At3g33187 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Composition: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
At3g33187 antibody; T14A11 antibody; Putative defensin-like protein 315 antibody
Target Names
At3g33187
Uniprot No.

Q&A

What is the optimal expression system for generating recombinant At3g33187 protein for antibody production?

Eukaryotic expression systems are highly recommended for generating recombinant proteins that will be used as antigens for antibody production, especially for heavily glycosylated proteins. Evidence suggests that using proteins expressed in eukaryotic cells as antigens is a practical way to generate monoclonal antibodies with robust affinity and specificity . For plant proteins like At3g33187, which may contain post-translational modifications, mammalian cell lines (HEK293 or CHO cells) or insect cell expression systems can preserve native protein structure and modifications. This approach typically requires only 1 cell fusion and 2 cyclic sub-cloning steps before obtaining antibodies with satisfactory performance .

What are the advantages of monoclonal versus polyclonal antibodies for At3g33187 detection?

Comparison of Antibody Types for At3g33187 Detection:

CharacteristicMonoclonal AntibodiesPolyclonal Antibodies
SpecificityHigher; recognizes single epitopeLower; recognizes multiple epitopes
Batch-to-batch consistencyExcellentVariable
Production complexityMore complex; requires hybridoma technologyLess complex; uses immunized animals
Detection sensitivityMay be lower for native proteinOften higher due to multiple epitope binding
Applications versatilityMay be limited to specific applicationsUsually works across multiple applications
CostHigherLower

Monoclonal antibodies offer superior specificity and consistency, which is critical for reproducible results in research applications. For highly specific detection of At3g33187, a monoclonal approach similar to the rabbit monoclonal antibody development described for APOBEC3B would be advisable . This approach enables effective use across multiple applications including ELISA, immunoblot, immunofluorescence microscopy, and immunohistochemistry.

How can I validate the specificity of an At3g33187 antibody?

Thorough validation of antibody specificity is essential before conducting experimental studies. A multi-method validation approach should include:

  • Western blot analysis comparing wild-type tissues/cells with At3g33187 knockout or knockdown samples

  • Immunoprecipitation followed by mass spectrometry identification

  • Immunohistochemistry or immunofluorescence comparing expression patterns with known localization data

  • ELISA testing against recombinant At3g33187 protein and closely related proteins

  • Pre-absorption tests using purified antigen to demonstrate signal reduction

For optimal validation, include positive and negative controls in each assay. When testing against recombinant proteins, it's crucial to ensure they contain proper post-translational modifications that may be present in the native protein .

How can natural antibody principles be applied to enhance At3g33187 antibody performance in complex biological samples?

Recent advances in understanding natural antibodies (nAbs) provide insights for antibody engineering. Research on natural antibodies to polysaccharide capsules demonstrates how structural recognition of specific moieties, such as β1-6-linked galactose branches, enables high-specificity binding . For At3g33187 antibodies, identifying conserved structural motifs or unique glycosylation patterns could enhance binding specificity.

Applying principles from nAb research could involve:

  • Mapping epitope structures of the At3g33187 protein

  • Engineering antibody binding domains to recognize specific structural elements

  • Incorporating complement-activating regions if the antibody is intended for functional studies

  • Optimizing antibody isotype selection based on intended application (similar to how nAbs enable Kupffer cells to capture bacteria through complement receptors)

This approach may be particularly valuable for detecting native At3g33187 in complex tissue samples where preserving physiological context is important.

What strategies can overcome cross-reactivity issues with At3g33187 antibodies in plant tissue samples?

Cross-reactivity represents a significant challenge when working with antibodies against plant proteins. To minimize this issue:

  • Epitope selection strategy: Choose unique epitopes from At3g33187 that have minimal homology with other plant proteins. Computational analysis of protein sequences can identify regions of low conservation.

  • Absorption protocols: Develop pre-absorption protocols using lysates from tissues that lack At3g33187 but contain potentially cross-reactive proteins.

  • Multi-antibody verification approach: Use multiple antibodies targeting different epitopes of At3g33187 and verify that they produce consistent results.

  • Knockout/knockdown controls: Always include tissues from At3g33187 knockout or knockdown plants as negative controls.

  • Selective expression testing: Compare antibody performance across tissues with known differential expression patterns of At3g33187.

These approaches have proven effective in addressing similar challenges with other plant protein antibodies, including those for heavily glycosylated proteins like pectins where novel rat monoclonal antibodies (LM18, LM19, LM20) demonstrated high specificity .

How can At3g33187 antibodies be optimized for detecting protein-protein interactions in plant cellular contexts?

Optimizing antibodies for protein-protein interaction studies requires specific modifications and validation steps:

  • Conjugation optimization: Carefully select conjugation methods that minimize interference with epitope recognition. Compare alkaline phosphatase and peroxidase conjugations to determine which preserves binding capacity better .

  • Antibody fragmentation: Consider using Fab fragments rather than whole IgG to reduce steric hindrance in crowded protein complexes.

  • Co-immunoprecipitation validation: Systematically test different buffer conditions, detergents, and salt concentrations to preserve protein-protein interactions while maintaining antibody specificity.

  • Proximity ligation assay (PLA) calibration: For in situ detection of protein interactions, calibrate PLA protocols specifically for plant tissues, which often require specialized cell wall digestion steps.

  • Cross-linking optimization: Develop optimized formaldehyde or DSP (dithiobis(succinimidyl propionate)) cross-linking protocols specific to the cellular compartment where At3g33187 is localized.

A validation matrix comparing different approaches should be created to identify the optimal combination of techniques for specific experimental questions.

What is the most effective immunization protocol for generating high-affinity At3g33187 antibodies?

Based on successful approaches with other challenging proteins, an optimized immunization protocol would include:

Recommended Immunization Protocol for At3g33187 Antibody Generation:

PhaseProcedureTimingNotes
Antigen preparationExpress full-length At3g33187 in eukaryotic cellsWeeks 1-3Ensure proper folding and post-translational modifications
Primary immunization50-100 μg antigen with complete Freund's adjuvantDay 0Subcutaneous injection
First boost50 μg antigen with incomplete Freund's adjuvantDay 21Multiple injection sites
Second boost50 μg antigen with incomplete Freund's adjuvantDay 42Multiple injection sites
Test bleedCollect serum sample for titer testingDay 52ELISA against recombinant protein
Final boost50 μg antigen in PBSDay 56Intravenous injection
Hybridoma generationCell fusion and selectionDay 60Using optimized PEG-based fusion protocol
ScreeningELISA, Western blot against native and recombinant proteinDays 67-90Include knockout/negative controls
SubcloningLimiting dilution of positive hybridomasWeeks 13-20Two rounds of subcloning

This approach would be similar to the process described for generating antibodies against heavily glycosylated proteins, which required just one cell fusion and two cyclic sub-cloning steps to obtain antibodies with satisfactory performance .

What purification methods are most effective for isolating At3g33187 antibodies while maintaining their activity?

The choice of purification method significantly impacts antibody performance. For At3g33187 antibodies:

  • Protein A/G affinity chromatography: Optimal for IgG isotypes, but buffer conditions must be optimized to prevent activity loss. Use gentle elution with 0.1 M glycine-HCl (pH 2.7) followed by immediate neutralization.

  • Antigen-specific affinity chromatography: Creates highest specificity antibodies but may reduce yield. Coupling recombinant At3g33187 to activated sepharose allows selective purification of only target-specific antibodies.

  • Size exclusion chromatography: Useful as a polishing step to remove aggregates that can cause high background in imaging applications.

  • Ion exchange chromatography: Provides additional purification based on antibody charge characteristics, which can separate antibody subpopulations with different binding properties.

For maintaining activity, all buffers should contain stabilizers (typically 0.1% BSA) and be kept at physiological pH except during specific elution steps. Similar approaches have been successfully used for purifying antibodies against other plant proteins .

How should At3g33187 antibody concentration be optimized for different detection methods?

Optimal antibody concentration varies significantly between applications and must be systematically determined:

Recommended Titration Ranges for At3g33187 Antibody Applications:

ApplicationStarting Dilution RangeOptimization MethodKey Considerations
Western blot1:500 to 1:5,000Serial dilutionBackground, specific band intensity ratio
ELISA1:1,000 to 1:30,000Checkerboard titrationSignal-to-noise ratio at different antigen concentrations
Immunofluorescence1:50 to 1:500Serial dilutionBackground, specific signal localization
Immunohistochemistry1:50 to 1:200Serial dilution with antigen retrieval variationsSignal specificity in tissues with/without target
Flow cytometry1:20 to 1:200Titration against positive/negative cellsSeparation between positive and negative populations

Always include proper controls: (1) secondary antibody only, (2) isotype control, and (3) known positive and negative samples. For immunohistochemistry applications, similar approaches to the APOBEC3B antibody characterization would be applicable, where the monoclonal antibody 5210-87-13 demonstrated superior performance in formalin-fixed paraffin-embedded tissues at specific concentrations .

How can batch-to-batch variation in At3g33187 antibodies be quantified and minimized?

Batch-to-batch variation is a critical concern for reproducible research. Implement these steps to monitor and minimize variations:

  • Standardized quality control panel: Develop a reference panel of positive and negative samples that is tested with each new antibody batch using quantitative methods (ELISA, quantitative Western blot).

  • Epitope mapping: Confirm that each batch recognizes the same epitope region through competition assays or epitope mapping.

  • Affinity measurements: Use surface plasmon resonance (SPR) or bio-layer interferometry (BLI) to quantify binding kinetics (kon, koff, KD) for each batch.

  • Performance metrics tracking: Maintain a database of key performance indicators for each batch:

    • Signal-to-noise ratio in Western blot

    • EC50 values in ELISA

    • Staining intensity and specificity scores in immunohistochemistry

  • Cell bank maintenance: Maintain frozen stocks of original hybridoma cells to enable return to original source if significant drift occurs.

  • Reference standard: Create a gold-standard antibody preparation as a reference to which all new batches are compared.

These approaches have proven effective with other antibodies, including those against heavily glycosylated proteins, where maintaining consistent recognition of specific structural features is crucial .

What are the most common causes of false-positive and false-negative results with At3g33187 antibodies, and how can they be addressed?

Understanding potential sources of error is essential for reliable antibody-based research:

Common Causes of False Results with At3g33187 Antibodies:

Error TypeCommon CausesPrevention Strategies
False PositivesCross-reactivity with related proteinsUse knockout controls; validate with multiple methods
Non-specific binding to plant cell wallOptimize blocking; use appropriate detergents
Endogenous peroxidase/phosphatase activityInclude enzyme inhibition steps
Fc receptor bindingUse F(ab')2 fragments or add normal serum
Secondary antibody cross-reactivityValidate secondary alone; use isotype controls
False NegativesEpitope masking by fixationTest multiple fixation methods; optimize antigen retrieval
Insufficient antibody concentrationTitrate systematically; consider sensitivity-enhancing methods
Protein degradation during sample preparationUse fresh samples; add protease inhibitors
Competitive inhibition by soluble antigenFilter samples; use optimized washing protocols
Sample buffer incompatibilityTest multiple buffer systems for extraction

Each of these issues should be systematically addressed during antibody validation. For plant samples specifically, special attention must be paid to endogenous enzyme activities and cell wall/membrane interactions that can interfere with antibody binding .

How can At3g33187 antibodies be optimized for use in plant species beyond Arabidopsis thaliana?

Extending antibody applications across plant species requires careful optimization:

  • Sequence homology analysis: Perform bioinformatic analysis of At3g33187 homologs across target plant species to predict cross-reactivity.

  • Epitope conservation testing: If the epitope sequence is known, synthesize peptides representing the corresponding sequences from target species and test antibody binding.

  • Sample preparation optimization: Different plant species may require modified extraction protocols due to varying levels of interfering compounds (phenolics, polysaccharides).

  • Fixation protocol adjustments: Cell wall composition varies significantly across plant families, requiring optimization of fixation and permeabilization protocols.

  • Blocking optimization: Test species-specific blocking reagents to minimize background (e.g., normal serum from the same species as the secondary antibody).

  • Signal amplification: For species with lower expression or reduced antibody affinity, implement amplification systems like tyramide signal amplification or polymer-based detection.

These approaches mirror successful strategies for extending antibody applications across multiple species in other research areas, such as with the rabbit monoclonal antibodies that demonstrated utility across multiple detection methods .

How can emerging antibody engineering technologies improve At3g33187 detection and functional studies?

Several cutting-edge approaches show promise for enhancing At3g33187 antibody performance:

  • Single-chain variable fragment (scFv) development: Creating smaller antibody fragments that maintain specificity while improving tissue penetration for in situ applications.

  • Nanobody generation: Single-domain antibodies derived from camelid heavy chains offer exceptional stability and small size, potentially improving access to sterically hindered epitopes.

  • Phage display libraries: Creating custom libraries enriched for plant protein recognition to identify novel binding domains with superior specificity.

  • Bispecific antibodies: Developing antibodies that simultaneously recognize At3g33187 and a second target to study protein interactions or improve detection specificity.

  • Site-specific conjugation: Using enzymatic or chemical approaches for controlled attachment of detection molecules at specific antibody sites to preserve binding properties.

These approaches build upon principles established in the development of other specialized antibodies, such as the natural antibodies that recognize specific carbohydrate structures with high specificity .

What approaches can integrate At3g33187 antibody detection with new imaging technologies?

Emerging imaging technologies offer new possibilities for At3g33187 visualization:

  • Super-resolution microscopy optimization: Develop specific labeling protocols for STORM, PALM, or STED microscopy to visualize At3g33187 distribution at nanoscale resolution.

  • Expansion microscopy protocols: Optimize hydrogel embedding and expansion protocols for plant tissues to achieve physical magnification of antibody-labeled structures.

  • Correlative light-electron microscopy (CLEM): Develop workflows that allow At3g33187 detection by light microscopy followed by ultrastructural analysis of the same sample.

  • Live cell imaging approaches: Engineer non-perturbing antibody fragments conjugated to bright, photostable fluorophores for dynamic studies in living plant cells.

  • Multiplexed imaging systems: Establish protocols for detecting At3g33187 alongside multiple other proteins using cycling methods or spectral unmixing approaches.

These advanced imaging approaches would build upon established immunofluorescence protocols, similar to those used successfully with other antibodies in the field .

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.