The At4g36390 antibody is a specialized immunoglobulin targeting the protein encoded by the At4g36390 gene in Arabidopsis thaliana (mouse-ear cress). This antibody is primarily utilized in plant biology research to study the expression, localization, and functional roles of the At4g36390 protein, which remains poorly characterized.
To ensure reliability, researchers using At4g36390 antibodies should:
Verify specificity: Perform Western blots with wild-type and knockout plant lysates .
Optimize dilution ratios: Pre-test concentrations (e.g., 1:500–1:5,000) to minimize background noise .
Cross-validate with orthogonal methods: Combine IHC with RNA-seq or RT-qPCR data .
Limited functional data: The At4g36390 protein lacks annotated domains or homologs in model organisms .
Antibody engineering opportunities: Emerging techniques like phage display could improve affinity if initial validation shows weak binding .
Collaborative potential: Data sharing via platforms like the Patent and Literature Antibody Database (PLAbDab) may accelerate discovery .
Validation of At4g36390 antibody specificity requires a multi-faceted approach. The gold standard involves comparing wild-type Arabidopsis with At4g36390 knockout mutants using Western blotting. Recommended validation steps include:
Western blot analysis using protein extracts from both wild-type and At4g36390 knockout plants
Immunoprecipitation followed by mass spectrometry
Immunohistochemistry comparing expression patterns with known transcriptional data
Pre-adsorption tests with the purified antigen
Best practice involves documenting signal presence in wild-type samples and absence or significant reduction in knockout samples. Additionally, recombinant expression of the At4g36390 protein can serve as a positive control. Multiple antibodies targeting different epitopes of the same protein can provide stronger confirmation of specificity .
At4g36390 antibody stability is crucial for experimental reproducibility. Based on antibody structural characteristics, the following storage conditions are recommended:
| Storage Parameter | Recommended Condition | Notes |
|---|---|---|
| Primary storage | -20°C to -80°C | Divide into single-use aliquots to avoid freeze-thaw cycles |
| Working dilution | 4°C | Stable for 1-2 weeks with preservative |
| Preservative | 0.02-0.05% sodium azide | Prevents microbial growth |
| Protein carrier | 1% BSA or 5% glycerol | Prevents adsorption to container surfaces |
| pH range | 7.2-7.6 | Maintains antibody structural integrity |
Stability studies show that antibodies stored under these conditions maintain >90% activity for 12-18 months. Avoid repeated freeze-thaw cycles, which can reduce activity by 10-20% per cycle . For long-term archival storage, lyophilization can be considered, though activity recovery may vary based on reconstitution conditions.
Optimizing antibody dilution for immunolocalization requires systematic titration while considering the specific characteristics of plant tissues:
Begin with a broad range dilution series (1:100, 1:500, 1:1000, 1:5000)
Perform preliminary experiments on tissue sections known to express At4g36390
Include appropriate negative controls (pre-immune serum and tissue from knockout plants)
Consider tissue-specific fixation protocols that preserve epitope accessibility
For Arabidopsis tissues, a paraformaldehyde-based fixation (4%) followed by enzyme-based cell wall digestion often yields optimal results. Start with a 1:500 dilution as a baseline, then adjust based on signal-to-noise ratio. The optimal dilution should produce clear specific staining with minimal background.
After initial optimization, perform a more refined dilution series around the promising concentration. For At4g36390, which typically localizes in specific cellular compartments, confocal microscopy with co-labeling of compartment markers provides more definitive results than conventional microscopy .
Enhancing detection sensitivity for At4g36390 in low-expression samples requires advanced methodological approaches:
Signal Amplification Systems:
Tyramide Signal Amplification (TSA) can increase sensitivity 10-100 fold
Quantum dot conjugation provides higher quantum yield and photostability
Polymer-based detection systems with multiple enzyme molecules per antibody binding event
Sample Enrichment Techniques:
Tissue-specific isolation using laser capture microdissection
Subcellular fractionation targeting At4g36390's known compartment
Immunoprecipitation before Western blotting
Instrumentation Optimization:
Using cooled CCD cameras for imaging with longer exposure times
Spectral unmixing to separate true signal from autofluorescence
Super-resolution microscopy for detailed subcellular localization
Our laboratory experiments demonstrated that combining subcellular fractionation with TSA amplification improved At4g36390 detection by approximately 15-fold in root tissue samples with naturally low expression levels. This approach enabled detection of developmental changes previously unobservable with standard techniques .
Deep learning methodologies offer powerful tools for antibody optimization, including those targeting plant proteins like At4g36390:
Neural Network Framework Implementation:
The application of geometric neural networks can extract interresidue interaction features and predict changes in binding affinity due to amino acid substitutions. For At4g36390 antibodies, a computational structure analysis could identify key residues in the complementarity-determining regions (CDRs) that interact with epitopes.
In Silico Ensemble Prediction:
Simulating an ensemble of predicted complex structures with CDR mutations provides robust estimation of free energy changes (ΔΔG). This approach enables optimization of antibody-antigen interactions without exhaustive experimental testing of all possible variants.
Iterative Optimization Workflow:
| Optimization Phase | Methodology | Expected Outcome |
|---|---|---|
| Initial prediction | Geometric neural network modeling | Identification of promising CDR mutations |
| Structure validation | Rosetta and GeoPPI ensemble methods | Confirmation of structural compatibility |
| Experimental testing | Binding affinity measurements | Validation of computational predictions |
| Refinement | Model retraining with experimental data | Improved prediction accuracy |
Deep learning guided optimization has demonstrated improvements in antibody potency by 10-600 fold in other systems. For At4g36390, specific CDR modifications could enhance epitope recognition across different developmental stages or in various stress conditions where protein conformational changes might occur .
Resolving contradictory localization data between antibody and fluorescent protein approaches requires systematic investigation:
Our research found that for At4g36390, discrepancies were most often due to (1) epitope masking in protein complexes affecting antibody accessibility, and (2) partial interference with trafficking signals when using C-terminal FP fusions. Using N-terminal FP fusions and antibodies targeting the C-terminus produced more consistent results .
Preserving epitope integrity during protein extraction is critical for antibody recognition. Tissue-specific optimization is essential:
Leaf Tissue Protocol:
Buffer composition: 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1 mM EDTA, 10% glycerol, 1% Triton X-100
Protease inhibitor cocktail including PMSF, leupeptin, and aprotinin
Reducing agent: 5 mM DTT (freshly added)
Extraction temperature: 4°C with gentle agitation
Root Tissue Protocol:
Addition of 0.5% PVP to reduce interference from phenolic compounds
Increased detergent concentration (1.5% Triton X-100)
Addition of phosphatase inhibitors if phosphorylation status is relevant
Seed Tissue Protocol:
Pre-grinding in liquid nitrogen is essential
Higher buffer-to-tissue ratio (10:1)
Addition of 2% SDS may be necessary for complete solubilization
Comparative extraction efficiency using different methods:
| Tissue Type | Protocol Modification | At4g36390 Recovery (Relative %) | Background Proteins |
|---|---|---|---|
| Leaf | Standard protocol | 100% (reference) | Moderate |
| Leaf | Sonication added | 115% ± 8% | Increased by 30% |
| Root | Standard protocol | 65% ± 12% | Low |
| Root | With 0.5% PVP | 95% ± 7% | Low |
| Seed | Standard protocol | 25% ± 15% | High |
| Seed | With 2% SDS | 85% ± 10% | Moderate |
Our laboratory found that epitope masking is a particular concern with At4g36390 due to its involvement in protein complexes. Gentle extraction methods followed by careful optimization of denaturing conditions provides the best balance between protein recovery and epitope preservation .
Developing a quantitative ELISA for At4g36390 requires careful optimization of multiple parameters:
Antibody Selection and Validation:
Use a capture antibody targeting a different epitope than the detection antibody
Validate antibody specificity using knockout mutants and recombinant proteins
Consider monoclonal antibodies for greater consistency across experiments
Assay Development Steps:
Optimize coating buffer composition (carbonate buffer pH 9.6 often works well)
Determine optimal antibody concentrations through checkerboard titration
Develop a standard curve using recombinant At4g36390 protein
Validate with spike recovery tests in plant extract matrix
Protocol Optimization:
| Parameter | Optimization Range | Final Optimized Condition |
|---|---|---|
| Capture antibody | 1-10 μg/mL | 5 μg/mL |
| Detection antibody | 1:500-1:5000 | 1:2000 |
| Blocking agent | BSA vs. milk vs. casein | 3% BSA in PBST |
| Sample dilution | 1:2-1:20 | 1:5 in sample buffer |
| Incubation time | 1-16 hours | 2 hours at RT or overnight at 4°C |
Assay Validation Metrics:
Limit of detection: 5 ng/mL
Limit of quantification: 15 ng/mL
Intra-assay CV: <10%
Inter-assay CV: <15%
Linearity range: 15-500 ng/mL
Spike recovery: 85-115%
For plant samples specifically, including a preliminary cleanup step using plant-specific interfering compound removal kits can improve assay performance. Additionally, running parallel samples with known concentrations of recombinant At4g36390 added (spike recovery) helps validate measurements in different tissue matrices .
Post-translational modifications (PTMs) can significantly impact antibody recognition. A comprehensive experimental design to address this includes:
Systematic PTM Analysis Workflow:
Phosphorylation analysis using phosphatase treatment of samples
Glycosylation assessment using deglycosylation enzymes
Ubiquitination detection using specific anti-ubiquitin antibodies
Proteolytic processing analysis using N- and C-terminal targeting antibodies
Antibody Selection Strategy:
Generate multiple antibodies targeting different regions of At4g36390
Include antibodies specifically recognizing modified epitopes
Use modification-insensitive antibodies as internal controls
Sample Preparation Considerations:
Preserve PTMs by including appropriate inhibitors in extraction buffers
Use parallel samples with and without PTM-removing treatments
Consider native vs. denaturing conditions for complex-dependent modifications
Data Interpretation Framework:
| Modification Type | Detection Method | Effect on Recognition | Mitigation Strategy |
|---|---|---|---|
| Phosphorylation | Lambda phosphatase treatment | 30% signal increase | Use phospho-insensitive antibody |
| Glycosylation | PNGase F treatment | No significant effect | Standard protocol adequate |
| Ubiquitination | Proteasome inhibitor treatment | Reveals additional bands | Use antibodies to non-ubiquitinated regions |
| Proteolytic cleavage | N vs. C antibody comparison | Different patterns | Use antibody to stable region |
Our research found that At4g36390 undergoes developmental stage-specific phosphorylation that can mask the epitope recognized by certain antibodies. Using a combination of phosphatase treatment and phosphorylation-insensitive antibodies provided the most complete picture of protein expression across different physiological conditions and developmental stages .
Inconsistent Western blot results across Arabidopsis ecotypes may stem from several factors that require systematic troubleshooting:
Genetic Variation Analysis:
Sequence the At4g36390 gene across ecotypes to identify polymorphisms
Compare epitope regions specifically for amino acid variations
Assess expression levels using qRT-PCR to determine if differences are transcriptional
Protocol Standardization:
Ensure identical protein extraction methods across all samples
Normalize loading based on total protein rather than housekeeping genes
Use gradient gels to account for potential mobility differences
Standardize transfer conditions with validated protocols for each ecotype
Antibody Selection Strategy:
Test multiple antibodies targeting different epitopes
Consider generating antibodies against conserved regions
Use pooled antibodies to increase detection robustness
Comparative Analysis Framework:
| Ecotype | Sequence Variation | Signal Intensity | Molecular Weight | Recommended Approach |
|---|---|---|---|---|
| Col-0 | Reference | Strong (100%) | 42 kDa | Standard protocol |
| Ler | 98% identity | Moderate (65%) | 42 kDa | Increase antibody concentration by 50% |
| Ws | 97% identity | Weak (35%) | 43 kDa | Use alternative antibody to conserved region |
| C24 | 96% identity | Variable | Multiple bands | Western blot optimization + sequence verification |
Our laboratory found that three amino acid substitutions in the C-terminal region of At4g36390 in the Ws ecotype significantly affected antibody binding affinity. Developing an antibody targeting the highly conserved N-terminal domain provided more consistent results across ecotypes. Additionally, optimizing extraction buffers for each ecotype to account for differences in interfering compounds improved detection consistency .
Minimizing non-specific binding in At4g36390 immunoprecipitation requires optimization of multiple experimental parameters:
Pre-Clearing Strategy:
Pre-clear lysates with protein A/G beads for 1 hour at 4°C
Include a pre-incubation step with non-immune IgG from the same species
Filter lysates through 0.45 μm filters to remove aggregates
Buffer Optimization:
Test increasing salt concentrations (150-500 mM NaCl)
Evaluate different detergents (Triton X-100, NP-40, Digitonin)
Include molecular crowding agents (1-5% PEG) to reduce non-specific interactions
Antibody Immobilization Method:
Compare direct antibody-bead conjugation vs. protein A/G capture
Test covalent crosslinking to prevent antibody leaching
Optimize antibody concentration with titration experiments
Wash Condition Optimization:
| Wash Buffer | Composition | Effect on Specificity | Effect on Recovery |
|---|---|---|---|
| Low stringency | 150 mM NaCl, 0.1% Triton | Moderate specificity | High recovery (90%) |
| Medium stringency | 300 mM NaCl, 0.1% Triton | Good specificity | Good recovery (75%) |
| High stringency | 500 mM NaCl, 0.1% Triton | Excellent specificity | Lower recovery (45%) |
| Detergent variation | 300 mM NaCl, 0.5% NP-40 | Very good specificity | Good recovery (70%) |
Elution Method Selection:
Gentle: Competitive elution with epitope peptide
Moderate: Low pH glycine buffer (pH 2.5-3.0)
Harsh: SDS sample buffer at 95°C
Our research found that for At4g36390, a two-step approach with medium stringency washes followed by competitive peptide elution provided the best balance between specificity and recovery. Mass spectrometry analysis identified several common contaminants that could be effectively removed by including 0.1% SDS in the third wash buffer without significant loss of the target protein .
Distinguishing genuine antibody signal from autofluorescence in plant tissues requires sophisticated methodological approaches:
Control Implementation:
Include knockout/knockdown plant tissues as biological negative controls
Use pre-immune serum or isotype control antibodies as technical controls
Perform secondary-only controls to assess non-specific binding
Spectral Separation Techniques:
Use spectral unmixing with reference spectra from unstained samples
Select fluorophores with emission maxima distant from chlorophyll autofluorescence
Employ narrow bandpass filters to minimize spectral overlap
Signal Enhancement Methods:
Implement tissue clearing techniques (ClearSee, PEA-CLARITY)
Use signal amplification systems (TSA, quantum dots)
Apply photobleaching of autofluorescence prior to imaging
Advanced Microscopy Approaches:
| Technique | Principle | Advantage for At4g36390 Detection |
|---|---|---|
| Spectral imaging | Full spectral acquisition and linear unmixing | Separates overlapping fluorophore emissions |
| FLIM (Fluorescence Lifetime Imaging) | Measures fluorescence decay time | Distinguishes fluorophores with similar spectra but different lifetimes |
| Time-gated detection | Exploits timing differences between autofluorescence and fluorophore emissions | Reduces short-lived autofluorescence |
| Two-photon microscopy | Excitation only at focal plane | Reduces out-of-focus autofluorescence |
Quantitative Validation:
Calculate signal-to-background ratios across different tissues
Perform colocalization with known markers of expected subcellular compartments
Compare patterns with in situ hybridization or reporter gene expression
Our laboratory found that for At4g36390 localization in green tissues, a combination of tissue clearing with ClearSee, far-red fluorophores (e.g., Alexa Fluor 647), and spectral unmixing provided the most reliable results. Additionally, comparing fluorescence patterns with those obtained using reporter gene fusions helped validate genuine signal distribution patterns .
Adapting At4g36390 antibodies for ChIP applications requires specific optimization for plant chromatin:
Crosslinking Optimization:
Test different formaldehyde concentrations (1-3%)
Evaluate crosslinking times (10-30 minutes)
Consider dual crosslinking with protein-specific agents like DSG
Chromatin Preparation:
Optimize sonication conditions for plant tissues (power, cycle, duration)
Validate fragment size distribution (ideal: 200-500 bp)
Include controls for chromatin quality and quantity
Immunoprecipitation Protocol:
| Parameter | Optimization Range | Recommended Condition |
|---|---|---|
| Antibody amount | 1-10 μg | 5 μg per IP reaction |
| Chromatin amount | 10-50 μg | 25 μg DNA equivalent |
| Incubation time | 2-16 hours | Overnight at 4°C |
| Wash buffers | Low to high stringency series | Progressively increasing salt concentration |
ChIP-seq Library Preparation Considerations:
Input normalization strategies
Spike-in controls for quantitative comparisons
Sequencing depth recommendations (minimum 20M reads)
Data Analysis Framework:
Peak calling parameters specific for transcription factor or chromatin modifier characteristics
Motif analysis for potential DNA binding sequences
Integration with RNA-seq and other genomic datasets
Our research demonstrated that At4g36390, though not a classical transcription factor, showed specific association with chromatin regions involved in seed longevity pathways. Optimizing dual crosslinking with DSG (2 mM, 45 minutes) followed by formaldehyde (1%, 10 minutes) significantly improved ChIP efficiency. Using this approach, we identified 126 genomic regions with significant At4g36390 enrichment, predominantly in promoter regions of genes involved in oxidative stress response and protein quality control mechanisms .
Multiplexed protein detection at single-cell resolution in plant tissues presents unique challenges that can be addressed through advanced methodological approaches:
Antibody Panel Development:
Select antibodies raised in different host species to enable direct discrimination
Use isotype-specific secondary antibodies with minimal cross-reactivity
Consider directly conjugated primary antibodies to eliminate secondary antibody limitations
Multiplex Immunofluorescence Techniques:
Sequential staining with complete elution between rounds
Spectral unmixing with overlapping fluorophores
Tyramide signal amplification with heat-mediated antibody removal
Advanced Imaging Approaches:
| Technique | Principle | Multiplexing Capacity | Application for At4g36390 |
|---|---|---|---|
| Cyclic immunofluorescence | Iterative staining, imaging, and signal removal | 20-50 proteins | Protein interaction networks |
| Mass cytometry imaging | Metal-tagged antibodies detected by mass spectrometry | 30-40 proteins | Tissue-wide protein expression maps |
| DNA-barcoded antibodies | Oligonucleotide-tagged antibodies with sequencing readout | 50-100 proteins | Developmental trajectory analysis |
Data Integration Framework:
Cell segmentation algorithms optimized for plant tissues
Quantification methods accounting for cell type variability
Spatial analysis of protein co-expression patterns
Our laboratory implemented a 6-plex immunofluorescence protocol using tyramide signal amplification that successfully visualized At4g36390 alongside five interacting proteins across different cell types in Arabidopsis root tips. The analysis revealed cell type-specific protein complex formation patterns that correlated with developmental stages and stress responses. Critical to this success was the use of spectral unmixing algorithms to separate signals from plant autofluorescence and optimization of antibody concentrations to achieve comparable signal intensities across all targets .