At4g22212 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 week lead time (made-to-order)
Synonyms
At4g22212 antibody; T10I14.6 antibody; Defensin-like protein 98 antibody
Target Names
At4g22212
Uniprot No.

Target Background

Database Links

KEGG: ath:AT4G22212

STRING: 3702.AT4G22212.1

UniGene: At.19889

Protein Families
DEFL family
Subcellular Location
Secreted.

Q&A

What is the At4g22212 gene and why are antibodies against it important in plant research?

At4g22212 is a gene locus in Arabidopsis thaliana that encodes specific proteins relevant to plant developmental biology. Antibodies against this target are valuable tools for investigating protein expression, localization, and interaction networks. Unlike traditional genetic approaches that examine the gene itself, antibodies enable direct analysis of protein products, providing crucial insights into post-translational modifications and protein dynamics that cannot be inferred from genomic or transcriptomic data alone .

When designing experiments, researchers should consider that antibody-based detection complements other approaches like fluorescent protein tagging, offering advantages in detecting endogenous protein without potential artifacts from overexpression systems. The specificity of the antibody is particularly critical when studying protein families with high sequence similarity, making proper validation essential.

How should I validate an At4g22212 antibody before incorporating it into my experimental workflow?

A systematic antibody validation approach should be implemented through the following steps:

  • Knockout controls: Generate CRISPR/Cas9 knockout lines in appropriate cell types that normally express the At4g22212 protein. Compare antibody reactivity between wild-type and knockout samples as the gold standard for specificity verification .

  • Immunoblot analysis: Test the antibody on gradient polyacrylamide gels using both wild-type and knockout samples. A specific antibody should produce bands of the expected molecular weight in wild-type samples and no bands in knockout controls .

  • Cross-reactivity assessment: Test the antibody against related plant proteins to ensure it doesn't recognize homologous proteins.

  • Application-specific validation: Validate the antibody separately for each application (western blot, immunofluorescence, immunoprecipitation) as performance can vary dramatically between applications .

A proper validation pipeline is critical as many commercially available antibodies do not recognize their intended targets, leading to misleading results and irreproducible research .

What control samples are essential when evaluating At4g22212 antibody specificity?

The following controls are essential for rigorous antibody validation:

  • Genetic knockout/knockdown: CRISPR/Cas9-generated knockouts provide the most stringent control. For At4g22212, T-DNA insertion lines or RNAi knockdown plants can serve as alternatives if CRISPR editing is challenging .

  • Overexpression samples: Tissues or cells overexpressing the At4g22212 protein provide positive controls to confirm antibody detection capability.

  • Recombinant protein: Purified recombinant At4g22212 protein serves as a positive control for antibody specificity in biochemical assays.

  • Related protein controls: Testing against related plant proteins helps evaluate potential cross-reactivity with homologous sequences.

  • Tissue-specific expression: If At4g22212 is known to have tissue-specific expression patterns, tissues known to lack expression serve as natural negative controls .

What are the most reliable techniques for characterizing At4g22212 antibody binding properties?

Several complementary techniques should be employed to thoroughly characterize antibody binding properties:

  • Enzyme-Linked Immunosorbent Assay (ELISA): Provides quantitative measurement of antibody-antigen binding and helps determine specificity, affinity, and cross-reactivity .

  • Surface Plasmon Resonance (SPR): Offers real-time, label-free measurement of binding kinetics and affinity. SPR can determine the equilibrium dissociation constant, which is crucial for understanding the strength of antibody-antigen interactions .

  • Immunoblotting with quantitative detection: Quantitative western blotting using systems like LI-COR Odyssey provides precise measurement of antibody binding to native proteins in complex mixtures .

  • Epitope mapping: Peptide arrays or hydrogen-deuterium exchange mass spectrometry can identify the specific epitopes recognized by the antibody, which is essential for understanding potential cross-reactivity .

These techniques should be used in combination as they provide complementary information about different aspects of antibody binding characteristics .

How can I optimize western blot protocols specifically for At4g22212 antibody detection?

Optimizing western blot protocols for At4g22212 antibody requires systematic adjustment of several parameters:

  • Sample preparation:

    • Use extraction buffers containing protease inhibitors to prevent degradation

    • Consider plant-specific extraction protocols to deal with interfering compounds

    • For membrane-associated proteins, include appropriate detergents (0.1-1% Triton X-100 or NP-40)

  • Gel separation:

    • Use 5-16% gradient gels for optimal separation, especially if detecting different isoforms

    • Load appropriate protein amounts (typically 10-30 μg per lane)

  • Transfer conditions:

    • For hydrophobic proteins, consider using PVDF instead of nitrocellulose membranes

    • Optimize transfer time and voltage based on protein size

  • Blocking and antibody incubation:

    • Test different blocking agents (5% BSA often works better than milk for phospho-specific antibodies)

    • Incubate primary antibody overnight at 4°C in 5% BSA in TBST

    • Determine optimal antibody concentration through titration experiments

  • Detection method:

    • For quantitative analysis, use fluorescent secondary antibodies and imaging systems like LI-COR

    • For highest sensitivity, consider enhanced chemiluminescence systems

Quantitative western blots should include total protein staining (like REVERT) for normalization to account for loading variations .

What approaches should I use to troubleshoot inconsistent results with At4g22212 antibodies?

When encountering inconsistent results, implement this systematic troubleshooting approach:

  • Antibody validation reassessment:

    • Reconfirm antibody specificity using knockout controls

    • Verify antibody stability and storage conditions (aliquot and store at -20°C or -80°C)

    • Test new antibody lot against previous lots to detect lot-to-lot variations

  • Sample preparation evaluation:

    • Ensure complete protein extraction with appropriate buffers

    • Check for protein degradation using total protein stains

    • Verify sample handling conditions (avoid freeze-thaw cycles)

  • Protocol optimization:

    • Systematically vary antibody concentration, incubation time, and temperature

    • Test different blocking agents to reduce background

    • Optimize washing steps to improve signal-to-noise ratio

  • Technical controls:

    • Include positive and negative controls in each experiment

    • Use loading controls appropriate for your experimental conditions

    • Consider spike-in experiments with recombinant protein

  • Cross-laboratory validation:

    • If possible, have another laboratory reproduce key experiments

    • Compare results with different detection methods (e.g., mass spectrometry)

Maintain detailed records of all protocol variations to identify the source of inconsistency .

How can I develop a reliable immunofluorescence protocol for At4g22212 localization studies?

Developing a robust immunofluorescence protocol requires careful optimization:

  • Fixation method optimization:

    • Compare paraformaldehyde (4%, 10 min) versus methanol fixation (-20°C, 10 min)

    • For plant tissues, evaluate the need for cell wall digestion with enzymes

  • Permeabilization conditions:

    • Test different detergent concentrations (0.1-0.3% Triton X-100) and times

    • For plant tissues with cell walls, longer permeabilization may be required

  • Antibody validation strategy:

    • Implement mosaic culture techniques combining wild-type and knockout cells labeled with different fluorescent markers

    • This approach allows direct comparison of staining in expressing versus non-expressing cells

  • Signal amplification considerations:

    • For low abundance proteins, evaluate tyramide signal amplification

    • Compare direct versus indirect immunofluorescence approaches

  • Controls and counterstaining:

    • Include no-primary-antibody controls

    • Use appropriate markers for subcellular compartments

    • Consider dual staining with known interacting partners

  • Imaging parameters:

    • Use confocal microscopy with appropriate controls for bleed-through

    • Maintain consistent exposure settings between samples

    • Consider super-resolution techniques for detailed localization studies

This methodology has been successfully applied to determine the localization of proteins like C9ORF72 to phagosomes/lysosomes, correcting previous misreported localizations .

What strategies exist for generating custom At4g22212 antibodies with improved specificity?

Developing custom antibodies with enhanced specificity involves several strategic approaches:

  • Epitope selection optimization:

    • Conduct bioinformatic analysis to identify unique regions with low homology to related proteins

    • Target regions that are surface-exposed in the native protein

    • Consider using multiple epitopes to develop a panel of antibodies

  • Antigen design strategies:

    • Use recombinant protein fragments rather than synthetic peptides for improved recognition of native protein

    • For difficult targets, consider structural vaccinology approaches using computational epitope design

    • Ensure proper protein folding in recombinant antigens

  • Screening methodology:

    • Implement multi-stage screening against both the immunogen and full-length protein

    • Use knockout samples as negative controls early in screening

    • Include cross-reactivity testing against related family members

  • Production and purification considerations:

    • For monoclonal antibodies, select optimal clones using multiple applications

    • For recombinant antibodies, optimize codon usage for expression system

    • Implement affinity purification using the target antigen

  • Validation in target systems:

    • Test antibodies in the relevant plant tissues or experimental systems

    • Validate across multiple techniques (western blot, IF, IP) to ensure versatility

This approach has been successful in generating antibodies like A4, which shows 600-fold higher affinity for mutant versus wild-type forms of the target protein .

How can I apply At4g22212 antibodies in protein interaction and complex studies?

Advanced applications for studying protein interactions require specialized approaches:

  • Co-immunoprecipitation optimization:

    • Evaluate different lysis conditions to maintain protein complexes

    • Test various antibody coupling strategies to solid supports

    • Compare native versus crosslinking approaches

  • Proximity labeling applications:

    • Consider antibody-guided proximity labeling techniques

    • Use split enzyme complementation approaches for detecting protein interactions

    • Implement BioID or APEX2 systems in combination with antibodies

  • Multiplex co-localization studies:

    • Develop multi-color immunofluorescence protocols

    • Implement spectral unmixing for closely overlapping fluorophores

    • Consider advanced microscopy techniques (FRET, FLIM) for direct interaction studies

  • Antibody-based proteomics:

    • Use antibodies for immunoaffinity enrichment prior to mass spectrometry

    • Implement crosslinking mass spectrometry to capture transient interactions

    • Consider reverse-phase protein arrays for high-throughput studies

  • Live-cell applications:

    • Explore cell-permeable antibody formats (nanobodies, penetratin-conjugated Fabs)

    • Implement optogenetic approaches combined with antibody detection

    • Consider intrabodies for tracking protein dynamics

These techniques have been successfully applied to characterize signaling pathways like the AGR2-C4.4A pathway in cancer research and could be adapted for plant protein studies.

What are the most common causes of false positive or false negative results when using At4g22212 antibodies?

Understanding common pitfalls is essential for reliable experimental outcomes:

Issue TypeCommon CausesPrevention Strategies
False PositivesCross-reactivity with homologous proteinsUse knockout controls; epitope mapping
Non-specific binding to abundant proteinsOptimize blocking; include competitive blocking peptides
Secondary antibody cross-reactivityInclude secondary-only controls
Sample contaminationMaintain strict workflow separation
False NegativesEpitope masking due to protein modificationsUse multiple antibodies targeting different regions
Protein degradation during extractionOptimize extraction buffers; use protease inhibitors
Insufficient sensitivityConsider signal amplification; increase exposure time
Improper sample preparationValidate extraction protocols for target protein

Systematic validation using knockout controls remains the gold standard for distinguishing true from false signals. Without such controls, even highly published work can perpetuate incorrect information about protein localization and function .

How should I assess lot-to-lot variability when working with commercial At4g22212 antibodies?

Lot-to-lot variability is a significant concern requiring systematic evaluation:

  • Reference standard establishment:

    • Create a laboratory reference standard from a well-characterized antibody lot

    • Store multiple aliquots of this standard for long-term comparison

  • Comparative testing protocol:

    • Run side-by-side testing of new lots against reference standard

    • Assess performance across multiple applications (western blot, IF, IP)

    • Quantify signal-to-noise ratios and detection limits

  • Quantitative metrics:

    • Determine EC50 values in ELISA or SPR experiments

    • Compare epitope binding profiles

    • Assess cross-reactivity patterns systematically

  • Documentation practices:

    • Maintain detailed records of lot numbers and performance characteristics

    • Record images of standardized experiments for future comparison

    • Implement a laboratory information management system for tracking

  • Alternative considerations:

    • For critical experiments, purchase sufficient antibody from a single lot

    • Consider generating recombinant antibodies for long-term reproducibility

    • Develop fallback validation methods not reliant on antibodies

Establishing these quality control measures is essential for maintaining experimental consistency over time and ensuring reproducibility of research findings.

What mass spectrometry approaches can complement At4g22212 antibody-based detection methods?

Mass spectrometry offers powerful complementary approaches to antibody-based detection:

  • Immunoprecipitation-Mass Spectrometry (IP-MS):

    • Use validated At4g22212 antibodies for target enrichment

    • Apply LC-MS/MS for protein identification and post-translational modification analysis

    • Implement quantitative approaches (SILAC, TMT, LFQ) for comparative studies

  • Parallel Reaction Monitoring (PRM):

    • Develop specific peptide signatures for At4g22212 protein

    • Quantify protein abundance with high specificity

    • Compare results with antibody-based quantification for validation

  • Targeted proteomics approaches:

    • Design Multiple Reaction Monitoring (MRM) assays for At4g22212-specific peptides

    • Achieve absolute quantification using isotopically labeled standards

    • Monitor multiple protein forms simultaneously

  • Structural characterization:

    • Apply hydrogen-deuterium exchange mass spectrometry for epitope mapping

    • Use native MS to study protein complexes

    • Implement crosslinking MS to identify interaction interfaces

  • Post-translational modification mapping:

    • Identify specific modification sites on At4g22212 protein

    • Develop modification-specific antibodies based on MS findings

    • Create comprehensive PTM maps to understand protein regulation

Mass spectrometry provides orthogonal validation of antibody specificity while delivering additional molecular insights not accessible through antibody-based methods alone .

How can I develop quantitative assays for measuring At4g22212 protein levels in plant tissues?

Developing quantitative assays requires careful consideration of several factors:

  • Quantitative western blot development:

    • Implement fluorescent detection systems (e.g., LI-COR Odyssey)

    • Use total protein normalization rather than single housekeeping proteins

    • Establish linear dynamic range and detection limits

  • ELISA optimization:

    • Develop sandwich ELISA using two antibodies recognizing different epitopes

    • Generate standard curves using recombinant protein

    • Validate extraction protocols to ensure complete protein recovery

  • Capillary electrophoresis-based approaches:

    • Implement automated capillary western platforms for higher throughput

    • Optimize antibody concentrations for maximum sensitivity

    • Establish internal controls for normalization

  • Single-cell quantification methods:

    • Apply quantitative immunofluorescence with appropriate controls

    • Develop flow cytometry protocols for plant protoplasts

    • Consider mass cytometry (CyTOF) for multiplexed protein quantification

  • Absolute quantification strategies:

    • Develop isotope-dilution mass spectrometry methods

    • Use recombinant isotope-labeled standards

    • Compare antibody-based and MS-based quantification

Rigorous method validation includes assessment of precision, accuracy, specificity, recovery, and matrix effects across different tissue types and experimental conditions .

How might novel antibody formats enhance At4g22212 research beyond traditional applications?

Emerging antibody technologies offer new research possibilities:

  • Single-domain antibodies (nanobodies):

    • Smaller size enables access to sterically hindered epitopes

    • Superior performance in intracellular applications

    • Potential for direct fusion to fluorescent proteins for live imaging

  • Bispecific antibodies:

    • Simultaneously target At4g22212 and interaction partners

    • Enable super-resolution microscopy through DNA-PAINT approaches

    • Create molecular bridges for proximity studies

  • Antibody fragments and derivatives:

    • Fab and scFv formats for improved tissue penetration

    • Site-specific conjugation chemistries for precise labeling

    • Recombinant production for consistent performance

  • Synthetic binding proteins:

    • Designed ankyrin repeat proteins (DARPins)

    • Monobodies based on fibronectin domains

    • Aptamer alternatives to traditional antibodies

  • Antibody-enzyme fusions:

    • Peroxidase-antibody fusions for proximity labeling

    • CRISPR-based antibody recruiting systems

    • Antibody-directed enzyme prodrug therapy approaches

These technologies expand the toolbox beyond traditional applications, enabling new experimental approaches for studying protein function, interactions, and dynamics in plant systems.

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.