetics , Ephrin-A5 , Gene Expression , Ligands , Membrane Proteins/genetics , Molecular Sequence Data , Proteins/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, EphA4 , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid
8.
Oncogene ; 19(49): 5614-9, 2000 Nov 20.
Article in English | MEDLINE | ID: mdl-11114742

ABSTRACT

Protein tyrosine kinase genes are the largest family of oncogenes. This is not surprising since tyrosine kinases are important components of signal transduction pathways that control cell shape, proliferation, differentiation, and migration. At 14 distinct members, the Eph kinases constitute the largest family of receptor tyrosine kinases. Although they have been most intensively studied for their roles in embryonic development, increasing evidence also implicates Eph family proteins in cancer. This review will address the recent progress in understanding the function of Eph receptors in normal development and how disregulation of these functions could promote tumorigenesis.


Subject(s)
Membrane Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Cell Transformation, Neoplastic , Humans , Ligands
9.
J Comp Neurol ; 426(2): 270-8, 2000 Oct 16.
Article in English | MEDLINE | ID: mdl-10982468

ABSTRACT

The avian auditory brainstem nuclei nucleus magnocellularis (NM) and nucleus laminaris (NL) display highly precise patterns of neuronal connectivity. NM projects tonotopically to the dorsal dendrites of ipsilateral NL neurons and to the ventral dendrites of contralateral NL neurons. The precision of this binaural segregation is evident at the earliest developmental stage at which connections can be observed. We have begun to examine the possibility that Eph receptor tyrosine kinase signaling is involved in establishing these spatially segregated connections. The expression of the EphA4 tyrosine kinase was examined at several developmental stages. EphA4 is expressed in rhombomere 5, which contains progenitors for both NM and NL. In this rhombomere, the labeling becomes striped during the time that precursor cells migrate to the auditory anlage. At the precise time when NM-NL projections are forming, EphA4 expression in NL is asymmetric, with markedly higher expression in the dorsal NL neuropil than in the ventral neuropil, suggesting a possible role in guiding growing axons to the appropriate region. At later embryonic ages EphA4 expression is symmetric around NL, and is absent in NM. As auditory function matures, EphA4 expression decreases so that by 4 days after hatch no EphA4 antibody labeling is evident in the auditory brainstem nuclei.


Subject(s)
Auditory Pathways/metabolism , Brain Stem/embryology , Chick Embryo/physiology , Chickens/growth & development , Chickens/metabolism , Fetal Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Animals, Newborn/growth & development , Animals, Newborn/metabolism , Chick Embryo/metabolism , Immunohistochemistry , Receptor, EphA4 , Time Factors
10.
J Cell Biochem ; 79(2): 213-24, 2000 Aug 02.
Article in English | MEDLINE | ID: mdl-10967549

ABSTRACT

A yeast two-hybrid screen was employed to identify ligands for the cytoplasmic domain of the NG2 chondroitin sulfate proteoglycan. Two overlapping cDNA clones selected in the screen are identical in sequence to a DNA segment coding for the most amino-terminal of the 13 PDZ domains found in the multi-PDZ-protein MUPP1. Antibodies made against recombinant polypeptides representing these two clones (NIP-2 and NIP-7) are reactive with the same 250-kDa molecule recognized by anti-MUPP1 antibodies, confirming the presence of the NIP-2 and NIP-7 sequences in the MUPP1 protein. NIP-2 and NIP-7 GST fusion proteins effectively recognize NG2 in pull-down assays, demonstrating the ability of these polypeptide segments to interact with the intact proteoglycan. The fusion proteins fail to bind NG2 missing the C-terminal half of the cytoplasmic domain, emphasizing the role of the NG2 C-terminus in the interaction with MUPP1. The existence of an NG2/MUPP1 interaction in situ is demonstrated by the ability of NG2 antibodies to co-immunoprecipitate both NG2 and MUPP1 from detergent extracts of cells expressing the two molecules. MUPP1 may serve as a multivalent scaffold that provides a means of linking NG2 with key structural and/or signaling components in the cytoplasm.


Subject(s)
Antigens/metabolism , Carrier Proteins/metabolism , Cytoplasm/metabolism , Proteoglycans/metabolism , Amino Acid Sequence , Antigens/genetics , Base Sequence , Carrier Proteins/genetics , Cell Membrane/metabolism , DNA, Complementary , Humans , Ligands , Membrane Proteins , Molecular Sequence Data , Precipitin Tests , Proteoglycans/genetics , Sequence Homology, Nucleic Acid , Signal Transduction , Tumor Cells, Cultured
11.
J Neurosci ; 20(17): 6488-500, 2000 Sep 01.
Article in English | MEDLINE | ID: mdl-10964955

ABSTRACT

Spatiotemporal expression patterns of six members of the Eph gene family (EphA4, EphA3, EphB2, ephrin-B1, ephrin-A2, and ephrin-A5) were characterized immunocytochemically at various stages of chick cerebellar development. EphA4 expression is observed in the cerebellar anlage as early as embryonic day 5 (E5) and continues in the posthatch cerebellum. During the early period of cerebellar development (E3-E8), complementarity is observed between EphA4 and ephrin-A5 expression within the cerebellar-isthmal region. By E8, differential expression of EphA4 in parasagittal Purkinje cell bands is evident, and the expression remains banded in the posthatch cerebellum. Banded expression of the ephrin-A5 ligand complements EphA4 expression during the middle period (E9-E15). During this period, ephrin-A2 and EphA3 are coexpressed in a banded pattern and with variable correlation to EphA4. Variability in the banding expression is observed for EphA4, EphA3, ephrin-A5, and ephrin-A2 across different lobes, and graded complementarity in the expression pattern of EphA3 and ephrin-A5 is observed in the external granular layer between the posterior and anterior lobes. Analysis of Purkinje cell birth date in correlation with Eph-ephrin expression during the middle period reveals that early-born cells express EphA4, whereas late-born cells express ephrin-A5. Finally, EphA4 expression domains are respected by migrating granule cell ribbons, which express both ephrin-B1 and EphB2. These expression patterns suggest multiple roles for the Eph-ephrin system in cerebellar development, including demarcation/enforcement of boundaries of the cerebellar anlage, formation/maintenance of Purkinje cell compartments, and restriction of the early phase of granule cell migration to ribbons.


Subject(s)
Body Patterning/physiology , Cerebellum/embryology , Fetal Proteins/genetics , Gene Expression Regulation, Developmental , Membrane Proteins/genetics , Neurons/physiology , Receptor Protein-Tyrosine Kinases/genetics , Transcription Factors/genetics , Animals , Cerebellum/cytology , Chick Embryo , Ephrin-A2 , Ephrin-B1 , Fetal Proteins/analysis , Membrane Proteins/analysis , Multigene Family , Nerve Tissue Proteins/genetics , Neurons/cytology , Receptor Protein-Tyrosine Kinases/analysis , Receptor, EphA4 , Receptor, EphB2 , Transcription Factors/analysis
12.
Dev Neurosci ; 22(3): 237-50, 2000.
Article in English | MEDLINE | ID: mdl-10894987

ABSTRACT

During neural development, spinal motor axons extend in a precise manner from the ventral portion of the developing spinal cord to innervate muscle targets in the limb. Although classical studies in avians have characterized the cellular interactions that influence motor axon pathfinding to the limb, less is known about the molecular mechanisms that mediate this developmental event. Here, we examine the spatiotemporal distributions of the EphA4 receptor tyrosine kinase (RTK) and its cognate ligands, ephrin-A2 and ephrin-A5, on motor neurons, their axons and their pathways to the avian hindlimb to determine whether these molecules may influence axonal projections. The expression patterns of EphA4, ephrin-A2 and ephrin-A5 mRNAs and proteins are highly complex and appear to exhibit some overlap during motor axon outgrowth and pathfinding to the hindlimb, reminiscent of the co-expression of Eph RTKs and ephrins in the retinotectal system. EphA4, similar to the carbohydrate moiety polysialic acid, strikingly marks the main dorsal, but not ventral, nerve trunk after axon sorting at the limb plexus region. Our results suggest that EphA4 RTK and its ligands may influence axon fasciculation and the sorting of axons at the limb plexus, contributing to the correct dorsoventral organization of nerve branches in the hindlimb.


Subject(s)
Axons/metabolism , Fetal Proteins/biosynthesis , Hindlimb/embryology , Hindlimb/innervation , Membrane Proteins/biosynthesis , Receptor Protein-Tyrosine Kinases/biosynthesis , Transcription Factors/biosynthesis , Animals , Chick Embryo , Ephrin-A2 , Ephrin-A5 , Fetal Proteins/genetics , Ganglia, Spinal/cytology , Ganglia, Spinal/embryology , Ganglia, Spinal/metabolism , Hindlimb/cytology , Immunohistochemistry , In Situ Hybridization , Ligands , Membrane Proteins/genetics , Motor Neurons/cytology , Motor Neurons/metabolism , Neurons, Afferent/cytology , Neurons, Afferent/metabolism , Notochord/cytology , Notochord/embryology , Notochord/metabolism , RNA, Messenger/biosynthesis , Receptor Protein-Tyrosine Kinases/genetics , Receptor, EphA4 , Transcription Factors/genetics
13.
J Neurobiol ; 42(4): 437-47, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10699981

ABSTRACT

The peripheral nervous system in vertebrates is composed of repeating metameric units of spinal nerves. During development, factors differentially expressed in a rostrocaudal pattern in the somites confine the movement of spinal motor axons and neural crest cells to the rostral half of the somitic sclerotome. The expression patterns of transmembrane ephrin-B ligands and interacting EphB receptors suggest that these proteins are likely candidates for coordinating the segmentation of spinal motor axons and neural crest cells. In vitro, ephrin-B1 has indeed been shown to repel axons extending from the rodent neural tube (Wang & Anderson, 1997). In avians, blocking interactions between EphB3 expressed by neural crest cells and ephrin-B1 localized to the caudal half of the somite in vivo resulted in loss of the rostrocaudal patterning of trunk neural crest migration (Krull et al., 1997). The role of ephrin-B1 in patterning spinal motor axon outgrowth in avian embryos was investigated. Ephrin-B1 protein was found to be expressed in the caudal half-sclerotome and in the dermomyotome at the appropriate time to interact with the EphB2 receptor expressed on spinal motor axons. Treatment of avian embryo explants with soluble ephrin-B1, however, did not perturb the segmental outgrowth of spinal motor axons through the rostral half-somite. In contrast, under the same treatment conditions with soluble ephrin-B1, neural crest cells migrated aberrantly through both rostral and caudal somite halves. These results indicate that the interaction between ephrin-B1 and EphB2 is not required for patterning spinal motor axon segmentation. Even though spinal motor axons traverse the same somitic pathway as neural crest cells, different molecular guidance mechanisms appear to influence their movement.


Subject(s)
Cell Movement/physiology , Motor Neurons/metabolism , Neural Crest/cytology , Somites/metabolism , Spinal Cord/cytology , Spinal Cord/embryology , Animals , Axons/physiology , Body Patterning/physiology , Chick Embryo , Culture Techniques , Ephrin-B1 , Ephrin-B2 , Humans , Immunoglobulin Fc Fragments/genetics , Immunohistochemistry , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Proteins/pharmacology , Motor Neurons/cytology , Neural Crest/embryology , RNA, Messenger/biosynthesis , Recombinant Fusion Proteins/metabolism
14.
Oncogene ; 19(2): 177-87, 2000 Jan 13.
Article in English | MEDLINE | ID: mdl-10644995

ABSTRACT

Eph receptor tyrosine kinases play key roles in pattern formation during embryonic development, but little is known about the mechanisms by which they elicit specific biological responses in cells. Here, we investigate the role of tyrosines 605 and 611 in the juxtamembrane region of EphB2, because they are conserved Eph receptor autophosphorylation sites and demonstrated binding sites for the SH2 domains of multiple signaling proteins. Mutation of tyrosines 605 and 611 to phenylalanine impaired EphB2 kinase activity, complicating analysis of their function as SH2 domain binding sites and their contribution to EphB2-mediated signaling. In contrast, mutation to the negatively charged glutamic acid disrupted SH2 domain binding without reducing EphB2 kinase activity. By using a panel of EphB2 mutants, we found that kinase activity is required for the changes in cell-matrix and cell - cell adhesion, cytoskeletal organization, and activation of mitogen-activated protein (MAP) kinases elicited by EphB2 in transiently transfected cells. Instead, the two juxtamembrane SH2 domain binding sites were dispensable for these effects. These results suggest that phosphorylation of tyrosines 605 and 611 is critical for EphB2-mediated cellular responses because it regulates EphB2 kinase activity.


Subject(s)
Amino Acid Substitution , Conserved Sequence , Glutamic Acid/physiology , Receptor Protein-Tyrosine Kinases/physiology , Tyrosine/physiology , src Homology Domains , 3T3 Cells , Actins/metabolism , Amino Acid Substitution/genetics , Animals , COS Cells , Cell Size/genetics , Enzyme Activation/genetics , Glutamic Acid/genetics , Humans , Mice , Mice, Knockout , Mitogen-Activated Protein Kinases/metabolism , Mutagenesis , Phenylalanine/genetics , Phenylalanine/metabolism , Phosphorylation , Protein Binding/genetics , Receptor Protein-Tyrosine Kinases/genetics , Receptor, EphB2 , Tyrosine/genetics , src Homology Domains/genetics , src-Family Kinases/metabolism
15.
Oncogene ; 19(52): 6043-52, 2000 Dec 07.
Article in English | MEDLINE | ID: mdl-11146556

ABSTRACT

Eph receptor tyrosine kinases and their ephrin ligands have been implicated in embryonic vascular development and in in vivo models of angiogenesis. Eph proteins may also regulate tumor neovascularization, but this role has not been previously investigated. To screen for Eph proteins expressed in tumor blood vessels, we used tumor xenografts grown in nude mice from MDA-MB-435 human breast cancer cells or KS1767 human Kaposi's sarcoma cells. By immunohistochemistry, the ephrin-A1 ligand and one of its receptors, EphA2, were detected throughout tumor vasculature. Double-labeling with anti-CD34 antibodies demonstrated that both ephrin-A1 and EphA2 were expressed in xenograft endothelial cells and also tumor cells. Furthermore, EphA2 was tyrosine-phosphorylated in the xenograft tumors, indicating that it was activated, presumably by interacting with ephrin-A1. Ephrin-A1 and EphA2 were also detected in both the vasculature and tumor cells of surgically removed human cancers. In an in vitro angiogenesis model, a dominant negative form of EphA2 inhibited capillary tube-like formation by human umbilical vein endothelial cells (HUVECs), demonstrating a requirement for EphA receptor signaling. These data suggest that ephrin-A1 and EphA2 play a role in human cancers, at least in part by influencing tumor neovascularization. Eph proteins may represent promising new targets for antiangiogenic cancer treatments.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/blood supply , Neovascularization, Pathologic/metabolism , Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Blotting, Western , Breast Neoplasms/blood supply , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Capillaries/growth & development , Capillaries/metabolism , Cells, Cultured , Collagen , Drug Combinations , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Ephrin-A1 , Female , Fluorescent Antibody Technique , Humans , Hyperplasia/metabolism , Hyperplasia/pathology , Laminin , Ligands , Mice , Mice, Nude , Neoplasm Transplantation , Neoplasms/metabolism , Neoplasms/pathology , Neovascularization, Pathologic/blood , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Phosphotyrosine/metabolism , Proteoglycans , Receptor Protein-Tyrosine Kinases/blood , Receptor Protein-Tyrosine Kinases/genetics , Receptor, EphA2 , Sarcoma, Kaposi/blood supply , Sarcoma, Kaposi/metabolism , Sarcoma, Kaposi/pathology , Signal Transduction , Transplantation, Heterologous , Tumor Cells, Cultured
16.
J Biol Chem ; 274(45): 31941-6, 1999 Nov 05.
Article in English | MEDLINE | ID: mdl-10542222

ABSTRACT

The Eph family of receptor tyrosine kinases has been implicated in many developmental patterning processes, including cell segregation, cell migration, and axon guidance. The cellular components involved in the signaling pathways of the Eph receptors, however, are incompletely characterized. Using a yeast two-hybrid screen, we have identified a novel signaling intermediate, SHEP1 (SH2 domain-containing Eph receptor-binding protein 1), which is expressed in the embryonic and adult brain. SHEP1 contains an Src homology 2 domain that binds to a conserved tyrosine-phosphorylated motif in the juxtamembrane region of the EphB2 receptor and may itself be a target of EphB2 kinase activity, since it becomes heavily tyrosine-phosphorylated in cells expressing activated EphB2. SHEP1 also contains a domain similar to Ras guanine nucleotide exchange factor domains and binds to the GTPases R-Ras and Rap1A, but not Ha-Ras or RalA. Thus, SHEP1 directly links activated, tyrosine-phosphorylated Eph receptors to small Ras superfamily GTPases.


Subject(s)
Adaptor Proteins, Signal Transducing , Carrier Proteins/metabolism , GTP Phosphohydrolases/metabolism , Nerve Tissue Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , rap1 GTP-Binding Proteins/metabolism , ras Proteins/metabolism , Amino Acid Sequence , Animals , Carrier Proteins/chemistry , Cell Line , Cloning, Molecular , Gene Library , Humans , Mice , Molecular Sequence Data , Nerve Tissue Proteins/chemistry , Polymerase Chain Reaction , Rabbits , Receptor, EphB2 , Signal Transduction
17.
Biochemistry ; 38(43): 14396-408, 1999 Oct 26.
Article in English | MEDLINE | ID: mdl-10572014

ABSTRACT

Autophosphorylation regulates the function of receptor tyrosine kinases. To dissect the mechanism by which Eph receptors transmit signals, we have developed an approach using matrix-assisted laser desorption-ionization (MALDI) mass spectrometry to map systematically their in vivo tyrosine phosphorylation sites. With this approach, phosphorylated peptides from receptors digested with various endoproteinases were selectively isolated on immobilized anti-phosphotyrosine antibodies and analyzed directly by MALDI mass spectrometry. Multiple in vivo tyrosine phosphorylation sites were identified in the juxtamembrane region, kinase domain, and carboxy-terminal tail of EphB2 and EphB5, and found to be remarkably conserved between these EphB receptors. A number of these sites were also identified as in vitro autophosphorylation sites of EphB5 by phosphopeptide mapping using two-dimensional chromatography. Only two in vitro tyrosine phosphorylation sites had previously been directly identified for Eph receptors. Our data further indicate that in vivo EphB2 and EphB5 are also extensively phosphorylated on serine and threonine residues. Because phosphorylation at each site can affect receptor signaling properties, the multiple phosphorylation sites identified here for the EphB receptors suggest a complex regulation of their functions, presumably achieved by autophosphorylation as well as phosphorylation by other kinases. In addition, we show that MALDI mass spectrometry can be used to determine the binding sites for Src homology 2 (SH2) domains by identifying the EphB2 phosphopeptides that bind to the SH2 domain of the Src kinase.


Subject(s)
Receptor Protein-Tyrosine Kinases/metabolism , Tyrosine/metabolism , Amino Acid Sequence , Animals , Binding Sites , Cells, Cultured , Chick Embryo , Humans , Kidney/cytology , Kidney/metabolism , Molecular Sequence Data , Mutagenesis, Site-Directed , Peptide Mapping , Phosphorylation , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/genetics , Receptor, EphB2 , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tyrosine/chemistry , src Homology Domains
18.
Proc Natl Acad Sci U S A ; 96(24): 13813-8, 1999 Nov 23.
Article in English | MEDLINE | ID: mdl-10570155

ABSTRACT

The ability of integrins to mediate cell attachment to extracellular matrices and to blood proteins is regulated from inside the cell. Increased ligand-binding activity of integrins is critical for platelet aggregation upon blood clotting and for leukocyte extravasation to inflamed tissues. Decreased adhesion is thought to promote tumor cell invasion. R-Ras, a small intracellular GTPase, regulates the binding of integrins to their ligands outside the cell. Here we show that the Eph receptor tyrosine kinase, EphB2, can control integrin activity through R-Ras. Cells in which EphB2 is activated become poorly adherent to substrates coated with integrin ligands, and a tyrosine residue in the R-Ras effector domain is phosphorylated. The R-Ras phosphorylation and loss of cell adhesion are causally related, because forced expression of an R-Ras variant resistant to phosphorylation at the critical site made cells unresponsive to the anti-adhesive effect of EphB2. This is an unusual regulatory pathway among the small GTPases. Reduced adhesiveness induced through the Eph/R-Ras pathway may explain the repulsive effect of the Eph receptors in axonal pathfinding and may facilitate tumor cell invasion and angiogenesis.


Subject(s)
Integrins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , ras Proteins/metabolism , 3T3 Cells , Animals , Cell Adhesion/physiology , Cell Line, Transformed , Chickens , Humans , Mice , Mutagenesis, Site-Directed , Phosphorylation , Receptor Protein-Tyrosine Kinases/genetics , Receptor, EphB2 , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , ras Proteins/genetics
19.
Mech Dev ; 87(1-2): 119-28, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10495276

ABSTRACT

The formation of the ten cerebellar lobules is an unsolved problem in brain development. We report a screen for the four subfamilies of Eph receptors and their ligands (ephrins) in developing mouse cerebellum, using soluble receptor-immunoglobulin and ligand-immunoglobulin fusion proteins, and antibodies against EphA and ephrin-B proteins. Our results identify Eph receptors and ephrins as the first molecules known to demarcate individual lobules during development. Staining for ephrin-A ligands is in lobule VIII as it forms, across the whole width of the cerebellum. Staining for three EphA receptors approximately coincides with presumptive lobules VI and/or VII before and just after birth, whereas a fourth EphA receptor (EphA4, which binds ligands of both subfamilies) has more widespread expression. Staining for EphB receptors is in lobules VII, VIII, and IX. Staining for ephrin-B ligands is much weaker, becomes detectable only after birth, and does not appear to be lobule-specific. Staining for all subfamilies spreads to at least some adjacent lobules as maturation proceeds. The lobule-specific patterns appear before the lobules form, and initially extend across the width of the cerebellum, in spite of the lesser conservation of the lateral extensions of the lobules. These expression patterns define previously unknown developmental units and suggest that Eph family proteins may contribute to cerebellar morphogenesis.


Subject(s)
Cerebellum/embryology , Cerebellum/metabolism , Membrane Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , Ephrin-A2 , Ephrin-A5 , Ephrin-B1 , Epitopes/metabolism , Fetal Proteins/metabolism , Immunoglobulin Fc Fragments/metabolism , Immunohistochemistry , In Situ Hybridization , Mice , Receptor, EphA4 , Receptor, EphA7 , Time Factors , Transcription Factors/metabolism
20.
Cell Tissue Res ; 298(1): 1-9, 1999 Oct.
Article in English | MEDLINE | ID: mdl-10555534

ABSTRACT

The Eph receptors are a unique family of receptor tyrosine kinases that enforce cellular position in tissues through mainly repulsive signals generated upon cell-cell contact. Together, Eph receptors and their membrane-anchored ligands. the ephrins, are key molecules for establishing tissue organization through signaling pathways that control axonal projection, cell migration, and the maintenance of cellular boundaries. Through their SH2 (Src Homology 2) and PDZ (postsynaptic density protein, disks large, zona occludens) domains, several signaling molecules have been demonstrated to interact with the activated cytoplasmic domain of Eph receptors by using the yeast two-hybrid system and in vitro biochemical assays. Most proteins found to interact with Eph receptors are well-known regulators of cytoskeletal organization and cell adhesion, and also cell proliferation. Promoting growth, however, does not appear to be a primary role of Eph receptors. Explaining which signaling interactions identified for the Eph receptors have physiological significance, how Eph receptor signaling cascades are propagated, and characterizing the intrinsic signaling properties of the ephrins are all exciting questions currently being investigated.


Subject(s)
Receptor Protein-Tyrosine Kinases/physiology , Signal Transduction/physiology , Animals , Cell Adhesion/physiology , Cell Communication/physiology , Enzyme Activation , Humans , Ligands , Membrane Proteins/physiology , Phosphorylation , Receptor Protein-Tyrosine Kinases/chemistry , Receptor, EphA1 , src Homology Domains
SELECTION OF CITATIONS
SEARCH DETAIL
...