ATG16L1 (Autophagy-Related 16-Like 1) is a critical protein in autophagy, a cellular degradation process. Phosphorylation of ATG16L1 at serine 278 (S278) serves as a biomarker for autophagy induction . The Anti-ATG16L1 (phospho S278) antibody [EPR19016] (ab195242) is a well-characterized monoclonal antibody used to detect this phosphorylation event .
Phosphorylation Site: S278 phosphorylation by ULK1 or IKKα regulates ATG16L1 stability and function .
Role in Disease: The Crohn’s disease-associated ATG16L1 T300A mutant shows enhanced cleavage under stress due to proximity to S278 .
Antibody Specificity:
Autophagy Monitoring: This antibody provides a direct readout of autophagy induction, unaffected by late-stage blocks .
Therapeutic Targets: Phospho-ATG16L1 levels correlate with autophagy rates in neurodegenerative and inflammatory diseases .
ATL16 (Arabidopsis Toxicos en Levadura 16) is a plant-specific protein expressed in Arabidopsis thaliana that functions as a RING-type E3 ubiquitin ligase. This protein plays critical roles in plant stress responses and developmental processes through the ubiquitin-proteasome system. ATL16's involvement in cellular regulatory mechanisms makes it an important research target for understanding plant adaptation to environmental conditions and development pathways .
For optimal Western blot results with ATL16 Antibody, prepare plant tissue samples using a buffer containing 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1% Triton X-100, 0.5% sodium deoxycholate, supplemented with protease inhibitors. Homogenize fresh or flash-frozen tissue samples thoroughly at 4°C, centrifuge at 12,000 × g for 15 minutes, and collect the supernatant. Load 20-30 μg of total protein per lane on SDS-PAGE gels. A dilution ratio of 1:1000 is typically recommended for detection, with expected molecular weight appearing higher than the predicted 56.4 kDa (approximately 70 kDa) due to post-translational modifications .
Store lyophilized ATL16 Antibody at -20°C for short-term (months) or -80°C for long-term (years) storage. After reconstitution with 50 μl of sterile water, create small aliquots to avoid repeated freeze-thaw cycles which can significantly degrade antibody performance. Before each use, briefly centrifuge tubes to collect any material adhering to the cap or walls. Working dilutions should be prepared fresh and used within the same day for best results .
When designing experiments with ATL16 Antibody, include both positive and negative controls. Use wild-type Arabidopsis thaliana samples as positive controls. For negative controls, utilize atl16 knockout/mutant lines when available, or samples from non-plant species where the protein is not expressed. Additionally, include a primary antibody omission control to assess non-specific binding of the secondary antibody. For loading controls in Western blot applications, anti-actin or anti-tubulin antibodies are recommended for normalization of protein expression data .
For studying ubiquitination pathways involving ATL16, implement a co-immunoprecipitation (co-IP) protocol using ATL16 Antibody to identify interaction partners within the ubiquitin-proteasome system. Treat plants with various stress conditions (e.g., drought, salinity, pathogen exposure) before tissue collection. After antibody-based pull-down, analyze the immunoprecipitated complexes using mass spectrometry to identify stress-responsive protein interactions. Compare ubiquitination profiles between treated and control samples by probing with both ATL16 Antibody and anti-ubiquitin antibodies in parallel Western blots. This approach allows for temporal mapping of ATL16-mediated protein degradation pathways under different environmental stressors .
For successful immunolocalization of ATL16 in plant tissues, fixation method selection is critical. Compare 4% paraformaldehyde with glutaraldehyde-based fixatives to determine optimal preservation of ATL16 epitopes. For tissue permeabilization, evaluate different concentrations of Triton X-100 (0.1-0.5%) or digitonin for maintaining structural integrity while allowing antibody penetration. Implement antigen retrieval techniques such as heat-induced epitope retrieval in citrate buffer (pH 6.0) to enhance signal detection. Use a blocking solution containing 5% BSA and 1% normal goat serum to minimize background. Test antibody at dilutions ranging from 1:100 to 1:500 to determine optimal signal-to-noise ratio. For signal amplification, consider tyramide signal amplification systems, especially when studying tissues with low ATL16 expression levels .
Validating ATL16 Antibody specificity across plant species requires a comprehensive approach. First, perform sequence alignment analysis of ATL16 with homologous proteins from target species to identify conserved epitope regions. Design peptide competition assays using synthesized peptides corresponding to the immunogen region of ATL16 to confirm specificity. Conduct Western blot analysis using protein extracts from multiple plant species with known ATL homologs, comparing banding patterns and molecular weights. For definitive validation, perform immunoprecipitation followed by mass spectrometry to confirm the identity of captured proteins. Additionally, test reactivity in atl16 knockout Arabidopsis lines as negative controls alongside wild-type samples to verify specificity within the ATL family of proteins .
When experiencing weak or absent signal with ATL16 Antibody, implement a systematic troubleshooting approach. First, verify protein extraction efficiency by analyzing total protein concentration and integrity on stained gels. Increase protein loading to 40-50 μg per lane and test higher antibody concentrations (1:500 instead of 1:1000). Extend primary antibody incubation time to overnight at 4°C with gentle agitation. For enhanced detection sensitivity, switch from conventional ECL to higher sensitivity chemiluminescent substrates or consider alternative detection systems like near-infrared fluorescent secondary antibodies. Evaluate different blocking agents (5% non-fat milk vs. 3-5% BSA) as milk proteins can sometimes interfere with plant antibody binding. Additionally, test different membrane types (PVDF vs. nitrocellulose) as protein transfer efficiency can vary depending on target properties .
To distinguish between specific and non-specific bands, implement a multi-faceted validation approach. Compare banding patterns between wild-type and atl16 mutant/knockout plants, with the specific band being absent or reduced in mutant samples. Perform peptide competition assays by pre-incubating the antibody with excess immunizing peptide before application to the membrane; specific bands should disappear while non-specific bands remain. For definitive identification, perform immunoprecipitation with ATL16 Antibody followed by mass spectrometry analysis of the enriched proteins. Additionally, compare band patterns across different tissues with known differential expression of ATL16 to correlate band intensity with expected expression patterns. Document molecular weight carefully, noting that the observed size (>70 kDa) differs from the predicted size (56.4 kDa) due to post-translational modifications .
For successful immunoprecipitation of ATL16 and its interacting partners, optimize buffer composition to preserve protein-protein interactions. Use a lysis buffer containing 50 mM Tris-HCl (pH 7.4), 150 mM NaCl, 1% NP-40, 0.5% sodium deoxycholate, supplemented with protease inhibitors, phosphatase inhibitors, and 10 mM N-ethylmaleimide to preserve ubiquitination. Pre-clear lysates with protein A/G beads to reduce non-specific binding. Cross-link antibody to beads using dimethyl pimelimidate to prevent antibody co-elution. Extend incubation times to 4-6 hours or overnight at 4°C with gentle rotation to enhance capture efficiency. For elution, compare different methods (low pH, high salt, or SDS) to determine which preserves interacting partners while efficiently releasing target proteins. Validate results using reciprocal co-immunoprecipitation with antibodies against suspected interaction partners .
| Antibody | Target | Species Reactivity | Applications | Sensitivity | Specificity | Background | Notes |
|---|---|---|---|---|---|---|---|
| ATL16 Antibody | ATL16 | A. thaliana | WB, IP | High | High with specific protocols | Low with optimized blocking | Requires optimization for ICC |
| ATL35 Antibody | ATL35 | A. thaliana | WB, IP, ICC | Moderate | High | Variable | Better for imaging studies |
| ATL69 Antibody | ATL69 | A. thaliana, Brassicaceae | WB, IP, ELISA | Moderate | Moderate | Low | Cross-reactivity with related species |
| ATL71 Antibody | ATL71 | A. thaliana | WB | High | High | Low | Limited application range |
When ATL16 Antibody protein detection results conflict with transcriptomic data, implement a systematic investigation process. First, validate both datasets independently: confirm antibody specificity through appropriate controls and verify RNA quality and sequence specificity of transcriptomic probes. Consider biological explanations for discrepancies, such as post-transcriptional regulation, protein stability differences, or condition-specific translation efficiency. Quantitatively compare protein levels across multiple biological replicates using densitometry normalized to appropriate loading controls. Perform time-course experiments to investigate temporal differences between transcript and protein expression. For definitive analysis, integrate proteomics approaches like targeted mass spectrometry (SRM/MRM) to provide transcript-independent quantification of ATL16 protein. Document experimental conditions meticulously, as differences in growth conditions, developmental stages, or stress exposures can lead to meaningful biological differences rather than technical artifacts .
To integrate ATL16 Antibody-derived data with machine learning for binding interaction predictions, researchers should adopt a multi-step approach. Begin by generating quantitative binding data through techniques like ELISA, surface plasmon resonance, or pull-down assays using ATL16 Antibody to identify and validate protein interactions. Structure this data in formats compatible with machine learning frameworks, ensuring proper labeling of positive and negative interaction pairs. Apply active learning strategies that iteratively expand the labeled dataset through targeted experiments, which has been shown to reduce the required number of experimental measurements by up to 35% compared to random sampling approaches . Implement cross-validation techniques specifically designed for out-of-distribution prediction scenarios to assess model generalizability. For optimal results, incorporate structural information about ATL16 and potential binding partners as additional features in the model. Balance the training dataset to avoid bias toward abundant interaction partners, and continuously refine predictions through validation experiments, creating a feedback loop between computational predictions and experimental verification .
Future advances in antibody engineering hold significant promise for developing enhanced ATL16 detection tools. Recombinant antibody technologies could enable production of single-chain variable fragments (scFvs) or nanobodies with improved tissue penetration for in situ applications in plant tissues. CRISPR-Cas9 epitope tagging of endogenous ATL16 would allow the use of highly specific commercial tag antibodies while maintaining native expression patterns and regulatory elements. Bispecific antibodies targeting both ATL16 and its substrate proteins could enable direct visualization of protein-protein interactions in plant cells. Additionally, proximity labeling approaches using ATL16 antibodies conjugated to enzymes like BioID or APEX2 would allow identification of transient interaction partners in their native cellular environment. These advanced tools would significantly enhance spatial and temporal resolution of ATL16 dynamics during plant development and stress responses .
When planning experiments combining ATL16 Antibody with CRISPR-modified plant lines, researchers must address several key considerations. First, epitope preservation must be verified - analyze CRISPR edit locations to ensure they don't alter the antibody recognition site. For knockout validation, ATL16 Antibody can confirm complete protein loss in knockout lines or quantify reduction in knockdown lines. When using CRISPR to introduce tagged versions of ATL16, compare antibody detection with tag detection to verify that the tag doesn't interfere with protein folding or epitope accessibility. For domain-specific mutations, use ATL16 Antibody to assess how structural alterations affect protein stability and expression levels. Additionally, researchers should characterize potential off-target effects by monitoring related ATL family proteins with appropriate controls. Finally, develop comprehensive genotyping and phenotyping pipelines that incorporate immunoblotting with ATL16 Antibody to correlate genetic modifications with protein expression levels and resulting phenotypes .
ATL16 Antibody can be instrumentally valuable for investigating plant immunity through several research approaches. By monitoring ATL16 protein abundance and modification patterns during pathogen infection, researchers can map temporal dynamics of this E3 ligase in immune signaling networks. Comparative studies between resistant and susceptible plant varieties using immunoprecipitation with ATL16 Antibody followed by mass spectrometry can identify differential interaction partners that may contribute to immunity. For spatiotemporal studies, combine immunolocalization using ATL16 Antibody with fluorescent pathogen tracking to visualize protein recruitment to infection sites. Researchers can develop experimental systems that pair ATL16 Antibody-based protein quantification with transcriptomics and metabolomics to build comprehensive models of immune pathway regulation. This multi-omics approach could identify how ATL16-mediated ubiquitination participates in the complex signaling networks activated during plant-pathogen interactions and reveal conserved mechanisms that could be targeted for developing broad-spectrum disease resistance in crops .
To ensure reproducibility across multiple studies using ATL16 Antibody, establish comprehensive quality control metrics. Implement antibody validation using positive controls (wild-type Arabidopsis) and negative controls (atl16 knockout lines) with each new antibody lot. Develop standard curves using recombinant ATL16 protein at defined concentrations to enable quantitative comparisons across experiments. Document batch information, including lot numbers and production dates, and maintain reference samples from successful experiments for comparative analysis. Establish acceptance criteria for signal-to-noise ratios and background levels in each application. For Western blot applications, implement densitometry standardization using housekeeping proteins and include internal reference samples across blots for normalization. Create detailed standard operating procedures (SOPs) for each application that specify sample preparation, antibody dilutions, incubation times and temperatures, and detection parameters. Finally, maintain a laboratory database of experimental conditions and outcomes to track antibody performance over time and identify potential variables affecting results .
For comprehensive epitope mapping of ATL16 Antibody, employ a multi-technique approach. Begin with in silico analysis using epitope prediction algorithms to identify potential binding regions within the ATL16 sequence. Generate a series of overlapping peptides spanning the ATL16 protein sequence and perform ELISA-based binding assays to narrow down the recognition region. For higher resolution mapping, express truncated versions of the protein and test reactivity by Western blot, progressively narrowing the epitope location. Use site-directed mutagenesis to introduce single amino acid substitutions within the suspected epitope region to identify critical binding residues. For structural insight, implement hydrogen-deuterium exchange mass spectrometry (HDX-MS) to identify protected regions upon antibody binding. This detailed epitope knowledge will inform experimental design, particularly when studying protein interactions or conformational changes that might mask or alter the epitope accessibility, and when evaluating cross-reactivity with related ATL family proteins .
When transitioning ATL16 Antibody applications from Arabidopsis to crop species, several methodological adaptations are essential. First, conduct sequence homology analysis between ATL16 and putative orthologs in target crop species to assess potential cross-reactivity. Optimize protein extraction protocols to address species-specific challenges such as higher levels of phenolic compounds, polysaccharides, or proteases in crop tissues. Test multiple extraction buffers with varying detergent compositions and protease inhibitor cocktails. Adjust antibody concentration and incubation conditions, as higher concentrations (1:500 instead of 1:1000) may be needed for cross-species detection. Implement more stringent washing procedures to reduce background in less characterized systems. Validate antibody specificity in each new species using techniques like immunoprecipitation followed by mass spectrometry. Consider developing species-specific blocking agents derived from knockout/knockdown lines of the target species. Document tissue-specific and developmental variations in protein expression patterns, as orthologous proteins may have divergent expression profiles across species despite sequence similarity .