Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 57
Filter
1.
Clin. transl. oncol. (Print) ; 24(7): 1274-1289, julio 2022.
Article in English | IBECS | ID: ibc-203828

ABSTRACT

Ephrin receptor A7 (EphA7) is a member of the Eph receptor family. It is widely involved in signal transduction between cells, regulates cell proliferation and differentiation, and participates in developing neural tubes and brain. In addition, EphA7 also has a dual role of tumor promoter and tumor suppressor. It can participate in cell proliferation, migration and apoptosis through various mechanisms, and affect tumor differentiation, staging and prognosis. EphA7 may be a potential diagnostic marker and tumor treatment target. This article reviews the effects of EphA7 on a variety of tumor biological processes and pathological characteristics, as well as specific effects and regulatory mechanisms.


Subject(s)
Humans , Apoptosis , Cell Proliferation , Receptor, EphA7/genetics , Receptor, EphA7/metabolism , Neoplasms/genetics , Signal Transduction
2.
BMC Cancer ; 22(1): 636, 2022 Jun 09.
Article in English | MEDLINE | ID: mdl-35681118

ABSTRACT

BACKGROUND: Aberrant methylation of EphA7 has been reported in the process of carcinogenesis but not in cervical cancer. Therefore, an integration study was performed to explore the association between EphA7 hypermethylation and cervical cancer and validate the potential value of EphA7 hypermethylation in the diagnosis of cervical cancer. METHODS: We performed an integration study to identify and validate the association between EphA7 methylation and cervical cancer. First, data on EphA7 methylation and expression in cervical cancer were extracted and analyzed via bioinformatics tools. Subsequently, CRISPR-based methylation perturbation tools (dCas9-Tet1/DNMT3a) were constructed to further demonstrate the association between DNA methylation and EphA7 expression. Ultimately, the clinical value of EphA7 methylation in cervical cancer was validated in cervical tissues and Thinprep cytologic test (TCT) samples by methylation-specific PCR (MSP) and quantitative methylation-specific PCR (QMSP), respectively. RESULTS: Pooled analysis showed that EphA7 promoter methylation levels were significantly increased in cervical cancer compared to normal tissues (P < 0.001) and negatively correlated with EphA7 expression. These prediction results were subsequently confirmed in cell lines; moreover, CRISPR-based methylation perturbation tools (dCas9-Tet1/DNMT3a) demonstrated that DNA methylation participates in the regulation of EphA7 expression directly. Consistent with these findings, the methylation level and the positive rate of EphA7 gradually increased with severity from normal to cancer stages in TCT samples (P < 0.01). CONCLUSIONS: EphA7 hypermethylation is present in cervical cancer and is a potential biomarker for the diagnosis of cervical cancer.


Subject(s)
DNA Methylation , Receptor, EphA7 , Uterine Cervical Neoplasms , Biomarkers, Tumor , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Receptor, EphA7/genetics , Uterine Cervical Neoplasms/diagnosis , Uterine Cervical Neoplasms/genetics
3.
Clin Transl Oncol ; 24(7): 1274-1289, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35112312

ABSTRACT

Ephrin receptor A7 (EphA7) is a member of the Eph receptor family. It is widely involved in signal transduction between cells, regulates cell proliferation and differentiation, and participates in developing neural tubes and brain. In addition, EphA7 also has a dual role of tumor promoter and tumor suppressor. It can participate in cell proliferation, migration and apoptosis through various mechanisms, and affect tumor differentiation, staging and prognosis. EphA7 may be a potential diagnostic marker and tumor treatment target. This article reviews the effects of EphA7 on a variety of tumor biological processes and pathological characteristics, as well as specific effects and regulatory mechanisms.


Subject(s)
Neoplasms , Receptor, EphA7 , Apoptosis , Cell Proliferation , Genes, Tumor Suppressor , Humans , Neoplasms/genetics , Receptor, EphA7/genetics , Receptor, EphA7/metabolism , Signal Transduction/physiology
4.
Anal Cell Pathol (Amst) ; 2022: 4220786, 2022.
Article in English | MEDLINE | ID: mdl-35103233

ABSTRACT

Basal cell carcinoma (BCC) is the most common malignancy worldwide, with increasing incidence. BCCs present low mortality but high morbidity, and its pathogenesis remains unclear. Eph receptors have been implicated in tumorigenesis. EphA7 plays a role as a tumor suppressor in certain cancers. We checked EphA7 expression levels and methylation status in a set of BCCs, benign skin diseases, and compound nevus tissue samples using immunohistochemistry. EphA7 protein was positively expressed in normal basal cells, benign skin diseases, and compound nevus cells, but lost in areas of BCC tissues. We detected hypermethylation in BCC tissue samples with reduced expression of EphA7. There is a significant relationship between the expression level of EphA7 receptor protein and the methylation status of CpG islands in the EphA7 promoter region (P < 0.001). To our knowledge, this is the first study to report the EphA7 expression profile and hypermethylation of EphA7 in BCC. The role of the EphA7 gene and the status of hypermethylation in tumorigenesis and treatment of BCC warrant further investigation.


Subject(s)
Carcinoma, Basal Cell , CpG Islands , DNA Methylation , Receptor, EphA7 , Skin Neoplasms , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/metabolism , Humans , Promoter Regions, Genetic , Receptor, EphA7/biosynthesis , Receptor, EphA7/genetics , Receptor, EphA7/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism
5.
Ann Med ; 53(1): 1358-1369, 2021 12.
Article in English | MEDLINE | ID: mdl-34396843

ABSTRACT

BACKGROUND: Triple-negative breast cancer (TNBC) represents about 19% of all breast cancer cases in the Chinese population. Lack of targeted therapy contributes to the poorer outcomes compared with other breast cancer subtypes. Comprehensive genomic profiling helps to explore the clinically relevant genomic alterations (CRGAs) and potential therapeutic targets in very-early-relapsed TNBC patients. METHODS: Formalin-fixed paraffin-embedded (FFPE) tumour tissue specimens from 23 patients with very-early-relapsed TNBC and 13 patients with disease-free survival (DFS) more than 36 months were tested by FoundationOne CDx (F1CDx) in 324 genes and select gene rearrangements, along with genomic signatures including microsatellite instability (MSI) and tumour mutational burden (TMB). RESULTS: In total, 137 CRGAs were detected in the 23 very-early-relapsed TNBC patients, averaging six alterations per sample. The mean TMB was 4 Muts/Mb, which was higher than that in non-recurrence patients, and is statistically significant. The top-ranked altered genes were TP53 (83%), PTEN (35%), RB1 (30%), PIK3CA (26%) and BRCA1 (22%). RB1 mutation carriers had shorter DFS. Notably, 100% of these patients had at least one CRGA, and 87% of patients had at least one actionable alteration. In pathway analysis, patients who carried a mutation in the cell cycle pathway were more likely to experience very early recurrence. Strikingly, we detected one patient with ERBB2 amplification and one patient with ERBB2 exon20 insertion, both of which were missed by immunohistochemistry (IHC). We also detected novel alterations of ROS1-EPHA7 fusion for the first time, which has not been reported in breast cancer before. CONCLUSIONS: The comprehensive genomic profiling can identify novel treatment targets and address the limited options in TNBC patients. Therefore, incorporating F1CDx into TNBC may shed light on novel therapeutic opportunities for these very-early-relapsed TNBC patients.


Subject(s)
Biomarkers, Tumor/genetics , Genomics/methods , Triple Negative Breast Neoplasms/genetics , Adult , Aged , China/epidemiology , Female , Genomic Instability/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Middle Aged , Molecular Targeted Therapy , Mutation , Neoplasm Staging , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Proteins/genetics , Receptor, EphA7/genetics , Recurrence , Triple Negative Breast Neoplasms/ethnology , Triple Negative Breast Neoplasms/pathology
6.
Biochem Biophys Res Commun ; 568: 62-67, 2021 09 03.
Article in English | MEDLINE | ID: mdl-34186436

ABSTRACT

Erythropoietin producing hepatocellular (Eph) forms the largest family of receptor tyrosine kinases (RTK). As a family, Eph regulates physiological events such as cell-cell interaction, cell migration, and adhesion. The Kinase domain is the catalytic core of the Eph receptor and is highly conserved sequentially. EphA7 has been recently regarded as a cancer driver gene and comprises several clinically important mutations. Three of the EphA7 mutations Gly656Glu, Gly656Arg, and Asp751His, present in the kinase domain, are predicted to be highly pathogenic. Furthermore, Gly656Glu and Gly656Arg are reported to be hotspot mutations. Considering the importance of mutations, crystals structure of EphA7 Gly656Glu, Gly656Arg, and Asp751His mutants has been explored. Changes in folding pattern and intramolecular interactions were observed in mutant structures. Secondary structural changes were observed in the hinge region of EphA7 Gly656Arg and Asp751His structure, affecting the transition of kinase domain between open and closed conformations. EphA7 Asp751His mutant structure shows a distorted nucleotide-binding groove. Differences were observed in hydrogen bonding and hydrophobic interactions between the catalytic and highly conserved DFG motif in the EphA7 mutants, which may influence the catalytic activity of kinase domain.


Subject(s)
Point Mutation , Receptor, EphA7/chemistry , Receptor, EphA7/genetics , Crystallography, X-Ray , Humans , Models, Molecular , Protein Conformation , Protein Domains
7.
Clin Genet ; 100(4): 396-404, 2021 10.
Article in English | MEDLINE | ID: mdl-34176129

ABSTRACT

Ephrin receptor and their ligands, the ephrins, are widely expressed in the developing brain. They are implicated in several developmental processes that are crucial for brain development. Deletions in genes encoding for members of the Eph/ephrin receptor family were reported in several neurodevelopmental disorders. The ephrin receptor A7 gene (EPHA7) encodes a member of ephrin receptor subfamily of the protein-tyrosine kinase family. EPHA7 plays a role in corticogenesis processes, determines brain size and shape, and is involved in development of the central nervous system. One patient only was reported so far with a de novo deletion encompassing EPHA7 in 6q16.1. We report 12 additional patients from nine unrelated pedigrees with similar deletions. The deletions were inherited in nine out of 12 patients, suggesting variable expressivity and incomplete penetrance. Four patients had tiny deletions involving only EPHA7, suggesting a critical role of EPHA7 in a neurodevelopmental disability phenotype. We provide further evidence for EPHA7 deletion as a risk factor for neurodevelopmental disorder and delineate its clinical phenotype.


Subject(s)
Genetic Association Studies , Genetic Predisposition to Disease , Haploinsufficiency , Neurodevelopmental Disorders/diagnosis , Neurodevelopmental Disorders/genetics , Phenotype , Receptor, EphA7/genetics , Chromosomes, Human, Pair 6 , Comparative Genomic Hybridization , Female , Genetic Association Studies/methods , Humans , In Situ Hybridization, Fluorescence , Inheritance Patterns , Male , Mutation , Pedigree , Exome Sequencing
8.
Mol Med Rep ; 23(1)2021 01.
Article in English | MEDLINE | ID: mdl-33236144

ABSTRACT

Micro (mi)RNAs serve crucial roles in cancer development although little is known about their cellular mechanisms in the pathogenesis of melanoma. The present study explored the regulatory roles of miR­18a­5p in melanoma cell proliferation, apoptosis and autophagy, in addition to its target gene in melanoma cells. miRNA and ephrin receptor A7 (EPHA7) mRNA were analyzed by reverse transcription­quantitative PCR. Cell Counting Kit­8 and colony formation assays were performed to examine the cell proliferation rate. Hoechst staining and flow cytometry were performed to investigate cell apoptosis. Western blotting was used to estimate the abundance of proteins. Dual-luciferase reporter assay verified the binding of miRNA with target gene sequences. Melanoma tissues and cell lines exhibited markedly elevated miR­18a­5p expression. miR­18a­5p inhibitor inhibited proliferation rates, and triggered apoptosis and autophagy marker protein expression in WM266­4 and A375 cells. It also negatively regulated EPHA7 expression in WM266­4 and A375 cells by directly binding at the 3'­untranslated region of EPHA7. miR­18a­5p mimics reversed the EPHA7 overexpression­induced suppression of proliferation, and the EPHA7 overexpression­induced promotion of apoptosis and autophagy. miR­18a­5p triggered proliferation of melanoma cells and inhibited apoptosis and autophagy by directly targeting and inhibiting EPHA7 expression. Thus, the present study aided our understanding of miRNA­mediated melanoma pathogenesis.


Subject(s)
Apoptosis , Autophagy , Cell Proliferation , Melanoma/metabolism , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , RNA, Neoplasm/metabolism , Receptor, EphA7/metabolism , Signal Transduction , Skin Neoplasms/metabolism , Cell Line, Tumor , Female , Humans , Male , Melanoma/genetics , Melanoma/pathology , MicroRNAs/genetics , Neoplasm Proteins/genetics , RNA, Neoplasm/genetics , Receptor, EphA7/genetics , Skin Neoplasms/genetics , Skin Neoplasms/pathology
9.
Eur Rev Med Pharmacol Sci ; 24(11): 6139-6149, 2020 06.
Article in English | MEDLINE | ID: mdl-32572879

ABSTRACT

OBJECTIVE: Non-small cell lung cancer (NSCLC) is a primary subtype of lung cancers which has a high morbidity and poor prognosis. Emerging evidence has demonstrated that aberrantly expressed microRNAs (miRNAs) were implicated in the regulatory functions of multiple processes during tumorigenesis. In the current study, we explored the functional roles and underlying mechanisms of miR-448 in NSCLC. PATIENTS AND METHODS: Quantitative real-time polymerase chain reaction assays were conducted to measure miR-448 expressions in 51 pairs of NSCLC tissues and corresponding normal tissues. Moreover, the relationship between miR-448 expressions and clinicopathological characteristics of NSCLC patients was also determined. We then performed transwell assays to explore the functions of miR-448 in NSCLC cell invasion and migration. As we had identified EPHA7 as a functional target of miR-448 in NSCLC cells, the clinical significance of EPHA7 in NSCLC patients was further investigated. Finally, we detected the influence of miR-448 on tumor growth rate and tumor size of NSCLC using tumor xenografts. RESULTS: Underexpressed miR-448 was identified in NSCLC, and low miR-448 expression was confirmed to be associated with the poor prognosis and adverse clinicopathologic features of NSCLC patients. Moreover, functional assays demonstrated that miR-448 overexpression suppressed NSCLC cell proliferation, invasion and migration. EPHA7 was identified as a direct target of miR-448. Additionally, miR-448 restoration suppressed in vivo NSCLC cell growth. Finally, our studies also indicated that miR-448 exerted anti-NSCLC functions via regulating PI3K/AKT signaling pathway and EMT. CONCLUSIONS: These results showed that miR-448/EPHA7 axis maybe one of the useful diagnostic and prognostic biomarkers for NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Cell Movement , Epithelial-Mesenchymal Transition , Lung Neoplasms/metabolism , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, EphA7/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line , Cell Proliferation , Humans , Lung Neoplasms/pathology , Mice , MicroRNAs/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Receptor, EphA7/genetics , Signal Transduction
10.
Am J Physiol Endocrinol Metab ; 319(1): E81-E90, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32396496

ABSTRACT

We have previously shown that systemic injection of erythropoietin-producing hepatocellular receptor A7 (EPHA7)-Fc raises serum luteinizing hormone (LH) levels before ovulation in female rats, indicating the induction of EPHA7 in ovulation. In this study, we aimed to identify the mechanism and hypothalamus-pituitary-ovary (HPO) axis level underlying the promotion of LH secretion by EPHA7. Using an ovariectomized (OVX) rat model, in conjunction with low-dose 17ß-estradiol (E2) treatment, we investigated the association between EPHA7-ephrin (EFN)A5 signaling and E2 negative feedback. Various rat models (OVX, E2-treated OVX, and abarelix treated) were injected with the recombinant EPHA7-Fc protein through the caudal vein to investigate the molecular mechanism underlying the promotion of LH secretion by EPHA7. Efna5 was observed strongly expressed in the arcuate nucleus of the female rat by using RNAscope in situ hybridization. Our results indicated that E2, combined with estrogen receptor (ER)α, but not ERß, inhibited Efna5 and gonadotropin-releasing hormone 1 (Gnrh1) expressions in the hypothalamus. In addition, the systemic administration of EPHA7-Fc restrained the inhibition of Efna5 and Gnrh1 by E2, resulting in increased Efna5 and Gnrh1 expressions in the hypothalamus as well as increased serum LH levels. Collectively, our findings demonstrated the involvement of EPHA7-EFNA5 signaling in the regulation of LH and the E2 negative feedback pathway in the hypothalamus, highlighting the functional role of EPHA7 in female reproduction.


Subject(s)
Ephrin-A5/metabolism , Estrogen Receptor alpha/metabolism , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Luteinizing Hormone/metabolism , Protein Precursors/metabolism , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Arcuate Nucleus of Hypothalamus/metabolism , Ephrin-A5/drug effects , Ephrin-A5/genetics , Estradiol/pharmacology , Estrogen Receptor beta/metabolism , Estrogens/pharmacology , Feedback, Physiological/drug effects , Feedback, Physiological/physiology , Female , Gonadotropin-Releasing Hormone/drug effects , Hormone Antagonists/pharmacology , Hypothalamo-Hypophyseal System/drug effects , Hypothalamo-Hypophyseal System/metabolism , Hypothalamus/drug effects , Luteinizing Hormone/drug effects , Oligopeptides/pharmacology , Ovariectomy , Ovary/drug effects , Ovary/metabolism , Protein Precursors/drug effects , Rats , Receptor, EphA7/genetics , Receptor, EphA7/metabolism , Receptor, EphA7/pharmacology , Recombinant Proteins
11.
Biochem Biophys Res Commun ; 526(2): 375-380, 2020 05 28.
Article in English | MEDLINE | ID: mdl-32222280

ABSTRACT

Receptor tyrosine kinase EphA7 is specifically expressed in otic region in Xenopus early development. However, its role in otocyst development remains unknown. Knockdown of EphA7 by a specific morpholino oligonucleotide (MO) reduced the size of the otocyst and triggered otic epithelial cell extrusion. Interestingly, EphA7 depletion attenuated the membrane level of the tight junction protein Claudin6 (CLDN6). Utilizing the Cldn6 MO, we further confirmed that CLDN6 attenuation also led to otic epithelial cell extrusion. Our work suggested that EphA7 modulates the otic epithelial homeostasis through stabilizing the CLDN6 membrane level.


Subject(s)
Claudins/genetics , Gene Expression Regulation, Developmental , Receptor, EphA7/genetics , Xenopus Proteins/genetics , Xenopus laevis/embryology , Animals , Gene Knockdown Techniques , Homeostasis , Xenopus laevis/genetics
12.
J Virol ; 93(15)2019 08 01.
Article in English | MEDLINE | ID: mdl-31118261

ABSTRACT

Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of Kaposi's sarcoma and is associated with two B cell malignancies, primary effusion lymphoma (PEL) and the plasmablastic variant of multicentric Castleman's disease. On several adherent cell types, EphA2 functions as a cellular receptor for the gH/gL glycoprotein complex of KSHV. KSHV gH/gL also has previously been found to interact weakly with other members of the Eph family of receptor tyrosine kinases (Ephs), and other A-type Ephs have been shown to be able to compensate for the absence of EphA2 using overexpression systems. However, whether these interactions are of functional consequence at endogenous protein levels has remained unclear so far. Here, we demonstrate for the first time that endogenously expressed EphA7 in BJAB B cells is critical for the cell-to-cell transmission of KSHV from producer iSLK cells to BJAB target cells. The BJAB lymphoblastoid cell line often serves as a model for B cell infection and expresses only low levels of all Eph family receptors other than EphA7. Endogenous EphA7 could be precipitated from the cellular lysate of BJAB cells using recombinant gH/gL, and knockout of EphA7 significantly reduced transmission of KSHV into BJAB target cells. Knockout of EphA5, the second most expressed A-type Eph in BJAB cells, had a similar, although less pronounced, effect on KSHV infection. Receptor function of EphA7 was conserved for cell-free infection by the related rhesus monkey rhadinovirus (RRV), which is relatively even more dependent on EphA7 for infection of BJAB cells.IMPORTANCE Infection of B cells is relevant for two KSHV-associated malignancies, the plasmablastic variant of multicentric Castleman's disease and PEL. Therefore, elucidating the process of B cell infection is important for the understanding of KSHV pathogenesis. While the high-affinity receptor for the gH/gL glycoprotein complex, EphA2, has been shown to function as an entry receptor for various types of adherent cells, the gH/gL complex can also interact with other Eph receptor tyrosine kinases with lower avidity. We analyzed the Eph interactions required for infection of BJAB cells, a model for B cell infection by KSHV. We identified EphA7 as the principal Eph receptor for infection of BJAB cells by KSHV and the related rhesus monkey rhadinovirus. While two analyzed PEL cell lines exhibited high EphA2 and low EphA7 expression, a third PEL cell line, BCBL-1, showed high EphA7 and low EphA2 expression, indicating a possible relevance for KSHV pathology.


Subject(s)
B-Lymphocytes/metabolism , Receptor, EphA7/metabolism , Receptors, Virus/metabolism , Rhadinovirus/physiology , Virus Internalization , Animals , B-Lymphocytes/pathology , B-Lymphocytes/virology , Cell Line, Tumor , Herpesvirus 8, Human/genetics , Herpesvirus 8, Human/metabolism , Herpesvirus 8, Human/physiology , Humans , Lymphoma, Primary Effusion/metabolism , Lymphoma, Primary Effusion/pathology , Macaca mulatta , Receptor, EphA7/genetics , Receptors, Virus/genetics , Rhadinovirus/genetics , Rhadinovirus/metabolism , Viral Envelope Proteins/genetics , Viral Envelope Proteins/metabolism
13.
Sci Rep ; 9(1): 263, 2019 01 22.
Article in English | MEDLINE | ID: mdl-30670742

ABSTRACT

Tumour sequencing identifies highly recurrent point mutations in cancer driver genes, but rare functional mutations are hard to distinguish from large numbers of passengers. We developed a novel computational platform applying a multi-modal approach to filter out passengers and more robustly identify putative driver genes. The primary filter identifies enrichment of cancer mutations in CATH functional families (CATH-FunFams) - structurally and functionally coherent sets of evolutionary related domains. Using structural representatives from CATH-FunFams, we subsequently seek enrichment of mutations in 3D and show that these mutation clusters have a very significant tendency to lie close to known functional sites or conserved sites predicted using CATH-FunFams. Our third filter identifies enrichment of putative driver genes in functionally coherent protein network modules confirmed by literature analysis to be cancer associated. Our approach is complementary to other domain enrichment approaches exploiting Pfam families, but benefits from more functionally coherent groupings of domains. Using a set of mutations from 22 cancers we detect 151 putative cancer drivers, of which 79 are not listed in cancer resources and include recently validated cancer associated genes EPHA7, DCC netrin-1 receptor and zinc-finger protein ZNF479.


Subject(s)
Neoplasms/genetics , Oncogenes/genetics , Protein Interaction Maps/genetics , Computational Biology/methods , DCC Receptor/genetics , DCC Receptor/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Databases, Genetic/statistics & numerical data , Datasets as Topic , Humans , Point Mutation , Protein Interaction Mapping/methods , Receptor, EphA7/genetics , Receptor, EphA7/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
14.
EBioMedicine ; 36: 539-552, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30292674

ABSTRACT

BACKGROUND: The ovulatory dysfunction mechanisms underlying polycystic ovary syndrome (PCOS) are not completely understood. And the roles of EPHA7 and EPHA7-regulated pathway factors in the pathogenesis of anovulation remain to be elucidated. METHODS: We used human granulosa cells (hGCs) of PCOS and non-PCOS patients to measure EPHA7 and other target gene expressions. We performed in vitro experiments in KGN cells to verify the molecular mechanisms. Additionally, we conducted in vivo loss- and gain-of-function studies using EPHA7 shRNA lentivirus and recombinant EPHA7-Fc protein injection to identify the ovulation effects of EPHA7. FINDINGS: EPHA7 functions as a critically positive upstream factor for the expression of ERK1/2-mediated C/EBPß. This protein, in turn, induced the expression of KLF4 and then ADAMTS1. Moreover, decreased abundance of EPHA7 was positively correlated with that of its downstream factors in hGCs of PCOS patients. Additionally, a 1-week functional EPHA7 shRNA lentivirus in rat ovaries contributed to decreased numbers of retrieved oocytes, and a 3-week functional lentivirus led to menstrual disorders and morphological polycystic changes in rat ovaries. More importantly, we found that EPHA7 triggered ovulation in rats, and it improved polycystic ovarian changes induced by DHEA in PCOS rats. INTERPRETATION: Our findings demonstrate a new role of EPHA7 in PCOS, suggesting that EPHA7 is an effective target for the development of innovative medicines to induce ovulation. FUND: National Key Research and Development Program of China, National Natural Science Foundation, Shanghai Municipal Education Commission--Gaofeng Clinical Medicine, and Shanghai Commission of Science and Technology.


Subject(s)
Ovulation/metabolism , Polycystic Ovary Syndrome/metabolism , Polycystic Ovary Syndrome/physiopathology , Receptor, EphA7/metabolism , Adult , Animals , Biomarkers , Cell Line , Disease Models, Animal , Female , Gene Expression , Gene Expression Regulation , Gene Silencing , Granulosa Cells/metabolism , Humans , Kruppel-Like Factor 4 , Ovary/metabolism , Ovary/pathology , Ovulation/genetics , Polycystic Ovary Syndrome/genetics , RNA Interference , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Receptor, EphA7/genetics , Young Adult
15.
Biochem Biophys Res Commun ; 495(2): 1580-1587, 2018 01 08.
Article in English | MEDLINE | ID: mdl-29223398

ABSTRACT

Eph/ephrin molecules are widely expressed during embryonic development, and function in a variety of developmental processes. Here we studied the roles of the Eph receptor EphA7 and its soluble form in Xenopus pronephros development. EphA7 is specifically expressed in pronephric tubules at tadpole stages and knockdown of EphA7 by a translation blocking morpholino led to defects in tubule cell differentiation and morphogenesis. A soluble form of EphA7 (sEphA7) was also identified. Interestingly, the membrane level of claudin6 (CLDN6), a tetraspan transmembrane tight junction protein, was dramatically reduced in the translation blocking morpholino injected embryos, but not when a splicing morpholino was used, which blocks only the full length EphA7. In cultured cells, EphA7 binds and phosphorylates CLDN6, and reduces its distribution at the cell surface. Our work suggests a role of EphA7 in the regulation of cell adhesion during pronephros development, whereas sEphA7 works as an antagonist.


Subject(s)
Claudins/metabolism , Pronephros/embryology , Receptor, EphA7/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Xenopus laevis/metabolism , Animals , Cell Membrane/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Oligodeoxyribonucleotides, Antisense/genetics , Pronephros/metabolism , Receptor, EphA7/antagonists & inhibitors , Receptor, EphA7/genetics , Solubility , Xenopus Proteins/antagonists & inhibitors , Xenopus Proteins/genetics , Xenopus laevis/genetics
16.
Cell Death Dis ; 8(10): e3122, 2017 10 12.
Article in English | MEDLINE | ID: mdl-29022918

ABSTRACT

The downregulation of receptor tyrosine kinase EphA7 is frequent in epithelial cancers and linked to tumor progression. However, the detailed mechanism of EphA7-mediated prostate tumor progression remains elusive. To test the role of EphA7 receptor in prostate cancer (PCa) progression directly, we generated EphA7 receptor variants that were either lacking the cytoplasmic domain or carrying a point mutation that inhibits its phosphorylation by site-directed mutagenesis. Overexpression of wild-type (WT) EphA7 in PCa cells resulted in decreased tumor volume and increased tumor apoptosis in primary tumors. In addition, ectopic expression of WT EphA7 both can delay PCa cell proliferation and could inhibit PCa cell migration and invasion. This protein can also induce PCa cell apoptosis that correlated with increasing the protein expression levels of Bax, elevating the caspase-3 activities, reducing the protein expression levels of Bcl-2 and facilitating the dephosphorylation of Akt, which is further increased by the stimulation of ephrinA5-Fc. However, expression of these EphA7 mutants in PCa cells has no effect in vivo and in vitro. The expression of EphA7 and ephrinA5 was significantly decreased in PCa specimens compared with BPH tissues or paired normal tissues. Moreover, the phosphorylation of EphA7 was positively related with ephrinA5 expression in human prostate tissues. In sum, receptor phosphorylation of EphA7, at least in part, suppress PCa tumor malignancy through targeting PI3K/Akt signaling pathways.


Subject(s)
Ephrin-A5/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-akt/metabolism , Receptor, EphA7/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Apoptosis/physiology , Caspase 3/metabolism , Cell Line, Tumor , Cell Movement/physiology , Cell Proliferation/physiology , Disease Progression , Down-Regulation , HEK293 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness/pathology , Phosphorylation/physiology , Receptor, EphA7/genetics , Signal Transduction/physiology , bcl-2-Associated X Protein/biosynthesis
17.
PLoS One ; 12(6): e0175553, 2017.
Article in English | MEDLINE | ID: mdl-28604772

ABSTRACT

Osteosarcoma is the most common type of malignant bone tumor, often affecting adolescents and children. MicroRNAs (miRNAs) are a group of small, non-protein coding, endogenous RNAs that play critical roles in osteosarcoma tumorigenesis. In our study, we demonstrated that miR-448 expression was downregulated in osteosarcoma tissues and cell lines. Overexpression of miR-448 suppressed osteosarcoma cell proliferation, colony formation and migration. Moreover, we found that EPHA7 was a direct target gene of miR-448 in osteosarcoma cells. We further demonstrated that the EPHA7 expression level was upregulated in osteosarcoma tissues. Interestingly, the expression level of EPHA7 was inversely correlated with the expression level of miR-448 in osteosarcoma tissues. In addition, elevated expression of miR-448 suppressed osteosarcoma cell proliferation and invasion through targeting EPHA7. Taken together, these findings suggest that miR-448 functioned as a tumor suppressor gene in the development of osteosarcoma through targeting EPHA7.


Subject(s)
Bone Neoplasms/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Osteosarcoma/genetics , RNA Interference , Receptor, EphA7/genetics , Base Pairing , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Humans , Osteosarcoma/pathology , Tumor Stem Cell Assay
18.
Chembiochem ; 18(13): 1317-1331, 2017 07 04.
Article in English | MEDLINE | ID: mdl-28509371

ABSTRACT

Human induced pluripotent stem-cell-derived cardiomyocytes (hiPSC CMs) may be used in regenerative medicine for individualized tissue transplants in the future. For application in patients, the generated CMs have to be highly pure and well characterized. In order to overcome the prevalent scarcity of CM-specific markers, we quantitatively assessed cell-surface-exposed sialo-glycoproteins and N-glycans of hiPSCs, CM progenitors, and CMs. Applying a combination of metabolic labeling and specific sialo-glycoprotein capture, we could highly enrich and quantify membrane proteins during cardiomyogenic differentiation. Among them we identified a number of novel, putative biomarkers for hiPSC CMs. Analysis of the N-glycome by capillary gel electrophoresis revealed three novel structures comprising ß1,3-linked galactose, α2,6-linked sialic acid and complex fucosylation; these were highly specific for hiPSCs. Bisecting GlcNAc structures strongly increased during differentiation, and we propose that they are characteristic of early, immature CMs.


Subject(s)
Cell Membrane/chemistry , Glycomics/methods , Induced Pluripotent Stem Cells/chemistry , Myocytes, Cardiac/chemistry , Polysaccharides/chemistry , Acetylglucosamine/chemistry , Acetylglucosamine/metabolism , Carbohydrate Sequence , Cell Differentiation , Cell Membrane/metabolism , Ciliary Neurotrophic Factor Receptor alpha Subunit/genetics , Ciliary Neurotrophic Factor Receptor alpha Subunit/metabolism , Fucose/chemistry , Fucose/metabolism , Galactose/chemistry , Galactose/metabolism , Gastrins/genetics , Gastrins/metabolism , Gene Expression Regulation , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Laminin/genetics , Laminin/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Polysaccharides/metabolism , Receptor, EphA7/genetics , Receptor, EphA7/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Sialic Acids/chemistry , Sialic Acids/metabolism , Staining and Labeling/methods
19.
Exp Hematol ; 48: 72-78, 2017 04.
Article in English | MEDLINE | ID: mdl-27988259

ABSTRACT

The proliferation, differentiation, adhesion, and migration of hematopoietic stem and progenitor cells (HSPCs) are dependent upon bone marrow stromal cells (BMSCs). In this study, we found that human primitive HSPCs (CD34+CD38-), but not lineage-committed hematopoietic cell populations, express the tyrosine kinase receptors EphA5 and EphA7. Moreover, we found that the ephrinA5 ligand, the high-affinity binding partner of EphA5 and EphA7, is highly expressed by primary human BMSCs. Previous studies have reported that interactions between EphA and ephrinA play important roles in hematopoietic cell trafficking; however, their role in BMSC support of hematopoiesis had not been described previously. Herein, we show that stimulating EphA5 and/or EphA7 forward signaling in HSPCs using soluble ephrinA5-Fc molecules promoted human HSPC-derived colony formation significantly and was associated with increased expression of granulocyte macrophage colony-stimulating factor receptor on HSPCs. Studies using functional blocking peptides to EphA5/7 found that disruption of EphA5/ephrinA5 and/or EphA7/ephrinA5 interactions inhibited HSPC function in BMSC-dependent long-term culture-initiating cell assays. Furthermore, the adhesion and migration of HSPCs was increased significantly in the presence of ephrinA5-Fc molecules compared with human immunoglobulin G-treated controls. Conversely, blocking EphA5 activation led to a reduction of HSPC adhesion, whereas inhibiting EphA5 and/or EphA7 activation hindered HSPC migration. Analysis of HSPC cultured in the presence of ephrinA5-Fc showed that EphA forward signaling stimulated Rac1 gene and protein expression and the Rac1 target molecule WAVE1. Moreover, a significant reduction of ephrinA5-mediated HSPC adhesion and migration was observed in the presence of Rac1 inhibitor. These findings suggest that interactions between EphA and ephrinA5 are important in maintaining the HSPC niche mediated in part by activation of Rac1 signaling.


Subject(s)
Cell Movement , Cell Self Renewal , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Receptor, EphA5/metabolism , Receptor, EphA7/metabolism , Signal Transduction , rac1 GTP-Binding Protein/metabolism , Cell Adhesion/genetics , Cell Communication , Cell Differentiation , Cell Movement/genetics , Cell Self Renewal/genetics , Gene Expression Profiling , Humans , Receptor, EphA5/genetics , Receptor, EphA7/genetics , Stem Cells , Stromal Cells/metabolism
20.
Biochem Biophys Res Commun ; 479(4): 759-765, 2016 Oct 28.
Article in English | MEDLINE | ID: mdl-27693790

ABSTRACT

Eph receptor tyrosine kinases (RTKs) and their ephrin ligands play multiple roles in the developing nervous system, including cell segregation, axon guidance and synaptic plasticity. Here we report the expression and function of EphA7 in Xenopus hindbrain development. EphA7 is specifically expressed in the hindbrain throughout neurulation in Xenopus embryos. Knockdown of EphA7 by specific morpholino oligonucleotide (MO) disrupted cranial neural tube closure and disturbed apical constriction of hindbrain neuroepithelial cells, indicating weakened cell surface tension. In neural plate explants, EphA7 knockdown inhibited apical filamentous actin (F-actin) accumulation. We further showed that EphA7 is involved in the phosphorylation and activation of focal adhesion kinase (FAK) in vivo and in vitro, a key regulator of actin assembly. Our findings reveal that EphA7 functions as a critical regulator of apical constriction of hindbrain neuroepithelial cells.


Subject(s)
Neurulation/physiology , Receptor, EphA7/metabolism , Xenopus Proteins/metabolism , Xenopus laevis/embryology , Xenopus laevis/metabolism , Animals , Animals, Genetically Modified , Cell Adhesion/genetics , Cell Adhesion/physiology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Neuroepithelial Cells/metabolism , Neurulation/genetics , Phosphorylation , Receptor, EphA7/antagonists & inhibitors , Receptor, EphA7/genetics , Rhombencephalon/embryology , Rhombencephalon/metabolism , Xenopus Proteins/antagonists & inhibitors , Xenopus Proteins/genetics , Xenopus laevis/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...