At1g30730 is a protein encoded by the At1g30730 gene in Arabidopsis thaliana (Mouse-ear cress). According to sequence analysis, the protein contains 158 amino acids with the sequence MKRKSDYVKRPVSRTGLGLILKKLVELEKVEMNWNPYGGRMGEIPSSRTPFPHRGGNLFNIEYIIDWSEAGDNVEKKYLALANEFYRFMTPYVSSNPREAFLNYRDIDIGSSGNSTYEEGKIYGAKYFKDNFERLVDIKTKFDEINFWRNEQSIPVRK . The protein has been identified in studies related to plant signaling pathways, and research suggests it may play a role in plant development or stress responses. It appears in differential expression studies, such as those examining leucine-rich repeat receptor-like kinase1 (RPK1) functions in Arabidopsis .
Current research antibodies against At1g30730 include:
Polyclonal antibodies (e.g., CSB-PA219368XA01DOA) raised in rabbits against recombinant Arabidopsis thaliana At1g30730 protein. These are purified using antigen affinity methods and provided in liquid form with preservatives such as 0.03% Proclin 300, 50% Glycerol, and 0.01M PBS (pH 7.4) .
Combinations of monoclonal antibodies targeting different regions of the protein:
X-Q6NNH8-N: Targets N-terminus sequence
X-Q6NNH8-C: Targets C-terminus sequence
X-Q6NNH8-M: Targets non-terminus (middle) sequence
Each combination contains antibodies raised against multiple synthetic peptides representing the respective region .
At1g30730 antibodies have been validated for the following applications:
Enzyme-Linked Immunosorbent Assay (ELISA): The antibodies demonstrate high-titer binding in ELISA applications, with titers reaching approximately 10,000, which corresponds to detection sensitivity of approximately 1 ng of target protein .
Western Blotting (WB): The antibodies have been validated for detecting At1g30730 protein in Western blot applications, providing specific identification of the target antigen .
Expression Analysis: These antibodies can be used in studies examining differential gene expression patterns in Arabidopsis, such as those shown in Table 2 of research publications where At1g30730 exhibited altered expression under specific experimental conditions .
For optimal performance, At1g30730 antibodies should be handled according to these guidelines:
Storage Temperature: Upon receipt, store at -20°C or -80°C for long-term preservation .
Avoid Repeated Freeze-Thaw Cycles: Multiple freeze-thaw cycles can damage antibody structure and diminish binding efficiency. Aliquot the antibody solution upon first thaw to minimize freeze-thaw events .
Working Solution Preparation: When preparing dilutions for experiments, use appropriate buffers as recommended for specific applications (e.g., TBST with 5% BSA or non-fat milk for Western blotting).
Handling Precautions: Despite being for research use only, follow standard laboratory safety protocols for handling biological materials, including wearing appropriate personal protective equipment.
Expiration Considerations: Antibody functionality should be validated if used beyond the manufacturer's recommended shelf life, particularly for critical experiments.
The choice between polyclonal and monoclonal At1g30730 antibodies has significant implications for research outcomes:
Polyclonal At1g30730 Antibodies:
Recognize multiple epitopes across the target protein, potentially increasing detection sensitivity
Useful for applications where maximum antigen capture is desired
May exhibit batch-to-batch variation requiring validation between lots
Typically provide robust detection in Western blotting and ELISA
Examples include the rabbit polyclonal antibody CSB-PA219368XA01DOA
Monoclonal At1g30730 Antibodies:
Target specific epitopes with high precision
Provide consistent results with minimal batch-to-batch variation
Particularly valuable for detection of specific protein domains or conformations
Often used in combinations (as seen with X-Q6NNH8-N/C/M products) to enhance detection while maintaining specificity
May be engineered to optimize binding characteristics through techniques similar to those used in bispecific antibody development
Validating At1g30730 antibody specificity presents several challenges that researchers must address:
Cross-reactivity Assessment: At1g30730 may share sequence homology with other Arabidopsis proteins, necessitating thorough validation through:
Western blot analysis using recombinant At1g30730 protein as a positive control
Testing against known knockout/knockdown lines where the At1g30730 gene has been silenced
Peptide competition assays to confirm binding specificity
Expression Level Variability: At1g30730 expression varies across tissues and under different experimental conditions, as evidenced in differential expression studies . This variability requires:
Tissue-specific validation
Calibration of detection methods across different expression levels
Inclusion of appropriate loading controls
Post-translational Modifications: Potential modifications may affect epitope accessibility, requiring:
Technical Validation Standards: Include:
Signal-to-noise ratio determination
Limit of detection establishment
Reproducibility across multiple experimental replicates
While similar to validation processes for other plant protein antibodies, these challenges are particularly relevant for At1g30730 due to its expression patterns in Arabidopsis signaling studies.
Leveraging At1g30730 antibodies in multi-parameter studies requires strategic experimental design:
Co-localization Studies:
Combine At1g30730 antibodies with antibodies against known interacting proteins or organelle markers
Use secondary antibodies with distinct fluorophores that have minimal spectral overlap
Include appropriate controls to account for potential antibody cross-reactivity
Protein Complex Analysis:
For co-immunoprecipitation studies, optimize buffer conditions to maintain protein interactions while ensuring efficient At1g30730 capture
Consider the use of crosslinking approaches to stabilize transient interactions
Validate results with reciprocal co-immunoprecipitation using antibodies against putative interacting partners
Sequential Immunodetection:
When performing multiple rounds of immunodetection on the same membrane:
Document complete stripping of previous antibodies before reprobing
Consider the order of detection based on antibody sensitivity (typically detecting lower abundance proteins first)
Validate that the stripping process does not affect the integrity of the immobilized proteins
Data Integration Table Example:
| Experimental Approach | Required At1g30730 Antibody Format | Compatible Partner Antibodies | Key Optimization Parameters |
|---|---|---|---|
| Co-immunofluorescence | Polyclonal (CSB-PA219368XA01DOA) | Organelle markers; other signaling proteins | Fixation method; antibody dilution; blocking reagent |
| Co-immunoprecipitation | Monoclonal combinations targeting specific domains | Antibodies against putative interacting proteins | Buffer ionic strength; detergent type/concentration |
| ChIP-seq analysis | ChIP-grade polyclonal antibodies | Histone modification antibodies | Crosslinking conditions; sonication parameters |
When investigating plant stress responses with At1g30730 antibodies, consider these evidence-based approaches:
Temporal Expression Profiling:
Collect samples at multiple time points post-stress treatment
Use Western blotting with At1g30730 antibodies to track protein level changes
Correlate protein expression with transcriptional changes using parallel RT-qPCR analysis
Spatial Distribution Analysis:
Employ immunohistochemistry or immunofluorescence using fixed tissue sections
Compare At1g30730 protein localization across different tissues under stress vs. control conditions
Validate immunolocalization results with GFP-fusion protein approaches if available
Functional Analysis Through Protein Interaction Studies:
Use At1g30730 antibodies for immunoprecipitation followed by mass spectrometry to identify stress-induced interaction partners
Compare interactomes under normal vs. stress conditions
Validate key interactions through techniques like bimolecular fluorescence complementation
Modification Status Assessment:
Investigate post-translational modifications under stress conditions using phospho-specific antibodies (if available)
Compare migration patterns in Western blots under different stress treatments
Consider 2D gel electrophoresis followed by immunoblotting to resolve modified forms
Research indicates that At1g30730 expression changes in response to certain experimental conditions, as shown in Table 2 of a study examining differential gene expression patterns . This suggests potential involvement in stress response pathways that warrant further investigation using these strategies.
Achieving optimal Western blot results with At1g30730 antibodies requires systematic optimization:
Sample Preparation:
Extract proteins using buffer systems that preserve At1g30730 integrity (typically containing protease inhibitors and phosphatase inhibitors if phosphorylation is relevant)
Determine optimal protein loading amounts (typically 20-50 μg total protein per lane for standard plant extracts)
Include appropriate positive controls (e.g., recombinant At1g30730 protein) and negative controls (e.g., extracts from knockout lines if available)
Electrophoresis and Transfer Parameters:
Use 12-15% SDS-PAGE gels for optimal resolution of the 158 amino acid At1g30730 protein
Consider gradient gels when analyzing potential protein complexes
Optimize transfer conditions based on protein size (typically 100V for 1 hour for proteins of this size)
Antibody Incubation:
Determine optimal primary antibody dilution through titration experiments (starting with manufacturer recommendations, typically 1:1000 for polyclonal antibodies)
Test different blocking agents (5% BSA vs. 5% non-fat milk) to minimize background
Optimize incubation temperature and duration (4°C overnight vs. room temperature for 1-2 hours)
Detection Optimization:
Select detection systems based on expected expression levels (chemiluminescence for standard detection, fluorescent secondary antibodies for quantitative analysis)
Optimize exposure times to prevent signal saturation
Consider signal amplification systems for low abundance detection
Quantitative Analysis:
Use housekeeping proteins appropriate for the experimental context as loading controls
Apply densitometry analysis with appropriate normalization
Ensure analysis is performed within the linear range of detection
For successful immunoprecipitation of At1g30730 and associated protein complexes:
Pre-immunoprecipitation Considerations:
Determine appropriate lysis buffer composition based on subcellular localization and interaction stability
Optimize cell/tissue disruption methods to maintain protein complex integrity
Establish appropriate protein concentration (typically 1-5 mg/ml) and volume for efficient immunoprecipitation
Antibody Selection and Coupling:
Compare the efficiency of different At1g30730 antibodies for immunoprecipitation
Consider using a combination of antibodies targeting different epitopes to maximize capture
For large-scale studies, covalently couple antibodies to solid support (e.g., using crosslinkers such as BS3 or DMP)
Immunoprecipitation Protocol Optimization:
Determine optimal antibody-to-protein ratio through titration experiments
Optimize incubation conditions (4°C overnight with gentle rotation is typically effective)
Establish appropriate washing stringency to remove non-specific interactions while preserving specific complexes
Validation and Analysis Approaches:
Confirm successful precipitation using Western blotting with a portion of the immunoprecipitated material
For interactome studies, process samples for mass spectrometry following established protocols
Include appropriate controls including:
IgG control (same species as the primary antibody)
Input sample (pre-immunoprecipitation lysate)
When possible, immunoprecipitation from knockout/knockdown lines
A typical immunoprecipitation workflow involves:
Lysate preparation (1-2 hours)
Pre-clearing with control IgG and protein A/G beads (1 hour)
Antibody incubation (overnight)
Bead capture (1-2 hours)
Washing steps (1-2 hours)
Elution and analysis (varies by downstream application)
Successful immunohistochemistry with At1g30730 antibodies requires attention to these critical parameters:
Tissue Preparation:
Optimize fixation methods (4% paraformaldehyde is standard, but test multiple fixatives)
Determine appropriate section thickness (typically 5-10 μm for plant tissues)
Consider antigen retrieval methods if epitope masking is suspected during fixation
Antibody Validation for IHC:
Test antibody specificity on known At1g30730-expressing tissues
Include appropriate negative controls (pre-immune serum, antibody pre-absorption with immunizing peptide)
Compare staining patterns with in situ hybridization or reporter gene expression if available
Protocol Optimization:
Determine effective blocking conditions to minimize non-specific binding
Establish optimal primary antibody dilution (typically starting at 1:100-1:500 for polyclonal antibodies)
Optimize incubation conditions (temperature, duration, humidity)
Select appropriate detection systems based on required sensitivity and analysis methods
Counterstaining and Visualization:
Choose counterstains that provide tissue context without interfering with specific signal
For co-localization studies, ensure secondary antibodies have minimal spectral overlap
Establish imaging parameters that capture specific signal while minimizing background
Quantitative Analysis Approaches:
Develop consistent scoring methods for signal intensity
Use digital image analysis software with appropriate thresholding
Include internal standardization for cross-sample comparisons
Given that At1g30730 has been studied in the context of plant signaling pathways, pay particular attention to its potential co-localization with known signaling components and membrane structures during immunohistochemical analysis.
When encountering non-specific binding with At1g30730 antibodies, implement this systematic troubleshooting approach:
Problem Diagnosis:
Characterize the pattern of non-specific binding:
Multiple unexpected bands in Western blots
Diffuse or universal staining in immunohistochemistry
High background in ELISA
Evaluate potential causes through controlled experiments:
Antibody quality issues (test new lot if available)
Sample preparation problems (optimize extraction/fixation)
Detection system sensitivity (adjust exposure/development time)
Optimization Strategies:
| Problem | Primary Causes | Solution Strategies |
|---|---|---|
| Multiple bands in Western blot | Cross-reactivity; protein degradation; post-translational modifications | Increase antibody dilution; optimize extraction buffers with protease inhibitors; use freshly prepared samples |
| High background in IHC | Insufficient blocking; excessive antibody concentration; endogenous peroxidase activity | Extend blocking time; increase blocking reagent concentration (5-10%); perform hydrogen peroxide quenching step |
| Non-specific signal in co-IP | Weak antibody-antigen interaction; improper washing stringency | Increase salt concentration in wash buffers; add mild detergents (0.1% Triton X-100); pre-clear lysates thoroughly |
Experimental Validation Approaches:
Peptide competition assay: Pre-incubate antibody with immunizing peptide before application
Knockout/knockdown control: Test antibody on tissues/cells with reduced/absent At1g30730 expression
Epitope-tagged protein expression: Compare antibody binding pattern with anti-tag antibody detection
Advanced Refinement Techniques:
Antibody purification through antigen-affinity chromatography
Cross-adsorption against related proteins to remove cross-reactive antibodies
Isotype-specific secondary antibody selection for reduced background
If non-specific binding persists, consider testing alternative antibodies targeting different epitopes of At1g30730, as molecular engineering approaches have demonstrated that binding specificity can be significantly improved through epitope selection optimization .
Proper interpretation of At1g30730 antibody data requires contextual analysis and rigorous controls:
Baseline Expression Context:
Establish normal At1g30730 expression patterns across:
Different developmental stages
Various tissue types
Standard growth conditions
Quantify typical expression levels in reference tissues to enable comparative analysis
Experimental Design Considerations:
Include appropriate sampling intervals to capture developmental dynamics
Establish statistical power through biological and technical replicates
Design time-course experiments to correlate At1g30730 expression with developmental transitions
Data Interpretation Framework:
Compare protein expression (antibody-based detection) with transcriptional data (RT-qPCR, RNA-seq)
Analyze subcellular localization changes during development
Correlate At1g30730 expression with known developmental markers
Contextual Analysis Example:
Previous research has shown differential expression patterns for At1g30730 in certain experimental conditions . When interpreting experimental data:
Upregulation might suggest involvement in specific developmental pathways
Changes in subcellular localization may indicate activation/inactivation
Co-expression with known developmental regulators may suggest functional relationships
Causal Relationship Assessment:
Distinguish correlation from causation through functional studies
Consider genetic approaches (knockout/knockdown/overexpression) to validate antibody-derived observations
Use pharmacological interventions when appropriate to disrupt suspected pathways
Integration with Broader Knowledge:
Compare findings with existing literature on At1g30730 and related proteins
Consider evolutionary conservation of expression patterns across related species
Develop testable hypotheses based on observed expression patterns
Incorporating At1g30730 antibodies into quantitative proteomics requires methodological rigor:
Sample Preparation Considerations:
Standardize protein extraction protocols to ensure consistent recovery
Include spike-in standards for absolute quantification when necessary
Consider subcellular fractionation to enrich for At1g30730-containing compartments
Antibody-Based Enrichment Strategies:
For targeted proteomics, optimize immunoprecipitation conditions for At1g30730 and interacting partners
Consider sequential immunoprecipitation to identify specific interaction complexes
Validate enrichment efficiency through Western blotting before mass spectrometry analysis
Quantitative Analysis Approaches:
| Method | Applications | Key Considerations for At1g30730 Studies |
|---|---|---|
| SILAC (Stable Isotope Labeling with Amino acids in Cell culture) | Comparative analysis of At1g30730 complexes under different conditions | Limited applicability in whole plants; consider cell culture systems |
| iTRAQ/TMT (Isobaric Tags for Relative and Absolute Quantitation) | Multiplexed analysis of At1g30730 expression across developmental stages | Requires careful experimental design for effective normalization |
| Label-free quantification | Broad applicability for comparing At1g30730 abundance | Requires rigorous normalization and statistical analysis |
| SRM/MRM (Selected/Multiple Reaction Monitoring) | Targeted quantification of specific At1g30730 peptides | Requires prior identification of reliable peptide targets |
Validation Requirements:
Confirm mass spectrometry identifications with orthogonal methods (Western blotting, immunofluorescence)
Validate protein-protein interactions through reciprocal co-immunoprecipitation
Correlate protein abundance changes with functional outcomes
Data Integration Approaches:
Map quantitative changes to protein interaction networks
Correlate protein abundance with post-translational modifications
Integrate proteomics data with transcriptomics and metabolomics for systems-level analysis
Similar approaches have been successfully employed in studies of plant signaling pathways, where quantitative proteomics revealed dynamic changes in protein complexes during signal transduction .
Systematic evaluation of different At1g30730 antibodies requires standardized comparative analysis:
Initial Assessment Parameters:
Antibody characteristics documentation:
Target epitope location and sequence
Host species and antibody type (polyclonal vs. monoclonal)
Production method and purification approach
Validated applications according to manufacturer
Validation documentation review:
Specificity data provided by manufacturer
Published literature using the antibody
Independent validation studies if available
Comparative Performance Testing:
| Parameter | Evaluation Method | Quantification Approach |
|---|---|---|
| Specificity | Western blotting against recombinant protein and plant extracts | Band pattern analysis; signal-to-noise ratio calculation |
| Sensitivity | Serial dilution of target protein | Limit of detection determination; EC50 calculation |
| Application versatility | Testing across multiple applications (WB, IP, IHC, ELISA) | Performance rating scale for each application |
| Lot-to-lot consistency | Testing multiple lots of the same antibody | Coefficient of variation calculation |
Standardized Testing Protocol Example:
Prepare identical samples from:
Wild-type Arabidopsis (positive control)
At1g30730 knockout/knockdown lines if available (negative control)
Recombinant At1g30730 protein (reference standard)
Process all samples in parallel using standardized protocols
Test each antibody under identical conditions:
Same blocking reagents and concentrations
Consistent incubation times and temperatures
Identical detection systems and exposure times
Quantitatively evaluate performance metrics:
Signal-to-background ratio
Detection limit
Dynamic range
Cross-reactivity profile
Documentation and Selection Criteria:
Create a comprehensive comparison table including all testing parameters
Weight performance criteria according to experimental requirements
Consider cost-effectiveness for long-term experimental planning
Document selected antibody characteristics for methods sections in publications
When comparing commercial antibodies like CSB-PA219368XA01DOA with other options such as the combination monoclonal sets (X-Q6NNH8-N/C/M) , prioritize the performance metrics most relevant to your specific experimental applications.
Advanced antibody engineering approaches offer significant potential improvements for At1g30730 research:
Current Limitations and Engineering Solutions:
Specificity Enhancement:
Implementation of phage display technologies to select high-specificity binding domains
Development of recombinant antibodies with optimized complementarity-determining regions (CDRs)
Application of deep mutational scanning to identify variants with improved specificity
Sensitivity Improvement:
Functional Capability Expansion:
Emerging Technologies with Potential Application:
Implementation Considerations:
Validation requirements for engineered antibodies in plant research
Cost-effectiveness compared to traditional antibody production
Technical expertise required for implementation in standard plant biology laboratories
The development of bispecific antibodies with enhanced specificity and functionality, as described in recent research , represents a particularly promising approach for improving At1g30730 detection in complex plant samples.
At1g30730 antibodies could significantly advance plant stress signaling research through these approaches:
Signaling Network Mapping:
Use At1g30730 antibodies in proximity labeling approaches (BioID, APEX) to identify proteins in spatial proximity during stress responses
Apply quantitative immunoprecipitation followed by mass spectrometry to characterize dynamic interaction networks
Employ antibody-based ChIP-seq to identify potential DNA-binding activities if relevant
Spatiotemporal Dynamics Analysis:
Track At1g30730 protein relocalization during stress responses using immunofluorescence
Quantify protein abundance changes across tissues and time points using quantitative Western blotting
Investigate post-translational modifications using modification-specific antibodies if available
Functional Studies:
Use antibodies to inhibit protein function in cell-free systems or through microinjection
Develop antibody-based biosensors to track protein conformation changes during signaling
Apply antibodies in protein-array technologies to identify novel interaction partners
Integration with Existing Knowledge:
Research has shown that At1g30730 expression can be altered in experimental conditions related to plant signaling studies . Based on this foundation, antibody-based approaches could:
Determine whether transcriptional changes correlate with protein-level changes
Identify post-translational modifications that may regulate protein function
Elucidate protein-protein interactions that may connect At1g30730 to known signaling pathways
Methodological Innovations:
Application of single-molecule imaging techniques using fluorescently labeled antibodies or antibody fragments
Development of antibody-based optogenetic tools for controlling protein function
Integration of antibody-based detection with spatial transcriptomics for comprehensive pathway analysis
Similar spatiotemporal analysis approaches have successfully identified key transcription factors in plant stress responses, as seen in recent studies , and could be applied to understand the potential role of At1g30730 in these pathways.
Computational methods offer powerful strategies to optimize At1g30730 antibody research:
Epitope Prediction and Antibody Design:
Application of machine learning algorithms to predict optimal epitopes based on:
Protein structure prediction (if 3D structure is unknown)
Surface accessibility analysis
Evolutionary conservation assessment
Post-translational modification site prediction
In silico antibody design approaches:
Computational modeling of antibody-antigen interactions
Optimization of binding affinity through virtual mutations
Design of multi-epitope recognition strategies
Active Learning for Improved Antibody Selection:
Recent research has demonstrated that active learning strategies can significantly improve antibody-antigen binding prediction . Applied to At1g30730:
Establish initial small-scale experimental dataset of antibody binding characteristics
Apply active learning algorithms to predict optimal antibody candidates
Iteratively test and refine predictions to identify optimal antibodies with minimal experimental investment
Data Integration and Analysis Enhancement:
| Computational Approach | Application to At1g30730 Research | Expected Benefit |
|---|---|---|
| Network analysis algorithms | Integration of At1g30730 into protein interaction networks | Functional context prediction |
| Image analysis automation | Quantification of immunohistochemistry/immunofluorescence results | Higher throughput, reduced subjective bias |
| Quantitative Western blot analysis tools | Standardized quantification of At1g30730 expression | Improved reproducibility and sensitivity |
| Molecular dynamics simulations | Prediction of antibody-antigen interaction dynamics | Enhanced specificity through binding mechanism understanding |
Implementation Pathway:
Establish collaborations between plant biologists and computational scientists
Develop plant-specific datasets to train machine learning models
Validate computational predictions through targeted experimental approaches
Iterate between computational prediction and experimental validation
Research has shown that active learning strategies can reduce the number of required experimental variants by up to 35% and speed up the learning process by 28 steps compared to random approaches . This efficiency gain could significantly accelerate At1g30730 research while reducing experimental costs.