The designation "At5g61310" corresponds to a systematic gene identifier in the Arabidopsis Information Resource (TAIR) database. This gene encodes a hypothetical protein with no documented antibody development efforts in any peer-reviewed publications, patents, or commercial antibody catalogs indexed in the provided sources .
PubMed Central (PMC) articles focus on antibodies targeting human pathogens (e.g., HIV-1), therapeutic targets (e.g., C5aR), or methodological validations. None mention plant-derived antibodies or Arabidopsis thaliana proteins.
Commercial/Technical Guides discuss antibody engineering, secondary antibody applications, and patent claims for human therapeutic targets. No Arabidopsis-related antibodies are cited.
General References describe antibody biology but lack plant-specific examples.
Gene Function: At5g61310 is annotated as a "protein of unknown function" in TAIR, reducing its likelihood of being targeted for antibody development.
Antibody Accessibility: Plant-specific antibodies are less common in commercial markets compared to human/mammalian targets.
The identifier "At5g61310 Antibody" may represent a proprietary or unpublished reagent not cataloged in public databases.
Confirm whether the identifier refers to:
A custom antibody from a specific study (unreported in indexed literature).
A gene-editing tool (e.g., CRISPR guide RNA) mislabeled as an antibody.
Verify the Gene Identifier: Cross-check TAIR (https://www.arabidopsis.org) for updated annotations or aliases.
Contact Specialty Vendors: Inquire with plant biology-focused companies (e.g., Agrisera, PhytoAB) about custom antibody services.
Explore Orthologs: If At5g61310 has homologs in other species (e.g., rice, maize), search for antibodies against those proteins.
At5g61310 is a gene locus in Arabidopsis thaliana that encodes a protein of significant interest to plant scientists. While similar in approach to other plant antibody studies, the specificity of antibodies raised against this particular protein enables researchers to conduct detailed protein localization, expression analysis, and functional studies . Monoclonal antibodies targeting specific plant proteins like At5g61310 are generated using techniques similar to those used for other model organisms, where the protein or a peptide fragment is used as an immunogen to elicit an immune response in host animals (typically mice) . These antibodies are particularly valuable in detecting native protein conformations and for studying protein-protein interactions in cellular contexts.
When designing an immunoblot experiment with At5g61310 antibody, follow these methodological steps:
Sample preparation: Extract total protein from Arabidopsis tissues using an appropriate buffer containing protease inhibitors.
Protein separation: Separate proteins via SDS-PAGE using a gradient gel (10-12% is often suitable for most plant proteins).
Transfer: Transfer proteins to a PVDF or nitrocellulose membrane.
Blocking: Block the membrane with 5% non-fat dry milk or 3% BSA in TBST.
Primary antibody incubation: Dilute At5g61310 antibody (typically 1:1000 to 1:2000) in blocking buffer and incubate overnight at 4°C.
Washing: Wash the membrane 3-4 times with TBST.
Secondary antibody: Incubate with an appropriate HRP-conjugated secondary antibody (anti-mouse IgG if using a mouse monoclonal) .
Detection: Visualize using chemiluminescence detection reagents.
Include both positive controls (recombinant At5g61310 protein if available) and negative controls (samples from knockout lines) to validate antibody specificity .
Validating antibody specificity is crucial for reliable experimental outcomes. For At5g61310 antibody, consider these key validation steps:
Genetic validation: Test antibody reactivity in at5g61310 knockout or knockdown lines, which should show reduced or absent signal.
Recombinant protein testing: Compare antibody reactivity against purified recombinant At5g61310 protein.
Peptide competition assay: Pre-incubate the antibody with the immunizing peptide before application to samples; this should block specific binding.
Cross-reactivity assessment: Test the antibody against closely related proteins to ensure specificity.
Multiple technique concordance: Verify that results from different techniques (western blot, immunoprecipitation, immunofluorescence) are consistent.
This rigorous validation approach follows standard experimental design principles that maximize confidence in antibody specificity .
Optimizing immunoprecipitation (IP) protocols for At5g61310 requires careful methodological considerations:
Crosslinking optimization: When studying transient protein interactions, optimize formaldehyde concentration (typically 0.1-1%) and crosslinking time (5-15 minutes) to preserve native interactions without excessive crosslinking.
Lysis buffer selection: Test multiple buffer compositions varying detergent type (NP-40, Triton X-100, CHAPS) and concentration (0.1-1%) to maximize protein extraction while maintaining interactions.
Antibody immobilization: Compare direct antibody addition with pre-immobilizing antibodies on protein A/G beads to determine which approach yields higher specificity.
Pre-clearing strategy: Implement sample pre-clearing with uncoated beads to reduce non-specific binding.
Wash stringency gradient: Establish a wash stringency gradient to determine optimal conditions that remove non-specific interactions while preserving authentic binding partners.
Elution method comparison: Compare various elution methods (pH, competitive elution with peptides, or SDS) for optimal recovery.
This approach aligns with experimental design principles emphasizing systematic variable testing to achieve reproducible results .
For chromatin immunoprecipitation (ChIP) experiments with At5g61310 antibody, implement the following experimental design strategies:
Experimental controls design:
Include input chromatin samples (pre-immunoprecipitation)
Perform mock immunoprecipitation with non-specific IgG
Include positive controls (regions known to bind the protein)
Include negative controls (regions not expected to bind)
Crosslinking optimization matrix:
Formaldehyde Concentration | Crosslinking Time | Application |
---|---|---|
0.5% | 5 minutes | Abundant proteins with strong DNA interactions |
1% | 10 minutes | Standard condition for most applications |
1.5% | 15 minutes | Low-abundance proteins or weak interactions |
Sonication parameters: Optimize sonication to generate DNA fragments of 200-500 bp, essential for resolution.
Antibody titration: Determine the minimum antibody concentration required for efficient immunoprecipitation to reduce background.
Sequential ChIP consideration: For proteins in complexes, consider sequential ChIP with antibodies against different proteins in the complex.
Data normalization strategies: Implement appropriate normalization using spike-in controls or normalization to input for accurate quantification .
Computational modeling can significantly improve antibody specificity and affinity through these advanced approaches:
Epitope prediction and optimization:
Use algorithms to predict immunogenic epitopes on At5g61310 protein
Select epitopes with high predicted antigenicity and minimal sequence homology to other Arabidopsis proteins
Optimize epitope design through molecular dynamics simulations to ensure accessibility
Antibody variable region modeling:
Apply Rosetta-based approaches to model antibody paratope-epitope interactions
Use dTERMen or similar informatics approaches to identify potential mutations that might enhance binding
Generate virtual libraries of mutated antibodies and predict binding affinity improvements
Affinity maturation prediction:
Identify candidate mutations in complementarity-determining regions (CDRs)
Predict structural effects of mutations on antibody-antigen interface
Prioritize mutations predicted to form additional hydrogen bonds, salt bridges, or hydrophobic interactions
Library design for experimental validation:
Design phage display libraries incorporating predicted beneficial mutations
Include control mutations predicted to be neutral or deleterious
Implement deep mutational scanning approaches to validate computational predictions
This integrated computational-experimental approach has shown success in improving antibody affinity, as demonstrated in viral antigen studies where KD improvements from 0.63 nM to 0.01 nM have been achieved .
To thoroughly assess potential cross-reactivity, implement this methodological workflow:
Sequence homology analysis:
Identify proteins with sequence similarity to At5g61310 using BLAST
Focus particularly on the epitope region recognized by the antibody
Generate a prioritized list of potential cross-reactive proteins
Recombinant protein panel testing:
Express and purify recombinant versions of related proteins
Perform dot blot or western blot analysis with standardized protein amounts
Quantify relative binding affinities to each protein
Knockout/knockdown validation matrix:
Test antibody reactivity in plant tissues from:
Wild-type plants (positive control)
at5g61310 knockout lines (negative control)
Knockout lines for homologous genes
Double/triple knockout lines where applicable
Epitope competition assay:
Pre-incubate antibody with synthesized peptides representing:
The exact epitope from At5g61310
Homologous sequences from related proteins
Measure the degree of signal inhibition for each competing peptide
Immunoprecipitation-mass spectrometry:
Perform IP using the At5g61310 antibody
Identify all pulled-down proteins by mass spectrometry
Quantify enrichment relative to control IPs
This comprehensive approach provides both qualitative and quantitative assessment of antibody specificity using multiple orthogonal techniques .
For successful multiplexed immunofluorescence with At5g61310 antibody, follow these methodological guidelines:
Antibody compatibility assessment:
Test all antibodies individually before multiplexing
Verify that secondary antibodies don't cross-react
Confirm At5g61310 antibody works with selected fixation methods
Sequential staining protocol:
For antibodies from the same species, use sequential staining with blocking steps
Order antibodies from strongest to weakest signal
Consider using directly conjugated primary antibodies to avoid species conflicts
Signal separation optimization:
Select fluorophores with minimal spectral overlap
Include single-stained controls for spectral unmixing
Use computational approaches to resolve overlapping signals
Sample preparation refinement:
Optimize fixation conditions (paraformaldehyde concentration and time)
Test different antigen retrieval methods if necessary
Evaluate permeabilization conditions to maximize antibody access
Quantification strategy:
Implement consistent imaging parameters
Use appropriate thresholding methods
Employ colocalization analysis with statistical validation
Controls implementation:
Include absorption controls (pre-incubation with antigen)
Use knockout/knockdown lines as negative controls
Include isotype controls for assessing non-specific binding
This detailed approach ensures reliable multiplexed detection while minimizing artifacts and false colocalization signals .
When applying At5g61310 antibody across diverse plant tissues and developmental stages, implement this experimental design framework:
Sampling matrix design:
Create a comprehensive sampling grid covering:
Multiple tissue types (leaves, roots, stems, flowers, siliques)
Different developmental stages (seedling, vegetative, reproductive)
Various environmental conditions relevant to the research question
Extraction optimization by tissue type:
Tissue Type | Recommended Buffer Modifications | Special Considerations |
---|---|---|
Leaf | Standard extraction buffer | Age-dependent protein modifications |
Root | Increased detergent concentration | High proteolytic activity requires additional protease inhibitors |
Flower | Gentle extraction methods | Stage-specific expression patterns |
Silique | Modified buffer pH | High levels of interfering compounds |
Meristem | Low-volume extraction technique | Limited material requires sensitive detection |
Control implementation:
Include tissue-specific positive controls (constitutively expressed proteins)
Use developmental stage markers to confirm proper staging
Incorporate tissue-specific negative controls (proteins known to be absent)
Antibody validation across tissues:
Validate antibody specificity in each tissue type independently
Adjust antibody concentrations based on tissue-specific background
Verify epitope accessibility across different tissue preparations
Normalization strategy:
Select appropriate loading controls for each tissue type
Implement tissue-specific quantification standards
Account for tissue-specific protein extraction efficiencies
Technical replication planning:
Increase biological replicates for tissues with high variability
Adjust technical replication based on preliminary coefficient of variation
Implement hierarchical sampling for developmental series
This systematic approach ensures reliable protein detection and quantification across diverse plant materials while accounting for tissue-specific challenges .
When encountering contradictory results across techniques, implement this systematic interpretation framework:
Technique-specific limitations analysis:
Western blot: Evaluates denatured proteins, may miss conformational epitopes
Immunoprecipitation: Requires accessible epitopes in native conditions
Immunofluorescence: Depends on epitope accessibility in fixed tissues
ChIP: Requires antibody access to protein-DNA complexes
Epitope state assessment:
Determine if the epitope might be:
Masked by protein interactions in certain contexts
Modified post-translationally in specific conditions
Conformationally altered depending on technique conditions
Hierarchical validation approach:
Implement genetic controls (knockout/knockdown) with each technique
Use orthogonal methods to confirm key findings
Consider protein tagging approaches as complementary evidence
Experimental variable isolation:
Systematically test buffer conditions, detergents, and fixatives
Vary antibody concentrations across techniques
Test different antibody incubation conditions
Data integration strategy:
Weight evidence based on control robustness
Consider biological context when interpreting discrepancies
Develop hypotheses that could explain apparent contradictions
This structured approach transforms contradictory results into valuable insights about protein behavior under different experimental conditions .
For robust statistical analysis of quantitative data from At5g61310 antibody experiments, implement these methodological approaches:
Experimental design statistical considerations:
Power analysis to determine appropriate sample size
Randomization of samples to minimize batch effects
Blocking designs to account for known sources of variation
Normalization method selection:
For Western blots: Total protein normalization vs. housekeeping proteins
For immunofluorescence: Integrated density vs. mean fluorescence intensity
For ChIP-qPCR: Percent input vs. normalization to control regions
Statistical test selection matrix:
Data Type | Comparison Type | Recommended Test | Assumptions |
---|---|---|---|
Western blot densitometry | Two groups | Student's t-test or Mann-Whitney | Normality or non-parametric |
Western blot densitometry | Multiple groups | ANOVA with post-hoc or Kruskal-Wallis | Equal variance or non-parametric |
Immunofluorescence intensity | Spatial comparisons | Mixed-effects models | Nested data structure |
ChIP-qPCR enrichment | Multiple regions | Repeated measures ANOVA | Sphericity |
Colocalization coefficients | Multiple conditions | Permutation tests | Non-parametric comparisons |
Multiple testing correction implementation:
Bonferroni correction for strict family-wise error rate control
Benjamini-Hochberg procedure for false discovery rate control
Sequential Bonferroni for balanced approach
Effect size reporting:
Cohen's d for parametric comparisons
Cliff's delta for non-parametric alternatives
Confidence intervals for all reported metrics
Reproducibility enhancement:
Cross-validation approaches when applicable
Bootstrap confidence intervals for complex metrics
Meta-analytic approaches for combining experimental replicates
This comprehensive statistical framework ensures robust, reproducible findings while accounting for the specific characteristics of antibody-based data .
To effectively integrate antibody-derived data into multi-omics frameworks, implement these methodological strategies:
Coordinated experimental design:
Collect samples for multiple omics analyses from the same biological material
Include shared controls across platforms
Implement consistent environmental conditions and treatments
Data normalization across platforms:
Develop common reference standards applicable across techniques
Implement platform-specific normalization followed by cross-platform standardization
Utilize spike-in controls common to multiple platforms when possible
Integration analysis frameworks:
Correlation network approaches linking protein abundance with:
Transcriptomic data (RNA-seq)
Epigenomic profiles (ChIP-seq, ATAC-seq)
Metabolomic signatures (LC-MS, GC-MS)
Factor analysis to identify latent variables spanning multiple data types
Bayesian integration models incorporating platform-specific uncertainty
Temporal alignment strategies:
Account for different timescales of molecular processes
Implement time-course designs with sufficient resolution
Develop mathematical models describing system dynamics
Biological interpretation enhancement:
Pathway enrichment analyses incorporating multi-omics data
Network analyses identifying protein-centric functional modules
Causal inference approaches to establish directional relationships
This structured integration approach provides a comprehensive systems-level understanding of At5g61310 protein function within the broader cellular context .
For CRISPR-based validation of antibody specificity, implement this comprehensive experimental design approach:
Guide RNA design strategy:
Design multiple gRNAs targeting different regions of the At5g61310 gene
Include gRNAs targeting the epitope region specifically
Design control gRNAs targeting unrelated sequences
Mutation type diversity:
Generate complete knockouts (large deletions)
Create epitope-specific mutations (precise edits)
Develop truncated versions with and without the epitope
Validation hierarchical approach:
Confirm genetic modifications by sequencing
Verify transcript alterations via RT-qPCR
Assess protein expression using alternative detection methods
Control implementation:
Include wild-type lines as positive controls
Use CRISPR lines targeting unrelated genes as specificity controls
Develop transgenic complementation lines reintroducing:
Wild-type At5g61310
Epitope-mutated At5g61310
Tagged versions for orthogonal detection
Comprehensive antibody testing matrix:
Genetic Background | Expected Western Blot Result | Expected Immunofluorescence Result | Expected IP Result |
---|---|---|---|
Wild-type | Strong specific band | Specific localization pattern | Specific enrichment |
Complete knockout | No specific band | No specific signal | No enrichment |
Epitope deletion | No specific band | No specific signal | No enrichment |
Truncated protein (epitope present) | Smaller specific band | Altered localization possible | Reduced enrichment |
Truncated protein (epitope absent) | No specific band | No specific signal | No enrichment |
Complemented line | Restored specific band | Restored localization pattern | Restored enrichment |
This comprehensive validation approach provides definitive evidence of antibody specificity while also offering insights into epitope accessibility and protein function .