EFNA1 Human, Sf9

Ephrin A1 Human Recombinant, Sf9
Shipped with Ice Packs
In Stock

Description

EFNA1 Human: Biological Significance

EFNA1 binds EphA receptors (e.g., EphA2, EphA4) to regulate bidirectional signaling, influencing cellular processes such as tumor suppression, vascular development, and neuronal morphogenesis . In cancer, EFNA1 exhibits dual roles: it suppresses glioblastoma by downregulating oncogenic EphA2 but promotes growth in breast and hepatocellular carcinomas .

Recombinant EFNA1 Production in Sf9 Cells

Sf9 insect cells are a preferred system for producing post-translationally modified eukaryotic proteins. Key features of EFNA1 expressed in Sf9 include:

Expression and Purification

  • Construct Design: Truncated EFNA1 (eA1-(19–175)) is generated by baculovirus-mediated expression, retaining the receptor-binding domain .

  • Tagging: An N-terminal 6xHis tag facilitates purification via affinity chromatography .

  • Yield: High-purity (>90%) protein is obtained, confirmed by SDS-PAGE .

ParameterDetails
Amino Acid Range19–175 (truncated extracellular domain)
Molecular Weight~21–24 kDa (varies by construct)
Expression SystemSf9 cells with recombinant baculovirus
Purification MethodNickel-affinity chromatography

Functional Characterization

EFNA1 produced in Sf9 retains full biological activity:

EphA2 Receptor Modulation

  • Receptor Downregulation: Sf9-derived EFNA1 induces EphA2 phosphorylation, internalization, and degradation in glioblastoma cells .

  • Morphological Changes: Treatment with 1 µg/mL EFNA1 triggers rapid cell rounding (within 15 minutes) in U-251 MG glioblastoma cells, mimicking effects of dimeric ephrinA1-Fc .

Signaling Pathways

  • ERK/AKT Inhibition: EFNA1 suppresses oncogenic signaling by reducing phosphorylated ERK and AKT levels .

  • Anti-Migratory Effects: Wild-type EFNA1 inhibits glioblastoma cell migration, while proteolytic-site mutants show impaired activity .

Applications in Research

  • Cancer Therapeutics: Truncated EFNA1 (eA1-(19–175)) is a candidate for targeting EphA2-overexpressing tumors .

  • Angiogenesis Studies: EFNA1’s role in vascular development is explored using Sf9-derived protein .

  • Structural Biology: Sf9 systems enable crystallographic studies of Ephrin-Eph complexes .

Comparison with Other Expression Systems

ParameterSf9 Insect CellsE. coli
Post-Translational ModificationsYes (e.g., disulfide bonds) No
GlycosylationNative-like (if required)Absent
YieldHigh (mg/L scale) Moderate
Functional ActivityFull receptor activation Partial (lacks modifications)

Challenges and Optimizations

  • Proteolytic Release: Soluble EFNA1 is generated via matrix metalloprotease cleavage at residues 174–181 . Mutants lacking this region remain membrane-bound, reducing therapeutic utility .

  • Storage Stability: Recombinant EFNA1 requires glycerol (10%) and low temperatures (-20°C) to prevent aggregation .

Future Directions

  • Therapeutic Engineering: Optimizing G-H loop mutants for enhanced EphA2 targeting .

  • Delivery Systems: Exploring nanoparticle conjugation for improved tumor penetration .

Product Specs

Introduction
EFNA1 is part of the ephrin (EPH) family, which constitutes the largest group of receptor protein kinases. These kinases play a crucial role in the development and functionality of the nervous system.
Description
Produced in Sf9 Baculovirus cells, EFNA1 is a single, glycosylated polypeptide chain. It comprises 406 amino acids (specifically, amino acids 19-182a.a.) and has a molecular mass of 46.6kDa. On SDS-PAGE, the molecular size will appear within the range of 40-57kDa. This EFNA1 protein is expressed with a 242 amino acid hIgG-His-tag located at the C-Terminus. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
A colorless solution that has undergone sterile filtration.
Formulation
The EFNA1 protein solution has a concentration of 0.5mg/ml. It is prepared with 10% glycerol and Phosphate Buffered Saline, maintaining a pH of 7.4.
Stability
For optimal storage, keep the solution at 4°C if the entire vial will be used within 2-4 weeks. For longer storage periods, freeze the solution at -20°C. Adding a carrier protein like 0.1% HSA or BSA is recommended for long-term storage. To preserve protein integrity, avoid repeated freeze-thaw cycles.
Purity
The purity of EFNA1 is determined to be greater than 90% based on SDS-PAGE analysis.
Synonyms

Ephrin A1, Immediate Early Response Protein B61, TNF Alpha-Induced Protein 4, Ephrin-A1, TNFAIP4, LERK-1, EPLG1, LERK1, Tumor Necrosis Factor, Alpha-Induced Protein 4, Eph-Related Receptor Tyrosine Kinase Ligand 1, EPH-Related Receptor Tyrosine Kinase Ligand 1, Tumor Necrosis Factor Alpha-Induced Protein 4, Ligand Of Eph-Related Kinase 1, ECKLG, EFL1, B61, EPH-related receptor tyrosine kinase ligand 1, LERK-1.

Source
Sf9, Baculovirus cells.
Amino Acid Sequence

ADPDRHTVFW NSSNPKFRNE DYTIHVQLND YVDIICPHYE DHSVADAAME QYILYLVEHE EYQLCQPQSK DQVRWQCNRP SAKHGPEKLS EKFQRFTPFT LGKEFKEGHS YYYISKPIHQ HEDRCLRLKV TVSGKITHSP QAHVNPQEKR LAADDPEVRV LHSIGHSLEP KSCDKTHTCP PCPAPELLGG PSVFLFPPKP KDTLMISRTP EVTCVVVDVS HEDPEVKFNW YVDGVEVHNA KTKPREEQYN STYRVVSVLT VLHQDWLNGK EYKCKVSNKA LPAPIEKTIS KAKGQPREPQ VYTLPPSRDE LTKNQVSLTC LVKGFYPSDI AVEWESNGQP ENNYKTTPPV LDSDGSFFLY SKLTVDKSRW QQGNVFSCSV MHEALHNHYT QKSLSLSPGK HHHHHH

 

Q&A

What is the molecular structure of EFNA1 and how does it differ from other ephrin family members?

EFNA1 is a member of the ephrin-A class of proteins that are anchored to the cell membrane via a glycosylphosphatidylinositol (GPI) linkage, distinguishing them from the transmembrane ephrin-B class. The human EFNA1 gene encodes a protein that binds primarily to EPHA2, EPHA4, EPHA5, EPHA6, and EPHA7 receptors . These binding interactions trigger bidirectional signaling cascades that regulate cellular processes including adhesion, migration, and proliferation. EFNA1's structural organization includes an extracellular ephrin domain responsible for receptor binding but lacks the cytoplasmic domain present in ephrin-B proteins, necessitating different mechanisms for reverse signaling. Unlike other ephrins, EFNA1 has been particularly implicated in cancer progression and angiogenesis, with significant associations to poor prognosis in multiple cancer types . The protein's unique binding preference for EPHA2 makes it distinct from other ephrin-A family members, which typically demonstrate broader receptor-binding patterns.

How does the EFNA1-EphA receptor interaction trigger bidirectional signaling cascades?

The EFNA1-EphA receptor interaction initiates a complex bidirectional signaling system where "forward signaling" occurs in the receptor-expressing cell while "reverse signaling" happens in the EFNA1-expressing cell . When EFNA1 binds to EphA receptors (primarily EphA2), it induces receptor clustering and autophosphorylation, activating downstream pathways including the Src/AKT/STAT3 axis . This activation leads to cytoskeletal reorganization, altered cell adhesion, and modified gene expression patterns. Recent research in cervical cancer demonstrates that EFNA1 cis-interaction with EphA2 leads to decreased EphA2 tyrosine phosphorylation but paradoxically activates the Src/AKT/STAT3 forward signaling pathway . In the EFNA1-expressing cell, reverse signaling occurs despite the lack of an intracellular domain through associated membrane proteins, affecting integrin function and cytoskeletal dynamics. This bidirectional communication creates context-dependent responses that regulate cell behavior through complex feedback mechanisms, making the EFNA1-EphA system a sophisticated mediator of cell-cell communication with significant implications for development and disease.

What experimental approaches can distinguish between forward and reverse signaling in the EFNA1-EphA system?

To distinguish between forward and reverse signaling in the EFNA1-EphA system, researchers must employ specialized experimental designs. For forward signaling assessment, one effective approach involves treating EphA-expressing cells with pre-clustered recombinant EFNA1-Fc fusion proteins and measuring receptor phosphorylation and downstream effector activation (Src, AKT, STAT3) . Phospho-specific antibodies against EphA receptors and downstream targets can be used in immunoblotting experiments to quantify activation levels. Conversely, reverse signaling can be studied by stimulating EFNA1-expressing cells with EphA-Fc fusion proteins while analyzing changes in cell adhesion molecules, cytoskeletal reorganization, and migration capacity. Cell-specific knockout models using CRISPR/Cas9 targeting either EFNA1 or EphA receptors enable more precise pathway dissection by isolating each component of the bidirectional system. Time-course studies are particularly valuable as they reveal the temporal dynamics of both signaling directions, which often operate on different timescales. Additionally, receptor mutants that preserve binding but lack kinase activity can help isolate reverse signaling effects from forward signaling interference, providing clearer experimental outcomes.

What are the optimal conditions for expressing functional human EFNA1 in the Sf9 baculovirus system?

For optimal expression of functional human EFNA1 in Sf9 cells, several critical parameters must be controlled. The baculovirus expression vector should contain the complete human EFNA1 coding sequence with its native signal peptide to ensure proper membrane targeting, though inclusion of a C-terminal purification tag (His6 or FLAG) is advisable if it doesn't interfere with the GPI-anchor attachment. Infection optimization requires maintaining a multiplicity of infection (MOI) between 2-5 to balance protein yield with cell viability. Temperature regulation is crucial—maintaining cultures at 27°C until infection, then reducing to 24-25°C post-infection can significantly improve proper folding and reduce aggregation. Harvest timing should be optimized through time-course experiments, typically occurring 48-72 hours post-infection before significant cell lysis occurs. Culture medium supplementation with trace elements and lipids supports GPI-anchor formation, while serum-free formulations like Sf-900™ III reduce background protein contamination. Expression should be verified through Western blotting against both EFNA1 and the purification tag, while functionality assessment requires EphA2 binding assays. For membrane-associated EFNA1, successful expression results in localization to the Sf9 cell membrane, which can be confirmed through immunofluorescence microscopy.

How do glycosylation patterns of EFNA1 in Sf9 cells differ from mammalian systems, and what are the functional implications?

The glycosylation patterns of human EFNA1 expressed in Sf9 cells significantly differ from those in mammalian systems, with important functional implications. Sf9 cells predominantly produce high-mannose, paucimannose, and non-complex N-glycans lacking terminal sialic acids that are typically present in mammalian cells. This glycosylation difference manifests in the following comparative aspects:

Glycosylation FeatureSf9-Expressed EFNA1Mammalian-Expressed EFNA1Functional Impact
N-glycan complexitySimple, high-mannoseComplex, with terminal sialic acidsMay affect protein half-life and receptor binding kinetics
O-glycosylationLimited or absentPresent where predictedPotential impact on protein stability and conformation
Phosphorylation of mannose residuesPresentRareMay alter recognition by mannose receptors
GPI-anchor compositionDifferent lipid compositionNative human lipid compositionCould affect membrane microdomain localization

These differences can impact EFNA1 functionality in several ways: altered receptor binding affinity or kinetics, modified protein stability and half-life, different clustering behavior in membrane microdomains, and potentially altered immunogenicity. For structural studies and initial functional characterization, Sf9-expressed EFNA1 is generally adequate, but for detailed signaling studies, particularly where glycan-dependent interactions are critical, mammalian expression systems might provide more physiologically relevant results. Researchers can partially address these limitations by enzymatic remodeling of Sf9-derived glycans or by using engineered Sf9 cell lines with humanized glycosylation machinery.

What approaches can overcome the challenge of purifying GPI-anchored EFNA1 from Sf9 membrane fractions?

Purifying GPI-anchored EFNA1 from Sf9 membrane fractions presents unique challenges requiring specialized approaches. A comprehensive purification strategy involves several key steps that must be carefully optimized:

  • Membrane isolation: Differential ultracentrifugation should be employed to separate membrane fractions (30,000-100,000g) after cell lysis using gentle methods like nitrogen cavitation or Dounce homogenization to preserve protein integrity.

  • Detergent solubilization: Critical detergent screening is essential—mild non-ionic detergents like n-dodecyl β-D-maltoside (DDM, 0.5-1%), CHAPS (0.5-1.5%), or octyl glucoside (0.5-2%) effectively solubilize EFNA1 while maintaining its native conformation. Temperature control (4°C) during solubilization prevents protein denaturation.

  • Affinity chromatography options:

    • For tagged EFNA1: Immobilized metal affinity chromatography (IMAC) for His-tagged protein

    • For untagged EFNA1: EphA2-Fc fusion protein coupled to Protein A/G resin enables selective capture of functional EFNA1

  • Size exclusion chromatography: As a polishing step to remove aggregates and achieve buffer exchange while maintaining detergent above critical micelle concentration.

  • GPI-anchor removal option: Phosphatidylinositol-specific phospholipase C (PI-PLC) treatment can release EFNA1 from membranes or detergent micelles if the soluble extracellular domain is sufficient for experiments.

High purity (>95%) typically requires a multi-step approach with careful optimization of detergent concentrations at each stage. Activity assays should be performed after each purification step to ensure functional integrity is maintained throughout the process.

What assays can reliably determine the functional activity of purified EFNA1 from Sf9 cells?

Verifying functional activity of Sf9-derived EFNA1 requires multiple complementary approaches:

Biochemical assays provide direct evidence of binding functionality. Surface Plasmon Resonance (SPR) or Bio-Layer Interferometry (BLI) can measure binding kinetics with recombinant EphA receptors—functional EFNA1 typically demonstrates nanomolar affinity for EPHA2 with characteristic association and dissociation rates. Enzyme-Linked Immunosorbent Assays (ELISA) using immobilized EphA-Fc fusion proteins provide quantitative binding assessment and can verify concentration-dependent interactions. Thermal shift assays can additionally confirm structural integrity by demonstrating proper protein folding.

Cell-based assays provide functional biological readouts. EphA2-expressing cell lines treated with purified EFNA1 should exhibit characteristic morphological changes (cell rounding/collapse) observable by microscopy within 15-30 minutes of treatment. Phosphorylation of EphA receptors and downstream signaling molecules (Src, AKT, STAT3) can be quantified by Western blotting using phospho-specific antibodies after EFNA1 stimulation. Migration inhibition assays using transwell chambers can demonstrate EFNA1's ability to inhibit cell motility, as it typically reduces migration by activating pathways that regulate cell attachment to extracellular matrix . Cell proliferation effects can be assessed in cancer cell lines, where functional EFNA1 often promotes proliferation as measured by MTT or EdU incorporation assays .

Comprehensive functional verification should include both binding parameters and cellular responses to ensure the purified protein maintains its biological activity.

How can researchers design experiments to study EFNA1's role in cancer progression?

Designing experiments to study EFNA1's role in cancer progression requires a multifaceted approach addressing expression, mechanistic function, and therapeutic potential:

Mechanistic investigations should employ both gain and loss-of-function approaches. CRISPR/Cas9-mediated EFNA1 knockout or shRNA-mediated knockdown in cancer cell lines allows assessment of proliferation (MTT/colony formation), migration (transwell assays), and invasion capabilities. Recent research in cervical cancer demonstrated that EFNA1 knockdown significantly inhibited tumor growth in xenograft mouse models, with sharp reductions in tumor volume, weight, and Ki-67 positive cells . Complementary EFNA1 overexpression studies confirm phenotypic effects through the same functional assays.

Signaling pathway analysis requires phosphorylation state assessment of downstream effectors (Src/AKT/STAT3) through Western blotting and immunoprecipitation to establish the molecular mechanisms of EFNA1-mediated cancer progression. In vivo models including orthotopic xenografts and patient-derived xenografts provide translational relevance, while selective pathway inhibitors can validate therapeutic targeting strategies.

What strategies can be employed to target the EFNA1-EphA axis for cancer therapy?

Based on current research, several promising strategies have emerged for therapeutically targeting the EFNA1-EphA axis in cancer:

  • Direct protein-protein interaction inhibition approaches include developing blocking antibodies against either EFNA1 or its receptors to prevent binding interactions, creating soluble decoy receptors (EphA-Fc fusion proteins) to sequester EFNA1, and designing peptide-based inhibitors that mimic binding interfaces but lack signaling capacity. These approaches directly interfere with the initial ligand-receptor engagement event.

  • Expression modulation strategies target EFNA1 at the genetic level. RNA interference technologies (siRNA/shRNA) delivered via nanoparticles have demonstrated efficacy in reducing EFNA1 expression levels in preclinical models . More recently, epigenetic approaches targeting super-enhancers that drive EFNA1 overexpression show promise—CRISPR/Cas9-mediated disruption of these regulatory elements or BET inhibitors can selectively reduce EFNA1 transcription in cancer cells .

  • Downstream signaling inhibition targets the effector pathways activated by EFNA1-EphA2 interaction. Small molecule inhibitors of Src (dasatinib, saracatinib), AKT (MK-2206), and STAT3 (stattic, napabucasin) have shown efficacy in attenuating EFNA1-driven tumor progression in preclinical models . Research demonstrates that inhibiting this pathway substantially reduces the tumorigenic capacity of EFNA1-overexpressing cancer cells in both in vitro and in vivo settings.

  • Combination approaches may offer the greatest therapeutic potential. Targeting EFNA1 signaling while simultaneously addressing complementary oncogenic pathways can overcome compensatory resistance mechanisms. Additionally, EFNA1-targeting approaches may sensitize tumors to conventional chemotherapy or radiotherapy by disrupting survival signaling.

How does super-enhancer regulation control EFNA1 expression in different cancer types?

Super-enhancer (SE) regulation of EFNA1 represents a sophisticated epigenetic control mechanism recently discovered in cancer biology. In cervical cancer, integrated epigenomic and transcriptomic profiling has identified a tumor-specific SE at the EFNA1 locus that drives its overexpression . This SE region spans approximately 10-15kb and is characterized by extensive H3K27ac enrichment, BRD4 occupancy, and open chromatin structure. Mechanistically, the EFNA1 SE contains multiple consensus binding sequences for the transcription factor FOSL2, whose knockdown significantly reduces luciferase activity and H3K27ac enrichment at the SE region .

Cancer-type specificity appears significant in SE-driven EFNA1 regulation. While the FOSL2-dependent mechanism has been well-characterized in cervical cancer, different transcription factor dependencies may exist in other cancer types like esophageal carcinoma where EFNA1 is also overexpressed. ChIP-seq analysis reveals distinctive enhancer-promoter interaction patterns across tumor types, suggesting tissue-specific regulatory circuits. The formation of these SEs appears to be an early oncogenic event that establishes feed-forward regulatory loops sustaining elevated EFNA1 expression.

The SE mechanism explains the pronounced overexpression of EFNA1 observed in tumors compared to normal tissues, providing a more nuanced understanding beyond simple gene amplification. Importantly, this regulatory mechanism presents a potential vulnerability that could be exploited therapeutically through epigenetic modifiers or transcription factor targeting.

What methodological challenges exist in studying bidirectional signaling in the EFNA1-EphA system?

Investigating bidirectional signaling in the EFNA1-EphA system presents several methodological challenges that researchers must address:

Membrane localization complexity represents a fundamental challenge as both EFNA1 (GPI-anchored) and EphA receptors (transmembrane) require a membrane environment for native conformation and clustering. Traditional biochemical methods often disrupt these membrane contexts, potentially altering signaling properties. Creating experimental systems that preserve native membrane organization while allowing controlled manipulation requires sophisticated approaches such as supported lipid bilayers or nanodiscs.

Signal isolation difficulties arise from the bidirectional nature of EFNA1-EphA signaling. Forward signaling through EphA receptors and reverse signaling through EFNA1 occur simultaneously, making it challenging to attribute observed effects to specific pathways. Researchers must design cell-specific knockout models using CRISPR/Cas9 targeting either EFNA1 or EphA receptors to isolate components. Alternatively, receptor mutants preserving binding but lacking kinase activity help separate reverse signaling effects.

Clustering dynamics present additional challenges as functional signaling requires receptor-ligand clustering rather than simple binary interactions. Techniques like single-molecule fluorescence microscopy or super-resolution imaging are needed to visualize these dynamic clustering events in real-time. Artificial clustering methods using antibodies or recombinant proteins must be carefully calibrated to reflect physiological conditions.

Context-dependent outcomes further complicate analysis, as identical molecular interactions can produce opposite effects depending on cellular context, expression levels, and signaling protein presence. Comprehensive experimental designs must incorporate multiple cell types and validate findings across diverse systems to establish generalizable mechanisms.

How can computational approaches advance our understanding of EFNA1 function and targeting?

Computational approaches offer powerful tools to advance EFNA1 research across multiple dimensions:

Structural biology computations can predict EFNA1-EphA receptor binding interfaces and conformational dynamics. Molecular dynamics simulations exploring the flexibility of EFNA1-EphA complexes in membrane environments can reveal transient interaction states not captured by static structural techniques. These simulations can identify allosteric sites for small molecule targeting and predict how post-translational modifications might alter binding characteristics. Structure-based virtual screening can then identify potential inhibitors of EFNA1-EphA interactions from large compound libraries.

Network analysis approaches integrate transcriptomic, proteomic, and phosphoproteomic data to construct comprehensive signaling networks downstream of EFNA1. Algorithms like WGCNA (Weighted Gene Co-expression Network Analysis) can identify gene modules correlated with EFNA1 expression across cancer datasets. Bayesian network modeling can infer causal relationships between signaling components, predicting how perturbations propagate through the network.

Multi-omics data integration allows researchers to correlate EFNA1 expression with genomic alterations, methylation patterns, and clinical outcomes across thousands of patient samples. Machine learning algorithms can identify biomarker signatures that predict response to EFNA1-targeting therapies and patient stratification approaches. For super-enhancer research, computational tools like ROSE (Rank Ordering of Super-Enhancers) algorithm can identify and quantify SE activity across the genome, pinpointing regulatory elements controlling EFNA1.

Single-cell computational analyses enable deconvolution of heterogeneous tumor cell populations, revealing how EFNA1-EphA signaling varies between cellular subpopulations and contributes to tumor heterogeneity, with potential implications for therapeutic resistance mechanisms.

What novel technologies could advance the understanding of EFNA1's role in the tumor microenvironment?

Emerging technologies present exciting opportunities to elucidate EFNA1's complex role in the tumor microenvironment:

Spatial transcriptomics and proteomics technologies, including Visium, Slide-seq, and imaging mass cytometry, can map EFNA1 expression and signaling activity within preserved tissue architecture. These approaches reveal how EFNA1-expressing cells interact with surrounding stromal and immune components, potentially uncovering previously unrecognized juxtacrine signaling networks. The spatial context is particularly relevant for EFNA1 research given its role in contact-dependent cell-cell communication.

Organoid and microfluidic systems enable controlled recreation of tumor-stroma interactions in three-dimensional contexts. Patient-derived organoids with EFNA1 genetic modifications can model cancer progression in systems that maintain physiological cell-cell contacts. Organ-on-chip platforms can incorporate multiple cell types (cancer, endothelial, immune) to study how EFNA1 influences their interactions under controlled conditions, including fluid flow and mechanical forces that affect ephrin signaling.

In vivo imaging approaches using advanced techniques like intravital microscopy with fluorescently tagged EFNA1 and EphA receptors allow real-time visualization of signaling events in living tumors. Optogenetic tools for precisely controlling EFNA1 activation in specific cell populations at defined timepoints can reveal temporal aspects of signaling that traditional genetic approaches cannot capture.

CRISPR-based epigenome editing offers unprecedented precision for manipulating the super-enhancers controlling EFNA1 expression. dCas9 fused to chromatin modifiers can target specific regulatory elements to understand how epigenetic changes at EFNA1 loci influence tumor progression and potentially identify synergistic combinations with existing therapies.

What is the potential for developing EFNA1 as a biomarker in precision oncology?

EFNA1 holds substantial promise as a biomarker in precision oncology, with several avenues for clinical translation:

Predictive biomarker potential exists for stratifying patients who might benefit from targeted therapies affecting the EFNA1-EphA signaling axis. Patients with super-enhancer-driven EFNA1 overexpression might respond particularly well to BET inhibitors or epigenetic modulators. Combined assessment of EFNA1 with its receptors and downstream effectors (phospho-Src, phospho-AKT, phospho-STAT3) could provide more refined prediction models.

Technical implementation requires standardized detection methods. Immunohistochemistry protocols with validated antibodies and scoring systems can be readily implemented in clinical pathology laboratories. For more quantitative assessment, RT-qPCR assays measuring EFNA1 mRNA levels offer higher sensitivity. Advanced approaches like digital spatial profiling could simultaneously assess EFNA1 expression and its spatial relationship with other tumor microenvironment components.

Clinical integration would involve incorporating EFNA1 testing into molecular tumor boards and treatment decision algorithms. Prospective clinical trials stratifying patients based on EFNA1 expression status would be needed to definitively establish its predictive value for specific therapeutic approaches.

How might the EFNA1-EphA signaling axis be exploited for immunotherapy applications?

The EFNA1-EphA signaling axis presents several innovative opportunities for enhancing cancer immunotherapy strategies:

Targeting the immunosuppressive tumor microenvironment represents a promising approach since EFNA1-EphA signaling influences immune cell trafficking and function. Recent research suggests that ephrin-EphA interactions regulate T-cell migration and activation. Disrupting EFNA1-EphA2 interactions could potentially enhance T-cell infiltration into tumors by modifying adhesion and migration properties of both cancer and immune cells. Combination strategies pairing EFNA1-targeting agents with immune checkpoint inhibitors could overcome resistance mechanisms by simultaneously addressing immune exhaustion and tumor microenvironment barriers.

Engineered immune cell approaches offer another avenue. CAR-T cells specifically targeting EFNA1-overexpressing tumor cells could provide selective killing of cancer cells with minimal off-target effects. Alternatively, T-cell engagers (BiTEs) incorporating EphA2-binding domains could redirect T-cell cytotoxicity to EFNA1-expressing tumors. The bidirectional nature of ephrin-Eph signaling could be exploited to simultaneously activate immune cells while disabling tumor cell defensive mechanisms.

Vaccine-based strategies might utilize EFNA1 as a tumor-associated antigen. Peptide vaccines derived from EFNA1 sequences or dendritic cell vaccines pulsed with EFNA1 could generate anti-tumor immune responses. The tumor-specific super-enhancer regulation of EFNA1 potentially creates neo-epitopes that could serve as highly selective vaccination targets.

Dual-function therapeutics could combine direct EFNA1-EphA blocking with immunomodulatory activities. Bispecific antibodies simultaneously targeting EFNA1/EphA2 and immune-activating receptors could localize immune stimulation to the tumor microenvironment, potentially improving efficacy while reducing systemic immune-related adverse events.

Product Science Overview

Structure and Production

Ephrin A1 produced in Sf9 Baculovirus cells is a single, glycosylated polypeptide chain containing 406 amino acids (19-182a.a.) and has a molecular mass of approximately 46.6 kDa . The molecular size on SDS-PAGE appears at approximately 40-57 kDa . This recombinant protein is expressed with a 242 amino acid hIgG-His-tag at the C-terminus and is purified using proprietary chromatographic techniques .

Physical Properties
  • Formulation: Ephrin A1 protein solution (0.5 mg/ml) contains 10% glycerol and Phosphate Buffered Saline (pH 7.4) .
  • Appearance: Sterile filtered colorless solution .
  • Stability: The protein should be stored at 4°C if used within 2-4 weeks. For longer storage, it should be frozen at -20°C, and it is recommended to add a carrier protein (0.1% HSA or BSA) to avoid multiple freeze-thaw cycles .
Biological Function

Ephrin A1 is involved in several key biological processes, including:

  • Nervous System Development: Ephrin A1 and its receptors are essential for the proper development and function of the nervous system .
  • Cell Signaling: Ephrin A1 interacts with Eph receptors to mediate cell signaling, which is crucial for cell positioning and tissue architecture .
Applications

Recombinant Ephrin A1 is widely used in research to study its role in cell signaling, development, and disease. It is particularly valuable in understanding the mechanisms of nervous system development and the potential therapeutic applications in neurodegenerative diseases and cancer .

Handling and Usage
  • Purity: Greater than 90% as determined by SDS-PAGE .
  • Safety: ProSpec’s products, including Ephrin A1, are intended for laboratory research use only and should not be used as drugs, agricultural or pesticidal products, food additives, or household chemicals .

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.