EPF2 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% ProClin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
EPF2 antibody; At1g34245 antibody; F23M19Protein EPIDERMAL PATTERNING FACTOR 2 [Cleaved into: MEPF2] antibody
Target Names
EPF2
Uniprot No.

Target Background

Function

EPF2 (Epidermal Patterning Factor 2) is a mobile signaling peptide that plays a crucial role in regulating stomatal development in plants. It controls stomatal patterning by inhibiting the transition of protodermal cells into meristemoid mother cells (MMCs), thus influencing the number of cells that enter and remain in the stomatal lineage. This regulatory function is non-cell-autonomous, meaning its effects extend beyond the cells where it's produced. EPF2 mediates stomatal development inhibition and acts as a key component of the MEPF2 (mobile signal controlling stomatal development) pathway. ERECTA serves as a major receptor for EPF2. The activity of EPF2 is regulated by cleavage via CRSP (a secreted protease), resulting in inactivation. Furthermore, EPF2 may compete with SOMATOGEN for binding to the TMM receptor, further influencing stomatal density.

Gene References Into Functions

The following studies provide further insight into EPF2's function:

  1. Hypermethylation of the EPF2 gene promoter in ROS1 mutants reduces EPF2 expression, leading to stomatal overproduction. PMID: 24898766
  2. The secreted protease CRSP cleaves the EPF2 pro-peptide, repressing stomatal development. PMID: 25043023
  3. Plants lacking EPF2 and EPFL9 exhibit a higher proportion of non-stomatal epidermal cells, indicating their role in regulating stomatal density. PMID: 22232766
  4. EPF2 expression precedes EPF1 during leaf development, suggesting a role in determining the number of cells entering and remaining in the stomatal lineage. PMID: 19398336
  5. EPF2 encodes a small secretory peptide expressed in MMCs, regulating epidermal cell density. PMID: 19435754
Database Links

KEGG: ath:AT1G34245

STRING: 3702.AT1G34245.1

UniGene: At.39780

Protein Families
Plant cysteine rich small secretory peptide family, Epidermal patterning factor subfamily
Subcellular Location
Secreted.
Tissue Specificity
Expressed in leaves, especially by the MMCs and their early descendants cells (stomatal lineage cells) including guard mother cells (GMCs).

Q&A

What is the difference between EPF2 and EphA2, and which antibodies should I use for my research?

EPF2 (Epidermal Patterning Factor 2) and EphA2 (Ephrin type-A receptor 2) are entirely different proteins functioning in distinct biological systems.

EPF2: A plant peptide involved in stomatal patterning on plant epidermis. EPF2 competes with Stomagen for binding to the ERECTA (ER) receptor and TMM (TOO MANY MOUTHS) co-receptor. EPF2 exhibits saturable binding to the ER ectodomain with dissociation constants in the nanomolar range .

EphA2: A receptor tyrosine kinase in mammals that binds ephrin ligands and is involved in cell positioning and morphogenesis. It's implicated in various epithelial cancers and has roles in neuronal development and repair .

For studying EPF2 in plant systems, use antibodies specifically raised against the mature EPF2 peptide. For EphA2 research in animal/human systems, numerous validated monoclonal and polyclonal antibodies are available from commercial sources with different binding properties .

How do I validate the specificity of my anti-EphA2 antibody?

Thorough validation is critical for ensuring reliable results. Implement these methodological approaches:

  • Positive and negative controls: Test antibody binding on cells with known high expression (like MiaPaCa2, 293/EphA2, or A431 cell lines) and compare with low/no expression models .

  • Receptor specificity panel: Test cross-reactivity against related Eph receptors by using transfected cells expressing EphA1, EphA3, EphA4, EphA5, and EphA7. Many commercial antibodies show some degree of cross-reactivity to EphA4 and should be avoided .

  • Receptor knockdown/knockout validation: Use siRNA or CRISPR to reduce/eliminate EphA2 expression and confirm decreased antibody signal.

  • Multiple detection methods: Validate using at least three different techniques (e.g., Western blot, immunoprecipitation, flow cytometry, and immunohistochemistry) .

  • Epitope competition: Pre-incubate with recombinant EphA2 protein to demonstrate specific blocking of antibody binding .

How can I determine if my anti-EphA2 antibody has agonistic or antagonistic properties, and which is better for my research?

Understanding the functional properties of anti-EphA2 antibodies is crucial for experimental design:

Methodological determination of antibody properties:

  • Receptor phosphorylation assay: Agonistic antibodies will induce EphA2 phosphorylation within 5-20 minutes of treatment, detectable by immunoprecipitation followed by anti-phosphotyrosine (4G10) western blotting .

  • Receptor internalization assay: Monitor antibody-induced internalization using fluorescently labeled antibodies and confocal microscopy or In Cell Analyzer quantification. Agonistic antibodies (like IgG25) promote rapid receptor endocytosis and degradation, while antagonistic antibodies (like IgG28) do not .

  • Ligand competition assay: Test if your antibody blocks binding of fluorescently labeled ephrinA1-Fc to EphA2-expressing cells. Antagonistic antibodies will reduce ligand binding in a dose-dependent manner .

Choosing between agonistic vs. antagonistic antibodies:

PropertyAgonistic AntibodiesAntagonistic Antibodies
MechanismPromote receptor activation, endocytosis and degradationBlock ligand binding
Best for studyingReceptor downregulation, signaling activationReceptor blockade, inhibition of signaling
Example effectsReduce EphA2 protein levels, increase FAK phosphorylationDecrease tumor vascularization, block ephrin-binding
Example modelsIgG25 shows strong anti-tumor effects in pancreatic cancer models IgG28 demonstrates anti-angiogenic properties

Interestingly, both approaches have shown comparable therapeutic efficacy in pancreatic xenograft models despite different mechanisms .

What are the challenges in using anti-EphA2 antibodies for studying EphA2 in primary tissues versus cell lines?

Primary tissues present unique challenges compared to cell lines:

Heterogeneous expression: Primary tissues show variable EphA2 expression levels. For example, human gastric organoids express EphA2 at levels comparable to cell lines but with patient-dependent variability . Flow cytometry analysis of fresh tissue requires careful gating strategies to account for this heterogeneity.

Accessibility issues: In 3D organoid cultures, antibody access may be limited compared to 2D cultures. Both apical and basolateral access should be tested as demonstrated in gastric organoid studies, where neither approach significantly improved detection .

Competition with endogenous ligands: Primary tissues often express endogenous ephrin ligands that may interfere with antibody binding. Pre-blocking with recombinant ephrins can help determine if this affects your results.

Growth factor effects: EGF treatment can increase EphA2 expression (approximately three-fold in AdAH cells) , potentially confounding results. Consider that organoid culture media typically contains EGF (50 ng/ml), which may alter baseline expression.

Methodology adjustments:

  • For flow cytometry: Single-cell suspensions must be carefully prepared without excessive digestion that might cleave surface EphA2

  • For immunohistochemistry: Optimize antigen retrieval methods specifically for EphA2

  • For functional studies: Consider that expression of EphA2 doesn't necessarily correlate with functional accessibility

How should I optimize co-immunoprecipitation protocols when studying EphA2 interactions with potential binding partners?

Co-immunoprecipitation (Co-IP) is valuable for studying EphA2 receptor complexes, but requires careful optimization:

Protocol optimization steps:

  • Cell stimulation conditions: Pre-incubate cells with antibodies (10 μg/mL) or ephrinA1-Fc (5 μg/mL) for 20 minutes at 4°C followed by 5 minutes at 37°C to capture transient interactions .

  • Lysis buffer selection: Use a buffer containing 1% NP-40 or Triton X-100 with phosphatase inhibitors (sodium orthovanadate, sodium fluoride) to preserve phosphorylation status.

  • Antibody selection for IP: Use antibodies targeting the EphA2 C-terminus rather than the ligand-binding domain to avoid interference with complex formation. Commercial antibodies from Santa Cruz have been successfully used (0.8 μg/sample) .

  • Bead selection: Protein G Sepharose beads (GE Healthcare) have been effectively used for EphA2 Co-IP studies .

  • Detection strategy: For detecting novel interactions, strip and reprobe membranes with antibodies against suspected binding partners after initial phosphotyrosine detection .

Validated Co-IP applications:

  • Detecting competitive binding: Co-IP can demonstrate competitive binding between different ligands. For example, increasing concentrations of Stomagen peptide can replace MEPF2 for ER-binding in plant systems .

  • Receptor-ligand interactions: Anti-EphA2 antibodies have been used to co-immunoprecipitate ephrinA1 ligand and demonstrating binding specificity .

  • Receptor phosphorylation studies: Anti-EphA2 IPs followed by phosphotyrosine blotting can quantify receptor activation after antibody or ligand treatment .

  • Negative controls: Always include isotype control antibodies (e.g., Ctrl IgG) and verify specificity by testing related receptors (e.g., FLS2 fails to immunoprecipitate Stomagen above background level) .

What are the optimal conditions for using anti-EphA2 antibodies in flow cytometry applications?

Flow cytometry is widely used for detecting cell surface EphA2 and requires specific optimization:

Sample preparation:

  • Use single-cell suspensions prepared with minimal enzymatic digestion to preserve EphA2 epitopes

  • For adherent cells, use non-enzymatic dissociation methods or very brief trypsinization

  • Keep cells at 4°C during processing to prevent receptor internalization

Staining protocol optimization:

  • Blocking: Use 1-5% BSA in PBS with 10mM HEPES (FACS buffer) to reduce non-specific binding

  • Primary antibody concentration: Titrate antibodies starting at 1 μg/mL for commercial monoclonal antibodies

  • Secondary detection: APC-conjugated or PE-conjugated anti-human Fc antibodies provide good signal-to-noise ratio

  • Positive controls: MiaPaCa2, A431, and AdAH cell lines express high levels of EphA2

  • Negative controls: Include isotype control antibodies (e.g., AB-108-C) to establish background levels

Applications in research:

  • Binding affinity determination: Calculate apparent Kd values by analyzing Mean Fluorescence Intensity (MFI) values with Sigma-Plot software after titrating antibody concentrations

  • Competition assays: For measuring ligand blocking, incubate cells with increasing antibody concentrations followed by labeled ephrinA1/Fc (30 nM)

  • Expression analysis: Compare EphA2 expression across different cell populations, as demonstrated in studies of gastric organoids

Technical considerations:

  • Since EphA2 can be rapidly internalized, avoid prolonged incubations at 37°C before analysis

  • Consider that growth factors like EGF can upregulate EphA2 expression

  • For cells with very low expression, signal amplification systems may be required

Why might I observe contradictory results when using different anti-EphA2 antibodies in functional studies?

Contradictory results with different anti-EphA2 antibodies are common and may arise from several factors:

Different binding epitopes:

  • Antibodies binding different domains of EphA2 can produce opposite effects

  • Extracellular domain-binding antibodies may be agonistic or antagonistic depending on exact epitope

  • Antibodies binding intracellular domains generally don't affect ligand binding but may interfere with downstream signaling

Functional differences between antibodies:

  • Agonistic antibodies (like IgG25) promote receptor phosphorylation and degradation

  • Antagonistic antibodies (like IgG28) block ligand binding

  • Some antibodies may have mixed effects or weak activity

Context-dependent EphA2 functions:

  • In pancreatic xenograft models, both agonistic and antagonistic antibodies showed comparable anti-tumor efficacy despite different mechanisms

  • In breast and colon tumor xenografts, agonistic antibodies failed to inhibit growth despite strong EphA2 downmodulation

  • Contradictions may reflect the complex, context-dependent role of EphA2 in different tumors

Experimental variables to control:

  • Standardize antibody concentrations (typically 1-10 μg/mL)

  • Control incubation times and temperatures

  • Verify antibody functionality before each experiment

  • Consider that cell culture conditions (serum starvation, confluency) affect baseline EphA2 activity

Conflicting results from literature:
Different studies have reported that EphA2 activation either weakly stimulates or inhibits Erk phosphorylation in MiaPaCa2 cells. These contradictions may result from differences in cell culture conditions, serum starvation protocols, and duration of ligand incubation .

What are the potential pitfalls when using anti-EphA2 antibodies for tissue staining in immunohistochemistry?

Immunohistochemical detection of EphA2 presents several challenges:

Epitope preservation issues:

  • Many anti-EphA2 antibodies recognize conformational epitopes and don't work with denatured protein

  • Formalin fixation can mask epitopes; test multiple antigen retrieval methods

  • EphA2's extracellular domain may be cleaved in some tissues, leading to false negatives with N-terminal-specific antibodies

Specificity concerns:

  • Cross-reactivity with other Eph receptors is common; validate specificity

  • High background can occur in tissues with endogenous Fc receptors; include blocking steps

  • Endogenous peroxidase activity in some tissues can cause false positives; use appropriate quenching

Interpretation challenges:

  • Heterogeneous expression within tumors may lead to sampling errors

  • Membranous versus cytoplasmic staining patterns have different biological implications

  • Secreted ephrins in the tissue may block antibody binding to EphA2

Technical recommendations:

  • Use positive control tissues with known EphA2 expression

  • Include appropriate negative controls (isotype antibodies)

  • Consider dual staining with epithelial markers to identify cell types

  • For mouse tissues, use non-mouse primary antibodies or M.O.M. kits to reduce background

  • Validate findings with multiple antibodies recognizing different EphA2 epitopes

How can anti-EphA2 antibodies be engineered for improved research and therapeutic applications?

Several engineering approaches show promise for enhancing anti-EphA2 antibodies:

Engineering strategies for research applications:

  • Bispecific antibodies: Targeting EphA2 and another receptor simultaneously could reveal synergistic signaling mechanisms

  • Fluorescently tagged antibodies: Direct conjugation with bright, photostable fluorophores improves live imaging capabilities

  • Conformation-specific antibodies: Developing antibodies that specifically recognize active versus inactive EphA2 conformations would advance signaling studies

  • Domain-specific antibodies: Creating a panel targeting distinct functional domains would help dissect complex signaling pathways

Therapeutic engineering approaches:

  • Antibody-drug conjugates (ADCs): Anti-EphA2 antibodies can deliver cytotoxic payloads specifically to EphA2-overexpressing tumors

  • Immune effector engagement: Engineering anti-EphA2 antibodies to recruit T cells (BiTEs) or NK cells could enhance anti-tumor activity

  • Tissue-penetrating modifications: Smaller formats like single-domain antibodies may improve tumor penetration

  • Combinatorial targeting: Developing antibodies that simultaneously block EphA2 and related receptors (e.g., other Eph family members)

Emerging applications:

  • Anti-EphA2 antibodies could be developed for detecting fungal infections, given EphA2's role as a receptor for fungal β-glucans in lung epithelial cells

  • Combining anti-EphA2 antibodies with other targeted therapies might overcome resistance mechanisms

  • Engineering pH-sensitive antibodies that preferentially bind in the tumor microenvironment could improve specificity

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 2025 TheBiotek. All Rights Reserved.