EFNA3 Human

Ephrin A3 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to EFNA3 Human

Ephrin-A3 (EFNA3) is a glycosylphosphatidylinositol (GPI)-anchored protein encoded by the EFNA3 gene in humans. It belongs to the ephrin (EPH) family, the largest subfamily of receptor protein-tyrosine kinases, which regulate developmental processes such as neural patterning, vascular remodeling, and immune cell interactions . EFNA3 interacts with Eph receptors to mediate bidirectional signaling, influencing cell adhesion, migration, and tissue boundary formation .

Gene and Protein Structure

  • Gene Location: Chromosome 1 (1q22) .

  • Protein:

    • Contains 217 amino acids (23–214 a.a.) with a molecular mass of 24 kDa in its non-glycosylated form .

    • Post-translational glycosylation increases apparent molecular weight to 60–65 kDa .

    • Includes a C-terminal human IgG1 Fc tag or N-terminal His-tag in recombinant forms for research applications .

Recombinant Production

ParameterACROBiosystems Prospec Bio
Expression SystemHEK293E. coli
Purity>90% (SDS-PAGE)>90% (SDS-PAGE)
TagFc tagHis-tag
Binding AffinityBinds EphA10 (0.1–2 ng/mL)N/A

Biological Functions and Pathways

EFNA3 plays critical roles in:

  • Axon Guidance: Mediates repulsive cues during neural development .

  • Immune Regulation: Modulates infiltration of B cells, T cells, macrophages, and dendritic cells in cancers .

  • Ras Signaling Pathway: Activates downstream effectors promoting cell proliferation and survival in hepatocellular carcinoma (HCC) .

Gastric Cancer (GC)

  • Immune Infiltration: High EFNA3 expression linked to reduced CD8+ T cells and increased regulatory T cells (Tregs) .

  • Pathway Enrichment: Associated with ribosome biogenesis and Ras signaling (p < 0.05) .

Therapeutic Potential

  • Immune Checkpoint Modulation: EFNA3 co-expresses with PDCD1 and CTLA4, suggesting combinatorial targeting could enhance immunotherapy efficacy .

  • Research Tools: Recombinant EFNA3 proteins (e.g., Fc-tagged or His-tagged) enable drug discovery and binding assays .

Key Research Findings

Study FocusKey ResultSource
HCC PrognosisEFNA3 amplification predicts poor survival
GC Immune MicroenvironmentEFNA3 inversely correlates with CD8+ T cells
Structural AnalysisGPI anchor critical for Eph receptor binding

Product Specs

Introduction
EFNA3 is part of the ephrin (EPH) family. Both ephrins and EPH-related receptors belong to the largest receptor protein-tyrosine kinases subfamily, known for their involvement in developmental processes, particularly in the nervous system and erythropoiesis. Ephrins are classified into two categories, ephrin-A (EFNA) and ephrin-B (EFNB), based on their structural and sequence similarities. EFNA class ephrins attach to the membrane via a glycosylphosphatidylinositol anchor, while EFNB class ephrins are transmembrane proteins.
Description
Recombinant human EFNA3, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 217 amino acids (residues 23-214). This protein has a molecular weight of 24 kDa. A 25 amino acid His-tag is fused to the N-terminus of EFNA3. Purification is achieved using proprietary chromatographic techniques.
Physical Appearance
A clear, colorless solution that has been sterilized by filtration.
Formulation
The EFNA3 protein solution is provided at a concentration of 1 mg/ml. It is formulated in a buffer containing 20 mM Tris-HCl (pH 8.0), 0.4 M Urea, and 10% glycerol.
Stability
For short-term storage (up to 4 weeks), keep the vial refrigerated at 4°C. For extended storage, freeze the product at -20°C. Adding a carrier protein like HSA or BSA (0.1%) is recommended for long-term storage. Avoid repeatedly freezing and thawing the product.
Purity
The purity of EFNA3 is determined by SDS-PAGE analysis to be greater than 90%.
Synonyms
Ephrin-A3, EFL2, Ehk1-L, EPLG3, LERK3, EPH-related receptor tyrosine kinase ligand 3.
Source
E.coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMQGPGG ALGNRHAVYW NSSNQHLRRE GYTVQVNVND YLDIYCPHYN SSGVGPGAGP GPGGGAEQYV LYMVSRNGYR TCNASQGFKR WECNRPHAPH SPIKFSEKFQ RYSAFSLGYE FHAGHEYYYI STPTHNLHWK CLRMKVFVCC ASTSHSGEKP VPTLPQFTMG PNVKINVLED FEGENPQVPK LEKSISG

Q&A

What is EFNA3 and what is its role in normal human physiology?

EFNA3 (Ephrin-A3) is a gene that encodes a member of the ephrin family of proteins that serve as ligands for Eph receptor tyrosine kinases. In normal physiology, ephrins like Ephrin-A3 regulate diverse biological processes by modulating cellular adhesion and repulsion . They play crucial roles in tissue development and cellular organization. To study EFNA3's normal physiological functions, researchers often use techniques such as immunohistochemistry on normal tissues, knockout models in developmental studies, and protein-protein interaction analyses to map its binding partners and downstream signaling effects.

How is EFNA3 gene expression regulated at the transcriptional level?

EFNA3 expression is regulated through multiple mechanisms, notably through at least two distinct promoter regions (Prm1 and Prm2) . The Prm2 promoter is strongly induced by hypoxia and the chemical inhibitor dimethyloxalylglycine (DMOG), which activates hypoxia-inducible factors (HIFs), while Prm1 remains largely unaffected by these conditions . Mechanistically, the hypoxic response of the Prm2 promoter is critically dependent on specific RCGTG motifs, as mutation of these elements completely abolishes hypoxic induction . To investigate EFNA3 transcriptional regulation, researchers should consider chromatin immunoprecipitation (ChIP) assays to detect RNA polymerase II binding to promoter regions, and luciferase reporter assays to evaluate promoter activity under different conditions.

What are the different RNA isoforms produced from the EFNA3 locus?

The EFNA3 gene locus produces multiple RNA isoforms, including the canonical protein-coding mRNA and several long noncoding RNAs (lncRNAs) . The expression levels and hypoxic induction vary significantly between these isoforms. The canonical coding isoform shows relatively low expression and minimal induction under hypoxic conditions, whereas the lncRNAs are more abundant and strongly upregulated in response to hypoxia . To accurately characterize these different isoforms, researchers should design primer pairs for all exons in the gene and use quantitative PCR (qPCR) with specific TaqMan probes to differentiate between the canonical mRNA and the lncRNAs. Additionally, RNA sequencing can provide comprehensive identification of all transcripts from this locus.

How does EFNA3 expression differ between normal tissues and various cancer types?

EFNA3 shows significantly higher expression in multiple cancer types compared to normal tissues. Particularly elevated expression has been observed in cervical squamous cell carcinoma, esophageal cancer, and bladder cancer . In bladder cancer specifically, immunohistochemical analysis has demonstrated EFNA3 protein expression in 57.4% of bladder urothelial carcinoma samples compared to only 31.3% of normal bladder tissues . Similarly, clear cell renal cell carcinoma (ccRCC) samples show increased EFNA3 expression compared to normal kidney tissue . To systematically evaluate EFNA3 expression across different cancer types, researchers should utilize comprehensive resources like TCGA data through platforms such as UALCAN, and validate findings through tissue microarrays and immunohistochemistry on patient samples.

What is the relationship between EFNA3 expression and clinical outcomes in cancer patients?

High EFNA3 expression correlates with poor clinical outcomes in multiple cancer types. In breast cancer, elevated EFNA3 expression strongly correlates with shorter metastasis-free survival . In bladder cancer, high EFNA3 expression is significantly associated with adverse clinicopathological features including larger tumor size, greater invasion depth, lymph node metastasis, distant metastasis, vascular invasion, and higher histological grade . Methodologically, researchers investigating this relationship should employ Kaplan-Meier survival analysis, logistic regression analysis, and Cox regression analysis to confirm the validity of EFNA3 as a predictor of patient prognosis and its significance in clinical pathology.

How does hypoxia affect EFNA3 expression in tumor cells?

Hypoxia strongly induces EFNA3 lncRNAs in tumor cells through an HIF-dependent mechanism, while having a marginal effect on the canonical EFNA3 mRNA . This induction is blocked by small interfering RNA (siRNA) directed against HIF1α, confirming the HIF-dependency of this response . In vivo studies using conditional VHL-knockout mouse models, which lead to constitutive HIF activity, further demonstrate increased EFNA3 lncRNA expression without significantly altering canonical EFNA3 mRNA levels . To study hypoxia-mediated regulation of EFNA3, researchers should use controlled hypoxic chambers, HIF stabilizing agents like DMOG, and genetic approaches to manipulate HIF activity, followed by transcript-specific expression analysis.

What bioinformatic resources are available for analyzing EFNA3 expression in human cancers?

Several bioinformatic resources provide valuable data for analyzing EFNA3 expression in human cancers. UALCAN (http://ualcan.path.uab.edu) offers access to TCGA data across 31 cancer types, allowing analysis of EFNA3 expression across tumor and normal samples, as well as various tumor subgroups based on cancer stage, tumor grade, and other clinicopathological features . LinkedOmics (http://www.linkedomics.org/login.php) can be used to examine relationships between EFNA3 expression and features such as methylation and mutation sites . The STRING search tool (http://string-db.org/) enables analysis of protein-protein interaction networks associated with EFNA3 . For pathway analysis, Gene Set Enrichment Analysis (GSEA) can identify potential pathways associated with EFNA3 expression and prognosis in different cancers .

What experimental techniques are most effective for studying EFNA3 protein function?

To effectively study EFNA3 protein function, researchers should employ a combination of techniques. Overexpression and knockdown studies using lentiviral vectors are valuable for assessing the effects of EFNA3 on cellular phenotypes . Protein interaction studies using co-immunoprecipitation can identify binding partners. For studying EFNA3's role in cancer progression, in vitro assays examining cell proliferation, migration, and invasion provide insights into cellular functions, while in vivo xenograft models are crucial for assessing metastatic potential . To investigate the regulatory mechanisms controlling EFNA3 protein levels, researchers should consider polysome profiling and ribosome profiling to examine translational efficiency, and study miRNA-mediated regulation, particularly focusing on miR-210, which has been implicated in regulating EFNA3 translation .

How can researchers effectively distinguish between EFNA3 mRNA and lncRNA expression in experimental settings?

Distinguishing between EFNA3 mRNA and lncRNA expression requires specific molecular approaches. Researchers should use commercially available TaqMan probes designed to amplify regions specific to the canonical EFNA3 mRNA (e.g., TaqMan1+2) and regions common to all RNA isoforms from this locus (e.g., TaqMan4+5) . Additionally, designing primer pairs for each exon in the gene allows determination of the response of every potential RNA isoform to experimental conditions such as hypoxia . For more comprehensive analysis, RNA sequencing with bioinformatic pipelines specifically designed to distinguish between coding and noncoding transcripts should be employed. Northern blotting with specific probes can also provide visual confirmation of the different RNA species.

How do EFNA3 lncRNAs regulate Ephrin-A3 protein levels without affecting mRNA expression?

EFNA3 lncRNAs regulate Ephrin-A3 protein levels through post-transcriptional mechanisms. Evidence suggests that these lncRNAs promote Ephrin-A3 protein accumulation without significantly affecting EFNA3 mRNA levels . One proposed mechanism involves miRNA sponging, particularly of miR-210, which is induced by hypoxia and known to prevent the translation of EFNA3 mRNA . The lncRNAs may increase EFNA3 mRNA translation by depleting miR-210 and other miRNAs that target the EFNA3 3′-untranslated region . To investigate this regulatory mechanism, researchers should perform experiments interfering with the miRNA processing machinery (e.g., by knocking down DGCR8), conduct RNA-RNA interaction studies to confirm direct binding between lncRNAs and miRNAs, and use translation efficiency assays to measure the impact on protein synthesis rates.

What are the molecular mechanisms underlying EFNA3's role in promoting cancer metastasis?

EFNA3 promotes cancer metastasis through several molecular mechanisms. In breast cancer, sustained expression of both Ephrin-A3 protein and EFNA3 lncRNAs increases the metastatic potential of tumor cells, potentially by enhancing their ability to extravasate from blood vessels into surrounding tissue . Rather than affecting vascularization, EFNA3 appears to exert a strong repulsive action that leads to increased intra- and extravasation, facilitating metastatic spread . To elucidate these mechanisms, researchers should employ transwell migration and invasion assays, 3D spheroid invasion models, and intra-vital microscopy in animal models to directly visualize tumor cell extravasation. Co-culture systems with endothelial cells can help assess tumor-endothelial interactions mediated by EFNA3.

How does the cross-talk between EFNA3 and other members of the Eph/ephrin family influence cancer progression?

The Eph/ephrin family includes multiple members that can interact and influence each other's functions in cancer progression. Interestingly, EFNA1, but not EFNA4, is also induced by hypoxia and shows increased expression in clear cell renal cell carcinoma, similar to EFNA3 . This suggests potential coordinated regulation of specific ephrin family members in response to tumor microenvironmental conditions. To investigate this cross-talk, researchers should perform comprehensive expression profiling of all Eph/ephrin family members across cancer types, conduct co-expression analysis to identify correlations, and use protein interaction studies to map physical interactions between family members. Functional studies with simultaneous manipulation of multiple family members (e.g., EFNA1 and EFNA3) would provide insights into their collective influence on cancer progression.

What controls and validation steps are essential when studying hypoxia-induced EFNA3 expression?

When studying hypoxia-induced EFNA3 expression, several critical controls and validation steps are essential. Researchers should include well-established hypoxia-responsive genes (e.g., P4HA) as positive controls and non-responsive genes (e.g., STT3A) as negative controls . Validation of HIF dependency should be performed using both pharmacological approaches (HIF stabilizers like DMOG) and genetic approaches (siRNA against HIF1α or HIF2α) . To confirm the specificity of promoter responses, mutation studies targeting putative HIF binding sites (RCGTG motifs) are crucial . Additionally, in vivo validation using genetic models such as conditional VHL-knockout mice provides important physiological context . Proper experimental design should include time-course studies to capture the dynamics of the hypoxic response and dose-response experiments with varying oxygen concentrations.

How should researchers design experiments to accurately assess EFNA3's role in tumor metastasis?

Designing experiments to accurately assess EFNA3's role in tumor metastasis requires careful consideration of multiple factors. First, researchers should use multiple cancer cell lines to ensure findings are not cell-type specific. Both gain-of-function (overexpression of EFNA3 or its lncRNAs) and loss-of-function (knockdown or knockout) approaches should be employed . In vivo metastasis models are essential, including both spontaneous metastasis models (primary tumor implantation followed by metastasis monitoring) and experimental metastasis models (direct injection of cells into circulation). Importantly, researchers should specifically examine extravasation ability, as evidence suggests this is a key mechanism by which EFNA3 promotes metastasis . Clinically relevant endpoints should be measured, including metastasis-free survival, which has been correlated with EFNA3 expression in patient data .

What therapeutic strategies targeting EFNA3 show the most promise for cancer treatment?

Targeting EFNA3 for cancer treatment holds significant therapeutic potential based on its role in promoting metastasis. Several approaches warrant investigation: (1) Small molecule inhibitors or blocking antibodies that disrupt Ephrin-A3 interaction with its receptors could prevent the repulsive action that facilitates tumor cell extravasation . (2) RNA-based therapeutics targeting EFNA3 lncRNAs, such as antisense oligonucleotides or siRNAs, could reduce Ephrin-A3 protein levels without affecting the canonical mRNA . (3) Combinatorial approaches targeting both EFNA3 and hypoxia-related pathways might provide synergistic effects, particularly in highly hypoxic tumors. When designing studies to evaluate these therapeutic strategies, researchers should include multiple cancer models, assess effects on both primary tumor growth and metastatic potential, and evaluate potential resistance mechanisms.

How might single-cell analysis advance our understanding of EFNA3's role in the tumor microenvironment?

Single-cell analysis offers tremendous potential to advance our understanding of EFNA3's role in the tumor microenvironment. This approach allows researchers to examine heterogeneity in EFNA3 expression within tumors and identify specific cell populations (both tumor and stromal) that express or respond to EFNA3. Single-cell RNA sequencing can reveal co-expression patterns with other genes at the individual cell level, potentially identifying novel signaling networks. Spatial transcriptomics can further contextualize EFNA3 expression within the architectural framework of the tumor, particularly in relation to hypoxic regions and vascular structures. To effectively implement single-cell approaches, researchers should combine multiple technologies (scRNA-seq, CyTOF, spatial transcriptomics) and develop computational pipelines specifically designed to analyze EFNA3-related gene signatures at the single-cell level.

What are the methodological challenges in translating preclinical findings on EFNA3 to clinical applications?

Translating preclinical findings on EFNA3 to clinical applications faces several methodological challenges. First, as with many preclinical studies, there's a risk of effect size inflation due to publication bias, which may lead to overestimation of EFNA3's importance . To address this, researchers should conduct meta-analyses of existing studies, employ standardized reporting guidelines, and preregister experimental protocols. Second, EFNA3's complex regulation through both coding and noncoding transcripts necessitates the development of clinical assays that can accurately detect all relevant RNA species. Third, establishing EFNA3 as a biomarker requires large-scale validation studies across diverse patient populations, with standardized methodologies for tissue collection, processing, and expression analysis. Finally, therapeutic targeting of EFNA3 must address challenges in drug delivery, particularly for RNA-based therapeutics, and potential off-target effects given the presence of multiple ephrin family members with potentially overlapping functions.

Product Science Overview

Introduction

Ephrin A3, also known as EFL-2, EHK1 ligand, EHK1-L, EPH-related receptor tyrosine kinase ligand 3, EFL2, EPLG3, and LERK3, is a member of the ephrin family of proteins. These proteins are cell surface glycosylphosphatidylinositol (GPI)-anchored ligands for Eph receptors, which are part of the largest subfamily of receptor protein-tyrosine kinases (RTKs). Ephrin A3 plays a crucial role in various biological processes, particularly in the nervous system and erythropoiesis .

Structure and Molecular Characteristics

Ephrin A3 is a GPI-bound ligand that binds promiscuously to Eph receptors on adjacent cells, leading to contact-dependent bidirectional signaling into neighboring cells . The recombinant human Ephrin A3 protein is typically expressed in human 293 cells (HEK293) and carries a human IgG1 Fc tag at the C-terminus. The protein has a calculated molecular weight of approximately 48.4 kDa, but it migrates as 60-65 kDa under reducing conditions due to glycosylation .

Biological Functions

Ephrin A3 is involved in several critical biological processes:

  • Axon Guidance and Topographic Map Formation: Ephrin A3 plays a significant role in the development of the nervous system by guiding axons to their appropriate targets and helping establish topographic maps.
  • Synaptic Plasticity: It is involved in the modulation of synaptic strength and plasticity, which are essential for learning and memory.
  • Angiogenesis: Ephrin A3 contributes to the formation of new blood vessels, a process vital for tissue growth and repair.
  • Tumor Growth and Metastasis: There is evidence suggesting that Ephrin A3 may play a role in tumor growth and metastasis, making it a potential target for cancer therapy .
Clinical and Translational Applications

Due to its involvement in various biological processes, Ephrin A3 has potential clinical and translational applications. It is being studied for its role in cancer therapy, particularly in targeting tumor growth and metastasis. Additionally, its role in angiogenesis makes it a potential target for therapies aimed at promoting tissue repair and regeneration .

Recombinant Production and Purity

Recombinant human Ephrin A3 is produced using human 293 cells (HEK293) and is typically purified to a high degree of purity (>90%) as determined by SDS-PAGE. The protein is lyophilized from a filtered solution and can be reconstituted for use in various applications. It is essential to follow the specific reconstitution protocol provided to ensure optimal performance .

Storage and Stability

For long-term storage, the recombinant human Ephrin A3 protein should be stored in its lyophilized state at -20°C or lower. After reconstitution, it is stable for up to 3 months at -70°C under sterile conditions. It is crucial to avoid repeated freeze-thaw cycles to maintain the protein’s stability and activity .

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.