Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Mol Biol Rep ; 47(10): 8037-8048, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32990903

ABSTRACT

The Eph (erythropoietin-producing human hepatocellular) receptors form the largest known subfamily of receptor tyrosine kinases. These receptors interact with membrane-bound ephrin ligands via direct cell-cell interactions resulting in bi-directional activation of signal pathways. Importantly, the Eph receptors play critical roles in embryonic tissue organization and homeostasis, and in the maintenance of adult processes such as long-term potentiation, angiogenesis, and stem cell differentiation. The Eph receptors also display properties of both tumor promoters and suppressors depending on the cellular context. Characterization of EphA2 receptor in regard to EphA2 dysregulation has revealed associations with various pathological processes, especially cancer. The analysis of various tumor types generally identify EphA2 receptor as overexpressed and/or mutated, and for certain types of cancers EphA2 is linked with poor prognosis and decreased patient survival. Thus, here we highlight the role of EphA2 in malignant tissues that are specific to cancer; these include glioblastoma multiforme, prostate cancer, ovarian and uterine cancers, gastric carcinoma, melanoma, and breast cancer. Due to its large extracellular domain, therapeutic targeting of EphA2 with monoclonal antibodies (mAbs), which may function as inhibitors of ligand activation or as molecular agonists, has been an oft-attempted strategy. Therefore, we review the most current mAb-based therapies against EphA2 expressing cancers currently in pre-clinical and/or clinical stages. Finally, we discuss the latest peptides and cyclical-peptides that function as selective agonists for EphA2 receptor.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Ephrin-A2/immunology , Immunotherapy , Neoplasm Proteins/immunology , Neoplasms/immunology , Neoplasms/therapy , Antineoplastic Agents, Immunological/immunology , Ephrin-A2/antagonists & inhibitors , Humans , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/pathology , Receptor, EphA2
2.
J Hematol Oncol ; 13(1): 114, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32811512

ABSTRACT

Eph receptors and the corresponding Eph receptor-interacting (ephrin) ligands jointly constitute a critical cell signaling network that has multiple functions. The tyrosine kinase EphA2, which belongs to the family of Eph receptors, is highly produced in tumor tissues, while found at relatively low levels in most normal adult tissues, indicating its potential application in cancer treatment. After 30 years of investigation, a large amount of data regarding EphA2 functions have been compiled. Meanwhile, several compounds targeting EphA2 have been evaluated and tested in clinical studies, albeit with limited clinical success. The present review briefly describes the contribution of EphA2-ephrin A1 signaling axis to carcinogenesis. In addition, the roles of EphA2 in resistance to molecular-targeted agents were examined. In particular, we focused on EphA2's potential as a target for cancer treatment to provide insights into the application of EphA2 targeting in anticancer strategies. Overall, EphA2 represents a potential target for treating malignant tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Ephrin-A2/antagonists & inhibitors , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Animals , Cancer Vaccines , Clinical Trials as Topic , Combined Modality Therapy , Dendritic Cells/immunology , Drug Delivery Systems , Enzyme Induction/drug effects , Ephrin-A1/physiology , Ephrin-A2/physiology , Humans , Immunoconjugates/therapeutic use , Immunoglobulin Fc Fragments/immunology , Immunotherapy, Adoptive , Mice , Mice, Nude , Nanocapsules , Neoplasm Proteins/physiology , Neoplasms/enzymology , Neoplasms/therapy , RNA Interference , RNA, Small Interfering/genetics , RNA, Small Interfering/therapeutic use , Receptor, EphA2 , Recombinant Fusion Proteins/metabolism , Substrate Specificity , Xenograft Model Antitumor Assays
3.
J Med Chem ; 63(8): 4107-4116, 2020 04 23.
Article in English | MEDLINE | ID: mdl-32202781

ABSTRACT

Bicycles are constrained bicyclic peptides that represent a promising binding modality for use in targeted drug conjugates. A phage display screen against EphA2, a receptor tyrosine kinase highly expressed in a number of solid tumors, identified a number of Bicycle families with low nanomolar affinity. A Bicycle toxin conjugate (BTC) was generated by derivatization of one of these Bicycles with the potent cytotoxin DM1 via a cleavable linker. This BTC demonstrated potent antitumor activity in vivo but was poorly tolerated, which was hypothesized to be the result of undesired liver uptake caused by poor physicochemical properties. Chemical optimization of a second Bicycle, guided by structural biology, provided a high affinity, metabolically stable Bicycle with improved physicochemical properties. A BTC incorporating this Bicycle also demonstrated potent antitumor activity and was very well tolerated when compared to the initial BTC. Phage display selection followed by chemical optimization of Bicycles can deliver potent drug conjugates with favorable pharmaceutical properties.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Cytotoxins/administration & dosage , Drug Delivery Systems/methods , Ephrin-A2/antagonists & inhibitors , Amino Acid Sequence , Animals , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Cytotoxins/chemistry , Cytotoxins/metabolism , Ephrin-A2/metabolism , Female , Humans , Liver/diagnostic imaging , Liver/drug effects , Liver/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Protein Structure, Secondary , Protein Structure, Tertiary , Receptor, EphA2 , Xenograft Model Antitumor Assays/methods
4.
Biochem Biophys Res Commun ; 519(4): 846-853, 2019 11 19.
Article in English | MEDLINE | ID: mdl-31558317

ABSTRACT

Small-cell lung cancer (SCLC) is characterized by one of neuroendocrine tumors, and is a clinically aggressive cancer due to its rapid growth, early dissemination, and rapid acquisition of multidrug resistance to chemotherapy. Moreover, the standard chemotherapeutic regimen in SCLC has not changed for three decades despite of the dramatic therapeutic improvement in non-SCLC. The development of a novel therapeutic strategy for SCLC has become a pressing issue. We found that expression of Eph receptor A2 (EphA2) is upregulated in three of 13 SCLC cell lines and five of 76 SCLC tumor samples. Genetic inhibition using siRNA of EphA2 significantly suppressed the cellular proliferation via induction of cell cycle arrest in SBC-5 cells. Furthermore, small molecule inhibitors of EphA2 (ALW-II-41-27 and dasatinib) also exclusively inhibited proliferation of EphA2-positive SCLC cells by the same mechanism. Collectively, EphA2 could be a promising candidate as a therapeutic target for SCLC.


Subject(s)
Antineoplastic Agents/pharmacology , Benzamides/pharmacology , Dasatinib/pharmacology , Ephrin-A2/antagonists & inhibitors , Lung Neoplasms/metabolism , Niacinamide/analogs & derivatives , Small Cell Lung Carcinoma/metabolism , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Ephrin-A2/genetics , Ephrin-A2/metabolism , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Niacinamide/pharmacology , Receptor, EphA2 , Small Cell Lung Carcinoma/drug therapy , Small Cell Lung Carcinoma/pathology , Structure-Activity Relationship , Tumor Cells, Cultured
5.
Mol Cancer Ther ; 18(4): 845-855, 2019 04.
Article in English | MEDLINE | ID: mdl-30824612

ABSTRACT

The EPHA2 tyrosine kinase receptor is implicated in tumor progression and targeted therapies resistance. We evaluated EPHA2 as a potential resistance marker to the antiepidermal growth factor receptor (EGFR) monoclonal antibody cetuximab in colorectal cancer. We studied activation of EPHA2 in a panel of human colorectal cancer cell lines sensitive or resistant to anti-EGFR drugs. The in vitro and in vivo effects of ALW-II-41-27 (an EPHA2 inhibitor) and/or cetuximab treatment were tested. Formalin-fixed paraffin-embedded tumor specimens from 82 RAS wild-type (WT) metastatic colorectal cancer patients treated with FOLFIRI + cetuximab as first-line therapy in the CAPRI-GOIM trial were assessed for EPHA2 expression by immunohistochemistry and correlated with treatment efficacy. EPHA2 was differentially activated in colorectal cancer cell lines. Combined treatment with ALW-II-41-27 plus cetuximab reverted primary and acquired resistance to cetuximab, causing cell growth inhibition, inducing apoptosis and cell-cycle G1-G2 arrest. In tumor xenograft models, upon progression to cetuximab, ALW-II-41-27 addition significantly inhibited tumor growth. EPHA2 protein expression was detected in 55 of 82 tumor samples, frequently expressed in less-differentiated and left-sided tumors. High levels of EPHA2 significantly correlated with worse progression-free survival [8.6 months; confidence interval (CI) 95%, 6.4-10.8; vs. 12.3 months; CI 95%, 10.4-14.2; P = 0.03] and with increased progression rate (29% vs. 9%, P = 0.02). A specific EPHA2 inhibitor reverts in vitro and in vivo primary and acquired resistance to anti-EGFR therapy. EPHA2 levels are significantly associated with worse outcome in patients treated with FOLFIRI + cetuximab. These results highlight EPHA2 as a potential therapeutic target in metastatic colorectal cancer.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Cetuximab/pharmacology , Cetuximab/therapeutic use , Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm , Ephrin-A2/metabolism , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Benzamides/pharmacology , Biomarkers, Tumor/metabolism , Camptothecin/analogs & derivatives , Camptothecin/therapeutic use , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cetuximab/administration & dosage , Colorectal Neoplasms/pathology , Ephrin-A2/antagonists & inhibitors , ErbB Receptors/antagonists & inhibitors , Female , Fluorouracil/therapeutic use , Humans , Leucovorin/therapeutic use , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Progression-Free Survival , RNA Interference , Receptor, EphA2 , Signal Transduction/drug effects , Transfection , Xenograft Model Antitumor Assays
6.
Cancer Res ; 78(17): 5023-5037, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29945963

ABSTRACT

Glioblastoma (GBM) carries a dismal prognosis and inevitably relapses despite aggressive therapy. Many members of the Eph receptor tyrosine kinase (EphR) family are expressed by GBM stem cells (GSC), which have been implicated in resistance to GBM therapy. In this study, we identify several EphRs that mark a therapeutically targetable GSC population in treatment-refractory, recurrent GBM (rGBM). Using a highly specific EphR antibody panel and CyTOF (cytometry by time-of-flight), we characterized the expression of all 14 EphR in primary and recurrent patient-derived GSCs to identify putative rGBM-specific EphR. EPHA2 and EPHA3 coexpression marked a highly tumorigenic cell population in rGBM that was enriched in GSC marker expression. Knockdown of EPHA2 and EPHA3 together led to increased expression of differentiation marker GFAP and blocked clonogenic and tumorigenic potential, promoting significantly higher survival in vivo Treatment of rGBM with a bispecific antibody against EPHA2/A3 reduced clonogenicity in vitro and tumorigenic potential of xenografted recurrent GBM in vivo via downregulation of AKT and ERK and increased cellular differentiation. In conclusion, we show that EPHA2 and EPHA3 together mark a GSC population in rGBM and that strategic cotargeting of EPHA2 and EPHA3 presents a novel and rational therapeutic approach for rGBM.Significance: Treatment of rGBM with a novel bispecific antibody against EPHA2 and EPHA3 reduces tumor burden, paving the way for the development of therapeutic approaches against biologically relevant targets in rGBM. Cancer Res; 78(17); 5023-37. ©2018 AACR.


Subject(s)
Ephrin-A2/genetics , Glioblastoma/genetics , Neoplasm Recurrence, Local/genetics , Receptor Protein-Tyrosine Kinases/genetics , Animals , Biomarkers, Tumor/genetics , Carcinogenesis/genetics , Cell Differentiation/genetics , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Ephrin-A2/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/genetics , Gene Knockdown Techniques , Glioblastoma/drug therapy , Glioblastoma/pathology , Glioblastoma/radiotherapy , Humans , Mice , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/radiotherapy , Neoplastic Stem Cells/pathology , Prognosis , Radiation , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor, EphA3 , Receptors, Eph Family/antagonists & inhibitors , Receptors, Eph Family/genetics , Temozolomide/pharmacology , Xenograft Model Antitumor Assays
7.
Cell Signal ; 49: 30-38, 2018 Sep.
Article in English | MEDLINE | ID: mdl-29793020

ABSTRACT

Endothelial cell activation by proinflammatory stimuli drives leukocyte recruitment through enhanced expression of counter-receptors such as vascular cell adhesion molecule-1 (VCAM-1). We previously demonstrated that activation of the receptor tyrosine kinase EphA2 with its ligand ephrin-A1 induces VCAM-1 expression. Here, we sought to characterize the proinflammatory signaling pathways involved. Analysis of over-represented transcription factors in ephrin-A1-induced genes identified multiple potential transcriptional regulators, including the Rel family members nuclear factor-κB (NF-κB/p65) and nuclear factor of activated T-cells (NFAT). While ephrin-A1 failed to induce endothelial NF-κB activation, NF-κB inhibitors prevented ephrin-A1-induced VCAM-1 expression, suggesting basal NF-κB activity is required. In contrast, ephrin-A1 induced a robust EphA2-dependent increase in NFAT activation, and mutation of the NF-κB/NFAT-binding sites in the VCAM-1 promoter blunted ephrin-A1-induced promoter activity. NFAT activation classically occurs through calcium-dependent calcineurin activation, and inhibiting NFAT signaling with calcineurin inhibitors (cyclosporine A, FK506) or direct NFAT inhibitors (A-285222) was sufficient to block ephrin-A1-induced VCAM-1 expression. Consistent with robust NFAT activation, ephrin-A1-induced an EphA2-dependent calcium influx in endothelial cells that was required for ephrin-A1-induced NFAT activation and VCAM-1 expression. This work provides the first data showing EphA2-dependent calcium influx and NFAT activation and identifies NFAT as a novel EphA2-dependent proinflammatory pathway in endothelial activation.


Subject(s)
Calcium/metabolism , Ephrin-A2/metabolism , NFATC Transcription Factors/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Calcium Signaling/drug effects , Cell Line , Cell Nucleus/metabolism , Cyclosporine/pharmacology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Ephrin-A2/antagonists & inhibitors , Ephrin-A2/genetics , Gene Expression Regulation/drug effects , Histone Deacetylases/metabolism , Humans , NF-kappa B/metabolism , NFATC Transcription Factors/genetics , RNA Interference , RNA, Small Interfering/metabolism , Receptor, EphA2 , Transcription Factors/genetics , Transcription Factors/metabolism
8.
Eur Rev Med Pharmacol Sci ; 22(5): 1290-1296, 2018 03.
Article in English | MEDLINE | ID: mdl-29565486

ABSTRACT

OBJECTIVE: Emerging evidence suggests aberrant microRNAs (miRNAs) expression is involved in cancer development through multiple. Although miR338 has shown to have tumor suppression ability and anti-migration effects in some cancers, its regulatory role and molecular mechanism in the development of gastric cancer cells yet remains little known. This work aims to investigate miR-338 in regulating Wnt/ß-catenin pathway in epithelial-mesenchymal transition (EMT) in gastric cancers. MATERIALS AND METHODS: Human gastric cancer cells were transfected with either miR-338 mimic or erythropoietin-producing hepatocellular (Eph)A2-targeting siRNA. The biological function of miR-338 in gastric cancer cells was investigated using a MTT assay and invasion assay. Western blot assay was used to measure the levels of EphA2, GSK-3ß, phospho-GSK-3ßSer9, c-Myc, E-cadherin, Vimentin, and ß-catenin of at protein level. RESULTS: Our data showed that miR-338 inhibited proliferation, migration and invasion of human gastric cancer cells. miR-338 affected the Wnt/ß-catenin pathway by increasing p-GSK-3ßSer9 and decreasing GSK-3ßSer9 and c-Myc at protein levels. EphA2 protein level was downregulated and positively correlated with EMT markers. Both silencing of EphA2 and transfection with miR-338 mimic resulted in the up-regulation of the EMT molecular marker E-cadherin and down-regulation of Vimentin and ß-catenin at protein levels. CONCLUSIONS: This study indicated that miR-338 is a potential tumor suppressor in gastric cancer and miR-338 inhibited EMT of gastric cancer cells through deactivation of Wnt/ß-catenin signaling targeting at EphA2.


Subject(s)
Cell Proliferation , MicroRNAs/metabolism , Wnt Signaling Pathway , Antagomirs/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , Ephrin-A2/antagonists & inhibitors , Ephrin-A2/genetics , Ephrin-A2/metabolism , Epithelial-Mesenchymal Transition , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3 beta/metabolism , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , RNA Interference , RNA, Small Interfering/metabolism , Receptor, EphA2 , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Vimentin/metabolism , beta Catenin/metabolism
9.
J Virol ; 91(20)2017 10 15.
Article in English | MEDLINE | ID: mdl-28768873

ABSTRACT

The predominant types of dendritic cells (DC) in the skin and mucosa are Langerhans cells (LC) and interstitial dermal DC (iDDC). LC and iDDC process cutaneous antigens and migrate out of the skin and mucosa to the draining lymph nodes to present antigens to T and B cells. Because of the strategic location of LC and iDDC and the ability of these cells to capture and process pathogens, we hypothesized that they could be infected with human herpesvirus 8 (HHV-8) (Kaposi's sarcoma [KS]-associated herpesvirus) and have an important role in the development of KS. We have previously shown that HHV-8 enters monocyte-derived dendritic cells (MDDC) through DC-SIGN, resulting in nonproductive infection. Here we show that LC and iDDC generated from pluripotent cord blood CD34+ cell precursors support productive infection with HHV-8. Anti-DC-SIGN monoclonal antibody (MAb) inhibited HHV-8 infection of iDDC, as shown by low expression levels of viral proteins and DNA. In contrast, blocking of both langerin and the receptor protein tyrosine kinase ephrin A2 was required to inhibit HHV-8 infection of LC. Infection with HHV-8 did not alter the cell surface expression of langerin on LC but downregulated the expression of DC-SIGN on iDDC, as we previously reported for MDDC. HHV-8-infected LC and iDDC had a reduced ability to stimulate allogeneic CD4+ T cells in the mixed-lymphocyte reaction. These results indicate that HHV-8 can target both LC and iDDC for productive infection via different receptors and alter their function, supporting their potential role in HHV-8 pathogenesis and KS.IMPORTANCE Here we show that HHV-8, a DNA tumor virus that causes Kaposi's sarcoma, infects three types of dendritic cells: monocyte-derived dendritic cells, Langerhans cells, and interstitial dermal dendritic cells. We show that different receptors are used by this virus to infect these cells. DC-SIGN is a major receptor for infection of both monocyte-derived dendritic cells and interstitial dermal dendritic cells, yet the virus fully replicates only in the latter. HHV-8 uses langerin and the ephrin A2 receptor to infect Langerhans cells, which support full HHV-8 lytic replication. This infection of Langerhans cells and interstitial dermal dendritic cells results in an impaired ability to stimulate CD4+ helper T cell responses. Taken together, our data show that HHV-8 utilizes alternate receptors to differentially infect and replicate in these tissue-resident DC and support the hypothesis that these cells play an important role in HHV-8 infection and pathogenesis.


Subject(s)
Dendritic Cells/virology , Herpesvirus 8, Human/physiology , Langerhans Cells/virology , Antigens, CD/metabolism , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Cell Differentiation , Cells, Cultured , Dendritic Cells/immunology , Ephrin-A2/antagonists & inhibitors , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/immunology , Herpesvirus 8, Human/pathogenicity , Humans , Langerhans Cells/immunology , Langerhans Cells/pathology , Lectins, C-Type/antagonists & inhibitors , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Lymphocyte Culture Test, Mixed , Mannose-Binding Lectins/antagonists & inhibitors , Mannose-Binding Lectins/metabolism , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Sarcoma, Kaposi/virology , Skin/cytology , Skin/immunology , Skin/virology , T-Lymphocytes, Helper-Inducer/immunology , Virus Replication
10.
Oncogene ; 36(40): 5620-5630, 2017 10 05.
Article in English | MEDLINE | ID: mdl-28581527

ABSTRACT

Basal-like/triple-negative breast cancers (TNBCs) are among the most aggressive forms of breast cancer, and disproportionally affects young premenopausal women and women of African descent. Patients with TNBC suffer a poor prognosis due in part to a lack of molecularly targeted therapies, which represents a critical barrier for effective treatment. Here, we identify EphA2 receptor tyrosine kinase as a clinically relevant target for TNBC. EphA2 expression is enriched in the basal-like molecular subtype in human breast cancers. Loss of EphA2 function in both human and genetically engineered mouse models of TNBC reduced tumor growth in culture and in vivo. Mechanistically, targeting EphA2 impaired cell cycle progression through S-phase via downregulation of c-Myc and stabilization of the cyclin-dependent kinase inhibitor p27/KIP1. A small molecule kinase inhibitor of EphA2 effectively suppressed tumor cell growth in vivo, including TNBC patient-derived xenografts. Thus, our data identify EphA2 as a novel molecular target for TNBC.


Subject(s)
Cell Cycle , Ephrin-A2/metabolism , Triple Negative Breast Neoplasms/metabolism , Animals , Benzamides/pharmacology , Cell Line, Tumor , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Ephrin-A2/antagonists & inhibitors , Ephrin-A2/genetics , Female , Gene Knockdown Techniques , Humans , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplasm Recurrence, Local , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-myb/metabolism , Receptor, EphA2 , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/therapy , Xenograft Model Antitumor Assays
11.
Development ; 141(2): 460-71, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24381199

ABSTRACT

Inhibitory interneurons control the flow of information and synchronization in the cerebral cortex at the circuit level. During embryonic development, multiple subtypes of cortical interneurons are generated in different regions of the ventral telencephalon, such as the medial and caudal ganglionic eminence (MGE and CGE), as well as the preoptic area (POA). These neurons then migrate over long distances towards their cortical target areas. Diverse families of diffusible and cell-bound signaling molecules, including the Eph/ephrin system, regulate and orchestrate interneuron migration. Ephrin A3 and A5, for instance, are expressed at the borders of the pathway of MGE-derived interneurons and prevent these cells from entering inappropriate regions via EphA4 forward signaling. We found that MGE-derived interneurons, in addition to EphA4, also express ephrin A and B ligands, suggesting Eph/ephrin forward and reverse signaling in the same cell. In vitro and in vivo approaches showed that EphA4-induced reverse signaling in MGE-derived interneurons promotes their migration and that this effect is mediated by ephrin A2 ligands. In EphA4 mutant mice, as well as after ephrin A2 knockdown using in utero electroporation, we found delayed interneuron migration at embryonic stages. Thus, besides functions in guiding MGE-derived interneurons to the cortex through forward signaling, here we describe a novel role of the ephrins in driving these neurons to their target via reverse signaling.


Subject(s)
Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Ephrin-A2/metabolism , Interneurons/physiology , Receptor, EphA4/metabolism , Telencephalon/embryology , Telencephalon/metabolism , Animals , Cell Movement/physiology , Cerebral Cortex/cytology , Ephrin-A2/antagonists & inhibitors , Ephrin-A2/genetics , Female , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Mice , Mice, Inbred C57BL , Mice, Mutant Strains , Mice, Transgenic , Molecular Motor Proteins/genetics , Molecular Motor Proteins/metabolism , Pregnancy , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, EphA4/genetics , Signal Transduction , Telencephalon/cytology
12.
PLoS Pathog ; 9(7): e1003510, 2013.
Article in English | MEDLINE | ID: mdl-23874206

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) interacts with human dermal endothelial cell surface tyrosine kinase EphrinA2 (EphA2) and integrins (α3ß1 and αVß3) in the lipid raft (LR) region, and EphA2 regulates macropinocytic virus entry by coordinating integrin-c-Cbl associated signaling. In contrast, KSHV enters human foreskin fibroblast (HFF) cells by LR-independent clathrin mediated endocytosis. The present studies conducted to identify the key molecules regulating KSHV entry in HFF cells showed that KSHV induces association with integrins (αVß5, αVß3 and α3ß1) and EphA2 in non-LR regions early during infection and activates EphA2, which in turn associates with phosphorylated c-Cbl, myosin IIA, FAK, Src, and PI3-K, as well as clathrin and its adaptor AP2 and effector Epsin-15 proteins. EphA2 knockdown significantly reduced these signal inductions, virus internalization and gene expression. c-Cbl knockdown ablated the c-Cbl mediated K63 type polyubiquitination of EphA2 and clathrin association with EphA2 and KSHV. Mutations in EphA2's tyrosine kinase domain (TKD) or sterile alpha motif (SAM) abolished its interaction with c-Cbl. Mutations in tyrosine kinase binding (TKB) or RING finger (RF) domains of c-Cbl resulted in very poor association of c-Cbl with EphA2 and decreased EphA2 polyubiquitination. These studies demonstrated the contributions of these domains in EphA2 and c-Cbl association, EphA2 polyubiquitination and virus-EphA2 internalization. Collectively, these results revealed for the first time that EphA2 influences the tyrosine phosphorylation of clathrin, the role of EphA2 in clathrin mediated endocytosis of a virus, and c-Cbl mediated EphA2 polyubiquitination directing KSHV entry in HFF cells via coordinated signal induction and progression of endocytic events, all of which suggest that targeting EphA2 and c-Cbl could block KSHV entry and infection.


Subject(s)
Clathrin-Coated Vesicles/metabolism , Endocytosis , Ephrin-A2/metabolism , Fibroblasts/virology , Herpesvirus 8, Human/physiology , Integrins/metabolism , Proto-Oncogene Proteins c-cbl/metabolism , Cells, Cultured , Ephrin-A2/agonists , Ephrin-A2/antagonists & inhibitors , Ephrin-A2/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Regulation, Viral , HEK293 Cells , Humans , Mutant Proteins/agonists , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Phosphorylation , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational , RNA Interference , RNA, Small Interfering , Signal Transduction , Ubiquitination , Up-Regulation , Viral Proteins/biosynthesis , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Internalization
13.
Biochem Biophys Res Commun ; 380(3): 569-74, 2009 Mar 13.
Article in English | MEDLINE | ID: mdl-19285002

ABSTRACT

The novel protein modification, O-linked N-acetylglucosamine (O-GlcNAc), plays an important role in various aspects of cell regulation. Although most of nuclear transcription regulatory factors are modified by O-GlcNAc, O-GlcNAc effects on transcription remain largely undefined yet. In this study, we show that O-GlcNAc inhibits a physical interaction between Sp1 and Elf-1 transcription factors, and negatively regulates transcription of placenta and embryonic expression oncofetal protein gene (Pem). These findings suggest that O-GlcNAc inhibits Sp1-mediated gene transcription possibly by interrupting Sp1 interaction with its cooperative factor.


Subject(s)
Ephrin-A2/antagonists & inhibitors , Gene Expression Regulation , Homeodomain Proteins/genetics , Sp1 Transcription Factor/antagonists & inhibitors , Transcription Factors/genetics , Acetylglucosamine , Animals , Cell Line, Tumor , Ephrin-A2/metabolism , Humans , Mice , Rats , Sp1 Transcription Factor/metabolism , Transcription, Genetic
14.
J Biol Chem ; 283(43): 29461-72, 2008 Oct 24.
Article in English | MEDLINE | ID: mdl-18728010

ABSTRACT

The erythropoietin-producing hepatocellular (Eph) family of receptor tyrosine kinases regulates a multitude of physiological and pathological processes. Despite the numerous possible research and therapeutic applications of agents capable of modulating Eph receptor function, no small molecule inhibitors targeting the extracellular domain of these receptors have been identified. We have performed a high throughput screen to search for small molecules that inhibit ligand binding to the extracellular domain of the EphA4 receptor. This yielded a 2,5-dimethylpyrrolyl benzoic acid derivative able to inhibit the interaction of EphA4 with a peptide ligand as well as the natural ephrin ligands. Evaluation of a series of analogs identified an isomer with similar inhibitory properties and other less potent compounds. The two isomeric compounds act as competitive inhibitors, suggesting that they target the high affinity ligand-binding pocket of EphA4 and inhibit ephrin-A5 binding to EphA4 with K(i) values of 7 and 9 mum in enzyme-linked immunosorbent assays. Interestingly, despite the ability of each ephrin ligand to promiscuously bind many Eph receptors, the two compounds selectively target EphA4 and the closely related EphA2 receptor. The compounds also inhibit ephrin-induced phosphorylation of EphA4 and EphA2 in cells, without affecting cell viability or the phosphorylation of other receptor tyrosine kinases. Furthermore, the compounds inhibit EphA4-mediated growth cone collapse in retinal explants and EphA2-dependent retraction of the cell periphery in prostate cancer cells. These data demonstrate that the Eph receptor-ephrin interface can be targeted by inhibitory small molecules and suggest that the two compounds identified will be useful to discriminate the activities of EphA4 and EphA2 from those of other co-expressed Eph receptors that are activated by the same ephrin ligands. Furthermore, the newly identified inhibitors represent possible leads for the development of therapies to treat pathologies in which EphA4 and EphA2 are involved, including nerve injuries and cancer.


Subject(s)
Ephrin-A2/antagonists & inhibitors , Ephrin-A4/antagonists & inhibitors , Receptor, EphA2/metabolism , Receptor, EphA4/metabolism , Animals , COS Cells , Chlorocebus aethiops , Drug Design , Humans , Inhibitory Concentration 50 , Kinetics , Ligands , Protein Binding/drug effects , Protein Structure, Tertiary , Tumor Necrosis Factor-alpha/metabolism
15.
Immunogenetics ; 60(10): 557-63, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18629488

ABSTRACT

The high-affinity receptor for immunoglobulin E (IgE), FcepsilonRI, is specifically expressed in mast cells and basophils and plays a key role in IgE-mediated allergic reactions. The transcription factor Elf-1 has been previously identified to bind to the promoter of the human FcepsilonRI alpha-chain, which is essential for the function and expression of FcepsilonRI. In the present study, Elf-1 siRNA was conducted to evaluate the effects of Elf-1 on FcepsilonRI alpha-chain expression in the primary mouse mast cells, bone marrow-derived mast cells (BMMC). Introduction of Elf-1 siRNA effectively reduced expression levels of Elf-1 mRNA and protein in BMMC. Transient reporter assay showed that the knockdown of Elf-1 by siRNA resulted in increased FcepsilonRI alpha-chain promoter activity, while overexpression of Elf-1 suppressed alpha-chain promoter activity in BMMC. Elf-1 siRNA-treated BMMC exhibited marked upregulation of FcepsilonRI alpha-chain transcription, whereas beta-chain mRNA was not affected by Elf-1 siRNA. Chromatin immunoprecipitation assay showed that the amount of transcription factor PU.1, recognizing the cis-element close to the Elf-1-site on the FcepsilonRI alpha-chain promoter, was significantly increased by introduction of Elf-1 siRNA. These results indicate that Elf-1 negatively regulates FcepsilonRI alpha-chain expression by suppressing PU.1-mediated transcription of the alpha-chain in BMMC.


Subject(s)
Ephrin-A2/antagonists & inhibitors , Gene Expression Regulation , Mast Cells/metabolism , RNA, Small Interfering/pharmacology , Receptors, IgE/genetics , Animals , Blotting, Western , Bone Marrow/metabolism , Chromatin Immunoprecipitation , Humans , Luciferases/metabolism , Mice , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, IgE/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription, Genetic , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...