The term "At5" appears in Source , but it refers to monoclonal antibody At5, which targets human brain antigens such as dMAG (a derivative of myelin-associated glycoprotein) and proteoglycans like phosphacan and neurocan. This antibody is unrelated to Arabidopsis proteins .
Antibodies are Y-shaped proteins produced by B cells that recognize specific antigens, enabling immune responses or experimental detection . They are widely used in research for:
Functional modulation of cellular pathways (e.g., malaria invasion inhibition by PfRH5 antibodies) .
Efforts to optimize antibodies include modifying Fc regions to enhance effector functions (e.g., complement activation) or prolong half-life . No such studies mention At5g62627.
No direct evidence of an antibody targeting At5g62627 exists in the provided sources.
Source lists antibodies used in a study but does not include At5g62627.
Source describes a patent database with ~150,000 antibody sequences but focuses on human therapeutics, not plant proteins.
If developed, such an antibody could be used to:
To obtain authoritative information about an "At5g62627 Antibody":
Consult specialized plant biology databases (e.g., TAIR, UniProt).
Review Arabidopsis-focused studies on DEFL protein characterization.
Contact commercial antibody suppliers (e.g., Agrisera, Thermo Fisher) for custom antibody development.
KEGG: ath:AT5G62627
STRING: 3702.AT5G62627.1
At5g62627 refers to a specific gene locus in Arabidopsis thaliana, a model plant organism widely used in molecular biology research. Researchers develop antibodies against the protein products of such genes to study protein expression, localization, interaction partners, and functional roles in plant biological processes. Antibodies enable visualization of proteins in their native context through techniques like immunohistochemistry and western blotting, providing insights not achievable through nucleic acid-based methods alone. Developing specific antibodies against plant proteins like At5g62627 requires careful consideration of protein structure, antigenicity, and cross-reactivity with other plant proteins.
Researchers can develop several types of antibodies for studying At5g62627 protein:
Polyclonal antibodies: Generated by immunizing animals with At5g62627 protein or peptide fragments, resulting in antibodies targeting multiple epitopes. These provide robust detection but may have higher cross-reactivity.
Monoclonal antibodies: Produced from single B-cell clones, offering high specificity to a single epitope, ideal for distinguishing between closely related proteins.
Recombinant antibodies: Engineered antibodies produced in expression systems, allowing precise control over antibody properties. Modern platforms enable sequencing from B-cells to obtain "immortal polyclonals" that preserve the diversity of immune responses .
Fragment antibodies: Smaller antibody fragments (Fab, scFv) that may provide better access to certain epitopes in complex plant tissues.
For plant proteins like At5g62627, recombinant approaches are increasingly favored as they overcome challenges related to plant-specific post-translational modifications and potential cross-reactivity.
Epitope selection for At5g62627 antibody development requires careful bioinformatic analysis:
Sequence analysis: Identify unique regions of At5g62627 with minimal homology to other Arabidopsis proteins to reduce cross-reactivity.
Structural prediction: Use protein structure prediction tools to identify surface-exposed regions likely to be accessible to antibodies.
Antigenicity assessment: Apply algorithms that predict antigenic determinants based on hydrophilicity, flexibility, and accessibility.
Post-translational modification (PTM) consideration: Avoid regions with potential PTMs that might block antibody recognition or create unwanted specificity.
Conservation analysis: For evolutionary studies, consider targeting either highly conserved or highly variable regions depending on research objectives.
Researchers should avoid transmembrane domains and signal peptides, instead focusing on unique loops or terminal regions that characterize the specific protein of interest.
Thorough validation of At5g62627 antibodies is critical to ensure experimental reliability:
Knockout/knockdown controls: Test the antibody in tissues from At5g62627 knockout or knockdown plants to confirm absence of signal.
Overexpression verification: Test in tissues overexpressing tagged At5g62627 to confirm recognition of the target.
Western blot analysis: Verify a single band of appropriate molecular weight, with disappearance in knockout samples.
Immunoprecipitation followed by mass spectrometry: Confirm that the antibody precipitates the intended protein.
Cross-reactivity testing: Test against closely related proteins or in tissues expressing various levels of related proteins.
Peptide competition assay: Pre-incubate antibody with the immunizing peptide to confirm signal disappearance.
Independent antibody comparison: Compare results with a second antibody targeting a different epitope of At5g62627.
These validation steps are particularly important for plant proteins, where antibody specificity can be compromised by the presence of large gene families with highly similar members.
Optimizing immunoprecipitation (IP) of At5g62627 from plant tissues requires addressing plant-specific challenges:
Tissue extraction buffer optimization: Test different detergents (CHAPS, NP-40, Triton X-100) and salt concentrations to maintain protein solubility while preserving antibody-antigen interactions.
Cell wall considerations: Include cell wall-degrading enzymes in initial extraction steps to improve protein release.
Secondary metabolite management: Add polyvinylpyrrolidone (PVP) or polyvinylpolypyrrolidone (PVPP) to adsorb phenolic compounds that can interfere with antibody binding.
Pre-clearing step: Include a pre-clearing step with non-immune serum or protein A/G beads to reduce non-specific binding.
Crosslinking consideration: For transient or weak interactions, consider crosslinking approaches (formaldehyde, DSP, or DTBP).
Antibody immobilization: Covalently link antibodies to beads to prevent antibody contamination in downstream analyses.
Gentle elution: Use epitope-competing peptides for gentle elution to maintain interaction partners.
For plant proteins like At5g62627, it's crucial to optimize conditions that account for the unique biochemical environment of plant cells while preserving native protein interactions.
Immunohistochemistry (IHC) in plant tissues presents unique challenges requiring specific adaptations for antibodies like those against At5g62627:
Fixation optimization: Test multiple fixatives (paraformaldehyde, glutaraldehyde) and concentrations to balance epitope preservation and tissue morphology.
Cell wall permeabilization: Include enzymatic digestion steps (cellulase, macerozyme) to facilitate antibody penetration through plant cell walls.
Antigen retrieval: Optimize heat-induced or enzymatic antigen retrieval to unmask epitopes potentially obscured during fixation.
Blocking optimization: Test plant-specific blocking agents to reduce background caused by endogenous peroxidases and biotin.
Signal amplification: Consider tyramide signal amplification for low-abundance proteins.
Autofluorescence management: Include specific steps to reduce plant tissue autofluorescence, such as sodium borohydride treatment or spectral unmixing during imaging.
Multichannel controls: Include controls for non-specific binding of secondary antibodies and autofluorescence in each channel.
For developmental studies, precise staging of plant tissues and comparison across different developmental timepoints can provide valuable insights into dynamic protein expression patterns.
Modern sequence-based antibody design can significantly enhance specificity for challenging targets like plant proteins:
Deep learning approaches: Recent advances like DyAb leverage pre-trained protein language models to predict antibody properties and design improved variants. These models can predict affinity differences between closely related sequences, enabling optimization with minimal experimental data .
Complementarity-determining region (CDR) optimization: Systematic mutation scanning of CDRs with natural amino acids (except cysteine) can identify critical residues for antigen binding. Comprehensive Substitution for Multidimensional Optimization (COSMO) experiments provide insights into important residues for antigen binding .
Combining beneficial mutations: Algorithms can identify combinations of beneficial point mutations to generate antibodies with improved specificity. DyAb successfully predicted multi-mutation variants with improved binding characteristics from datasets with as few as 100 labeled datapoints .
Plant-specific epitope targeting: Algorithms can identify unique surface regions of At5g62627 with minimal homology to other plant proteins.
Humanization considerations: For in vivo applications, computational approaches can guide humanization while preserving binding properties.
This approach has yielded high success rates, with over 85% of designed antibody variants successfully expressing and binding their targets, and most showing improved affinity compared to parent antibodies .
Cross-reactivity is a significant challenge for plant protein antibodies due to gene duplication and families of related proteins:
Epitope refinement: Computational analysis of the At5g62627 protein sequence compared to related Arabidopsis proteins can identify unique regions for targeting.
Negative selection approaches: Pre-absorb antibodies with recombinant proteins of related family members to deplete cross-reactive antibodies.
Subtractive screening: Screen antibody libraries against both At5g62627 and closely related proteins, selecting only those that bind exclusively to At5g62627.
Genetic algorithm optimization: Apply genetic algorithms to sample the vast design space and iteratively improve antibody specificity. The DyAb approach demonstrated that this can produce novel antibody variants with dramatically improved target specificity .
Deep mutational scanning: Systematic evaluation of all possible amino acid substitutions at key positions can identify mutations that enhance specificity.
Structural guided design: If structural data is available, focus mutations on key interface residues to enhance specificity.
Cross-validation in multiple systems: Test antibodies in multiple experimental systems, including plants with varying expression levels of related proteins.
Implementing these strategies can significantly reduce cross-reactivity issues, enabling more precise detection of At5g62627 even in complex plant proteomes.
Integrating computational and experimental approaches creates a powerful framework for antibody optimization:
Iterative design-build-test cycles: Begin with computational predictions, experimentally test top candidates, and feed results back into refined models. For example, the DyAb approach demonstrates how incorporating experimental data from initial designs back into the training set led to antibodies with further improved binding properties (up to 50-fold improvement in affinity) .
Structure-guided optimization: Use computational structural predictions or experimental structures to guide rational mutation design, focusing on the antibody-antigen interface.
Machine learning with minimal data: Leverage language models pre-trained on antibody sequences (like AntiBERTy or LBSTER) to predict binding properties from limited experimental data. Models trained on antibody-specific datasets consistently outperform general protein language models in prediction tasks .
Conformational dynamics analysis: Use molecular dynamics simulations to understand binding mechanisms and identify stabilizing mutations.
Combining beneficial mutations: Experimentally test point mutations individually, then use computational models to predict optimal combinations. This approach has yielded binding rates >85% for novel antibody variants .
High-throughput screening integration: Design smart libraries based on computational predictions to focus experimental efforts on promising candidates.
This integrated approach enables efficient optimization of antibodies even with limited initial experimental data, making it particularly valuable for challenging targets like plant proteins.
Contradictory results with different antibody preparations require systematic investigation:
Epitope mapping comparison: Determine if different antibodies recognize distinct epitopes of At5g62627, which might be differentially accessible in various experimental contexts.
Validation re-assessment: Re-evaluate validation data for each antibody, including knockout controls and specificity tests.
Post-translational modification influence: Consider whether contradictory results might reflect detection of different post-translationally modified forms of the protein.
Protein complex interference: Investigate whether protein-protein interactions might mask epitopes in certain cellular contexts.
Fixation and preparation effects: Test whether different sample preparation methods affect epitope accessibility.
Quantitative comparison: Perform side-by-side quantitative comparisons under identical conditions, including concentration normalization and standardized protocols.
Independent validation approaches: Employ non-antibody methods (mass spectrometry, RNA expression) to resolve contradictions.
Documenting these systematic investigations provides valuable information for the research community even when contradictions cannot be fully resolved.
Statistical analysis of antibody-based protein quantification requires thoughtful consideration:
Normalization strategies:
For Western blots: Normalize to consistent loading controls (housekeeping proteins)
For immunohistochemistry: Consider cell number, tissue area, or total protein normalization
For flow cytometry: Use appropriate fluorescence standards
Experimental design considerations:
Include biological and technical replicates (minimum n=3 for each)
Randomize sample processing order
Include gradient controls for quantification calibration
Statistical tests:
| Analysis Type | Recommended Tests | Application Context |
|---|---|---|
| Two-condition comparison | t-test (parametric) or Mann-Whitney (non-parametric) | Comparing mutant vs. wild-type |
| Multi-condition comparison | ANOVA with post-hoc tests (Tukey, Dunnett) | Comparing multiple treatments |
| Correlation analysis | Pearson or Spearman correlation | Relating protein levels to phenotypic measurements |
| Time-course data | Repeated measures ANOVA or mixed models | Developmental studies |
| Spatial distribution | Spatial statistics (Moran's I, Getis-Ord) | Tissue localization patterns |
Data transformation considerations: Log transformation of Western blot data often improves normality for statistical testing.
Multiple testing correction: Apply appropriate corrections (Bonferroni, Benjamini-Hochberg) when performing multiple comparisons.
Rigorous statistical analysis enhances reproducibility and facilitates meaningful comparisons between different experimental conditions.
Integrating antibody-based protein data with other omics datasets provides comprehensive biological insights:
Multi-omics correlation analysis:
Correlate protein levels with transcript levels to identify post-transcriptional regulation
Compare protein abundance with metabolite profiles to establish functional relationships
Correlate protein localization with chromatin accessibility data for transcription factors
Network integration approaches:
Construct protein-protein interaction networks using immunoprecipitation data
Overlay expression data on pathway maps
Perform gene set enrichment analysis using protein co-expression patterns
Temporal integration strategies:
Align time-course data across different omics layers
Identify lead-lag relationships between transcript and protein changes
Construct dynamic regulatory models incorporating protein data
Data normalization considerations:
Standardize data across platforms before integration
Consider batch effects and technical variations
Apply appropriate transformation methods for each data type
Visualization approaches:
Heat maps showing correlation across data types
Network diagrams with multi-omics overlay
Principal component analysis of integrated datasets
Causal inference methods:
Bayesian network modeling to infer causal relationships
Granger causality analysis for time-series data
Intervention-based approaches using genetic perturbations
This integrated analysis can reveal regulatory mechanisms and functional relationships that might not be apparent from antibody-based studies alone.
Next-generation sequencing (NGS) is revolutionizing antibody development for challenging targets:
B-cell receptor sequencing: Direct sequencing of B-cell receptors from immunized animals can identify the full repertoire of antibodies against At5g62627, enabling more comprehensive antibody discovery .
Hybridoma sequencing: NGS approaches to hybridoma sequencing provide comprehensive results, allowing immortalization of antibody sequences for consistent production .
Single B-cell sequencing: Isolating and sequencing individual B-cells allows pairing of heavy and light chains that recognize At5g62627.
Serum antibody sequencing: Advanced techniques enable sequencing from serum to obtain immortal polyclonal antibodies, preserving the diversity of immune responses .
Epitope mapping by phage display with NGS: Deep sequencing of phage display outputs can comprehensively map epitopes on At5g62627.
Computational pairing algorithms: Machine learning approaches can predict optimal heavy and light chain combinations from sequencing data.
These approaches provide an "antibody sequence database" that can be mined for optimal binders and engineered for improved properties, offering significant advantages over traditional hybridoma methods for plant protein targets.
Multispecific antibodies offer powerful capabilities for studying protein interactions:
Format selection: Consider various formats (bispecific IgG, diabodies, dual-variable-domain antibodies) based on the size and spatial relationship of target proteins.
Target selection strategy:
Pair At5g62627 with interacting proteins for co-localization studies
Combine with subcellular markers for precise localization
Pair with enzymatic domains for proximity labeling applications
Orientation considerations: Test multiple configurations of binding domains to optimize dual binding capacity.
Linker optimization: Systematically test linker lengths and compositions to accommodate the spatial arrangement of epitopes.
Expression system selection: Consider plant-based expression systems for proper folding of plant-optimized multispecific antibodies.
Validation strategies:
Confirm binding to individual targets separately
Verify simultaneous binding capability
Assess potential steric hindrance effects
Modern engineering platforms enable the production of custom multispecific antibodies with precise specifications, opening new avenues for studying protein complexes and signaling pathways involving At5g62627 .
CRISPR and other genome editing technologies provide powerful tools for antibody validation:
Knockout line generation: Create complete At5g62627 knockout lines in Arabidopsis as negative controls for antibody validation.
Epitope tagging: Insert small epitope tags into the endogenous At5g62627 locus to enable parallel detection with established tag antibodies.
Allele-specific modifications: Introduce subtle mutations that should affect antibody binding to confirm epitope specificity.
Humanized plant models: Develop plant lines expressing human-optimized versions of At5g62627 for therapeutic antibody testing.
Conditional knockouts: Create inducible knockout systems to observe dynamic changes in antibody staining upon target depletion.
Domain-specific modifications: Selectively remove specific domains to map antibody binding regions.
Reporter fusions: Create endogenous fusions with fluorescent proteins as independent verification of antibody staining patterns.
These approaches create precisely defined genetic backgrounds that serve as gold standard controls for antibody validation, overcoming the challenges of potential cross-reactivity with related plant proteins.