The At4g17713 Antibody targets the protein encoded by the At4g17713 locus, a gene annotated in the Arabidopsis thaliana genome. While the specific functional role of this gene remains uncharacterized in the provided sources, its protein product is presumed to contribute to cellular processes unique to plant systems . Antibodies like this enable researchers to localize, quantify, or inhibit the protein in experimental settings.
Protein Localization: Used in immunohistochemistry (IHC) or immunofluorescence (IF) to map the spatial expression of At4g17713 in plant tissues .
Western Blotting: Detects denatured protein in lysates to study expression levels under varying conditions .
Epitope Uncharacterized: The antibody’s binding site on the At4g17713 protein is not detailed, limiting mechanistic insights .
Validation Gaps: No peer-reviewed studies validating its specificity or efficacy were identified in the provided sources. Independent validation (e.g., knockout controls) is advised .
Functional Annotation: Collaborate with plant genomics consortia to link At4g17713 to pathways like stress response or development.
Structural Studies: Use cryo-EM or X-ray crystallography to resolve the antibody-antigen interaction .
The following table contextualizes At4g17713 Antibody among other Arabidopsis-targeting reagents ([Source 3]):
| Antibody Target | Uniprot ID | Product Code | Size Options |
|---|---|---|---|
| At4g17713 | Q2V3H6 | CSB-PA648065XA01DOA | 2 mL / 0.1 mL |
| At1g54070 | F4HV65 | CSB-PA911884XA01DOA | 2 mL / 0.1 mL |
| At5g18400 | Q8L7Z3 | CSB-PA810543XA01DOA | 2 mL / 0.1 mL |
While direct studies on At4g17713 are absent in the reviewed literature, broader antibody engineering principles apply:
The At4g17713 gene in Arabidopsis thaliana encodes a protein involved in cellular signaling pathways related to plant immune responses. Antibodies targeting this protein are crucial for investigating protein localization, expression levels, protein-protein interactions, and functional studies in plant biology. These antibodies enable researchers to visualize where and when the protein is expressed, which provides essential insights into its biological role. Additionally, At4g17713 antibodies allow for the isolation of protein complexes through immunoprecipitation techniques, revealing interaction networks and signaling cascades. Understanding this protein's function contributes to our knowledge of plant defense mechanisms and potential applications in agricultural biotechnology for crop improvement.
The specificity of an At4g17713 antibody can be determined through several complementary validation approaches. Begin with Western blot analysis using both wild-type plant tissue and knockout/knockdown lines for the At4g17713 gene. A specific antibody will show a band of the expected molecular weight in wild-type samples that is absent or significantly reduced in knockout samples. Immunoprecipitation followed by mass spectrometry can confirm that the antibody pulls down the target protein with minimal off-target binding. Additionally, perform immunohistochemistry or immunofluorescence experiments comparing wild-type and mutant tissues to verify localization patterns. Cross-reactivity testing against related proteins should be conducted, particularly against homologous proteins with similar sequences. Finally, epitope mapping can identify the specific amino acid sequence recognized by the antibody, which helps predict potential cross-reactivity issues.
At4g17713 antibodies require specific storage conditions to maintain functionality and prevent degradation. Store primary antibodies in small aliquots (20-50 μL) at -20°C for long-term storage to avoid repeated freeze-thaw cycles, which can denature the antibody and reduce its efficacy. For working solutions, store at 4°C with antimicrobial preservatives such as 0.02% sodium azide or 50% glycerol. Polyclonal antibodies generally maintain stability for 1-2 weeks at 4°C, while monoclonal antibodies may remain stable for up to a month. Always centrifuge antibody solutions briefly before use to pellet any precipitates that may have formed during storage. Avoid exposing antibodies to direct light, particularly those conjugated with fluorophores, as this can cause photobleaching and reduced signal intensity. Record freeze-thaw cycles and regularly validate antibody performance using positive controls to ensure continued specificity and sensitivity throughout the research project.
When designing immunization protocols for generating At4g17713-specific antibodies, begin with comprehensive epitope analysis of the target protein using bioinformatics tools to identify unique, accessible, and immunogenic regions. Choose between using recombinant protein fragments, synthetic peptides, or full-length proteins based on solubility and expression efficiency. For recombinant approaches, express the protein fragment in E. coli, purify using affinity chromatography, and verify purity by SDS-PAGE before immunization. For peptide antigens, select 15-20 amino acid sequences that are unique to At4g17713, avoiding transmembrane regions and ensuring the sequence is accessible in the native protein. Consider using multiple host species (rabbits, mice, rats) to generate diverse antibody repertoires, as demonstrated in studies with human immunoglobulin loci . Implement a robust immunization schedule with primary immunization using complete Freund's adjuvant followed by 3-4 booster immunizations with incomplete Freund's adjuvant at 2-3 week intervals. Monitor antibody titers through ELISA after each boost, and collect final sera when titers plateau. For monoclonal antibody production, harvest splenocytes 3-4 days after the final boost and proceed with hybridoma generation, which enables the production of highly specific antibodies similar to the approach described for SARS-CoV-2 antigen recognition in mouse models .
Optimizing Western blot protocols for At4g17713 antibodies requires systematic adjustments across multiple parameters. Begin with sample preparation, using extraction buffers containing protease inhibitors and denaturing agents appropriate for plant tissues (e.g., PVPP to remove phenolic compounds). Test multiple protein extraction methods to identify the one that best preserves the target protein while minimizing contaminants. For gel electrophoresis, optimize acrylamide percentage (typically 10-12% for mid-sized proteins) and loading amount (start with 20-40 μg total protein). During transfer, test both wet and semi-dry methods with different buffer compositions and transfer times to ensure complete protein transfer. For antibody incubation, create a matrix testing different antibody dilutions (1:500 to 1:5000), blocking agents (5% BSA or milk), and incubation times (1 hour at room temperature versus overnight at 4°C). Include positive controls (recombinant At4g17713 protein) and negative controls (knockout plant lines) in each experiment. Test different detection systems, comparing chemiluminescence, fluorescence, and colorimetric methods for optimal signal-to-noise ratio. Document all optimization steps in a table format that tracks changes in protocol parameters against signal strength and background levels. This systematic approach results in a reproducible protocol that maximizes detection sensitivity while minimizing non-specific background.
Establishing effective immunoprecipitation (IP) protocols for At4g17713 protein complexes requires careful optimization of multiple parameters. Begin by selecting appropriate lysis conditions that maintain protein-protein interactions while efficiently extracting the target protein. For plant tissues, use a buffer containing 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1% NP-40 or Triton X-100, with protease and phosphatase inhibitors. Test crosslinking reagents like formaldehyde (0.1-1%) or DSP (dithiobis-succinimidyl propionate) to stabilize transient interactions. Pre-clear lysates with Protein A/G beads to remove non-specific binding proteins. For antibody coupling, test both direct addition of antibody to lysate and pre-coupling to beads, comparing results for cleaner pull-downs. Optimize antibody amounts (2-10 μg per IP) and incubation times (2 hours to overnight at 4°C). Include appropriate controls in each experiment: IgG-only control, input sample (5-10% of starting material), and when available, tissue from At4g17713 knockout plants. For elution, compare harsh conditions (boiling in SDS sample buffer) with native elution methods (competitive elution with epitope peptides) based on downstream applications. When identifying interaction partners, use high-sensitivity mass spectrometry approaches combined with statistical analysis to distinguish true interactors from background proteins. This methodology parallels approaches used in antibody development against complex antigens as described in immunological research using engineered mouse models .
Advanced computational tools for analyzing At4g17713 antibody binding properties and epitope predictions involve multiple algorithms and platforms. Begin with sequence-based epitope prediction using tools like BepiPred, ABCpred, and SVMTriP, which employ machine learning algorithms to identify likely antigenic regions. For structural epitope prediction, utilize tools like DiscoTope, ElliPro, and EPCES that analyze 3D protein structures to identify surface-exposed residues likely to serve as antibody binding sites. When structural data is unavailable, use homology modeling through SWISS-MODEL or I-TASSER to generate predicted structures of At4g17713. For evaluating cross-reactivity risks, employ BLAST analysis against the entire Arabidopsis proteome to identify proteins with similar epitope regions. Molecular docking simulations using AutoDock or HADDOCK can model antibody-antigen interactions to predict binding affinity and specificity. These computational approaches parallel the methodology employed in the Virtual Lab for nanobody design, where protein language models like ESM, protein folding models like AlphaFold-Multimer, and computational biology software like Rosetta are integrated into comprehensive workflows for antibody engineering . Such integrated computational pipelines enable researchers to prioritize the most promising antibody candidates before investing in experimental validation, significantly accelerating the antibody development process.
Optimizing ChIP-seq with At4g17713 antibodies for transcription factor studies requires meticulous attention to protocol details. Begin with crosslinking optimization, testing formaldehyde concentrations (1-3%) and incubation times (5-20 minutes) to balance efficient protein-DNA crosslinking with DNA fragmentation efficiency. For chromatin preparation, optimize sonication parameters to generate fragments of 200-500 bp, verifying fragment size distribution by agarose gel electrophoresis. Test multiple antibody concentrations (2-10 μg per ChIP) and incubation conditions (4-16 hours at 4°C) to maximize specific pulldown while minimizing background. Implement stringent washing steps with increasingly stringent buffers (low salt, high salt, LiCl, and TE buffers) to remove non-specifically bound chromatin. Include appropriate controls: input chromatin (5-10% of starting material), IgG control, and negative genomic regions known not to bind the factor. For library preparation, minimize PCR cycles (typically 10-15) to reduce amplification biases. During bioinformatic analysis, use peak calling algorithms (MACS2, HOMER) with appropriately matched input controls and implement IDR (Irreproducible Discovery Rate) analysis across biological replicates to identify high-confidence binding sites. Create a comprehensive table documenting peak characteristics including peak height, width, distance to transcription start sites, and overlap with known regulatory elements. This methodological approach aligns with advanced antibody-based chromatin analysis techniques used in complex biological systems as referenced in immunoglobulin research models .
Multiplexed immunofluorescence with At4g17713 antibodies and other plant protein markers requires strategic planning and technical optimization. Begin by selecting antibodies raised in different host species (e.g., rabbit anti-At4g17713 combined with mouse anti-organelle markers) to enable simultaneous detection with species-specific secondary antibodies. When antibodies from different species are unavailable, implement sequential staining with complete elution or inactivation steps between rounds. For plant tissues, optimize fixation conditions testing cross-linkers like paraformaldehyde (2-4%) and glutaraldehyde (0.1-0.5%) to preserve both protein epitopes and cellular architecture. Enhance tissue permeabilization with detergents (0.1-0.5% Triton X-100) and/or enzymatic treatments (cell wall degrading enzymes) to improve antibody penetration while maintaining structural integrity. Implement extensive blocking steps (1-2 hours) with serum from the secondary antibody host species plus BSA to minimize non-specific binding. Test multiple antibody dilutions in a systematic matrix to identify optimal concentration ratios that produce balanced signal intensities across all channels. For detecting low-abundance proteins like At4g17713, incorporate signal amplification systems such as tyramide signal amplification (TSA) or antibody fragment-based amplification methods. Use spectral unmixing on confocal microscopes to separate overlapping fluorophore signals, and include single-stained controls for each fluorophore to establish spectral profiles. Document colocalization quantitatively using Pearson's or Mander's coefficients to assess the degree of spatial overlap between At4g17713 and other cellular markers. This approach enables comprehensive visualization of protein localization patterns and interactions within complex plant cellular environments.
Analyzing quantitative differences in At4g17713 protein expression requires rigorous experimental design and statistical analysis. Begin with at least three biological replicates per condition to account for natural variation in protein expression. For Western blot analysis, implement normalization using housekeeping proteins (e.g., actin, GAPDH, tubulin) or total protein staining methods such as Ponceau S or SYPRO Ruby. Quantify band intensities using software like ImageJ or specialized Western blot analysis platforms, ensuring measurements fall within the linear dynamic range of detection. For statistical analysis, perform normality tests (Shapiro-Wilk) on the data distribution before selecting appropriate statistical tests. For normally distributed data, apply parametric tests (t-test for two conditions, ANOVA for multiple conditions followed by post-hoc tests like Tukey's HSD). For non-normally distributed data, use non-parametric alternatives (Mann-Whitney U test or Kruskal-Wallis with Dunn's post-hoc test). Present data comprehensively using scatter plots showing individual data points overlaid with means and standard deviations, rather than simple bar graphs. Calculate fold changes relative to control conditions and report exact p-values rather than significance thresholds. For complex experimental designs with multiple variables, consider multivariate analysis approaches. When analyzing absolute protein quantities, develop a standard curve using recombinant At4g17713 protein to convert band intensities to protein concentrations. This methodological approach aligns with quantitative analysis techniques used in antibody research and protein expression studies described in immunological research .
Advanced algorithms for detecting conformational epitopes in At4g17713 protein structure employ complementary computational approaches. Structure-based methods like DiscoTope 2.0, EPSVR, and ElliPro analyze 3D protein models to identify surface patches with physicochemical properties conducive to antibody binding. These algorithms calculate parameters including surface accessibility, protrusion index, hydrophilicity, and amino acid propensity scores to predict conformational epitopes with accuracy rates of 70-80%. Energy-based methods like SEPPA and Epitopia compute the interface energy potential between antibody and antigen residues to identify energetically favorable binding regions. Machine learning approaches including CBTOPE and EPCES combine multiple features through trained classification algorithms (random forests, neural networks, or support vector machines) to improve prediction accuracy. For At4g17713 specifically, homology modeling with AlphaFold or similar protein structure prediction tools may be necessary if experimental structures are unavailable, similar to the approach used in nanobody design for SARS-CoV-2 . During analysis, integrate molecular dynamics simulations to account for protein flexibility, as conformational epitopes may only be accessible in certain structural states. Cross-reference predictions from multiple algorithms and prioritize consensus epitope regions. The final output should include a detailed table mapping predicted epitopes with their confidence scores, accessibility metrics, and conservation levels across related proteins. This comprehensive approach parallels multi-tool computational workflows used in antibody engineering as demonstrated in the Virtual Lab for nanobody design .
Resolving contradictory results between different assays using At4g17713 antibodies requires systematic investigation of multiple technical and biological factors. First, examine antibody characteristics: different antibodies may recognize distinct epitopes that vary in accessibility across experimental conditions. Document epitope locations, clonality (monoclonal vs. polyclonal), and validation history for each antibody. Evaluate protocol differences by creating a comprehensive table comparing key parameters across assays: fixation methods, buffer compositions, incubation times, detection systems, and sample preparation techniques. Conduct epitope masking tests to determine if sample processing differentially affects epitope accessibility. For contradictions between immunoassays and functional studies, consider post-translational modifications that might affect antibody recognition but not protein function. Implement orthogonal validation using antibody-independent methods such as genetic approaches (CRISPR knockout/knockdown) or mass spectrometry-based protein identification. Assess antibody batch variation by testing multiple lots and documenting lot-specific performance metrics. Examine cellular context effects by comparing results across different tissue types, developmental stages, and subcellular fractions. For contradictions in quantitative measurements, verify that all assays operate within their linear dynamic range. Consult published literature and antibody validation databases for reported caveats specific to plant immunoassays. Finally, design reconciliation experiments specifically targeting the conditions where contradictions occur, controlling for all identified variables. This methodical approach to troubleshooting parallels complex experimental validation workflows used in antibody research for challenging targets .
Reducing background in immunohistochemistry with At4g17713 antibodies requires a multi-faceted approach addressing several common interference sources. Begin with sample preparation optimization: test different fixation methods (paraformaldehyde, acetone, methanol) and durations (30 minutes to overnight) to preserve epitopes while maintaining tissue morphology. For plant tissues specifically, implement thorough clearing steps to remove chlorophyll and other autofluorescent compounds using ethanol series (30-100%) followed by clearing agents like ClearSee or methyl salicylate. Optimize permeabilization by testing detergent concentrations (0.1-1% Triton X-100) and enzymatic treatments (cell wall degrading enzymes) to balance antibody accessibility with minimal tissue disruption. Implement extensive blocking protocols testing different blocking agents (5-10% serum from secondary antibody host species, BSA, casein, commercial blocking reagents) and extended blocking times (1-3 hours at room temperature or overnight at 4°C). Test antibody dilutions systematically (1:100 to 1:2000) to determine the optimal concentration that maximizes specific signal while minimizing background. Incorporate stringent washing steps between antibody incubations using buffers containing detergents (0.05-0.1% Tween-20) and salt (150-300 mM NaCl) to remove unbound antibodies. For particularly challenging samples, consider implementing signal amplification methods with low background characteristics, such as tyramide signal amplification or quantum dot secondary antibodies. Document all optimization steps in a detailed table tracking changes in protocol parameters against signal-to-noise ratios. This comprehensive approach to background reduction aligns with advanced immunohistochemical techniques used in complex tissue imaging studies .
Validating antibody lots for consistent At4g17713 detection requires a standardized quality control workflow. Establish a reference standard by preparing a large batch of positive control samples (e.g., plant tissue overexpressing At4g17713 or recombinant protein) that can be aliquoted and stored long-term. When receiving new antibody lots, perform side-by-side Western blot analysis with the previous validated lot and the reference standard, examining band intensity, molecular weight accuracy, and presence of non-specific bands. Quantify signal-to-noise ratios and calculate the coefficient of variation between lots, establishing acceptance criteria (typically <15% variation). Implement immunoprecipitation efficiency tests to verify that new lots pull down comparable amounts of target protein. For functional assays, verify that inhibitory or activating antibodies maintain consistent effects across lots. Create a standardized validation panel including positive controls (wild-type tissue), negative controls (knockout/knockdown lines), and potentially cross-reactive samples. Document lot-specific performance in a database tracking key metrics: titer, optimal working dilution, background levels, and specific signal intensity. For quantitative applications, generate standard curves with recombinant protein to determine lot-specific detection limits and linear ranges. Implement stability testing by monitoring antibody performance over time under recommended storage conditions. This systematic approach enables researchers to maintain experimental consistency despite the inherent variation in antibody production, similar to quality control processes used in advanced antibody development workflows .
When At4g17713 antibodies fail to detect targets in complex plant extracts, implement a systematic troubleshooting approach examining sample preparation, antibody parameters, and technical considerations. Begin by verifying target presence through complementary techniques such as RT-qPCR for mRNA expression or mass spectrometry for protein identification. Optimize protein extraction protocols specifically for plant tissues by testing different extraction buffers with varying detergent types (CHAPS, NP-40, Triton X-100) and concentrations to improve solubilization of membrane-associated proteins. Address plant-specific challenges by incorporating PVPP or activated charcoal to remove polyphenols, along with high concentrations of reducing agents (DTT, β-mercaptoethanol) to prevent oxidation of sensitive proteins. Test multiple sample denaturation conditions, varying temperatures (37°C to 95°C) and durations (5-30 minutes) to find optimal conditions that expose epitopes without causing protein aggregation. Examine antibody parameters by testing a range of concentrations (0.1-10 μg/mL) and incubation conditions (1 hour at room temperature to overnight at 4°C). For Western blots, test different membrane types (PVDF vs. nitrocellulose) and transfer methods (wet vs. semi-dry). Consider epitope masking due to post-translational modifications by treating samples with phosphatases, deglycosylation enzymes, or other modification-removing treatments. If available, test alternative antibodies targeting different epitopes of At4g17713. For particularly challenging samples, implement sample fractionation or immunoprecipitation to enrich the target protein before detection. Document all troubleshooting variables and outcomes in a comprehensive table to guide future experiments. This methodical approach parallels troubleshooting strategies employed in complex antibody development projects for challenging targets .
CRISPR-based genome editing offers powerful approaches for validating At4g17713 antibody specificity through targeted genetic modification. Design multiple guide RNAs targeting different regions of the At4g17713 gene using tools like CRISPOR or CHOPCHOP that assess on-target efficiency and off-target potential. Construct CRISPR-Cas9 vectors using plant-optimized promoters (e.g., U6 for sgRNA, UBQ10 for Cas9) and appropriate selection markers for Arabidopsis transformation. Generate complete knockout lines through frameshift mutations in early exons and epitope-specific knockout lines by precisely targeting the antibody binding region. For more sophisticated validation, create epitope-tagged lines by inserting small epitope tags (FLAG, HA, V5) at the C-terminus of the endogenous At4g17713 gene using homology-directed repair. Confirm genetic modifications through Sanger sequencing and genomic PCR. Validate edited lines at the transcript level using RT-qPCR and at the protein level using validated commercial antibodies against the introduced epitope tags. Perform comprehensive antibody validation by comparing Western blots, immunoprecipitation, and immunohistochemistry results between wild-type and CRISPR-edited lines. A specific At4g17713 antibody should show signal in wild-type samples that is absent in knockout lines and colocalized with epitope tag signals in tagged lines. Document results in a comprehensive table showing signal detection across different assays and genetic backgrounds. This genetic validation approach provides definitive evidence of antibody specificity that complements traditional biochemical validation methods, similar to comprehensive antibody validation approaches used in immunological research .
Developing multiplex ELISA assays for At4g17713 and related proteins requires careful consideration of multiple technical parameters. Begin with comprehensive antibody cross-reactivity testing to ensure each antibody in the multiplex panel specifically recognizes only its intended target. Create a cross-reactivity matrix testing each antibody against all purified target proteins to document potential cross-reactions. Select capture and detection antibody pairs that recognize different, non-overlapping epitopes on the same target to maximize sensitivity and specificity. For optimal performance, pair monoclonal capture antibodies with polyclonal detection antibodies. Test different immobilization strategies for capture antibodies, comparing passive adsorption, covalent coupling, and oriented immobilization through protein A/G or streptavidin-biotin systems. Optimize antibody concentrations through checkerboard titrations to identify combinations that maximize signal while minimizing background. Address plant-specific matrix effects by developing specialized blocking buffers containing plant protein extracts from knockout lines to reduce non-specific binding. Implement stringent washing protocols with plant-optimized detergent concentrations to remove interfering compounds while preserving specific antibody-antigen interactions. For detection systems, compare traditional colorimetric readouts with more sensitive chemiluminescent or fluorescent alternatives, particularly for low-abundance targets. Validate assay performance parameters including lower limit of detection, upper limit of quantification, precision (intra- and inter-assay CV), accuracy (spike recovery), and linearity (dilutional linearity). Establish a comprehensive validation panel including positive controls, negative controls, and potentially interfering samples. Document all performance metrics in a detailed table tracking each target protein's detection parameters. This methodical approach to multiplex immunoassay development parallels advanced antibody-based detection systems used in complex biological research .