The At2g37700 Antibody is produced using recombinant DNA technology, a method that enables precise control over antibody specificity and scalability . Key attributes include:
| Attribute | Details |
|---|---|
| Uniprot ID | F4IR05 |
| Species | Arabidopsis thaliana (Mouse-ear cress) |
| Vendor | Cusabio |
| Product Code | CSB-PA260651XA01DOA |
| Size | 2ml/0.1ml (concentration not specified) |
| Format | Likely polyclonal or monoclonal (exact type unspecified) |
While the antibody’s production details are not explicitly described in available literature, recombinant antibody production typically involves cloning gene sequences into expression vectors, followed by heterologous expression in bacterial or mammalian systems .
The At2g37700 gene is implicated in stress responses and metabolic pathways. Notably, it has been linked to:
Aliphatic glucosinolate biosynthesis: Glucosinolates (GLSs) are sulfur-containing compounds critical for plant defense and stress tolerance .
Drought tolerance: Studies indicate that At2g37700 expression is modulated during dehydration stress, particularly in auxin-sensitive mutants .
A genome-wide association study (GWAS) and functional genomics experiments identified At2g37700 as differentially expressed under dehydration stress :
Gene Expression Patterns: At2g37700 expression is downregulated in iaa5,6,19 triple mutants (auxin-sensitive lines) during desiccation, suggesting its role in stress-responsive pathways .
Statistical Significance: RNA-seq data revealed a p-value of 0.0005 for At2g37700 in dehydration-stressed seedlings, highlighting its importance in drought adaptation .
The At2g37700 Antibody is employed to:
Quantify Protein Abundance: Western blotting or immunoprecipitation assays to measure At2g37700 protein levels under stress conditions.
Study Subcellular Localization: Immunofluorescence microscopy to determine spatial distribution within plant tissues.
Map Protein Interactions: Co-immunoprecipitation (Co-IP) assays to identify interacting partners in stress-response pathways.
While At2g37700 has been associated with glucosinolate biosynthesis and drought responses, several questions remain unresolved:
Epitope Specificity: The antibody’s binding site (paratope) and epitope (on At2g37700) are not characterized, limiting insights into its cross-reactivity .
Functional Mechanism: The precise biochemical role of At2g37700 in GLS synthesis or drought tolerance requires further investigation.
The At2g37700 Antibody’s utility can be contextualized against other Arabidopsis antibodies targeting stress-related genes:
Validating antibody specificity for At2g37700 (HDA9) requires a binary validation approach that tests both positive and negative expression systems. This approach confirms that the antibody recognizes the target antigen in its native environment without cross-reacting with other biomolecules.
For HDA9 antibody validation, implement the following strategy:
Endogenous Expression Testing: Compare antibody reactivity in tissues/cells known to express HDA9 versus those with low or no expression. This approach provides a clear yes/no answer regarding specific antigen recognition .
Genetic Knockout Controls: Use hda9 mutant lines alongside wild-type Arabidopsis as definitive positive and negative controls. The complete absence of signal in the knockout confirms antibody specificity .
Orthogonal Method Verification: Verify expression data using secondary techniques such as RT-qPCR or RNA-seq to confirm that observed antibody reactivity patterns match transcript levels .
Application-Specific Validation: Test the antibody separately for each intended application (Western blot, ChIP, immunofluorescence), as specificity in one application doesn't guarantee performance in others .
When performing Western blot validation, always include proper loading controls (such as β-Actin) to ensure sample quality doesn't lead to misinterpretation of results. For immunohistochemistry, tissues containing both HDA9-expressing and non-expressing cells provide powerful internal controls .
Robust experimental design for At2g37700 (HDA9) antibody applications requires carefully selected controls to ensure reliable interpretation of results.
Recommended positive controls:
Wild-type Arabidopsis tissues with confirmed HDA9 expression (based on published RNA-seq data)
Transgenic lines expressing tagged HDA9 (such as pHDA9::HDA9-3xFLAG) that can be detected with both the HDA9 antibody and anti-FLAG antibodies
Cell types with known high expression based on tissue-specific transcriptome databases
Recommended negative controls:
hda9 knockout mutant lines showing complete absence of the protein
Tissues where HDA9 expression is naturally absent or minimal
Pre-immune serum or isotype control antibodies to establish baseline non-specific binding
Additional validation controls:
Peptide competition assays where the antibody is pre-incubated with the immunizing peptide
Secondary antibody-only controls to detect non-specific binding
For ChIP experiments, include input controls and IgG controls to establish background signal levels
Include loading controls (β-Actin, GAPDH, or total histone H3) when performing Western blot analyses to normalize protein levels across samples and verify sample quality .
The successful application of At2g37700 (HDA9) antibodies across different experimental techniques requires specific optimization for each method:
Western Blot Optimization:
Blocking conditions: 5% non-fat dry milk in TBST is typically sufficient, but BSA may reduce background for phospho-specific antibodies
Antibody dilution: Start with manufacturer recommendations (typically 1:1000), then optimize
Incubation time/temperature: Primary antibody incubation at 4°C overnight often improves specific binding
Detection system: ECL Plus Western Blotting Detection System works effectively for plant proteins
Chromatin Immunoprecipitation (ChIP) Optimization:
Crosslinking conditions: 1% formaldehyde for 10 minutes at room temperature is standard for histone modifications
Sonication parameters: Optimize to achieve chromatin fragments of 200-500 bp
Antibody amount: Typically 2-5 μg per IP reaction
Washing stringency: Critical for reducing background without losing specific signal
Library preparation: The Ovation Ultralow DR Multiplex System has been successfully used for HDA9 ChIP-seq
Immunohistochemistry/Immunofluorescence Optimization:
Fixation method: 4% paraformaldehyde is standard, but may require adjustment
Antigen retrieval: May be necessary for some plant tissues
Antibody dilution: Typically requires higher concentration than Western blot
Detection system: Fluorescence-based detection offers quantification advantages
For quantitative applications like ChIP-seq, replicates are essential, and advanced normalization strategies may be required to account for technical variation between samples .
Investigating protein-protein interactions involving At2g37700 (HDA9) requires sophisticated approaches that build upon the successful identification of the HDA9-PWR complex. Based on published methodologies, the following integrated strategy is recommended:
Immunoaffinity Purification coupled with Mass Spectrometry (IP-MS):
Generate transgenic Arabidopsis lines expressing epitope-tagged HDA9 under its native promoter (pHDA9::HDA9-3xFLAG) in the hda9 mutant background to ensure functionality
Perform affinity purification using anti-FLAG beads to isolate HDA9 and its associated proteins
Analyze purified proteins using multidimensional protein identification technology mass spectrometry
Include wild-type Col-0 plants as negative controls to identify non-specific binding proteins
Validation of Interactions using Reciprocal Co-Immunoprecipitation:
Generate additional transgenic lines expressing epitope tags on candidate interacting proteins (such as PWR-FLAG)
Create F1 plants expressing both HA-tagged HDA9 and FLAG-tagged interacting proteins
Perform co-IP experiments using anti-FLAG beads to pull down the interacting protein
Detect co-precipitation of HDA9 with anti-HA antibody on Western blots
Functional Validation through ChIP Analysis:
To determine if potential interacting proteins co-localize with HDA9 on chromatin:
Perform ChIP-qPCR using antibodies against both HDA9 and the interacting protein
Test for enrichment at the same genomic loci, focusing on promoter regions of active genes
Compare histone modification patterns (particularly H3K27ac) in wild-type, hda9, and interacting protein mutant lines
This comprehensive approach has successfully identified PWR as a functional partner of HDA9, demonstrating that they bind to the same genomic regions and cooperatively regulate H3K27 acetylation levels .
When faced with contradictory ChIP-seq data regarding histone modifications regulated by At2g37700 (HDA9), researchers should implement a systematic troubleshooting and validation strategy:
Technical Validation:
Antibody Specificity Re-evaluation:
ChIP Protocol Assessment:
Systematically analyze each step of the ChIP protocol (crosslinking, sonication, immunoprecipitation, washing, library preparation)
Evaluate sonication efficiency by analyzing fragment size distribution
Test multiple ChIP protocols in parallel to identify variables affecting results
Biological Validation:
Orthogonal Approaches:
Correlate ChIP-seq results with RNA-seq data to evaluate functional relevance
Perform ChIP-qPCR on selected targets to verify ChIP-seq findings
Use alternative techniques like CUT&RUN or CUT&Tag that may offer higher signal-to-noise ratios
Genetic Validation:
Data Analysis Refinement:
Apply multiple peak-calling algorithms (MACS, HOMER, etc.) and compare results
Utilize more stringent statistical thresholds (p<0.001 instead of p<0.05)
Implement bias correction for factors like GC content and chromatin accessibility
When analyzing contradictory results, focus particularly on the finding that HDA9 preferentially associates with active genes rather than silent genes, contrary to the conventional view of HDACs as transcriptional repressors . This unexpected association with DNase I hypersensitive sites and active promoters may explain some apparent contradictions in the data .
Optimizing epitope tagging strategies for At2g37700 (HDA9) requires careful consideration to maintain native protein functionality while enabling sensitive detection. Based on successful published approaches, consider the following optimization framework:
Tag Selection and Positioning:
Tag Type Selection:
Tag Positioning:
Functional Validation:
Complementation Testing:
Molecular Function Assessment:
Expression Control:
Promoter Selection:
Expression Level Monitoring:
Quantify tagged protein expression relative to endogenous levels
Too high expression may cause artifacts, while too low may be insufficient for detection
Western blot analysis with antibodies against both the tag and HDA9 can verify expression levels
This comprehensive approach ensures that the tagged HDA9 protein behaves as similarly as possible to the native protein, providing confidence in experimental results while enabling powerful techniques like ChIP-seq and IP-MS .
Performing ChIP-seq with At2g37700 (HDA9) antibodies requires specific protocol adaptations compared to standard histone modification ChIP-seq:
Crosslinking and Chromatin Preparation:
Crosslinking Conditions:
HDA9 typically requires stronger crosslinking than histone modifications
Implement dual crosslinking: 1.5 mM EGS (ethylene glycol bis-succinimidyl succinate) for 30 minutes followed by 1% formaldehyde for 10 minutes
This enhances capture of protein-protein interactions in addition to protein-DNA interactions
Chromatin Fragmentation:
Optimize sonication parameters for slightly larger fragments (300-500 bp) than typical histone ChIP (200-300 bp)
Monitor fragmentation efficiency using agarose gel electrophoresis
Over-sonication can destroy epitopes on transcription factors and chromatin modifiers
Immunoprecipitation:
Antibody Amounts:
HDA9 ChIP typically requires 3-5 μg of antibody per IP reaction (vs. 1-2 μg for histone modifications)
Pre-clearing chromatin with protein A/G beads reduces background
Include IgG controls at the same concentration as the HDA9 antibody
Incubation Conditions:
Extend primary antibody incubation to overnight at 4°C with gentle rotation
Use low-binding tubes to prevent antibody loss
Consider adding BSA (0.1-0.5%) to reduce non-specific binding
Washing and Elution:
Washing Stringency:
Implement more stringent washing conditions than histone ChIP
Include additional high-salt washes (500 mM NaCl) to reduce background
Monitor wash stringency carefully, as over-washing can reduce signal
DNA Recovery:
Implement RNase treatment before proteinase K digestion
Consider phenol-chloroform extraction for higher purity
Include carrier (glycogen) during precipitation to improve recovery of low-abundance DNA
Library Preparation and Sequencing:
Library Construction:
Data Analysis:
These modifications address the specific challenges of performing ChIP-seq with transcriptional regulators like HDA9 compared to abundant histone proteins.
Integrating RNA-seq and ChIP-seq data provides powerful insights into At2g37700 (HDA9) function in gene regulation. The following methodological framework optimizes this integration:
Experimental Design for Integration:
Sample Consistency:
Key Comparisons:
Data Analysis Pipeline:
Individual Data Processing:
Integrative Analysis:
Categorize genes into direct targets (HDA9 bound + differentially expressed) and indirect targets
Generate genome browser visualization showing RNA-seq and ChIP-seq tracks at key loci
Calculate correlation between H3K27ac changes and expression changes
Functional Correlation Analysis:
Gene Categorization by HDA9 Binding and Expression:
| Category | HDA9 Binding | Expression in hda9 | Number of Genes | Percentage |
|---|---|---|---|---|
| Direct activated | Yes | Decreased | Calculate | Calculate |
| Direct repressed | Yes | Increased | Calculate | Calculate |
| Indirect | No | Changed | Calculate | Calculate |
| Bound not regulated | Yes | Unchanged | Calculate | Calculate |
| Not bound or regulated | No | Unchanged | Calculate | Calculate |
Histone Acetylation Correlation:
Quantify H3K27ac changes at HDA9-bound vs. non-bound genes
Compare acetylation changes between hda9 and pwr mutants at shared targets
Correlate acetylation changes with expression changes
Gene Ontology Analysis:
Perform GO analysis using agriGO (http://bioinfo.cau.edu.cn/agriGO/)[3]
Compare GO terms between direct and indirect targets
Identify biological processes most affected by HDA9 regulation
This integrated approach has revealed that HDA9 preferentially binds to active genes rather than silent genes, contrary to the conventional view of HDACs as transcriptional repressors . Furthermore, it showed that HDA9 and PWR bind to the same genomic regions and cooperatively regulate H3K27 acetylation levels, providing mechanistic insight into their function .
Optimizing immunoprecipitation (IP) efficiency with At2g37700 (HDA9) antibodies is crucial for successful protein complex identification. Based on published methodologies that successfully identified the HDA9-PWR complex, the following parameters should be systematically optimized:
Extract Preparation:
Plant Material Selection:
Young, actively growing tissue typically yields better results for chromatin-associated proteins
Consider developmental stages where HDA9 is most active
Scale up material (5-10g) to compensate for potentially low abundance
Extraction Buffer Optimization:
Test multiple extraction buffers with varying salt concentrations (100-300 mM)
Include protease inhibitors (complete, EDTA-free) to prevent degradation
Add phosphatase inhibitors if studying phosphorylation status
Consider including low concentrations of detergents (0.1% NP-40) to aid solubilization
Immunoprecipitation Conditions:
Antibody Selection and Amount:
Incubation Parameters:
Test different incubation times (2h vs. overnight)
Compare incubation temperatures (4°C vs. room temperature)
Use gentle rotation rather than shaking to preserve complexes
Washing and Elution:
Washing Stringency Optimization:
Develop a washing strategy with gradually increasing stringency
Test different salt concentrations (150-500 mM)
Compare detergent concentrations in wash buffers (0.1-1% Triton X-100)
Elution Method Comparison:
For tagged proteins: competitive elution with FLAG peptide
For native IP: low pH glycine, SDS buffer, or boiling in sample buffer
Monitor elution efficiency with multiple elution steps
Validation and Analysis:
Western Blot Verification:
Analyze 5-10% of IP sample by Western blot before mass spectrometry
Probe for HDA9 to confirm successful precipitation
Include input, unbound, and IP fractions to assess efficiency
Mass Spectrometry Sample Preparation:
In-gel digestion vs. on-bead digestion protocols
Optimize peptide recovery and cleanup methods
Consider cross-linking antibodies to beads for cleaner samples
IP Efficiency Measurement:
The successful application of these optimized parameters enabled the identification of PWR as the most enriched interacting protein with HDA9, with 27 unique peptides detected in the IP-MS analysis . Reciprocal IP-MS of PWR-FLAG also purified HDA9, confirming the interaction .
Integrating At2g37700 (HDA9) antibodies into multi-omics approaches provides comprehensive insights into epigenetic regulation networks. The following methodological framework outlines how to effectively combine multiple techniques:
ChIP-seq and RNA-seq Integration:
Unified Experimental Design:
Correlation Analysis:
Proteomics Integration:
IP-MS for Protein Complex Identification:
Validation Through Reciprocal IP:
Chromatin Accessibility Integration:
ATAC-seq or DNase-seq Correlation:
Data Integration Framework:
| Data Type | Primary Analysis | Integration Points | Biological Insight |
|---|---|---|---|
| ChIP-seq | HDA9 binding sites | Overlay with expression changes | Direct vs. indirect regulation |
| RNA-seq | Differential expression | Correlate with binding and acetylation | Functional outcomes of binding |
| IP-MS | Protein interactions | Connect to genomic targets | Mechanism of target selection |
| ATAC-seq | Accessibility changes | Compare with binding patterns | Chromatin state influence on binding |
| ChIP-seq (H3K27ac) | Acetylation changes | Link to expression changes | Mechanism of transcriptional regulation |
This integrated approach has revealed unexpected insights into HDA9 function, including its preferential association with active genes rather than silent genes, contrary to the conventional view of HDACs as transcriptional repressors . Furthermore, integration of ChIP-seq data for HDA9 and histone acetylation revealed that HDA9-bound genes show significantly higher increases in H3K27ac in hda9 and pwr mutants compared to non-bound genes, demonstrating the direct deacetylase activity of the complex at its genomic targets .
When using At2g37700 (HDA9) antibodies in plant chromatin studies, implementing robust controls is critical for generating reliable and interpretable data. The following comprehensive control framework addresses the specific challenges of plant chromatin research:
Antibody Specificity Controls:
Binary Validation Controls:
Cross-Reactivity Assessment:
Test antibody reactivity against closely related HDACs in Arabidopsis
Perform peptide competition assays to confirm binding specificity
Include isotype control antibodies matched to the HDA9 antibody
ChIP-seq Technical Controls:
Input Controls:
Process chromatin before immunoprecipitation as a reference for genomic DNA abundance
Use input for normalization of ChIP signal to account for biases in chromatin preparation
Sequence input at similar depth to ChIP samples
Negative IP Controls:
Perform parallel IPs with non-specific IgG matched to the host species of the HDA9 antibody
Include no-antibody controls to assess bead background
For tagged HDA9, perform IPs in untagged plants with the same tag antibody
Positive Controls:
Biological Validation Controls:
Genetic Complementation:
Known Target Validation:
Perform ChIP-qPCR on established HDA9 targets before proceeding to ChIP-seq
Include both positive regions (HDA9-bound) and negative regions (unbound)
Validate ChIP-seq peaks by targeted ChIP-qPCR on selected loci
Data Analysis Controls:
Peak Calling Stringency:
Replication and Validation:
This comprehensive control framework addresses the specific challenges of plant chromatin studies, including tissue heterogeneity, cell wall barriers to crosslinking, and potential plant-specific artifacts, ensuring robust and reproducible results when using At2g37700 antibodies.
When researchers encounter inconsistent results with At2g37700 (HDA9) antibodies across different experimental platforms, a systematic troubleshooting approach is essential. The following decision tree and methodological framework address common sources of inconsistency:
Antibody-Related Factors:
Epitope Accessibility Variation:
Antibody Quality Assessment:
Lot-to-lot variation can cause inconsistent results
Solution: Document lot numbers and perform validation for each new lot
Consider creating large batches of validated antibody for long-term projects
Sample Preparation Variables:
Fixation and Extraction Optimization:
Crosslinking efficiency varies between applications and can affect epitope recognition
Solution: Systematically test multiple fixation protocols (duration, concentration)
For IP applications, compare native extraction versus crosslinked chromatin
Tissue/Cell Type Differences:
HDA9 expression and complex formation may vary between tissues
Solution: Verify HDA9 expression in each tissue type via RT-qPCR
Maintain strict consistency in developmental stage and growth conditions
Technical Platform Adjustments:
Platform-Specific Protocol Modifications:
| Platform | Common Issue | Optimization Strategy |
|---|---|---|
| Western Blot | Low signal | Increase antibody concentration; optimize transfer conditions |
| ChIP-seq | High background | More stringent washing; pre-adsorption of antibody |
| Immunostaining | Non-specific signal | Optimize blocking; use fluorescence over chromogenic detection |
| IP-MS | Low recovery | Scale up input material; optimize extraction buffer |
Cross-Platform Validation Approach:
Implement a multi-method validation for key findings
Confirm protein interactions by both co-IP and bimolecular fluorescence complementation
Validate ChIP-seq results with targeted ChIP-qPCR
Systematic Troubleshooting Decision Tree:
Define the inconsistency precisely:
Is it complete failure in one platform or quantitative differences?
Is it specific to certain tissues or conditions?
Is it reproducible or sporadic?
Isolate variables through controlled experiments:
Test the same sample preparation with different antibody lots
Test the same antibody with different sample preparations
Include known positive controls that should work across all platforms
Implement reference standards:
For Western blot: Include recombinant HDA9 protein as a standard
For ChIP: Include spike-in control chromatin from another species
For all applications: Process wild-type and hda9 mutant samples in parallel
Consider biological complexities:
When researchers have identified the source of inconsistency, they should document the optimized protocols thoroughly, including all critical parameters that influence reproducibility. This approach has successfully resolved contradictions in HDA9 studies, particularly regarding its unexpected association with active rather than repressed genes .
Emerging antibody technologies offer promising avenues to overcome current limitations in At2g37700 (HDA9) research, potentially revealing new insights into epigenetic regulation in plants. These innovative approaches include:
Single-Domain Antibodies and Nanobodies:
Advantages for HDA9 Research:
Smaller size (15 kDa vs. 150 kDa for conventional antibodies) enables better penetration into plant tissues
Greater stability under varying buffer conditions enhances versatility across applications
Recognition of epitopes inaccessible to conventional antibodies may reveal hidden aspects of HDA9 function
Implementation Strategy:
Generate camelid nanobodies against purified HDA9 protein
Create intrabodies that can be expressed in vivo to track HDA9 in living plant cells
Develop nanobody-based proximity labeling to map HDA9 interactome with higher resolution
Recombinant Antibody Fragments:
Application to HDA9 Studies:
Implementation Benefits:
Standardization reduces batch-to-batch variation
Epitope tagging can be minimized or eliminated
Antibody properties can be engineered for specific applications
Proximity Labeling Technologies:
Integration with Antibody Approaches:
Implementation Strategy:
Compare proximity labeling in wild-type vs. hda9 mutant as specificity control
Identify condition-specific interactors across development or stress responses
Map the spatial organization of HDA9-PWR complex on chromatin
Antibody-Free Alternatives:
CRISPR-Based Tagging:
Endogenous tagging of HDA9 to avoid overexpression artifacts
Create allelic series with domain-specific tags to dissect protein function
Implement degron tags for inducible protein degradation to study temporal dynamics
DNA-Programmed Affinity Reagents:
Develop aptamers with specificity for HDA9
Create SOMAmers (Slow Off-rate Modified Aptamers) for higher affinity
Implement peptide nucleic acid (PNA)-based affinity reagents for improved stability
These emerging technologies will enable researchers to address fundamental questions about HDA9 function that remain challenging with conventional antibodies, particularly regarding the temporal dynamics of complex formation, the chromatin environment at binding sites, and the complete interactome under various conditions.
Standardization is critical for improving reproducibility in At2g37700 (HDA9) antibody-based experiments across different laboratories. The following comprehensive standardization framework addresses key areas requiring harmonization:
Antibody Validation Standards:
Mandatory Validation Criteria:
Validation Reporting Requirements:
Standardized validation data format including western blots showing specificity
Antibody registry entries with unique identifiers
Recombinant Antibody Network integration where applicable
Detailed methods sections in publications that include catalog numbers, lot numbers, and dilutions
Experimental Protocol Standardization:
ChIP-seq Protocol Harmonization:
Standardized crosslinking conditions (concentration, time, temperature)
Consistent chromatin fragmentation parameters (size range verification)
Uniform IP conditions (antibody amount, incubation time)
Standardized washing stringency and number of washes
Western Blot Protocol Standardization:
Defined sample preparation (extraction buffer, protein quantification)
Standardized blocking conditions (agent, concentration, time)
Specified antibody dilutions and incubation parameters
Consistent detection methods and image acquisition settings
Data Analysis Standardization:
ChIP-seq Analysis Pipeline:
Quantitative Western Blot Analysis:
Linear dynamic range verification
Standardized normalization to validated loading controls
Uniform quantification methods for band intensity
Statistical analysis requirements for replicates
Reference Materials and Controls:
Shared Reference Standards:
Standardized Control Framework:
Implementation Strategy:
Community Standards Development:
Establish working groups through plant research organizations
Create detailed protocols through collaborative efforts
Develop benchmarking datasets for analytical pipeline validation
Standardization Enforcement Mechanisms:
Journal requirement for standardized validation documentation
Data repositories requiring standardized metadata
Funding agency mandates for standardization adherence
Training and Education:
Workshops on standardized protocols
Online resources and video protocols
Interlaboratory proficiency testing