• Title/Summary/Keyword: EphA

Search Result 41, Processing Time 0.03 seconds

Identification of EphA7 BAC clone containing a long-range dorsal midline-specific enhancer

  • Kim, Yu-Jin;Park, Soo-Chul
    • BMB Reports
    • /
    • v.44 no.2
    • /
    • pp.113-117
    • /
    • 2011
  • Previous studies suggest that EphA7 plays a critical role in neural tube closure or cortical progenitor apoptosis. In this report, enhancer trap assay was used to modify various EphA7 BAC clones and screen a large genomic region spanning 570 kb downstream of the EphA7 gene. We found that the dorsal midline-specific EphA7 enhancer resides on the 457D20 EphA7 BAC clone and is localized to a 35 kb genomic region in between +345.7 kb to +379.8 kb downstream of the EphA7 transcription start site. Identification of the EphA7 BAC clone containing a long-range dorsal midline enhancer may constitute a useful tool for investigating the biological functions of EphA7 in vivo.

EphA Receptors Form a Complex with Caspase-8 to Induce Apoptotic Cell Death

  • Lee, Haeryung;Park, Sunjung;Kang, Young-Sook;Park, Soochul
    • Molecules and Cells
    • /
    • v.38 no.4
    • /
    • pp.349-355
    • /
    • 2015
  • EphA7 has been implicated in the regulation of apoptotic cell death in neural epithelial cells. In this report, we provide evidence that EphA7 interacts with caspase-8 to induce apoptotic cell signaling. First, a pull-down assay using biotinylated ephrinA5-Fc showed that EphA7 co-precipitated with wild type caspase-8 or catalytically inactive caspase-8 mutant. Second, co-transfection of EphA7 with caspase-8 significantly increased the number of cleaved caspase-3 positive apoptotic cells under an experimental condition where transfection of EphA7 or caspase-8 alone did not affect cell viability or apoptosis. EphA4 also had a causative role in inducing apoptotic cell death with caspase-8, whereas EphA8 did not. Third, caspase-8 catalytic activity was essential for the apoptotic signaling cascade, whereas tyrosine kinase activity of the EphA4 receptor was not. Interestingly, we found that kinase-inactive EphA4 was well co-localized at the plasma membrane with catalytically inactive caspase-8, suggesting that an interaction between these mutant proteins was more stable. Finally, we observed that the extracellular region of the EphA7 receptor was critical for interacting with caspase-8, whereas the cytoplasmic region of EphA7 was not. Therefore, we propose that Eph receptors physically associate with a transmembrane protein to form an apoptotic signaling complex and that this unidentified receptor-like protein acts as a biochemical linker between the Eph receptor and caspase-8.

Engineering lacZ Reporter Gene into an ephA8 Bacterial Artificial Chromosome Using a Highly Efficient Bacterial Recombination System

  • Kim, Yu-Jin;Song, Eun-Sook;Choi, Soon-Young;Park, Soo-Chul
    • BMB Reports
    • /
    • v.40 no.5
    • /
    • pp.656-661
    • /
    • 2007
  • In this report, we describe an optimized method for generation of ephA8 BAC transgenic mice expressing the lacZ reporter gene under ephA8 regulatory sequences. First, we constructed a targeting vector that carries a 1.2 kb ephA8 DNA upstream of its first exon, a lacZ expression cassette, a kanamycin cassette, and a 0.7 kb ephA8 DNA downstream of its first exon. Second, the targeting vector was electroporated into cells containing the ephA8 BAC and pKOBEGA, in which recombinases induce a homologous recombination between the ephA8 BAC DNA and the targeting vector. Third, the FLP plasmid expressing the Flipase was electroporated into these bacteria to eliminate a kanamycin cassette from the recombinant BAC DNA. The appropriate structures of the modified ephA8 BAC DNA were confirmed by Southern analysis. Finally, BAC transgenic mouse embryos were generated by pronuclear injection of the recombinant BAC DNA. Whole mount X-gal staining revealed that the lacZ reporter expression is restricted to the anterior region of the developing midbrain in each transgenic embryo. These results indicate that the ephA8 BAC DNA contains most, if not all, regulatory sequences to direct temporal and spatial expression of the lacZ gene in vivo.

The EphA8 Receptor Phosphorylates and Activates Low Molecular Weight Phosphotyrosine Protein Phosphatase in Vitro

  • Park, Soo-Chul
    • BMB Reports
    • /
    • v.36 no.3
    • /
    • pp.288-293
    • /
    • 2003
  • Low molecular weight phosphotyrosine protein phosphatase (LMW-PTP) has been implicated in modulating the EphB1-mediated signaling pathway. In this study, we demonstrated that the EphA8 receptor phosphorylates LMW-PTP in vitro. In addition, we discovered that mixing these two proteins leads to EphA8 dephosphorylation in the absence of phosphatase inhibitors. Finally, we demonstrated that LMW-PTP, modified by the EphA8 autokinase activity, possesses enhanced catalytic activity in vitro. These results suggest that LMW-PTP may also participate in a feedback-control mechanism of the EphA8 receptor autokinase activity in vivo.

In-silico and structure-based assessment to evaluate pathogenicity of missense mutations associated with non-small cell lung cancer identified in the Eph-ephrin class of proteins

  • Shubhashish Chakraborty;Reshita Baruah;Neha Mishra;Ashok K Varma
    • Genomics & Informatics
    • /
    • v.21 no.3
    • /
    • pp.30.1-30.13
    • /
    • 2023
  • Ephs belong to the largest family of receptor tyrosine kinase and are highly conserved both sequentially and structurally. The structural organization of Eph is similar to other receptor tyrosine kinases; constituting the extracellular ligand binding domain, a fibronectin domain followed by intracellular juxtamembrane kinase, and SAM domain. Eph binds to respective ephrin ligand, through the ligand binding domain and forms a tetrameric complex to activate the kinase domain. Eph-ephrin regulates many downstream pathways that lead to physiological events such as cell migration, proliferation, and growth. Therefore, considering the importance of Eph-ephrin class of protein in tumorigenesis, 7,620 clinically reported missense mutations belonging to the class of variables of unknown significance were retrieved from cBioPortal and evaluated for pathogenicity. Thirty-two mutations predicted to be pathogenic using SIFT, Polyphen-2, PROVEAN, SNPs&GO, PMut, iSTABLE, and PremPS in-silico tools were found located either in critical functional regions or encompassing interactions at the binding interface of Eph-ephrin. However, seven were reported in nonsmall cell lung cancer (NSCLC). Considering the relevance of receptor tyrosine kinases and Eph in NSCLC, these seven mutations were assessed for change in the folding pattern using molecular dynamic simulation. Structural alterations, stability, flexibility, compactness, and solvent-exposed area was observed in EphA3 Trp790Cys, EphA7 Leu749Phe, EphB1 Gly685Cys, EphB4 Val748Ala, and Ephrin A2 Trp112Cys. Hence, it can be concluded that the evaluated mutations have potential to alter the folding pattern and thus can be further validated by in-vitro, structural and in-vivo studies for clinical management.

Identification of the 187 bp EphA7 Genomic DNA as the Dorsal Midline-Specific Enhancer of the Diencephalon and Mesencephalon

  • Kim, Yujin;Park, Eunjeong;Park, Soochul
    • Molecules and Cells
    • /
    • v.38 no.11
    • /
    • pp.1007-1012
    • /
    • 2015
  • EphA7 is a key molecule in regulating the development of the dien- and mesencephalon. To get insight into the mechanism of how EphA7 gene expression is regulated during the dorsal specification of the dien- and mesencephalon, we investigated the cis-acting regulatory sequence driving EphA7 to the dorsal midline of the dien- and mesencephalon. Transgenic LacZ reporter analysis, using overlapping EphA7 BACs, was used to narrow down the dorsal midline-specific enhancer, revealing the 25.3 kb genomic region as the enhancer candidate. Strikingly, this genomic DNA was located far downstream of the EphA7 transcription start site, +302.6 kb to +327.9 kb. Further enhancer mapping, using comparative genomic analysis and transgenic methods, showed that the 187 bp genomic DNA alone, approximately 305 kb downstream of the EphA7 transcription start site, was sufficient to act as the dorsal midline-specific enhancer of EphA7. Importantly, our results indicate that the 187 bp dorsal midline-specific enhancer is critically regulated by homeobox transcription factors during the development of the dien- and mesencephalon.

The p110${\gamma}$ PI-3 Kinase is Required for the Mechanism by Which the EphA8-induced Neurites are Modulated by Ephrin-A5 Engagement

  • Park, Soo-Chul
    • Animal cells and systems
    • /
    • v.8 no.1
    • /
    • pp.57-63
    • /
    • 2004
  • This study provides evidence that expression of EphA8 receptor in NG108-15 cells results in a substantial increase in the number of neurite-bearing cells. However, the EphA8-induced neurite outgrowth does not require either ephrin-A5 stimulation or ectopic expression of $p110{\gamma}$ PI-3 kinase. In contrast, co-expression of a lipid kinase-inactive $p110{\gamma}$ mutant together with EphA8 causes neurite retraction in the presence of ephrin-A5 stimulation. This effect was not observed in the absence of ephrin-A5 stimulation. Significantly, the tyrosine kinase activity of EphA8 is not important for either of these processes. Taken together, our results strongly suggest that $p110{\gamma}$ PI-3 kinase is critically involved in the regulatory process by which ephrin-A5 exerts effects on the EphA8-induced neurite outgrowth.

Expression Pattern of EphB2 in Gastric Cancer (위암에시 EphB2 단백의 발현 양상)

  • Song, Jae-Hwi;Kim, Chang-Jae;Cho, Young-Gu;Park, Cho-Hyun;Nam, Suk-Woo;Yoo, Nam-Jin;Lee, Jung-Young;Park, Won-Sang
    • Journal of Gastric Cancer
    • /
    • v.6 no.1
    • /
    • pp.25-30
    • /
    • 2006
  • Purpose: The EphB2 receptor, a member of the receptor tyrosine kinase family, is a target gene of the Wnt signaling pathway and may achieve a tumor suppressor function through regulation of cell growth and migration. Our aim was to determine whether an altered expression of EphB2 might be associated with gastric cancer development and, if so, to determine to which pathologic parameter it is linked. Materials and Methods: For the construction of the gastric cancer tissue microarray, 83 paraffin-embedded tissues containing gastric cancer areas were cored 3 times and transferred to the recipient master block. The expression patterns of EphB2 were examined on tissue microarray slides by using immunohistochemistry and were compared using pathologic parameters, including histological type, depth of invasion, lymph node metastatsis, and peritoneal dissemination. Results: The EphB2 protein was expressed in the normal gastric mucosal epithelium, especially in the bottom of the mucosa. We found loss of EphB2 expression in 30 (36.1%) of the 83 gastric cancer tissues. Statistically, loss of EphB2 expression was more common in gastric cancer with lymph-node metastasis. There was no significant correlation between EphB2 expression and depth of invasion, histologic type, or peritoneal dissemination. Conclusion: Our findings suggest that loss of EphB2 expression may represent a critical step in gastric carcinogenesis.

  • PDF

EphA2 Receptor Signaling Mediates Inflammatory Responses in Lipopolysaccharide-Induced Lung Injury

  • Hong, Ji Young;Shin, Mi Hwa;Chung, Kyung Soo;Kim, Eun Young;Jung, Ji Ye;Kang, Young Ae;Kim, Young Sam;Kim, Se Kyu;Chang, Joon;Park, Moo Suk
    • Tuberculosis and Respiratory Diseases
    • /
    • v.78 no.3
    • /
    • pp.218-226
    • /
    • 2015
  • Background: Eph receptors and ephrin ligands have several functions including angiogenesis, cell migration, axon guidance, fluid homeostasis, oncogenesis, inflammation and injury repair. The EphA2 receptor potentially mediates the regulation of vascular permeability and inflammation in response to lung injury. Methods: Mice were divided into 3 experimental groups to study the role of EphA2 signaling in the lipopolysaccharide (LPS)-induced lung injury model i.e., IgG+phosphate-buffered saline (PBS) group (IgG instillation before PBS exposure), IgG+LPS group (IgG instillation before LPS exposure) and EphA2 monoclonal antibody (mAb)+LPS group (EphA2 mAb pretreatment before LPS exposure). Results: EphA2 and ephrinA1 were upregulated in LPS-induced lung injury. The lung injury score of the EphA2 mAb+LPS group was lower than that of the IgG+LPS group ($4.30{\pm}2.93$ vs. $11.45{\pm}1.20$, respectively; p=0.004). Cell counts (EphA2 mAb+LPS: $11.33{\times}10^4{\pm}8.84{\times}10^4$ vs. IgG+LPS: $208.0{\times}10^4{\pm}122.6{\times}10^4$; p=0.018) and total protein concentrations (EphA2 mAb+LPS: $0.52{\pm}0.41mg/mL$ vs. IgG+LPS: $1.38{\pm}1.08mg/mL$; p=0.192) were decreased in EphA2 mAb+LPS group, as compared to the IgG+LPS group. In addition, EphA2 antagonism reduced the expression of phospho-p85, phosphoinositide 3-kinase $110{\gamma}$, phospho-Akt, nuclear factor ${\kappa}B$, and proinflammatory cytokines. Conclusion: This results of the study indicated a role for EphA2-ephrinA1 signaling in the pathogenesis of LPS-induced lung injury. Furthermore, EphA2 antagonism inhibits the phosphoinositide 3-kinase-Akt pathway and attenuates inflammation.

Identification of WDR7 as a Novel Downstream Target of the EphA8-Odin Signaling Complex

  • Park, Eun-Jeong;Park, Soo-Chul
    • Animal cells and systems
    • /
    • v.13 no.1
    • /
    • pp.9-15
    • /
    • 2009
  • Eph receptors and their ephrin ligands have been implicated in a variety of cellular processes such as cellular morphogenesis and motility. Our previous studies demonstrated that Odin, one of the Anks family proteins, functions as a scaffolding protein of the EphA8 signaling pathway leading to modulation of cell migration or axonal outgrowth. Here we show that WDR7 is associated with Odin and that it is possibly implicated in the EphA8 signaling pathway. WD40 repeats present in the COOH-terminal region of WDR7 appear to be crucial for its association with Odin, whereas the binding motif of Odin is located in between ankyrin repeats and PTB domain. Co-immunoprecipitation experiments revealed that association of WDR7 with Odin is enhanced by ephrin ligand treatment, possibly through forming large protein complexes including both EphA8 and ephrin-A5. Consistently, immunofluorescence staining experiments suggested that WDR7 constitute a component of the large protein complexes containing Odin, EphA8 and ephrin-A5. Taken together, our results suggest the WDR7-Odin complexes might be involved in the signaling pathway downstream of the EphA8 receptor.