Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Article in English | MEDLINE | ID: mdl-39297461

ABSTRACT

OBJECTIVE: This study aimed to investigate the differential expression of mitophagyrelated genes in osteosarcoma patients with distinct prognostic outcomes and explore potential molecular regulatory mechanisms. METHODS: We analyzed microarray data from metastatic and nonmetastatic osteosarcoma patients using the UCSC dataset. Differential gene screening and intersection of mitophagy-related genes were performed using NetworkAnalyst. Random forest and LASSO regression were employed to screen selected genes and establish a risk prediction model. Functional enrichment analysis, protein- protein interaction (PPI) networks, immunoassays, and in vitro experiments were conducted to validate the findings. RESULTS: Seven differentially expressed genes were identified, and a robust risk prediction model was developed (AUC=0.886). PPI and functional enrichment analyses provided insights into relevant molecules and regulatory pathways. The immunoassay results revealed differences in the immune environment between the metastatic and nonmetastatic groups. Immunohistochemistry demonstrated significant downregulation of EPHA3 expression in the metastatic group, and in vitro experiments indicated that inhibiting EPHA3 increased the proliferative activity and migration ability of osteosarcoma cells. CONCLUSION: Our study suggests that the downregulation of EPHA3 may contribute to mitochondrial autophagy dysfunction, thereby increasing the risk of osteosarcoma metastasis.

2.
Int Immunopharmacol ; 142(Pt A): 113028, 2024 Sep 02.
Article in English | MEDLINE | ID: mdl-39226824

ABSTRACT

BACKGROUND: Pulmonary hypertension is a serious disease. Emerging studies have shown that M2 macrophages play an essential role in pulmonary hypertension; however, their mechanism of action is uncertain. METHODS: Four GEO datasets were downloaded. The differentially expressed genes (DEGs) were obtained using the limma package. Simultaneously, the Cell-type Identification By Estimating Relative Subsets Of RNA Transcripts (CIBERSORT) algorithm and weighted gene co-expression network analysis (WGCNA) were used to get the information about M2 macrophage-related modules. Potential key genes were obtained by intersecting DEGs with M2 macrophage-related module genes (M2MRGs), and finally the area under the curve (AUC) was calculated. Rats were exposed to hypoxia condition (10 % O2) for 4 weeks to induce PH. Subsequently, potential key genes with AUC>0.7 were analyzed by quantitative real-time polymerase chain reaction and Western blot using normoxia and hypoxia rat lungs. We knocked down EPHA3 in Raw264.7 cells and detected the protein expression of M2 macrophage markers including arginase 1 (ARG1) and interleukin 10 (IL-10), phospho-protein kinase B (P-Akt), and protein kinase B (Akt) to explore the downstream pathways of EPHA3. RESULTS: Seven potential hub genes were detected by intersecting M2MRGs and DEGs. Six genes with AUC values above 0.7 were used for further exploration. The expression of EPHA3 mRNA and protein was significantly more upregulated in rats with hypoxia than in rats with normoxia. The expression levels of IL10, ARG1, and P-Akt/Akt decreased after knocking down EPHA3. CONCLUSIONS: This study suggested that the activation of the P-Akt/Akt signaling pathway promoted by EPHA3 played an essential role in the progression of pulmonary hypertension.

3.
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
4.
J Exp Clin Cancer Res ; 42(1): 164, 2023 Jul 12.
Article in English | MEDLINE | ID: mdl-37434266

ABSTRACT

BACKGROUND: The receptor for advanced glycation-end products (RAGE) and its ligands have been implicated in obesity and associated inflammatory processes as well as in metabolic alterations like diabetes. In addition, RAGE-mediated signaling has been reported to contribute to the metastatic progression of breast cancer (BC), although mechanistic insights are still required. Here, we provide novel findings regarding the transcriptomic landscape and the molecular events through which RAGE may prompt aggressive features in estrogen receptor (ER)-positive BC. METHODS: MCF7 and T47D BC cells stably overexpressing human RAGE were used as a model system to evaluate important changes like cell protrusions, migration, invasion and colony formation both in vitro through scanning electron microscopy, clonogenic, migration and invasion assays and in vivo through zebrafish xenografts experiments. The whole transcriptome of RAGE-overexpressing BC cells was screened by high-throughput RNA sequencing. Thereafter, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses allowed the prediction of potential functions of differentially expressed genes (DEGs). Flow cytometry, real time-PCR, chromatin immunoprecipitation, immunofluorescence and western blot assays were performed to investigate the molecular network involved in the regulation of a novel RAGE target gene namely EphA3. The clinical significance of EphA3 was explored in the TCGA cohort of patients through the survivALL package, whereas the pro-migratory role of EphA3 signaling was ascertained in both BC cells and cancer-associated fibroblasts (CAFs). Statistical analysis was performed by t-tests. RESULTS: RNA-seq findings and GSEA analysis revealed that RAGE overexpression leads to a motility-related gene signature in ER-positive BC cells. Accordingly, we found that RAGE-overexpressing BC cells exhibit long filopodia-like membrane protrusions as well as an enhanced dissemination potential, as determined by the diverse experimental assays. Mechanistically, we established for the first time that EphA3 signaling may act as a physical mediator of BC cells and CAFs motility through both homotypic and heterotypic interactions. CONCLUSIONS: Our data demonstrate that RAGE up-regulation leads to migratory ability in ER-positive BC cells. Noteworthy, our findings suggest that EphA3 may be considered as a novel RAGE target gene facilitating BC invasion and scattering from the primary tumor mass. Overall, the current results may provide useful insights for more comprehensive therapeutic approaches in BC, particularly in obese and diabetic patients that are characterized by high RAGE levels.


Subject(s)
Breast Neoplasms , Receptor for Advanced Glycation End Products , Receptor, EphA3 , Animals , Female , Humans , Breast Neoplasms/genetics , Receptor, EphA3/genetics , Signal Transduction , Zebrafish/genetics
5.
J Neurogenet ; 37(3): 93-102, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37129498

ABSTRACT

A great amount of reaches have confirmed that circular RNAs (circRNAs) are novel regulators in glioma progression. Here, our work aimed to probe the specific role of circ_CLIP2 in glioma. The mRNA and protein expressions were analyzed by qRT-PCR and western blot, respectively. Cell viability, migration, invasion and apoptosis were examined by MTT assay, tranwell and flow cytometry assays, respectively. Moreover, the binding relationships between circ_CLIP2, microRNA (miR)-641 and erythropoietin-producing human hepatocellular (Eph)A3 were verified by dual luciferase reporter gene assay and/or RIP assay. The following data showed that circ_CLIP2 and EPHA3 were markedly increased in glioma tissues and cells, while miR-647 was downregulated. Gain- and loss-of-function experiments discovered that circ_CLIP2 knockdown remarkably inhibited cell proliferation, migration and invasion and promoted cell apoptosis of glioma cells, while these effects of circ_CLIP2 knockdown were abolished by miR-641 inhibition. Circ_CLIP2 was proved as a sponge of miR-641 to competitively upregulate EPHA3 expression. In addition, EPHA3 overexpression could abolish the inhibitory effects of miR-641 overexpression on the malignant behaviors of glioma cells by activating the signal transducer and activator of transcription 3 (STAT3). These findings elucidated that circ_CLIP2 knockdown suppressed glioma development by regulation of the miR-641/EP HA3/STAT3 axis, which provided a novel mechanism for understanding the pathogenesis of glioma.

6.
Open Med (Wars) ; 17(1): 2098-2108, 2022.
Article in English | MEDLINE | ID: mdl-36578556

ABSTRACT

Melanoma is a rare, fatal type of skin tumor. Although EPH receptor A3 (EphA3) is deregulated in melanoma, its detailed role remained uncharacterized. Using real time quantitative PCR analysis and western blotting, EphA3 was identified to be upregulated in melanoma tissues and cells, while miR-3666 showed an opposite expression trend. Cell counting kit-8, scratch wound, and in vivo assays proved that EphA3 silence inhibited the melanoma cell proliferation and migration and retarded tumor growth in vivo. Furthermore, western blotting results displayed that EphA3 silence resulted in a low expression of p38-MAPK and p-ERK1/2. Mechanically, miR-3666 was proved to target EphA3 3'UTR by the luciferase reporter assay. Furthermore, miR-3666 mimic compromised the driven melanoma cell proliferation and migration by EphA3 overexpression. In addition, induction of ERK1/2 and p38 MAPK pathways offset the positive effect of EphA3 overexpression on melanoma cells. In conclusion, miR-3666 downregulated EphA3 expression and retarded melanoma malignancy via inactivating ERK1/2 and p38 MAPK pathways. Hence, miR-3666/EphA3 axis may represent a druggable target against melanoma progression.

7.
Front Pharmacol ; 13: 965789, 2022.
Article in English | MEDLINE | ID: mdl-36059989

ABSTRACT

Introduction: Temozolomide (TMZ) is the first-line drug for glioblastoma (GBM), but it is limited in clinical use due to the drug resistance, poor brain targeting, and side effects. Temozolomide hexadecyl ester (TMZ16e), a TMZ derivative with high lipophilicity, membrane permeability, and high anti-glioma properties, has the potential to reverse drug resistance. In this study, anti-ephrin type-A receptor 3 (EphA3) modified TMZ16e loaded nanoparticles (NPs) were prepared for targeted GBM therapy via intranasal administration to deliver TMZ16e to the brain, treat drug-resistant glioma effectively, and reduce peripheral toxicity. Methods: TMZ16e loaded NPs were prepared by emulsion solvent evaporation method followed by modified with anti-EphA3 (anti-EphA3-TMZ16e-NPs). In vitro evaluations were performed by an MTT assay and flow cytometry analysis. The orthotopic nude mice models were used to evaluate the anti-glioma effect in vivo. Additionally, we investigated the anti-drug resistant mechanism by western blot analysis. Results: The particle size of the prepared NPs was less than 200 nm, and the zeta potential of TMZ16e-NPs and anti-EphA3-TMZ16e-NPs were -23.05 ± 1.48 mV and -28.65 ± 1.20mV, respectively, which is suitable for nasal delivery. In vitro studies have shown that anti-EphA3 modification increased the cellular uptake of nanoparticles in T98G cells. The cytotoxicity in the anti-EphA3-TMZ16e-NPs treated group was significantly higher than that of the TMZ16e-NPs, TMZ16e, and TMZ groups (p < 0.01), and the cell cycle was blocked. Western blotting analysis showed that the TMZ16e-loaded NPs were able to effectively downregulate the expression level of O6-methylguanine-deoxyribonucleic acid-methyltransferase (MGMT) protein in T98G cells and reverse drug resistance. In vivo studies showed that the median survival time of tumor-bearing nude mice in the anti-EphA3-TMZ16e-NPs group was extended to 41 days, which was 1.71-fold higher than that of the saline group and the TUNEL staining results of the brain tissue section indicated that the TMZ16e-loaded NPs could elevate apoptosis in T98G cells. Conclusion: In conclusion, the TMZ16e-loaded NPs can be effectively delivered to the brain and targeted to gliomas, exhibiting better anti-glioma activity, indicating they possess great potential in the treatment of drug-resistant glioma.

8.
Antioxidants (Basel) ; 11(5)2022 Apr 19.
Article in English | MEDLINE | ID: mdl-35624662

ABSTRACT

The development of targeted therapies (BRAF/MEK inhibitors) and immunotherapy have had a major impact on the treatment of melanoma. However, the majority of patients with advanced melanomas succumb to their disease. The mechanisms of resistance to both targeted therapies and immunotherapies are numerous and have been well-described. These include the alternative activation of BRAF/MEK signaling, novel compensating mutations in additional oncogenes, and loss of neoantigens. There has been limited development of small molecules that target alternative pathways in melanoma in the last two decades. We have previously identified triphenylmethanes as a class that shows activity against a wide variety of tumors. We have synthesized a novel triphenylmethane, indolium 1, and demonstrated its efficacy against an aggressive vemurafenib-resistant melanoma in vivo. Indolium 1 has a novel mechanism of action against melanoma, in that it results in induction of the tumor-suppressor EPHA3. We believe that pre-IND studies are warranted for this novel compound, given its mechanism of action and ability to inhibit the growth of vemurafenib resistant melanoma in vivo.

9.
J Biomed Res ; 37(3): 179-193, 2022 Nov 28.
Article in English | MEDLINE | ID: mdl-37013864

ABSTRACT

Erythropoietin-producing hepatocellular carcinoma A3 (EphA3) is a member of the largest subfamily of tyrosine kinase receptors-Eph receptors. Previous studies have shown that EphA3 is associated with tissue development. Recently, we have found that the expression of EphA3 is elevated in the hypothalamus of mice with diet-induced obesity (DIO). However, the role of EphA3 in hypothalamic-controlled energy metabolism remains unclear. In the current study, we demonstrated that the deletion of EphA3 in the hypothalamus by CRISPR/Cas9-mediated gene editing promotes obesity in male mice with high-fat diet feeding rather than those with normal chow diet feeding. Moreover, the deletion of hypothalamic EphA3 promotes high-fat DIO by increasing food intake and reducing energy expenditure. Knockdown of EphA3 leads to smaller intracellular vesicles in GT1-7 cells. The current study reveals that hypothalamic EphA3 plays important roles in promoting DIO.

10.
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
11.
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
12.
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
13.
Curr Cancer Drug Targets ; 20(1): 76-83, 2020.
Article in English | MEDLINE | ID: mdl-31433759

ABSTRACT

BACKGROUND: Src associated with mitosis of 68 kDa (Sam68), is often highly expressed in human cancers. Overexpression of Sam68 has been shown to be correlated with poor survival prognosis in some cancer patients. However, little is known whether Sam68 plays a role in promoting metastasis in breast cancer. MATERIALS AND METHODS: The expression of Sam68 protein in breast cancer tissue was detected by immunohistochemistry. Trans-well assay, wound-healing, real-time PCR and Western blotting analysis were used to detect the effect of Sam68 on promoting EMT or metastasis of breast cancer. Next-generation RNA sequencing was used to analyze genes that may be regulated by Sam68. RESULTS: Sam68 plays a positive role in promoting breast cancer metastasis. Sam68 was found to be overexpressed in breast cancer along with lymph node metastasis. MMP-9 was also found to be overexpressed in breast cancer tissue and was correlated to the expression of Sam68 (P<0.01). Xenograft in NOD/SCID mice and in vitro experiments confirmed that the invasion and metastatic ability of breast cancer cells were regulated by Sam68. And EPHA3 could be up-regulated by Sam68 in breast cancer. CONCLUSION: High expression of Sam68 participates in breast cancer metastasis by up-regulating the EPHA3 gene.


Subject(s)
Adaptor Proteins, Signal Transducing/physiology , Breast Neoplasms/pathology , DNA-Binding Proteins/physiology , RNA-Binding Proteins/physiology , Receptor, EphA3/physiology , Adult , Aged , Animals , Cell Line, Tumor , Disease Progression , Epithelial-Mesenchymal Transition , Female , Humans , Mice , Middle Aged , Neoplasm Invasiveness , Neoplasm Metastasis , Receptor, EphA3/genetics
14.
Acta Neuropathol ; 138(6): 1033-1052, 2019 12.
Article in English | MEDLINE | ID: mdl-31463571

ABSTRACT

Glioblastomas (GBMs) are malignant central nervous system (CNS) neoplasms with a very poor prognosis. They display cellular hierarchies containing self-renewing tumourigenic glioma stem cells (GSCs) in a complex heterogeneous microenvironment. One proposed GSC niche is the extracellular matrix (ECM)-rich perivascular bed of the tumour. Here, we report that the ECM binding dystroglycan (DG) receptor is expressed and functionally glycosylated on GSCs residing in the perivascular niche. Glycosylated αDG is highly expressed and functional on the most aggressive mesenchymal-like (MES-like) GBM tumour compartment. Furthermore, we found that DG acts to maintain an MES-like state via tight control of MAPK activation. Antibody-based blockade of αDG induces robust ERK-mediated differentiation leading to reduced GSC potential. DG was shown to be required for tumour initiation in MES-like GBM, with constitutive loss significantly delaying or preventing tumourigenic potential in-vivo. These findings reveal a central role of the DG receptor, not only as a structural element, but also as a critical factor promoting MES-like GBM and the maintenance of GSCs residing in the perivascular niche.


Subject(s)
Brain Neoplasms/metabolism , Dystroglycans/metabolism , Glioma/metabolism , Neoplastic Stem Cells/metabolism , Tumor Microenvironment/physiology , Animals , Brain Neoplasms/blood supply , Brain Neoplasms/surgery , Cell Transformation, Neoplastic , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Glioma/blood supply , Glioma/surgery , Humans , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation
15.
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
16.
J Clin Lab Anal ; 33(5): e22871, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30958616

ABSTRACT

BACKGROUND: This study aimed to preliminarily assess the relationship between erythropoietin-producing hepatocellular carcinoma receptor A3 (EphA3) and androgen receptor (AR) protein expression levels and prognosis in prostate cancer (PCa) to better understand the role of EphA3 in the prognosis and progression of PCa. MATERIALS: We investigated the expression of EphA3 and AR in human PCa by immunohistochemistry. RESULTS: EphA3 and AR were both significantly upregulated in PCa, with expression mainly localized to the nucleus. A high level of AR expression was found in 48.4% of 64 tumor samples, which was significantly more than in the adjacent tissue samples (15.6%) (P < 0.01). The percentage of samples expressing a high level of EphA3 was significantly greater in the PCa samples (54.7%) than in the adjacent tissue samples (20.3%) for the 64 tumors (P < 0.01). The high levels of EphA3 and AR expression in the PCa tissue samples were both correlated with the pathological stage, bladder and rectal invasion, distant metastasis, and preoperative PSA level (both P < 0.05). The survival time was significantly shorter in high levels of AR expression of patients. (P < 0.01). A high level of EphA3 in PCa patients suggests a poor prognosis (P < 0.05). Biochemical recurrence, distant metastasis, and the final scores of EphA3 and AR expression were significantly correlated with the prognosis of PCa (P < 0.05). CONCLUSIONS: Increased EphA3 expression is an independent prognostic factor for a poor outcome and decreased survival in PCa.


Subject(s)
Prostatic Neoplasms/mortality , Prostatic Neoplasms/surgery , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Androgen/metabolism , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Prostatectomy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptor, EphA3 , Retrospective Studies
17.
Dig Dis Sci ; 64(6): 1514-1522, 2019 06.
Article in English | MEDLINE | ID: mdl-30560328

ABSTRACT

BACKGROUND: EphA3 is a member of Eph receptors, which is involved in tumorigenesis. The expression and clinical significance of EphA3 in colorectal cancer (CRC) have not been fully investigated. METHODS: Four colon cancer cell lines and a set of CRC tissues were examined for EphA3 expression. The methylation status of a CpG island within the EphA3 promoter, the presence of four somatic EPHA3 mutations, and EPHA3 gene copy number variations were also analyzed in colon cancer cell lines. RESULTS: EphA3 expression was lost in all colon cancer cell lines examined. EphA3 expression was lower in tumor tissues when compared with normal intestinal tissues (P < 0.001). A comparison of EphA3 immunohistochemical scores for tumor and matched normal intestinal tissues revealed that the protein was downregulated in 82/164 (50.0%), unchanged in 52/164 (31.7%), and upregulated in 30/164 (18.3%) cases of CRC. EphA3 expression was negatively associated with lymph node metastasis (P =0.014, rs=- 0.192) and TNM stage (P =0.001, rs=- 0.260). Downregulation of expression was more common in older patients (P =0.013, rs=0.193). Methylated promoter DNA was detected in all four colon cancer cell lines. Somatic mutations or EphA3 gene deletion was not detected. CONCLUSIONS: EphA3 was downregulated in the majority of CRC. Hypermethylation of a CpG island within the EPHA3 promoter provides a possible mechanism. Loss of EphA3 expression was associated with lymph node metastasis and TNM stage and may therefore prove useful as a predictor for tumor spread in CRC.


Subject(s)
Colorectal Neoplasms/genetics , DNA Methylation , Receptor Protein-Tyrosine Kinases/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , CpG Islands , Down-Regulation , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , Promoter Regions, Genetic , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, EphA3
18.
Int J Oncol ; 54(2): 722-732, 2019 02.
Article in English | MEDLINE | ID: mdl-30483759

ABSTRACT

Eph receptor tyrosine kinases are critical for cell­cell communication during normal and oncogenic development. Eph receptor A3 (EphA3) expression is associated with tumor promotion in certain types of cancer; however, it acts as a tumor suppressor in others. The expression levels of EphA3 and its effects on tumor progression in esophageal squamous cell carcinoma (ESCC) cell lines were determined using reverse transcription­quantitative polymerase chain reaction analysis and a Transwell invasion assay. The present study demonstrated that EphA3 expression was decreased in ESCC tissues and cell lines. Treatment with the DNA methylation inhibitor 5­aza­2'­deoxycytidine increased the mRNA expression levels of EphA3 in the ESCC cell lines KYSE510 and KYSE30. In addition, overexpression of EphA3 in KYSE450 and KYSE510 cells inhibited cell migration and invasion. EphA3 overexpression also decreased RhoA GTPase. Furthermore, EphA3 overexpression induced mesenchymal­epithelial transition, as demonstrated by epithelial­like morphological alterations, increased expression of epithelial proteins (E­cadherin and the tight junction protein 1 zonula occludens­1) and decreased expression of mesenchymal proteins (Vimentin, N­cadherin and Snail). Conversely, silencing EphA3 in KYSE410 cells triggered epithelial­mesenchymal transition, and promoted cell migration and invasion. These results suggested that EphA3 may serve a tumor­suppressor role in ESCC.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Esophageal Squamous Cell Carcinoma/genetics , Receptor Protein-Tyrosine Kinases/genetics , rhoA GTP-Binding Protein/genetics , Apoptosis/genetics , Cadherins/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , DNA Methylation/genetics , Esophageal Squamous Cell Carcinoma/pathology , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Humans , Neoplasm Invasiveness/genetics , RNA, Messenger/genetics , Receptor, EphA3
19.
Biochem Biophys Res Commun ; 508(3): 715-721, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30528229

ABSTRACT

EphA3, a member of the Eph family of receptor tyrosine kinases, has been reported to be overexpressed in some human cancers including glioblastoma. Here, we found that expression of EphA3 is up-regulated in response to epidermal growth factor (EGF) stimulation and promotes formation of cell aggregates in suspension culture of glioblastoma cells. Suppression of EphA3 expression by short hairpin RNA-mediated knockdown or CRISPR/Cas9-mediated gene deletion inhibited EGF-induced promotion of cell aggregate formation, whereas overexpression of EphA3 promoted formation of cell aggregates in suspension culture. EGF-induced EphA3 expression and promotion of cell aggregate formation required Akt activity. Furthermore, N-cadherin, whose expression was regulated by EGF and EphA3, contributed to the formation of cell aggregates in suspension culture. These results suggest that the regulation of EphA3 expression plays a critical role in glioblastoma cell growth in non-adherent conditions.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Epidermal Growth Factor/pharmacology , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/pathology , Receptor Protein-Tyrosine Kinases/genetics , Up-Regulation/genetics , Cadherins/metabolism , Cell Aggregation/drug effects , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Proto-Oncogene Proteins c-akt/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, EphA3 , Suspensions , Up-Regulation/drug effects
20.
Cancers (Basel) ; 10(12)2018 Dec 17.
Article in English | MEDLINE | ID: mdl-30562956

ABSTRACT

The EphA3 receptor has recently emerged as a functional tumour-specific therapeutic target in glioblastoma (GBM). EphA3 is significantly elevated in recurrent disease, is most highly expressed on glioma stem cells (GSCs), and has a functional role in maintaining self-renewal and tumourigenesis. An unlabelled EphA3-targeting therapeutic antibody is currently under clinical assessment in recurrent GBM patients. In this study, we assessed the efficacy of EphA3 antibody drug conjugate (ADC) and radioimmunotherapy (RIT) approaches using orthotopic animal xenograft models. Brain uptake studies, using positron emission tomography/computed tomography (PET/CT) imaging, show EphA3 antibodies are effectively delivered across the blood-tumour barrier and accumulate at the tumour site with no observed normal brain reactivity. A robust anti-tumour response, with no toxicity, was observed using EphA3, ADC, and RIT approaches, leading to a significant increase in overall survival. Our current research provides evidence that GBM patients may benefit from pay-loaded EphA3 antibody therapies.

SELECTION OF CITATIONS
SEARCH DETAIL