The AT4G04930 gene encodes a delta4-desaturase, an enzyme that introduces double bonds into sphingolipid fatty acid chains. This modification is essential for regulating membrane fluidity and signaling pathways in plants .
| Property | Value | Source |
|---|---|---|
| Gene ID | AT4G04930 | |
| Protein Name | Sphingolipid delta4-desaturase | |
| Uniprot ID | Q9ZPH4 | |
| Species | Arabidopsis thaliana | |
| Antibody Type | Polyclonal | |
| Antibody Format | Liquid (2ml/0.1ml) |
While not directly related to At4g04930, broader antibody studies highlight key principles relevant to its use:
Fc Engineering: Modifications to antibody Fc regions (e.g., IgG1 vs. IgG4) influence immune effector functions, as seen in cancer immunotherapy .
Polyreactivity: Antibodies with flexible CDR3 regions may bind multiple antigens, affecting specificity in assays .
Developability: Therapeutic antibodies are optimized for stability (e.g., Tm ≥68.5°C) and low aggregation .
Current data on the At4g04930 Antibody is largely product-oriented. Future studies could:
Validate Specificity: Confirm cross-reactivity with homologs in other plant species.
Functional Assays: Use the antibody to inhibit delta4-desaturase activity in vivo and assess lipidome changes.
Stress-Response Profiling: Map lipid remodeling during pathogen infection or osmotic stress.
At4g04930 is annotated as a fatty acid desaturase family protein in Arabidopsis thaliana . This protein belongs to a class of enzymes involved in lipid metabolism, specifically in the modification of fatty acid chains through the introduction of double bonds. Researchers develop antibodies against such proteins for several critical research applications: (1) to study protein localization within cellular compartments, (2) to quantify protein expression levels under different experimental conditions, (3) to investigate protein-protein interactions through co-immunoprecipitation experiments, and (4) to examine post-translational modifications. For membrane-associated proteins like desaturases, antibodies provide one of the few reliable methods to study their expression patterns and regulatory mechanisms.
For optimal detection of membrane-associated proteins like At4g04930, researchers should employ a multi-step protein extraction protocol:
Harvest fresh plant tissue (preferably 7-day-old seedlings for highest expression) and grind in liquid nitrogen
Extract using a membrane protein-optimized buffer (50mM HEPES-KOH buffer containing 250mM sucrose, 5% glycerol, 50mM NaPP, 1mM NaMo, 25mM NaF, 10mM EDTA, 0.5% PVP, 3mM DTT, 1mM PMSF, with protease inhibitors)
Fractionate through differential centrifugation to separate soluble (S100) and microsomal (P100) fractions at 100,000 × g for 30 minutes at 4°C
Enrich membrane proteins using detergent solubilization (1% n-dodecyl-β-D-maltoside works well for many plant membrane proteins)
Denature samples at moderate temperatures (65°C for 5 minutes rather than boiling) to prevent aggregation of membrane proteins
This protocol minimizes protein degradation while maximizing recovery of membrane-associated proteins that are typically present at lower abundance than soluble proteins.
Antibody validation requires multiple complementary approaches to ensure specificity:
Western blot analysis using wild-type and knockout/knockdown lines: Compare protein detection between wild-type Arabidopsis and mutant lines with confirmed knockout or knockdown of At4g04930. Absence or significant reduction of the target band in mutant samples provides strong evidence for antibody specificity .
Pre-absorption controls: Incubate the antibody with the immunizing peptide or recombinant protein prior to immunodetection. This should abolish or significantly reduce signal if the antibody is specific.
Heterologous expression system validation: Express the At4g04930 protein with an epitope tag (e.g., FLAG, His, or GFP) in a heterologous system, then perform parallel detection with both the developed antibody and an antibody against the epitope tag.
Mass spectrometry verification: Immunoprecipitate the protein using the antibody and confirm identity through mass spectrometry analysis.
Cross-reactivity testing: Test the antibody against protein extracts from related plant species to assess specificity against orthologs and potential cross-reactivity with other desaturase family members.
A comprehensive Western blot experiment for At4g04930 should include the following controls:
| Control Type | Purpose | Implementation |
|---|---|---|
| Positive control | Confirms antibody functionality | Recombinant At4g04930 protein or extract from tissues with known high expression |
| Negative control | Confirms specificity | Extract from At4g04930 knockout line (if available) |
| Loading control | Normalizes for protein loading differences | Antibody against a housekeeping protein (e.g., actin, GAPDH) |
| Subcellular fraction control | Confirms proper fractionation | Antibodies against known compartment markers (e.g., BiP for ER, H+-ATPase for plasma membrane) |
| Peptide competition | Verifies epitope specificity | Antibody pre-incubated with immunizing peptide |
| Cross-reactivity control | Assesses potential false positives | Extracts from non-target tissues or related Arabidopsis genes (other desaturases) |
Proper implementation of these controls helps distinguish true signals from artifacts and enables accurate interpretation of experimental results in antibody-based detection assays.
Optimizing immunolocalization for membrane proteins like At4g04930 requires careful attention to fixation and permeabilization steps:
Fixation optimization: Test both aldehyde-based (4% paraformaldehyde) and solvent-based (methanol/acetone) fixation methods, as membrane proteins may require different approaches than soluble proteins.
Antigen retrieval: For paraffin-embedded sections, incorporate a citrate buffer (pH 6.0) heat-mediated antigen retrieval step to expose epitopes potentially masked during fixation.
Permeabilization optimization: Test a gradient of detergent concentrations (0.1-0.5% Triton X-100 or 0.05-0.1% saponin) to determine optimal membrane permeabilization without destroying antigenicity.
Blocking optimization: Use 3-5% BSA with 0.1% cold fish skin gelatin to reduce background while maintaining specific binding.
Antibody concentration: Perform a titration series (1:100 to 1:5000) to determine optimal primary antibody concentration that maximizes specific signal while minimizing background.
Signal amplification: For low-abundance proteins, incorporate tyramide signal amplification or quantum dot-based detection systems to enhance visualization of weak signals.
Co-localization markers: Include antibodies against known subcellular compartment markers to verify expected localization patterns.
Success can be evaluated using positive controls of known localization pattern and negative controls (pre-immune serum or secondary antibody only).
Discrepancies between protein and transcript levels are common in biological research and may reflect important regulatory mechanisms. When At4g04930 protein levels (detected by antibody) do not correlate with mRNA levels (detected by RT-PCR), researchers should systematically investigate:
Post-transcriptional regulation: Examine microRNA-mediated regulation by using target prediction tools and validating through reporter assays. Based on similar studies of desaturase family proteins, miRNA regulation can cause substantial differences between transcript and protein levels.
Protein stability differences: Perform cycloheximide chase assays to determine protein half-life under different conditions, as protein stability can vary significantly between treatments while transcript levels remain similar.
Translational efficiency: Conduct polysome profiling to assess whether the transcript is efficiently translated under various conditions.
Technical limitations: Validate both detection methods by using alternative approaches:
For protein: Try different antibodies if available or use targeted proteomics (SRM/MRM)
For transcript: Use different primer pairs and quantification methods (digital droplet PCR)
Temporal dynamics: Perform time-course experiments, as transcript and protein levels may peak at different times following stimulus.
Understanding these discrepancies often reveals important regulatory mechanisms affecting gene expression at different levels and should be viewed as an opportunity for deeper biological insights rather than simply technical obstacles.
Distinguishing specific from non-specific binding requires sophisticated approaches:
Two-dimensional Western blotting: Separate proteins first by isoelectric point and then by molecular weight to better resolve potential cross-reactive proteins sharing similar molecular weights.
Epitope competition assays with concentration gradients: Perform blocking with increasing concentrations of the immunizing peptide to demonstrate dose-dependent reduction in signal intensity for specific bands.
Analysis of band patterns across genetic variants: Compare antibody binding patterns between wild-type, heterozygous, and homozygous mutant lines to confirm genotype-dependent signal changes.
Sequential immunoprecipitation validation: Perform multiple rounds of immunoprecipitation to deplete the specific target and demonstrate corresponding signal reduction.
Orthogonal binding site antibodies: Use multiple antibodies raised against different epitopes of the same protein to confirm consistent detection.
Mass spectrometry analysis of immunoprecipitated proteins: Identify all proteins pulled down by the antibody to catalog potential cross-reactive proteins.
The most definitive demonstration of specificity comes from showing absence of signal in verified knockout lines alongside proper controls for loading and sample preparation.
Robust quantification of Western blot or immunohistochemistry data requires appropriate statistical methods:
Normalization approaches:
For Western blots: Normalize band intensity to total protein (using stain-free technology or Ponceau S) rather than single housekeeping proteins, which may vary across conditions
For immunohistochemistry: Use ratio-based methods comparing target protein to a stable reference protein within the same cell/tissue
Experimental design considerations:
Statistical analysis recommendations:
Apply log-transformation to Western blot densitometry data to better approximate normal distribution
Use ANOVA with appropriate post-hoc tests for multiple condition comparisons
Consider non-parametric alternatives (Kruskal-Wallis) when normality cannot be assumed
Report effect sizes (Cohen's d) alongside p-values to indicate biological significance
Present 95% confidence intervals rather than standard error to better represent uncertainty
Reproducibility practices:
Adapting antibodies for ChIP requires specialized approaches, particularly for membrane-associated proteins like At4g04930 that may have transcription factor activity:
Crosslinking optimization: Test dual crosslinking protocols using both formaldehyde (1%) and protein-specific crosslinkers (DSG, 2 mM) to capture indirect DNA associations.
Chromatin fragmentation: Optimize sonication parameters specifically for plant tissue to achieve 200-500 bp fragments, with careful monitoring via agarose gel electrophoresis.
Epitope availability assessment: Compare antibodies raised against different regions of At4g04930, as crosslinking may mask certain epitopes.
Stringency balancing: Test multiple wash buffer compositions with varying salt concentrations (150-500 mM NaCl) to determine optimal conditions that maintain specific interactions while reducing background.
Sequential ChIP approach: For proteins with weak or transient DNA interactions, consider tandem ChIP approaches where the initial immunoprecipitation is followed by a second round using the same or different antibody.
Control recommendations:
Input chromatin (pre-immunoprecipitation)
Non-specific IgG immunoprecipitation
Immunoprecipitation from knockout/knockdown lines
Biologically relevant negative control regions for qPCR validation
Data analysis considerations:
Use spike-in normalization with exogenous chromatin
Apply appropriate peak calling algorithms accounting for the expected binding pattern
The success of ChIP experiments should be validated using known or predicted DNA binding regions identified through bioinformatic analysis of the At4g04930 protein sequence.
Genetic compensation assessment: Examine expression of homologous desaturase family members in knockout lines using RT-qPCR and proteomics to identify potential compensatory mechanisms.
Antibody epitope verification: Confirm that the antibody epitope is truly absent in the knockout line through genomic PCR and sequencing of the target region, as some T-DNA insertions may allow partial protein expression.
Conditional phenotyping: Analyze protein expression and phenotypes under stress conditions (temperature, drought, salt) where redundancy may be reduced, revealing roles masked under standard conditions.
Temporal and spatial resolution: Use tissue-specific and inducible knockdown approaches (CRISPR interference or artificial microRNA) to overcome developmental compensation that may occur in constitutive knockouts.
Post-translational modification focus: Develop antibodies specific to modified forms of At4g04930 (phosphorylated, ubiquitinated) to determine if the relevant biological activity depends on specific protein states.
Proteomics validation: Perform targeted proteomics on wild-type and knockout lines to verify complete protein absence and identify any truncated versions or alternative isoforms.
Systematic validation in genetic backgrounds: Test antibody detection and phenotypes across multiple knockout line alleles and complementation lines to rule out background effects.
Optimizing co-immunoprecipitation (co-IP) for membrane proteins like At4g04930 requires specific adaptations:
Membrane protein solubilization optimization:
Test detergent panel (digitonin, DDM, CHAPS) at different concentrations
Consider using membrane-specific IP kits designed to maintain protein-protein interactions
Validate that solubilization conditions maintain known protein interactions as positive controls
Crosslinking considerations:
For transient interactions, incorporate reversible crosslinkers (DSP, DTBP)
Optimize crosslinker concentration (0.5-2 mM) and time (5-30 minutes) to balance capture efficiency with specificity
Controls and validation:
Perform reciprocal co-IPs when possible
Include negative controls (IgG, unrelated antibody, protein extract from knockout lines)
Use tagged version of At4g04930 (GFP, FLAG) for orthogonal validation
Analysis approaches:
For novel interactors, confirm with alternative methods (Y2H, BiFC, FRET)
Use quantitative proteomics (SILAC, TMT) to distinguish enriched proteins from background
Apply stringent statistical filtering (>2-fold enrichment, p<0.05, present in multiple replicates)
Biological relevance verification:
Test interaction under different physiological conditions (stress, developmental stages)
Perform domain mapping to identify interaction regions
Validate functional significance through genetic analysis of interaction partners
By combining these approaches, researchers can minimize false positives while identifying biologically relevant protein complexes involving At4g04930.
Quantitative analysis of immunofluorescence data requires rigorous methodology:
Image acquisition standardization:
Use identical microscope settings (exposure time, gain, offset) for all compared samples
Capture multiple representative fields per sample (minimum 5-10) selected using unbiased criteria
Include fluorescence calibration standards in imaging sessions
Systematic processing workflow:
Process all compared images identically (same thresholding algorithm, no individual image adjustments)
Use automated analysis pipelines in ImageJ/FIJI or CellProfiler to reduce bias
Document all processing steps for reproducibility
Quantification approaches:
Single-cell analysis: Measure fluorescence intensity within defined cellular compartments
Co-localization analysis: Calculate Pearson's or Manders' coefficients with proper controls
Spatial distribution analysis: Perform radial profile analysis from defined reference points
Statistical considerations:
Apply hierarchical statistical approaches that account for multiple cells per field and multiple fields per sample
Use appropriate transformations for fluorescence intensity data (log-transformation often required)
Incorporate biological replicate variation into statistical models
Visualization best practices:
Present representative images alongside quantification
Use colorblind-friendly lookup tables (avoid rainbow color maps)
Include scale bars and normalization information
Modern image analysis should leverage machine learning approaches for complex pattern recognition while maintaining transparent analysis parameters that can be shared with published results.
Multi-omics integration requires sophisticated approaches:
Data normalization strategies:
Apply platform-specific normalization first (e.g., quantile normalization for microarrays, TPM/FPKM for RNA-seq)
Consider batch effect correction using ComBat or similar tools
Implement data scaling methods appropriate for integration (z-scores, min-max scaling)
Correlation analysis approaches:
Calculate protein-transcript correlations using appropriate methods (Pearson, Spearman, or distance correlation)
Identify discordant patterns that may indicate post-transcriptional regulation
Develop time-lagged correlations to account for delays between transcription and translation
Pathway-based integration:
Map all dataset components to common pathway databases (KEGG, MapMan)
Perform overrepresentation analysis to identify enriched functional categories
Apply pathway topology tools that consider molecular interactions (SPIA, PathwayExpress)
Network analysis methods:
Construct correlation networks across omics layers
Use partial correlation methods to distinguish direct from indirect associations
Implement Bayesian network approaches to infer causal relationships
Visualization techniques:
Create multi-omics heatmaps with hierarchical clustering
Develop pathway visualizations with multi-omics overlay
Use dimension reduction techniques (PCA, t-SNE, UMAP) for integrated data exploration
Validation approaches:
Test predictions through targeted experiments
Implement leave-one-out validation of integration models
Compare findings to published literature on related desaturase family proteins
The most insightful integration analyses often focus on condition-specific or time-resolved datasets that capture dynamic processes related to fatty acid metabolism.
Several cutting-edge technologies show promise for advancing research on At4g04930 and other plant membrane proteins:
Proximity labeling approaches:
BioID or TurboID fusion proteins to identify proximity-based interactors in living cells
APEX2 technology for subcellular localization with electron microscopy-level resolution
Single-cell proteomics:
Adapting mass cytometry (CyTOF) for plant tissue analysis
Development of single-cell Western blot technologies for plant cells
Advanced microscopy techniques:
Super-resolution microscopy (STORM, PALM) for nanoscale localization
Lattice light-sheet microscopy for live-cell dynamics with minimal phototoxicity
CRISPR-based technologies:
CRISPRi for tuneable knockdown without complete protein elimination
CRISPR activation for overexpression studies
Base editing for introducing specific mutations without double-strand breaks
Computational approaches:
Enhanced structural prediction with AlphaFold2 for antibody epitope engineering
Machine learning integration of multi-omics datasets for functional prediction
Protein engineering strategies:
Split protein complementation systems adapted for membrane proteins
Optogenetic tools for temporal control of protein activity