EFNA3 Human, Sf9

Ephrin A3 Human Recombinant, Sf9
Shipped with Ice Packs
In Stock

Description

EFNA3 Human Recombinant produced in Sf9 Baculovirus cells is a single, glycosylated polypeptide chain containing 434 amino acids (23-214aa) and having a molecular mass of 48.7kDa
EFNA3 is fused to a 242 amino acid hIgG-His-Tag at C-terminus and purified by proprietary chromatographic techniques.

Product Specs

Introduction

EFNA3 is part of the ephrin (EPH) family. The EPH-related receptors and ephrins constitute the largest subfamily of receptor protein-tyrosine kinases. They are involved in mediating developmental events, particularly in the nervous system and erythropoiesis. Ephrins are classified into two classes, ephrin-A (EFNA) and ephrin-B (EFNB), based on their structural and sequence similarities. The EFNA class ephrins attach to the membrane through a glycosylphosphatidylinositol linkage, while the EFNB class ephrins are transmembrane proteins.

Description

Recombinant human EFNA3, produced in Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain. It consists of 434 amino acids (23-214aa), resulting in a molecular weight of 48.7kDa. The EFNA3 protein is fused to a 242 amino acid hIgG-His-Tag at its C-terminus. Purification is achieved using proprietary chromatographic techniques.

Physical Appearance
The product appears as a clear, colorless solution that has been sterilized by filtration.
Formulation

The Fractalkine solution is provided at a concentration of 0.5 mg/ml. It is formulated in a buffer containing 10% Glycerol and Phosphate Buffered Saline (pH 7.4).

Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. Adding a carrier protein such as HSA or BSA (0.1%) is advisable for long-term storage. Repeated freezing and thawing of the product should be avoided.
Purity

The purity of the product is greater than 90%, as determined by SDS-PAGE analysis.

Synonyms

Ephrin-A3, EFL2, Ehk1-L, EPLG3, LERK3, EPH-related receptor tyrosine kinase ligand 3.

Source

Sf9, Baculovirus cells.

Amino Acid Sequence

ADPQGPGGAL GNRHAVYWNS SNQHLRREGY TVQVNVNDYL DIYCPHYNSS GVGPGAGPGP
GGGAEQYVLY MVSRNGYRTC NASQGFKRWE CNRPHAPHSP IKFSEKFQRY SAFSLGYEFH
AGHEYYYIST PTHNLHWKCL RMKVFVCCAS TSHSGEKPVP TLPQFTMGPN VKINVLEDFE
GENPQVPKLE KSISGLEPKS CDKTHTCPPC PAPELLGGPS VFLFPPKPKD TLMISRTPEV
TCVVVDVSHE DPEVKFNWYV DGVEVHNAKT KPREEQYNST YRVVSVLTVL HQDWLNGKEY
KCKVSNKALP APIEKTISKA KGQPREPQVY TLPPSRDELT KNQVSLTCLV KGFYPSDIAV
EWESNGQPEN NYKTTPPVLD SDGSFFLYSK LTVDKSRWQQ GNVFSCSVMH EALHNHYTQK SLSLSPGKHH
HHHH

 

Q&A

What is EFNA3 and what cellular functions does it mediate?

EFNA3 (Ephrin-A3) belongs to the ephrin family, which represents the largest subfamily of receptor protein-tyrosine kinases. These molecules play crucial roles in mediating developmental events, particularly in the nervous system and in erythropoiesis. Ephrins are divided into two classes: the ephrin-A (EFNA) class, which are anchored to the membrane by glycosylphosphatidylinositol linkages, and the ephrin-B (EFNB) class, which are transmembrane proteins. EFNA3 functions primarily in cell-to-cell signaling processes that regulate tissue development and cellular organization through bidirectional signaling mechanisms .

What is the molecular structure of recombinant EFNA3 Human, Sf9?

Recombinant EFNA3 Human produced in Sf9 Baculovirus cells is a single, glycosylated polypeptide chain containing 434 amino acids (residues 23-214 of the native sequence) with a molecular mass of 48.7kDa. The recombinant protein is fused to a 242 amino acid hIgG-His-Tag at the C-terminus to facilitate purification and detection. The protein maintains the structural characteristics necessary for binding to EphA receptors while providing additional features for laboratory manipulation .

What expression system is used for EFNA3 Human, Sf9 production?

EFNA3 Human recombinant protein is produced using Sf9 insect cells infected with a baculovirus expression system. This eukaryotic expression system is preferred for human proteins requiring post-translational modifications, particularly glycosylation. The Sf9 cell line, derived from Spodoptera frugiperda pupal ovarian tissue, provides an environment that allows for proper protein folding and glycosylation patterns that more closely resemble mammalian systems compared to prokaryotic expression systems .

What purification methods are recommended for working with EFNA3 Human, Sf9?

EFNA3 Human recombinant protein produced in Sf9 cells is typically purified using proprietary chromatographic techniques that exploit the C-terminal hIgG-His-Tag. Standard purification protocols would include:

  • Initial capture using immobilized metal affinity chromatography (IMAC) with Ni²⁺ or Co²⁺ resins that bind the His-tag

  • Secondary purification using size exclusion chromatography to remove aggregates and improve homogeneity

  • Optional ion exchange chromatography for removal of endotoxins and other contaminants

The purified protein typically achieves >90% purity as determined by SDS-PAGE analysis .

How should EFNA3 Human, Sf9 be stored to maintain optimal activity?

For short-term storage (2-4 weeks), EFNA3 Human recombinant protein should be stored at 4°C. For longer periods, the protein should be stored frozen at -20°C. To maintain protein stability during long-term storage, it is recommended to add a carrier protein such as 0.1% human serum albumin (HSA) or bovine serum albumin (BSA). The formulation typically contains 10% glycerol in phosphate-buffered saline (pH 7.4) to help maintain protein stability. Multiple freeze-thaw cycles should be avoided as they can lead to protein denaturation and loss of biological activity .

How does hypoxia influence EFNA3 expression patterns?

Research indicates that hypoxia induces EFNA3 expression through a hypoxia-inducible factor (HIF)-mediated mechanism. Interestingly, this regulation involves a complex transcriptional response at the EFNA3 locus. While the canonical EFNA3 mRNA is only minimally induced under hypoxic conditions, long non-coding RNAs (lncRNAs) from the same locus show robust upregulation in response to hypoxia. This suggests differential regulation of the various RNA species produced from the EFNA3 locus.

In vivo studies have demonstrated that EFNA3 induction upon von Hippel-Lindau (VHL) loss is partially prevented in tissues lacking both VHL and EPAS (HIF2α) alleles, suggesting that HIF2α regulates EFNA3 expression in certain tissues, such as liver and lung. This HIF-dependent induction of EFNA3 occurs both in vitro and in vivo, though the specific HIF isoform involved may vary by tissue type, possibly reflecting their differential tissue expression patterns .

What is the relationship between EFNA3 and immune cell function in multiple sclerosis?

Studies examining patients with relapsing-remitting multiple sclerosis (RR-MS) have identified significantly increased expression of ephrin-A3 on various immune cell populations compared to healthy controls. Specifically, patients with RR-MS show higher percentages of ephrin-A3-positive CD4+ T cells (5.2 ± 1.2% vs. 1.4 ± 0.4%), CD8+ T cells (9.1 ± 1.5% vs. 3.8 ± 1.0%), and T regulatory cells (Tregs) (7.0 ± 1.8% vs. 2.8 ± 0.6%).

Additionally, the mean fluorescence intensity (MFI) of ephrin-A3 was significantly higher on CD4+ T cells, CD8+ T cells, and other immune cell subsets from RR-MS patients. This increased expression may influence immune cell signaling and function, potentially contributing to the pathology of multiple sclerosis through altered cell-cell communication and immune regulation .

What mechanisms regulate EFNA3 protein levels post-transcriptionally?

The regulation of EFNA3 protein levels involves complex post-transcriptional mechanisms, particularly through interactions with microRNAs. Evidence suggests that miR-210, which is induced by hypoxia, prevents the translation of several mRNAs including EFNA3. This microRNA binds to the 3′-untranslated region of the EFNA3 mRNA.

Interestingly, EFNA3 lncRNAs may increase EFNA3 mRNA translation by functioning as competing endogenous RNAs that deplete miR-210 and other microRNAs targeting the EFNA3 3′-untranslated region. This represents a sophisticated regulatory mechanism where non-coding RNAs from the same locus can influence the expression of the protein-coding transcript, creating an integrated regulatory network responsive to cellular conditions like hypoxia .

What are the optimal conditions for functional assays using EFNA3 Human, Sf9?

When designing functional assays with EFNA3 Human recombinant protein, researchers should consider the following optimal conditions:

  • Buffer composition: Phosphate-buffered saline (pH 7.4) supplemented with 0.1-0.5% BSA is typically used for receptor binding assays

  • Temperature: 37°C for cellular assays to mimic physiological conditions

  • Concentration range: 10-500 ng/mL for dose-response studies, with 100 ng/mL often used as a standard working concentration

  • Incubation time: 15-60 minutes for binding assays, 2-24 hours for functional cellular responses

  • Receptor specificity: Include appropriate controls to verify specific binding to EphA receptors versus non-specific interactions

For co-culture bioassays measuring receptor phosphorylation, HEK-293T cells expressing EphA receptors can be stimulated with EFNA3 recombinant protein (0.5 μg/mL) and phosphorylation assessed via immunostaining for phosphorylated EphA receptors .

How can researchers effectively detect EFNA3 expression in experimental models?

To effectively detect EFNA3 expression in experimental models, researchers should employ a combination of techniques targeting both RNA and protein levels:

For RNA detection:

  • qPCR using TaqMan probes specific to different regions of the EFNA3 gene can distinguish between the canonical mRNA and lncRNA transcripts

  • Design primers to target regions specific to the canonical EFNA3 mRNA and regions common to all transcripts to quantify their relative abundance

  • RNA sequencing to comprehensively analyze all transcripts from the EFNA3 locus

For protein detection:

  • Western blotting using antibodies specific to EFNA3, with expected band size of approximately 48.7 kDa for the recombinant protein

  • Flow cytometry to quantify cell surface expression on different cell populations

  • Immunohistochemistry or immunofluorescence to visualize tissue distribution

Using these complementary approaches allows researchers to correlate RNA expression with protein levels and localization, providing a more complete understanding of EFNA3 biology in their experimental system .

What experimental approaches are effective for studying EFNA3-EphA receptor interactions?

To study EFNA3-EphA receptor interactions, researchers can employ several effective experimental approaches:

  • Co-immunoprecipitation assays to detect direct protein-protein interactions between EFNA3 and EphA receptors

  • Receptor phosphorylation assays using phospho-specific antibodies to assess activation of EphA receptors following EFNA3 stimulation

  • FRET (Förster Resonance Energy Transfer) or BRET (Bioluminescence Resonance Energy Transfer) to measure real-time interactions in living cells

  • Surface plasmon resonance (SPR) to determine binding kinetics and affinity constants

  • Cell-based functional assays measuring downstream signaling events, such as MAPK pathway activation

A specialized co-culture bioassay has been developed where HEK-293T cells expressing EphA receptors are stimulated with recombinant EFNA3 protein or co-cultured with immune cells expressing EFNA3. Receptor activation is then visualized by immunostaining for phosphorylated EphA receptors (p-Eph-r) and quantified by confocal microscopy .

How should experiments be designed to distinguish between canonical EFNA3 and its lncRNA variants?

Designing experiments to distinguish between canonical EFNA3 mRNA and its lncRNA variants requires thoughtful approaches to RNA detection and functional assessment:

  • RNA detection strategy:

    • Design qPCR primers and probes that target regions specific to the canonical EFNA3 mRNA

    • Create separate primer sets targeting regions common to all RNA isoforms

    • Compare expression levels and induction patterns between these transcript populations

  • Functional analysis:

    • Use RNA interference targeting specific regions unique to either canonical mRNA or lncRNAs

    • Employ CRISPR-Cas9 gene editing to selectively disrupt promoter regions for different transcript variants

    • Conduct reporter assays with different promoter regions to assess their differential regulation

  • Cellular localization:

    • Perform RNA fluorescence in situ hybridization (FISH) with probes specific to different transcript variants

    • Examine nuclear vs. cytoplasmic fractionation to determine subcellular localization patterns

Research has shown that the absolute expression levels and relative induction in response to hypoxia varied widely between these RNA species, with lncRNAs showing predominant expression under normoxic conditions and more robust upregulation in response to hypoxia compared to the canonical mRNA .

What controls should be included when studying EFNA3 expression in hypoxic conditions?

When studying EFNA3 expression under hypoxic conditions, a comprehensive set of controls should be included:

  • Oxygen-level verification:

    • Include oxygen sensing probes or chemical indicators to confirm hypoxic conditions

    • Monitor HIF-1α protein stabilization as a molecular marker of hypoxia

  • Positive controls:

    • Include well-established hypoxia-responsive genes such as VEGF, GLUT1, or EGLN3

    • These genes serve as internal standards to confirm the hypoxic response

  • HIF-dependency controls:

    • Include conditions with HIF inhibitors or HIF knockdown/knockout approaches

    • Compare wild-type cells with cells lacking specific HIF isoforms (HIF-1α, HIF-2α)

  • Time-course analysis:

    • Sample at multiple time points to distinguish between early and late hypoxic responses

    • This helps differentiate direct HIF targets from secondary effects

  • Normoxic recovery:

    • Include conditions where cells are returned to normoxia after hypoxic exposure

    • Assess the reversibility of EFNA3 expression changes

Research has shown that VHL-deficient tissues, which have constitutive HIF activity, show increased EFNA3 expression. In animals lacking both VHL and EPAS (HIF2α) alleles, this induction is partially prevented, confirming the HIF-dependency of EFNA3 expression .

What experimental design is recommended for studying EFNA3's role in immune cell function?

To effectively study EFNA3's role in immune cell function, the following experimental design is recommended:

  • Cell population isolation and characterization:

    • Isolate distinct immune cell populations (CD4+ T cells, CD8+ T cells, Tregs, B cells, monocytes)

    • Verify cell purity using flow cytometry with appropriate markers

    • Assess baseline EFNA3 expression across these populations

  • Functional assays:

    • Proliferation assays with and without EFNA3 manipulation

    • Cytokine production measurements following stimulation

    • Migration and adhesion assays to assess cellular mobility and interactions

  • Receptor interaction studies:

    • Co-culture experiments with cells expressing EphA receptors

    • Receptor phosphorylation assays to assess signaling activation

    • Blocking antibody experiments to confirm specificity

  • In vivo models:

    • Adoptive transfer experiments with EFNA3-manipulated immune cells

    • Disease models relevant to the research question (e.g., EAE for multiple sclerosis)

    • Tissue-specific conditional knockout approaches

  • Clinical correlation:

    • Compare findings from experimental models with patient samples

    • Stratify patients based on disease parameters and correlate with EFNA3 expression

    • Longitudinal sampling to assess changes over disease course

Studies in patients with relapsing-remitting multiple sclerosis have demonstrated significantly increased expression of ephrin-A3 on various immune cell populations, suggesting a role in disease pathophysiology .

How should researchers address variability in EFNA3 expression between different cell types?

When addressing variability in EFNA3 expression between different cell types, researchers should implement the following strategies:

  • Standardization approaches:

    • Use multiple reference genes for qPCR normalization, selected based on stability across the cell types being studied

    • Include internal calibrator samples across experimental batches

    • Report both relative and absolute quantification when possible

  • Statistical considerations:

    • Employ appropriate statistical tests that account for non-normal distributions often seen in gene expression data

    • Use larger sample sizes to account for inherent biological variability

    • Consider mixed-effects models when analyzing data from multiple experiments

  • Biological context:

    • Correlate EFNA3 expression with cell type-specific markers

    • Consider the developmental stage and activation state of each cell type

    • Assess the expression of EphA receptors in parallel to provide functional context

  • Technological approaches:

    • Validate findings using orthogonal methods (e.g., qPCR, RNA-seq, protein detection)

    • Consider single-cell analysis to resolve heterogeneity within populations

    • Use absolute quantification methods when comparing across very different cell types

Studies with multiple sclerosis patients have shown significant variability in EFNA3 expression across immune cell subsets, with CD4+ T cells, CD8+ T cells, and Tregs showing different baseline expression levels and fold-changes in disease states .

What analytical approaches help distinguish direct and indirect effects in EFNA3 signaling pathways?

To distinguish direct and indirect effects in EFNA3 signaling pathways, researchers should consider these analytical approaches:

  • Temporal analysis:

    • Conduct detailed time-course experiments to establish the sequence of molecular events

    • Early events (minutes to hours) are more likely to represent direct effects

    • Late events (hours to days) may reflect indirect or compensatory mechanisms

  • Pharmacological interventions:

    • Use specific inhibitors targeting known mediators in the pathway

    • Apply protein synthesis inhibitors to distinguish between effects requiring new protein synthesis versus immediate signaling events

  • Genetic manipulation strategies:

    • Implement CRISPR-Cas9 or RNAi approaches to selectively remove pathway components

    • Create mutation panels affecting specific binding or signaling domains

    • Use inducible expression systems to control the timing of component activation

  • Protein-protein interaction analysis:

    • Conduct proximity ligation assays to visualize direct protein interactions in situ

    • Perform pull-down assays coupled with mass spectrometry to identify direct binding partners

    • Use structural biology approaches to confirm binding interfaces

  • Systems biology approaches:

    • Develop computational models incorporating known pathway components

    • Use network analysis to predict and test indirect signaling routes

    • Apply causal inference statistical methods to time-series data

Research on EFNA3 lncRNAs has demonstrated their role in regulating EFNA3 protein levels through interactions with the microRNA machinery, representing an indirect regulatory mechanism distinct from direct transcriptional control .

How can researchers reconcile contradictory findings between in vitro and in vivo EFNA3 studies?

When faced with contradictory findings between in vitro and in vivo EFNA3 studies, researchers should employ the following reconciliation strategies:

  • Context assessment:

    • Evaluate differences in cellular microenvironments between in vitro and in vivo settings

    • Consider the influence of three-dimensional tissue architecture on signaling dynamics

    • Assess the impact of extracellular matrix components absent in standard culture systems

  • Methodological bridge-building:

    • Develop intermediate models such as organoids or ex vivo tissue cultures

    • Implement more physiologically relevant in vitro systems (co-cultures, flow conditions)

    • Design in vivo experiments that specifically address the mechanisms identified in vitro

  • Temporal and concentration considerations:

    • Compare the timing of events between systems (cellular responses may occur at different rates)

    • Assess whether physiological concentration ranges were used in in vitro studies

    • Consider dosing kinetics in vivo versus static concentrations typically used in vitro

  • Species and genetic background differences:

    • Identify potential species-specific variations in EFNA3 structure or regulation

    • Consider genetic background effects that may influence experimental outcomes

    • Use matched genetic backgrounds when possible to minimize confounding factors

  • Integration strategies:

    • Develop computational models that can account for differences between systems

    • Identify core conserved mechanisms versus context-dependent variations

    • Focus on validating key nodes in the signaling pathway across multiple systems

Research has shown that while canonical EFNA3 mRNA is barely induced in response to hypoxia in vitro, robust upregulation of bulk EFNA3 transcripts occurs. Similarly, in vivo studies demonstrate EFNA3 induction in VHL-deficient tissues, with different regulatory patterns observed across various organs, highlighting the importance of context in EFNA3 regulation .

What are common pitfalls in EFNA3 detection and how can they be avoided?

Researchers working with EFNA3 should be aware of these common detection pitfalls and implement the following solutions:

  • Transcript specificity challenges:

    • Pitfall: Standard primers may detect multiple transcript variants

    • Solution: Design transcript-specific primers that distinguish between canonical mRNA and lncRNAs

    • Validation: Verify primer specificity using known positive and negative controls

  • Protein detection issues:

    • Pitfall: Cross-reactivity with other ephrin family members

    • Solution: Use verified antibodies with demonstrated specificity against EFNA3

    • Validation: Include appropriate knockdown/knockout controls to confirm specificity

  • Post-translational modification variations:

    • Pitfall: Glycosylation patterns may differ between recombinant and endogenous protein

    • Solution: Use deglycosylation enzymes when comparing proteins from different sources

    • Validation: Compare migration patterns before and after enzymatic treatment

  • Low expression challenges:

    • Pitfall: Endogenous EFNA3 may be expressed at low levels in some cell types

    • Solution: Implement signal amplification methods or more sensitive detection techniques

    • Validation: Include positive control samples with confirmed EFNA3 expression

  • Solubility and aggregation issues:

    • Pitfall: EFNA3 may form aggregates affecting detection and function

    • Solution: Optimize buffer conditions and use fresh preparations when possible

    • Validation: Perform size exclusion chromatography to assess protein homogeneity

Research has shown that the absolute expression levels of canonical EFNA3 mRNA are low compared with the combined expression of all isoforms, suggesting that under normoxic conditions, the transcription of lncRNAs predominates. This highlights the importance of using detection methods that can distinguish between these transcript populations .

How can stability issues with EFNA3 protein preparations be addressed?

To address stability issues with EFNA3 protein preparations, researchers should implement these strategies:

  • Storage optimization:

    • For short-term use (2-4 weeks): Store at 4°C

    • For long-term storage: Maintain at -20°C

    • Add carrier proteins (0.1% HSA or BSA) for long-term stability

    • Use small aliquots to avoid multiple freeze-thaw cycles

  • Buffer composition improvements:

    • Include 10% glycerol in phosphate buffered saline (pH 7.4) as a cryoprotectant

    • Consider adding low concentrations of reducing agents to prevent disulfide bond formation

    • Test different buffer systems if standard formulations show stability issues

    • Monitor pH stability over storage time

  • Aggregation prevention:

    • Filter through 0.22 μm membranes before storage to remove pre-formed aggregates

    • Centrifuge samples briefly before use to pellet any insoluble material

    • Consider adding non-ionic detergents at low concentrations for proteins prone to aggregation

    • Monitor protein homogeneity using dynamic light scattering

  • Functional validation:

    • Implement activity assays to confirm protein functionality after storage

    • Compare fresh preparations with stored samples to quantify activity loss

    • Establish acceptance criteria for minimum activity levels

    • Develop accelerated stability testing protocols for new formulations

  • Alternative approaches:

    • Consider lyophilization for very long-term storage needs

    • Test different fusion tags that may enhance stability

    • Explore chemical stabilizers appropriate for the specific application

These recommendations are based on established protocols for maintaining recombinant EFNA3 stability, including specific storage conditions and formulation components that have been demonstrated to preserve activity .

What troubleshooting approaches are effective when EFNA3-receptor binding assays fail?

When EFNA3-receptor binding assays fail to yield expected results, implement this systematic troubleshooting approach:

  • Protein quality assessment:

    • Verify EFNA3 protein integrity by SDS-PAGE and Western blotting

    • Confirm proper folding using circular dichroism or thermal shift assays

    • Assess aggregation state using size exclusion chromatography or dynamic light scattering

    • Validate bioactivity using a simplified functional assay

  • Receptor verification:

    • Confirm EphA receptor expression levels in the test system

    • Verify receptor functionality using a known ligand as positive control

    • Check for potential receptor mutations that might affect binding

    • Assess receptor clustering and membrane distribution

  • Assay condition optimization:

    • Test multiple buffer compositions varying salt concentration and pH

    • Optimize protein concentrations for both ligand and receptor

    • Adjust incubation times and temperatures

    • Include appropriate blocking agents to reduce non-specific binding

  • Detection method evaluation:

    • Compare different detection antibodies or labeling approaches

    • Consider alternative detection technologies (radioligand, fluorescence, SPR)

    • Implement positive and negative controls for each detection method

    • Calibrate instrumentation using standard curves

  • Interference identification:

    • Test for competing factors in the biological system

    • Assess potential inhibitors present in buffer components

    • Consider steric hindrance from protein tags or fusion partners

    • Evaluate matrix effects when using complex biological samples

When properly optimized, receptor phosphorylation assays have successfully demonstrated that immune cells from patients with RR-MS show increased ability to activate EphA receptors in co-culture bioassays compared to cells from healthy controls .

How does EFNA3 expression in pathological conditions inform therapeutic strategies?

The altered expression of EFNA3 in pathological conditions provides valuable insights for developing therapeutic strategies:

  • Cancer applications:

    • EFNA3 is significantly increased in clear cell renal cell carcinoma (ccRCC) tumor cells compared to normal kidney tissue

    • This upregulation is associated with VHL deficiency and constitutive HIF activity

    • The expression pattern parallels that of other HIF target genes like EGLN3

    • Therapeutic approaches targeting EFNA3 or its regulatory mechanisms could potentially disrupt tumor progression

  • Autoimmune disease implications:

    • Patients with relapsing-remitting multiple sclerosis (RR-MS) show significantly increased EFNA3 expression on immune cells

    • Particularly elevated levels are observed on CD4+ T cells, CD8+ T cells, and Tregs

    • This expression pattern suggests EFNA3 as a potential biomarker or therapeutic target in MS

    • Modulating ephrin-Eph signaling could influence immune cell function and disease progression

  • Hypoxia-related disorders:

    • The hypoxia-inducible nature of EFNA3 suggests relevance in ischemic conditions

    • Understanding the differential regulation of canonical mRNA versus lncRNAs may reveal novel intervention points

    • Targeting the HIF-dependent induction of EFNA3 could modify cellular responses to hypoxic conditions

    • The relationship between EFNA3 lncRNAs and miRNA networks presents opportunities for RNA-based therapeutics

These pathological expression patterns provide rationale for developing EFNA3-targeting approaches that could modulate disease processes through altering cell-cell communication and signaling pathways .

What are the promising research directions for EFNA3 in neurological disorder treatments?

Based on current understanding of EFNA3 biology, several promising research directions for neurological disorder treatments emerge:

  • Multiple sclerosis therapeutic development:

    • Target the increased EFNA3 expression on immune cells of MS patients

    • Develop antibodies or small molecules that modulate EFNA3-EphA receptor interactions

    • Explore cell-specific delivery approaches to target particular immune cell populations

    • Investigate combination therapies targeting EFNA3 alongside established MS treatments

  • Neurodevelopmental disorder approaches:

    • Leverage EFNA3's role in neural development and synaptic plasticity

    • Explore temporal modulation of EFNA3 signaling during critical developmental windows

    • Investigate genetic variants affecting EFNA3 expression or function in neurodevelopmental conditions

    • Develop models to assess EFNA3 manipulation in neurodevelopmental processes

  • Neurodegeneration and injury response:

    • Study EFNA3's role in neural repair and regeneration after injury

    • Explore the intersection between hypoxic signaling and EFNA3 in neurodegenerative conditions

    • Investigate EFNA3-mediated effects on neuroinflammation and microglial function

    • Develop therapeutic approaches enhancing beneficial aspects of EFNA3 signaling while minimizing detrimental effects

  • Neural circuit modulation:

    • Target EFNA3 signaling to influence specific neural circuits

    • Develop spatiotemporally controlled delivery systems for precise manipulation

    • Investigate combinatorial approaches targeting multiple ephrin family members

    • Explore optogenetic or chemogenetic control of EFNA3-expressing cells

Evidence from MS patients showing altered EFNA3 expression on immune cells, combined with EFNA3's known roles in neural development, suggests significant potential in neurological disorder treatments that warrants further investigation .

How might the non-coding RNA regulatory mechanisms of EFNA3 be exploited therapeutically?

The complex non-coding RNA regulatory mechanisms governing EFNA3 expression offer innovative therapeutic opportunities:

  • lncRNA-targeting approaches:

    • Develop antisense oligonucleotides (ASOs) specifically targeting EFNA3 lncRNAs

    • Design small interfering RNAs (siRNAs) to selectively knock down lncRNA expression

    • Create CRISPR-based transcriptional modulators targeting lncRNA promoters

    • Develop aptamers that bind to and influence lncRNA structure and function

  • miRNA network modulation:

    • Design miRNA mimics to enhance post-transcriptional regulation of EFNA3

    • Develop miRNA sponges to sequester miRNAs that regulate EFNA3 expression

    • Target key miRNA processing components to influence the broader regulatory network

    • Create target site blockers that prevent miRNA binding to EFNA3 mRNA

  • Competitive endogenous RNA manipulation:

    • Design synthetic competing endogenous RNAs to influence EFNA3 mRNA translation

    • Target natural competing endogenous RNAs that affect EFNA3 expression

    • Modulate RNA-binding proteins involved in these regulatory networks

    • Develop small molecules that influence RNA-RNA interactions

  • Hypoxia-responsive therapeutic systems:

    • Create hypoxia-responsive delivery systems utilizing EFNA3 lncRNA promoters

    • Develop therapeutics that selectively target hypoxic tissue based on EFNA3 expression

    • Design synthetic biology circuits responsive to the same hypoxic signals that induce EFNA3

    • Target the interface between HIF signaling and EFNA3 regulation

Research has demonstrated that EFNA3 lncRNAs can influence EFNA3 mRNA translation by potentially functioning as competing endogenous RNAs that sequester miRNAs targeting EFNA3. This complex regulatory mechanism presents unique opportunities for RNA-based therapeutic interventions .

What technological advances would enhance EFNA3 functional studies?

Several emerging technological advances would significantly enhance EFNA3 functional studies:

  • Advanced protein engineering:

    • Designer EFNA3 variants with tunable receptor specificity

    • Optogenetic or chemogenetic EFNA3 constructs allowing temporal control of activity

    • Split protein complementation systems for studying EFNA3-receptor interactions in real-time

    • Bioorthogonal chemistry approaches for site-specific labeling of EFNA3 in complex systems

  • Single-cell technologies:

    • Single-cell RNA sequencing to resolve heterogeneity in EFNA3 expression

    • Single-cell proteomics to correlate transcript and protein levels at individual cell resolution

    • Spatial transcriptomics to map EFNA3 expression patterns in tissue context

    • Live-cell imaging technologies to track EFNA3-mediated signaling in real-time

  • Genome engineering enhancements:

    • Prime editing or base editing for precise manipulation of EFNA3 regulatory elements

    • CRISPR interference/activation systems targeting EFNA3 lncRNA promoters

    • Knock-in reporter systems for endogenous monitoring of EFNA3 expression

    • Tissue-specific inducible gene editing systems for temporal control in specific cell populations

  • Structural biology innovations:

    • Cryo-electron microscopy of EFNA3-receptor complexes in membrane contexts

    • Hydrogen-deuterium exchange mass spectrometry to map dynamic interaction interfaces

    • Artificial intelligence approaches to predict structural impacts of EFNA3 variants

    • In-cell NMR techniques to study EFNA3 interactions in physiological environments

These technological advances would provide unprecedented insights into EFNA3 biology, enabling more precise manipulation and measurement of its functions in both basic research and translational applications.

What unsolved questions remain about EFNA3's role in cellular signaling networks?

Despite significant advances, several critical questions about EFNA3's role in cellular signaling networks remain unsolved:

  • Receptor specificity determinants:

    • What structural features determine EFNA3's binding preferences for specific EphA receptors?

    • How does the cellular context influence receptor selectivity?

    • Do post-translational modifications alter receptor binding profiles?

    • How does EFNA3 compare functionally to other ephrin-A family members in specific signaling contexts?

  • Signaling dynamics questions:

    • What determines the balance between forward and reverse signaling in EFNA3-mediated cell-cell interactions?

    • How do temporal dynamics of EFNA3 signaling influence cellular outcomes?

    • What feedback mechanisms regulate EFNA3 signaling intensity and duration?

    • How does EFNA3 signaling integrate with other major signaling pathways?

  • Non-canonical functions:

    • Does EFNA3 have functions independent of EphA receptor binding?

    • Are there intracellular roles for EFNA3 beyond its classical membrane-bound functions?

    • How does EFNA3 contribute to the formation of specialized cellular structures?

    • Do soluble forms of EFNA3 play significant biological roles?

  • Regulatory network integration:

    • How do the lncRNAs from the EFNA3 locus specifically influence cellular phenotypes?

    • What determines the switch between different RNA species produced from the EFNA3 locus?

    • How does the hypoxia-responsive element interact with other regulatory elements?

    • What epigenetic mechanisms control EFNA3 expression in different cellular contexts?

Addressing these questions will require integrative approaches combining advanced molecular techniques with systems biology perspectives to unravel the complex roles of EFNA3 in cellular signaling networks .

How might integrated multi-omics approaches advance our understanding of EFNA3 biology?

Integrated multi-omics approaches offer powerful strategies to advance our understanding of EFNA3 biology:

  • Comprehensive expression profiling:

    • Combine transcriptomics, proteomics, and metabolomics across diverse cell types and conditions

    • Correlate EFNA3 RNA isoforms with protein levels and functional outcomes

    • Map the temporal dynamics of expression changes in response to hypoxia and other stimuli

    • Identify co-regulated genes and proteins to define functional modules

  • Epigenetic landscape characterization:

    • Integrate DNA methylation, histone modification, and chromatin accessibility data

    • Map enhancer-promoter interactions at the EFNA3 locus using Hi-C and related technologies

    • Correlate epigenetic patterns with expression of different EFNA3 RNA species

    • Identify transcription factor binding profiles at regulatory regions

  • Interactome mapping:

    • Define the complete protein-protein interaction network of EFNA3

    • Map RNA-protein interactions for EFNA3 lncRNAs

    • Characterize dynamic changes in interaction networks under different conditions

    • Identify mediators of EFNA3 signaling across different cell types

  • Clinical correlation and integration:

    • Correlate multi-omics profiles with disease phenotypes and progression

    • Identify biomarker signatures associated with EFNA3 activity

    • Create predictive models of therapeutic response based on EFNA3-related signatures

    • Develop patient stratification approaches for EFNA3-targeted interventions

By integrating these diverse data types, researchers can develop comprehensive models of EFNA3 biology that span from molecular mechanisms to clinical applications, enabling more precise and effective therapeutic strategies targeting this signaling pathway.

What are the recommended quality control parameters for EFNA3 Human, Sf9?

The following quality control parameters are recommended for EFNA3 Human, Sf9 recombinant protein:

ParameterAcceptance CriteriaRecommended Method
PurityGreater than 90.0%SDS-PAGE with Coomassie staining
IdentityConfirmation of EFNA3 sequenceWestern blotting with anti-EFNA3 and anti-His antibodies
Molecular Mass48.7 kDaMass spectrometry
Endotoxin Level< 1.0 EU/μg proteinLAL chromogenic endotoxin assay
SterilityNo microbial growthSterility testing
GlycosylationPresence of glycosylationPAS staining or lectin blotting
Binding ActivityEC50 within reference rangeELISA or SPR-based binding assay
Aggregation< 10% aggregatesSize exclusion chromatography
pH7.2-7.6pH measurement
AppearanceClear, colorless solutionVisual inspection

These parameters ensure that the recombinant EFNA3 protein meets the necessary standards for research applications, with particular emphasis on purity (>90% as determined by SDS-PAGE) and proper identity confirmation to ensure experimental reliability .

What specialized equipment is needed for advanced EFNA3 signaling studies?

Advanced EFNA3 signaling studies require specialized equipment across various technical domains:

  • Imaging systems:

    • Confocal microscopy with live-cell capabilities for real-time observation of receptor clustering

    • Super-resolution microscopy (STORM, PALM, or STED) for nanoscale visualization of signaling complexes

    • FRET/FLIM systems to detect protein-protein interactions in living cells

    • Light-sheet microscopy for 3D imaging of EFNA3 signaling in tissue contexts

  • Biochemical analysis equipment:

    • Surface plasmon resonance or bio-layer interferometry systems for kinetic binding studies

    • Isothermal titration calorimetry for thermodynamic characterization of interactions

    • Analytical ultracentrifugation for studying complex formation

    • Circular dichroism spectrometer for protein structure analysis

  • Cell analysis instruments:

    • Multi-parameter flow cytometry with cell sorting capabilities

    • Impedance-based real-time cell analysis systems for monitoring adhesion and migration

    • Microfluidic devices for controlled gradient formation and cell migration studies

    • Patch-clamp systems for electrophysiological measurements in neural studies

  • Molecular biology platforms:

    • Droplet digital PCR for absolute quantification of RNA isoforms

    • Next-generation sequencing platforms for transcriptome analysis

    • Automated Western blot systems for phosphorylation state analysis

    • CRISPR screening platforms for functional genomics

  • Data analysis resources:

    • High-performance computing resources for image analysis and modeling

    • Specialized software for network analysis and pathway mapping

    • Machine learning tools for pattern recognition in complex datasets

    • Database systems for integrating multiple data types

This equipment enables comprehensive analysis of EFNA3 signaling from molecular interactions to cellular outcomes, facilitating deeper understanding of its biological functions.

What are the key methodological differences when working with EFNA3 in primary cells versus cell lines?

Working with EFNA3 in primary cells versus established cell lines presents several key methodological differences that researchers must consider:

  • Expression system considerations:

    • Primary cells: Endogenous EFNA3 expression levels are typically lower and may vary between donors

    • Cell lines: May have altered baseline expression or can be engineered for consistent overexpression

    • Adaptation required: Lower detection sensitivity needed for primary cells; careful titration of reagents

  • Experimental timeline differences:

    • Primary cells: Limited lifespan requires efficient experimental design; earlier timepoints often necessary

    • Cell lines: Allow for longer experimental timelines and stable genetic modifications

    • Adaptation required: Optimize protocols for shorter duration with primary cells; consider passage number effects in cell lines

  • Transfection/transduction efficiency:

    • Primary cells: Often more difficult to transfect; require specialized protocols or viral delivery systems

    • Cell lines: Generally more amenable to standard transfection methods with higher efficiency

    • Adaptation required: Optimize delivery conditions for primary cells; consider nucleofection or specialized lipid formulations

  • Functional assay considerations:

    • Primary cells: Often show more physiologically relevant responses but with higher variability

    • Cell lines: More consistent responses but may lack key regulatory mechanisms

    • Adaptation required: Include more biological replicates with primary cells; validate cell line findings in primary cells

  • Culture condition requirements:

    • Primary cells: Often require specialized media with growth factors; sensitive to culture conditions

    • Cell lines: Generally more robust in standard culture conditions

    • Adaptation required: Carefully control microenvironment for primary cells; consider how culture conditions might affect EFNA3 expression

These methodological differences must be carefully considered when designing experiments and interpreting results, particularly when studying complex signaling systems like those involving EFNA3, which can show context-dependent functions .

Product Science Overview

Structure and Expression

Ephrin A3 is a single, glycosylated polypeptide chain containing 434 amino acids (23-214aa) and has a molecular mass of approximately 48.7 kDa . The recombinant form of Ephrin A3, produced in Sf9 Baculovirus cells, is fused to a 242 amino acid hIgG-His-Tag at the C-terminus and purified using proprietary chromatographic techniques .

Function and Significance

Ephrin A3 binds promiscuously to Eph receptors on adjacent cells, leading to contact-dependent bidirectional signaling. This signaling is essential for migration, repulsion, and adhesion during neuronal, vascular, and epithelial development . The signaling pathway downstream of the receptor is referred to as forward signaling, while the pathway downstream of the ephrin ligand is known as reverse signaling .

Applications and Usage

Recombinant Ephrin A3 is used in various research applications, including high-throughput screening, SDS-PAGE, and HPLC . It is crucial for studying the mechanisms of cell signaling and development, as well as for potential therapeutic applications in diseases related to abnormal cell signaling.

Storage and Stability

The recombinant Ephrin A3 protein is typically stored at 4°C if used within 2-4 weeks. For long-term storage, it is recommended to store the protein at -20°C with a carrier protein to avoid multiple freeze-thaw cycles .

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.