At5g19315 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
At5g19315 antibody; F7K24Putative defensin-like protein 23 antibody
Target Names
At5g19315
Uniprot No.

Target Background

Database Links
Protein Families
DEFL family
Subcellular Location
Secreted.

Q&A

What essential characteristics should be analyzed when characterizing a newly developed monoclonal antibody?

A thorough characterization of any monoclonal antibody, including those targeting plant proteins like At5g19315, must assess three fundamental properties: structural integrity, specificity, and potency .

Structural integrity analysis should employ a combination of techniques including SDS-PAGE, isoelectric focusing (IEF), HPLC, and mass spectrometry to verify the antibody is not fragmented, aggregated, or otherwise modified (e.g., through loss of carbohydrate side chains) . Side-by-side comparisons with a well-characterized in-house reference standard are essential for establishing consistency .

Specificity must be rigorously demonstrated through direct binding assays incorporating both positive and negative controls. At minimum, include an isotype-matched irrelevant control antibody and negative antigen controls with similar chemical properties but no antigenic relationship to your target . Whenever possible, biochemically define the protein, glycoprotein, or other molecule bearing the reactive epitope, and determine the antigenic epitope itself .

Potency characterization requires quantitative measurement of binding activity using techniques like ELISA, RIA, or flow cytometry, with activity expressed as specific antigen-binding units per mg of antibody . The potency assay should ideally relate closely to the antibody's physiological/pharmacological activity and be sufficiently sensitive to detect functionally significant differences .

How can researchers validate the specificity of a monoclonal antibody for their target of interest?

Validating antibody specificity requires a multi-method approach with rigorous controls. First, perform direct binding assays with both positive and negative controls, including at least one isotype-matched, irrelevant control antibody . For plant protein targets like those encoded by At5g19315, include closely related proteins as negative controls.

Second, conduct fine specificity studies using defined structural preparations (e.g., peptides representing different domains of your target protein) to characterize binding through inhibition or other techniques . For complex biological mixtures, standardize test antigen lots used for direct binding tests .

Third, quantify binding affinity, avidity, and immunoreactivity using established methods appropriate to your experimental system . Consider competitive binding studies with known ligands of your target protein to further confirm specificity.

The experience with monoclonal antibody At5 provides an instructive example. Though initially developed against sturgeon chordin, researchers demonstrated through protein sequencing and immunoblotting that it recognized dMAG in human brain, along with proteoglycans phosphacan and neurocan . This cross-species reactivity highlights the importance of comprehensive specificity testing across diverse samples.

What methodological approaches can determine whether a monoclonal antibody recognizes native versus denatured forms of the target protein?

This distinction is crucial for selecting appropriate applications. To evaluate recognition of native conformations, employ:

  • Flow cytometry with live cells expressing the target protein

  • Immunoprecipitation from non-denaturing lysates

  • ELISA using purified protein in non-denaturing buffers

  • Native gel electrophoresis followed by Western blotting

For denatured protein recognition, assess binding using:

  • Western blotting with samples boiled in reducing SDS buffer

  • Immunohistochemistry on fixed tissues using different fixatives

  • Dot blots with denatured protein samples

The case of monoclonal antibody At5 illustrates these considerations. Initially raised against chordin, it demonstrated different reactivity patterns in fixed tissues versus biochemical preparations . In mixed oligoastrocytoma sections, At5 showed intense staining in oligodendroglioma areas while astrocytoma regions lacked immunoreactivity , suggesting conformation-dependent epitope recognition.

To comprehensively characterize your antibody, conduct parallel experiments using both native and denatured sample preparations, documenting which conditions yield optimal specific binding . This information will guide appropriate application selection and experimental design.

How should researchers design cross-reactivity testing for monoclonal antibodies targeting plant proteins?

Cross-reactivity testing for plant protein antibodies requires systematic assessment across taxonomically diverse plant tissues and related protein families. Implement the following methodological approach:

  • Phylogenetic analysis: Test antibody binding across evolutionary diverse plant species, focusing on both closely and distantly related taxa to the original target organism. Include representatives spanning monocots, dicots, and non-flowering plants when possible.

  • Tissue panel screening: Examine reactivity across different tissue types (leaves, roots, stems, reproductive structures) within the same species, as protein modifications may vary by tissue context. The approach used with At5 antibody, which showed differential reactivity in oligodendroglioma versus astrocytoma regions of mixed tumors , exemplifies the importance of tissue-specific testing.

  • Protein family analysis: Test against related proteins within the same family as your target. For At5g19315, this would include proteins with similar domain structures or functions within Arabidopsis.

  • Epitope competition assays: Perform inhibition studies using synthetic peptides corresponding to the presumed epitope region and related sequences from homologous proteins .

  • Multi-technique validation: Confirm cross-reactivity patterns using complementary methods (Western blot, ELISA, immunohistochemistry) as epitope accessibility may differ between techniques .

Document all cross-reactivity in a comprehensive table showing percent cross-reactivity relative to the primary target across all tested species, tissues, and related proteins. This data is essential for accurately interpreting experimental results and identifying potential false positives.

What methodological strategies can overcome challenges in detecting low-abundance plant proteins using monoclonal antibodies?

Detecting low-abundance plant proteins requires specialized approaches to enhance sensitivity while maintaining specificity:

  • Sample enrichment protocols:

    • Subcellular fractionation to concentrate compartment-specific proteins

    • Immunoprecipitation with polyclonal antibodies followed by detection with monoclonal antibodies

    • Size-exclusion chromatography to separate target proteins from abundant contaminants

  • Signal amplification methods:

    • Tyramide signal amplification (TSA) for immunohistochemistry

    • Polymer-based detection systems that carry multiple secondary antibodies

    • Biotin-streptavidin systems with optimized blocking to minimize background

  • Instrumentation optimization:

    • Confocal microscopy with photomultiplier tube gain optimization

    • Flow cytometry with enhanced voltage settings for fluorescence channels

    • Chemiluminescence detection with extended exposure times and cooled CCD cameras

  • Validation controls:

    • Transgenic overexpression lines as positive controls

    • Knockout/knockdown lines as negative controls

    • Concentration gradients of recombinant protein to establish detection limits

  • Background reduction strategies:

    • Pre-adsorption of antibodies with plant tissue extracts from knockout lines

    • Optimized blocking with plant-specific blocking agents (e.g., non-fat milk can contain plant-reactive components)

    • Extended washing steps with detergent-optimized buffers

The principles employed in analyzing the heterogeneous anti-Neu5Gc antibody response could be adapted here, particularly the approach of using structurally similar but antigenically distinct controls . Just as Neu5Ac-containing glycans served as background controls for Neu5Gc detection , similar plant glycoproteins lacking your specific epitope would provide optimal background controls.

How can researchers design studies to determine whether post-translational modifications affect antibody recognition of plant proteins?

Post-translational modifications (PTMs) can significantly impact epitope accessibility and antibody binding. Implement this systematic approach:

  • Modification-specific comparative analysis:

    • Phosphorylation: Compare antibody binding before and after phosphatase treatment

    • Glycosylation: Test binding after treatment with specific glycosidases

    • Ubiquitination: Compare binding to native protein versus deubiquitinated samples

    • Methylation/acetylation: Compare binding to chemically demethylated/deacetylated samples

  • Recombinant protein panel testing:

    • Generate protein variants with site-directed mutagenesis at known or predicted modification sites

    • Express proteins in systems with different modification capabilities (E. coli vs. insect cells vs. plant expression systems)

    • Create a panel of proteins with defined modifications through in vitro enzymatic treatments

  • Parallel antibody strategy:

    • Compare binding patterns of multiple antibodies recognizing different epitopes on the same protein

    • Include modification-specific antibodies (e.g., anti-phospho antibodies) in parallel experiments

    • Develop a binding matrix documenting reactivity patterns across antibody-modification combinations

  • Mass spectrometry correlation:

    • Perform immunoprecipitation followed by mass spectrometry to identify modifications on specifically bound proteins

    • Correlate modification states with antibody binding efficiency

    • Generate quantitative data on preferential binding to specific modified forms

This approach is informed by studies of complex antigens like those recognized by HNK-1 family antibodies, which bind to glycolipids and high molecular weight glycoconjugates . The At5 antibody's differential binding to proteoglycans phosphacan and neurocan exemplifies how modifications affect recognition patterns.

What systematic approach should researchers use to optimize immunohistochemistry protocols for plant tissue sections using monoclonal antibodies?

Optimizing immunohistochemistry for plant tissues requires systematic variation of key parameters while controlling for plant-specific challenges:

  • Fixation optimization matrix:

    Fixative TypeConcentration RangeDuration RangeTemperature
    Paraformaldehyde2-4%1-24 hours4°C, RT
    Glutaraldehyde0.1-2.5%1-4 hours4°C, RT
    Ethanol70-100%1-24 hoursRT
    Combined PFA/GlutVarious ratios1-4 hours4°C, RT
  • Antigen retrieval method comparison:

    • Heat-induced epitope retrieval (pressure cooker, microwave)

    • Enzymatic retrieval (proteinase K, trypsin)

    • pH-modified buffers (citrate pH 6.0, EDTA pH 8.0, Tris pH 9.0)

    • No retrieval (baseline control)

  • Blocking optimization:

    • Plant-specific blocking agents (plant-derived albumins)

    • Standard blocking solutions (BSA, normal sera, commercial blockers)

    • Pre-adsorption with plant material lacking target

    • Hydrogen peroxide treatment for peroxidase quenching

  • Antibody incubation parameters:

    • Concentration gradient (typically 0.1-10 μg/mL)

    • Duration (1 hour to overnight)

    • Temperature (4°C, room temperature)

    • Buffer composition (salt concentration, detergent type/amount)

  • Detection system comparison:

    • Direct fluorescence vs. enzymatic (HRP, AP) methods

    • Polymer-based vs. avidin-biotin systems

    • Amplification systems (tyramide) vs. conventional detection

    • Counterstaining compatibility assessment

For each combination, quantitatively assess signal-to-noise ratio and specific staining pattern. The experience with At5 antibody, which showed differential staining in oligodendroglioma versus astrocytoma regions , illustrates how optimization can reveal important biological differences in antibody reactivity patterns.

What strategies can effectively minimize non-specific binding when using monoclonal antibodies in plant protein research?

Non-specific binding presents particular challenges in plant systems due to endogenous biotin, peroxidases, and secondary metabolites. Implement these evidence-based approaches:

  • Pre-adsorption techniques:

    • Pre-incubate antibodies with acetone powder preparations from plants lacking the target protein

    • For recombinant antibodies, express and purify the antibody using plant-based expression systems to remove cross-reactive clones

    • Perform subtractive screening against related plant proteins during antibody development

  • Buffer optimization:

    • Increase detergent concentration (0.1-0.3% Triton X-100 or Tween-20)

    • Add competing proteins (1-5% BSA or casein)

    • Adjust salt concentration (150-500 mM NaCl)

    • Include reducing agents (1-5 mM DTT) to minimize disulfide-based interactions

  • Secondary reagent optimization:

    • Use highly cross-adsorbed secondary antibodies

    • Consider F(ab')2 fragments to avoid Fc receptor binding

    • Test secondary antibodies from different host species

    • For plant tissues, use secondaries pre-adsorbed against plant proteins

  • Blocking endogenous activities:

    • Quench endogenous peroxidases (3% H2O2, 30 minutes)

    • Block endogenous biotin (commercial biotin-blocking kits)

    • Inhibit endogenous alkaline phosphatase (levamisole)

    • Pre-treat samples with appropriate enzymes to remove interfering molecules

  • Control implementation:

    • Include isotype-matched control antibodies at identical concentrations

    • Use target-knockout plant material as a negative control

    • Include secondary-only controls to assess direct binding

    • Implement absorption controls with immunizing peptide

The importance of these controls is exemplified by specificity studies of monoclonal antibodies like At5, where multiple control approaches revealed true binding patterns versus background .

What troubleshooting decision tree should researchers follow when a monoclonal antibody fails to detect the target protein in plant samples?

When antibody detection fails, a systematic troubleshooting approach based on testing key variables is essential:

  • Sample preparation issues:

    • Is the target protein present? → Verify by mass spectrometry or alternative detection method

    • Is the protein intact? → Check extraction methods for protease inhibitors and denaturing conditions

    • Is the epitope accessible? → Try multiple extraction methods and buffers

    • Are interfering compounds present? → Test purification steps (dialysis, desalting)

  • Antibody concerns:

    • Is the antibody functional? → Test with positive control (recombinant protein)

    • Is the concentration appropriate? → Perform titration experiments (0.1-10 μg/mL)

    • Does the antibody recognize the species variant? → Check sequence homology in epitope region

    • Has the antibody degraded? → Check storage conditions, prepare fresh dilutions

  • Technical factors:

    • Is the detection system working? → Include system control (directly labeled standard)

    • Are buffers correct? → Verify pH, salt concentration, and detergent content

    • Are blocking conditions excessive? → Test reduced blocking stringency

    • Is the incubation adequate? → Extend time or adjust temperature

  • Target protein variables:

    • Is post-translational modification blocking the epitope? → Test deglycosylation, dephosphorylation

    • Is the protein expression level too low? → Implement concentration steps or signal amplification

    • Is the target in complex with other proteins? → Try more stringent lysis conditions

    • Is the protein localized unexpectedly? → Check multiple cellular fractions

  • Method-specific adjustments:

    • Western blotting: Try different transfer conditions, membrane types

    • ELISA: Adjust coating buffer, blocking agent

    • IHC/ICC: Test multiple fixation and antigen retrieval methods

    • IP: Modify lysis buffer composition, test different bead types

Document all troubleshooting steps systematically, as the experience with characterizing the diverse anti-Neu5Gc antibody response demonstrates how methodical optimization eventually reveals specific binding patterns that were initially obscured .

How should researchers quantitatively analyze and statistically validate immunoblotting data using monoclonal antibodies?

Rigorous quantitative analysis of immunoblot data requires standardized approaches:

  • Image acquisition controls:

    • Capture images within the linear dynamic range of the detection system

    • Include exposure series to confirm signal linearity

    • Use identical acquisition settings for all comparable samples

    • Include molecular weight markers in all images

  • Quantification methodology:

    • Use densitometry software with background subtraction

    • Normalize target protein bands to loading controls (e.g., GAPDH, actin, total protein stain)

    • Include calibration curves using purified protein standards

    • Report results as relative units or absolute quantities when standards are used

  • Statistical analysis framework:

    • Perform experiments with minimum n=3 biological replicates

    • Apply appropriate statistical tests based on data distribution:

      • For normally distributed data: t-tests (two groups) or ANOVA (multiple groups)

      • For non-parametric data: Mann-Whitney U (two groups) or Kruskal-Wallis (multiple groups)

    • Report both p-values and effect sizes

    • Use error bars representing standard deviation or standard error consistently

  • Validation controls:

    • Technical replicates to assess method precision

    • Biological replicates to assess natural variation

    • Positive and negative controls for antibody specificity

    • Gradient controls for quantification linearity

  • Result presentation standards:

    • Show representative blots with molecular weight markers

    • Include all relevant controls in figures

    • Present quantitative data as charts with appropriate statistics

    • Provide detailed methods including antibody dilutions and exposure times

This approach aligns with the rigorous quantification methods used in characterizing diverse anti-Neu5Gc antibodies, where serial dilutions of human IgG provided accurate quantification standards .

What criteria should researchers use to evaluate contradictory results from experiments using different monoclonal antibodies against the same target?

When different antibodies yield contradictory results, apply this methodical evaluation framework:

  • Antibody characterization assessment:

    • Epitope mapping: Do the antibodies recognize different epitopes?

    • Binding conditions: Do the antibodies require different conditions for optimal binding?

    • Cross-reactivity profiles: Have specificity tests been equally rigorous for both antibodies?

    • Validation methods: Were knockout/knockdown controls used for both antibodies?

  • Technical factors analysis:

    • Detection methods: Were different detection systems used?

    • Sample preparation: Were samples prepared identically for both antibodies?

    • Protocol optimization: Was each antibody used with its optimized protocol?

    • Batch effects: Were experiments performed simultaneously or separately?

  • Biological considerations:

    • Protein isoforms: Could the antibodies recognize different isoforms?

    • Post-translational modifications: Do modifications affect one antibody's binding but not the other's?

    • Protein complexes: Does one antibody recognize the free form while another detects the complex?

    • Subcellular localization: Are the antibodies accessing different cellular compartments?

  • Resolution strategy:

    • Independent method verification: Use non-antibody methods (mass spectrometry, RNA analysis)

    • Third antibody testing: Introduce additional antibodies with well-characterized properties

    • Recombinant expression: Test binding to defined recombinant constructs

    • Genetic validation: Use CRISPR-edited lines lacking the target protein

The case of At5 antibody is instructive here. Initially developed against chordin, additional characterization revealed it recognized diverse neural antigens including dMAG, phosphacan, and neurocan . Without this extensive characterization, contradictory results between At5 and other antibodies might have been misinterpreted.

How can researchers determine whether apparent changes in protein expression detected by monoclonal antibodies represent genuine biological differences versus technical artifacts?

Distinguishing true biological changes from technical artifacts requires systematic validation:

  • Technical replication controls:

    • Repeat experiments with identical and varied technical parameters

    • Test multiple antibody lots and dilutions

    • Compare results across different detection methods

    • Vary sample preparation methods while maintaining biological conditions

  • Biological validation approaches:

    • Correlate protein changes with mRNA levels (qRT-PCR, RNA-seq)

    • Verify with alternative antibodies recognizing different epitopes

    • Test in multiple biological models or conditions

    • Employ genetic approaches (overexpression, knockout) to confirm causality

  • Quantitative assessment:

    • Establish detection thresholds based on signal-to-noise ratios

    • Determine intra-assay and inter-assay coefficients of variation

    • Calculate minimal detectable differences based on system variability

    • Implement hierarchical statistical models accounting for technical and biological variation

  • Controls for specific confounding factors:

    • Epitope masking: Test denatured versus native samples

    • Protein relocalization: Examine multiple cellular fractions

    • Processing changes: Analyze protein size variants

    • Protease sensitivity: Include protease inhibitor controls

  • Documentation standards:

    • Record all experimental parameters systematically

    • Document antibody validation data comprehensively

    • Report all tested conditions, not only supportive results

    • Provide raw data alongside processed results

This approach is informed by the heterogeneity observed in anti-Neu5Gc antibody responses, where careful methodology revealed true biological variation in antibody levels and specificities between individuals , demonstrating how rigorous controls can distinguish genuine biological differences from technical artifacts.

What methodological approaches can researchers use to develop antibodies with improved specificity for plant-specific post-translational modifications?

Developing highly specific antibodies for plant-specific modifications requires specialized strategies:

  • Immunogen design optimization:

    • Use synthetic peptides containing the exact modification of interest

    • Include the surrounding amino acid sequence specific to the plant protein

    • Create multiple immunogens with the modification in different sequence contexts

    • Develop carrier protein conjugation methods that preserve modification integrity

  • Screening methodology enhancement:

    • Implement parallel screening against modified and unmodified antigens

    • Include structurally similar but distinct modifications as specificity controls

    • Develop high-throughput plant tissue microarrays for screening

    • Use competition assays with defined modified and unmodified peptides

  • Production and purification optimization:

    • Express antibodies in plant-based systems to remove cross-reactive clones

    • Develop negative selection steps against common plant epitopes

    • Implement affinity maturation strategies to enhance specificity

    • Purify using modified antigen columns with stringent wash conditions

  • Validation matrix implementation:

    • Test against tissues from wildtype and mutant plants lacking the modification

    • Assess cross-reactivity with related plant species

    • Evaluate performance across multiple experimental platforms

    • Validate using mass spectrometry confirmation of modifications

  • Antibody engineering approaches:

    • Generate recombinant antibody fragments with enhanced specificity

    • Create bispecific antibodies requiring dual epitope recognition

    • Develop synthetic binding proteins as alternatives to traditional antibodies

    • Implement computational design to minimize cross-reactivity with common plant epitopes

The approach used in characterizing the specificity of monoclonal antibody At5, which demonstrated similarities to antibodies of the HNK-1 family , provides a model for how to rigorously establish specificity profiles for antibodies against complex modifications.

How can researchers effectively employ monoclonal antibodies to study protein-protein interactions in plant systems?

Studying plant protein-protein interactions with antibodies requires specialized approaches addressing plant-specific challenges:

  • Co-immunoprecipitation optimization:

    • Develop gentle lysis buffers preserving native plant protein complexes

    • Implement crosslinking strategies appropriate for plant cell walls

    • Optimize antibody coupling to beads to minimize leaching during plant extract incubation

    • Include appropriate controls for plant-specific non-specific binding

  • Proximity labeling methods:

    • Adapt BioID or APEX2 proximity labeling for plant systems

    • Optimize biotin-ligase fusion expression in plant cells

    • Develop plant-specific extraction methods for biotinylated proteins

    • Validate interaction partners using reciprocal approaches

  • In situ interaction detection:

    • Implement proximity ligation assays optimized for plant tissues

    • Develop clearing methods compatible with antibody epitope preservation

    • Optimize FRET/FLIM approaches for plant autofluorescence backgrounds

    • Adapt BiFC techniques for plant-specific subcellular compartments

  • Quantitative analysis frameworks:

    • Establish control datasets for random protein associations in plant cells

    • Develop scoring systems accounting for plant-specific contaminants

    • Implement mass spectrometry normalization for plant protein abundance

    • Create visualization tools for plant protein interaction networks

  • Validation strategies:

    • Genetic confirmation through co-expression, knockout, and mutation studies

    • Structural validation through domain mapping and mutation analysis

    • Functional assessment through biochemical assays of co-purified complexes

    • Computational prediction integration with experimental results

The principles used in studying complex glycoconjugates recognized by antibodies like At5, which identified interactions with proteoglycans phosphacan and neurocan , demonstrate how antibody-based approaches can reveal complex molecular interactions when rigorously validated.

What emerging technologies are improving the specificity and utility of monoclonal antibodies for plant protein research?

Several cutting-edge technologies are transforming plant antibody research:

  • Next-generation recombinant antibody platforms:

    • Nanobodies (VHH antibodies) with enhanced tissue penetration for plant tissues

    • Plant-expressed synthetic scaffolds avoiding mammalian glycosylation patterns

    • Computationally designed paratopes targeting plant-specific epitopes

    • Yeast surface display for rapid selection of plant-compatible antibodies

  • Integrated multi-omics validation approaches:

    • Correlation of antibody binding with proteomics and transcriptomics data

    • Epitope confirmation through hydrogen-deuterium exchange mass spectrometry

    • Cross-linking mass spectrometry to confirm binding sites

    • Systems biology frameworks integrating antibody-based datasets with other omics data

  • Advanced imaging applications:

    • Expansion microscopy protocols optimized for plant cell walls

    • Multi-epitope ligand cartography for plant tissue mapping

    • Adaptive optics for deep tissue imaging with labeled antibodies

    • Light-sheet microscopy approaches for whole-organ antibody labeling

  • Quantitative binding analysis technologies:

    • Surface plasmon resonance optimized for plant protein complexes

    • Bio-layer interferometry with plant extract compatibility

    • Microfluidic diffusional sizing for plant protein interactions

    • Single-molecule pull-down assays for rare plant protein complexes

  • Antibody-guided enzyme technologies:

    • CRISPR-directed modification of specific protein populations

    • Antibody-directed proximity labeling for subcellular proteomics

    • Targeted protein degradation using antibody-degron fusions

    • Spatial transcriptomics guided by antibody recognition

These technologies build upon the fundamental principles of antibody specificity demonstrated in studies of antibodies like At5, which showed differential tissue reactivity patterns , and the heterogeneous anti-Neu5Gc antibody response requiring sensitive detection methods . The implementation of these advanced approaches will continue to improve the specificity, sensitivity, and utility of antibodies in plant research.

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.