EGC1 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% ProClin 300; Constituents: 50% Glycerol, 0.01M Phosphate-Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
14-16 weeks (made-to-order)
Synonyms
EGC1 antibody; EXPR2 antibody; PNP-B antibody; At4g30380 antibody; F17I23_280Putative EG45-like domain containing protein 1 antibody; Plant natriuretic peptide B antibody; AtEXPR2 antibody; AtPNP-B antibody; Ath-ExpGamma-1.1 antibody
Target Names
EGC1
Uniprot No.

Target Background

Function
Potentially plays a systemic role in water and solute homeostasis.
Database Links
Subcellular Location
Secreted.

Q&A

What is the EGC1 gene and what protein does it encode?

The EGC1 gene (Entrez Gene ID: 30986144) encodes a glycoside hydrolase family 5 protein in Suhomyces tanzawaensis NRRL Y-17324. This protein-coding gene is associated with carbohydrate metabolism and has been studied in comparative genomics of biotechnologically important yeasts . The gene's mRNA accession number is XM_020212008.1, and the corresponding protein accession is XP_020061973.1. Understanding the target protein's function is essential for developing effective antibody-based detection strategies.

What are the recommended validation steps for EGC1 antibodies before using them in experiments?

Comprehensive validation of EGC1 antibodies requires a multi-step approach:

  • Context-specific validation: Validate the antibody specifically for your intended research application rather than relying solely on manufacturer claims .

  • Western blotting validation: Confirm specificity by checking that the antibody detects a band of the expected molecular weight (based on the EGC1 protein sequence).

  • Negative controls: Test the antibody in samples where EGC1 is known to be absent or knocked down.

  • Cross-reactivity assessment: Evaluate potential cross-reactivity with similar glycoside hydrolase family proteins.

  • Application-specific validation: Perform validation tests specific to your intended application (immunohistochemistry, flow cytometry, etc.) .

Proper validation minimizes experimental artifacts and ensures the reliability of your research findings.

What expression vectors are recommended for generating recombinant EGC1 proteins for antibody production?

Based on available information about EGC1 clones, the following expression systems are recommended:

Vector SystemFeaturesApplications
pcDNA3.1-C-(k)DYKMammalian expression, C-terminal tagCell-based assays, protein production for immunization
Custom vectors with TEV cleavage sitesRemovable tags for native protein structureAdvanced structural studies, high-purity antigen preparation

The selection of an appropriate vector depends on your specific requirements for antigen preparation. The EGC1 ORF is 1452bp in length and can be cloned into a variety of expression vectors . When preparing antigens for antibody production, consider the impact of tags on protein folding and epitope accessibility.

What are the key controls to include when testing a new EGC1 antibody in Western blot applications?

When validating an EGC1 antibody for Western blot applications, the following controls are essential:

  • Positive control: Lysate from cells overexpressing EGC1 or recombinant EGC1 protein

  • Negative control: Lysate from cells where EGC1 is known to be absent or knocked down

  • Loading control: Detection of a housekeeping protein to ensure equal loading

  • Isotype control: A non-specific antibody of the same isotype to identify non-specific binding

  • Blocking peptide control: Pre-incubation of the antibody with the immunizing peptide should abolish specific signal

These controls help distinguish specific from non-specific signals and validate the antibody's performance in your experimental system, following standard practice for antibody validation in molecular research .

How can computational modeling be used to predict and enhance EGC1 antibody specificity?

Advanced computational modeling can significantly enhance EGC1 antibody specificity through several approaches:

  • Binding mode identification: Computational models can identify distinct binding modes associated with particular ligands, helping disentangle modes associated with chemically similar targets .

  • Custom specificity profiles: Biophysics-informed modeling combined with selection experiment data can be used to design antibodies with customized specificity profiles for EGC1, either with high specificity for a particular epitope or with cross-specificity for multiple epitopes .

  • Energy function optimization: For generating new EGC1-specific antibody sequences, optimization of energy functions associated with each binding mode can be performed. For enhanced specificity, minimize energy functions associated with desired epitopes while maximizing those for undesired targets .

  • Integration with experimental data: These computational approaches are most effective when trained on data from phage display experiments, allowing the model to learn from selection patterns observed experimentally .

This computational strategy extends beyond what's experimentally feasible, allowing researchers to design EGC1 antibodies with binding properties tailored to specific research needs.

What approaches are recommended to resolve longitudinal variations in antibody responses to EGC1 protein?

Longitudinal studies of antibody responses often reveal temporal variations that can complicate data interpretation. Based on similar studies with other protein targets, the following methodological approach is recommended:

  • Sequential sampling protocol: Collect 6-13 serum samples over 18-42 months to capture the full range of temporal variation .

  • Multiple epitope tracking: Monitor antibody responses to at least 5-7 distinct epitopes of the EGC1 protein, as responses to individual epitopes may vary independently over time .

  • Quantitative analysis of response patterns: Some epitopes will show stable responses (detectable in 80-100% of samples), while others will show variable detection (33-83% of samples), providing insight into antigenic response diversity .

  • Marker epitope identification: Identify specific epitopes that consistently distinguish between study groups (e.g., disease vs. control) to serve as reliable biomarkers .

This approach accounts for the natural biological variability in antibody responses while enabling identification of consistent markers for research applications.

How can EGC1 antibodies be developed for potential therapeutic applications through ADCC mechanisms?

While not directly studied for EGC1, the development of antibodies with Antibody-Dependent Cellular Cytotoxicity (ADCC) potential follows these research steps:

  • IgG1 isotype selection: Design antibodies of the IgG1 isotype, which is particularly effective at engaging FcγR on effector cells to trigger ADCC .

  • Effector function assessment: Test the antibody's ability to induce ADCC using peripheral blood mononuclear cells (PBMCs) at various effector:target ratios (20:1 to 80:1 is recommended) .

  • Correlation analysis: Establish the relationship between target expression levels and ADCC efficiency to predict therapeutic potential .

  • In vivo validation: Confirm ADCC activity in humanized mouse models before proceeding to clinical applications .

  • Patient-derived cell testing: Validate that PBMCs from relevant patient populations can effectively perform ADCC when combined with the therapeutic antibody .

This structured approach moves from basic in vitro assessment to clinically relevant models, providing a comprehensive evaluation of therapeutic potential.

What methods are optimal for identifying conformational epitopes of the EGC1 protein for antibody generation?

Identification of conformational epitopes in the EGC1 glycoside hydrolase requires specialized approaches:

  • X-ray crystallography of antibody-antigen complexes: Provides atomic-level resolution of the interaction interface but requires crystallization of the complex.

  • Hydrogen-deuterium exchange mass spectrometry (HDX-MS): Identifies regions of the protein that are protected from exchange upon antibody binding, indicating epitope locations.

  • Cryo-electron microscopy (Cryo-EM): Increasingly used to map conformational epitopes for larger protein complexes without crystallization.

  • Alanine scanning mutagenesis: Systematic replacement of surface residues with alanine to identify critical binding residues.

  • Computational epitope prediction: Utilize structure-based algorithms that consider surface accessibility, hydrophobicity, and electrostatic properties to predict likely epitope regions.

These complementary approaches provide a comprehensive map of conformational epitopes, enabling the design of antibodies targeting specific functional domains of the EGC1 protein.

What strategies can resolve specificity issues when working with EGC1 antibodies in complex biological samples?

Enhancing specificity when working with EGC1 antibodies in complex samples requires a multi-faceted approach:

  • Pre-adsorption protocol: Pre-incubate antibodies with related proteins to remove cross-reactive antibodies before use in experiments.

  • Optimization of blocking conditions: Test different blocking agents (BSA, normal serum, casein) at various concentrations to minimize non-specific binding.

  • Titration experiments: Determine the optimal antibody concentration that maximizes specific signal while minimizing background.

  • Two-antibody validation approach: Use two antibodies recognizing different epitopes of EGC1 to confirm specificity; signals that co-localize are more likely to be genuine .

  • Competitive binding assays: Include excess unlabeled antibody or antigen to demonstrate signal reduction, confirming specificity.

These methods collectively enhance confidence in the specificity of observed signals, particularly important when studying EGC1 in heterogeneous biological samples where related glycoside hydrolases may be present.

How can researchers troubleshoot inconsistent results when using EGC1 antibodies across different experimental platforms?

Inconsistent results across platforms often stem from platform-specific factors. The following troubleshooting workflow is recommended:

  • Platform-specific validation: Validate the EGC1 antibody separately for each experimental platform (Western blot, IHC, ELISA, flow cytometry) .

  • Buffer optimization: Test different buffer systems for each platform, as buffer components can significantly affect antibody-antigen interactions.

  • Sample preparation assessment: Evaluate how different sample preparation methods affect epitope accessibility:

    • For Western blotting: Compare different lysis buffers and reducing/non-reducing conditions

    • For IHC/IF: Test multiple fixation protocols and antigen retrieval methods

    • For flow cytometry: Compare different permeabilization techniques

  • Cross-platform concordance analysis: When results differ between platforms, determine which platform more reliably reflects the biological reality by correlating with functional assays or orthogonal detection methods.

  • Lot-to-lot validation: Test new antibody lots against reference samples with established results to ensure consistent performance over time.

This systematic approach identifies the source of inconsistency and establishes reliable protocols for each experimental platform.

What are the recommended approaches for quantifying EGC1 protein levels using antibody-based techniques?

Accurate quantification of EGC1 protein levels requires careful selection of methods based on research needs:

MethodSensitivityDynamic RangeSample RequirementsKey Considerations
Quantitative Western BlotModerate10-fold10-50 μg total proteinRequires validated linear range and standard curve
ELISAHigh1000-fold1-5 μg total proteinSandwich format recommended for complex samples
ImmunofluorescenceModerate20-foldFixed cells/tissuesBest for relative quantification and localization
Flow CytometryHigh10,000-foldSingle cell suspensionsIdeal for population analysis and heterogeneity assessment

For absolute quantification, include a standard curve using recombinant EGC1 protein of known concentration. For relative quantification, normalize to appropriate housekeeping proteins or total protein staining. The choice of method depends on the required sensitivity, sample availability, and whether spatial information is needed.

How can researchers generate monoclonal antibodies with high specificity for EGC1 using phage display technology?

Generating high-specificity monoclonal antibodies against EGC1 using phage display involves these methodological steps:

  • Antigen preparation: Express and purify the EGC1 protein (glycoside hydrolase family 5) from Suhomyces tanzawaensis, ensuring proper folding for presentation of native epitopes .

  • Selection strategy design: Implement a multi-round selection process with increasing stringency:

    • Early rounds: High antigen concentration to capture diverse binders

    • Middle rounds: Decreased antigen concentration to select for higher affinity

    • Final rounds: Include competitors or related proteins for negative selection to enhance specificity

  • Computational analysis of selection outcomes: Apply biophysics-informed modeling to identify distinct binding modes associated with specific epitopes of interest .

  • Specificity engineering: Optimize antibody sequences computationally to enhance specificity by minimizing energy functions associated with desired epitopes while maximizing those for undesired targets .

  • Validation of selected clones: Test monoclonal antibodies for specificity using multiple techniques including Western blotting, immunoprecipitation, and testing against knockout or knockdown samples .

This integrated experimental and computational approach leverages the advantages of phage display while addressing its limitations through sophisticated selection design and downstream analysis.

What experimental approaches best characterize the affinity and kinetics of EGC1 antibody-antigen interactions?

Comprehensive characterization of EGC1 antibody-antigen interactions requires multiple biophysical techniques:

  • Surface Plasmon Resonance (SPR): Provides real-time analysis of binding kinetics, determining:

    • Association rate constant (kon)

    • Dissociation rate constant (koff)

    • Equilibrium dissociation constant (KD = koff/kon)

  • Bio-Layer Interferometry (BLI): Alternative to SPR with advantages for crude samples and simpler setup.

  • Isothermal Titration Calorimetry (ITC): Measures thermodynamic parameters including:

    • Binding enthalpy (ΔH)

    • Entropy changes (ΔS)

    • Stoichiometry of interaction

  • Microscale Thermophoresis (MST): Useful for difficult targets with minimal sample consumption.

  • Enzyme-Linked Immunosorbent Assay (ELISA): For high-throughput screening and approximate KD determination.

Select methods based on available instrumentation, sample properties, and required parameters. For the most complete characterization, combine multiple techniques to verify consistency of binding parameters across different experimental conditions.

How can researchers determine if an EGC1 antibody recognizes a linear or conformational epitope?

Distinguishing between linear and conformational epitopes requires a systematic analytical approach:

  • Denaturation test: Compare antibody binding to native versus denatured EGC1 protein:

    • Linear epitope: Similar binding to both forms

    • Conformational epitope: Significantly reduced binding to denatured protein

  • Peptide mapping: Screen overlapping synthetic peptides spanning the EGC1 sequence:

    • Linear epitope: Strong binding to specific peptide(s)

    • Conformational epitope: Weak or no binding to individual peptides

  • Proteolytic fragmentation: Digest the EGC1 protein with proteases and identify fragments that retain antibody binding.

  • Glycosylation impact assessment: Compare binding to glycosylated versus deglycosylated EGC1, as glycans may be part of conformational epitopes.

  • Mutational analysis: Create point mutations in potential epitope regions and assess their impact on antibody binding.

This multi-method approach provides comprehensive evidence for epitope classification, guiding application-specific optimizations and interpretation of experimental results.

How can CRISPR/Cas9 gene editing be used to validate EGC1 antibody specificity?

CRISPR/Cas9 technology offers powerful approaches for antibody validation:

  • Gene knockout validation: Generate complete EGC1 knockout cell lines or model organisms to create true negative controls for antibody testing . The absence of signal in knockout samples provides definitive evidence of antibody specificity.

  • Epitope modification: Use CRISPR to introduce specific mutations in the epitope region recognized by the antibody while maintaining protein expression. This approach can precisely map the binding site and confirm specificity.

  • Tagged endogenous protein: Insert epitope tags into the endogenous EGC1 gene locus, allowing parallel detection with both the EGC1 antibody and tag-specific antibodies for cross-validation.

  • Isogenic cell line panels: Create a series of cell lines with varying EGC1 expression levels to establish the quantitative relationship between protein levels and antibody signal.

  • Cross-reactivity assessment: Knockout related glycoside hydrolase family genes to assess potential cross-reactivity with homologous proteins.

This genetic validation approach provides the highest standard for antibody validation, establishing unambiguous controls for specificity assessment.

What are the emerging approaches for developing antibodies that can distinguish between closely related glycoside hydrolase family members?

Developing highly specific antibodies that can distinguish between closely related glycoside hydrolase family members requires cutting-edge approaches:

  • Structural biology-guided epitope selection: Use structural comparisons between EGC1 and related glycoside hydrolases to identify unique surface epitopes suitable for specific antibody development.

  • Negative selection strategies: Implement rigorous phage display selection protocols that include counter-selection against related family members to eliminate cross-reactive antibodies .

  • Computational specificity engineering: Apply machine learning algorithms trained on antibody-antigen interaction data to predict sequence modifications that will enhance specificity for EGC1 over related proteins .

  • Single B-cell sorting techniques: Isolate B cells from immunized animals using both positive selection for EGC1 binding and negative selection against related proteins.

  • Affinity maturation optimization: Conduct in vitro evolution focusing not only on improved affinity but also on increased specificity through directed evolution approaches.

These advanced approaches can generate antibodies with the discrimination capacity required for research applications involving closely related protein family members.

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.