At3g42722 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
14-16 week lead time (made-to-order)
Synonyms
At3g42722 antibody; F7P3Putative F-box protein At3g42722 antibody
Target Names
At3g42722
Uniprot No.

Q&A

What critical factors should be considered when selecting antibodies for plant protein targets like At3g42722?

When selecting antibodies for plant protein targets, researchers should prioritize several key factors to ensure experimental success:

Antibody format significantly impacts experimental outcomes. Monoclonal antibodies provide higher specificity but may recognize fewer epitopes, whereas polyclonal antibodies can bind multiple epitopes, potentially providing stronger signals but with higher cross-reactivity risk. For novel targets like At3g42722, initial screening with polyclonal antibodies followed by monoclonal validation often provides complementary advantages.

Epitope accessibility varies considerably in plant proteins depending on their subcellular localization and conformational states. Consider whether the epitope is likely to be exposed in the protein's native conformation for applications requiring detection of non-denatured proteins. For transmembrane or membrane-associated proteins, targeting extracellular domains generally improves accessibility.

Validation data should be carefully scrutinized, particularly ensuring the antibody has been tested in plant systems rather than exclusively in mammalian models. Cross-reactivity profiles should be thoroughly examined to confirm specificity against potential homologs in your experimental system.

Application compatibility must be verified for your specific experimental design. An antibody that performs well in Western blots may not necessarily work for immunoprecipitation or immunofluorescence applications due to differences in epitope accessibility and binding conditions .

How should antibody titration be optimized for detection of plant proteins?

Optimizing antibody concentration is critical for achieving high signal-to-noise ratios. Recent research on antibody titration has revealed:

Most antibodies used at concentrations at or above 2.5 μg/mL show high background signal with minimal additional benefit to specific signal detection. Studies demonstrate that a fourfold reduction in concentration from these levels typically produces minimal loss in sensitivity while significantly reducing background .

Contrary to vendor recommendations of 5-10 μg/mL starting concentrations, evidence suggests beginning with concentrations in the 0.625-2.5 μg/mL range, which often provides sufficient signal without excessive background. This approach has demonstrated a 33.6-fold reduction in antibody costs compared to vendor recommendations while maintaining or improving signal quality .

For highly expressed targets, antibodies can often be used at even lower concentrations without compromising the ability to distinguish positive from negative signals. Antibodies used at concentrations below 0.625 μg/mL typically show linear response to concentration changes, making titration particularly effective in this range .

Sequential dilution testing is recommended, evaluating at least four concentration points (e.g., 2.5, 0.625, 0.156, and 0.039 μg/mL) to identify optimal conditions for each application. This systematic approach allows researchers to categorize antibodies based on their titration response and optimize accordingly .

What comprehensive validation protocols are essential for confirming At3g42722 antibody specificity?

A robust validation approach should include multiple complementary techniques:

Western blot analysis using positive controls (tissues known to express At3g42722) alongside negative controls (knockout lines or tissues lacking expression). The observed molecular weight should match the predicted size, accounting for any post-translational modifications. Signal should disappear in knockout samples and be reduced or absent in peptide competition assays.

Immunoprecipitation followed by mass spectrometry provides powerful validation by confirming the identity of the captured protein. This approach can also identify potential cross-reactive proteins or interaction partners that may complicate interpretation of results.

Cross-reactivity testing against closely related proteins, particularly homologs or family members, is essential for confirming specificity. This can be performed using recombinant proteins or extracts from tissues expressing different family members.

Immunohistochemistry should be performed with appropriate positive and negative tissue controls, with signal distribution matching the expected localization pattern of the target protein. Subcellular localization can provide additional confidence in antibody specificity.

How can researchers minimize non-specific binding when using antibodies in plant systems?

Non-specific binding represents a significant challenge in plant systems due to complex tissue matrices and abundant secondary metabolites. Effective strategies include:

Optimized blocking protocols are essential, with buffer composition tailored to the specific plant tissue. A combination of serum proteins (3-5%), BSA (1-3%), and mild detergents (0.1-0.3% Triton X-100 or Tween-20) often provides effective blocking. For tissues with high endogenous biotin, adding avidin or streptavidin to blocking solutions can reduce background.

Pre-adsorption of antibodies against plant extracts lacking the target protein can significantly reduce non-specific binding. This approach is particularly valuable for polyclonal antibodies and can be implemented by incubating diluted antibody with acetone powder prepared from knockout or wild-type tissues known not to express the target.

Concentration optimization is critical, as research demonstrates that antibodies used at concentrations above 2.5 μg/mL show disproportionately high background signal. Systematic titration can identify the optimal concentration that maximizes the signal-to-noise ratio .

Secondary antibody selection also impacts background, with highly cross-adsorbed secondary antibodies generally providing better specificity. When working with plant tissues that express endogenous immunoglobulins or proteins with Fc-binding domains, using F(ab')2 fragments rather than whole IgG molecules can reduce non-specific interactions.

What approaches should be used to quantitatively analyze antibody-based detection results?

Robust quantitative analysis requires:

Standard curves using recombinant protein at known concentrations should be included whenever possible to establish the relationship between signal intensity and protein quantity. This approach is particularly important for ELISA and other binding assays.

Appropriate normalization controls are essential, including loading controls (e.g., GAPDH, actin, or tubulin) for Western blots and housekeeping proteins for immunohistochemistry. The selected control should have stable expression across experimental conditions.

Technical replicates (minimum of three) and biological replicates (different plant samples) are necessary for statistical validity. Technical replicates assess measurement precision, while biological replicates account for natural variation between samples.

Dynamic range considerations are critical, as detection systems have finite linear ranges. Samples should be diluted or concentrated as needed to ensure measurements fall within this range, and saturation must be avoided for accurate quantification.

Statistical analysis should be appropriate to the data distribution, with parametric tests used only when normality assumptions are met. Nonparametric alternatives should be employed when data does not follow a normal distribution.

How can antibody-based approaches be integrated with other techniques for comprehensive protein analysis?

Multi-modal approaches provide the most complete characterization:

Antibody-based detection combined with mass spectrometry offers powerful validation and characterization capabilities. Immunoprecipitation followed by LC-MS/MS can confirm antibody specificity while identifying interaction partners and post-translational modifications.

Integration with genomic data can provide context for antibody results, particularly when working with novel targets like At3g42722. Expression patterns determined by RNA-seq can be compared with protein detection patterns to identify potential discrepancies between transcription and translation.

Functional assays paired with antibody detection can link protein presence to activity. For enzymes, combining activity assays with immunodetection in fractionated samples can confirm the identity of active protein species.

Multiparameter imaging combining antibody detection with subcellular markers allows precise localization studies. Recent developments in multiplexed antibody staining enable simultaneous detection of multiple proteins, providing insights into co-localization and complex formation.

How can engineered antibodies be utilized to modulate plant protein interactions?

Antibody engineering offers powerful tools for functional studies:

Bispecific antibodies targeting two different epitopes can be valuable tools for studying protein complexes. Research has shown that bispecific antibodies can outperform individually delivered antibodies in certain contexts, potentially by bringing target proteins into proximity or by engaging multiple signaling pathways simultaneously .

Domain-specific targeting can be achieved by designing antibodies against functional domains rather than simply accessible regions. This approach can selectively modulate protein activity without complete inhibition, similar to strategies employed with the SIRPα antibody KWAR23, which disrupts CD47 binding while preserving other functions .

Intrabodies (intracellularly expressed antibodies) can be designed to target specific protein domains within living cells, providing temporal and spatial control over protein function. This approach requires antibodies that fold correctly in the reducing intracellular environment, typically using specialized frameworks like camelid-derived single-domain antibodies.

Nanobodies, derived from camelid heavy-chain antibodies, offer unique advantages for plant research due to their small size (approximately 15 kDa), stability, and ability to access epitopes that conventional antibodies cannot reach. Their single-domain nature also simplifies engineering and recombinant expression.

What methodological considerations are critical when using antibodies in multimodal single-cell analysis of plant tissues?

Multimodal analysis requires careful optimization:

Antibody panel design should balance detection of diverse targets while minimizing interference. Research demonstrates that antibodies used at excessive concentrations (above 2.5 μg/mL) can disproportionately consume sequencing resources without providing additional biological information. In one study, just 4 out of 52 antibodies used at 10 μg/mL accounted for more than 20% of total UMI counts without showing clear positive populations .

Concentration optimization is particularly critical in multiplexed applications. Studies show that reducing the concentration of high-background antibodies from 10 μg/mL to 0.667 μg/mL can dramatically improve signal quality while using 79% fewer sequencing reads. This optimization allowed researchers to achieve a 57% increase in median positive signal alongside a 43% reduction in background signal .

Staining protocols should be carefully optimized, with research showing that reducing antibody concentration has a greater impact on signal quality than reducing staining volume. When working with limited sample material, maintaining adequate staining volume while reducing antibody concentration typically yields better results than reducing volume with higher concentrations .

How should researchers approach development of functional antibodies that modulate plant defense responses?

Developing functional modulatory antibodies requires:

Epitope selection should focus on domains involved in critical protein-protein interactions or signaling functions. Structural biology and protein interaction data can guide identification of these regions. Similar to strategies employed with KWAR23, which disrupts SIRPα-CD47 interaction to enhance immune responses , plant defense modulators should target key regulatory interactions.

Effector function engineering can enhance antibody activity. In mammalian systems, antibodies like PGT145 exhibit potent antibody-dependent cellular cytotoxicity (ADCC) that contributes to viral containment . While classical ADCC isn't relevant in plants, engineering antibodies to recruit specific plant defense components could provide novel regulatory mechanisms.

Combination approaches may be necessary for optimal effects. Research shows that while KWAR23 is inert when administered alone, it significantly enhances tumor killing when combined with tumor-opsonizing antibodies . Similarly, antibodies targeting plant defense proteins might show synergistic effects when used in specific combinations.

Delivery and stability considerations are particularly important in plant systems. Antibodies must be stabilized against plant proteases and delivered effectively to target tissues. Recombinant expression within the plant itself using transgenic approaches may provide advantages over exogenous application.

What is the optimal protocol for antibody titration to maximize signal-to-noise ratio?

Based on recent research, an effective titration protocol includes:

Concentration LevelRange (μg/mL)Expected ResponseRecommended Action
Very High>2.5Minimal response to dilution, high backgroundReduce concentration 4-fold or more
High0.625-2.5Non-linear response to dilutionReduce concentration 2-fold
Moderate0.156-0.625Linear or near-linear responseMaintain or adjust based on signal quality
Low<0.156Linear response, may lose signal with dilutionMaintain or increase if signal is weak

Initial testing should evaluate at least four concentration points across this range. Research has demonstrated that antibodies used at very high concentrations (>2.5 μg/mL) typically show minimal loss of specific signal upon 4-fold reduction while significantly decreasing background .

Signal assessment should quantify both positive signal intensity and background levels. For flow cytometry applications, calculate the ratio of median signal in positive versus negative populations. For imaging applications, measure signal-to-background ratios across multiple fields.

Categorization of antibodies based on their titration response allows rational optimization:

  • Category A: High background that decreases with dilution without loss of specific signal

  • Category B: Good signal-to-noise ratio but signal could be reduced to balance panel

  • Category C: Optimal signal-to-noise ratio at current concentration

  • Category D: Weak signal that decreases with dilution

  • Category E: No clear positive population

Final validation should test optimized concentrations in the full experimental setup, verifying that positive and negative populations remain clearly distinguishable .

What approaches are most effective for troubleshooting poor antibody performance in plant systems?

Systematic troubleshooting should address:

Sample preparation issues often underlie poor antibody performance. For recalcitrant plant tissues, test alternative extraction buffers with different detergents (CHAPS, NP-40, Triton X-100) and salt concentrations. Consider using specialized plant protein extraction kits that address challenges like high polyphenol content.

Cross-reactivity can be evaluated through peptide competition assays and testing in knockout lines. If cross-reactivity is confirmed, more stringent washing conditions or higher dilutions may improve specificity, though potentially at the cost of sensitivity.

Detection system optimization may be necessary if the primary antibody is functioning but signal remains weak. For enzyme-linked detection, extend substrate development time or switch to more sensitive substrates. For fluorescence-based detection, consider signal amplification systems or longer exposure times.

How can researchers effectively develop antibodies against challenging plant-specific epitopes?

For challenging plant targets:

Immunogen design is critical. For proteins like At3g42722 with limited characterization, computational prediction of surface-exposed, antigenic regions can guide peptide design. Multiple peptides from different protein regions should be tested in parallel to identify the most immunogenic epitopes.

Expression system selection significantly impacts protein folding and post-translational modifications. For plant proteins, plant-based expression systems (N. benthamiana, BY-2 cells) often provide more native-like proteins than bacterial or mammalian systems, potentially yielding antibodies that better recognize the native protein.

Selection strategies should include negative selection against similar proteins to enhance specificity. For antibody development against At3g42722, pre-adsorption against extracts from knockout plants or competitive elution strategies can enrich for highly specific antibodies.

Validation in multiple assay formats is essential, as antibodies may perform well in some applications but poorly in others. Testing should include Western blot, immunoprecipitation, immunofluorescence, and when possible, knockout/knockdown controls to confirm specificity.

How might emerging antibody technologies advance research on plant proteins like At3g42722?

Emerging technologies offer new research opportunities:

Single-domain antibodies (nanobodies) derived from camelids provide unique advantages for plant research due to their small size, stability in diverse conditions, and ability to access epitopes that conventional antibodies cannot reach. Their single-domain nature also simplifies engineering and recombinant expression within plant systems.

Proximity-labeling applications combine antibodies with enzymes like BirA or APEX2 to catalyze biotinylation of proteins in close proximity to the target. This approach can identify interacting partners or proximal proteins in native cellular contexts, providing insights into protein function and complex formation.

Intracellular antibody expression (intrabodies) allows targeting of proteins within living cells, potentially modulating function in a temporally and spatially controlled manner. This approach is particularly valuable for studying dynamic processes and has been successfully applied in plant systems using nanobody frameworks.

Bispecific antibody formats that simultaneously target two different epitopes enable novel research applications. Studies in mammalian systems have demonstrated that bispecific antibodies can outperform individually delivered antibodies in certain contexts , suggesting potential applications in plant research for studying protein complexes or signaling pathways.

What considerations are important when designing antibodies for studying protein-protein interactions in plant signaling pathways?

Effective design requires:

Epitope selection should avoid regions involved in critical interactions unless the goal is to disrupt those interactions. Structural data, when available, can guide selection of accessible epitopes that do not interfere with function unless such interference is desired.

Binding kinetics significantly impact the utility of antibodies for interaction studies. High-affinity antibodies may disrupt weak interactions, while lower-affinity antibodies may fail to maintain stable complexes during isolation procedures. Titration experiments are essential to identify optimal conditions .

Format considerations include using smaller antibody fragments (Fab, scFv) that cause less steric hindrance when studying large protein complexes. For in vivo applications, camelid single-domain antibodies offer advantages due to their small size and stability.

Validation approaches should include tests that specifically address whether the antibody preserves or disrupts known protein interactions. Co-immunoprecipitation of known interaction partners followed by Western blot or mass spectrometry analysis can confirm complex integrity.

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.