ATL58 (Q570X5) is a protein found in Arabidopsis thaliana (Mouse-ear cress), a model organism widely used in plant molecular biology research. It belongs to the ATL (Arabidopsis Tóxicos en Levadura) family of proteins, which are RING-H2 type E3 ubiquitin ligases involved in various cellular processes including stress responses and development. The protein contains specific domains that facilitate protein-protein interactions and ubiquitination activities. When studying ATL58, it's important to consider its subcellular localization and potential interaction partners to properly interpret experimental results .
The ATL58 Antibody (CSB-PA681984XA01DOA) is a polyclonal antibody raised in rabbits against recombinant Arabidopsis thaliana ATL58 protein. Its specificity is limited to Arabidopsis thaliana samples. The antibody is supplied in liquid form, non-conjugated, and purified using antigen affinity purification methods. The storage buffer consists of 0.03% Proclin 300 as a preservative, 50% Glycerol, and 0.01M PBS at pH 7.4. Being a polyclonal IgG antibody, it recognizes multiple epitopes on the target protein, which can provide enhanced detection sensitivity in various applications .
The ATL58 Antibody has been specifically tested and validated for Enzyme-Linked Immunosorbent Assay (ELISA) and Western Blot (WB) applications. When employing this antibody in Western blotting, researchers should expect to detect the ATL58 protein at its predicted molecular weight, though post-translational modifications may affect observed migration patterns. It is important to note that the antibody has not been validated for other applications such as immunohistochemistry, immunofluorescence, or immunoprecipitation. Any use beyond the validated applications would require thorough optimization and validation by the researcher .
To maintain optimal activity, the ATL58 Antibody should be stored at either -20°C or -80°C immediately upon receipt. Repeated freeze-thaw cycles should be avoided as they can significantly degrade antibody quality and performance. When working with the antibody, it's advisable to aliquot it into smaller volumes before freezing to minimize freeze-thaw cycles. The storage buffer containing 50% glycerol helps stabilize the antibody during freeze-thaw, but proper handling remains essential for long-term preservation of activity .
When designing experiments using the ATL58 Antibody, several controls should be incorporated to ensure reliable interpretation of results:
Control Type | Description | Purpose |
---|---|---|
Positive Control | Arabidopsis thaliana wild-type tissue/cells expressing ATL58 | Confirms antibody functionality |
Negative Control | Non-Arabidopsis samples or ATL58 knockout lines | Assesses potential cross-reactivity |
Secondary Antibody Control | Samples treated with secondary antibody only | Evaluates non-specific binding |
Loading/Housekeeping Control | Detection of constitutively expressed proteins (e.g., actin, tubulin) | Normalizes for sample loading variations |
Pre-absorption Control | Antibody pre-incubated with immunizing peptide | Confirms specificity of detected signal |
These controls are essential for proper validation and interpretation of experimental results, especially when employing the antibody in novel contexts or with modified protocols .
To validate ATL58 Antibody specificity in your experimental system, apply a multi-pronged approach following the "five pillars" of antibody validation:
Genetic strategy: Utilize ATL58 knockout or knockdown Arabidopsis lines as negative controls. The absence or reduction of signal in these samples strongly supports antibody specificity. CRISPR-Cas9 or RNAi techniques can generate appropriate negative control samples.
Orthogonal strategy: Compare ATL58 protein levels detected by the antibody with mRNA levels measured by RT-qPCR. Correlation between protein and transcript levels under various conditions provides evidence for antibody specificity.
Independent antibody strategy: If available, compare results with a different antibody targeting ATL58 at a distinct epitope. Concordant results between different antibodies increase confidence in specificity.
Recombinant expression strategy: Overexpress tagged ATL58 and confirm detection by both the antibody and tag-specific detection methods. This approach can also help determine detection limits.
Immunoprecipitation-MS strategy: Perform immunoprecipitation followed by mass spectrometry to identify all proteins captured by the antibody. This comprehensive approach identifies potential cross-reactivity issues .
Documentation of these validation steps is critical for publication and ensuring reproducible research. Remember that antibody performance may vary across different experimental conditions, requiring validation for each specific application and context .
Optimizing Western blotting with ATL58 Antibody requires systematic adjustment of multiple parameters:
Sample preparation:
Extract proteins using buffers containing protease inhibitors to prevent degradation
Include reducing agents (e.g., DTT, β-mercaptoethanol) to ensure proper protein denaturation
Heat samples at 95°C for 5 minutes for complete denaturation
Antibody dilution optimization:
Begin with manufacturer's recommended dilution (typically 1:1000)
Test serial dilutions (e.g., 1:500, 1:1000, 1:2000) to determine optimal signal-to-noise ratio
Record precise antibody lot numbers for reproducibility
Blocking optimization:
Test different blocking agents (BSA, non-fat milk, commercial blockers)
Determine optimal blocking time and temperature
Consider using the same blocking agent in antibody dilution buffer
Membrane selection:
PVDF membranes typically provide better protein retention and sensitivity
Optimize transfer conditions for the predicted molecular weight of ATL58
Signal development:
For low abundance targets, consider enhanced chemiluminescence or fluorescent detection systems
Optimize exposure times for chemiluminescence detection
Document all optimization steps methodically in a laboratory notebook for reproducibility. Remember that optimization is an iterative process, and conditions may need adjustment for different experimental systems or sample types .
Non-specific binding is a common challenge when working with polyclonal antibodies. To troubleshoot and minimize this issue with ATL58 Antibody:
Adjust blocking conditions:
Increase blocking time (from 1 hour to overnight)
Test different blocking agents (5% BSA often reduces background compared to milk for phospho-specific detection)
Add 0.1-0.3% Tween-20 to washing and antibody incubation buffers
Optimize antibody concentration:
Dilute the antibody further if background is high
Reduce incubation time or temperature
Implement additional washing steps:
Increase the number and duration of washes
Use more stringent washing buffers (higher salt concentration or detergent)
Pre-absorb the antibody:
Incubate with proteins from non-target species or tissues
Use lysates from ATL58 knockout plants to pre-absorb non-specific antibodies
Cross-adsorption technique:
Identify the molecular weight of non-specific bands
Use knockout verification to confirm which bands are specific
If possible, cut membranes to isolate target molecular weight regions
For complex plant samples with high endogenous peroxidase activity, consider hydrogen peroxide pre-treatment or alternative detection methods. Document all troubleshooting steps methodically to establish a reproducible protocol for your specific experimental system .
For accurate quantification of ATL58 expression, several methodological approaches are recommended:
Densitometry analysis:
Capture images within the linear dynamic range of detection
Use analysis software (ImageJ, ImageLab) to quantify band intensity
Always normalize to loading controls (actin, tubulin, total protein stain)
Include a standard curve with known quantities of recombinant protein
Quantitative immunoblotting:
Use fluorescently-labeled secondary antibodies for more accurate quantification
Include a dilution series of control samples to ensure measurements within linear range
Employ multiplexing to detect target and loading control simultaneously
ELISA-based quantification:
Develop a sandwich ELISA using ATL58 Antibody as capture or detection antibody
Generate a standard curve using recombinant ATL58 protein
Ensure sample preparation methods maintain native protein conformation
Normalization approaches:
For accurate quantification across multiple samples, normalize to:
Total protein (measured by stain-free technology or Ponceau S)
Housekeeping proteins (after validating their stability under your experimental conditions)
Multiple reference proteins rather than a single housekeeping gene
Quantification Method | Advantages | Limitations | Best Application |
---|---|---|---|
Western Blot Densitometry | Simple, widely available | Semi-quantitative, narrow dynamic range | Relative expression changes |
Fluorescent Western | Higher dynamic range, more precise | Requires specialized equipment | Accurate relative quantification |
ELISA | High throughput, good sensitivity | Requires native protein conformation | Absolute quantification |
Document all quantification parameters, including exposure settings, analysis methods, and normalization strategies, to ensure reproducibility and reliable comparative analyses across experiments .
The polyclonal nature of the ATL58 Antibody has several significant implications for experimental design and data interpretation:
Epitope recognition diversity:
Polyclonal antibodies recognize multiple epitopes on the target protein
Advantages: Enhanced sensitivity, better tolerance to minor protein denaturation or modification
Limitations: Increased potential for cross-reactivity with similar epitopes on non-target proteins
Lot-to-lot variability:
Different production lots may contain varying antibody compositions
Critical consideration: Document lot numbers for reproducibility
Recommendation: Purchase sufficient quantity of a single lot for entire project duration
Post-translational modification detection:
Some antibodies in the polyclonal mixture may recognize epitopes affected by PTMs
This can result in detecting multiple bands corresponding to different forms of ATL58
Verification approach: Use dephosphorylation or deglycosylation treatments to confirm band identity
Signal interpretation:
Higher sensitivity may detect low-abundance forms of the protein not visible with monoclonal antibodies
May require additional controls to distinguish specific from non-specific signals
Consider complementary approaches for critical findings (e.g., mass spectrometry)
To mitigate these challenges, implement thorough validation using genetic controls (knockout/knockdown lines), include reciprocal confirmation with orthogonal techniques, and maintain consistent experimental conditions across studies. For publications, explicitly report antibody validation methods, lot numbers, and any observed limitations in specificity or reproducibility .
While the ATL58 Antibody has not been specifically validated for immunofluorescence applications, researchers may adapt it for this purpose following these best practices:
Sample preparation optimization:
Test multiple fixation methods (4% paraformaldehyde, methanol, acetone)
Evaluate different permeabilization protocols (0.1-0.5% Triton X-100, saponin)
Consider antigen retrieval techniques if working with fixed tissue sections
Antibody validation for IF:
Begin with positive controls (tissues known to express ATL58)
Include negative controls (ATL58 knockout tissue or non-plant tissue)
Compare staining patterns with published subcellular localization data
Signal enhancement techniques:
Implement tyramide signal amplification for low-abundance targets
Use high-sensitivity detection systems (e.g., quantum dots, highly cross-adsorbed secondaries)
Optimize signal-to-noise ratio through careful titration of primary and secondary antibodies
Counterstaining and colocalization:
Use well-established organelle markers to determine subcellular localization
Employ nuclear counterstains (DAPI, Hoechst) for orientation
Apply appropriate controls for spectral bleed-through in multi-color imaging
Image acquisition and analysis:
Capture images using consistent settings across experimental conditions
Implement blind analysis where possible to prevent bias
Use appropriate colocalization analysis methods and statistics
Since this application extends beyond the manufacturer's validated uses, thorough controls and stepwise optimization are essential. Document the entire protocol development process carefully and consider conducting parallel experiments with orthogonal approaches (e.g., fluorescent protein tagging) to confirm observed localization patterns .
Integrating ATL58 Antibody data with complementary molecular techniques provides a more comprehensive understanding of ATL58 biology. Consider these methodological approaches:
Multi-omics integration:
Correlate protein expression data (Western blot/ELISA) with transcriptomics (RNA-seq, qRT-PCR)
Compare protein abundance with promoter activity (reporter assays)
Integrate with metabolomics data to identify functional pathways affected by ATL58
Protein-protein interaction studies:
Use ATL58 Antibody for co-immunoprecipitation followed by mass spectrometry
Validate interactions using reciprocal pulldowns or proximity ligation assays
Correlate interaction data with functional assays (e.g., ubiquitination assays)
Functional genomics correlation:
Compare phenotypes from ATL58 mutant/overexpression lines with protein expression patterns
Correlate protein expression with physiological or developmental parameters
Integrate with ChIP-seq data if studying transcriptional regulation
Spatio-temporal analysis:
Combine tissue-specific Western blot quantification with in situ hybridization
Correlate developmental expression patterns with known developmental markers
Compare protein stability/turnover (pulse-chase) with functional readouts
Data integration frameworks:
Employ computational approaches to integrate multiple data types
Use pathway analysis tools to place ATL58 in functional networks
Apply machine learning for pattern recognition across diverse datasets
When employing ATL58 Antibody for plant stress response studies, several methodological considerations are critical:
Baseline expression profiling:
Establish normal ATL58 expression patterns across tissues and developmental stages
Document diurnal variations in expression under standard conditions
Create a reference dataset before introducing stress variables
Stress treatment standardization:
Precisely define stress parameters (duration, intensity, application method)
Include appropriate positive controls (known stress-responsive proteins)
Consider time-course experiments to capture dynamic responses
Sample preparation adaptations:
Modify extraction buffers for stress-treated samples (may contain different interfering compounds)
Adjust protein extraction protocols for tissues with altered physical properties post-stress
Include additional protease inhibitors for tissues with stress-induced proteolytic activity
Post-translational modification analysis:
Assess potential stress-induced modifications of ATL58 (phosphorylation, ubiquitination)
Use phosphatase treatments to determine if multiple bands represent phosphorylated forms
Consider specialized techniques (Phos-tag gels) for better resolution of modified forms
Data normalization challenges:
Traditional housekeeping genes may vary under stress conditions
Implement multiple normalization strategies (total protein, multiple reference proteins)
Validate stability of reference genes under your specific stress conditions
For meaningful interpretation, compare ATL58 expression patterns with known stress markers and integrate findings with physiological measurements. Document all environmental parameters during experiments, as minor variations can significantly impact stress responses in plants .
Epitope mapping provides valuable insights into the specific regions of ATL58 recognized by the antibody, which can inform experimental design and interpretation. Follow these methodological approaches:
Peptide array mapping:
Synthesize overlapping peptides spanning the entire ATL58 sequence
Probe arrays with the ATL58 Antibody
Identify peptides generating positive signals to localize epitope regions
Truncation mutant analysis:
Generate recombinant fragments of ATL58 with sequential deletions
Perform Western blot analysis to determine which fragments maintain antibody recognition
Narrow down the epitope region through systematic deletion mapping
Point mutation strategy:
Once approximate epitope regions are identified, introduce point mutations
Analyze changes in antibody binding affinity with mutated proteins
Identify critical amino acids essential for antibody recognition
Competitive binding assays:
Synthesize candidate epitope peptides
Perform competition experiments where peptides block antibody binding
Quantify inhibition to confirm epitope regions
Cross-species reactivity analysis:
Compare ATL58 sequences across related plant species
Test antibody against homologs from different species
Correlate recognition patterns with sequence conservation
Understanding epitope regions has practical applications for:
Predicting potential cross-reactivity with related proteins
Determining if the antibody can detect denatured vs. native protein
Assessing whether post-translational modifications might affect recognition
Interpreting negative results when epitopes might be masked by protein interactions
Document epitope information thoroughly to provide context for experimental results and facilitate method development by other researchers .
Multiplexing with ATL58 Antibody requires careful methodological considerations to ensure reliable simultaneous detection of multiple targets:
Antibody compatibility assessment:
Verify that ATL58 Antibody can be used alongside other primary antibodies
Test for potential cross-reactivity between antibody pairs
Ensure primary antibodies are raised in different host species for compatible secondary detection
Multiplex Western blotting approaches:
Fluorescent Western blotting with spectrally distinct secondary antibodies
Sequential reprobing with stripping between detections (validate complete stripping)
Size-based multiplexing when target proteins have sufficiently different molecular weights
Multiplex immunoassay optimization:
For bead-based multiplex assays, validate ATL58 Antibody performance in multiplex format
Test for potential cross-reactivity with other capture or detection antibodies
Establish appropriate dilution factors that work across all targets
Signal separation strategies:
Spectral unmixing for fluorescent detection systems
Sequential scanning for narrowly separated emission spectra
Spatial separation strategies for tissue section analysis
Data analysis considerations:
Implement appropriate normalization for each target in the multiplex panel
Account for potential signal spillover in closely related detection channels
Apply statistical methods appropriate for multi-parameter data
Multiplexing Method | Advantages | Limitations | Best Application |
---|---|---|---|
Fluorescent WB | Simultaneous detection, quantitative | Requires specialized equipment | Protein co-expression analysis |
Sequential reprobing | Uses standard equipment, flexible | Time-consuming, potential incomplete stripping | When fluorescent detection unavailable |
Multiplex ELISA | High-throughput, quantitative | Complex optimization, potential cross-reactivity | Population screening, biomarker panels |
For all multiplex applications, comprehensive validation with appropriate single-plex controls is essential. Document all optimization steps and validation experiments thoroughly to ensure reproducible results and proper interpretation of complex datasets .