Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 505
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Rev Mol Cell Biol ; 17(4): 240-56, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26790531

ABSTRACT

Eph receptor Tyr kinases and their membrane-tethered ligands, the ephrins, elicit short-distance cell-cell signalling and thus regulate many developmental processes at the interface between pattern formation and morphogenesis, including cell sorting and positioning, and the formation of segmented structures and ordered neural maps. Their roles extend into adulthood, when ephrin-Eph signalling regulates neuronal plasticity, homeostatic events and disease processes. Recently, new insights have been gained into the mechanisms of ephrin-Eph signalling in different cell types, and into the physiological importance of ephrin-Eph in different organs and in disease, raising questions for future research directions.


Subject(s)
Ephrins/physiology , Receptors, Eph Family/metabolism , Signal Transduction , Animals , Growth and Development/physiology , Humans
2.
Mol Cell ; 79(3): 390-405.e7, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32619402

ABSTRACT

Despite their apparent lack of catalytic activity, pseudokinases are essential signaling molecules. Here, we describe the structural and dynamic properties of pseudokinase domains from the Wnt-binding receptor tyrosine kinases (PTK7, ROR1, ROR2, and RYK), which play important roles in development. We determined structures of all pseudokinase domains in this family and found that they share a conserved inactive conformation in their activation loop that resembles the autoinhibited insulin receptor kinase (IRK). They also have inaccessible ATP-binding pockets, occluded by aromatic residues that mimic a cofactor-bound state. Structural comparisons revealed significant domain plasticity and alternative interactions that substitute for absent conserved motifs. The pseudokinases also showed dynamic properties that were strikingly similar to those of IRK. Despite the inaccessible ATP site, screening identified ATP-competitive type-II inhibitors for ROR1. Our results set the stage for an emerging therapeutic modality of "conformational disruptors" to inhibit or modulate non-catalytic functions of pseudokinases deregulated in disease.


Subject(s)
Cell Adhesion Molecules/chemistry , Protein Kinase Inhibitors/pharmacology , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Tyrosine Kinase-like Orphan Receptors/chemistry , Amino Acid Sequence , Animals , Baculoviridae/genetics , Baculoviridae/metabolism , Binding Sites , Cell Adhesion Molecules/antagonists & inhibitors , Cell Adhesion Molecules/genetics , Cell Adhesion Molecules/metabolism , Cell Line , Cloning, Molecular , Crystallography, X-Ray , Gene Expression , Humans , Mice , Models, Molecular , Precursor Cells, B-Lymphoid/cytology , Precursor Cells, B-Lymphoid/metabolism , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Kinase Inhibitors/chemistry , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/antagonists & inhibitors , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Receptors, Eph Family/antagonists & inhibitors , Receptors, Eph Family/chemistry , Receptors, Eph Family/genetics , Receptors, Eph Family/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sf9 Cells , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Spodoptera , Structural Homology, Protein , Substrate Specificity
3.
Proc Natl Acad Sci U S A ; 121(19): e2322934121, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38701119

ABSTRACT

EPH receptors (EPHs), the largest family of tyrosine kinases, phosphorylate downstream substrates upon binding of ephrin cell surface-associated ligands. In a large cohort of endometriotic lesions from individuals with endometriosis, we found that EPHA2 and EPHA4 expressions are increased in endometriotic lesions relative to normal eutopic endometrium. Because signaling through EPHs is associated with increased cell migration and invasion, we hypothesized that chemical inhibition of EPHA2/4 could have therapeutic value. We screened DNA-encoded chemical libraries (DECL) to rapidly identify EPHA2/4 kinase inhibitors. Hit compound, CDD-2693, exhibited picomolar/nanomolar kinase activity against EPHA2 (Ki: 4.0 nM) and EPHA4 (Ki: 0.81 nM). Kinome profiling revealed that CDD-2693 bound to most EPH family and SRC family kinases. Using NanoBRET target engagement assays, CDD-2693 had nanomolar activity versus EPHA2 (IC50: 461 nM) and EPHA4 (IC50: 40 nM) but was a micromolar inhibitor of SRC, YES, and FGR. Chemical optimization produced CDD-3167, having picomolar biochemical activity toward EPHA2 (Ki: 0.13 nM) and EPHA4 (Ki: 0.38 nM) with excellent cell-based potency EPHA2 (IC50: 8.0 nM) and EPHA4 (IC50: 2.3 nM). Moreover, CDD-3167 maintained superior off-target cellular selectivity. In 12Z endometriotic epithelial cells, CDD-2693 and CDD-3167 significantly decreased EFNA5 (ligand) induced phosphorylation of EPHA2/4, decreased 12Z cell viability, and decreased IL-1ß-mediated expression of prostaglandin synthase 2 (PTGS2). CDD-2693 and CDD-3167 decreased expansion of primary endometrial epithelial organoids from patients with endometriosis and decreased Ewing's sarcoma viability. Thus, using DECL, we identified potent pan-EPH inhibitors that show specificity and activity in cellular models of endometriosis and cancer.


Subject(s)
Protein Kinase Inhibitors , Humans , Female , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Endometriosis/drug therapy , Endometriosis/metabolism , Endometriosis/pathology , DNA/metabolism , Receptors, Eph Family/metabolism , Receptors, Eph Family/antagonists & inhibitors , Receptor, EphA2/metabolism , Receptor, EphA2/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Small Molecule Libraries/chemistry , Cell Movement/drug effects
4.
Biochem Biophys Res Commun ; 720: 150072, 2024 08 06.
Article in English | MEDLINE | ID: mdl-38749187

ABSTRACT

The Eph receptor, a prototypically large receptor protein tyrosine kinase, interacts with ephrin ligands, forming a bidirectional signaling system that impacts diverse brain functions. Eph receptors and ephrins mediate forward and reverse signaling, affecting neurogenesis, axon guidance, and synaptic signaling. While mammalian studies have emphasized their roles in neurogenesis and synaptic plasticity, the Drosophila counterparts are less studied, especially in glial cells, despite structural similarities. Using RNAi to modulate Eph/ephrin expression in Drosophila neurons and glia, we studied their roles in brain development and sleep and circadian behavior. Knockdown of neuronal ephrin disrupted mushroom body development, while glial knockdown had minimal impact. Surprisingly, disrupting ephrin in neurons or glial cells altered sleep and circadian rhythms, indicating a direct involvement in these behaviors independent from developmental effects. Further analysis revealed distinct sleep phenotypes between neuronal and glial knockdowns, underscoring the intricate interplay within the neural circuits that govern behavior. Glia-specific knockdowns showed altered sleep patterns and reduced circadian rhythmicity, suggesting an intricate role of glia in sleep regulation. Our findings challenge simplistic models of Eph/ephrin signaling limited to neuron-glia communication and emphasize the complexity of the regulatory networks modulating behavior. Future investigations targeting specific glial subtypes will enhance our understanding of Eph/ephrin signaling's role in sleep regulation across species.


Subject(s)
Circadian Rhythm , Ephrins , Mushroom Bodies , Neuroglia , Neurons , Signal Transduction , Sleep , Animals , Neuroglia/metabolism , Sleep/physiology , Sleep/genetics , Circadian Rhythm/physiology , Neurons/metabolism , Ephrins/metabolism , Ephrins/genetics , Mushroom Bodies/metabolism , Drosophila Proteins/metabolism , Drosophila Proteins/genetics , Receptors, Eph Family/metabolism , Receptors, Eph Family/genetics , Drosophila melanogaster/metabolism , Drosophila melanogaster/physiology , Drosophila melanogaster/genetics , Drosophila/metabolism
5.
Cell Commun Signal ; 22(1): 299, 2024 May 29.
Article in English | MEDLINE | ID: mdl-38811954

ABSTRACT

Eph receptors constitute the largest family of receptor tyrosine kinases, comprising 14 distinct members classified into two subgroups: EphAs and EphBs.. Despite their essential functions in normal physiological processes, accumulating evidence suggests that the involvement of the Eph family in cancer is characterized by a dual and often contradictory nature. Research indicates that Eph/ephrin bidirectional signaling influences cell-cell communication, subsequently regulating cell migration, adhesion, differentiation and proliferation. The contradictory functionalities may arise from the diversity of Eph signaling pathways and the heterogeneity of different cancer microenvironment. In this review, we aim to discuss the dual role of the Eph receptors in tumor development, attempting to elucidate the paradoxical functionality through an exploration of Eph receptor signaling pathways, angiogenesis, immune responses, and more. Our objective is to provide a comprehensive understanding of the molecular mechanisms underlying tumor development. Additionally, we will explore the evolving landscape of utilizing Eph receptors as potential targets for tumor therapy and diagnostic tools.


Subject(s)
Neoplasms , Neovascularization, Pathologic , Receptors, Eph Family , Signal Transduction , Humans , Neoplasms/pathology , Neoplasms/metabolism , Neoplasms/immunology , Neovascularization, Pathologic/metabolism , Receptors, Eph Family/metabolism , Animals , Disease Progression , Immunity , Angiogenesis
6.
Dev Dyn ; 252(1): 10-26, 2023 01.
Article in English | MEDLINE | ID: mdl-35705527

ABSTRACT

Acoustic communication relies crucially on accurate interpretation of information about the intensity, frequency, timing, and location of diverse sound stimuli in the environment. To meet this demand, neurons along different levels of the auditory system form precisely organized neural circuits. The assembly of these precise circuits requires tight regulation and coordination of multiple developmental processes. Several groups of axon guidance molecules have proven critical in controlling these processes. Among them, the family of Eph receptors and their ephrin ligands emerge as one group of key players. They mediate diverse functions at multiple levels of the auditory pathway, including axon guidance and targeting, topographic map formation, as well as cell migration and tissue pattern formation. Here, we review our current knowledge of how Eph and ephrin molecules regulate different processes in the development and maturation of central auditory circuits.


Subject(s)
Auditory Pathways , Ephrins , Auditory Pathways/metabolism , Neurons/metabolism , Receptors, Eph Family/metabolism , Signal Transduction/physiology
7.
FASEB J ; 36(1): e22076, 2022 01.
Article in English | MEDLINE | ID: mdl-34856019

ABSTRACT

A distinct boundary exists between the progenitor cells in the basal limbal epithelium and the more differentiated corneal epithelial basal cells. We have shown that reciprocal expression patterns of EphA2 and Ephrin-A1 are likely to contribute to normal limbal-corneal epithelial compartmentalization as well as play a role in response to injury. How this signaling axis is regulated remains unclear. We have demonstrated that microRNAs (miRNAs) play critical roles in corneal epithelial wound healing and several miRNAs (e.g. miR-210) have been predicted to target ephrins. Previous expression profiling experiments demonstrated that miR-210 is prominently expressed in corneal epithelial cells. RNA-seq data acquired from miR-210-depleted HCECs showed up-regulation of genes involved in cellular migration. In addition, miR-210 is decreased after corneal injury while EphA2 is increased. Moreover, antago-210-treated HCECs markedly enhanced wound closure in a scratch wound assay. Antago-210 treatment resulted in increased EphA2 protein levels as well as pS897-EphA2, the pro-migratory form of EphA2. As expected, Ephrin-A1 levels were reduced, while levels of a well-known target of miR-210, Ephrin-A3, were increased by antago-210 treatment. The increase in migration with antago-210 could be inhibited by Ephrin-A1 overexpression, Ephrin-A1-Fc treatment or siRNA depletion of EphA2. However, depletion of Ephrin-A3 did not have effects on the antago-210-induced increase in migration. In addition, Ephrin-A1 overexpression and siEphA2 dampened EGFR signaling, which is increased by antago-210. Our data clearly demonstrate a link between miR-210 and EphA2/Ephrin-A1 signaling that regulates, in part, corneal epithelial migration. This interaction might potentially control the limbal-corneal epithelial boundary.


Subject(s)
Cell Movement , Cornea/metabolism , Epithelial Cells/metabolism , Gene Expression Regulation , MicroRNAs/metabolism , Receptors, Eph Family/metabolism , Humans , MicroRNAs/genetics , RNA-Seq , Receptors, Eph Family/genetics
8.
Mol Pharm ; 20(12): 6066-6078, 2023 Dec 04.
Article in English | MEDLINE | ID: mdl-37906960

ABSTRACT

Erythropoietin-producing hepatocellular (Eph) receptors and their ligands, ephrins, are the largest subfamily of receptor tyrosine kinases (RTKs) that have emerged as a new class of cancer biomarkers due to their aberrant expression in cancer progression. The activation of Eph receptors either due to their hyperexpression or via high affinity binding with their respective ephrin ligands initiates a cascade of signals that impacts cancer development and progression. In prostate cancer, the overexpression of the EphA6 receptor has been correlated with increased metastatic potential. Azurin, a small redox protein, is known to prevent tumor progression by binding to cell surface Eph receptors, inhibiting its autophosphorylation in the kinase domain and thereby disrupting Eph-ephrin signaling. Hence, a self-assembled, theranostic nanosystem of recombinant fusion protein his6EGFP-azu (80-128) was designed by conjugating enhanced green fluorescent protein (EGFP) with the C-terminal region of azurin. This design was inspired by the in silico binding study, where the analogue of ephrinA, his6EGFP-azu (80-128) showed higher binding affinity for the EphA6 receptor than the ephrinA ligands. The his6EGFP-azu (80-128) nanosystem which assembled as nanoparticles was tested for its ability to simultaneously detect and kill the prostate cancer cells, LNCaP. This was achieved by specifically targeting EphA6 receptors overexpressed on the cancer cell surface via C-terminal peptide, azu (80-128). Herein, we report antiproliferative, apoptotic, antimigratory, and anti-invasive effects of this nanosystem on LNCaP cells, while having no similar effects on EphA6 negative human normal lung cells, WI-38.


Subject(s)
Azurin , Prostatic Neoplasms , Receptor, EphA6 , Male , Humans , Receptors, Eph Family/chemistry , Receptors, Eph Family/metabolism , Azurin/genetics , Precision Medicine , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Ephrins/chemistry , Ephrins/metabolism
9.
Cells Tissues Organs ; 212(2): 125-137, 2023.
Article in English | MEDLINE | ID: mdl-34695818

ABSTRACT

The ephrin-B family of membrane-bound ligands is involved in skeletal patterning, osteogenesis, and bone homeostasis. Yet, despite the increasing collection of data affirming their importance in bone, the Eph tyrosine kinases that serve as the receptors for these ephrins in osteoblast stem cell niches remain unidentified. Here we report the expression of EphB3 at sites of bone growth in the embryo, especially at the calvaria suture fronts, periosteum, chondrocytes, and trabeculae of developing long bones. Strong EphB3 expression persisted in the adult calvarial sutures and in the proliferative chondrocytes of long bones, both of which are documented niches for osteoblastic stem cells. We observed EphB3-positive cells in the tissue filling a created calvarial injury, further implying EphB3 involvement in bone healing. Genetic knockout of EphB3 caused an increase in the bone tissue volume as a fraction of total volume in 6-week-old calvaria and in femoral trabecular density, compared to wild type controls. This difference resolved by 12 weeks of age, when we instead observed an increase in the bone volume of femoral trabeculae and in trabecular thickness. Our data identify EphB3 as a candidate regulator of osteogenesis either alone or in combination with other bone-expressed Ephs, and indicate that it appears to function as a limiter of bone growth.


Subject(s)
Osteogenesis , Receptors, Eph Family , Receptors, Eph Family/metabolism , Ephrins/genetics , Ephrins/metabolism , Skull , Osteoblasts
10.
Cell ; 133(6): 949-51, 2008 Jun 13.
Article in English | MEDLINE | ID: mdl-18555770

ABSTRACT

Dominantly inherited mutations in an endoplasmic reticulum protein called VAPB have been found in a subset of patients with a rare familial form of amyotrophic lateral sclerosis (ALS). In this issue, Tsuda et al. (2008) identify a secreted form of VAPB that binds directly to Eph receptors inducing their activation and signaling, providing fresh insights into ALS pathogenesis, including non-neuronal aspects of this disorder.


Subject(s)
Amyotrophic Lateral Sclerosis/metabolism , Receptors, Eph Family/metabolism , Vesicular Transport Proteins/metabolism , Animals , Humans , Signal Transduction
11.
Cell ; 133(1): 38-52, 2008 Apr 04.
Article in English | MEDLINE | ID: mdl-18394988

ABSTRACT

Receptor tyrosine kinases of the Eph family bind to cell surface-associated ephrin ligands on neighboring cells. The ensuing bidirectional signals have emerged as a major form of contact-dependent communication between cells. New findings reveal that Eph receptors and ephrins coordinate not only developmental processes but also the normal physiology and homeostasis of many adult organs. Imbalance of Eph/ephrin function may therefore contribute to a variety of diseases. The challenge now is to better understand the complex and seemingly paradoxical signaling mechanisms of Eph receptors and ephrins, which will enable effective strategies to target these proteins in the treatment of diseases such as diabetes and cancer.


Subject(s)
Cell Communication , Ephrins/metabolism , Receptors, Eph Family/metabolism , Animals , Diabetes Mellitus/metabolism , Humans , Neoplasms/metabolism , Nervous System/embryology , Signal Transduction
12.
Cell ; 133(6): 963-77, 2008 Jun 13.
Article in English | MEDLINE | ID: mdl-18555774

ABSTRACT

VAP proteins (human VAPB/ALS8, Drosophila VAP33, and C. elegans VPR-1) are homologous proteins with an amino-terminal major sperm protein (MSP) domain and a transmembrane domain. The MSP domain is named for its similarity to the C. elegans MSP protein, a sperm-derived hormone that binds to the Eph receptor and induces oocyte maturation. A point mutation (P56S) in the MSP domain of human VAPB is associated with Amyotrophic lateral sclerosis (ALS), but the mechanisms underlying the pathogenesis are poorly understood. Here we show that the MSP domains of VAP proteins are cleaved and secreted ligands for Eph receptors. The P58S mutation in VAP33 leads to a failure to secrete the MSP domain as well as ubiquitination, accumulation of inclusions in the endoplasmic reticulum, and an unfolded protein response. We propose that VAP MSP domains are secreted and act as diffusible hormones for Eph receptors. This work provides insight into mechanisms that may impact the pathogenesis of ALS.


Subject(s)
Caenorhabditis elegans Proteins/metabolism , Drosophila Proteins/metabolism , Membrane Proteins/metabolism , Receptors, Eph Family/metabolism , Vesicular Transport Proteins/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Animals , Animals, Genetically Modified , Caenorhabditis elegans/metabolism , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/genetics , Cell Line , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Drosophila melanogaster/metabolism , Endoplasmic Reticulum/metabolism , Humans , Ligands , Membrane Proteins/chemistry , Membrane Proteins/genetics , Protein Folding , Protein Structure, Tertiary , Ubiquitination , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics
13.
Cell Mol Life Sci ; 79(11): 583, 2022 Nov 05.
Article in English | MEDLINE | ID: mdl-36334147

ABSTRACT

Eph receptors and their ligands, Ephrins, are involved in the thymocyte-thymic epithelial cell (TEC) interactions, key for the functional maturation of both thymocytes and thymic epithelium. Several years ago, we reported that the lack of EphA4, a Eph of the subfamily A, coursed with reduced proportions of double positive (DP) thymocytes apparently due to an altered thymic epithelial stroma [Munoz et al. in J Immunol 177:804-813, 2006]. In the present study, we reevaluate the lymphoid, epithelial, and extracellular matrix (ECM) phenotype of EphA4-/- mice grouped into three categories with respect to their proportions of DP thymocytes. Our results demonstrate a profound hypocellularity, specific alterations of T cell differentiation that affected not only DP thymocytes, but also double negative and single positive T cell subsets, as well as the proportions of positively and negatively selected thymocytes. In correlation, thymic histological organization changed markedly, especially in the cortex, as well as the proportions of both Ly51+UEA-1- cortical TECs and Ly51-UEA-1+ medullary TECs. The alterations observed in the expression of ECM components (Fibronectin, Laminin, Collagen IV), integrin receptors (VLA-4, VLA-6), chemokines (CXCL12, CCL25, CCL21) and their receptors (CXCR4, CCR7, CCR9) and in vitro transwell assays on the capacity of migration of WT and mutant thymocytes suggest that the lack of EphA4 alters T-cell differentiation by presumably affecting cell adhesion between TECs and T-TEC interactions rather than by thymocyte migration.


Subject(s)
Thymocytes , Thymus Gland , Mice , Animals , Thymocytes/metabolism , Thymus Gland/metabolism , Lymphocyte Activation , Epithelial Cells/metabolism , Cell Differentiation , Receptors, Eph Family/metabolism , Extracellular Matrix
14.
PLoS Genet ; 16(2): e1008300, 2020 02.
Article in English | MEDLINE | ID: mdl-32092051

ABSTRACT

Craniofrontonasal syndrome (CFNS) is a rare X-linked disorder characterized by craniofacial, skeletal, and neurological anomalies and is caused by mutations in EFNB1. Heterozygous females are more severely affected by CFNS than hemizygous males, a phenomenon called cellular interference that results from EPHRIN-B1 mosaicism. In Efnb1 heterozygous mice, mosaicism for EPHRIN-B1 results in cell sorting and more severe phenotypes than Efnb1 hemizygous males, but how craniofacial dysmorphology arises from cell segregation is unknown and CFNS etiology therefore remains poorly understood. Here, we couple geometric morphometric techniques with temporal and spatial interrogation of embryonic cell segregation in mouse mutant models to elucidate mechanisms underlying CFNS pathogenesis. By generating EPHRIN-B1 mosaicism at different developmental timepoints and in specific cell populations, we find that EPHRIN-B1 regulates cell segregation independently in early neural development and later in craniofacial development, correlating with the emergence of quantitative differences in face shape. Whereas specific craniofacial shape changes are qualitatively similar in Efnb1 heterozygous and hemizygous mutant embryos, heterozygous embryos are quantitatively more severely affected, indicating that Efnb1 mosaicism exacerbates loss of function phenotypes rather than having a neomorphic effect. Notably, neural tissue-specific disruption of Efnb1 does not appear to contribute to CFNS craniofacial dysmorphology, but its disruption within neural crest cell-derived mesenchyme results in phenotypes very similar to widespread loss. EPHRIN-B1 can bind and signal with EPHB1, EPHB2, and EPHB3 receptor tyrosine kinases, but the signaling partner(s) relevant to CFNS are unknown. Geometric morphometric analysis of an allelic series of Ephb1; Ephb2; Ephb3 mutant embryos indicates that EPHB2 and EPHB3 are key receptors mediating Efnb1 hemizygous-like phenotypes, but the complete loss of EPHB1-3 does not fully recapitulate the severity of CFNS-like Efnb1 heterozygosity. Finally, by generating Efnb1+/Δ; Ephb1; Ephb2; Ephb3 quadruple knockout mice, we determine how modulating cumulative receptor activity influences cell segregation in craniofacial development and find that while EPHB2 and EPHB3 play an important role in craniofacial cell segregation, EPHB1 is more important for cell segregation in the brain; surprisingly, complete loss of EPHB1-EPHB3 does not completely abrogate cell segregation. Together, these data advance our understanding of the etiology and signaling interactions underlying CFNS dysmorphology.


Subject(s)
Cell Movement/genetics , Craniofacial Abnormalities/genetics , Ephrin-B1/genetics , Neural Crest/embryology , Skull/abnormalities , Animals , Craniofacial Abnormalities/diagnosis , Disease Models, Animal , Embryo, Mammalian , Embryonic Development/genetics , Ephrin-B1/metabolism , Female , Heterozygote , Humans , Male , Mice , Mice, Knockout , Mosaicism , Mutation , Neural Crest/cytology , Phenotype , Receptors, Eph Family/genetics , Receptors, Eph Family/metabolism , Severity of Illness Index , Sex Factors , Skull/embryology , X Chromosome/genetics
15.
Int J Mol Sci ; 24(3)2023 Feb 03.
Article in English | MEDLINE | ID: mdl-36769332

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a major concern for health care systems worldwide, since its mortality remains unaltered despite the surge in cutting-edge science. The EPH/ephrin signaling system was first investigated in the 1980s. EPH/ephrins have been shown to exert bidirectional signaling and cell-to-cell communication, influencing cellular morphology, adhesion, migration and invasion. Recent studies have highlighted the critical role of the EPH/ephrin system in various physiologic processes, including cellular proliferation, survival, synaptic plasticity and angiogenesis. Thus, it has become evident that the EPH/ephrin signaling system may have compelling effects on cell homeostasis that contribute to carcinogenesis. In particular, the EPH/ephrins have an impact on pancreatic morphogenesis and development, whereas several EPHs and ephrins are altered in PDAC. Several clinical and preclinical studies have attempted to elucidate the effects of the EPH/ephrin pathway, with multilayered effects on PDAC development. These studies have highlighted its highly promising role in the diagnosis, prognosis and therapeutic management of PDAC. The aim of this review is to explore the obscure aspects of the EPH/ephrin system concerning the development, physiology and homeostasis of the pancreas.


Subject(s)
Adenocarcinoma , Ephrins , Humans , Ephrins/metabolism , Receptors, Eph Family/metabolism , Signal Transduction/physiology , Pancreas/metabolism
16.
Dev Dyn ; 251(7): 1138-1155, 2022 07.
Article in English | MEDLINE | ID: mdl-35025117

ABSTRACT

BACKGROUND: Variation in facial shape may arise from the combinatorial or overlapping actions of paralogous genes. Given its many members, and overlapping expression and functions, the EPH receptor family is a compelling candidate source of craniofacial morphological variation. We performed a detailed morphometric analysis of an allelic series of E14.5 Ephb1-3 receptor mutants to determine the effect of each paralogous receptor gene on craniofacial morphology. RESULTS: We found that Ephb1, Ephb2, and Ephb3 genotypes significantly influenced facial shape, but Ephb1 effects were weaker than Ephb2 and Ephb3 effects. Ephb2-/- and Ephb3-/- mutations affected similar aspects of facial morphology, but Ephb3-/- mutants had additional facial shape effects. Craniofacial differences across the allelic series were largely consistent with predicted additive genetic effects. However, we identified a potentially important nonadditive effect where Ephb1 mutants displayed different morphologies depending on the combination of other Ephb paralogs present, where Ephb1+/- , Ephb1-/- , and Ephb1-/- ; Ephb3-/- mutants exhibited a consistent deviation from their predicted facial shapes. CONCLUSIONS: This study provides a detailed assessment of the effects of Ephb receptor gene paralogs on E14.5 mouse facial morphology and demonstrates how the loss of specific receptors contributes to facial dysmorphology.


Subject(s)
Ephrin-B1 , Maxillofacial Development , Receptor, EphB1 , Receptor, EphB3 , Receptors, Eph Family , Animals , Ephrin-B1/genetics , Ephrin-B1/metabolism , Face , Mice , Mutation , Receptor, EphB1/genetics , Receptor, EphB2/genetics , Receptor, EphB3/genetics , Receptors, Eph Family/metabolism
17.
Eur J Neurosci ; 56(9): 5532-5546, 2022 11.
Article in English | MEDLINE | ID: mdl-34989046

ABSTRACT

Erythropoietin-producing hepatoma (Eph) receptors belong to a family of tyrosine kinase receptors that plays a pivotal role in the development of the brain. Eph can be divided broadly into two groups, namely, EphA and EphB, comprising nine and five members, respectively. In recent years, the role of EphA-4 has become increasingly apparent in the onset of Alzheimer's disease (AD). Emerging evidence suggests that EphA-4 results in synaptic dysfunction, which in turn promotes the progression of AD. Moreover, pharmacological or genetic ablation of EphA-4 in the murine model of AD can alleviate the symptoms. The current review summarizes different pathways by which EphA-4 can influence pathogenesis. Since, majority of the studies had reported the protective effect of EphA-4 inhibition during AD, designing therapeutics based on decreasing its enzymatic activity might be necessary for introducing the novel interventions. Therefore, the review described peptide and nanobodies inhibitors of EphA-4 that exhibit the potential to modulate EphA-4 and could be used as lead molecules for the targeted therapy of AD.


Subject(s)
Alzheimer Disease , Animals , Humans , Mice , Alzheimer Disease/drug therapy , Brain/metabolism , Receptors, Eph Family/chemistry , Receptors, Eph Family/metabolism
18.
Biochem J ; 478(17): 3351-3371, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34431498

ABSTRACT

EphB6 and EphA10 are two poorly characterised pseudokinase members of the Eph receptor family, which collectively serves as mediators of contact-dependent cell-cell communication to transmit extracellular cues into intracellular signals. As per their active counterparts, EphB6 and EphA10 deregulation is strongly linked to proliferative diseases. However, unlike active Eph receptors, whose catalytic activities are thought to initiate an intracellular signalling cascade, EphB6 and EphA10 are classified as catalytically dead, raising the question of how non-catalytic functions contribute to Eph receptor signalling homeostasis. In this study, we have characterised the biochemical properties and topology of the EphB6 and EphA10 intracellular regions comprising the juxtamembrane (JM) region, pseudokinase and SAM domains. Using small-angle X-ray scattering and cross-linking-mass spectrometry, we observed high flexibility within their intracellular regions in solution and a propensity for interaction between the component domains. We identified tyrosine residues in the JM region of EphB6 as EphB4 substrates, which can bind the SH2 domains of signalling effectors, including Abl, Src and Vav3, consistent with cellular roles in recruiting these proteins for downstream signalling. Furthermore, our finding that EphB6 and EphA10 can bind ATP and ATP-competitive small molecules raises the prospect that these pseudokinase domains could be pharmacologically targeted to counter oncogenic signalling.


Subject(s)
Receptors, Eph Family/chemistry , Receptors, Eph Family/metabolism , Signal Transduction/genetics , Sterile Alpha Motif/genetics , src Homology Domains/genetics , Adenosine Triphosphate/metabolism , Animals , Humans , Phosphorylation , Protein Binding , Protein Conformation, alpha-Helical , Protein Kinase Inhibitors/metabolism , Receptors, Eph Family/genetics , Recombinant Proteins/metabolism , Sf9 Cells , Spodoptera/cytology , Tyrosine/metabolism
19.
J Allergy Clin Immunol ; 147(3): 941-954, 2021 03.
Article in English | MEDLINE | ID: mdl-33039479

ABSTRACT

BACKGROUND: Asthma exacerbations are associated with heightened asthma symptoms, which can result in hospitalization in severe cases. However, the molecular immunologic processes that determine the course of an exacerbation remain poorly understood, impeding the progression of development of effective therapies. OBJECTIVE: Our aim was to identify candidate genes that are strongly associated with asthma exacerbation at a cellular level. METHODS: Subjects with asthma exacerbation and healthy control subjects were recruited, and bronchoalveolar lavage fluid was isolated from these subjects via bronchoscopy. Cells were isolated through fluorescence-activated cell sorting, and single-cell RNA sequencing was performed on enriched cell populations. RESULTS: We showed that the levels of monocytes, CD8+ T cells, and macrophages are significantly elevated in the bronchoalveolar lavage fluid of patients. A set of cytokines and intracellular transduction regulators are associated with asthma exacerbations and are shared across multiple cell clusters, forming a complicated molecular framework. An additional group of core exacerbation-associated modules is activated, including eukaryotic initiation factor 2 signaling, ephrin receptor signaling, and C-X-C chemokine receptor type 4 signaling in the subpopulations of CD8+ T cells (C1-a) and monocyte clusters (C7 clusters), which are associated with infection. CONCLUSION: Our study identified a significant number of severe asthma-associated genes that are differentially expressed by multiple cell clusters.


Subject(s)
Asthma/genetics , CD8-Positive T-Lymphocytes/immunology , Lung/physiology , Macrophages/immunology , Monocytes/immunology , Adult , Asthma/immunology , Cells, Cultured , Disease Progression , Eukaryotic Initiation Factor-2/genetics , Eukaryotic Initiation Factor-2/metabolism , Female , Gene Expression Profiling , Humans , Male , Middle Aged , Phenotype , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Receptors, Eph Family/genetics , Receptors, Eph Family/metabolism , Sequence Analysis, RNA , Signal Transduction , Single-Cell Analysis
20.
Int J Mol Sci ; 23(5)2022 Mar 02.
Article in English | MEDLINE | ID: mdl-35269901

ABSTRACT

The EPH/ephrin system constitutes a bidirectional signaling pathway comprised of a family of tyrosine kinase receptors in tandem with their plasma membrane-bound ligand (ephrins). Its significance in a wide variety of physiologic and pathologic processes has been recognized during the past decades. In carcinogenesis, EPH/ephrins coordinate a wide spectrum of pathologic processes, such as angiogenesis, vessel infiltration, and metastasis. Despite the recent advances in colorectal cancer (CRC) diagnosis and treatment, it remains a leading cause of death globally, accounting for 9.2% of all cancer deaths. A growing body of literature has been published lately revitalizing our scientific interest towards the role of EPH/ephrins in pathogenesis and the treatment of CRC. The aim of the present review is to present the recent CRC data which might lead to clinical practice changes in the future.


Subject(s)
Colorectal Neoplasms , Ephrins , Ephrins/metabolism , Humans , Neovascularization, Pathologic , Receptors, Eph Family/metabolism , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL