EFNA1 Human, HEK

Ephrin A1 Human Recombinant, HEK
Shipped with Ice Packs
In Stock

Description

Physiological Functions

  • Neuronal Development: Mediates axon guidance and synaptic plasticity via interactions with Eph receptors .

  • Angiogenesis: Promotes vascular endothelial cell migration through Rac1 GTPase activation .

  • Cell Adhesion/Repulsion: Facilitates bidirectional signaling at cell-cell junctions .

Oncogenic Mechanisms

EFNA1 is implicated in cervical cancer (CC) through the FOSL2-EFNA1-EphA2-Src/AKT/STAT3 axis:

  • SE-Driven Overexpression: Super-enhancers (SEs) in CC tumors upregulate EFNA1 transcription, correlating with poor prognosis .

  • Tumor Growth: Knockdown of EFNA1 in xenograft models reduced tumor volume by 60% and weight by 55% .

  • Signaling Pathway Activation: EFNA1 binding to EphA2 triggers phosphorylation of Src, AKT, and STAT3, driving proliferation and metastasis .

Key Studies

  • In Vitro: EFNA1 knockdown in CC cells (SiHa, HCC-94) suppressed proliferation (↓70% colony formation) and migration (↓80% invasion) .

  • In Vivo: Overexpression increased tumor growth by 2.5-fold, while inhibitors of AKT (MK2206) and STAT3 (Stattic) reversed this effect .

  • Clinical Correlation: High EFNA1 expression in CC patients correlates with advanced tumor staging (FIGO III/IV) and reduced 5-year survival (35% vs. 75% in low-expression cohorts) .

Table: EFNA1 in Cervical Cancer

ParameterEFNA1 High ExpressionEFNA1 Low Expression
Tumor Volume (mm³)1,200 ± 150450 ± 90
Ki-67 Proliferation85% positive cells25% positive cells
5-Year Survival35%75%
Data derived from CC patient cohorts and xenograft models .

Applications in Research

EFNA1 Human, HEK is widely used as a reagent in:

  • Cancer Studies: Investigating SE-driven oncogene regulation and therapeutic targeting .

  • Neurobiology: Mapping Eph receptor-ephrin interactions in neural networks .

  • Drug Development: Screening inhibitors of the Src/AKT/STAT3 pathway .

Future Directions

  • Therapeutic Targeting: Small-molecule inhibitors of EFNA1-EphA2 binding (e.g., ALW-II-41-27) show preclinical efficacy .

  • Biomarker Potential: EFNA1 expression in liquid biopsies could predict CC recurrence .

Product Specs

Introduction

EFNA1, a member of the ephrin (EPH) family, plays a crucial role in the central nervous system, contributing to its development and functionality. The EPH subfamily, known as the largest group of receptor protein kinases, is integral to these processes.

Description

Recombinant EFNA1 Human, produced in HEK293 cells, is a single, glycosylated polypeptide chain with a molecular weight of 20.2kDa (calculated). It comprises 170 amino acids, including a 6 a.a C-terminal His tag, spanning from amino acid positions 19 to 182.

Physical Appearance
White lyophilized powder, filtered for purity.
Formulation

EFNA1 is supplied as a lyophilized powder, filtered through a 0.4 μm filter. The protein was initially prepared in a phosphate buffered saline solution at a pH of 7.5 with a 5% (w/v) trehalose concentration before lyophilization. The initial concentration before lyophilization was 0.5mg/ml.

Solubility

To create a working stock solution, add deionized water to the lyophilized pellet until it reaches a concentration of approximately 0.5mg/ml. Ensure the pellet is fully dissolved before use. This product is not sterile; therefore, filter it through a sterile filter before introducing it to cell cultures.

Stability

Store the lyophilized protein at -20°C. After reconstitution, aliquot the protein solution to minimize freeze-thaw cycles. The reconstituted protein remains stable at 4°C for a limited period and exhibits consistent quality for at least one week when stored at this temperature.

Purity

The purity of this product is greater than 95.0%, as determined by SDS-PAGE analysis.

Synonyms

Ephrin-A1, EPLG1, TNFAIP4, LERK1, EFL1, ECKLG, EPH-related receptor tyrosine kinase ligand 1, Immediate early response protein B61, Tumor necrosis factor alpha-induced protein 4, TNF alpha-induced protein 4, ligand of eph-related kinase 1, tumor necrosis factor, alpha-induced protein 4.

Source

HEK293 cells.

Amino Acid Sequence

DRHTVFWNSS NPKFRNEDYT IHVQLNDYVD IICPHYEDHS VADAAMEQYI LYLVEHEEYQ LCQPQSKDQV RWQCNRPSAK HGPEKLSEKF QRFTPFTLGK EFKEGHSYYY ISKPIHQHED RCLRLKVTVS GKITHSPQAH DNPQEKRLAA DDPEVRVLHS IGHS HHHHHH.

Q&A

What is the role of EFNA1 in human cancer progression?

EFNA1 functions as a tumor-promoting protein in several cancer types, most notably in cervical cancer (CC) where it demonstrates tumor-specific expression patterns. Research has revealed that EFNA1 knockdown significantly inhibits tumor growth in xenograft mouse models, with measurable reductions in both tumor volume and weight compared to control groups . Additionally, EFNA1 overexpression promotes cell proliferation, migration, and invasion in cervical cancer cells, as demonstrated through colony formation, EdU staining, and transwell assays . In hepatocellular carcinoma (HCC), EFNA1 is overexpressed in approximately 90% of tumors compared to corresponding non-tumor tissues, and its expression is positively associated with alpha-fetoprotein (AFP) in most hepatoma cell lines .

How is EFNA1 expression regulated in human cells?

EFNA1 expression is primarily regulated through an epigenetic mechanism involving super-enhancers (SEs). ChIP-seq analysis has identified two enhancer constituents (E1-E2) within the EFNA1 SE locus that show significant enrichment of H3K27ac occupancy in cervical cancer samples compared to normal tissues . This regulatory mechanism is BRD4-dependent, as treatment with JQ1 (a small-molecule inhibitor blocking BRD4 binding to H3K27ac) reduces EFNA1 expression at both mRNA and protein levels . Transcription factor analysis has identified FOSL2 as a key regulator of EFNA1, with knockdown of FOSL2 resulting in decreased EFNA1 expression, while overexpression increases EFNA1 levels .

What experimental approaches are used to study EFNA1 function in HEK cells?

HEK 293T cells are frequently employed as a model system for studying EFNA1 function through several key experimental approaches:

  • Luciferase reporter assays: Using HEK 293T cells transfected with luciferase reporter plasmids containing exogenous EFNA1 super-enhancer constituents (EFNA1-P-E1~2) to assess the regulatory impact of enhancer elements on EFNA1 expression .

  • Transcription factor studies: Testing the regulatory relationship between transcription factors (like FOSL2) and EFNA1 expression through both knockdown and overexpression experiments followed by luciferase activity measurements .

  • Protein interaction studies: Investigating the interaction between EFNA1 and its receptor EphA2, as well as downstream signaling pathways.

  • Expression systems: Using HEK cells for recombinant production of EFNA1 for functional studies.

How can the super-enhancer-driven regulation of EFNA1 be experimentally validated?

Super-enhancer (SE) driven regulation of EFNA1 requires multi-layered experimental validation:

  • Chromatin immunoprecipitation (ChIP) analysis: Perform ChIP-seq using antibodies against H3K27ac (active enhancer mark), H3K4me1 (enhancer mark), and BRD4 (a key co-activator that binds to acetylated histones) to map enhancer regions. Data from cervical cancer samples shows significant enrichment of H3K27ac at the EFNA1 locus compared to normal tissues .

  • CRISPR/Cas9-mediated deletion: Delete individual enhancer constituents (E1, E2) and the promoter within the EFNA1 SE using CRISPR/Cas9 technology. In SiHa cells, deletion of these elements significantly reduced EFNA1 expression at both mRNA and protein levels, confirming their regulatory function .

  • Chromosome conformation capture: Utilize Hi-C assays to confirm direct physical interaction between the EFNA1 SE locus and the EFNA1 promoter, as demonstrated in SiHa and HCC-94 cells .

  • Pharmacological inhibition: Treat cells with BRD4 inhibitors like JQ1 to disrupt SE function and assess impact on EFNA1 expression. Research shows JQ1 treatment reduces H3K27ac occupancy at SE regions and decreases EFNA1 expression .

  • Exogenous enhancer testing: Introduce luciferase reporter plasmids containing the enhancer elements to assess their regulatory capacity in heterologous systems like HEK 293T cells .

What are the technical challenges in differentiating EFNA1 functions from other ephrin family members in HEK expression systems?

Several technical challenges exist when attempting to isolate EFNA1-specific functions:

  • Sequence homology: Ephrin family members share significant sequence homology, making selective targeting challenging. Researchers must design highly specific primers for qPCR and RNA interference experiments.

  • Receptor promiscuity: EFNA1 can bind multiple EphA receptors with varying affinities, while these receptors can also interact with other ephrin ligands. To address this, researchers should:

    • Use receptor-blocking antibodies specific to individual Eph receptors

    • Create receptor-specific knockdown/knockout cell lines

    • Employ competitive binding assays with recombinant proteins

  • Compensatory mechanisms: Knockdown of EFNA1 may lead to compensatory upregulation of other ephrin family members. Transcriptome analysis following EFNA1 manipulation is essential to identify such effects.

  • Functional divergence: Despite structural similarities, ephrin family members can have opposing functions. For example, while EFNA1 promotes tumor growth in cervical cancer, EFNA2 and EFNA5 overexpression significantly reduces cell proliferation and migration .

How can researchers resolve contradictory data regarding EFNA1 and EphA2 expression patterns in different cancer models?

Resolving contradictory expression patterns requires systematic investigation:

  • Tissue-specific regulation: The EFNA1 super-enhancer mechanism appears to be cancer-type specific. Pan-cancer analysis across 24 primary cancer types showed only about 25% of cancers manifest the EFNA1 super-enhancer signature . Researchers should verify whether regulatory mechanisms are conserved across their specific cancer model.

  • Receptor-ligand inverse correlation: In hepatoma cell lines, EFNA1 protein expression was inversely associated with EphA2 expression despite both being potential biomarkers for HCC . This apparent contradiction can be explained by:

    • Ligand-induced receptor internalization and degradation

    • Differential regulation at transcriptional versus post-translational levels

    • Context-dependent feedback mechanisms

  • Methodological approach integration:

    • Compare protein expression (western blot, IHC) with mRNA expression (qPCR, RNA-seq)

    • Conduct time-course experiments to capture dynamic expression changes

    • Perform single-cell analysis to identify cell population heterogeneity that might mask opposing expression patterns

  • Validation across multiple models: Combine results from cell lines, patient samples, and animal models to establish consistent patterns, as exemplified by the integration of ChIP-seq data from clinical samples with in vitro cell line experiments for EFNA1 .

What are the optimal conditions for expressing functional EFNA1 in HEK cell systems?

For optimal expression of functional EFNA1 in HEK cell systems:

  • Expression vector considerations:

    • Include the native EFNA1 signal peptide sequence for proper secretion

    • Consider adding a C-terminal tag (His, FLAG) that doesn't interfere with receptor binding

    • Use a vector with an appropriate promoter (CMV for high expression, EF1α for sustained expression)

  • Transfection optimization:

    • For transient expression: Use lipid-based transfection (Lipofectamine) or PEI at cell densities of 70-80% confluence

    • For stable cell lines: Select optimal antibiotic concentration through kill curve analysis

    • Consider using HEK293T cells (containing SV40 large T antigen) for enhanced expression levels

  • Post-translational modifications:

    • EFNA1 requires GPI-anchor attachment for proper membrane localization

    • Enable proper protein folding by growing cells at 32-34°C for 24-48 hours post-transfection

    • Add 10 mM sodium butyrate to enhance protein expression while maintaining proper folding

  • Validation of functional activity:

    • Confirm membrane localization through cell surface biotinylation

    • Verify receptor binding through co-immunoprecipitation with EphA receptors

    • Assess downstream signaling pathway activation (Src/AKT/STAT3) to confirm biological activity

How can researchers effectively measure the impact of EFNA1 on the Src/AKT/STAT3 signaling pathway?

To comprehensively evaluate EFNA1's impact on the Src/AKT/STAT3 pathway:

  • Phosphorylation status analysis:

    • Perform western blot analysis focusing on phosphorylated forms of key proteins: p-SRC, p-AKT, p-STAT3

    • Use human phosphorylated kinase arrays to simultaneously detect multiple phosphorylation events, as demonstrated in SiHa cells where EFNA1 knockdown decreased phosphorylation of several components in the AKT and STAT3 pathways

    • Include downstream targets like CCND1 and Vimentin to confirm pathway activation

  • Pathway manipulation approaches:

    • Use specific inhibitors (Dasatinib for Src, MK2206 for AKT, Stattic for STAT3) to block individual pathway components

    • Apply genetic approaches (siRNA, CRISPR) to selectively modulate pathway components

    • Rescue experiments with constitutively active forms of pathway components in EFNA1-knockdown conditions

  • Transcriptome analysis:

    • Conduct RNA-seq following EFNA1 manipulation to identify differentially expressed genes

    • Apply pathway enrichment analysis to comprehensively evaluate affected signaling networks

    • Compare transcriptome changes in EFNA1-manipulated cells with those treated with pathway-specific inhibitors

  • Functional validation:

    • Test whether pathway inhibitors can reverse EFNA1-induced phenotypes (proliferation, migration, invasion)

    • Confirm pathway involvement through xenograft models treated with pathway inhibitors

What controls are essential when studying EFNA1 expression in HEK cell models?

Essential controls for EFNA1 studies in HEK cells include:

  • Expression controls:

    • Empty vector transfection: Accounts for transfection-induced cellular stress

    • Related ephrin family member expression (EFNA2, EFNA5): Distinguishes EFNA1-specific effects from general ephrin effects

    • Dose-dependent expression: Establishes relationship between expression level and observed phenotypes

  • Knockdown/knockout controls:

    • Multiple shRNA/siRNA sequences targeting EFNA1: Minimizes off-target effects

    • Non-targeting shRNA/siRNA: Controls for RNA interference machinery activation

    • Rescue experiments: Reintroducing EFNA1 to confirm phenotype specificity

  • Pathway analysis controls:

    • Parallel analysis of upstream activators and downstream effectors

    • Inclusion of pathway inhibitors at effective concentrations

    • Time-course analysis to capture dynamic signaling events

  • Cell system controls:

    • Comparison with primary cells when available

    • Analysis across multiple HEK cell derivatives (HEK293, HEK293T, HEK293F)

    • Confirmation in disease-relevant cell lines (like SiHa and HCC-94 for cancer studies)

How can researchers accurately measure secreted versus membrane-bound EFNA1 in experimental systems?

Differentiating and quantifying secreted versus membrane-bound EFNA1 requires specialized techniques:

  • For secreted EFNA1:

    • ELISA: Develop sandwich ELISA using antibodies against different EFNA1 epitopes

    • Western blot of concentrated cell culture supernatant

    • Proximity ligation assay in fixed cells to detect secreted EFNA1 bound to cellular receptors

    • Detection of EFNA1 in patient serum samples as demonstrated in HCC studies

  • For membrane-bound EFNA1:

    • Cell surface biotinylation followed by pulldown with streptavidin beads

    • Flow cytometry with non-permeabilized cells using antibodies against extracellular EFNA1 domains

    • Membrane fractionation followed by western blot analysis

    • Immunofluorescence microscopy without cell permeabilization

  • Quantitative comparison:

    • Establish standard curves using recombinant EFNA1 protein

    • Express results as ratio of secreted to membrane-bound protein

    • Analyze changes in this ratio under different experimental conditions

  • Validation approaches:

    • Use PI-PLC enzyme treatment to cleave GPI-anchored proteins from the cell surface

    • Employ mutations in the GPI anchor sequence to create exclusively secreted versions

    • Compare findings with those reported for HCC, where EFNA1 levels were detected in culture supernatants and patient serum

How should researchers interpret contradictory results between in vitro and in vivo EFNA1 studies?

When confronted with contradictory results between experimental systems:

  • Consider context-dependent factors:

    • Microenvironment influence: In vivo systems include stromal cells, immune cells, and extracellular matrix components absent in vitro

    • Receptor availability: Expression patterns of EphA receptors may differ between culture systems and tissues

    • Concentration gradients: EFNA1 functions in concentration-dependent manner that may not be replicated in vitro

  • Methodological reconciliation:

    • Establish whether the same EFNA1 variants/isoforms were studied across systems

    • Compare protein expression levels between systems using quantitative approaches

    • Evaluate whether experimental timeframes were appropriate for observed endpoints

  • Integrated analysis approaches:

    • Patient-derived xenograft models to bridge in vitro and in vivo findings

    • 3D organoid cultures that better recapitulate tissue architecture

    • Ex vivo tissue slice cultures that maintain tissue structure while allowing manipulation

  • Documentation and reporting:

    • Clearly report all experimental parameters that could influence outcomes

    • Discuss potential reasons for disparities between systems

    • Consider whether contradictions reveal novel biology rather than experimental artifacts

What bioinformatic approaches are most effective for analyzing EFNA1 expression patterns across cancer datasets?

For comprehensive bioinformatic analysis of EFNA1 in cancer:

  • Multi-cancer analysis strategies:

    • Pan-cancer analysis across tumor types, as demonstrated in the study examining EFNA1 super-enhancer presence across 24 primary cancer types

    • Comparison of expression patterns across squamous cell carcinomas from different tissues

    • Integration of protein expression data (RPPA, mass spectrometry) with transcriptomic data

  • Single-cell transcriptomics applications:

    • Deconvolution of bulk tumor transcriptomes to identify cell-specific expression patterns

    • Direct single-cell RNA-seq analysis to reveal heterogeneity in EFNA1 expression

    • Trajectory analysis to map EFNA1 expression changes during disease progression

  • Correlation analysis approaches:

    • Identify transcription factors correlated with EFNA1 expression (similar to the identification of FOSL2)

    • Perform pathway enrichment analysis on genes co-expressed with EFNA1

    • Construct gene regulatory networks centered on EFNA1

  • Clinical correlation methods:

    • Survival analysis using Kaplan-Meier curves stratified by EFNA1 expression levels

    • Multivariate analysis to determine independent prognostic value

    • Correlation with clinical parameters like tumor stage and differentiation status

How can researchers differentiate between direct and indirect effects of EFNA1 manipulation in experimental systems?

Distinguishing direct versus indirect effects requires systematic experimental design:

  • Temporal analysis:

    • Perform time-course experiments to identify primary (early) versus secondary (late) effects

    • Use inducible expression systems to control the timing of EFNA1 expression/knockdown

    • Employ rapid inhibition techniques (e.g., degrader technologies) to capture immediate effects

  • Pathway dissection:

    • Use specific inhibitors of known EFNA1 downstream pathways (Src/AKT/STAT3) to block indirect effects

    • Combine EFNA1 manipulation with knockdown of potential mediators

    • Perform epistasis experiments by sequential manipulation of EFNA1 and downstream factors

  • Protein interaction approaches:

    • Use EFNA1 mutants that selectively disrupt specific protein interactions

    • Perform co-immunoprecipitation to identify direct binding partners

    • Apply proximity ligation assays to visualize direct interactions in situ

  • Mechanistic validation:

    • Construct EFNA1 dominant-negative variants to competitively inhibit specific interactions

    • Perform domain swapping between EFNA1 and other family members to map functional regions

    • Use recombinant EFNA1 protein treatment to identify direct effects in the absence of expression changes

What are best practices for integrating ChIP-seq and RNA-seq data to understand EFNA1 regulation?

For optimal integration of ChIP-seq and RNA-seq data:

  • Experimental design considerations:

    • Perform both assays on the same biological samples when possible

    • Include multiple time points to capture dynamic regulatory changes

    • Compare tumor and normal tissues to identify cancer-specific alterations

  • Data processing workflows:

    • Apply consistent quality control and normalization methods across datasets

    • Use specialized algorithms like ROSE to identify super-enhancers from H3K27ac ChIP-seq data

    • Correlate H3K27ac signal intensity at enhancers with transcript levels of associated genes

  • Integrative analysis strategies:

    • Map physical chromatin interactions (Hi-C data) to connect enhancers with target genes

    • Identify transcription factor binding motifs within active enhancer regions

    • Perform transcription factor ChIP-seq (e.g., FOSL2) to confirm regulatory mechanisms

  • Validation approaches:

    • CRISPR/Cas9-mediated deletion of enhancer elements followed by expression analysis

    • Luciferase reporter assays to test enhancer activity

    • Functional assays following manipulation of identified regulatory elements

What emerging technologies could advance our understanding of EFNA1 function in human disease models?

Several cutting-edge technologies hold promise for EFNA1 research:

  • CRISPR-based epigenome editing:

    • CRISPRa/CRISPRi systems to selectively activate or repress EFNA1 enhancers

    • Base editing to introduce specific mutations in regulatory elements

    • CRISPR screens targeting enhancer elements to identify critical regions

  • Advanced imaging approaches:

    • Live-cell imaging of EFNA1-receptor interactions using fluorescent protein fusions

    • Super-resolution microscopy to visualize EFNA1 clustering and signaling complex formation

    • Intravital microscopy to observe EFNA1 dynamics in living tissues

  • Single-cell multi-omics:

    • Integrated single-cell RNA-seq and ATAC-seq to correlate chromatin accessibility with expression

    • Single-cell proteomics to measure EFNA1 protein levels alongside posttranslational modifications

    • Spatial transcriptomics to map EFNA1 expression patterns within tissue architecture

  • Organoid and advanced 3D culture systems:

    • Patient-derived organoids to study EFNA1 in personalized disease models

    • Microfluidic organ-on-chip systems to study EFNA1 in tissue-specific contexts

    • Co-culture systems to investigate EFNA1-mediated cell-cell communication

How can researchers design experiments to uncover novel therapeutic strategies targeting the EFNA1 signaling axis?

Strategic experimental design for therapeutic development:

  • Target identification approaches:

    • Screen for small molecules that disrupt EFNA1-EphA receptor interactions

    • Identify druggable nodes in the EFNA1 signaling pathway using phosphoproteomics

    • Explore BRD4 inhibitors that selectively target the EFNA1 super-enhancer

  • Combination therapy investigation:

    • Test EFNA1 pathway inhibitors in combination with standard chemotherapeutics

    • Combine targeting of EFNA1 with inhibitors of compensatory pathways

    • Investigate synergistic effects between EFNA1 inhibition and immune checkpoint blockade

  • Biomarker development:

    • Validate EFNA1 as a serum biomarker for early cancer detection

    • Develop assays to measure EFNA1/EphA2 ratio as a prognostic indicator

    • Identify patient subgroups most likely to benefit from EFNA1-targeted therapies

  • Preclinical validation models:

    • Generate patient-derived xenografts with varying EFNA1 expression levels

    • Develop genetically engineered mouse models with tissue-specific EFNA1 alterations

    • Establish organoid biobanks from patients with EFNA1-driven tumors for drug screening

Product Science Overview

Discovery and Structure

Ephrin A1 was first discovered in a human carcinoma cell line. It is a glycosylated protein comprising 175 amino acids with a predicted molecular mass of 20.8 kDa. Due to glycosylation, it migrates as an approximately 26 kDa band in SDS-PAGE under reducing conditions .

Expression and Function

Ephrin A1 is ubiquitously expressed in various tissues and cells, including those of the immune system. It interacts with Eph receptors to mediate changes in cellular shape, motility, migration, and proliferation. These interactions are essential for normal cellular processes during development and adult tissue homeostasis .

Recombinant Production

Recombinant human Ephrin A1 is produced using HEK293 cells, a human embryonic kidney cell line. The DNA sequence encoding human Ephrin A1 (NP_004419.2) is expressed with a polyhistidine tag at the C-terminus. The recombinant protein is typically lyophilized from sterile PBS, pH 7.4, and may include protectants such as trehalose, mannitol, and Tween80 before lyophilization .

Applications in Research

Ephrin A1 and its interactions with Eph receptors are studied extensively for their roles in immune cell development, activation, and migration. These studies have implications for understanding disease pathogenesis and developing future immunotherapies .

Stability and Storage

Recombinant human Ephrin A1 is stable for up to twelve months when stored at -20°C to -80°C under sterile conditions. It is recommended to aliquot the protein to avoid repeated freeze-thaw cycles .

Ephrin A1’s diverse roles in cellular processes and its potential as a therapeutic target make it a significant focus of biomedical research.

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.