Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Genes Dev ; 31(10): 1054-1065, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28637694

ABSTRACT

Proper function of the neural network results from the precise connections between axons and dendrites of presynaptic and postsynaptic neurons, respectively. In the Drosophila olfactory system, the dendrites of projection neurons (PNs) stereotypically target one of ∼50 glomeruli in the antennal lobe (AL), the primary olfactory center in the brain, and form synapses with the axons of olfactory receptor neurons (ORNs). Here, we show that Eph and Ephrin, the well-known axon guidance molecules, instruct the dendrodendritic segregation during the discrete olfactory map formation. The Eph receptor tyrosine kinase is highly expressed and localized in the glomeruli related to reproductive behavior in the developing AL. In one of the pheromone-sensing glomeruli (DA1), the Eph cell-autonomously regulates its dendrites to reside in a single glomerulus by interacting with Ephrins expressed in adjacent PN dendrites. Our data demonstrate that the trans interaction between dendritic Eph and Ephrin is essential for the PN dendritic boundary formation in the DA1 olfactory circuit, potentially enabling strict segregation of odor detection between pheromones and the other odors.


Subject(s)
Drosophila melanogaster/physiology , Receptor, EphA1/metabolism , Animals , Dendrites/enzymology , Dendrites/physiology , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/embryology , Drosophila melanogaster/enzymology , Drosophila melanogaster/genetics , Gene Expression Profiling , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Membrane Proteins/genetics , Membrane Proteins/metabolism , Olfactory Receptor Neurons/cytology , Olfactory Receptor Neurons/enzymology , Olfactory Receptor Neurons/physiology , RNA Interference , Receptor, EphA1/genetics
2.
Hum Genomics ; 15(1): 9, 2021 01 30.
Article in English | MEDLINE | ID: mdl-33516273

ABSTRACT

Genome-wide association studies (GWAS) have identified several susceptibility loci of Alzheimer's disease (AD), which were mainly located in noncoding regions of the genome. Meanwhile, the putative biological mechanisms underlying AD susceptibility loci were still unclear. At present, identifying the functional variants of AD pathogenesis remains a major challenge. Herein, we first used summary data-based Mendelian randomization (SMR) with AD GWAS summary and expression quantitative trait loci (eQTL) data to identify variants who affects expression levels of nearby genes and contributed to the risk of AD. Using the SMR integrative analysis, we totally identified 14 SNPs significantly affected the expression level of 16 nearby genes in blood or brain tissues and contributed to the AD risk. Then, to confirm the results, we replicated the GWAS and eQTL results across multiple samples. Totally, four risk SNP (rs11682128, rs601945, rs3935067, and rs679515) were validated to be associated with AD and affected the expression level of nearby genes (BIN1, HLA-DRA, EPHA1-AS1, and CR1). Besides, our differential expression analysis showed that the BIN1 gene was significantly downregulated in the hippocampus (P = 2.0 × 10-3) and survived after multiple comparisons. These convergent lines of evidence suggest that the BIN1 gene identified by SMR has potential roles in the pathogenesis of AD. Further investigation of the roles of the BIN1 gene in the pathogenesis of AD is warranted.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Alzheimer Disease/genetics , Genetic Predisposition to Disease , Nuclear Proteins/genetics , Quantitative Trait Loci/genetics , Tumor Suppressor Proteins/genetics , Alzheimer Disease/pathology , Female , Gene Expression Regulation/genetics , Genome-Wide Association Study , HLA-DR alpha-Chains/genetics , Humans , Male , Mendelian Randomization Analysis , Polymorphism, Single Nucleotide/genetics , RNA, Long Noncoding/genetics , Receptor, EphA1/genetics , Receptors, Complement 3b/genetics , Risk Factors
3.
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
4.
J Biol Chem ; 294(1): 341-350, 2019 01 04.
Article in English | MEDLINE | ID: mdl-30401746

ABSTRACT

Gap junctions are intercellular conduits that permit the passage of ions, small metabolites, and signaling molecules between cells. Connexin32 (Cx32) is a major gap junction protein in the liver and brain. Phosphorylation is integral to regulating connexin assembly, degradation, and electrical and metabolic coupling, as well as to interactions with molecular partners. Cx32 contains two intracellular tyrosine residues, and tyrosine phosphorylation of Cx32 has been detected after activation of the epidermal growth factor receptor; however, the specific tyrosine residue and the functional implication of this phosphorylation remain unknown. To address the limited available information on Cx32 regulation by tyrosine kinases, here we used the Cx32 C-terminal (CT) domain in an in vitro kinase-screening assay, which identified ephrin (Eph) receptor family members as tyrosine kinases that phosphorylate Cx32. We found that EphB1 and EphA1 phosphorylate the Cx32CT domain residue Tyr243 Unlike for Cx43, the tyrosine phosphorylation of the Cx32CT increased gap junction intercellular communication. We also demonstrated that T-cell protein-tyrosine phosphatase dephosphorylates pTyr243 The data presented above along with additional examples throughout the literature of gap junction regulation by kinases, indicate that one cannot extrapolate the effect of a kinase on one connexin to another.


Subject(s)
Connexins/metabolism , Gap Junctions/metabolism , Receptor, EphA1/metabolism , Receptor, EphB1/metabolism , Caco-2 Cells , Connexin 43/genetics , Connexin 43/metabolism , Connexins/genetics , Gap Junctions/genetics , HeLa Cells , Humans , Protein Tyrosine Phosphatase, Non-Receptor Type 2 , Receptor, EphA1/genetics , Receptor, EphB1/genetics , Gap Junction beta-1 Protein
5.
Brain Inj ; 33(10): 1385-1401, 2019.
Article in English | MEDLINE | ID: mdl-31319723

ABSTRACT

Primary Objective: Eph/Ephrin signaling is inhibitory for developing axons and blocking Eph pathways enhances regeneration after spinal cord injury. It was hypothesized that inhibition of Eph signaling promotes cellular and behavioral recovery after traumatic brain injury (TBI). Research design: Lateral fluid percussion (LFP) injury was performed on wildtype (WT) and EphA6 knockout (KO) mice. EphA6-Fc, Ephrin-A5-Fc fusion proteins, and sodium orthovanadate were used to alter the signaling pathway. Immunohistochemistry and tissue explants revealed cellular changes. Rotarod tests demonstrated vestibulomotor function. Outcomes: The EphA6 receptor expression is upregulated following LFP. Uninjured EphA6 KO mice exhibit greater neurite density and clustered Ephrin-A5-Fc causes growth cone collapse in vitro. After LFP, EphA6 KO mice demonstrate longer neurites and decreased neuronal cell death and astrocytosis compared to WT mice. Blocking EphA signaling by soluble EphA6-Fc fusion protein reduces cell death and improves motor function following LFP whereas clustered Ephrin-A5-Fc exacerbates cell death and neurodegeneration. Sodium orthovanadate rescues growth cone collapse in vitro as well as cell death and neurodegeneration in vivo. Conclusions: Eph/Ephrin signaling plays an inhibitory role following TBI. Targeting the Eph signaling pathway with Fc fusion proteins and pharmacological agents can be a novel strategy to counter the damaging effects of TBI. Abbreviations: LFP: lateral fluid percussion; TBI: traumatic brain injury; KO: knockout; WT: wildtype; PTP2: protein phosphotyrosine phosphatase 2; Tg: transgenic; YFP: yellow fluorescent protein; ATM: atmospheres; RT-qPCR: Real-time-quantitative PCR; dpi: days post injury; GAPDH: glyceraldehyde-3-phosphate dehydrogenase; DAPI: 4',6-diamidino-2-phenylindole; PBS: phosphate buffered saline; GFAP: glial fibrillary acidic protein; FLJC: fluorojade C; CA: cornu ammonis; SEM: standard error of the mean; ANOVA: analysis of variance; PLSD: posthoc least significant difference.


Subject(s)
Brain Injuries, Traumatic/drug therapy , Brain Injuries, Traumatic/therapy , Genetic Therapy/methods , Receptor, EphA1/antagonists & inhibitors , Receptor, EphA1/genetics , Animals , Astrocytes/pathology , Brain Injuries, Traumatic/pathology , Cell Death , Immunoglobulin G/pharmacology , Male , Mice , Mice, Knockout , Nerve Degeneration/genetics , Nerve Degeneration/prevention & control , Neurites/pathology , Neurons/metabolism , Postural Balance , Receptor, EphA1/biosynthesis , Signal Transduction/drug effects , Signal Transduction/genetics , Vanadates/therapeutic use
6.
Reprod Domest Anim ; 53(4): 937-946, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29740882

ABSTRACT

The objective of this study was to ascertain whether mRNA and protein expressions of implantation-related genes (erythropoietin-producing hepatocellular receptor-ligand A1, Eph-ephrin A1 and leptin receptor-leptin, LEPR-LEP) differed between pigs with high and low number of embryos, and whether these differences in gene expression might affect embryo implantation. Experimental pig groups (n = 24) for high and low number of embryos were prepared by altering the number of eggs ovulated in pre-pubertal gilts treated with 1.5 × (High) or 1.0 × (Low) PG600 ([400 IU PMSG + 200 IU hCG]/dose, AKZO-NOBEL). Gilts expressing oestrus were artificially inseminated twice and maintained in breeding and gestation until the reproductive tract was collected on day 22 of pregnancy. At slaughter, the reproductive tracts from each pregnant gilt from each treatment were immediately processed to collect samples for RNA and protein analysis. Within each gilt, three conceptus points were sampled, one from each horn and then a random conceptus within the tract. At each conceptus point, endometrial attachment site, chorion-allantois and embryo were collected and immediately frozen in liquid nitrogen. Number of corpus luteum (CL) (35.4 vs. 12.6) and total embryo number (18.8 vs. 10.2) were greater in the high-embryo compared to the low-embryo group, respectively (p < .05). Real-time qPCR results showed that Eph-ephrin A1 mRNA expression was less in the high-embryo (p < .05) compared to the low-embryo group. In addition, Western blotting analysis indicated that Eph-ephrin A1 and LEP protein expression at endometrial attachment site in high-embryo was less (p < .05) compared to low-embryo group. It was also noted that mRNA expression of Eph-ephrin A1 and LEPR-LEP was greater in pregnant than non-pregnant gilts (p < .05). Moreover, mRNA expression of Eph-ephrin A1 (p < .05) and LEPR-LEP was greatest at endometrial attachment site among all three tissues. There was a positive correlation between expressions of Eph-ephrin A1, LEPR-LEP and embryo length with the correlation coefficient 0.31-0.59. For Eph-ephrin A1, the highest correlation coefficient appeared between Eph A1 expression and normal embryo number, between ephrin A1 expression and embryo length. For LEPR-LEP, the highest correlation coefficient appeared between LEPR-LEP expression and ovary weight (0.79 for both, p < .05), followed by embryo length and weight. The results of this study suggest that low expression of Eph-ephrin A1 and LEPR-LEP is somehow related to increased embryo number during implantation and that endometrial attachment site might be the main target tissue of these gene products. Yet, the increased expression of Eph-ephrin A1 and LEPR-LEP appeared associated with increased embryo growth (length and weight) and ovary weight, Eph-ephrin A1 and LEPR-LEP might play roles in the regulation of embryo implantation in pigs.


Subject(s)
Ephrin-A1/metabolism , Leptin/metabolism , Receptor, EphA1/metabolism , Receptors, Leptin/metabolism , Swine/embryology , Animals , Embryo Implantation/physiology , Ephrin-A1/genetics , Female , Gene Expression Regulation/physiology , Leptin/genetics , Pregnancy , RNA, Messenger , Receptor, EphA1/genetics , Receptors, Leptin/genetics
7.
Am J Med Genet B Neuropsychiatr Genet ; 177(8): 727-735, 2018 12.
Article in English | MEDLINE | ID: mdl-30378268

ABSTRACT

Mild behavioral impairment (MBI) describes the emergence of later-life neuropsychiatric symptoms (NPS) as an at-risk state for incident cognitive decline and dementia, and for some as a potential manifestation of prodromal dementia. How NPS mechanistically link to the development of mild cognitive impairment and Alzheimer's disease (AD) is not fully understood, with potential mechanisms including shared risk factors related to both NPS and cognitive impairment, or AD pathology promoting NPS. This is the first exploratory study to examine whether AD genetic loci as a genetic risk score (GRS), or individually, are a shared risk factor with MBI. Participants were 1,226 older adults (aged 72-79; 738 males; 763 normal cognition) from the Personality and Total Health Through Life project. MBI was approximated in accordance with Criterion 1 of the ISTAART-AA diagnostic criteria using a transformation algorithm for the neuropsychiatric inventory. A GRS was constructed from 25 AD risk loci. Binomial logistic regression adjusting for age, gender, and education examined the association between GRS and MBI. A higher GRS and APOE*ε4 were associated with increased likelihood of affective dysregulation. Nominally significant associations were observed between MS4A4A-rs4938933*C and MS4A6A-rs610932*G with a reduced likelihood of affective dysregulation; ZCWPW1-rs1476679*C with a reduced likelihood of social inappropriateness and abnormal perception/thought content; BIN1-rs744373*G and EPHA1-rs11767557*C with higher likelihood of abnormal perception/thought content; NME8-rs2718058*G with a reduced likelihood of decreased motivation. These preliminary findings suggest a common genetic etiology between MBI and traditionally recognized cognitive problems observed in AD and improve our understanding of the pathophysiological features underlying MBI.


Subject(s)
Alzheimer Disease/genetics , Cognitive Dysfunction/genetics , Adaptor Proteins, Signal Transducing/genetics , Aged , Cognition , Disease Progression , Female , Genetic Association Studies/methods , Genetic Loci , Genetic Predisposition to Disease , Humans , Male , Membrane Proteins/genetics , Neuropsychological Tests , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide/genetics , Preliminary Data , Receptor, EphA1/genetics , Risk Factors , Tumor Suppressor Proteins/genetics
8.
Biochem Biophys Res Commun ; 486(3): 693-699, 2017 05 06.
Article in English | MEDLINE | ID: mdl-28341121

ABSTRACT

The Eph family of receptor tyrosine kinases serves as key modulators of various cellular functions, including inflammation, hypertrophy and fibrosis. Recent analyses have revealed that a member of the Eph family, EphA1, plays a pivotal role in regulating insulin metabolism and kidney injury. However, the importance of EphA1 in diabetic nephropathy has not been recognized. We established a diabetic nephropathy mouse model using a high-fat diet and streptozotocin (STZ) injection. Then, the recombinant adeno-associated virus type 9 (AAV9) overexpressing EphA1 or a negative control was injected locally into the kidney. Metabolite testing and histopathological analyses of kidney fibrosis, pancreatic islet function and signaling pathways were evaluated. Our study showed that hyperglycemia, insulin resistance, and renal fibrosis accompanied the deterioration of kidney function in diabetic mice. The overexpression of EphA1 in the kidney attenuated renal fibrosis and improved kidney function but did not affect systemic glucose metabolism and pancreatic islet function. Furthermore, the overexpression of EphA1 decreased the phosphorylation of ERK1/2, JNK and MYPT1 (a substrate of Rho kinase). The overexpression of EphA1 can be therapeutically targeted to inhibit diabetic renal fibrosis, which suggests that the EphA1-Epha receptor axis may be a novel therapy target for diabetic nephropathy. Mechanistically, the overexpression of EphA1 could inhibit MAPK and the Rho pathway in diabetic kidneys.


Subject(s)
Diabetes Mellitus, Experimental/therapy , Diabetic Nephropathies/therapy , Ephrin-A1/genetics , Genetic Vectors/therapeutic use , Hyperglycemia/therapy , Receptor, EphA1/genetics , Animals , Dependovirus/genetics , Dependovirus/metabolism , Diabetes Mellitus, Experimental/chemically induced , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetic Nephropathies/chemically induced , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Diet, High-Fat , Ephrin-A1/agonists , Ephrin-A1/metabolism , Fibrosis , Gene Expression Regulation , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Hyperglycemia/chemically induced , Hyperglycemia/genetics , Hyperglycemia/pathology , Insulin Resistance , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/pathology , MAP Kinase Kinase 4/genetics , MAP Kinase Kinase 4/metabolism , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Myosin-Light-Chain Phosphatase/genetics , Myosin-Light-Chain Phosphatase/metabolism , Receptor, EphA1/agonists , Receptor, EphA1/metabolism , Signal Transduction , Streptozocin
9.
Dev Dyn ; 244(3): 417-30, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25529479

ABSTRACT

BACKGROUND: NR4A nuclear receptors are a conserved, functionally diverse group of nuclear receptors that regulate multiple cellular processes including proliferation and differentiation. The gene nhr-6 encodes the sole Caenorhabditis elegans NR4A nuclear receptor homolog with an essential role in reproduction by regulating morphogenesis of the spermatheca, a somatic gonad organ involved in ovulation and fertilization. RESULTS: Here, we identify the spermatheca cell lineage defects that occur in nhr-6 mutants. Utilizing cell marker analysis, we find that nhr-6 is required for cell cycle progression and that the cell proliferation phenotype is not due to premature cell cycle exit. We also show that loss of the negative cell cycle regulators fzr-1 and lin-35 suppresses the cell proliferation defects. We further demonstrate that NHR-6 activity intersects with Eph receptor signaling during spermatheca cell proliferation. CONCLUSIONS: NHR-6 has an essential function in promoting cell cycle progression during G1 phase in a specific spermatheca cell lineage. Genetic suppression of the proliferation phenotype does not affect the differentiation phenotypes observed in nhr-6 mutants, indicating a dualistic role for nhr-6 in regulating cell proliferation and cell differentiation during spermatheca organogenesis.


Subject(s)
Animal Structures/embryology , Caenorhabditis elegans Proteins/metabolism , Caenorhabditis elegans/embryology , G1 Phase/physiology , Organogenesis/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans Proteins/genetics , Receptor, EphA1/genetics , Receptor, EphA1/metabolism , Receptors, Cytoplasmic and Nuclear/genetics
10.
Am J Hum Genet ; 90(4): 720-6, 2012 Apr 06.
Article in English | MEDLINE | ID: mdl-22482808

ABSTRACT

Recent genome-wide association studies have identified a number of susceptibility loci for Alzheimer disease (AD). To understand the functional consequences and potential interactions of the associated loci, we explored large-scale data sets interrogating the human genome for evidence of positive natural selection. Our findings provide significant evidence for signatures of recent positive selection acting on several haplotypes carrying AD susceptibility alleles; interestingly, the genes found in these selected haplotypes can be assembled, independently, into a molecular complex via a protein-protein interaction (PPI) network approach. These results suggest a possible coevolution of genes encoding physically-interacting proteins that underlie AD susceptibility and are coexpressed in different tissues. In particular, PICALM, BIN1, CD2AP, and EPHA1 are interconnected through multiple interacting proteins and appear to have coordinated evidence of selection in the same human population, suggesting that they may be involved in the execution of a shared molecular function. This observation may be AD-specific, as the 12 loci associated with Parkinson disease do not demonstrate excess evidence of natural selection. The context for selection is probably unrelated to AD itself; it is likely that these genes interact in another context, such as in immune cells, where we observe cis-regulatory effects at several of the selected AD loci.


Subject(s)
Alzheimer Disease/genetics , Genetic Loci , Selection, Genetic , Adaptor Proteins, Signal Transducing/genetics , Age of Onset , Cytoskeletal Proteins/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Haplotypes , Humans , Monomeric Clathrin Assembly Proteins/genetics , Nuclear Proteins/genetics , Polymorphism, Single Nucleotide , Protein Interaction Maps/genetics , Receptor, EphA1/genetics , Tumor Suppressor Proteins/genetics
11.
Ann Surg Oncol ; 22(7): 2329-35, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25391265

ABSTRACT

BACKGROUND: Erythropoietin-producing hepatocellular (Eph) receptors are the largest subfamily of receptor tyrosine kinases that have been investigated as a possible target for molecular targeted therapy of various cancers. METHODS: Patients (n = 222) who underwent gastrectomy for primary gastric cancer were enrolled in this study. Tumor protein expression of EphA1 and EphB6 in surgically resected specimen was investigated using immunohistochemistry. The associations between expression of EphA1 and EphB6 and clinicopathological factors and prognosis were analyzed. RESULTS: High expression of EphA1 was associated with undifferentiated histology (P = 0.002), depth of tumor (P < 0.001), lymph node metastasis (P = 0.001), venous invasion (P = 0.015), stage (P = 0.001), and remote metastasis or recurrence (P < 0.001). In univariate analysis, patients with high expression of EphA1 had significantly poorer overall survival and relapse-free survival compared with patients with low EphA1 expression. The expression level of EphB6 was not associated with any clinicopathological factors and patient survival. Multivariate analysis indicated that depth of tumor [hazard ratio (HR) 9.26, 95 % confidence interval (CI) 0.03-0.46, P = 0.003], lymph node metastasis (HR 9.26, 95 % CI 0.07-0.39, P < 0.001), and high expression of EphA1 (HR 1.86, 95 % CI 0.29-0.99, P = 0.048) are independent prognostic factors for relapse-free survival. CONCLUSIONS: EphA1 is a possible target of molecular targeted therapy of gastric cancer.


Subject(s)
Adenocarcinoma/secondary , Biomarkers, Tumor/metabolism , Neoplasm Recurrence, Local/pathology , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, EphA1/metabolism , Stomach Neoplasms/pathology , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Aged , Biomarkers, Tumor/genetics , Female , Follow-Up Studies , Humans , Immunoenzyme Techniques , In Situ Hybridization, Fluorescence , Lymphatic Metastasis , Male , Neoplasm Invasiveness , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/mortality , Neoplasm Staging , Prognosis , Receptor Protein-Tyrosine Kinases/genetics , Receptor, EphA1/genetics , Receptors, Eph Family , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Survival Rate
12.
Proc Natl Acad Sci U S A ; 107(24): 10860-5, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20505120

ABSTRACT

Eph receptor tyrosine kinases and their ephrin ligands regulate cell navigation during normal and oncogenic development. Signaling of Ephs is initiated in a multistep process leading to the assembly of higher-order signaling clusters that set off bidirectional signaling in interacting cells. However, the structural and mechanistic details of this assembly remained undefined. Here we present high-resolution structures of the complete EphA2 ectodomain and complexes with ephrin-A1 and A5 as the base unit of an Eph cluster. The structures reveal an elongated architecture with novel Eph/Eph interactions, both within and outside of the Eph ligand-binding domain, that suggest the molecular mechanism underlying Eph/ephrin clustering. Structure-function analysis, by using site-directed mutagenesis and cell-based signaling assays, confirms the importance of the identified oligomerization interfaces for Eph clustering.


Subject(s)
Receptor, EphA1/chemistry , Amino Acid Sequence , Binding Sites , Cell Line , Crystallography, X-Ray , Ephrin-A1/chemistry , Ephrin-A1/genetics , Ephrin-A1/metabolism , Ephrin-A5/chemistry , Ephrin-A5/genetics , Ephrin-A5/metabolism , Humans , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes , Protein Structure, Secondary , Protein Structure, Tertiary , Receptor, EphA1/genetics , Receptor, EphA1/metabolism , Receptor, EphA2/chemistry , Receptor, EphA2/genetics , Receptor, EphA2/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction
13.
J Mol Biol ; 435(19): 168243, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37619706

ABSTRACT

The Eph (erythropoietin-producing human hepatocellular) receptor family, the largest subclass of receptor tyrosine kinases (RTKs), plays essential roles in embryonic development and neurogenesis. The intracellular Sterile Alpha Motif (SAM) domain presents a critical structural feature that distinguishes Eph receptors from other RTKs and participates in recruiting and binding downstream molecules. This study identified SASH1 (SAM and SH3 domain containing 1) as a novel Eph receptor-binding partner through SAM-SAM domain interactions. Our comprehensive biochemical analyses revealed that SASH1 selectively interacts with Eph receptors via its SAM1 domain, displaying the highest affinity for EphA8. The high-resolution crystal structure of the EphA8-SASH1 complex provided insights into the specific intermolecular interactions between these proteins. Cellular assays confirmed that EphA8 and SASH1 co-localize and co-precipitate in mammalian cells, with cancer mutations (EphA8 R942H or G978D) impairing this interaction. We demonstrated that SAM-SAM interaction is critical for SASH1-mediated regulation of EphA8 kinase activity, shedding new light on the Eph signaling pathway and expanding our understanding of the molecular basis of the tumor suppressor gene SASH1.


Subject(s)
Receptor, EphA1 , Sterile Alpha Motif , Tumor Suppressor Proteins , Animals , Female , Humans , Pregnancy , Embryonic Development , Receptor, EphA1/genetics , Receptors, Eph Family/genetics , Signal Transduction
14.
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
15.
Biol Open ; 11(10)2022 10 15.
Article in English | MEDLINE | ID: mdl-36214254

ABSTRACT

While the typical role of receptor tyrosine kinases is to receive and transmit signals at the cell surface, in some cellular contexts (particularly transformed cells) they may also act as nuclear proteins. Aberrant nuclear localization of receptor tyrosine kinases associated with transformation often enhances the transformed phenotype (i.e. nuclear ErbBs promote tumor progression in breast cancer). Rhabdomyosarcoma (RMS), the most common soft tissue tumor in children, develops to resemble immature skeletal muscle and has been proposed to derive from muscle stem/progenitor cells (satellite cells). It is an aggressive cancer with a 5-year survival rate of 33% if it has metastasized. Eph receptor tyrosine kinases have been implicated in the development and progression of many other tumor types, but there are only two published studies of Ephs localizing to the nucleus of any cell type and to date no nuclear RTKs have been identified in RMS. In a screen for protein expression of Ephs in canine RMS primary tumors as well as mouse and human RMS cell lines, we noted strong expression of EphA1 in the nucleus of interphase cells in tumors from all three species. This localization pattern changes in dividing cells, with EphA1 localizing to the nucleus or the cytoplasm depending on the phase of the cell cycle. These data represent the first case of a nuclear RTK in RMS, and the first time that EphA1 has been detected in the nucleus of any cell type.


Subject(s)
Receptor, EphA1 , Rhabdomyosarcoma , Animals , Child , Dogs , Humans , Mice , Muscle, Skeletal , Nuclear Proteins , Receptor, EphA1/genetics , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Tyrosine
16.
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
17.
Nat Neurosci ; 10(1): 67-76, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17143272

ABSTRACT

The development of dendritic spines is thought to be crucial for synaptic plasticity. Dendritic spines are retracted upon Eph receptor A4 (EphA4) activation, but the mechanisms that control this process are not well understood. Here we report an important function of cyclin-dependent kinase 5 (Cdk5) in EphA4-dependent spine retraction in mice. We found that blocking Cdk5 activity inhibits ephrin-A1-triggered spine retraction and reduction of mEPSC frequency at hippocampal synapses. The activation of EphA4 resulted in the recruitment of Cdk5 to EphA4, leading to the tyrosine phosphorylation and activation of Cdk5. EphA4 and Cdk5 then enhanced the activation of ephexin1, a guanine-nucleotide exchange factor that regulates activation of the small Rho GTPase RhoA. The association between EphA4 and ephexin1 was significantly reduced in Cdk5(-/-) brains and Cdk5-dependent phosphorylation of ephexin1 was required for the ephrin-A1-mediated regulation of spine density. These findings suggest that ephrin-A1 promotes EphA4-dependent spine retraction through the activation of Cdk5 and ephexin1, which in turn modulates actin cytoskeletal dynamics.


Subject(s)
Cyclin-Dependent Kinase 5/physiology , Dendritic Spines/physiology , Guanine Nucleotide Exchange Factors/physiology , Receptor, EphA4/metabolism , Animals , Animals, Newborn , Cells, Cultured , Cyclin-Dependent Kinase 5/deficiency , Dendritic Spines/drug effects , Embryo, Mammalian , Enzyme Activation , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , Humans , In Vitro Techniques , Luminescent Proteins/metabolism , Mice , Mice, Knockout , Nerve Tissue Proteins/metabolism , Neurons/cytology , Patch-Clamp Techniques/methods , Phosphorylation , Receptor, EphA1/genetics , Receptor, EphA1/metabolism , Transfection/methods , Tyrosine/metabolism , rhoA GTP-Binding Protein/metabolism
18.
Sichuan Da Xue Xue Bao Yi Xue Ban ; 42(2): 179-84, 2011 Mar.
Article in Zh | MEDLINE | ID: mdl-21500549

ABSTRACT

OBJECTIVE: To investigate the expression and prognostic significance of EphA2 and EphrinA-1 in ovarian serous carcinomas. METHODS: Ninety five tumors from the patients with ovarian serous carcinomas and 2 ovarian cancer cell lines were recruited. The expressions of EphA2 and EphrinA-1 were examined by means of immunohistochemistry. The relationships among protein expression and clinicopathological features, survival of patients were analyzed. The mRNA and protein expressions of EphA2 and EphrinA-1 in ovarian cancer cell lines were measured with semiquantitative polymerase chain reaction and western blotting. RESULTS: The protein expressions of EphA2 and EphrinA-1 in tumor were 92.6% (88/95)and 97.9% (93/95) respectively, while were 40%(8/20) and 30% (6/20) in adjacent ovarian tissue (P = 0.000). EphA2 immunohistochemical staining could be observed in both tumour and vascular endothelial cells, and EphrinA-1 mainly localized in the tumor cells. The expression of EphA2 was significantly associated with the expression of EphrinA-1 (r = 0.98, P = 0.02). There was no significant correlation between the expressions of EphA2/EphrinA-1 and age, FIGO stage, residual tumour size and histological grade. High levels of both EphA2 and EphrinA-1 protein expression were significantly associated with a shorter overall survival in multivariate analysis (P < 0.05). High levels of EphA2 and EphrinA-1 mRNA and protein were detected in OVCAR3 and SKOV3 cell lines. CONCLUSION: There are over-expression of EphA2 and EphrinA-1 in ovarian serous carcinomas and ovarian cancer cell lines. Tumours with higher expression levels of both EphA2 and EphrinA-1 significantly associated with poorer clinical outcome.


Subject(s)
Cystadenocarcinoma, Serous/metabolism , Ovarian Neoplasms/metabolism , Receptor, EphA1/metabolism , Receptor, EphA2/metabolism , Adult , Aged , Aged, 80 and over , Female , Humans , Middle Aged , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, EphA1/genetics , Receptor, EphA2/genetics
19.
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
20.
J Mol Biol ; 433(21): 167241, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34508727

ABSTRACT

Autonomous heavy-chain variable (VH) domains are the smallest functional antibody fragments, and they possess unique features, including small size and convex paratopes, which provide enhanced targeting of concave epitopes that are difficult to access with larger conventional antibodies. However, human VH domains have evolved to fold and function with a light chain partner, and alone, they typically suffer from low stability and high aggregation propensity. Development of autonomous human VH domains, in which aggregation propensity is reduced without compromising antigen recognition, has proven challenging. Here, we used an autonomous human VH domain as a scaffold to construct phage-displayed synthetic libraries in which aspartate was systematically incorporated at different paratope positions. In selections, the library yielded many anti-EphA1 receptor VH domains, which were characterized in detail. Structural analyses of a parental anti-EphA1 VH domain and an improved variant provided insights into the effects of aspartate and other substitutions on preventing aggregation while retaining function. Our naïve libraries and in vitro selection procedures offer a systematic approach to generating highly functional autonomous human VH domains that resist aggregation and could be used for basic research and biomedical applications.


Subject(s)
Aspartic Acid/chemistry , Binding Sites, Antibody , Complementarity Determining Regions/chemistry , Immunoglobulin Heavy Chains/chemistry , Peptide Library , Amino Acid Sequence , Aspartic Acid/metabolism , Binding Sites , Cloning, Molecular , Complementarity Determining Regions/genetics , Complementarity Determining Regions/metabolism , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Humans , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/metabolism , Kinetics , Models, Molecular , Protein Aggregates , Protein Binding , Protein Conformation , Protein Folding , Protein Interaction Domains and Motifs , Receptor, EphA1/genetics , Receptor, EphA1/immunology , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL