Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
Add more filters










Publication year range
1.
eNeuro ; 10(11)2023 Nov.
Article in English | MEDLINE | ID: mdl-37852780

ABSTRACT

It has been suggested that stochasticity acts in the formation of topographically ordered maps in the visual system through the opposing chemoaffinity and neural activity forces acting on the innervating nerve fibers being held in an unstable equilibrium. Evidence comes from the Islet2-EphA3 knock-in mouse, in which ∼50% of the retinal ganglion cells, distributed across the retina, acquire the EphA3 receptor, thus having an enhanced density of EphA which specifies retinotopic order along the rostrocaudal (RC) axis of the colliculus. Sampling EphA3 knock-in maps in heterozygotes at different positions along the mediolateral (ML) extent of the colliculus had found single 1D maps [as in wild types (WTs)], double maps (as in homozygous knock-ins) or both single and double maps. We constructed full 2D maps from the same mouse dataset. We found either single maps or maps where the visual field projects rostrally, with a part-projection more caudally to form a double map, the extent and location of this duplication varying considerably. Contrary to previous analyses, there was no strict demarcation between heterozygous and homozygous maps. These maps were replicated in a computational model where, as the level of EphA3 was increased, there was a smooth transition from single to double maps. Our results suggest that the diversity in these retinotopic maps has its origin in a variability over the retina in the effective amount of EphA3, such as through variability in gene expression or the proportion of EphA3+ retinal ganglion cells, rather than the result of competing mechanisms acting at the colliculus.


Subject(s)
Superior Colliculi , Visual Pathways , Mice , Animals , Receptor, EphA3/genetics , Receptor, EphA3/metabolism , Superior Colliculi/metabolism , Visual Pathways/physiology , Retina/metabolism , Retinal Ganglion Cells/metabolism
2.
Sci Rep ; 12(1): 3840, 2022 03 09.
Article in English | MEDLINE | ID: mdl-35264657

ABSTRACT

The EPHA3 protein tyrosine kinase, a member of the ephrin receptor family, regulates cell fate, cell motility, and cell-cell interaction. These cellular events are critical for tissue development, immunological responses, and the processes of tumorigenesis. Earlier studies revealed that signaling via the STK4-encoded MST1 serine-threonine protein kinase, a core component of the Hippo pathway, attenuated EPHA3 expression. Here, we investigated the mechanism by which MST1 regulates EPHA3. Our findings have revealed that the transcriptional regulators YAP1 and TEAD1 are crucial activators of EPHA3 transcription. Silencing YAP1 and TEAD1 suppressed the EPHA3 protein and mRNA levels. In addition, we identified putative TEAD enhancers in the distal EPHA3 promoter, where YAP1 and TEAD1 bind and promote EPHA3 expression. Furthermore, EPHA3 knockout by CRISPR/Cas9 technology reduced cell-cell interaction and cell motility. These findings demonstrate that EPHA3 is transcriptionally regulated by YAP1/TEAD1 of the Hippo pathway, suggesting that it is sensitive to cell contact-dependent interactions.


Subject(s)
Adaptor Proteins, Signal Transducing , YAP-Signaling Proteins , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Protein Serine-Threonine Kinases/genetics , Receptor, EphA3/genetics , Receptor, EphA3/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
3.
JCI Insight ; 6(11)2021 06 08.
Article in English | MEDLINE | ID: mdl-33945505

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is characterized by aberrant repair that diminishes lung function via mechanisms that remain poorly understood. CC chemokine receptor (CCR10) and its ligand CCL28 were both elevated in IPF compared with normal donors. CCR10 was highly expressed by various cells from IPF lungs, most notably stage-specific embryonic antigen-4-positive mesenchymal progenitor cells (MPCs). In vitro, CCL28 promoted the proliferation of CCR10+ MPCs while CRISPR/Cas9-mediated targeting of CCR10 resulted in the death of MPCs. Following the intravenous injection of various cells from IPF lungs into immunodeficient (NOD/SCID-γ, NSG) mice, human CCR10+ cells initiated and maintained fibrosis in NSG mice. Eph receptor A3 (EphA3) was among the highest expressed receptor tyrosine kinases detected on IPF CCR10+ cells. Ifabotuzumab-targeted killing of EphA3+ cells significantly reduced the numbers of CCR10+ cells and ameliorated pulmonary fibrosis in humanized NSG mice. Thus, human CCR10+ cells promote pulmonary fibrosis, and EphA3 mAb-directed elimination of these cells inhibits lung fibrosis.


Subject(s)
Idiopathic Pulmonary Fibrosis/metabolism , Mesenchymal Stem Cells/metabolism , Receptor, EphA3/metabolism , Receptors, CCR10/metabolism , Alveolar Epithelial Cells/metabolism , Animals , Antibodies, Monoclonal/pharmacology , CRISPR-Cas Systems , Chemokines, CC/metabolism , Fibroblasts/metabolism , Gene Knockout Techniques , Humans , Idiopathic Pulmonary Fibrosis/pathology , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred NOD , Mice, SCID
4.
Biomolecules ; 11(4)2021 04 18.
Article in English | MEDLINE | ID: mdl-33919657

ABSTRACT

EPHA3, a member of the EPH family, is overexpressed in various cancers. We demonstrated previously that EPHA3 is associated with radiation resistance in head and neck cancer via the PTEN/Akt/EMT pathway; the inhibition of EPHA3 significantly enhances the efficacy of radiotherapy in vitro and in vivo. In this study, we investigated the mechanisms of PTEN regulation through EPHA3-related signaling. Increased DNA methyltransferase 1 (DNMT1) and enhancer of zeste homolog 2 (EZH2) levels, along with increased histone H3 lysine 27 trimethylation (H3K27me3) levels, correlated with decreased levels of PTEN in radioresistant head and neck cancer cells. Furthermore, PTEN is regulated in two ways: DNMT1-mediated DNA methylation, and EZH2-mediated histone methylation through EPHA3/C-myc signaling. Our results suggest that EPHA3 could display a novel regulatory mechanism for the epigenetic regulation of PTEN in radioresistant head and neck cancer cells.


Subject(s)
Epigenetic Repression , Head and Neck Neoplasms/genetics , PTEN Phosphohydrolase/genetics , Radiation Tolerance , Receptor, EphA3/genetics , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferase 1/genetics , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , DNA Methylation , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/metabolism , Head and Neck Neoplasms/metabolism , Head and Neck Neoplasms/radiotherapy , Histone Code , Humans , PTEN Phosphohydrolase/metabolism , Receptor, EphA3/metabolism
5.
Mol Pharm ; 18(3): 915-927, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33417456

ABSTRACT

Glioblastoma multiforme (GBM) is a highly lethal and aggressive tumor of the brain that carries a poor prognosis. Temozolomide (TMZ) has been widely used as a first-line treatment for GBM. However, poor brain targeting, side effects, and drug resistance limit its application for the treatment of GBM. We designed a Temozolomide-conjugated gold nanoparticle functionalized with an antibody against the ephrin type-A receptor 3 (anti-EphA3-TMZ@GNPs) for targeted GBM therapy via intranasal administration. The system can bypass the blood-brain barrier and target active glioma cells to improve the glioma targeting of TMZ and enhance the treatment efficacy, while reducing the peripheral toxicity and drug resistance. The prepared anti-EphA3-TMZ@GNPs were 46.12 ± 2.0 nm and suitable for intranasal administration, which demonstrated high safety to the nasal mucosa in a toxicity assay. In vitro studies showed that anti-EphA3-TMZ@GNPs exhibited significantly enhanced cellular uptake and toxicity, and a higher cell apoptosis ratio has been seen compared with that of TMZ (54.9 and 14.1%, respectively) toward glioma cells (C6). The results from experiments on TMZ-resistant glioma cells (T98G) demonstrated that the IC50 of anti-EphA3-TMZ@GNPs (64.06 ± 0.16 µM) was 18.5-fold lower than that of TMZ. In addition, Western blot analysis also revealed that anti-EphA3-TMZ@GNPs effectively down-modulated expression of O6-methylguanine-DNA methyltransferase and increased chemosensitivity of T98G to TMZ. The antiglioma efficacy in vivo was investigated in orthotopic glioma-bearing rats, and the results demonstrated that the anti-EphA3-TMZ@GNPs prolonged the median survival time to 42 days and increased tumor-cell apoptosis dramatically compared with TMZ. In conclusion, anti-EphA3-TMZ@GNPs could serve as an intranasal drug delivery system for efficacious treatment of GBM.


Subject(s)
Brain Neoplasms/drug therapy , Glioblastoma/drug therapy , Glioma/drug therapy , Gold/chemistry , Metal Nanoparticles/chemistry , Receptor, EphA3/metabolism , Temozolomide/pharmacology , Administration, Intranasal/methods , Animals , Apoptosis/drug effects , Brain Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Glioblastoma/metabolism , Glioma/metabolism , Humans , Male , Rats , Rats, Sprague-Dawley , Xenograft Model Antitumor Assays/methods
6.
Mol Biol Rep ; 47(7): 5523-5533, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32621117

ABSTRACT

The erythropoietin-producing human hepatocellular (Eph) receptors are transmembrane glycoprotein members of the tyrosine kinase receptors family. The Ephs may bind to various ephrin ligands resulting in the phosphorylation of their tyrosine kinase domain and the activation of the Eph receptor. In this review we focus on EphA3, one receptor of the 14 different Ephs, as it carries out both redundant and restricted functions in the germline development of mammals and in the maintenance of various adult tissues. The loss of EphA3 regulation is correlated with various human malignancies, the most notable being cancer. This receptor is overexpressed and/or mutated in multiple tumors, and is also associated with poor prognosis and decreased survival in patients. Here we highlight the role of EphA3 in normal and malignant tissues that are specific to cancer; these include hematologic disorders, gastric cancer, glioblastoma multiforme, colorectal cancer, lung cancer, renal cell carcinoma, and prostate cancer. Moreover, various anticancer agents against EphA3 have been developed to either inhibit its kinase domain activity or to function as agonists. Thus, we examine the most potent small molecule drugs and mAb-based therapeutics against EphA3 that are currently in pre-clinical or clinical stages.


Subject(s)
Neoplasms/genetics , Receptor, EphA3/genetics , Receptor, EphA3/metabolism , Animals , Antineoplastic Agents/pharmacology , Carcinoma, Renal Cell , Colorectal Neoplasms , Female , Gene Expression Regulation, Neoplastic/drug effects , Glioblastoma , Humans , Kidney Neoplasms , Lung Neoplasms , Male , Neoplasms/drug therapy , Phosphorylation , Prostatic Neoplasms , Protein Binding , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism
7.
BMC Cancer ; 20(1): 154, 2020 Feb 24.
Article in English | MEDLINE | ID: mdl-32093644

ABSTRACT

BACKGROUND: Cancer recurrence is one of the most concerning clinical problems of cholangiocarcinoma (CCA) patients after treatment. However, an identification of predictive factor on Opisthorchis viverrini (OV)-associated CCA recurrence is not well elucidated. In the present study, we aimed to investigate the correlation of twelve targeted protein kinases with CCA recurrence. METHODS: Twelve protein kinases, epidermal growth factor receptor (EGFR), human epidermal growth factor receptor 2, 3, 4 (HER2, HER3, HER4), vascular endothelial growth factor receptor 3 (VEGFR3), vascular endothelial growth factor-C (VEGF-C), erythropoietin-producing hepatocellular carcinoma receptor type-A3 (EphA3), EphrinA1, phosphor-serine/threonine kinase 1 (p-Akt1), serine/threonine kinase 1 (Akt1), beta-catenin and protein Wnt5a (Wnt5a) were examined using immunohistochemistry. Pre-operative serum tumor markers, CA19-9 and CEA were also investigated. RESULTS: Among twelve protein kinases, EGFR, HER4, and EphA3 were associated with tumor recurrence status, recurrence-free survival (RFS) and overall survival (OS). Multivariate cox regression demonstrated that EGFR, HER4, EphA3 or the panel of high expression of these proteins was an independent prognostic factor for tumor recurrence. The combination of high expression of these proteins with a high level of CA19-9 could improve the predictive ability on tumor recurrence. Moreover, the patients were stratified more accurately when analyzed using the combination of high expression of these proteins with primary tumor (T) or lymph node metastasis (N) status. CONCLUSION: EGFR, HER4, EphA3 or the panel of high expression of these proteins is an independent prognostic factor for post-operative CCA recurrence.


Subject(s)
Bile Duct Neoplasms/surgery , Biomarkers, Tumor/metabolism , Cholangiocarcinoma/surgery , Neoplasm Recurrence, Local/diagnosis , Postoperative Complications/diagnosis , Animals , Bile Duct Neoplasms/metabolism , Bile Duct Neoplasms/pathology , CA-19-9 Antigen/blood , Cholangiocarcinoma/metabolism , Cholangiocarcinoma/pathology , ErbB Receptors/metabolism , Female , Humans , Incidence , Lymphatic Metastasis , Male , Neoplasm Recurrence, Local/epidemiology , Neoplasm Recurrence, Local/metabolism , Neoplasm Staging , Postoperative Complications/epidemiology , Postoperative Complications/metabolism , Progression-Free Survival , Receptor, EphA3/metabolism , Receptor, ErbB-4/metabolism , Thailand/epidemiology
8.
Nat Commun ; 10(1): 5686, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31831727

ABSTRACT

Diabetes mellitus affects one in eleven adults worldwide. Most suffer from Type 2 Diabetes which features elevated blood glucose levels and an inability to adequately secrete or respond to insulin. Insulin producing ß-cells have primary cilia which are implicated in the regulation of glucose metabolism, insulin signaling and secretion. To better understand how ß-cell cilia affect glucose handling, we ablate cilia from mature ß-cells by deleting key cilia component Ift88. Here we report that glucose homeostasis and insulin secretion deteriorate over 12 weeks post-induction. Cilia/basal body components are required to suppress spontaneous auto-activation of EphA3 and hyper-phosphorylation of EphA receptors inhibits insulin secretion. In ß-cells, loss of cilia/basal body function leads to polarity defects and epithelial-to-mesenchymal transition. Defective insulin secretion from IFT88-depleted human islets and elevated pEPHA3 in islets from diabetic donors both point to a role for cilia/basal body proteins in human glucose homeostasis.


Subject(s)
Cilia/metabolism , Diabetes Mellitus, Type 2/metabolism , Endosomes/metabolism , Glucose/metabolism , Homeostasis , Insulin-Secreting Cells/metabolism , Receptors, Eph Family/metabolism , Aged , Animals , Blood Glucose , Glucose Tolerance Test , Guanine Nucleotide Exchange Factors , Humans , Insulin/metabolism , Insulin Secretion , Islets of Langerhans/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neuropeptides/metabolism , Phosphorylation , Receptor, EphA3/genetics , Receptor, EphA3/metabolism , Signal Transduction , T-Lymphoma Invasion and Metastasis-inducing Protein 1/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , rac1 GTP-Binding Protein/metabolism
9.
Elife ; 82019 10 02.
Article in English | MEDLINE | ID: mdl-31577226

ABSTRACT

EphA/ephrin signaling regulates axon growth and guidance of neurons, but whether this process occurs also independently of ephrins is unclear. We show that presenilin-1 (PS1)/γ-secretase is required for axon growth in the developing mouse brain. PS1/γ-secretase mediates axon growth by inhibiting RhoA signaling and cleaving EphA3 independently of ligand to generate an intracellular domain (ICD) fragment that reverses axon defects in PS1/γ-secretase- and EphA3-deficient hippocampal neurons. Proteomic analysis revealed that EphA3 ICD binds to non-muscle myosin IIA (NMIIA) and increases its phosphorylation (Ser1943), which promotes NMIIA filament disassembly and cytoskeleton rearrangement. PS1/γ-secretase-deficient neurons show decreased phosphorylated NMIIA and NMIIA/actin colocalization. Moreover, pharmacological NMII inhibition reverses axon retraction in PS-deficient neurons suggesting that NMIIA mediates PS/EphA3-dependent axon elongation. In conclusion, PS/γ-secretase-dependent EphA3 cleavage mediates axon growth by regulating filament assembly through RhoA signaling and NMIIA, suggesting opposite roles of EphA3 on inhibiting (ligand-dependent) and promoting (receptor processing) axon growth in developing neurons.


Subject(s)
Axons/physiology , Nonmuscle Myosin Type IIA/metabolism , Presenilin-1/metabolism , Receptor, EphA3/metabolism , Animals , Cells, Cultured , Humans , Mice , Signal Transduction , rhoA GTP-Binding Protein/metabolism
10.
Biosci Rep ; 39(7)2019 07 31.
Article in English | MEDLINE | ID: mdl-31262977

ABSTRACT

Background: Endometriosis is a chronic fibrotic disease characterized by agonizing pelvic pain and low fertility, mainly affecting middle-aged women. The aim of the present study is to assess the potential effects of erythropoietin-producing hepatocellular carcinoma A3 (EPHA3) on endometriosis, with emphasis on the autophagy and apoptosis of macrophages via inhibition of the mammalian target of rapamycin (mTOR) signaling pathway.Methods: The mouse models of endometriosis were established followed by culturing the macrophages and macrophage transfection via the EPHA3 vector, siRNA EPHA3, and RAPA (an inhibitor of the mTOR signaling pathway). The expression of EPHA3, related factors in the mTOR signaling pathway, macrophage autophagy (autophagy-related gene 3 (Atg3), light chain 3-I (LC3-I), light chain 3-II (LC3-II) and Beclin1) and apoptosis (B-cell lymphoma-2 (bcl-2), bax and fas) were all detected and documented, respectively. The changes of autophagic lysosomes and the apoptosis of macrophages in each group following transfection were also inspected and detected.Results: The results of the in silico analysis ascertained EPHA3 to be a candidate gene of endometriosis. After successful modeling, the uterine tissues of endometriosis mice presented with a low expression of EPHA3 and activated mTOR signaling pathway. Overexpression of EPHA3 inhibited the activation of the mTOR signaling pathway, down-regulated bcl-2 expression, up-regulated the expression of Atg3, LC3-II/LC3-I, Beclin1, bax and fas, and also promoted the autophagy and apoptosis of macrophages in endometriosis mice.Conclusion: Altogether, EPHA3 could potentially promote the autophagy and apoptosis of macrophages in endometriosis via inhibition of the mTOR signaling pathway, highlighting the potential of EPHA3 as the target to treat endometriosis.


Subject(s)
Apoptosis , Autophagic Cell Death , Endometriosis/metabolism , Macrophages/metabolism , Receptor, EphA3/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Animals , Disease Models, Animal , Endometriosis/genetics , Endometriosis/pathology , Female , Macrophages/pathology , Mice , Mice, Inbred BALB C , Receptor, EphA3/genetics , TOR Serine-Threonine Kinases/genetics
11.
Anticancer Res ; 38(3): 1685-1693, 2018 03.
Article in English | MEDLINE | ID: mdl-29491103

ABSTRACT

BACKGROUND/AIM: Erythropoietin-producing hepatocellular carcinoma receptor A (EphA) is associated with angiogenesis and invasive tumor progression. In this study, we evaluated the EphA1-4 expression levels in advanced gastric cancer. PATIENTS AND METHODS: Tumor tissues obtained from 114 patients with advanced gastric adenocarcinoma who underwent gastrectomy were analyzed. In addition, the impact of EPHA 1-4 mRNA expression on survival was analyzed using the Kaplan-Meier plotter database on the website. RESULTS: High EphA 1, 2, and 4 expression levels were significantly related to recurrence (p<0.01, p=0.04, and p<0.01). Both high EphA 1 and 4 expression levels were independent predictors of relapse-free interval (hazard ratio [HR]=2.0, p=0.03; HR=2.4, p=0.03) and disease-specific survival (HR=2.0, 95% p=0.03; HR=2.5, p=0.02) on multivariate analysis. In the Kaplan-Meier plotter database, high EPHA2 mRNA expression was significantly associated with poor survival in patients with gastric cancer (p=0.0098), and high expression levels of EPHA1 and 4 tended to be associated with poor survival (p=0.050, p=0.052). CONCLUSION: EphA 1, 2, and 4 may play key roles in recurrence and survival in patients with advanced gastric cancer.


Subject(s)
Adenocarcinoma/genetics , Gene Expression Regulation, Neoplastic , Receptor, EphA1/genetics , Receptor, EphA2/genetics , Receptor, EphA3/genetics , Receptor, EphA4/genetics , Stomach Neoplasms/genetics , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Aged , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Multivariate Analysis , Neoplasm Recurrence, Local , Outcome Assessment, Health Care/methods , Outcome Assessment, Health Care/statistics & numerical data , Prognosis , Proportional Hazards Models , Receptor, EphA1/metabolism , Receptor, EphA2/metabolism , Receptor, EphA3/metabolism , Receptor, EphA4/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery
12.
Front Neural Circuits ; 11: 73, 2017.
Article in English | MEDLINE | ID: mdl-29066954

ABSTRACT

Throughout the visual system, different subtypes of neurons are tuned to distinct aspects of the visual scene, establishing parallel circuits. Defining the mechanisms by which such tuning arises has been a long-standing challenge for neuroscience. To investigate this, we have focused on the retina's projection to the superior colliculus (SC), where multiple visual neuron subtypes have been described. The SC receives inputs from a variety of retinal ganglion cell (RGC) subtypes; however, which RGCs drive the tuning of different SC neurons remains unclear. Here, we pursued a genetic approach that allowed us to determine the tuning properties of neurons innervated by molecularly defined subpopulations of RGCs. In homozygous Islet2-EphA3 knock-in (Isl2EA3/EA3) mice, Isl2+ and Isl2- RGCs project to non-overlapping sub-regions of the SC. Based on molecular and anatomic data, we show that significantly more Isl2- RGCs are direction-selective (DS) in comparison with Isl2+ RGCs. Targeted recordings of visual responses from each SC sub-region in Isl2EA3/EA3 mice revealed that Isl2- RGC-innervated neurons were significantly more DS than those innervated by Isl2+ RGCs. Axis-selective (AS) neurons were found in both sub-regions, though AS neurons innervated by Isl2+ RGCs were more tightly tuned. Despite this segregation, DS and AS neurons innervated by Isl2+ or Isl2- RGCs did not differ in their spatial summation or spatial frequency (SF) tuning. Further, we did not observe alterations in receptive field (RF) size or structure of SC neurons innervated by Isl2+ or Isl2- RGCs. Together, these data show that innervation by Isl2+ and Isl2- RGCs results in distinct tuning in the SC and set the stage for future studies investigating the mechanisms by which these circuits are built.


Subject(s)
LIM-Homeodomain Proteins/metabolism , Retinal Ganglion Cells/cytology , Retinal Ganglion Cells/metabolism , Superior Colliculi/cytology , Superior Colliculi/physiology , Transcription Factors/metabolism , Visual Perception/physiology , Action Potentials , Animals , Immunohistochemistry , LIM-Homeodomain Proteins/genetics , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Transgenic , Microelectrodes , Neuroanatomical Tract-Tracing Techniques , Photic Stimulation , Receptor, EphA3/genetics , Receptor, EphA3/metabolism , Receptors, Thyrotropin-Releasing Hormone/genetics , Receptors, Thyrotropin-Releasing Hormone/metabolism , Transcription Factors/genetics , Visual Pathways/cytology , Visual Pathways/physiology
13.
Cell Oncol (Dordr) ; 40(5): 483-496, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28721629

ABSTRACT

PURPOSE: Multiple myeloma (MM) is a hematologic malignancy characterized by a clonal expansion of plasma cells (PCs) in the bone marrow (BM). Since MM has so far remained incurable, further insights into its pathogenesis and the concomitant identification of new therapeutic targets are urgently needed. The tyrosine kinase receptor EphA3 is known to be involved in various cellular processes including cell viability, cell movement and cell-cell interactions. Recently, EphA3 has emerged as a potential therapeutic target in several hematologic and solid tumors. Here, we aimed to uncover the role of EphA3 in MM. METHODS: EphA3 mRNA and protein expression in primary MM bone marrow plasma cells (BMPCs), in MM-derived cell lines and in healthy controls (HCs) was assessed using qRT-PCR, Western blotting and flow cytometry. The effects of siRNA-mediated EphA3 silencing and anti EphA3 antibody (EphA3mAb) treatment on MM PC trafficking and viability were evaluated using in vitro assays. The effects of EphA3mAb treatment were also assessed in two MM-derived mouse xenograft models. RESULTS: We found that EphA3 was overexpressed in primary MM BMPCs and MM-derived cell lines compared to HCs. We also found that siRNA-mediated EphA3 silencing and EphA3mAb treatment significantly inhibited the ability of MM PCs to adhere to fibronectin and stromal cells and to invade in vitro, without affecting cell proliferation and viability. Gene expression profiling showed that EphA3 silencing resulted in expression modulation of several molecules that regulate adhesion, migration and invasion processes. Importantly, we found that EphA3mAb treatment significantly inhibited in vivo tumor growth and angiogenesis in two MM-derived mouse xenograft models. CONCLUSIONS: Our findings suggest that EphA3 plays an important role in the pathogenesis of MM and provide support for the notion that its targeting may represent a novel therapeutic opportunity for MM.


Subject(s)
Cell Movement/genetics , Multiple Myeloma/genetics , Neovascularization, Pathologic/genetics , Receptor, EphA3/genetics , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Cell Adhesion/genetics , Cell Line, Tumor , Cells, Cultured , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Humans , Mice, Inbred NOD , Mice, SCID , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Neovascularization, Pathologic/metabolism , RNA Interference , Receptor, EphA3/immunology , Receptor, EphA3/metabolism , Xenograft Model Antitumor Assays
14.
Leukemia ; 31(8): 1779-1787, 2017 08.
Article in English | MEDLINE | ID: mdl-27922598

ABSTRACT

The human EphA3 gene was discovered in a pre-B acute lymphoblastic leukemia (pre-B-ALL) using the EphA3-specific monoclonal antibody (mAb), IIIA4, which binds and activates both human and mouse EphA3. We use two models of human pre-B-ALL to examine EphA3 function, demonstrating effects on pre-B-cell receptor signaling. In therapeutic targeting studies, we demonstrated antitumor effects of the IIIA4 mAb in EphA3-expressing leukemic xenografts and no antitumor effect in the xenografts with no EphA3 expression providing evidence that EphA3 is a functional therapeutic target in pre-B-ALL. Here we show that the therapeutic effect of the anti-EphA3 antibody was greatly enhanced by adding an α-particle-emitting 213Bismuth payload.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Receptor, EphA3/immunology , Animals , Bismuth , Cell Line, Tumor , Humans , Immunotherapy , Mice , Receptor, EphA3/metabolism , Xenograft Model Antitumor Assays
15.
J Biol Chem ; 291(51): 26262-26272, 2016 Dec 16.
Article in English | MEDLINE | ID: mdl-27803162

ABSTRACT

Establishment of a proper balance of excitatory and inhibitory connectivity is achieved during development of cortical networks and adjusted through synaptic plasticity. The neural cell adhesion molecule (NCAM) and the receptor tyrosine kinase EphA3 regulate the perisomatic synapse density of inhibitory GABAergic interneurons in the mouse frontal cortex through ephrin-A5-induced growth cone collapse. In this study, it was demonstrated that binding of NCAM and EphA3 occurred between the NCAM Ig2 domain and EphA3 cysteine-rich domain (CRD). The binding interface was further refined through molecular modeling and mutagenesis and shown to be comprised of complementary charged residues in the NCAM Ig2 domain (Arg-156 and Lys-162) and the EphA3 CRD (Glu-248 and Glu-264). Ephrin-A5 induced co-clustering of surface-bound NCAM and EphA3 in GABAergic cortical interneurons in culture. Receptor clustering was impaired by a charge reversal mutation that disrupted NCAM/EphA3 association, emphasizing the importance of the NCAM/EphA3 binding interface for cluster formation. NCAM enhanced ephrin-A5-induced EphA3 autophosphorylation and activation of RhoA GTPase, indicating a role for NCAM in activating EphA3 signaling through clustering. NCAM-mediated clustering of EphA3 was essential for ephrin-A5-induced growth cone collapse in cortical GABAergic interneurons, and RhoA and a principal effector, Rho-associated protein kinase, mediated the collapse response. This study delineates a mechanism in which NCAM promotes ephrin-A5-dependent clustering of EphA3 through interaction of the NCAM Ig2 domain and the EphA3 CRD, stimulating EphA3 autophosphorylation and RhoA signaling necessary for growth cone repulsion in GABAergic interneurons in vitro, which may extend to remodeling of axonal terminals of interneurons in vivo.


Subject(s)
GABAergic Neurons/metabolism , Growth Cones/metabolism , Neural Cell Adhesion Molecules/metabolism , Receptor, EphA3/metabolism , Signal Transduction/physiology , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases/metabolism , Animals , Ephrin-A5/genetics , Ephrin-A5/metabolism , Mice , Mice, Mutant Strains , Neural Cell Adhesion Molecules/genetics , Phosphorylation/physiology , rho GTP-Binding Proteins/genetics , rho-Associated Kinases/genetics , rhoA GTP-Binding Protein
16.
J Comp Pathol ; 154(2-3): 180-5, 2016.
Article in English | MEDLINE | ID: mdl-26895888

ABSTRACT

Ephrin A3 (EphA3), a member of the ephrin receptor tyrosine kinase family, is involved in a variety of functions in normal cells, especially during embryonic development, and alterations in its expression profile have been observed in several human cancers. However, there are no reports of the expression of EphA3 in normal, hyperplastic or neoplastic canine prostate tissue or in other types of canine tumours. Six normal, 15 hyperplastic and 21 neoplastic canine prostates were examined immunohistochemically with a polyclonal antibody specific for human EphA3. The percentage of positive cells in all prostatic carcinomas was increased, with a mean of 89.28 ± 5.18% compared with normal (9.17 ± 6.72%) and hyperplastic prostates (20.00 ± 8.28%). EphA3 expression was not correlated with the histological subtypes of prostate cancer or with the Gleason score. The increase in EphA3 expression in canine prostatic carcinomas suggests the involvement of this receptor in prostatic carcinogenesis and its potential use as a target for new therapeutic strategies.


Subject(s)
Adenocarcinoma/veterinary , Dog Diseases/metabolism , Prostatic Neoplasms/veterinary , Receptor, EphA3/metabolism , Adenocarcinoma/metabolism , Animals , Dogs , Humans , Immunohistochemistry , Male , Prostatic Neoplasms/metabolism , Receptor, EphA3/analysis
17.
J Cell Biol ; 211(5): 1077-91, 2015 Dec 07.
Article in English | MEDLINE | ID: mdl-26644518

ABSTRACT

Each adult mammalian skeletal muscle has a unique complement of fast and slow myofibers, reflecting patterns established during development and reinforced via their innervation by fast and slow motor neurons. Existing data support a model of postnatal "matching" whereby predetermined myofiber type identity promotes pruning of inappropriate motor axons, but no molecular mechanism has yet been identified. We present evidence that fiber type-specific repulsive interactions inhibit innervation of slow myofibers by fast motor axons during both postnatal maturation of the neuromuscular junction and myofiber reinnervation after injury. The repulsive guidance ligand ephrin-A3 is expressed only on slow myofibers, whereas its candidate receptor, EphA8, localizes exclusively to fast motor endplates. Adult mice lacking ephrin-A3 have dramatically fewer slow myofibers in fast and mixed muscles, and misexpression of ephrin-A3 on fast myofibers followed by denervation/reinnervation promotes their respecification to a slow phenotype. We therefore conclude that Eph/ephrin interactions guide the fiber type specificity of neuromuscular interactions during development and adult life.


Subject(s)
Muscle, Skeletal/growth & development , Muscle, Skeletal/innervation , Neurogenesis/physiology , Receptor, EphA3/metabolism , Animals , Axons/physiology , Female , Gene Expression Regulation, Developmental , Immunohistochemistry , Ligands , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Motor Neurons/physiology , Muscle, Skeletal/embryology , Myofibrils/metabolism , Neuromuscular Junction/physiology , Neuronal Plasticity , Phenotype , Receptor, EphA8/metabolism , Schwann Cells/metabolism , Sciatic Nerve/physiology
18.
PLoS One ; 10(5): e0127081, 2015.
Article in English | MEDLINE | ID: mdl-25993310

ABSTRACT

The Eph receptor tyrosine kinase/ephrin ligand system regulates a wide spectrum of physiological processes, while its dysregulation has been implicated in cancer progression. The human EphA3 receptor is widely upregulated in the tumor microenvironment and is highly expressed in some types of cancer cells. Furthermore, EphA3 is among the most highly mutated genes in lung cancer and it is also frequently mutated in other cancers. We report the structure of the ligand-binding domain of the EphA3 receptor in complex with its preferred ligand, ephrin-A5. The structure of the complex reveals a pronounced tilt of the ephrin-A5 ligand compared to its orientation when bound to the EphA2 and EphB2 receptors and similar to its orientation when bound to EphA4. This tilt brings an additional area of ephrin-A5 into contact with regions of EphA3 outside the ephrin-binding pocket thereby enlarging the size of the interface, which is consistent with the high binding affinity of ephrin-A5 for EphA3. This large variation in the tilt of ephrin-A5 bound to different Eph receptors has not been previously observed for other ephrins.


Subject(s)
Ephrin-A5/chemistry , Ephrin-A5/metabolism , Receptor, EphA3/chemistry , Receptor, EphA3/metabolism , Binding Sites , Calorimetry , Crystallography, X-Ray , Humans , Models, Molecular , Protein Binding , Protein Structure, Secondary , Protein Structure, Tertiary , Surface Properties , Thermodynamics
19.
Dis Model Mech ; 8(4): 393-401, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25713296

ABSTRACT

Treatment of non-small cell lung cancer (NSCLC) is based on histological analysis and molecular profiling of targetable driver oncogenes. Therapeutic responses are further defined by the landscape of passenger mutations, or loss of tumor suppressor genes. We report here a thorough study to address the physiological role of the putative lung cancer tumor suppressor EPH receptor A3 (EPHA3), a gene that is frequently mutated in human lung adenocarcinomas. Our data shows that homozygous or heterozygous loss of EphA3 does not alter the progression of murine adenocarcinomas that result from Kras mutation or loss of Trp53, and we detected negligible postnatal expression of EphA3 in adult wild-type lungs. Yet, EphA3 was expressed in the distal mesenchyme of developing mouse lungs, neighboring the epithelial expression of its Efna1 ligand; this is consistent with the known roles of EPH receptors in embryonic development. However, the partial loss of EphA3 leads only to subtle changes in epithelial Nkx2-1, endothelial Cd31 and mesenchymal Fgf10 RNA expression levels, and no macroscopic phenotypic effects on lung epithelial branching, mesenchymal cell proliferation, or abundance and localization of CD31-positive endothelia. The lack of a discernible lung phenotype in EphA3-null mice might indicate lack of an overt role for EPHA3 in the murine lung, or imply functional redundancy between EPHA receptors. Our study shows how biological complexity can challenge in vivo functional validation of mutations identified in sequencing efforts, and provides an incentive for the design of knock-in or conditional models to assign the role of EPHA3 mutation during lung tumorigenesis.


Subject(s)
Carcinogenesis/genetics , Genes, Tumor Suppressor , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Morphogenesis/genetics , Receptor, EphA3/genetics , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Animals , Carcinogenesis/pathology , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Humans , Lung/embryology , Lung/pathology , Mesoderm/metabolism , Mice , Mutation , Proto-Oncogene Proteins p21(ras)/metabolism , Receptor, EphA3/metabolism , Tumor Suppressor Protein p53/metabolism
20.
PLoS One ; 9(11): e112106, 2014.
Article in English | MEDLINE | ID: mdl-25420155

ABSTRACT

Eph and ephrin proteins are essential cell guidance cues that orchestrate cell navigation and control cell-cell interactions during developmental tissue patterning, organogenesis and vasculogenesis. They have been extensively studied in animal models of embryogenesis and adult tissue regeneration, but less is known about their expression and function during human tissue and organ regeneration. We discovered the hypoxia inducible factor (HIF)-1α-controlled expression of EphA3, an Eph family member with critical functions during human tumour progression, in the vascularised tissue of regenerating human endometrium and on isolated human endometrial multipotent mesenchymal stromal cells (eMSCs), but not in other highly vascularised human organs. EphA3 affinity-isolation from human biopsy tissue yielded multipotent CD29+/CD73+/CD90+/CD146+ eMSCs that can be clonally propagated and respond to EphA3 agonists with EphA3 phosphorylation, cell contraction, cell-cell segregation and directed cell migration. EphA3 silencing significantly inhibited the ability of transplanted eMSCs to support neovascularisation in immunocompromised mice. In accord with established roles of Eph receptors in mediating interactions between endothelial and perivascular stromal cells during mouse development, our findings suggest that HIF-1α-controlled expression of EphA3 on human MSCs functions during the hypoxia-initiated early stages of adult blood vessel formation.


Subject(s)
Mesenchymal Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Neovascularization, Physiologic , Receptor, EphA3/genetics , Adult , Animals , Blotting, Western , Cell Hypoxia , Cells, Cultured , Endometrium/cytology , Female , Gene Expression , Heterografts/blood supply , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mesenchymal Stem Cell Transplantation/methods , Mice, Inbred BALB C , Mice, Nude , Microscopy, Fluorescence , Multipotent Stem Cells/transplantation , RNA Interference , Receptor, EphA3/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...