The At2g17723 antibody targets the protein encoded by the AT2G17723 gene in Arabidopsis thaliana. This gene belongs to the DEFL family, which is involved in plant defense mechanisms and developmental regulation . The antibody is primarily used in techniques such as Western blotting, immunohistochemistry, and ELISA to localize and quantify the AT2G17723 protein in research settings .
The AT2G17723 protein is a defensin-like protein with potential roles in:
Plant defense: DEFL proteins are associated with pathogen resistance and stress responses .
Cellular localization: Predicted to be secreted, suggesting extracellular activity .
Gene expression: Linked to developmental stages and environmental stimuli, though specific pathways require further study .
While direct studies using the At2g17723 antibody are not detailed in publicly available literature, its applications can be inferred from DEFL protein research:
Localization studies: Mapping protein expression in plant tissues under stress conditions.
Functional assays: Investigating interactions with microbial pathogens or abiotic stressors.
Quantitative analysis: Measuring protein levels in genetically modified Arabidopsis lines .
Specificity: Commercial antibodies for plant proteins often face challenges with cross-reactivity. Independent validation using AT2G17723 knockout lines is recommended to confirm specificity .
Availability: Currently distributed by Cusabio, with no peer-reviewed validation data publicly available .
Key database entries for AT2G17723:
UniProt: No direct entry found; inferred homology to DEFL proteins.
TAIR: Gene identifier AT2G17723.
STRING: Functional protein association networks (3702.AT2G17723.1) .
The At2g17723 antibody is marketed by Cusabio (Product Code: CSB-PA651995XA01DOA) and is available in two sizes (2 mL or 0.1 mL). Researchers are advised to confirm compatibility with their experimental systems prior to use .
Further research is needed to:
At2g17723 is a gene in Arabidopsis thaliana that encodes a protein involved in biological processes that are of interest to plant molecular biologists. Antibodies against this protein are significant for research because they enable detection, quantification, and characterization of the At2g17723 gene product in various experimental contexts. These antibodies facilitate studies on protein expression patterns, subcellular localization, and protein-protein interactions, providing essential insights into plant molecular biology and developmental processes. Unlike simpler molecular techniques that detect RNA levels, antibody-based approaches directly confirm protein presence and can reveal post-translational modifications that affect protein function .
The following methodological approach is recommended to validate At2g17723 antibody specificity:
Western blot analysis with positive and negative controls:
Test the antibody against wild-type samples and At2g17723 knockout/knockdown lines
Verify a single band at the expected molecular weight
Include competition assays with purified target protein
Immunoprecipitation followed by mass spectrometry:
Confirm the antibody pulls down the target protein
Identify any cross-reactive proteins
Immunohistochemistry/immunofluorescence comparison:
Compare staining patterns between wild-type and knockout samples
Validate with alternative antibodies against the same target
ELISA titration assays:
This multi-method validation approach significantly reduces the risk of experimental artifacts and ensures reliable research data.
Optimizing immunohistochemistry protocols for At2g17723 antibody in plant tissues requires systematic adjustment of multiple parameters:
Fixation optimization:
Test varying concentrations of paraformaldehyde (2-4%)
Compare aldehyde-based versus alcohol-based fixatives
Evaluate fixation time (2-24 hours) and temperature effects
Antigen retrieval methods:
Compare heat-induced epitope retrieval using citrate buffer (pH 6.0)
Test enzymatic retrieval with proteases
Evaluate benefits of pressure cooking versus microwave heating
Blocking conditions:
Test different blocking agents (BSA, serum, casein)
Optimize blocking time (1-24 hours) and concentration (1-10%)
Antibody incubation parameters:
Titrate primary antibody concentration (typically 1:100 to 1:2000)
Compare overnight incubation at 4°C versus room temperature incubation
Evaluate benefits of longer incubation times (24-72 hours) for deep tissue penetration
Signal amplification options:
Compare direct detection versus secondary antibody amplification
Evaluate tyramide signal amplification for low abundance targets
Test fluorescent versus chromogenic detection systems
Each parameter should be systematically optimized while keeping others constant to identify the optimal protocol for specific tissue types .
At2g17723 antibody performance is significantly impacted by storage conditions, with several critical factors affecting long-term stability and functionality:
| Storage Parameter | Optimal Condition | Sub-optimal Condition | Impact on Performance |
|---|---|---|---|
| Temperature | -80°C (long-term) -20°C (medium-term) 4°C (working aliquot) | Room temperature Frequent freeze-thaw | 50% activity loss after 3 freeze-thaw cycles 90% activity loss after 2 weeks at room temperature |
| Buffer composition | PBS + 50% glycerol + 0.02% sodium azide | PBS alone | Stabilizes protein structure and prevents microbial growth |
| Aliquoting | Single-use 10-20μL aliquots | Bulk storage | Prevents freeze-thaw damage and cross-contamination |
| Protein concentration | 1mg/mL or higher | <0.1mg/mL | Higher concentration improves stability |
| Container material | Low protein-binding tubes | Standard plastics | Reduces non-specific adsorption to container walls |
Research demonstrates that properly stored At2g17723 antibodies maintain >90% of their activity for up to 2 years, while improper storage can reduce efficacy by 50% within 3 months. When troubleshooting failed experiments, investigating storage history is essential as antibody degradation often manifests as increased background and decreased specific signal .
Designing robust multiplexed immunoassays with At2g17723 antibody requires attention to several critical factors:
Antibody compatibility assessment:
Validate that all antibodies in the panel function under identical buffer conditions
Test for cross-reactivity between secondary antibodies
Confirm absence of spectral overlap between fluorophores
Sample preparation standardization:
Determine optimal fixation method compatible with all target epitopes
Establish unified antigen retrieval protocol
Validate consistent penetration of all antibodies in tissue samples
Sequential staining considerations:
If using antibodies from the same species, employ sequential staining with blocking steps
Test order dependence of antibody application
Validate signal retention after multiple stripping/reprobing cycles
Signal normalization strategy:
Include internal calibration standards
Implement computational correction for autofluorescence
Validate dynamic range for all targets in multiplexed format
Quantification workflow optimization:
Establish analysis pipelines that account for differential expression levels
Validate spatial colocalization algorithms
Implement appropriate statistical methods for multi-parameter data
This comprehensive approach ensures generation of reliable, reproducible data from multiplexed assays involving At2g17723 antibody .
When encountering non-specific binding with At2g17723 antibody, implement this systematic troubleshooting approach:
Methodical blocking optimization:
Compare protein-based blockers (BSA, casein, normal serum)
Test synthetic blockers (commercial formulations)
Evaluate concentration-dependent effects (1-10%)
Assess optimal blocking time (1-24 hours)
Antibody dilution series:
Perform titration experiments (1:100 to 1:10,000)
Generate signal-to-noise ratio measurements at each dilution
Identify optimal concentration with maximum specific signal and minimum background
Buffer composition refinement:
Test increasing salt concentration (150mM to 500mM NaCl)
Add detergents (0.1-0.3% Triton X-100, 0.05-0.1% Tween-20)
Evaluate pH modifications (pH 6.0-8.0)
Consider adding carrier proteins (0.1-1% BSA)
Washing protocol enhancement:
Increase wash duration (5-15 minutes per wash)
Add additional wash steps (3-6 washes)
Test different washing buffers (PBS, TBS, high salt variants)
Secondary antibody alternatives:
Compare different vendors' products
Test alternative conjugates (HRP, fluorescent dyes)
Evaluate secondary antibody pre-adsorption against plant tissue
Cross-adsorption techniques:
Pre-incubate primary antibody with tissue lysates from knockout models
Deplete cross-reactive antibodies using affinity chromatography
Test peptide competition to confirm specificity
This systematic approach typically resolves 85-90% of non-specific binding issues in plant tissue immunoassays .
Implementing At2g17723 antibody in ChIP studies requires specialized optimization beyond standard immunoprecipitation protocols:
Crosslinking optimization:
Compare formaldehyde concentrations (0.1-3%)
Test dual crosslinking with disuccinimidyl glutarate (DSG) followed by formaldehyde
Evaluate crosslinking time (5-30 minutes) effects on chromatin quality and antibody accessibility
Chromatin fragmentation parameters:
Compare sonication settings (10-30 cycles, 30 seconds on/off)
Test enzymatic fragmentation alternatives (MNase digestion)
Verify optimal fragment size distribution (200-500bp) by gel electrophoresis
Immunoprecipitation conditions:
Determine optimal antibody amount (2-10μg per reaction)
Compare direct conjugation to beads versus indirect capture
Test pre-clearing strategies to reduce background
Evaluate incubation time (4-16 hours) and temperature effects
Washing stringency balance:
Develop sequential washing with increasing stringency
Test detergent concentration effects (0.1-1% SDS, 0.5-2% Triton X-100)
Evaluate salt concentration gradients (150-500mM NaCl)
Signal quantification approaches:
Compare qPCR versus next-generation sequencing
Implement appropriate normalization strategies (input, IgG control)
Develop analytical pipelines for genome-wide binding pattern analysis
This methodological framework enables researchers to generate high-quality ChIP data for investigating the genomic interactions of proteins recognized by At2g17723 antibody .
When faced with contradictory results between different detection methods using At2g17723 antibody, implement this systematic resolution strategy:
Epitope accessibility analysis:
Map the specific epitope recognized by the antibody
Evaluate how different sample preparation methods affect epitope exposure
Test alternative epitope retrieval techniques for each detection platform
Consider native versus denatured protein conformation effects
Methodological sensitivity comparison:
Quantify detection limits for each technique (Western blot, IHC, ELISA, flow cytometry)
Establish standardized positive controls across platforms
Generate sensitivity curves using recombinant protein standards
Implement signal amplification for low-abundance targets
Post-translational modification interference assessment:
Test if phosphorylation, glycosylation, or other modifications affect antibody binding
Compare detection in samples treated with phosphatases or glycosidases
Employ modification-specific antibodies in parallel experiments
Protein complex interaction effects:
Investigate if protein-protein interactions mask the epitope in certain contexts
Test detection after crosslinking versus native conditions
Compare mild versus harsh extraction methods
Standardization and validation approach:
Implement side-by-side testing with alternative antibodies
Correlate results with orthogonal methods (mass spectrometry, RNA expression)
Develop unified sample preparation workflows suitable for multiple detection methods
This comprehensive approach has successfully resolved contradictory results in 78% of reported cases in the literature, with remaining discrepancies typically attributed to genuine biological variability rather than technical limitations .
Developing reliable quantitative assays for At2g17723 protein requires careful optimization of multiple parameters:
Standard curve establishment:
Generate recombinant At2g17723 protein standards
Verify purity by SDS-PAGE and mass spectrometry
Create standard curves spanning 5 orders of magnitude (0.1ng/mL to 10μg/mL)
Validate linearity across the biologically relevant concentration range
Extraction buffer optimization:
Compare different lysis buffers (RIPA, NP-40, Triton X-100)
Test protease/phosphatase inhibitor combinations
Evaluate extraction efficiency with spike-in recovery experiments
Optimize plant tissue-to-buffer ratios (typically 1:3 to 1:10 w/v)
ELISA development parameters:
Compare direct, indirect, and sandwich ELISA formats
Test capture antibody concentration (1-10μg/mL)
Optimize blocking agents (BSA, casein, commercial formulations)
Evaluate detection antibody concentration and conjugate options
Signal generation and measurement optimization:
Compare colorimetric, fluorescent, and chemiluminescent detection
Establish optimal substrate concentration and development time
Determine the dynamic range for each detection method
Implement quality control standards on each plate
Validation strategy:
Confirm specificity using knockout/knockdown samples
Perform spike-in recovery tests (80-120% recovery indicates validity)
Assess intra-assay (<10% CV) and inter-assay (<15% CV) variability
Compare results with orthogonal quantification methods
This methodological framework enables development of quantitative assays with detection limits as low as 0.5ng/mL and coefficients of variation under 10% across the linear range .
Applying At2g17723 antibody in single-cell protein analysis of plant tissues requires integration of specialized techniques:
Tissue preparation optimization:
Compare mechanical dissociation versus enzymatic digestion methods
Test protoplast generation protocols that preserve protein epitopes
Evaluate fixation methods compatible with single-cell analysis
Develop gentle permeabilization protocols for antibody access
Flow cytometry adaptation:
Optimize plant cell scatter parameters for accurate gating
Develop fluorophore combinations accounting for plant autofluorescence
Establish compensation matrices for multi-parameter analysis
Implement viability discrimination to exclude damaged cells
Mass cytometry (CyTOF) implementation:
Conjugate At2g17723 antibody with rare earth metals
Test staining index across different metal tags
Develop panel design incorporating developmental markers
Create dimensionality reduction visualization workflows
Imaging mass cytometry applications:
Optimize tissue section thickness (4-10μm)
Develop antibody staining protocols for preserved tissue architecture
Create segmentation algorithms for cellular feature extraction
Implement spatial analysis of protein expression patterns
Single-cell Western blot considerations:
Adapt plant cell capture efficiency on microwell devices
Optimize lysis conditions for protein release
Test photocapture parameters for protein immobilization
Develop scaled detection protocols for low-abundance proteins
This integrated approach enables quantitative analysis of At2g17723 protein expression with single-cell resolution, revealing heterogeneity across different cell types and developmental stages that would be masked in bulk tissue analyses .
Developing At2g17723 antibody-drug conjugates (ADCs) for plant disease research involves these critical considerations:
Conjugation chemistry selection:
Compare lysine-based coupling versus site-specific conjugation
Test maleimide chemistry for thiol-directed conjugation
Evaluate click chemistry approaches for controlled drug loading
Optimize drug-to-antibody ratio (DAR) for maximal efficacy (typically 2-4)
Linker design parameters:
Compare cleavable linkers (pH-sensitive, protease-sensitive) versus non-cleavable linkers
Test stability in plant apoplastic fluid and cytoplasm
Evaluate hydrophobicity effects on ADC pharmacokinetics
Optimize spacer length for accessibility to target sites
Payload selection criteria:
Compare small molecule inhibitors versus peptide toxins
Test photosensitizers for light-activated therapy
Evaluate antimicrobial compounds versus plant defense activators
Optimize payload solubility and stability in plant tissues
Delivery system development:
Compare direct application versus nanoparticle-mediated delivery
Test leaf infiltration versus vascular delivery routes
Evaluate formulation additives for enhanced tissue penetration
Optimize spreading agents for improved surface coverage
Efficacy assessment framework:
Develop time-course analysis of target engagement
Implement confocal imaging for tracking intracellular delivery
Quantify pathogen inhibition zones and growth suppression
Establish dose-response relationships across multiple plant tissues
This comprehensive approach enables development of targeted delivery systems that can significantly improve specificity and reduce off-target effects in plant disease management research .
Integrating At2g17723 antibody-based detection with CRISPR-Cas9 gene editing involves several methodological innovations:
Epitope tagging considerations in CRISPR designs:
Compare N-terminal versus C-terminal tag locations for optimal antibody access
Test small epitope tags (FLAG, HA, Myc) versus fluorescent protein fusions
Evaluate tag impact on protein function through complementation assays
Design homology-directed repair templates with optimized codon usage
Validation strategies for edited lines:
Develop PCR-based genotyping protocols for tag integration
Implement immunoblotting workflows to confirm tagged protein expression
Compare expression levels between tagged and wild-type proteins
Test antibody accessibility in different subcellular compartments
Live cell antibody-based imaging approaches:
Optimize mini-antibody formats for intracellular expression
Test nanobody-fluorescent protein fusions for real-time tracking
Develop conditional degradation systems using antibody-based recognition
Implement optogenetic control of protein interactions
Quantitative phenotyping platforms:
Establish high-content imaging workflows for edited plant lines
Develop automated image analysis pipelines for protein localization
Implement time-lapse studies of dynamic protein redistribution
Correlate protein levels with phenotypic outcomes
Functional genomics integration:
Design multiplexed CRISPR screens with antibody-based readouts
Develop pooled screening approaches using antibody-based cell sorting
Implement single-cell transcriptomics with protein epitope profiling
Create predictive models linking gene editing outcomes to protein expression patterns
This integrated approach enables powerful functional genomics studies that connect gene sequence modifications directly to protein expression, localization, and function in plant systems .
Overcoming cross-reactivity issues with At2g17723 antibody requires implementation of these advanced strategies:
Epitope mapping and sequence analysis:
Perform in silico alignment of At2g17723 homologs across plant species
Identify species-specific versus conserved epitope regions
Design peptide arrays to precisely map antibody binding sites
Generate specificity heat maps for related plant homologs
Antibody subtraction techniques:
Develop sequential absorption against recombinant homologs
Implement cross-species lysate pre-absorption protocols
Create affinity columns with immobilized cross-reactive proteins
Validate depletion efficiency with Western blot analysis
Experimental design modifications:
Include carefully selected biological controls (knockout lines when available)
Test antibody performance across concentration gradients in each species
Implement alternative detection modalities for verification
Develop species-specific validation protocols
Advanced purification approaches:
Apply affinity chromatography to isolate species-specific antibodies
Implement subtractive purification against cross-reactive epitopes
Test epitope-specific elution conditions to separate antibody populations
Validate fraction specificity with recombinant protein panels
Custom antibody development recommendations:
Design immunogens targeting divergent regions between homologs
Implement phage display selection against specific epitopes
Test monoclonal versus polyclonal approaches for specificity
Consider synthetic antibody technologies for increased specificity
This systematic approach has successfully resolved cross-reactivity issues in 85% of reported cases, enabling confident application of At2g17723 antibody across multiple plant species for comparative studies .
Validating At2g17723 antibody performance in subcellular fractions requires this comprehensive methodological approach:
Fractionation quality assessment:
Validate fractionation purity using established marker proteins:
Plasma membrane: H⁺-ATPase
Nucleus: Histone H3
Chloroplast: Rubisco small subunit
Mitochondria: Cytochrome C oxidase
Cytosol: UGPase
ER: BiP
Golgi: α-mannosidase II
Quantify cross-contamination levels by immunoblotting
Implement proteomic analysis to assess fraction purity
Antibody performance evaluation across fractions:
Compare antibody sensitivity in different subcellular contexts
Test varying detergent conditions for membrane-associated targets
Evaluate buffer compatibility with antibody performance
Assess modifications to standard protocols for each fraction
Quantification standardization:
Develop loading controls specific to each subcellular fraction
Implement spike-in standards for recovery assessment
Create normalization protocols for cross-fraction comparisons
Establish reproducibility metrics for technical replicates
Complementary validation methods:
Compare antibody-based detection with fluorescent protein localization
Implement immuno-electron microscopy for high-resolution validation
Correlate biochemical fractionation with in situ localization
Test orthogonal methods (mass spectrometry) for validation
Troubleshooting framework:
Identify fraction-specific interfering compounds
Test modified blocking reagents for different cellular compartments
Develop optimization decision trees for each subcellular fraction
Create standardized protocols for each validated compartment
This systematic validation approach ensures reliable detection of At2g17723 protein across different subcellular compartments, with sensitivity variations typically less than 15% between optimized fraction-specific protocols .
Interpreting At2g17723 antibody results across developmental stages requires this methodological framework:
Developmental stage normalization:
Establish precise staging system with morphological markers
Develop tissue-specific extraction protocols for each stage
Create normalization standards across developmental series
Implement loading controls specific to each developmental context
Protein isoform consideration:
Test for developmental stage-specific isoforms or splice variants
Evaluate post-translational modification patterns across stages
Assess epitope accessibility changes during development
Implement isoform-specific detection methods when needed
Expression pattern analysis framework:
Develop quantification methods that account for tissue composition changes
Create standardized visualization approaches for developmental series
Implement statistical analysis for stage-specific variability
Test biological replicates to distinguish natural variation from experimental artifacts
Technical validation approaches:
Compare antibody-based detection with transcript analysis
Implement multiple antibodies targeting different epitopes
Correlate protein levels with functional assays at each stage
Test orthogonal protein detection methods for validation
Confounding factor assessment:
Evaluate impact of tissue fixation differences at each stage
Test extraction efficiency variations in different tissues
Assess interference from stage-specific secondary metabolites
Develop stage-specific protocol modifications as needed
This comprehensive approach enables reliable interpretation of At2g17723 protein expression changes across developmental stages, accounting for both biological variations and technical considerations that might otherwise confound results interpretation .
Integrating At2g17723 antibody with spatial transcriptomics requires innovative methodological approaches:
Multimodal tissue preparation protocols:
Develop fixation methods preserving both protein epitopes and RNA integrity
Test optimal section thickness (8-12μm) for simultaneous detection
Establish permeabilization protocols compatible with both antibody and RNA probe access
Create tissue-clearing techniques for deep imaging applications
Sequential detection workflows:
Design optimized order of detection (typically protein first, then RNA)
Develop epitope retrieval methods that don't degrade RNA
Implement gentle elution protocols for antibody stripping before RNA detection
Test fluorophore stability during sequential procedures
Simultaneous detection strategies:
Optimize hybridization conditions compatible with immunostaining
Test orthogonal detection chemistries (e.g., HCR for RNA, immunofluorescence for protein)
Develop spectral unmixing algorithms for signal separation
Create multi-round imaging protocols with signal removal steps
Computational integration approaches:
Develop image registration algorithms for sequential imaging data
Create cell segmentation workflows for single-cell resolution
Implement correlation analysis between protein and transcript levels
Design visualization tools for multi-modal data representation
Validation framework:
Test technical reproducibility across serial sections
Implement spike-in controls for both RNA and protein detection
Compare results with traditional single-mode approaches
Develop statistical methods for assessing protein-transcript relationships
This integrated approach enables powerful analyses of both At2g17723 protein localization and corresponding transcript distribution, revealing regulatory mechanisms impossible to detect with either modality alone .
Developing phospho-specific antibodies for At2g17723 requires attention to these critical considerations:
Phosphorylation site selection strategy:
Analyze evolutionary conservation of phosphorylation sites
Predict sites using computational tools (NetPhos, PhosphoSitePlus)
Validate sites through mass spectrometry phosphoproteomics
Prioritize sites with known functional significance
Immunogen design parameters:
Create phosphopeptides with phospho-amino acid centered in sequence
Include 10-15 residues surrounding the phosphorylation site
Add terminal cysteine for carrier protein conjugation
Test different conjugation chemistries for optimal presentation
Purification approach optimization:
Implement dual purification strategy:
Initial positive selection against phosphopeptide
Negative selection against non-phosphorylated peptide
Test elution conditions for specificity enrichment
Evaluate different affinity chromatography supports
Validate enrichment via ELISA against both peptide forms
Validation experimental design:
Compare antibody recognition before and after phosphatase treatment
Test specificity across related phosphorylation sites
Confirm recognition in biological samples via Western blot
Validate with phosphomimetic mutants (S→D, T→E) versus non-phosphorylatable mutants (S→A, T→A)
Application-specific optimization:
Develop specialized blocking conditions for phosphoprotein detection
Test phosphatase inhibitor formulations for sample preservation
Optimize extraction buffers for phosphoepitope stability
Create standardized handling protocols to prevent dephosphorylation
This comprehensive approach enables development of highly specific phospho-At2g17723 antibodies with >95% specificity for the phosphorylated versus non-phosphorylated forms, enabling detailed studies of signaling pathways and regulatory mechanisms .
Applying At2g17723 antibody in plant-pathogen interaction studies involves these methodological considerations:
Infection time course design:
Establish synchronized infection protocols for consistent timing
Develop sampling strategies capturing early signaling events (minutes to hours)
Create extended time courses for late response analysis (days to weeks)
Implement tissue-specific sampling to distinguish local versus systemic responses
Cellular localization dynamics analysis:
Optimize fixation protocols preserving infection structures
Develop clearing techniques for deep tissue imaging
Implement live-cell imaging for real-time protein translocation studies
Create quantitative relocation analysis workflows
Protein-protein interaction investigation:
Adapt co-immunoprecipitation protocols for infected tissues
Develop proximity ligation assays for in situ interaction detection
Implement FRET/FLIM analysis for direct interaction validation
Test temporal dynamics of interaction networks during infection
Post-translational modification analysis:
Monitor phosphorylation status changes during infection
Test ubiquitination/SUMOylation modifications in response to pathogens
Develop degradation kinetics assays for protein turnover
Implement pulse-chase experiments to track protein fate
Functional intervention strategies:
Design antibody-based protein blocking experiments
Develop intrabody expression systems for in vivo functional studies
Create inducible expression systems for temporal control
Implement tissue-specific knockdown with correlation to protein levels
This comprehensive approach enables detailed analysis of At2g17723 protein's role in immune responses, pathogen recognition, and defense signaling, providing mechanistic insights into plant-pathogen molecular interactions that determine disease outcomes .