Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 95
Filter
1.
Alzheimers Dement ; 20(3): 2016-2033, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38184788

ABSTRACT

INTRODUCTION: Genome-wide association studies link susceptibility to late-onset Alzheimer's disease (LOAD) with EphA1. Sequencing identified a non-synonymous substitution P460L as a LOAD risk variant. Other Ephs regulate vascular permeability and immune cell recruitment. We hypothesized that P460L dysregulates EphA1 receptor activity and impacts neuroinflammation. METHODS: EphA1/P460L receptor activity was assayed in isogenic Human Embryonic Kidney (HEK) cells. Soluble EphA1/P460L (sEphA1/sP460L) reverse signaling in brain endothelial cells was assessed by T-cell recruitment and barrier function assays. RESULTS: EphA1 and P460L were expressed in HEK cells, but membrane and soluble P460L were significantly reduced. Ligand engagement induced Y781 phosphorylation of EphA1 but not P460L. sEphA1 primed brain endothelial cells for increased T-cell recruitment; however, sP460L was less effective. sEphA1 decreased the integrity of the brain endothelial barrier, while sP460L had no effect. DISCUSSION: These findings suggest that P460L alters EphA1-dependent forward and reverse signaling, which may impact blood-brain barrier function in LOAD. HIGHLIGHTS: EphA1-dependent reverse signaling controls recruitment of T cells by brain endothelial cells. EphA1-dependent reverse signaling remodels brain endothelial cell contacts. LOAD-associated P460L variant of EphA1 shows reduced membrane expression and reduced ligand responses. LOAD-associated P460L variant of EphA1 fails to reverse signal to brain endothelial cells.


Subject(s)
Alzheimer Disease , Humans , Alzheimer Disease/genetics , Blood-Brain Barrier , Endothelial Cells , Genome-Wide Association Study , Ligands , Receptor, EphA1/metabolism
2.
Life Sci ; 326: 121796, 2023 Aug 01.
Article in English | MEDLINE | ID: mdl-37230378

ABSTRACT

Chronic pain is a common and debilitating condition with a huge social and economic burden worldwide. Currently, available drugs in clinics are not adequately effective and possess a variety of severe side effects leading to treatment withdrawal and poor quality of life. The ongoing search for new therapeutics with minimal side effects for chronic pain management remains a high research priority. Erythropoietin-producing human hepatocellular carcinoma cell receptor (Eph) is a tyrosine kinase receptor that is involved in neurodegenerative disorders, including pain. The Eph receptor interacts with several molecular switches, such as N methyl d-aspartate receptor (NMDAR), mitogen-activated protein kinase (MAPK), calpain 1, caspase 3, protein kinase a (PKA), and protein kinase Cy (PKCy), which in turn regulates pathophysiology of chronic pain. Here we highlight the emerging evidence of the Ephs/ephrin system as a possible near-future therapeutic target for the treatment of chronic pain and discuss the various mechanism of its involvement. We critically analyse the present status of Eph receptor system and conclude that extrapolating the pharmacological and genetic approaches using a strong therapeutic development framework could serve as next-generation analgesics for the management of chronic pain.


Subject(s)
Chronic Pain , Ephrins , Humans , Ephrins/metabolism , Receptor, EphA1/metabolism , Chronic Pain/drug therapy , Quality of Life , Signal Transduction
3.
Curr Med Chem ; 30(20): 2340-2353, 2023.
Article in English | MEDLINE | ID: mdl-35996244

ABSTRACT

BACKGROUND: Eph receptors tyrosine kinase (RTK) were identified in 1987 from hepatocellular carcinoma cell lines and were the largest known subfamily of RTK. Eph receptors can be divided into two categories, EphA and EphB, based on their structure and receptor-ligand specificity. EphA can be divided into 10 species (EphA 1-10) and EphB into 6 species (EphB1-6). Similarly, the ligands of Eph receptors are Ephrins. Ephrins also can be divided into Ephrin A and Ephrin B, of which there are five species(Ephrin-A1-5) and three species(Ephrin-B1-3). Among the Eph receptors, EphA1 has been the least studied so far. As far as we know, Eph receptors are involved in multiple pathologies, including cancer progression, tumor angiogenesis, intestinal environmental stability, the lymph node system, neurological disease, and inhibition of nerve regeneration after injury. There is a link between EphA1, integrin and ECM- related signal pathways. Ephrin-A1 is a ligand of the EphA1 receptor. EphA1 and ephrin-A1 functions are related to tumor angiogenesis. EphA1 and ephrin-A1 also play roles in gynecological diseases. Ephrin-A1 and EphA1 receptors regulate the follicular formation, ovulation, embryo transport, implantation and placental formation, which are of great significance for the occurrence of gynecological tumor diseases. EphA1 has been identified as an oncoprotein in various tumors and has been associated with the prognosis of various tumors in recent years. EphA1 is considered a driver gene in tumor genomics. There are significant differences in EphA1 expression levels in different types of normal tissues and tumors and even in different stages of tumor development, suggesting its functional diversity. Changes at the gene level in cell biology are often used as biological indicators of cancer, known as biomarkers, which can be used to provide diagnostic or prognostic information and are valuable for improving the detection, monitoring and treatment of tumors. However, few prognostic markers can selectively predict clinically significant tumors with poor prognosis. These malignancies are more likely to progress and lead to death, requiring more aggressive treatment. Currently available treatments for advanced cancer are often ineffective, and treatment options are mainly palliative. Therefore, early identification and treatment of those at risk of developing malignant tumors are crucial. Although pieces of evidence have shown the role of EphA1 in tumorigenesis and development, its specific mechanism is still unknown to a great extent. OBJECTIVE: This review reveals the changes and roles of EphA1 in many tumors and cancers. The change of EphA1 expression can be used as a biological marker of cancer, which is valuable for improving tumor detection, monitoring and treatment and can be applied to imaging. Studies have shown that structural modification of EphA1 could make it an effective new drug. EphA1 is unique in that it can be considered a prognostic marker in many tumors and is of important meaning for clinical diagnosis and operative treatment. At the same time, the study of the specific mechanism of EphA1 in tumors can provide a new way for targeted therapy. METHODS: Relevant studies were retrieved and collected through the PubMed system. After determining EphA1 as the research object, by analyzing research articles on EphA1 in the PubMed system in recent 10 years, we found that EphA1 was closely connected with the occurrence and development of tumors and further determined the references according to the influencing factors for review and analysis. RESULTS: EphA1 has been identified as a cancer protein in various tumors, such as hepatocellular carcinoma, nasopharyngeal carcinoma, ovarian cancer, gastric cancer, colorectal cancer, clear cell renal cell carcinoma, esophageal squamous cell carcinoma, breast cancer, prostate cancer and uveal melanoma. EphA1 is abnormally expressed in these tumor cells, which mainly plays a role in cancer progression, tumor angiogenesis, intestinal environmental stability, the lymph node system, nervous system diseases and gynecological diseases. In a narrow sense, EphA1 is especially effective in breast cancer in terms of gynecological diseases. However, the specific mechanism of EphA1 leading to the change of cancer cells in some tumors is not clear, which needs further research and exploration. CONCLUSION: RTK EphA1 can be used as a biomarker for tumor diagnosis (especially a prognostic marker), an indispensable therapeutic target for new anti-tumor therapies, and a novel anti-tumor drug.


Subject(s)
Breast Neoplasms , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Receptor, EphA2 , Pregnancy , Male , Humans , Female , Receptor, EphA1/genetics , Receptor, EphA1/analysis , Receptor, EphA1/metabolism , Ephrin-A1/metabolism , Ligands , Placenta/chemistry , Placenta/metabolism , Ephrins/genetics , Ephrins/analysis , Ephrins/metabolism , Receptors, Eph Family/genetics , Receptors, Eph Family/metabolism , Biomarkers , Receptor, EphA2/metabolism
4.
Cells ; 11(20)2022 10 19.
Article in English | MEDLINE | ID: mdl-36291158

ABSTRACT

The eye lens is a transparent, ellipsoid organ in the anterior chamber of the eye that is required for fine focusing of light onto the retina to transmit a clear image. Cataracts, defined as any opacity in the lens, remains the leading cause of blindness in the world. Recent studies in humans and mice indicate that Eph-ephrin bidirectional signaling is important for maintaining lens transparency. Specifically, mutations and polymorphisms in the EphA2 receptor and the ephrin-A5 ligand have been linked to congenital and age-related cataracts. It is unclear what other variants of Ephs and ephrins are expressed in the lens or whether there is preferential expression in epithelial vs. fiber cells. We performed a detailed analysis of Eph receptor and ephrin ligand mRNA transcripts in whole mouse lenses, epithelial cell fractions, and fiber cell fractions using a new RNA isolation method. We compared control samples with EphA2 knockout (KO) and ephrin-A5 KO samples. Our results revealed the presence of transcripts for 12 out of 14 Eph receptors and 8 out of 8 ephrin ligands in various fractions of lens cells. Using specific primer sets, RT-PCR, and sequencing, we verified the variant of each gene that is expressed, and we found two epithelial-cell-specific genes. Surprisingly, we also identified one Eph receptor variant that is expressed in KO lens fibers but is absent from control lens fibers. We also identified one low expression ephrin variant that is only expressed in ephrin-A5 control samples. These results indicate that the lens expresses almost all Ephs and ephrins, and there may be many receptor-ligand pairs that play a role in lens homeostasis.


Subject(s)
Cataract , Lens, Crystalline , Receptor, EphA2 , Humans , Mice , Animals , Ephrins/genetics , Ephrins/metabolism , Receptor, EphA1/metabolism , Ephrin-A5/genetics , Ephrin-A5/metabolism , Ligands , Receptor, EphA2/genetics , Receptor, EphA2/metabolism , Lens, Crystalline/metabolism , Receptors, Eph Family/genetics , Receptors, Eph Family/metabolism , Cataract/genetics , RNA, Messenger/metabolism
5.
J Immunol ; 209(9): 1796-1805, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36130827

ABSTRACT

The receptor tyrosine kinase EPHB2 (EPH receptor B2) is highly expressed in many human cancer types, especially in gastrointestinal cancers, such as colorectal cancer. Several coding mutations of the EPHB2 gene have been identified in many cancer types, suggesting that EPHB2 plays a critical role in carcinogenesis. However, the exact functional mechanism of EPHB2 in carcinogenesis remains unknown. In this study, we find that EPHB2 is required for TNF-induced signaling activation and proinflammatory cytokine production in colorectal epithelial cells. Mechanistically, after TNF stimulation, EPHB2 is ubiquitinated by its E3 ligase RNF186. Then, ubiquitinated EPHB2 recruits and further phosphorylates TAB2 at nine tyrosine sites, which is a critical step for the binding between TAB2 and TAK1. Due to defects in TNF signaling in RNF186-knockout colorectal epithelial cells, the phenotype of colitis-propelled colorectal cancer model in RNF186-knockout mice is significantly reduced compared with that in wild-type control mice. Moreover, we find that a genetic mutation in EPHB2 identified in a family with colorectal cancer is a gain-of-function mutation that promoted TNF signaling activation compared with wild-type EPHB2. We provide evidence that the EPHB2-RNF186-TAB2-TAK1 signaling cascade plays an essential role in TNF-mediated signal transduction in colorectal epithelial cells and the carcinogenesis of colorectal cancer, which may provide potential targets for the treatment of colorectal cancer.


Subject(s)
Colorectal Neoplasms , Receptor, EphA1 , Animals , Humans , Mice , Carcinogenesis , Colorectal Neoplasms/genetics , Cytokines , Epithelial Cells/metabolism , Receptor, EphA1/metabolism , Signal Transduction , Tyrosine , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Receptor, EphB2
7.
Genomics ; 114(3): 110341, 2022 05.
Article in English | MEDLINE | ID: mdl-35283197

ABSTRACT

OBJECTIVE: This study intends to conquer the mystery of microRNA-16-5p/erythropoietin-producing hepatocellular A1/nuclear factor-κB signaling (miR-16-5p/EPHA1/NF-κB signaling) in breast cancer. METHODS: Expression of miR-16-5p, EPHA1 and NF-κB signaling-related proteins were detected. Gene overexpression or silencing was used to examine the biological roles of bone marrow mesenchymal stem cells (BMSCs)-derived exo-miR-16-5p in breast cancer. The effect of exo-miR-16-5p on tumorigenesis of breast cancer was confirmed by the xenograft nude mouse model. RESULTS: Low miR-16-5p and high EPHA1 expression were examined in breast cancer. BMSCs-derived exosomes, up-regulated miR-16-5p or down-regulated EPHA1 restrained epithelial-mesenchymal transition (EMT) of breast cancer cells and tumor growth in nude mice. Down-regulated miR-16-5p or up-regulated EPHA1 activated NF-κB signaling. Knockdown of EPHA1 or inhibition of NF-κB signaling reversed the effects of down-regulated miR-16-5p on breast cancer cells. CONCLUSION: BMSCs-derived exosomal miR-16-5p hinders breast cancer cells progression via EPHA1/NF-κB signaling axis.


Subject(s)
Mesenchymal Stem Cells , MicroRNAs , Neoplasms , Animals , Humans , Mice , Disease Models, Animal , Epithelial-Mesenchymal Transition , Mesenchymal Stem Cells/metabolism , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasms/metabolism , NF-kappa B/metabolism , Receptor, EphA1/metabolism
8.
Contact Dermatitis ; 86(2): 71-79, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34812515

ABSTRACT

BACKGROUND: No biomarkers have been identified that can classify subtypes of hand eczema (HE). Although skin biopsies represent the gold standard for investigations of the skin, the invasive technique is not favorable when investigating skin from sensitive areas. Recent advances in the use of skin-tape strips for molecular investigations enable noninvasive investigations of HE. OBJECTIVE: By using whole transcriptome sequencing (WTS), the molecular profile of HE according to different localizations on the hands, etiologies, and clinical/morphological subtypes was investigated. METHODS: Thirty adult, Danish HE patients, 12 with and 18 without concurrent atopic dermatitis (AD), as well as 16 controls were included. Tape strip samples were collected from lesional, nonlesional, and healthy skin. Total RNA was extracted and WTS was performed. RESULTS: The largest molecular difference of HE patients with and without AD was found in nonlesional skin areas and included a downregulation of CXCL8 for HE patients without AD. Differences between allergic and irritant contact dermatitis included epidermal biomarkers such as EPHA1. CONCLUSION: Skin tape strip samples could be used to assess the gene expression profile of HE on different localizations of the hands. The skin tape strip method identified new molecular markers that showed promising result for the identification of HE subtypes.


Subject(s)
Hand Dermatoses/diagnosis , Hand Dermatoses/genetics , Specimen Handling/methods , Surgical Tape , Transcriptome , Adult , Aged , Biomarkers/metabolism , Dermatitis, Allergic Contact/diagnosis , Dermatitis, Allergic Contact/genetics , Dermatitis, Atopic/diagnosis , Dermatitis, Atopic/genetics , Dermatitis, Irritant/diagnosis , Dermatitis, Irritant/genetics , Diagnosis, Differential , Down-Regulation , Female , Hand Dermatoses/immunology , Humans , Interleukin-8/metabolism , Male , Middle Aged , Receptor, EphA1/metabolism , Skin/immunology , Skin/metabolism , Exome Sequencing
9.
Int J Mol Sci ; 22(16)2021 Aug 05.
Article in English | MEDLINE | ID: mdl-34445116

ABSTRACT

Erythropoietin-producing human hepatocellular receptors (EPHs) compose the largest known subfamily of receptor tyrosine kinases (RTKs). They bind and interact with the EPH family receptor interacting proteins (ephrins). EPHs/ephrins are implicated in a variety of physiological processes, as well as in cancer pathogenesis. With neoplastic disease remaining a leading cause of death world-wide, the development of novel biomarkers aiding in the field of diagnosis, prognosis, and disease monitoring is of utmost importance. A multitude of studies have proven the association between the expression of members of the EPH/ephrin system and various clinicopathological parameters, including disease stage, tumor histologic grade, and patients' overall survival. Besides their utilization in timely disease detection and assessment of outcome, EPHs/ephrins could also represent possible novel therapeutic targets. The aim of the current review of the literature was to present the existing data regarding the association between EPH/ephrin system expression and the clinical characteristics of malignant tumors.


Subject(s)
Ephrins/metabolism , Neoplasms/metabolism , Receptor, EphA1/metabolism , Biomarkers, Tumor/metabolism , Humans , Signal Transduction/physiology
10.
PLoS One ; 16(3): e0247335, 2021.
Article in English | MEDLINE | ID: mdl-33770085

ABSTRACT

Eph receptors are the largest group amongst the receptor tyrosine kinases and are divided into two subgroups, A and B, based on ligand binding specificities and sequence conservation. Through ligand-induced and ligand-independent activities, Ephs play central roles in diverse biological processes, including embryo development, regulation of neuronal signaling, immune responses, vasculogenesis, as well as tumor initiation, progression, and metastasis. The Eph extracellular regions (ECDs) are constituted of multiple domains, and previous structural studies of the A class receptors revealed how they interact with ephrin ligands and simultaneously mediate Eph-Eph clustering necessary for biological activity. Specifically, EphA structures highlighted a model, where clustering of ligand-bound receptors relies on two distinct receptor/receptor interfaces. Interestingly, most unliganded A class receptors also form an additional, third interface, between the ligand binding domain (LBD) and the fibronectin III domain (FN3) of neighboring molecules. Structures of B-class Eph ECDs, on the other hand, have never been reported. To further our understanding of Eph receptor function, we crystallized the EphB6-ECD and determined its three-dimensional structure using X-ray crystallography. EphB6 has important functions in both normal physiology and human malignancies and is especially interesting because this atypical receptor innately lacks kinase activity and our understanding of the mechanism of action is still incomplete. Our structural data reveals the overall EphB6-ECD architecture and shows EphB6-LBD/FN3 interactions similar to those observed for the unliganded A class receptors, suggesting that these unusual interactions are of general importance to the Eph group. We also observe unique structural features, which likely reflect the atypical signaling properties of EphB6, namely the need of co-receptor(s) for this kinase-inactive Eph. These findings provide new valuable information on the structural organization and mechanism of action of the B-class Ephs, and specifically EphB6, which in the future will assist in identifying clinically relevant targets for cancer therapy.


Subject(s)
Receptor, EphB6/ultrastructure , Receptors, Eph Family/ultrastructure , Cell Line , Crystallography, X-Ray/methods , Ephrins/metabolism , Fibronectins/metabolism , Humans , Ligands , Phosphorylation , Protein Binding/physiology , Protein Domains/physiology , Receptor, EphA1/metabolism , Receptor, EphA1/ultrastructure , Receptor, EphB6/metabolism , Receptors, Eph Family/metabolism , Signal Transduction
11.
Eur J Med Chem ; 216: 113318, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33730624

ABSTRACT

Identifying a pharmacological agent that targets only one of more than 500 kinases present in humans is an important challenge. One potential solution to this problem is the development of bivalent kinase inhibitors, which consist of two connected fragments, each bind to a dissimilar binding site of the bisubstrate enzyme. The main advantage of bivalent (type V) kinase inhibitors is generating more interactions with target enzymes that can enhance the molecules' selectivity and affinity compared to single-site inhibitors. Earlier type V inhibitors were not suitable for the cellular environment and were mostly used in in vitro studies. However, recently developed bivalent compounds have high kinase affinity, high biological and chemical stability in vivo. This review summarized the hetero-bivalent kinase inhibitors described in the literature from 2014 to the present. We attempted to classify the molecules by serine/threonine and tyrosine kinase inhibitors, and then each target kinase and its hetero-bivalent inhibitor was assessed in depth. In addition, we discussed the analysis of advantages, limitations, and perspectives of bivalent kinase inhibitors compared with the monovalent kinase inhibitors.


Subject(s)
Protein Kinase Inhibitors/chemistry , Protein Serine-Threonine Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/metabolism , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Receptor, EphA1/antagonists & inhibitors , Receptor, EphA1/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/metabolism
12.
Cancer Gene Ther ; 28(3-4): 307-320, 2021 04.
Article in English | MEDLINE | ID: mdl-33199830

ABSTRACT

Gastric cancer (GC) is a common cancer and a leading cause of cancer-related deaths worldwide. Recent studies have supported the important role of long non-coding RNAs (lncRNAs) in GC progression. This study identified functional significance of X inactive specific transcript (XIST) in GC. The expression of XIST and EPHA1 in GC tissues and cells was measured. Then, dual luciferase reporter gene assay, RNA immunoprecipitation (RIP) assay and Chromatin Immunoprecipitation (ChIP) assay were performed to explore the interaction among XIST, EPHA1 and HNF4A. The effects of XIST on GG progression were evaluated by determining expression of proliferation- and invasion-related proteins (Ki67, PCNA, MMP-2, and MMP-9). Further, the functional role of XIST in GC with the involvement of NFκB pathway was also analyzed. Subsequently, the tumor growth in nude mice was evaluated. High expression of XIST and EPHA1 was observed in GC. XIST elevated EPHA1 expression by recruiting HNF4A. In addition, silencing of XIST inhibited GC progression in vitro and in vivo. Overexpressed XIST and EPHA1 yielded a reversed effect on cell proliferation and invasion. SN50 treatment (inhibitor of NFκB pathway) counteracted the promotive effect on GC cell proliferation and invasion mediated by XIST. The present study unveils that XIST increases the enrichment of HNF4A in the promoter region of EPHA1, thus promoting the deterioration of GC.


Subject(s)
Hepatocyte Nuclear Factor 4/antagonists & inhibitors , Hepatocyte Nuclear Factor 4/metabolism , NF-kappa B/metabolism , RNA, Long Noncoding/genetics , Receptor, EphA1/metabolism , Stomach Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Proliferation/physiology , Disease Progression , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/biosynthesis , RNA, Long Noncoding/metabolism , Receptor, EphA1/genetics , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Transcription, Genetic
13.
Mol Neurobiol ; 58(3): 913-925, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33057926

ABSTRACT

There is increasing evidence that EphA1 is involved in the function and development of the central nervous system, especially in neuroinflammation. It has been found to affect the disease progression of Alzheimer's disease (AD) by regulating the neuroinflammatory process. Neuroinflammation has always been regarded as the mechanism of the development of Parkinson's disease (PD) and possible therapeutic targets. Therefore, it is worth studying whether EphA1 has a potential therapeutic value for PD. The purpose of this study is to investigate the effect of EphA1 in mice and PD cell models and its mechanism.In this study, we verified the difference in expression of EphA1 and the effect and mechanism of EphA1 on neuropathological changes through Parkinson's patient samples, Parkinson's mice model, and Parkinson's model prepared from SH-SY5Y cells in vitro.EphA1 was highly expressed in the substantia nigra (SN) region of Parkinson mice and the Parkinson cell model, while the expression of tyrosine hydroxylase (TH) in the SN region of Parkinson mice was significantly reduced. After silenced EphA1 in the SH-SY5Y cell PD model, the expression levels of α-synuclein, inflammatory factors, and microglia-activated chemokine decreased. The co-immunoprecipitation experiment proved that EphA1 overexpression could promote the binding of CXCL12 and CXCR4. However, after silenced EphA1 and CXCL12 at the same time, the above effects brought by silenced EphA1 were suppressed. The same result appeared in mice with PD.EphA1 improves the inflammatory responses and neuropathological changes of the PD model in vivo and in vitro through the CXCL12/CXCR4 signaling pathway. Graphical abstract.


Subject(s)
Brain/pathology , Chemokine CXCL12/metabolism , Parkinson Disease/pathology , Receptor, EphA1/metabolism , Receptors, CXCR4/metabolism , Signal Transduction , Animals , Brain/metabolism , Cell Line, Tumor , Disease Models, Animal , Humans , Inflammation/complications , Inflammation/pathology , Male , Mice, Inbred C57BL , Neurotoxins/toxicity , Parkinson Disease/blood , Receptor, EphA1/blood , alpha-Synuclein/metabolism
14.
Development ; 147(15)2020 08 14.
Article in English | MEDLINE | ID: mdl-32665244

ABSTRACT

Gastrulation is the first major morphogenetic event during animal embryogenesis. Ascidian gastrulation starts with the invagination of 10 endodermal precursor cells between the 64- and late 112-cell stages. This process occurs in the absence of endodermal cell division and in two steps, driven by myosin-dependent contractions of the acto-myosin network. First, endoderm precursors constrict their apex. Second, they shorten apico-basally, while retaining small apical surfaces, thereby causing invagination. The mechanisms that prevent endoderm cell division, trigger the transition between step 1 and step 2, and drive apico-basal shortening have remained elusive. Here, we demonstrate a conserved role for Nodal and Eph signalling during invagination in two distantly related ascidian species, Phallusia mammillata and Ciona intestinalis Specifically, we show that the transition to step 2 is triggered by Nodal relayed by Eph signalling. In addition, our results indicate that Eph signalling lengthens the endodermal cell cycle, independently of Nodal. Finally, we find that both Nodal and Eph signals are dispensable for endoderm fate specification. These results illustrate commonalities as well as differences in the action of Nodal during ascidian and vertebrate gastrulation.


Subject(s)
Ciona intestinalis/embryology , Endoderm/embryology , Gastrulation/physiology , Nodal Protein/metabolism , Receptor, EphA1/metabolism , Animals , Endoderm/cytology
15.
Exp Cell Res ; 392(1): 112009, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32305326

ABSTRACT

One of the reasons for recurrence following treatment of high grade serous ovarian carcinoma (HGSOC) is the persistence of residual cancer stem cells (CSCs). There has been variability between laboratories in the identification of CSC markers for HGSOC. We have identified new surface markers (CD24, CD9 and EPHA1) in addition to those previously known (CD44, CD117 and CD133) using a bioinformatics approach. The expression of these surface markers was evaluated in ovarian cancer cell lines, primary malignant cells (PMCs), normal ovary and HGSOC. There was no preferential expression of any of the markers or a combination. All the markers were expressed at variable levels in ovarian cancer cell lines and PMCs. Only CD117 and CD9 were expressed in the normal ovarian surface epithelium and fallopian tube. Both ALDEFLUOR (ALDH1A1) and side population assays identified a small proportion of cells (<3%) separately that did not overlap with little variability in cell lines and PMCs. All surface markers were co-expressed in ALDH1A1+ cells without preference for one combination. The cell cycle analysis of ALDH1A1+ cells alone revealed that majority of them reside in G0/G1 phase of cell cycle. Further separation of G0 and G1 phases showed that ALDH1A1+ cells reside in G1 phase of the cell cycle. Xenograft assays showed that the combinations of ALDH1A1 + cells co-expressing CD9, CD24 or EPHA1 were more tumorigenic and aggressive with respect to ALDH1A1-cells. These data suggest that a combined approach could be more useful in identifying CSCs in HGSOC.


Subject(s)
Aldehyde Dehydrogenase 1 Family/metabolism , Biomarkers, Tumor/metabolism , Cystadenocarcinoma, Serous/pathology , Neoplastic Stem Cells/physiology , Ovarian Neoplasms/pathology , Retinal Dehydrogenase/metabolism , Aldehyde Dehydrogenase 1 Family/genetics , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Biomarkers, Tumor/genetics , CD24 Antigen/genetics , CD24 Antigen/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Female , Heterografts , Humans , Mice , Mice, Nude , Mice, Transgenic , Neoplasm Invasiveness , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Receptor, EphA1/genetics , Receptor, EphA1/metabolism , Retinal Dehydrogenase/genetics , Tetraspanin 29/genetics , Tetraspanin 29/metabolism
16.
Cancer Biomark ; 28(3): 397-408, 2020.
Article in English | MEDLINE | ID: mdl-32224528

ABSTRACT

BACKGROUND: There has been variability between laboratories in the identification of cancer stem cells (CSCs) markers for epithelial ovarian cancer (EOC). We have evaluated three new surface markers for EOC to identify CSCs precisely. METHODS: Three new putative CSCs specific surface markers CD9, CD24 and EPHA1 identified by a bioinformatics approach were evaluated in normal ovary, fallopian tube and ovarian tumours. RESULTS: The expression of CD9 alone was observed in normal ovarian surface epithelium and fallopian tube whereas CD24 and EPHA1 were not expressed (n= 5). CD24 was expressed in all tumours (N= 101) while CD9 and EPHA1 were expressed in 89 and 71 tumours, respectively. The statistical analysis showed significant correlation of the stage of the disease (p< 0.0001), type of surgery (p< 0.0001) and residual disease (p< 0.0001) with overall survival. Although expression of CD9, CD24 and EPHA1 was observed in the majority of tumours there was no significant correlation with outcome. In patients who underwent primary surgery, increased expression of CD24 significantly correlated with poor survival. The expression of CD24 was significantly reduced (p< 0.002) upon analysis of paired sections from patients prior to surgery and at interval debulking surgery (n= 16). CONCLUSION: These findings suggest that overexpression of these new markers may be useful in identifying and targeting ovarian CSCs and CD24 may be a putative CSCs marker in ovarian cancer.


Subject(s)
Biomarkers, Tumor/metabolism , CD24 Antigen/metabolism , Carcinoma, Ovarian Epithelial/pathology , Ovarian Neoplasms/pathology , Receptor, EphA1/metabolism , Tetraspanin 29/metabolism , Adult , Carcinoma, Ovarian Epithelial/mortality , Carcinoma, Ovarian Epithelial/therapy , Chemotherapy, Adjuvant/methods , Computational Biology , Disease-Free Survival , Feasibility Studies , Female , Follow-Up Studies , Humans , Middle Aged , Neoadjuvant Therapy/methods , Neoplasm Staging , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/mortality , Ovarian Neoplasms/therapy , Ovariectomy , Ovary/cytology , Ovary/pathology , Ovary/surgery
17.
Neoplasma ; 67(4): 794-801, 2020 07.
Article in English | MEDLINE | ID: mdl-32266818

ABSTRACT

Although the incidence of nasopharyngeal carcinoma (NPC) is relatively low, the mortality is very high and the patients have a poor prognosis. Thus, it is urgent to find a novel biomarker and a new therapeutic strategy. Suppressor of cytokine signaling-2 (SOCS2) was reported to be associated with various malignancies. However, the exact role of SOCS2 in NPC still remains largely unsure. In the present study, we showed that the expression of SOCS2 was significantly upregulated in NPC patients and cells. And the high expression of SOCS2 predicted a worse outcome in NPC patients. Moreover, the in vivo experiments indicated that knockout of SOCS2 inhibits the proliferation, migration, and invasion of NPC cells. Besides, we found a positive relationship between SOCS2 and EphA1 in NPC tissues. The rescue experiments indicated that SOCS2 affected the malignancy of NPC cells by regulating the expression of EphA1. Altogether, our data uncovered the ontogenetic role of SOCS2 dysregulation during the tumorigenesis of NPC. SOCS2 might serve as a biomarker during the diagnosis and treatment of NPC. And targeting SOCS2 might provide a novel treatment strategy for NPC patients.


Subject(s)
Cell Movement , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms , Suppressor of Cytokine Signaling Proteins , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Neoplasms/genetics , Neoplasm Invasiveness , Receptor, EphA1/metabolism , Suppressor of Cytokine Signaling Proteins/physiology
18.
Cell Oncol (Dordr) ; 43(4): 655-667, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32291572

ABSTRACT

PURPOSE: Upregulation of receptor tyrosine kinase EphA2 has been found to be associated with a poor prognosis in many types of cancer and is considered an attractive therapeutic target. As yet, few efforts have been focused on its tumor suppressive activity triggered by its ligand, ephrinA1. Here, we aimed to determine the potential of ephrinA1 as an important player in melanoma metastasis. METHODS: Data from the Cancer Genome Atlas (TCGA) and the Cancer Cell Line Encyclopedia (CCLE) were analyzed to explore the expression and prognostic implications of EphA2 and ephrinA1 in melanoma. Western blotting, shRNA, colony formation and immunofluorescence assays, as well as two in vivo xenograft models (subcutaneous and metastatic) were used to evaluate the role of EphA2 in melanoma progression. Akt inhibition and ephrinA1-Fc were used to confirm the influence of Akt activation and ephrinA1 levels on the EphA2 effects. Immunohistochemistry (IHC) was performed on xenograft and patient melanoma tissues. RESULTS: We found that high levels of ephrinA1, but not EphA2, were negatively correlated with melanoma metastasis. The expression levels of EphA2 and ephrinA1 were not correlated. After EphA2 downregulation, colony forming abilities and lung metastatic growth were reduced in melanoma cell lines with a low ephrinA1 expression, but were increased in melanoma cell lines with a high ephrinA1 expression. EphA2-mediated colony formation in EphA2-high/ephrinA1-low cells was found to be Akt-dependent and to be inhibited by the addition of ephrinA1-Fc. IHC staining of primary melanoma specimens revealed that EphA2-high/ephrinA1-low patients exhibited poorer outcomes than EphA2-high/ephrinA1-high patients. CONCLUSIONS: From our data we conclude that evaluation of ephrinA1 levels may be helpful for the application of EphA2-targeted therapies and for prognostic predictions in melanoma patients.


Subject(s)
Melanoma/metabolism , Melanoma/pathology , Receptor, EphA1/metabolism , Receptor, EphA2/metabolism , Animals , Heterografts , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude
19.
Sci Rep ; 10(1): 4211, 2020 03 06.
Article in English | MEDLINE | ID: mdl-32144278

ABSTRACT

Neurofibromatosis type 2 (NF2) is an inherited disorder characterized by bilateral vestibular schwannomas (VS) that arise from neoplastic Schwann cells (SCs). NF2-associated VSs are often accompanied by meningioma (MN), and the majority of NF2 patients show loss of the NF2 tumor suppressor. mTORC1 and mTORC2-specific serum/glucocorticoid-regulated kinase 1 (SGK1) are constitutively activated in MN with loss of NF2. In a recent high-throughput kinome screen in NF2-null human arachnoidal and meningioma cells, we showed activation of EPH RTKs, c-KIT, and SFK members independent of mTORC1/2 activation. Subsequently, we demonstrated in vitro and in vivo efficacy of combination therapy with the dual mTORC1/2 inhibitor AZD2014 and the multi-kinase inhibitor dasatinib. For these reasons, we investigated activated mTORC1/2 and EPH receptor-mediated signaling in sporadic and NF2-associated VS. Using primary human VS cells and a mouse allograft model of schwannoma, we evaluated the dual mTORC1/2 inhibitor AZD2014 and the tyrosine kinase inhibitor dasatinib as monotherapies and in combination. Escalating dose-response experiments on primary VS cells grown from 15 human tumors show that combination therapy with AZD2014 and dasatinib is more effective at reducing metabolic activity than either drug alone and exhibits a therapeutic effect at a physiologically reasonable concentration (~0.1 µM). In vivo, while AZD2014 and dasatinib each inhibit tumor growth alone, the effect of combination therapy exceeds that of either drug. Co-targeting the mTOR and EPH receptor pathways with these or similar compounds may constitute a novel therapeutic strategy for VS, a condition for which there is no FDA-approved pharmacotherapy.


Subject(s)
Benzamides/pharmacology , Dasatinib/pharmacology , Disease Models, Animal , Morpholines/pharmacology , Neurofibromin 2/physiology , Neuroma, Acoustic/drug therapy , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , TOR Serine-Threonine Kinases/antagonists & inhibitors , Animals , Drug Therapy, Combination , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurofibromin 2/genetics , Neurofibromin 2/metabolism , Neuroma, Acoustic/metabolism , Neuroma, Acoustic/pathology , Receptor, EphA1/metabolism
20.
Med Sci Monit ; 26: e923409, 2020 Mar 27.
Article in English | MEDLINE | ID: mdl-32218416

ABSTRACT

BACKGROUND With the growing global burden of gastric carcinoma (GC) and the urgent need for biomolecular targeted therapies, this study aimed to elucidate the relationship between EphA1 and the tumor microenvironment (focusing primarily on the key inflammatory cytokines IL-6 and tumor angiogenic cytokine VEGF) to identify a new potential therapeutic target. MATERIAL AND METHODS IHC and qRT-PCR were performed to quantify the protein and gene expression levels of EphA1, IL-6, and VEGF in normal mucosal tissues, carcinoma tissues, and paracarcinomatous tissues from 57 GC patients. Spearman's rank correlation test was performed to determine the relationship between EphA1, IL-6, and VEGF expression levels. The relationships of EphA1 with clinicopathologic parameter and survival in GC patients were also evaluated. RESULTS The protein and gene expression levels of EphA1 were all attenuated gradually from carcinoma tissues to paracarcinomatous tissues and then to normal mucosal tissues in GC patients. Additionally, significant correlations between the overexpression of EphA1 with aggressive clinicopathological features and shorter survival time of GC patients were verified. In particular, we found a significant positive correlation between the expression of EphA1 and tumor microenvironment hallmark proteins IL-6 and VEGF in carcinoma tissues and paracarcinomatous tissues. CONCLUSIONS EphA1 can promote the occurrence and development of GC by its selective high expression in cancer tissues and its relationship with malignant clinical features and prognosis of GC patients. The underlying potential mechanism appears to involve enhancement of the tumor microenvironment, which via drives the expression of tumor microenvironment hallmark proteins IL-6 and VEGF.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma/pathology , Receptor, EphA1/metabolism , Stomach Neoplasms/pathology , Tumor Microenvironment , Adult , Aged , Aged, 80 and over , Biopsy , Carcinoma/genetics , Carcinoma/mortality , Female , Gastric Mucosa/pathology , Gene Expression Profiling , Humans , Immunohistochemistry , Interleukin-6/metabolism , Kaplan-Meier Estimate , Male , Middle Aged , Prognosis , Stomach Neoplasms/genetics , Stomach Neoplasms/mortality , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...