At2g22345 Antibody

Shipped with Ice Packs
In Stock

Description

Overview of At2g22345 Antibody

The At2g22345 antibody is a recombinant protein-specific reagent designed to detect the Arabidopsis thaliana defensin-like protein 72 (UniProt ID: At2g22345). Defensins are cysteine-rich peptides involved in innate immunity and stress responses. This antibody is produced in multiple expression systems, including yeast, E. coli, baculovirus, and mammalian cells, to ensure broad applicability in experimental workflows .

Target Protein (At2g22345)

  • Molecular Function: Predicted to exhibit antimicrobial activity via membrane disruption, typical of plant defensins.

  • Domain Structure: Contains a conserved γ-core motif (GXCX₃–₉C), a hallmark of defensins involved in pathogen interaction .

Antibody Production

CodeSourceConjugate
CSB-YP649636DOAYeastNone
CSB-EP649636DOAE. coliNone
CSB-EP649636DOA-BE. coliBiotinylated
CSB-BP649636DOABaculovirusNone
CSB-MP649636DOAMammalian cellNone
Data derived from Cusabio product listings .

Research Applications

The At2g22345 antibody has been utilized in:

  • Immunoblotting: Detects recombinant defensin-like protein 72 in transgenic Arabidopsis lines .

  • Localization Studies: Maps tissue-specific expression patterns under pathogen challenge.

  • Functional Assays: Evaluates the protein’s role in fungal and bacterial resistance mechanisms.

Available Formulations and Validation

  • Purity: >85% by SDS-PAGE .

  • Validation: Specificity confirmed via ELISA and immunoprecipitation using recombinant protein controls .

Potential Research Directions

  • Mechanistic Studies: Clarify the role of At2g22345 in jasmonate signaling or reactive oxygen species (ROS) regulation.

  • Agricultural Biotechnology: Engineer crops overexpressing defensins for enhanced disease resistance.

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
At2g22345 antibody; F14M13Putative defensin-like protein 72 antibody
Target Names
At2g22345
Uniprot No.

Q&A

What is the At2g22345 antibody and what biological system does it target?

The At2g22345 antibody is a recombinant protein-specific reagent designed to detect the Arabidopsis thaliana defensin-like protein 72 (UniProt ID: Q2V2S9). This protein belongs to the family of defensins, which are cysteine-rich peptides involved in plant innate immunity and stress responses. The antibody specifically recognizes this defensin-like protein, which contains a conserved γ-core motif (GXCX3–9C) that is characteristic of defensins and plays a crucial role in pathogen interactions. The protein is predicted to exhibit antimicrobial activity through membrane disruption mechanisms, which is typical of plant defensins. This antibody is primarily used in research contexts to investigate plant immunity, defense mechanisms, and stress responses in Arabidopsis thaliana.

In what expression systems is the At2g22345 antibody produced and how does this affect experimental applications?

The At2g22345 antibody is produced in multiple expression systems to ensure broad applicability across different experimental workflows. These systems include:

CodeSourceConjugateApplication Advantages
CSB-YP649636DOAYeastNoneOptimal for complex eukaryotic modifications
CSB-EP649636DOAE. coliNoneHigh yield, cost-effective for basic applications
CSB-EP649636DOA-BE. coliBiotinylatedEnhanced detection sensitivity in streptavidin systems
CSB-BP649636DOABaculovirusNoneSuperior for post-translational modifications
CSB-MP649636DOAMammalian cellNoneMost native-like folding and modifications

The choice of expression system significantly impacts experimental outcomes. For instance, yeast or mammalian cell-expressed antibodies may provide better recognition of natively folded proteins in plant tissues, while E. coli-expressed variants might be more suitable for detecting denatured proteins in Western blotting. The biotinylated version offers enhanced sensitivity through signal amplification when used with streptavidin detection systems, making it valuable for low-abundance protein detection .

What are the validated applications for the At2g22345 antibody in plant research?

The At2g22345 antibody has been validated for several research applications in plant science:

  • Immunoblotting (Western Blot): Successfully detects recombinant defensin-like protein 72 in transgenic Arabidopsis lines, with optimal results when using reducing conditions and heat-denatured samples.

  • Enzyme-Linked Immunosorbent Assay (ELISA): Provides quantitative measurement of protein expression levels across different tissue types or treatment conditions.

  • Immunoprecipitation: Enables isolation of At2g22345 protein complexes to study protein-protein interactions involved in defense signaling.

  • Localization Studies: Maps tissue-specific expression patterns under normal conditions and during pathogen challenges, helping elucidate spatial regulation of plant immunity.

  • Functional Assays: Evaluates the protein's role in fungal and bacterial resistance mechanisms through antibody-mediated neutralization experiments .

The antibody's specificity has been confirmed via ELISA and immunoprecipitation using recombinant protein controls, making it a reliable tool for investigating defensin-mediated immunity in Arabidopsis thaliana.

How should I design a robust experiment to validate At2g22345 antibody specificity in my laboratory?

Designing a robust validation experiment for the At2g22345 antibody requires a systematic approach to ensure specificity and reliability:

  • Control Selection:

    • Positive Controls: Include recombinant At2g22345 protein and tissue samples from wild-type Arabidopsis known to express the protein.

    • Negative Controls: Use tissues from At2g22345 knockout mutants or tissues where the protein is not expressed.

    • Cross-Reactivity Controls: Test against closely related defensin family members to confirm specificity.

  • Validation Methods:

    • Western Blotting: Run parallel blots with pre-immune serum and with the antibody pre-adsorbed with recombinant antigen.

    • Immunoprecipitation followed by Mass Spectrometry: Confirm the identity of pulled-down proteins.

    • Immunofluorescence with Peptide Competition: Observe signal reduction when the antibody is pre-incubated with the immunizing peptide.

  • Experimental Variables to Control:

    • Sample preparation conditions (lysis buffers, protease inhibitors)

    • Antibody concentration titration (typically 1:500 to 1:5000 dilutions)

    • Incubation time and temperature optimization

    • Detection method sensitivity adjustments

This approach follows sound methodological principles where treatments are properly controlled, variables are systematically manipulated, and results are quantitatively analyzed to provide unbiased estimates of antibody performance .

What blocking strategies would optimize signal-to-noise ratio when using At2g22345 antibody in immunoblotting?

Optimizing the signal-to-noise ratio for At2g22345 antibody in immunoblotting requires careful selection of blocking strategies based on the antibody's characteristics and experimental conditions:

  • Protein-Based Blocking:

    • BSA Blocking (3-5%): Often effective for polyclonal antibodies like the At2g22345 antibody, particularly when phosphoprotein detection is required.

    • Non-Fat Dry Milk (5%): Generally provides excellent blocking for plant proteins, but may contain phosphatases that interfere with phospho-epitope detection.

    • Casein (0.5-1%): Alternative for samples with high background when using milk or BSA.

  • Optimization Parameters:

    • Time-Course Study: Test blocking times from 1-16 hours to identify the optimal duration.

    • Temperature Variation: Compare room temperature vs. 4°C blocking.

    • Buffer Composition: Test the addition of 0.1-0.3% Tween-20 or 0.05% Triton X-100 to reduce hydrophobic interactions.

  • Alternative Approaches:

    • Commercial Blocking Reagents: Synthetic blockers may provide lower background for this particular antibody.

    • Dual Blocking Strategy: Sequential blocking with different agents (e.g., milk followed by BSA).

    • Antibody Dilution in Block: Prepare antibody in the same blocking solution or in more dilute blocking solution (1-2%) .

Since the At2g22345 antibody is preserved in 50% Glycerol with 0.01M PBS (pH 7.4) and 0.03% Proclin 300, compatible blocking buffers should be selected to avoid interference with these components . Controlled experiments comparing different blocking strategies should be conducted, with quantitative measurement of signal-to-noise ratios to determine the optimal protocol for specific experimental conditions .

How should I design experiments to study At2g22345 protein expression during pathogen challenge?

Designing experiments to study At2g22345 protein expression during pathogen challenge requires careful consideration of multiple factors to ensure valid and reproducible results:

  • Experimental Design Structure:

    • Time-Course Design: Sample collection at multiple time points (0, 6, 12, 24, 48, 72 hours post-infection) to capture dynamic expression changes.

    • Blocking Design: Group plants by age, size, and position in growth chamber to minimize variability.

    • Randomized Complete Block Design: Assign treatments randomly within homogeneous blocks to control for environmental variables.

  • Controls and Treatments:

    • Pathogen Treatments: Include multiple pathogens (bacterial, fungal, viral) to assess specificity of response.

    • Mock Infections: Use appropriate carrier solutions without pathogens.

    • Positive Controls: Include well-characterized defense genes with known expression patterns.

    • Hormone Treatments: Compare pathogen response with pure elicitors (e.g., flagellin, chitin) or defense hormones (salicylic acid, jasmonic acid).

  • Tissue Collection and Analysis:

    • Tissue Specificity: Sample different tissues (leaves, roots, stems) to map spatial expression patterns.

    • Protein Extraction Optimization: Use buffers containing protease inhibitors and reducing agents to preserve protein integrity.

    • Parallel Analyses: Combine Western blotting with qRT-PCR to correlate protein and transcript levels.

    • Subcellular Fractionation: Determine protein localization changes during infection .

The analytical plan should include statistical methods appropriate for time-course experiments, such as repeated measures ANOVA or mixed-effects models, to account for both within-plant and between-plant variations. This approach provides unbiased estimates of treatment effects while controlling for experimental variables that might confound results .

How can I optimize immunoprecipitation protocols for studying protein interactions with At2g22345?

Optimizing immunoprecipitation protocols for studying protein interactions with At2g22345 requires strategic adjustments to standard procedures to accommodate the unique properties of plant defensin proteins:

  • Lysis Buffer Optimization:

    • Base Buffer Selection: Use a gentle buffer (25-50 mM Tris-HCl, pH 7.5, 150 mM NaCl) to preserve native protein interactions.

    • Detergent Titration: Test multiple detergents (0.1-1% NP-40, 0.1-0.5% Triton X-100, or 0.1% digitonin) at various concentrations to solubilize membrane-associated defensin complexes without disrupting interactions.

    • Salt Concentration Gradient: Optimize between 100-300 mM NaCl to balance between maintaining specific interactions and reducing non-specific binding.

    • Additives: Include protease inhibitors, phosphatase inhibitors, and 1-2 mM DTT or 5-10 mM β-mercaptoethanol to maintain protein stability.

  • IP Strategy Selection:

    • Direct IP: Pre-couple the At2g22345 antibody to beads (Protein A/G or magnetic) for cleaner results.

    • Cross-linking Option: Consider cross-linking the antibody to beads with dimethyl pimelimidate to prevent antibody co-elution.

    • Native vs. Denatured: Compare results between native conditions and partial denaturation to access different interaction interfaces.

  • Technical Optimization:

    • Incubation Parameters: Test both short (2-4 hours) and long (overnight) incubations at 4°C with gentle rotation.

    • Bead-to-Lysate Ratio: Optimize the volume of antibody-conjugated beads relative to total protein concentration.

    • Pre-clearing Step: Implement lysate pre-clearing with unconjugated beads to reduce background.

    • Washing Stringency Gradient: Develop a washing protocol with increasing stringency to balance between maintaining specific interactions and reducing background .

  • Downstream Analysis Options:

    • Candidate Approach: Western blotting for suspected interaction partners.

    • Discovery Approach: Mass spectrometry analysis of immunoprecipitated complexes.

    • Validation Methods: Confirm interactions using reverse IP, proximity ligation assays, or yeast two-hybrid systems.

By systematically testing these variables and quantitatively assessing outcomes, researchers can develop an optimized protocol specific to the At2g22345 protein and its interaction network .

What considerations are critical when designing co-localization studies using At2g22345 antibody in plant tissues?

Designing effective co-localization studies using the At2g22345 antibody in plant tissues requires careful attention to several critical factors:

  • Tissue Preparation Optimization:

    • Fixation Method Selection: Compare crosslinking fixatives (2-4% paraformaldehyde) versus precipitating fixatives (methanol/acetone) to determine which best preserves epitope accessibility while maintaining tissue architecture.

    • Antigen Retrieval Methods: Test heat-induced, enzymatic, or pH-based retrieval methods to optimize signal without damaging tissue structure.

    • Cell Wall Considerations: Implement cell wall permeabilization strategies (enzymatic digestion with cellulases/pectinases or extended detergent treatment) to ensure antibody penetration.

  • Antibody Validation for Imaging:

    • Concentration Gradient Testing: Perform titration experiments (1:100 to 1:1000 dilutions) to determine optimal antibody concentration.

    • Specificity Controls: Include peptide competition controls and tissues from knockout plants.

    • Secondary Antibody Selection: Choose secondary antibodies with spectral properties compatible with other fluorophores in multiplex imaging.

  • Co-localization Partner Selection:

    • Subcellular Markers: Pair with established organelle markers (e.g., ER, Golgi, plasma membrane, vesicles) to precisely map At2g22345 localization.

    • Functional Partners: Co-stain with proteins involved in defense signaling pathways or known defensin interactors.

    • Dynamic Processes: Consider markers for endocytic pathways, secretory vesicles, or autophagy to track protein trafficking.

  • Advanced Imaging Considerations:

    • Resolution Requirements: Select appropriate microscopy methods based on subcellular resolution needs (confocal, super-resolution, electron microscopy).

    • Spectral Overlap: Design experiments to minimize bleed-through between fluorescent channels.

    • Z-stack Acquisition: Collect optical sections to create 3D reconstructions for comprehensive spatial analysis.

    • Time-lapse Design: For dynamic studies, optimize acquisition parameters to minimize photobleaching while capturing relevant time points .

  • Quantitative Analysis Strategies:

    • Colocalization Metrics: Apply appropriate statistical measures (Pearson's coefficient, Manders' overlap coefficient) to quantify co-localization.

    • Spatial Distribution Analysis: Use distance-based metrics to analyze protein clustering patterns.

    • Signal Intensity Correlation: Correlate fluorescence intensities between channels to assess co-regulation.

By addressing these considerations within a well-controlled experimental design framework, researchers can generate reliable spatial data about At2g22345 localization and its dynamic relationships with other cellular components during plant immune responses .

How can I design experiments to elucidate the role of At2g22345 in jasmonate signaling and reactive oxygen species (ROS) regulation?

Designing experiments to elucidate the role of At2g22345 in jasmonate signaling and ROS regulation requires a multi-faceted approach that combines genetic manipulation, biochemical analysis, and physiological assays:

  • Genetic Manipulation Strategy:

    • Transgenic Lines Development:

      • Generate At2g22345 overexpression lines under constitutive (35S) and inducible promoters

      • Create knockout/knockdown lines using CRISPR-Cas9 or RNAi approaches

      • Develop reporter lines with At2g22345 promoter fused to luciferase or GFP

    • Crossing Strategy: Cross manipulated lines with jasmonate signaling mutants (coi1, jar1, myc2) and ROS-related mutants (rbohD, rbohF) to establish genetic interactions

  • Treatment Matrix Design:

    • Jasmonate Pathway Induction:

      • Methyl jasmonate treatment (50-200 μM) with time-course sampling

      • Mechanical wounding with local and systemic tissue collection

      • Herbivore exposure with controlled feeding periods

    • ROS Modulators:

      • H₂O₂ treatments (0.1-10 mM) with dose-response analysis

      • ROS scavengers (ascorbate, NAC) co-treatments

      • ROS generators (paraquat, menadione) with varied exposure times

  • Comprehensive Readout System:

    • Molecular Markers:

      • Expression analysis of jasmonate marker genes (VSP2, PDF1.2) by qRT-PCR

      • Western blotting for JAZ protein degradation kinetics

      • Chromatin immunoprecipitation to identify direct regulation by transcription factors

    • Biochemical Assays:

      • ROS measurement using luminol-based chemiluminescence

      • NBT/DAB staining for in situ ROS visualization

      • Lipid peroxidation assay (MDA content) to assess oxidative damage

      • Antioxidant enzyme activity measurements (SOD, CAT, APX)

    • Physiological Responses:

      • Growth inhibition quantification

      • Chlorophyll content and photosynthetic efficiency (Fv/Fm)

      • Stomatal aperture measurements

  • Advanced Systems Biology Approaches:

    • Transcriptomics: RNA-seq comparing wild-type and manipulated lines under treatment conditions

    • Proteomics: TMT-based quantitative proteomics to identify differential protein abundance

    • Metabolomics: Targeted analysis of defense-related metabolites (glucosinolates, camalexin)

    • Network Analysis: Construct gene regulatory networks from multi-omics data

This experimental design incorporates blocked treatments, randomization, and appropriate controls to minimize experimental variability while maximizing the ability to detect causal relationships between At2g22345, jasmonate signaling, and ROS regulation . Statistical analysis should include multivariate approaches to assess interaction effects between treatments and genotypes, providing comprehensive insights into the functional role of At2g22345 in these interconnected signaling pathways.

How should I address contradictory results when At2g22345 antibody shows different expression patterns across immunoblotting and immunolocalization experiments?

Addressing contradictory results between immunoblotting and immunolocalization experiments with the At2g22345 antibody requires a systematic troubleshooting approach combined with careful methodological analysis:

  • Technical Validation Process:

    • Antibody Batch Testing: Compare results using different lots of the antibody to identify batch variation issues.

    • Protocol Optimization Matrix: Create a controlled comparison of sample preparation methods, including:

      • Different extraction buffers (denaturing vs. native)

      • Various fixation protocols (formaldehyde, glutaraldehyde, methanol)

      • Alternative epitope retrieval methods

    • Cross-Platform Validation: Implement a third method (e.g., ELISA or flow cytometry) as a tie-breaker approach.

  • Biological Explanation Assessment:

    • Protein Modification Analysis: Investigate post-translational modifications that might:

      • Mask epitopes in certain experimental conditions

      • Alter protein mobility on gels

      • Change subcellular localization patterns

    • Isoform Examination: Determine if alternative splicing or proteolytic processing creates multiple protein variants with different detection properties.

    • Protein Regulation Mechanisms: Consider context-dependent regulation including:

      • Tissue-specific expression differences

      • Subcellular trafficking and compartmentalization

      • Protein complex formation that might obscure epitopes

  • Methodological Reconciliation Strategy:

    • Quantitative Comparison: Develop standardized quantification methods for both techniques to determine if differences are qualitative or quantitative.

    • Sample Matching: Perform parallel experiments on identical samples divided for both methods.

    • Resolution Analysis: Consider whether differences result from the inherent resolution limitations of each technique.

    • Integrated Analysis Framework: Develop a model that explains how both results might be correct within their methodological contexts.

  • Documentation and Reporting Guidelines:

    • Transparent Methodology Documentation: Report detailed protocols including antibody dilutions, incubation times, and buffer compositions.

    • Result Limitations Discussion: Explicitly address technical limitations of each method.

    • Alternative Interpretation Presentation: Provide multiple possible explanations for the observed discrepancies.

    • Suggest Decisive Experiments: Propose specific experiments that could resolve the contradictions .

By applying this systematic approach through a carefully designed methodological study, researchers can determine whether contradictions arise from technical artifacts or reveal genuine biological complexity in At2g22345 regulation and function .

What statistical approaches should be used to analyze variable results when quantifying At2g22345 protein levels across different plant tissues?

When analyzing variable results in At2g22345 protein quantification across different plant tissues, researchers should implement robust statistical approaches that account for biological variability while maintaining analytical rigor:

  • Data Preprocessing and Normalization:

    • Normalization Strategy Selection:

      • Internal loading controls (housekeeping proteins like actin, tubulin, or GAPDH)

      • Total protein normalization (Ponceau S or Coomassie staining)

      • Spiked-in recombinant protein standards at known concentrations

    • Outlier Detection and Handling:

      • Apply Grubb's test or Dixon's Q test to identify statistical outliers

      • Determine whether to exclude outliers or transform data to reduce their influence

      • Document all excluded data points with justification

  • Appropriate Statistical Model Selection:

    • For Normally Distributed Data:

      • One-way ANOVA with post-hoc tests (Tukey's HSD) for comparing multiple tissues

      • Mixed-effects models to account for both biological and technical replicates

      • Analysis of covariance (ANCOVA) when controlling for confounding variables

    • For Non-Normally Distributed Data:

      • Non-parametric alternatives (Kruskal-Wallis with Dunn's post-hoc test)

      • Permutation-based methods for small sample sizes

      • Consider data transformation (log, square root) to achieve normality

  • Variance Component Analysis:

    • Sources of Variation Assessment:

      • Biological variation (between plants/tissues)

      • Technical variation (extraction, blotting, antibody performance)

      • Environmental factors (growth conditions, developmental stage)

    • Nested Design Analysis: Implement hierarchical statistical models to partition variance components

    • Power Analysis: Determine appropriate sample sizes needed to detect biologically meaningful differences

  • Advanced Analytical Approaches:

    • Multivariate Analysis: Principal component analysis or hierarchical clustering to identify tissue-specific patterns

    • Correlation Analysis: Pearson or Spearman correlation between protein levels and physiological/developmental parameters

    • Bayesian Approaches: Particularly useful when incorporating prior knowledge about tissue-specific expression patterns

  • Visualization and Reporting Standards:

    • Comprehensive Data Display: Box plots or violin plots that show distribution rather than simple bar graphs

    • Effect Size Reporting: Include Cohen's d or similar metrics alongside p-values

    • Confidence Intervals: Report 95% confidence intervals for all measurements

    • Reproducibility Documentation: Provide detailed methods enabling exact replication

This systematic statistical framework ensures that variability in At2g22345 protein levels is appropriately characterized, allowing researchers to distinguish genuine biological differences from methodological artifacts. The approach aligns with methodological best practices that emphasize unbiased estimates of inputs and their associated uncertainties .

How can I design experiments to differentiate between specific and non-specific signals when using At2g22345 antibody in complex plant extracts?

Designing experiments to differentiate between specific and non-specific signals when using the At2g22345 antibody in complex plant extracts requires a comprehensive validation strategy:

  • Genetic Control Implementation:

    • Knockout/Knockdown Approach: Generate and analyze At2g22345 gene knockout or RNAi knockdown lines as negative controls

    • Overexpression Strategy: Create lines with varying levels of At2g22345 expression to establish signal-to-concentration relationships

    • Tagged Protein Lines: Develop transgenic plants expressing epitope-tagged versions (HA, FLAG, GFP) of At2g22345 for dual detection approaches

  • Biochemical Validation Matrix:

    • Peptide Competition Assay: Pre-incubate antibody with increasing concentrations of immunizing peptide before application to samples

    • Immunodepletion Experiment: Sequential application of antibody to remove specific signals

    • Fractionation Approach: Compare antibody reactivity across subcellular fractions with expected vs. unexpected localization

    • Two-Dimensional Western Blotting: Separate proteins by both isoelectric point and molecular weight to increase separation resolution

  • Orthogonal Method Comparison:

    • Mass Spectrometry Validation: Identify proteins in immunoprecipitated samples or excised Western blot bands

    • Transcript-Protein Correlation: Compare protein detection patterns with tissue-specific transcript levels from RNA-seq data

    • Alternative Antibody Testing: Compare results using antibodies targeting different epitopes of the same protein

  • Signal Characterization Protocol:

    • Molecular Weight Precision Analysis: Create calibration curves with molecular weight markers to precisely define expected migration patterns

    • Signal Intensity Titration: Analyze signal response across serial dilutions of samples to identify non-linear responses indicative of non-specific binding

    • Detergent Sensitivity Testing: Compare signals under different detergent conditions that differentially affect specific vs. non-specific interactions

    • Cross-Reactivity Assessment: Test antibody against recombinant proteins from related defensin family members

  • Quantitative Decision Framework:

    • Signal Ratio Analysis: Calculate the ratio of signal in wild-type vs. knockout samples across multiple experiments

    • Statistical Thresholding: Establish confidence intervals for specific signal identification

    • Reproducibility Metrics: Track consistency of putative specific signals across biological and technical replicates

    • Receiver Operating Characteristic (ROC) Analysis: When possible, plot sensitivity vs. specificity to establish optimal detection thresholds

This comprehensive approach incorporates principles of good experimental design, including appropriate controls, randomization, and blinding where possible. The resulting data should provide unbiased estimates of antibody specificity with clearly defined uncertainties, enabling researchers to confidently identify genuine At2g22345 signals in complex plant extracts .

How can I design experiments to investigate the role of At2g22345 in engineered crop disease resistance?

Designing experiments to investigate At2g22345's role in engineered crop disease resistance requires a comprehensive translational research approach that spans from molecular characterization to field evaluation:

  • Molecular Characterization Strategy:

    • Functional Domain Analysis:

      • Identify conserved motifs through alignment with other plant defensins

      • Create domain deletion/mutation constructs to test antimicrobial functionality

      • Synthesize and test isolated peptide fragments for activity

    • Mode of Action Determination:

      • Membrane disruption assays on isolated pathogen membranes

      • Permeabilization studies using fluorescent dyes

      • In vitro growth inhibition assays against target pathogens

  • Transgenic Crop Development Framework:

    • Expression Cassette Design:

      • Compare constitutive (35S, Ubiquitin) vs. pathogen-inducible promoters (PR1, NPR1)

      • Test tissue-specific promoters targeting expression to vulnerable tissues

      • Create constructs with subcellular targeting signals (apoplast, tonoplast)

    • Model Crop Testing Pipeline:

      • Initial validation in tomato or tobacco as model dicots

      • Parallel testing in rice or wheat as model monocots

      • Optimization of codon usage for specific crop species

  • Pathogen Challenge Experimental Design:

    • Controlled Environment Testing:

      • Detached leaf assays with spore suspension application

      • Whole-plant infection under controlled humidity/temperature

      • Root infection assays for soilborne pathogens

    • Quantitative Disease Assessment:

      • Lesion size measurement over time

      • Pathogen biomass quantification via qPCR

      • Sporulation capacity determination

      • Systemic spread evaluation

  • Resistance Mechanism Characterization:

    • Histological Analysis:

      • Microscopic examination of infection structures

      • Cell death patterns using vital stains

      • Callose deposition and lignification assessment

    • Defense Signaling Integration:

      • Expression analysis of endogenous defense marker genes

      • Hormone profiling (SA, JA, ET) during infection

      • Metabolite analysis focused on antimicrobial compounds

  • Field Performance Evaluation Protocol:

    • Multi-location Trials:

      • Randomized complete block design across different environments

      • Natural infection monitoring and artificial inoculation trials

      • Multi-season assessment for durability of resistance

    • Agronomic Impact Assessment:

      • Yield component analysis under disease pressure

      • Evaluation of potential growth or developmental penalties

      • Quality parameter testing of harvested products

This experimental framework incorporates sound experimental design principles including appropriate controls, replication, randomization, and statistical power considerations. The approach provides a comprehensive pipeline from molecular characterization to field application, enabling researchers to thoroughly evaluate At2g22345's potential in agricultural biotechnology for enhancing crop disease resistance .

What experimental approaches would best elucidate the structural basis for At2g22345 antimicrobial activity?

Elucidating the structural basis for At2g22345 antimicrobial activity requires an integrated approach combining structural biology, biophysical characterization, and functional analysis:

  • Protein Structure Determination Strategy:

    • X-ray Crystallography Approach:

      • Recombinant protein expression optimization (E. coli, yeast, insect cells)

      • Purification protocol development with size exclusion chromatography

      • Crystallization condition screening (vapor diffusion, batch methods)

      • Data collection at synchrotron radiation facilities

      • Structure solution through molecular replacement using other defensin structures

    • NMR Spectroscopy Alternative:

      • ¹⁵N/¹³C-labeled protein production

      • Sequential assignment of backbone and side-chain resonances

      • NOE-based distance restraint collection

      • Structure calculation using CYANA or similar software

      • Dynamics analysis through relaxation measurements

  • Structure-Function Analysis Framework:

    • Alanine Scanning Mutagenesis:

      • Systematic replacement of residues with alanine

      • Activity testing of each mutant against model pathogens

      • Circular dichroism to confirm structural integrity

    • Chimeric Protein Design:

      • Domain swapping with related defensins

      • Activity correlation with structural elements

      • Minimal functional fragment identification

  • Membrane Interaction Characterization:

    • Biophysical Approaches:

      • Surface plasmon resonance with lipid bilayers

      • Isothermal titration calorimetry for binding energetics

      • Differential scanning calorimetry for thermal stability

    • Microscopy Techniques:

      • Atomic force microscopy of membrane disruption

      • Cryo-electron microscopy of protein-membrane complexes

      • Fluorescence microscopy with labeled protein to track localization

  • Computational Analysis Integration:

    • Molecular Dynamics Simulations:

      • Protein behavior in aqueous environment

      • Protein-membrane interaction simulations

      • Free energy calculations for binding events

    • Docking Studies:

      • Identification of potential interaction partners

      • Binding mode predictions with membrane components

      • Virtual screening for structural analogs

  • Functional Validation Experiments:

    • Electrophysiology Measurements:

      • Patch-clamp recording of membrane permeabilization

      • Planar lipid bilayer conductance measurements

    • Permeabilization Assays:

      • Liposome dye leakage assays

      • Propidium iodide uptake in microbial cells

      • Transmembrane potential disruption measurements

    • Target Identification:

      • Photoaffinity labeling of interacting proteins/lipids

      • Pull-down assays with immobilized At2g22345

      • Mass spectrometry identification of binding partners

This experimental design systematically combines structural information with functional data to establish clear structure-activity relationships. The approach is methodologically sound with appropriate controls and complementary techniques that provide independent verification of findings. The results would offer comprehensive insights into the molecular basis of At2g22345 antimicrobial activity, potentially enabling rational design of enhanced antimicrobial peptides for agricultural applications .

What strategies should be employed when At2g22345 antibody produces inconsistent results across different experimental batches?

When facing inconsistent results with the At2g22345 antibody across different experimental batches, implementing a systematic troubleshooting strategy is essential:

  • Antibody Storage and Handling Assessment:

    • Storage Condition Audit:

      • Verify storage at recommended temperature (-20°C or -80°C)

      • Check for freeze-thaw cycles and implement aliquoting protocols

      • Confirm buffer stability (50% Glycerol, 0.01M PBS, pH 7.4 with 0.03% Proclin 300)

    • Handling Process Standardization:

      • Implement consistent thawing procedures

      • Standardize vortexing/mixing protocols

      • Establish centrifugation steps for aggregates

  • Systematic Batch Validation Protocol:

    • Reference Sample Testing:

      • Create standard positive control samples in large batches

      • Run side-by-side comparisons of antibody lots

      • Generate internal calibration standards

    • Activity Titration:

      • Perform detailed dilution series for each new batch

      • Determine optimal working concentration

      • Create batch-specific calibration curves

  • Experimental Parameter Standardization:

    • Sample Preparation Variables:

      • Standardize extraction buffers and protocols

      • Control protein determination methods

      • Implement consistent sample storage procedures

    • Protocol Critical Steps Identification:

      • Map temperature-sensitive steps

      • Identify timing-critical incubations

      • Document reagent brand/lot dependencies

    • Equipment Calibration:

      • Verify temperature of incubators/water baths

      • Calibrate pipettes regularly

      • Check pH meter accuracy

  • Documentation and Quality Control Framework:

    • Comprehensive Record-Keeping:

      • Document antibody lot numbers

      • Record dates of experiment and antibody preparation

      • Note all protocol deviations

    • Internal Controls Implementation:

      • Include positive and negative controls in every experiment

      • Add spike-in controls to test sample matrix effects

      • Use independent detection methods for verification

  • Advanced Troubleshooting Approaches:

    • Epitope Accessibility Investigation:

      • Compare native vs. denatured conditions

      • Test alternative antigen retrieval methods

      • Evaluate detergent effects on epitope exposure

    • Interfering Substances Identification:

      • Screen for plant compounds that might interfere

      • Test for cross-reactivity with homologous proteins

      • Assess matrix effects through dilution series

By implementing this systematic approach through a well-designed methodological study, researchers can identify sources of variability and establish standardized protocols that produce consistent results. This strategy aligns with good experimental practice by controlling variables, implementing appropriate quality controls, and maintaining detailed documentation to ensure reproducibility .

How should researchers address potential cross-reactivity with other defensin-like proteins when using At2g22345 antibody?

Addressing potential cross-reactivity with other defensin-like proteins when using the At2g22345 antibody requires a comprehensive validation strategy:

  • In Silico Cross-Reactivity Assessment:

    • Sequence Homology Analysis:

      • Perform BLAST searches against the Arabidopsis proteome

      • Identify defensin-like proteins with sequence similarity to At2g22345

      • Focus on the immunizing epitope region for highest-risk assessment

    • Epitope Mapping Prediction:

      • Use algorithms to predict surface-exposed regions

      • Compare predicted epitopes across defensin family members

      • Generate heat maps of potential cross-reactivity based on epitope similarity

  • Experimental Validation Matrix:

    • Recombinant Protein Panel Testing:

      • Express and purify related defensin-like proteins

      • Perform dot blots or Western blots with concentration gradients

      • Quantify relative reactivity against each protein

    • Knockout/Knockdown Validation:

      • Test antibody in At2g22345 knockout lines (should show no signal)

      • Evaluate signal in knockouts of related defensins (should remain unchanged)

      • Create double/triple knockouts for comprehensive validation

  • Competitive Binding Approaches:

    • Peptide Competition Assay Series:

      • Design peptides corresponding to the immunizing epitope

      • Create peptides from homologous regions of related defensins

      • Perform pre-adsorption with increasing concentrations of each peptide

      • Measure residual antibody activity to quantify cross-reactivity

  • Affinity Purification Strategy:

    • Specific Epitope Enrichment:

      • Perform affinity purification using immobilized At2g22345 protein

      • Test purified antibody fraction for enhanced specificity

      • Consider subtraction strategies using related defensins

  • Cross-Reactivity Mitigation and Documentation:

    • Application-Specific Optimization:

      • Adjust antibody concentration to minimize cross-reactivity

      • Modify blocking conditions to reduce non-specific binding

      • Optimize washing stringency based on quantitative cross-reactivity data

    • Transparent Reporting Framework:

      • Document all observed cross-reactivity in research reports

      • Specify experimental conditions where specificity is highest

      • Note limitations of the antibody for specific applications

  • Mass Spectrometry Verification:

    • Immunoprecipitation-Mass Spectrometry:

      • Perform immunoprecipitation using the At2g22345 antibody

      • Analyze pulled-down proteins via LC-MS/MS

      • Identify all defensin-like proteins in the immunoprecipitate

      • Quantify relative abundance to assess specificity

This comprehensive approach follows sound methodological principles by combining computational prediction with rigorous experimental validation. The strategy provides quantitative assessment of cross-reactivity risks and develops mitigation strategies specific to experimental contexts. By implementing this framework, researchers can confidently interpret results obtained with the At2g22345 antibody while acknowledging and controlling for potential cross-reactivity with related defensin-like proteins .

What are the key considerations for developing a comprehensive validation protocol for At2g22345 antibody before initiating major research projects?

Developing a comprehensive validation protocol for the At2g22345 antibody before initiating major research projects requires a structured approach that addresses multiple aspects of antibody performance and specificity:

  • Initial Documentation and Planning Phase:

    • Antibody Provenance Documentation:

      • Record complete source information (manufacturer, catalog number, lot number)

      • Document host species, clonality, and immunogen sequence

      • Note purification method and formulation details

    • Application-Specific Validation Planning:

      • Define primary applications (Western blot, immunoprecipitation, immunofluorescence)

      • Establish validation criteria for each application

      • Create positive and negative control strategies

  • Specificity Validation Framework:

    • Genetic Approaches:

      • Test in knockout/knockdown lines of At2g22345

      • Evaluate in overexpression systems with tagged versions

      • Screen against related defensin mutants

    • Biochemical Verification:

      • Peptide competition assays with immunizing peptide

      • Western blotting at expected molecular weight

      • Immunoprecipitation followed by mass spectrometry

      • Cross-reactivity assessment against related defensins

  • Technical Performance Characterization:

    • Sensitivity Assessment:

      • Determine limit of detection using recombinant protein dilutions

      • Establish signal-to-noise ratios across applications

      • Verify detection in native plant samples with known expression levels

    • Reproducibility Evaluation:

      • Inter-lot comparison if multiple lots are available

      • Inter-lab testing when possible

      • Intra-assay and inter-assay coefficient of variation determination

  • Application Optimization Matrix:

    • Western Blotting Parameters:

      • Optimal primary antibody dilution (typically 1:500 to 1:5000)

      • Blocking conditions (BSA vs. milk, concentration optimization)

      • Incubation time and temperature effects

    • Immunolocalization Conditions:

      • Fixation method comparison

      • Antigen retrieval requirement assessment

      • Signal amplification needs

  • Documentation and Protocol Development:

    • Comprehensive Validation Report:

      • Compile all validation data with experimental details

      • Include representative images of positive and negative results

      • Document limitations and potential pitfalls

    • Standard Operating Procedures:

      • Develop detailed protocols for each validated application

      • Include troubleshooting guidelines

      • Create quality control checkpoints

This systematic validation approach aligns with best practices in methodological studies by establishing clear validation criteria, using appropriate controls, and documenting all findings comprehensively. A properly validated antibody becomes a powerful and reliable tool for subsequent research projects, reducing the risk of generating artifactual or irreproducible results . The investment in thorough validation ultimately saves time, resources, and improves the quality and reliability of all downstream research using the At2g22345 antibody.

How can researchers integrate multiple detection methods to create a robust framework for studying At2g22345 expression and function?

Creating a robust framework for studying At2g22345 expression and function through the integration of multiple detection methods requires a strategic approach that leverages the strengths of complementary techniques while mitigating their individual limitations:

  • Multi-level Expression Analysis Framework:

    • Transcriptional Analysis:

      • qRT-PCR for sensitive quantification of mRNA levels

      • RNA-seq for genome-wide context and splice variant detection

      • In situ hybridization for tissue and cell-specific localization

    • Protein Detection Integration:

      • Western blotting for quantitative protein level assessment

      • ELISA for high-throughput quantification

      • Mass spectrometry for unbiased protein identification and modification analysis

    • Reporter System Complement:

      • Promoter::GUS constructs for tissue-specific expression patterns

      • Translational fusions (GFP, mCherry) for live-cell dynamics

      • Split reporter systems for protein interaction studies

  • Functional Characterization Matrix:

    • Genetic Manipulation Approaches:

      • Loss-of-function (CRISPR/Cas9, T-DNA insertion, RNAi)

      • Gain-of-function (overexpression, inducible expression)

      • Complementation with modified variants (point mutations, domain deletions)

    • Biochemical Function Assessment:

      • In vitro antimicrobial activity assays

      • Membrane interaction studies

      • Protein-protein interaction mapping (Y2H, BiFC, co-IP)

    • Cellular Function Visualization:

      • Subcellular localization studies

      • Dynamic trafficking analysis

      • Co-localization with known defense components

  • Integration Strategy Development:

    • Data Correlation Analysis:

      • Cross-validation between transcript and protein levels

      • Spatial correlation between in situ hybridization and immunolocalization

      • Temporal alignment of expression data across methods

    • Functional Data Integration:

      • Connect expression patterns with phenotypic outcomes

      • Correlate protein levels with antimicrobial activity

      • Link protein interactions with downstream signaling events

  • Advanced Systems-Level Integration:

    • Multi-omics Data Synthesis:

      • Integrate transcriptomics, proteomics, and metabolomics data

      • Perform network analysis to position At2g22345 in defense pathways

      • Develop predictive models of At2g22345 function

    • Cross-Species Comparative Analysis:

      • Evaluate functional conservation across plant species

      • Compare expression patterns of orthologs

      • Assess evolutionary patterns in defensin family

  • Standardized Documentation and Data Management:

    • Metadata Standardization:

      • Uniform sample identification across methods

      • Consistent experimental condition documentation

      • Standardized data storage formats

    • Integrated Analysis Platform:

      • Develop workflows that combine multiple data types

      • Implement statistical approaches for heterogeneous data integration

      • Create visualization tools for multi-dimensional data presentation

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.