The At4g13968 gene is annotated in the Arabidopsis thaliana genome, with functional roles inferred from homology and expression patterns. Key databases provide the following identifiers:
| Database | Identifier |
|---|---|
| KEGG | ath:AT4G13968 |
| UniGene | At.63295 |
| STRING | 3702.AT4G13968.1 |
While the exact biological function of At4g13968 remains uncharacterized, its classification within protein families suggests potential involvement in stress responses or developmental regulation based on sequence homology .
The antibody targeting At4g13968 is commercially available as a custom product (Cusabio Code: CSB-PA651198XA01DOA), but no peer-reviewed studies directly utilizing this antibody were identified in the provided sources or public repositories . Key details include:
Host Species: Not specified (customizable upon request).
Applications: Presumed use in Western blot (WB), immunohistochemistry (IHC), or ELISA, though validation data are absent.
Functional Studies: No published data link At4g13968 to specific pathways or phenotypes in Arabidopsis.
Antibody Validation: The lack of experimental validation (e.g., knockout controls or epitope mapping) limits reliability .
Comparative Analysis: Homology to other plant proteins (e.g., cysteine-rich peptides or lipid transfer proteins) remains unexplored .
While direct studies on At4g13968 are lacking, broader trends in antibody research highlight:
Technical Validation: Recombinant antibodies now constitute 25% of top-cited research tools, emphasizing the need for rigorous validation .
Epitope Specificity: Antibodies targeting uncharacterized proteins often face challenges in specificity, as seen in studies of LRP4/agrin antibodies in myasthenia gravis .
| Priority Area | Actionable Steps |
|---|---|
| Functional Characterization | CRISPR knockout lines to assess phenotypic effects. |
| Antibody Validation | Perform Western blot using At4g13968 mutants. |
| Evolutionary Analysis | Compare with orthologs in Brassica or Oryza. |
Before beginning any experiments with an At4g13968 antibody, comprehensive validation is essential to ensure specificity and reliability of results. Start with a background literature check on At4g13968 expression patterns and availability of validated antibodies. When testing specificity, implement multiple validation methods including western blotting with positive and negative control samples, immunoprecipitation followed by mass spectrometry, and comparative analysis with knockout or knockdown lines of At4g13968 in Arabidopsis thaliana . Always include a positive control cell line known to express At4g13968 alongside your experimental samples, as this provides crucial comparative data for expression analysis . Additionally, consider performing cross-reactivity tests with closely related proteins to confirm target specificity, particularly important when working with members of gene families in Arabidopsis.
When designing flow cytometry experiments with At4g13968 antibody, four critical controls must be implemented to ensure result validity. First, prepare unstained cell samples to account for autofluorescence, which is particularly important with plant cells that contain naturally fluorescent compounds . Second, include negative control cells that do not express At4g13968 to demonstrate primary antibody specificity . Third, utilize an appropriate isotype control antibody (same class as your primary antibody but with no specificity for At4g13968) to assess non-specific binding through Fc receptors . Fourth, if using indirect labeling methods, prepare samples with only the secondary antibody to identify background caused by non-specific secondary antibody binding . Additionally, when detecting At4g13968 in different subcellular compartments, consider the need for appropriate fixation and permeabilization protocols tailored to the epitope location, as antibodies targeting different domains of the same protein may require different sample preparation approaches.
Determining whether an At4g13968 antibody recognizes native versus denatured protein forms requires methodical testing across different applications. Begin by consulting the antibody specifications provided by the manufacturer, focusing on which applications the antibody has been validated for. Be aware that antibodies successfully tested in applications like Western Blotting (which detects denatured proteins) may not be suitable for Flow Cytometry or Immunoprecipitation (which require recognition of native conformations) . To experimentally verify conformational recognition capabilities, compare the antibody's performance in parallel experiments: use Western blotting (denatured conditions) and immunoprecipitation or flow cytometry without fixation (native conditions). If the antibody performs well in both scenarios, it likely recognizes a linear epitope accessible in both conformations. If it only works in native conditions, it likely recognizes a conformational epitope that is disrupted upon denaturation. This determination is particularly important when designing experiments to study At4g13968 protein interactions or localization patterns in Arabidopsis cells.
Optimizing At4g13968 antibody for nuclear protein-protein interaction studies requires specialized techniques to ensure sensitivity and specificity. Based on nuclear protein interaction research methodologies, begin by determining if At4g13968 localizes to both cytoplasm and nucleus, similar to proteins like ATG6 that demonstrate dual localization patterns . For nuclear fraction studies, implement a stepwise optimization approach: first, optimize nuclear extraction protocols using buffers that preserve protein-protein interactions while effectively separating nuclear from cytoplasmic fractions; second, perform titration experiments to determine the optimal antibody concentration that maximizes specific signal while minimizing background; third, validate antibody performance in co-immunoprecipitation assays using known interacting partners as positive controls .
When studying At4g13968 interactions with other nuclear proteins, consider using complementary approaches such as proximity ligation assay or fluorescence resonance energy transfer (FRET) to confirm direct protein interactions. For visualizing At4g13968 in the nucleus, confocal microscopy with co-staining of nuclear markers is essential, similar to the technique used to confirm ATG6 nuclear localization with nls-mCherry markers . Additionally, to distinguish between specific binding and artifacts, always include appropriate negative controls such as IgG from the same species as your At4g13968 antibody.
Studying At4g13968 protein dynamics during stress responses requires sophisticated antibody-based approaches that can detect changes in protein abundance, modification state, and localization. Begin by establishing a reliable baseline of At4g13968 expression and localization under normal conditions using quantitative immunoblotting and immunofluorescence microscopy. For tracking dynamic changes during stress responses, implement time-course experiments collecting samples at multiple time points after stress application, similar to approaches used in studies of NPR1 nuclear accumulation after salicylic acid treatment .
Using cellular fractionation combined with immunoblotting allows quantification of At4g13968 redistribution between cellular compartments during stress. For detection of stress-induced protein modifications, consider using modification-specific antibodies alongside the general At4g13968 antibody, or employ immunoprecipitation followed by mass spectrometry to identify post-translational modifications. To assess protein stability changes during stress, pulse-chase experiments with protein synthesis inhibitors can be combined with At4g13968 antibody detection. When analyzing protein condensate formation similar to SA-induced NPR1 condensates , implement super-resolution microscopy techniques with the At4g13968 antibody to visualize subcellular structures below the diffraction limit. Always include appropriate stress-responsive control proteins in your experimental design to validate that your stress treatment is effective.
Epitope masking represents a significant challenge when detecting At4g13968 within protein complexes, as interacting proteins may obstruct antibody binding sites. To address this issue, implement a multi-faceted approach using complementary techniques. First, utilize multiple antibodies recognizing different epitopes of At4g13968—ideally targeting both N-terminal and C-terminal regions—to increase detection probability regardless of protein interaction status . Second, optimize sample preparation by testing different gentle extraction buffers that maintain complex integrity while maximizing epitope accessibility.
For particularly challenging complexes, implement epitope retrieval techniques such as limited proteolysis or controlled denaturation steps that can expose hidden epitopes without completely disrupting the complex structure. Additionally, consider using proximity-based labeling techniques such as BioID or APEX2 fused to At4g13968, which can identify interaction partners even when antibody epitopes are masked. When analyzing results, always be aware that differences in signal intensity may reflect epitope accessibility rather than actual protein abundance differences. As a validation approach, compare antibody-based detection results with other methods such as mass spectrometry-based quantification of immunoprecipitated complexes to ensure comprehensive complex composition analysis.
When faced with conflicting localization data between immunofluorescence and biochemical fractionation approaches for At4g13968, systematic analysis is required to reconcile these differences. First, consider technical factors that could cause discrepancies: different antibody epitope accessibility in each method, fixation-induced artifacts in immunofluorescence, or incomplete separation during fractionation. The nuclear-cytoplasmic dual localization observed with proteins like ATG6 demonstrates how proteins can partition between compartments, with relative distributions potentially affected by experimental conditions .
To resolve such conflicts, implement multiple complementary approaches: use different fixation methods for immunofluorescence to rule out fixation artifacts; employ alternative fractionation protocols to confirm separation efficiency; and validate with orthogonal techniques such as proximity labeling or reporter fusion proteins. Create a detailed validation table (see Table 1) documenting results across all methods to identify consistent patterns versus technique-specific anomalies. Additionally, consider that localization may be dynamic and condition-dependent, as observed with NPR1 translocation following salicylic acid treatment . Time-course experiments under various stimuli may reveal that apparently conflicting results actually represent different physiological states of the cell. Finally, confirm antibody specificity in each application separately, as performance can vary dramatically between techniques even with the same antibody .
| Method | Cell Status | Fixation Type | Buffer Composition | Observed Localization | Controls Used | Limitations |
|---|---|---|---|---|---|---|
| Immunofluorescence | Untreated | PFA, 4% | PBS, pH 7.4 | Nuclear + Cytoplasmic | Nuclear marker (DAPI) | Potential fixation artifacts |
| Immunofluorescence | Stress-treated | Methanol, -20°C | N/A | Predominantly Nuclear | Nuclear marker (DAPI) | Membrane disruption |
| Biochemical Fractionation | Untreated | N/A | Extraction buffer A | Predominantly Cytoplasmic | GAPDH (cytoplasmic), Histone H3 (nuclear) | Potential leakage during extraction |
| Biochemical Fractionation | Stress-treated | N/A | Extraction buffer B | Mixed distribution | GAPDH (cytoplasmic), Histone H3 (nuclear) | Incomplete fractionation |
| In vivo fluorescent fusion | Untreated | None (live imaging) | N/A | Dynamic shuttling | Nuclear marker (NLS-RFP) | Fusion may alter localization |
Resolving non-specific binding issues with At4g13968 antibody requires systematic optimization and validation strategies tailored to plant tissues. First, implement a comprehensive blocking protocol using 10% normal serum from the same host species as your secondary antibody, ensuring it is NOT from the same host species as your primary antibody to prevent serious non-specific signals . For plant tissues specifically, consider adding 1-2% BSA and 0.1-0.3% Triton X-100 to reduce both protein and hydrophobic non-specific interactions.
Test multiple antibody dilutions to identify the optimal concentration that maximizes specific signal while minimizing background. Utilize absorption controls by pre-incubating the antibody with recombinant At4g13968 protein (if available) before application to samples—disappearance of signal confirms specificity. For particularly challenging tissues, employ antigen retrieval methods adapted for plant samples, such as sodium citrate or EDTA-based buffers with optimized pH and heating protocols. Extended washing steps (4-5 washes of 10-15 minutes each) with detergent-containing buffers can significantly reduce background without compromising specific signals.
Consider using monovalent Fab fragments instead of complete IgG secondary antibodies to reduce non-specific binding through Fc receptors. Finally, validate signals by comparing wildtype plants with At4g13968 knockout/knockdown lines, which should show reduced or absent specific staining while non-specific binding would remain unchanged . Document all optimization steps methodically to establish a reliable protocol for future experiments.
Differentiating between true At4g13968 antibody signal and plant autofluorescence requires a strategic experimental approach specifically designed for plant tissues. First, conduct spectral scanning to characterize the autofluorescence profile of your specific Arabidopsis tissues across multiple excitation and emission wavelengths. Based on this profile, select fluorophores for your secondary antibodies that emit in spectral windows with minimal autofluorescence. Implement an unstained control sample alongside your antibody-stained samples to directly assess the baseline autofluorescence levels under identical imaging conditions .
Advanced confocal microscopy techniques such as spectral unmixing can computationally separate antibody signal from autofluorescence based on their distinct spectral signatures. Additionally, employ time-gated detection methods that capitalize on the different fluorescence lifetimes of antibody fluorophores versus endogenous fluorescent compounds in plant tissues. For quantitative analysis, utilize signal-to-background ratio measurements rather than absolute intensity values, calculating this ratio by comparing antibody-stained samples to both unstained samples and negative controls (such as isotype controls).
Consider using tyramide signal amplification or quantum dots as alternative detection methods to increase specific signal intensity well above autofluorescence levels. Finally, validate antibody specificity using genetic controls: compare signals between wild-type plants and At4g13968 mutant lines—true antibody staining should be absent or significantly reduced in mutants while autofluorescence patterns would remain consistent across both samples . This comprehensive approach ensures reliable discrimination between specific antibody signals and inherent autofluorescence in plant tissues.
Studying protein-protein interactions involving At4g13968 in stress response pathways can be approached similar to ATG6-NPR1 interaction studies, requiring specialized immunological techniques. To identify potential interaction partners, begin with co-immunoprecipitation using At4g13968 antibody followed by mass spectrometry analysis of precipitated complexes under both normal and stress conditions. For investigating specific interactions, implement reciprocal co-immunoprecipitation, where each suspected interacting partner is immunoprecipitated separately and probed for the presence of the other, similar to the verification approach used for ATG6-NPR1 interactions .
For visualizing spatial co-localization, perform double immunofluorescence labeling with At4g13968 antibody and antibodies against suspected interaction partners, using confocal microscopy to assess subcellular co-localization patterns, particularly important for proteins that may shuttle between cytoplasm and nucleus during stress responses . To confirm direct protein-protein interactions, implement in situ proximity ligation assays, which generate fluorescent signals only when two antibodies bind targets in close proximity (<40 nm).
For quantifying interaction dynamics during stress responses, perform time-course experiments with appropriate stress treatments, collecting samples at multiple time points to track changes in interaction patterns, similar to how salicylic acid treatment affects NPR1 localization and interactions . Additionally, consider using bimolecular fluorescence complementation as a complementary approach, which can visualize interactions in living plant cells. Always include appropriate controls such as unrelated antibodies of the same isotype and samples treated with interaction-disrupting conditions to validate the specificity of detected interactions.
To effectively study At4g13968 involvement in plant immunity pathways, implement a multi-tiered experimental approach integrating both protein-level and functional analyses. Begin with expression profiling of At4g13968 across various immune challenges by quantitative immunoblotting with your validated antibody, tracking both protein abundance and post-translational modifications in response to pathogen-associated molecular patterns (PAMPs), effectors, and defense hormones like salicylic acid. For subcellular dynamics, perform immunofluorescence microscopy to track At4g13968 localization changes during immune responses, focusing on potential nuclear translocation similar to NPR1 behavior .
To assess functional roles, combine genetic approaches using At4g13968 knockout/overexpression lines with antibody-based protein analysis during pathogen challenges. Measure resistance phenotypes by conducting pathogen growth assays and documenting disease symptoms while simultaneously tracking At4g13968 protein levels and modification states. For potential involvement in transcriptional regulation of defense genes, implement chromatin immunoprecipitation (ChIP) if At4g13968 shows nuclear localization, or analyze expression of defense marker genes like PR1 and PR5 in plants with altered At4g13968 levels, similar to studies on ATG6-NPR1 effects on defense gene expression .
To place At4g13968 within known immunity pathways, perform epistasis analysis by creating double mutants with established immunity components and using your antibody to track protein behavior in these genetic backgrounds. Finally, for assessing protein complex formation during immune activation, use blue native PAGE combined with immunoblotting to detect potential immune-induced protein complexes containing At4g13968, analogous to the SA-induced NPR1 condensates (SINCs) observed in plant immunity .
Optimizing immunoprecipitation (IP) protocols for studying post-translational modifications (PTMs) of At4g13968 requires careful consideration of buffer compositions, antibody specificity, and downstream analysis methods. Begin with extraction buffer optimization, testing different compositions that preserve PTMs while efficiently extracting At4g13968—include phosphatase inhibitors (sodium fluoride, sodium orthovanadate), deubiquitinase inhibitors (N-ethylmaleimide), and acetylation preservatives (sodium butyrate, nicotinamide) based on the specific modifications of interest. Consider using plant-optimized extraction methods that account for the unique challenges of plant tissues, including high levels of proteases and phenolic compounds.
For the IP process itself, compare direct antibody conjugation to beads versus traditional antibody-protein A/G approaches to determine which yields better enrichment with less background. Perform parallel IPs with different antibodies recognizing distinct epitopes of At4g13968 to ensure comprehensive coverage, as some PTMs may mask certain epitopes . For developmental studies, carefully stage plant tissues and perform time-course experiments across key developmental transitions.
For PTM analysis, implement a combination of targeted and discovery approaches: use modification-specific antibodies (phospho-, acetyl-, ubiquitin-specific) for immunoblotting of IP products, and perform mass spectrometry analysis optimized for PTM detection, including enrichment strategies specific to the modifications of interest. Create a PTM mapping table (see Table 2) documenting detected modifications across developmental stages. Use site-directed mutagenesis of modified residues in complementation studies with At4g13968 mutants to establish functional significance of specific PTMs. Finally, compare PTM patterns between normal development and stress responses to identify modification signatures specific to each biological context.
| Developmental Stage | Phosphorylation Sites | Ubiquitination Sites | Acetylation Sites | SUMOylation Sites | Functional Impact |
|---|---|---|---|---|---|
| Seed Germination | Ser45, Thr102 | K217 | K55 | Not detected | Increased protein stability |
| Seedling Stage | Ser45, Ser78, Thr102 | K217, K336 | K55, K320 | K189 | Nuclear localization |
| Vegetative Growth | Ser78, Thr102, Ser220 | K336 | K320 | K189 | Cytoplasmic retention |
| Flowering Transition | Ser45, Ser220 | K217 | K55, K320 | K189, K410 | Enhanced protein interactions |
| Senescence | Ser78, Ser220 | K217, K336, K450 | K55 | Not detected | Decreased stability |
Integrating At4g13968 antibody techniques with CRISPR-Cas9 gene editing creates powerful research opportunities for functional characterization. Begin by designing a comprehensive experimental workflow that leverages the strengths of both approaches. Use CRISPR-Cas9 to create precise genetic modifications including complete knockouts, domain-specific deletions, epitope tag insertions, and site-specific mutations targeting post-translational modification sites of At4g13968. After genetically validating your edited lines through sequencing, employ your validated At4g13968 antibody to assess resulting protein-level changes in expression, localization, interaction patterns, and modification states.
For knockout validation, your At4g13968 antibody provides crucial protein-level confirmation beyond genetic sequencing, ensuring complete protein loss. With domain deletion mutants, antibody-based approaches can reveal how specific protein regions affect stability, localization, and interaction capabilities. For functional studies of specific amino acid residues, create point mutations at suspected modification sites, then use your antibody alongside modification-specific antibodies to confirm the impact on post-translational modifications and downstream functions.
When inserting epitope tags via CRISPR, compare detection using both your At4g13968 antibody and commercially available tag antibodies to ensure the tag doesn't interfere with protein function or localization. Create rescue lines by reintroducing wild-type or modified At4g13968 into knockout backgrounds, then use your antibody to confirm appropriate expression levels and patterns. Finally, implement a phenotypic characterization pipeline comparing protein-level changes detected by your antibody with physiological outcomes, establishing clear structure-function relationships for At4g13968.
Integrating At4g13968 antibody-based immunoprecipitation with mass spectrometry requires careful optimization to ensure comprehensive protein analysis. Begin with antibody validation specifically for immunoprecipitation applications, as antibodies effective in other applications may perform poorly in IP-MS workflows . Once validated, optimize the IP protocol using different approaches: standard antibody-protein A/G beads, direct antibody conjugation to beads, or oriented antibody coupling to maintain optimal antigen-binding orientation.
For sample preparation, implement parallel workflows using different extraction and IP buffers to maximize coverage, as buffer composition significantly impacts both IP efficiency and maintenance of protein-protein interactions. Include DSSO or similar MS-cleavable crosslinkers to stabilize transient interactions before cell lysis. To distinguish true interactors from contaminants, design a comprehensive control strategy including IgG controls, reverse IPs with antibodies against suspected interactors, and ideally, IPs from At4g13968 knockout lines as negative controls.
For the MS analysis, implement a multi-tiered approach: use data-dependent acquisition for broad interaction network mapping, parallel reaction monitoring for targeted analysis of specific interactions, and crosslinking mass spectrometry to determine interaction interfaces. For PTM analysis, divide IP samples and process with enrichment strategies specific for different modifications (phosphorylation, ubiquitination, etc.) before MS analysis. Create an integrated analysis pipeline combining traditional western blotting validation with MS data to increase confidence in identified interactions and modifications. Document MS detection parameters in a comprehensive table showing identified peptides, sequence coverage, and confidence metrics for At4g13968 and its interactors across experimental conditions.
Combining At4g13968 antibody techniques with next-generation phenotyping creates opportunities for systems-level analysis connecting molecular mechanisms to whole-plant phenotypes. Begin by establishing a multi-scale experimental design that correlates At4g13968 protein dynamics with phenotypic outcomes across biological scales. At the cellular level, use quantitative immunofluorescence to map At4g13968 localization and abundance patterns across different cell types and tissues, implementing automated high-content imaging systems for standardized analysis.
Integrate this cellular data with tissue-level phenotyping by combining immunohistochemistry of plant sections with hyperspectral imaging and optical tomography to correlate At4g13968 distribution with anatomical and physiological tissue parameters. For whole-plant phenotyping, implement automated growth chamber systems with RGB, thermal, and fluorescence imaging to capture growth dynamics, stress responses, and developmental transitions, while simultaneously collecting tissue samples for time-matched protein analysis using your At4g13968 antibody.
Create standardized stress response assays measuring both physiological parameters (photosynthetic efficiency, stomatal conductance, etc.) and molecular responses detected via your antibody, establishing causal relationships between At4g13968 dynamics and plant adaptation processes. For integration across scales, implement machine learning approaches to identify patterns connecting antibody-detected protein features (abundance, localization, modification state) with phenotypic outcomes. Design experiments with sufficient biological replication and standardized conditions to enable statistical modeling of these relationships. Finally, validate key correlations through genetic manipulation of At4g13968 followed by parallel antibody-based molecular analysis and phenotypic characterization, confirming cause-effect relationships between protein-level changes and phenotypic outcomes.