Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 138
Filter
1.
Acta Oncol ; 62(12): 1757-1766, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37738252

ABSTRACT

BACKGROUND: Our previous study has revealed that EphA7 was upregulated in patient-derived esophageal squamous cell carcinoma (ESCC) xenografts with hyper-activated STAT3, but its mechanism was still unclear. MATERIALS AND METHODS: To assess the association between EphA7 and STAT3, western blotting, immunofluorescence, ChIP assay, and qRT-PCR were conducted. Truncated mutation and luciferase assay were performed to examine the promoter activity of EphA7. CCK-8 assay and colony formation were performed to assess the proliferation of ESCC. Cell-derived xenograft models were established to evaluate the effects of EphA7 on ESCC tumor growth. RNA-seq analyses were used to assess the effects of EphA7 on related signals. RESULTS: In this study, EphA7 was found upregulated in ESCC cell lines with high STAT3 activation, and immunofluorescence also showed that EphA7 was co-localized with phospho-STAT3 in ESCC cells. Interestingly, suppressing STAT3 activation by the STAT3 inhibitor Stattic markedly inhibited the protein expression of EphA7 in ESCC cells, in contrast, activation of STAT3 by IL-6 obviously upregulated the protein expression of EphA7. Moreover, the transcription of EphA7 was also mediated by the activation of STAT3 in ESCC cells, and the -2000∼-1500 region was identified as the key promoter of EphA7. Our results also indicated that EphA7 enhanced the cell proliferation of ESCC, and silence of EphA7 significantly suppressed ESCC tumor growth. Moreover, EphA7 silence markedly abolished STAT3 activation-derived cell proliferation of ESCC. Additionally, RNA-seq analyses indicated that several tumor-related signaling pathways were significantly changed after EphA7 downregulation in ESCC cells. CONCLUSION: Our results showed that the transcriptional expression of EphA7 was increased by activated STAT3, and the STAT3 signaling may act through EphA7 to promote the development of ESCC.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Receptor, EphA7 , STAT3 Transcription Factor , Humans , Cell Line, Tumor , Cell Movement , Cell Proliferation/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Gene Expression Regulation, Neoplastic , Signal Transduction , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Receptor, EphA7/metabolism
2.
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
3.
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
4.
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
5.
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
6.
Arch Physiol Biochem ; 128(5): 1181-1187, 2022 Oct.
Article in English | MEDLINE | ID: mdl-32421395

ABSTRACT

MicroRNAs (miRNAs) have critical roles in colorectal cancer (CRC) tumorigenesis and development. It has been reported that Eph receptor A7 (EphA7) was a potential target of miR-944 which is transcriptionally activated in cancer. The aim of this study was to explore the expression profile of miR-944 and its target gene EPHA7 in the serum of Egyptian CRC patients. 150 CRC patients, 50 adenomatous polyps (AP) patients, and 100 healthy controls were included. Serum miR-944 was downregulated (0.304 ± 0.0512) while serum EPHA7 was upregulated (3.163 ± 0.610) in CRC and AP patients versus controls and discriminated aganst these groups by Receiver operating characteristic curve (ROC) analysis. miR-944 presented the highest diagnostic accuracy for CRC patients from control (AUC = 0.90). Moreover obvious prognostic power in distinguishing AP from CRC (AUC = 0.87). In conclusion, miR-944 and EPHA7 are potential genetic markers of CRC predisposition and novel potential non-invasive diagnostic biomarkers for CRC.


Subject(s)
Circulating MicroRNA , Colorectal Neoplasms , MicroRNAs , Biomarkers, Tumor/genetics , Circulating MicroRNA/genetics , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Genetic Markers , Humans , MicroRNAs/genetics , Receptor, EphA1/genetics , Receptor, EphA7
7.
Cereb Cortex ; 32(11): 2321-2331, 2022 05 30.
Article in English | MEDLINE | ID: mdl-34546353

ABSTRACT

Soma spacing and dendritic arborization during brain development are key events for the establishment of proper neural circuitry and function. Transcription factor Satb2 is a molecular node in regulating the development of the cerebral cortex, as shown by the facts that Satb2 is required for the regionalization of retrosplenial cortex, the determination of callosal neuron fate, and the regulation of soma spacing and dendritic self-avoidance of cortical pyramidal neurons. In this study, we explored downstream effectors that mediate the Satb2-implicated soma spacing and dendritic self-avoidance. First, RNA-seq analysis of the cortex revealed differentially expressed genes between control and Satb2 CKO mice. Among them, EphA7 transcription was dramatically increased in layers II/III of Satb2 CKO cortex. Overexpression of EphA7 in the late-born cortical neurons of wild-type mice via in utero electroporation resulted in soma clumping and impaired self-avoidance of affected pyramidal neurons, which resembles the phenotypes caused by knockdown of Satb2 expression. Importantly, the phenotypes by Satb2 knockdown was rescued by reducing EphA7 expression in the cortex. Finally, ChIP and luciferase reporter assays indicated a direct suppression of EphA7 expression by Satb2. These findings provide new insights into the complexity of transcriptional regulation of the morphogenesis of cerebral cortex.


Subject(s)
Cerebral Cortex , Neurons , Animals , Cell Body/metabolism , Cerebral Cortex/metabolism , Matrix Attachment Region Binding Proteins , Mice , Neurons/metabolism , Pyramidal Cells/metabolism , Receptor, EphA7 , Transcription Factors/metabolism
8.
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
9.
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
10.
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
11.
J Neurosci ; 41(22): 4795-4808, 2021 06 02.
Article in English | MEDLINE | ID: mdl-33906900

ABSTRACT

Coordination of skilled movements and motor planning relies on the formation of regionally restricted brain circuits that connect cortex with subcortical areas during embryonic development. Layer 5 neurons that are distributed across most cortical areas innervate the pontine nuclei (basilar pons) by protrusion and extension of collateral branches interstitially along their corticospinal extending axons. Pons-derived chemotropic cues are known to attract extending axons, but molecules that regulate collateral extension to create regionally segregated targeting patterns have not been identified. Here, we discovered that EphA7 and EfnA5 are expressed in the cortex and the basilar pons in a region-specific and mutually exclusive manner, and that their repulsive activities are essential for segregating collateral extensions from corticospinal axonal tracts in mice. Specifically, EphA7 and EfnA5 forward and reverse inhibitory signals direct collateral extension such that EphA7-positive frontal and occipital cortical areas extend their axon collaterals into the EfnA5-negative rostral part of the basilar pons, whereas EfnA5-positive parietal cortical areas extend their collaterals into the EphA7-negative caudal part of the basilar pons. Together, our results provide a molecular basis that explains how the corticopontine projection connects multimodal cortical outputs to their subcortical targets.SIGNIFICANCE STATEMENT Our findings put forward a model in which region-to-region connections between cortex and subcortical areas are shaped by mutually exclusive molecules to ensure the fidelity of regionally restricted circuitry. This model is distinct from earlier work showing that neuronal circuits within individual cortical modalities form in a topographical manner controlled by a gradient of axon guidance molecules. The principle that a shared molecular program of mutually repulsive signaling instructs regional organization-both within each brain region and between connected brain regions-may well be applicable to other contexts in which information is sorted by converging and diverging neuronal circuits.


Subject(s)
Axon Guidance/physiology , Ephrin-A5/metabolism , Neocortex/embryology , Neural Pathways/embryology , Pons/embryology , Receptor, EphA7/metabolism , Animals , Female , Male , Mice , Mice, Inbred C57BL , Neocortex/metabolism , Neural Pathways/metabolism , Pons/pathology
12.
BMC Med ; 19(1): 26, 2021 02 02.
Article in English | MEDLINE | ID: mdl-33526018

ABSTRACT

BACKGROUND: A critical and challenging process in immunotherapy is to identify cancer patients who could benefit from immune checkpoint inhibitors (ICIs). Exploration of predictive biomarkers could help to maximize the clinical benefits. Eph receptors have been shown to play essential roles in tumor immunity. However, the association between EPH gene mutation and ICI response is lacking. METHODS: Clinical data and whole-exome sequencing (WES) data from published studies were collected and consolidated as a discovery cohort to analyze the association between EPH gene mutation and efficacy of ICI therapy. Another independent cohort from Memorial Sloan Kettering Cancer Center (MSKCC) was adopted to validate our findings. The Cancer Genome Atlas (TCGA) cohort was used to perform anti-tumor immunity and pathway enrichment analysis. RESULTS: Among fourteen EPH genes, EPHA7-mutant (EPHA7-MUT) was enriched in patients responding to ICI therapy (FDR adjusted P < 0.05). In the discovery cohort (n = 386), significant differences were detected between EPHA7-MUT and EPHA7-wildtype (EPHA7-WT) patients regarding objective response rate (ORR, 52.6% vs 29.1%, FDR adjusted P = 0.0357) and durable clinical benefit (DCB, 70.3% vs 42.7%, FDR adjusted P = 0.0200). In the validation cohort (n = 1144), significant overall survival advantage was observed in EPHA7-MUT patients (HR = 0.62 [95% confidence interval, 0.39 to 0.97], multivariable adjusted P = 0.0367), which was independent of tumor mutational burden (TMB) and copy number alteration (CNA). Notably, EPHA7-MUT patients without ICI therapy had significantly worse overall survival in TCGA cohort (HR = 1.33 [95% confidence interval, 1.06 to 1.67], multivariable adjusted P = 0.0139). Further gene set enrichment analysis revealed enhanced anti-tumor immunity in EPHA7-MUT tumor. CONCLUSIONS: EPHA7-MUT successfully predicted better clinical outcomes in ICI-treated patients across multiple cancer types, indicating that EPHA7-MUT could serve as a potential predictive biomarker for immune checkpoint inhibitors.


Subject(s)
Immune Checkpoint Inhibitors/metabolism , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Receptor, EphA7/metabolism , Biomarkers, Tumor/genetics , Cohort Studies , DNA Copy Number Variations , Humans , Immunotherapy , Male , Middle Aged , Mutation
13.
Braz J Med Biol Res ; 54(2): e9161, 2021.
Article in English | MEDLINE | ID: mdl-33439936

ABSTRACT

Patients with osteosarcoma (OS) usually have poor overall survival because of frequent metastasis. Long non-coding RNAs (lncRNAs) have been reported to be associated with tumorigenesis and metastasis. In this study, we investigated the expression and roles of lncRNA human histocompatibility leukocyte antigen (HLA) complex P5 (HCP5) in OS, aiming to provide a novel molecular mechanism for OS. HCP5 was up-regulated both in OS tissues and cell lines and high expression of HCP5 was associated to low survival in OS patients. Down-regulation of HCP5 inhibited cell proliferation, migration, and invasion, suggesting its carcinogenic role in OS. miR-101 was targeted by HCP5 and its expression was decreased in OS. The inhibitor of miR-101 reversed the impact of HCP5 down-regulation on cell proliferation, apoptosis, and metastasis in OS. Ephrin receptor 7 (EPHA7) was proved to be a target of miR-101 and had ability to recover the effects of miR-101 inhibitor in OS. In conclusion, lncRNA HCP5 knockdown suppressed cell proliferation, migration, and invasion, and induced apoptosis through depleting the expression of EPHA7 by binding to miR-101, providing a potential therapeutic strategy of HCP5 in OS.


Subject(s)
Bone Neoplasms , MicroRNAs/metabolism , Osteosarcoma , RNA, Long Noncoding/genetics , Receptor, EphA7/metabolism , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Cell Line, Tumor , Cell Movement , Cell Proliferation , Down-Regulation , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Osteosarcoma/genetics , Osteosarcoma/pathology
14.
Mol Cell Biochem ; 476(1): 213-220, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32914261

ABSTRACT

A soluble form of EphA7 (sEphA7) has been found to antagonize the role of full-length EphA7 (EphA7-FL) to stabilize the membrane level of the tight junction protein Claudin6 (CLDN6) during Xenopus pronephros development. However, the mechanism underlying this antagonistic effect remains unclear. In this study, we identified Nicalin, a Nicastrin-like protein, as a novel sEphA7-interacting protein using immunoprecipitation (IP)/mass spectrometry (MS). In HEK293 cells, Nicalin interacted with sEphA7 and they predominantly co-localized in the endoplasmic reticulum (ER). Interestingly, Nicalin diminished the protein level of sEphA7 in the membranous fraction but increased that in the insoluble cytoplasmic fraction with a reduced molecular weight, suggesting that Nicalin restricts the entry of sEphA7 into the ER for further modification. sEphA7 probably acted as a chaperone and enhanced the membrane level of EphA7-FL and the formation of EphA7 complex, however, this effect was reversed by Nicalin. Our work suggested that Nicalin limits sEphA7 secretion, thereby preventing the formation of EphA7 complex. These results demonstrated the potential role of Nicalin in regulating EphA7 expression and revealed a potential mechanism underlying the antagonistic effect between sEphA7 and EphA7-FL.


Subject(s)
Claudins/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Receptor, EphA7/metabolism , Animals , Biotinylation , Cell Membrane/metabolism , Cytoplasm/metabolism , Endoplasmic Reticulum/metabolism , HEK293 Cells , Humans , Signal Transduction , Xenopus , Xenopus Proteins/metabolism
15.
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
16.
Braz. j. med. biol. res ; 54(2): e9161, 2021. graf
Article in English | LILACS | ID: biblio-1153511

ABSTRACT

Patients with osteosarcoma (OS) usually have poor overall survival because of frequent metastasis. Long non-coding RNAs (lncRNAs) have been reported to be associated with tumorigenesis and metastasis. In this study, we investigated the expression and roles of lncRNA human histocompatibility leukocyte antigen (HLA) complex P5 (HCP5) in OS, aiming to provide a novel molecular mechanism for OS. HCP5 was up-regulated both in OS tissues and cell lines and high expression of HCP5 was associated to low survival in OS patients. Down-regulation of HCP5 inhibited cell proliferation, migration, and invasion, suggesting its carcinogenic role in OS. miR-101 was targeted by HCP5 and its expression was decreased in OS. The inhibitor of miR-101 reversed the impact of HCP5 down-regulation on cell proliferation, apoptosis, and metastasis in OS. Ephrin receptor 7 (EPHA7) was proved to be a target of miR-101 and had ability to recover the effects of miR-101 inhibitor in OS. In conclusion, lncRNA HCP5 knockdown suppressed cell proliferation, migration, and invasion, and induced apoptosis through depleting the expression of EPHA7 by binding to miR-101, providing a potential therapeutic strategy of HCP5 in OS.


Subject(s)
Humans , Bone Neoplasms/genetics , Bone Neoplasms/pathology , Osteosarcoma/genetics , Osteosarcoma/pathology , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , Down-Regulation , Gene Expression Regulation, Neoplastic , Cell Movement , Receptor, EphA7/metabolism , Cell Line, Tumor , Cell Proliferation , Neoplasm Invasiveness
17.
PLoS One ; 15(12): e0231561, 2020.
Article in English | MEDLINE | ID: mdl-33275600

ABSTRACT

The shape of a neuron facilitates its functionality within neural circuits. Dendrites integrate incoming signals from axons, receiving excitatory input onto small protrusions called dendritic spines. Therefore, understanding dendritic growth and development is fundamental for discerning neural function. We previously demonstrated that EphA7 receptor signaling during cortical development impacts dendrites in two ways: EphA7 restricts dendritic growth early and promotes dendritic spine formation later. Here, the molecular basis for this shift in EphA7 function is defined. Expression analyses reveal that EphA7 full-length (EphA7-FL) and truncated (EphA7-T1; lacking kinase domain) isoforms are dynamically expressed in the developing cortex. Peak expression of EphA7-FL overlaps with dendritic elaboration around birth, while highest expression of EphA7-T1 coincides with dendritic spine formation in early postnatal life. Overexpression studies in cultured neurons demonstrate that EphA7-FL inhibits both dendritic growth and spine formation, while EphA7-T1 increases spine density. Furthermore, signaling downstream of EphA7 shifts during development, such that in vivo inhibition of mTOR by rapamycin in EphA7-mutant neurons ameliorates dendritic branching, but not dendritic spine phenotypes. Finally, direct interaction between EphA7-FL and EphA7-T1 is demonstrated in cultured cells, which results in reduction of EphA7-FL phosphorylation. In cortex, both isoforms are colocalized to synaptic fractions and both transcripts are expressed together within individual neurons, supporting a model where EphA7-T1 modulates EphA7-FL repulsive signaling during development. Thus, the divergent functions of EphA7 during cortical dendrite development are explained by the presence of two variants of the receptor.


Subject(s)
Cerebral Cortex/embryology , Dendrites/metabolism , Receptor, EphA7/metabolism , Animals , Axons/metabolism , Cells, Cultured , Cerebral Cortex/metabolism , Dendritic Spines/metabolism , Male , Mice, Inbred C57BL/embryology , Neurons/metabolism , Organogenesis , Protein Isoforms/physiology , Rats , Rats, Sprague-Dawley/embryology , Receptor, EphA7/physiology , Signal Transduction
18.
Article in English | MEDLINE | ID: mdl-32843430

ABSTRACT

Although cutaneous squamous cell carcinoma (cSCC) is treatable in the majority of cases, deadly invasive and metastatic cases do occur. To date there are neither reliable predictive biomarkers of disease progression nor FDA-approved targeted therapies as standard of care. To address these issues, we screened patient-derived primary cultured cells from invasive/metastatic cSCC with 107 small-molecule inhibitors. In-house bioinformatics tools were used to cross-analyze drug responses and DNA mutations in tumors detected by whole-exome sequencing (WES). Aberrations in molecular pathways with evidence of potential drug targets were identified, including the Eph-ephrin and neutrophil degranulation signaling pathways. Using a screening panel of siRNAs, we identified EPHA6 and EPHA7 as targets within the Eph-ephrin pathway responsible for mitigating decreased cell viability. These studies form a plausible foundation for detecting biomarkers of high-risk progressive disease applicable in dermatopathology and for patient-specific therapeutic options for invasive/metastatic cSCC.


Subject(s)
Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/metabolism , Cell Survival/genetics , Disease Progression , Gene Expression/genetics , Gene Expression Regulation, Neoplastic/genetics , Genomics/methods , Humans , Male , Middle Aged , Mutation/genetics , Receptor, EphA6/antagonists & inhibitors , Receptor, EphA6/metabolism , Receptor, EphA7/antagonists & inhibitors , Receptor, EphA7/metabolism , Signal Transduction/genetics , Skin Neoplasms/genetics , Small Molecule Libraries/pharmacology , Exome Sequencing/methods
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...