The At2g43620 locus in Arabidopsis thaliana corresponds to a chitinase gene critical for hydrolyzing chitin, a key component of fungal cell walls. Its expression is induced during pathogen-associated molecular pattern (PAMP)-triggered immunity (PTI) and effector-triggered immunity (ETI) .
| Gene ID | Protein Class | Function | Expression Trigger |
|---|---|---|---|
| At2g43620 | Chitinase | Fungal cell wall degradation | Pathogen infection, flg22/elf18 |
While direct commercial or experimental data on the At2g43620 antibody is limited in publicly available literature, its hypothetical uses align with standard practices for plant immune protein detection:
Western Blotting: Quantify protein levels in mutants (e.g., dde2, ein2, pad4, sid2) under immune challenges .
Immunohistochemistry: Localize chitinase expression in plant tissues during fungal infections.
qRT-PCR Validation: Cross-validate mRNA levels with protein abundance .
Studies measuring At2g43620 expression reveal its role in immune signaling networks:
flg22: Bacterial flagellin-derived peptide; FB1: Fungal toxin fumonisin B1.
Quadruple mutants lack JA, ET, SA, and phytoalexin pathways, highlighting At2g43620's dependency on these sectors .
Plant antibody specificity is critical due to structural similarities between proteins. For example:
Commercial antibodies for unrelated targets (e.g., angiotensin II AT2 receptor) often show nonspecific binding .
Epitope mapping and inhibition assays (as in SARS-CoV-2 antibody studies) are recommended to confirm At2g43620 antibody specificity .
Chitinases like At2g43620 are promising targets for enhancing crop resistance. Lessons from monoclonal antibodies in human therapeutics (e.g., ARG2 inhibitors ) suggest that plant antibody engineering could optimize immune responses without off-target effects.
At2g43620 is a gene locus on chromosome 2 of Arabidopsis thaliana that encodes a protein of interest in plant molecular biology. Antibodies targeting this protein enable researchers to:
Track protein expression patterns across different tissues and developmental stages
Determine subcellular localization through microscopy-based techniques
Study protein-protein interactions in native cellular environments
Investigate post-translational modifications and protein turnover dynamics
These antibodies bridge the gap between genomic data and functional proteomics, allowing researchers to confirm whether transcript abundance correlates with actual protein levels. For reliable results, validation of antibody specificity is essential, requiring both positive and negative controls in direct binding assays, including at least one isotype-matched, irrelevant (negative) control antibody .
Comprehensive validation of At2g43620 antibodies requires multiple complementary approaches:
Specificity testing:
Western blotting against wild-type and knockout/knockdown plant tissues
Immunoprecipitation followed by mass spectrometry identification
Cross-reactivity testing against related proteins in the same family
Binding characterization:
Affinity determination using titration experiments across multiple dilutions
Epitope mapping to confirm the antibody targets the intended region
Competitive binding assays with purified antigens
Application-specific validation:
Optimization for each experimental technique (Western blot, immunoprecipitation, immunostaining)
Reproducibility testing across different sample preparations
Comparison of results from multiple antibodies targeting different epitopes
According to best practices, the protein bearing the reactive epitope should be biochemically defined and the antigenic epitope determined whenever possible . For complex samples, standardized test antigens and inhibitors should be used, with quantitative measurement of antibody binding inhibition by soluble antigen or other antibodies .
Proper storage and handling of At2g43620 antibodies is crucial for maintaining reactivity and specificity:
Long-term storage:
Store concentrated antibody stocks at -20°C or -80°C in small aliquots to avoid freeze-thaw cycles
Include cryoprotectants such as glycerol (30-50%) to prevent freezing damage
Maintain records of antibody performance from each aliquot to track potential degradation
Working dilutions:
Prepare fresh working dilutions for each experiment when possible
Store diluted antibodies at 4°C for short periods only (1-2 weeks)
Add preservatives such as sodium azide (0.02%) to prevent microbial growth
Handling precautions:
Avoid repeated freeze-thaw cycles which can cause antibody denaturation
Centrifuge after thawing to remove aggregates before use
Use clean, nuclease-free tubes and pipette tips to prevent contamination
Quality control:
Periodically test antibody function using consistent positive controls
Document lot-to-lot variation if using commercial antibodies
Consider including stabilizing proteins (BSA, gelatin) for dilute solutions
Proper storage and handling practices significantly impact experimental reproducibility and validity of results across different studies.
Optimizing Western blot protocols for At2g43620 antibody requires attention to several key parameters:
Sample preparation:
Extract proteins in buffers containing appropriate detergents (RIPA or NP-40)
Include protease inhibitors to prevent degradation
Denature samples in SDS loading buffer at 95°C for 5 minutes
Gel electrophoresis:
Use 10-12% polyacrylamide gels for optimal separation
Load 20-50 μg of total protein per lane
Include molecular weight markers to confirm target protein size
Transfer conditions:
Transfer to PVDF membrane at 100V for 1 hour or 30V overnight
Confirm transfer efficiency with reversible protein staining
Blocking:
Antibody incubation:
Detection:
Use HRP-conjugated secondary antibody matched to primary antibody species
Develop with enhanced chemiluminescence substrate
Expose to film or digital imager for appropriate duration
Each of these parameters may require optimization for specific experimental contexts to achieve the optimal balance between specific signal and background.
Determining optimal antibody dilution requires systematic titration experiments:
Initial screening:
Begin with a broad range of dilutions (1:500, 1:1000, 1:2000, 1:5000)
Use consistent positive control samples across all dilutions
Evaluate both signal intensity and background levels
Concentration-based considerations:
Fine-tuning based on initial results:
Narrow testing range around promising dilutions (e.g., 1:800, 1:1000, 1:1200)
Assess signal-to-noise ratio rather than absolute signal strength
Consider exposure times when comparing different dilutions
Application-specific adjustments:
Different techniques (Western blot, immunofluorescence, ELISA) require different optimal dilutions
Adjust based on detection method sensitivity (chemiluminescence, fluorescence, colorimetric)
Consider increasing dilution to reduce background if specific signal remains adequate
For advanced techniques like single-cell analysis, data indicates that antibodies targeting highly abundant epitopes perform better regardless of concentration, while optimal dilution depends on balancing signal strength against economic considerations .
Proper experimental controls are crucial for accurate interpretation of results:
Positive controls:
Wild-type Arabidopsis tissue with known expression of At2g43620
Recombinant At2g43620 protein (if available)
Tissues with confirmed high expression levels
Negative controls:
Procedural controls:
Secondary antibody only (omitting primary antibody)
Primary antibody pre-absorbed with immunizing peptide
Blocking peptide competition assay
Normalization controls:
Housekeeping proteins (tubulin, actin) for Western blots
Total protein staining (Ponceau S, SYPRO Ruby) as loading controls
Internal reference proteins with stable expression
Application-specific controls:
For immunoprecipitation: pre-immune serum or IgG controls
For immunofluorescence: autofluorescence controls and secondary-only controls
For ChIP experiments: input controls and IgG controls
According to established guidelines, direct binding assays should include chemically similar but antigenically unrelated compounds as negative antigen controls when available .
Using At2g43620 antibody for ChIP requires specific considerations:
Experimental design:
Design positive control primers targeting known binding regions
Include negative control regions (gene deserts or unexpressed genes)
Plan appropriate replicates to ensure statistical validity
Sample preparation:
Optimize crosslinking conditions (1% formaldehyde, 10-15 minutes)
Ensure efficient cell lysis and chromatin fragmentation
Verify DNA fragment size (200-500 bp optimal for most applications)
Immunoprecipitation optimization:
Pre-clear chromatin with protein A/G beads to reduce background
Determine optimal antibody amount through titration experiments
Include appropriate controls (IgG, input DNA)
Washing and elution:
Use increasingly stringent wash buffers to reduce non-specific binding
Monitor wash efficiency through pilot experiments
Optimize elution conditions to maximize recovery
Analysis approaches:
qPCR for targeted analysis of specific genomic regions
ChIP-seq for genome-wide binding site identification
Integrate with RNA-seq data to correlate binding with expression
The quality of ChIP results depends critically on antibody specificity and affinity for the native protein in crosslinked chromatin, requiring validation with known positive controls before proceeding to genome-wide analyses.
Adapting At2g43620 antibody for single-cell analysis requires specialized approaches:
Oligo-conjugation strategies:
Conjugate antibodies with DNA barcodes for CITE-seq or similar technologies
Validate conjugated antibodies against unconjugated versions
Confirm that conjugation doesn't compromise binding properties
Concentration optimization:
Protocol adaptations:
Background assessment and mitigation:
Analysis considerations:
Set appropriate thresholds based on background assessment
Consider the distribution of signals in positive and negative populations
Apply appropriate normalization and batch correction methods
Optimizing antibody panels for single-cell analysis can yield significant improvements, with research showing properly adjusted concentrations can increase median positive signal by 57% while reducing background by 43% .
Resolving discrepancies requires systematic troubleshooting:
Antibody-related factors:
Different antibodies may recognize distinct epitopes with varying accessibility
Antibody performance may differ between denatured (Western) and native (IP) conditions
Lot-to-lot variability can affect consistency of results
Sample preparation differences:
Protein extraction methods influence protein solubility and epitope exposure
Fixation protocols for immunohistochemistry affect antigen preservation
Buffer compositions may preserve or disrupt protein-protein interactions
Technical considerations:
Detection sensitivity varies across methods (Western blot vs. immunofluorescence)
Signal amplification approaches alter detection thresholds
Image acquisition parameters impact apparent signal intensity
Biological variability:
Protein modifications may mask epitopes in context-dependent manner
Protein interactions might block antibody access in some conditions
Expression levels vary with developmental stage and environmental conditions
Systematic approach to reconciliation:
Use multiple antibodies targeting different epitopes
Apply orthogonal detection methods (mass spectrometry)
Implement controlled experiments varying one factor at a time
When discrepancies persist, they often reveal important biological insights about protein regulation, modification, or complex formation that merit further investigation.
Rigorous quantitative analysis requires systematic approaches:
Image acquisition:
Capture images within the linear range of detection
Use consistent exposure settings across all samples
Include standard curves when possible for absolute quantification
Software-based quantification:
Use dedicated analysis software (ImageJ, Image Lab, etc.)
Apply consistent background subtraction methods
Define regions of interest systematically across samples
Normalization strategies:
Normalize to loading controls (housekeeping proteins)
Consider total protein normalization (Ponceau S, SYPRO Ruby)
Account for lane-to-lane variations in transfer efficiency
Statistical analysis:
Perform experiments with sufficient biological replicates (minimum n=3)
Apply appropriate statistical tests based on data distribution
Report both raw and normalized data with error bars
Validation approaches:
Confirm key findings with alternative detection methods
Use knockout/knockdown controls to validate specificity
Consider absolute quantification with purified protein standards
Quantitative Western blot analysis should always be performed within the linear range of detection, as signal saturation can lead to underestimation of differences between samples.
High background is a common challenge that can be addressed through systematic optimization:
Antibody dilution adjustment:
Blocking optimization:
Test different blocking agents (milk, BSA, commercial blockers)
Increase blocking time (2 hours to overnight)
Add 0.1-0.3% Tween-20 to reduce hydrophobic interactions
Washing modifications:
Increase number and duration of wash steps
Use higher salt concentration in wash buffers (150-500 mM NaCl)
Consider adding low concentrations of detergents to wash buffers
Sample preparation improvements:
Pre-clear lysates with Protein A/G beads
Filter samples to remove aggregates
Optimize protein extraction to reduce interfering compounds
Technical adjustments:
Reduce secondary antibody concentration
Use fresh reagents and buffers
Optimize incubation temperatures
For single-cell analysis techniques, background signal can be assessed using empty droplets, with research showing markers exhibiting high background generally have high UMI cutoffs regardless of whether they show cell-type-specific signal .
Reconciling contradictory results requires systematic investigation:
Antibody characterization:
Confirm antibody specificity with appropriate controls
Check for lot-to-lot variation if using commercial antibodies
Compare results using antibodies targeting different epitopes
Experimental variables:
Document and standardize all buffer compositions
Control environmental conditions (temperature, humidity)
Standardize timing for critical steps
Sample-specific factors:
Account for developmental stages of plant tissues
Consider growth conditions that might affect protein expression
Evaluate potential post-translational modifications
Technical considerations:
Assess detection method sensitivity limits
Verify equipment calibration and performance
Implement standard operating procedures for consistency
Biological interpretation:
Consider context-dependent protein regulation
Investigate potential protein isoforms or modifications
Examine tissue-specific expression patterns
When contradictions persist despite thorough troubleshooting, they often reveal genuine biological complexity rather than technical artifacts, potentially uncovering novel regulatory mechanisms worthy of focused investigation.