KEGG: ath:AT4G30067
UniGene: At.63312
LCR63 Antibody belongs to the family of monoclonal antibodies (mAbs) developed for specific antigen recognition in research applications. Like other well-characterized mAbs such as anti-V mAbs described in plague research, LCR63 Antibody binds to a specific conformational epitope on its target protein . The binding mechanism involves precise molecular interactions at the antigen-antibody interface, which determines its specificity and utility in various experimental applications.
To characterize LCR63 Antibody's target recognition properties, researchers should employ multiple complementary approaches, including:
ELISA assays with purified target protein (typically at 10 μg/mL concentration)
Western blotting with both native and denatured protein samples
Immunoprecipitation followed by mass spectrometry
Surface plasmon resonance to determine binding kinetics
The binding specificity should be validated using both positive controls (cells/tissues known to express the target) and negative controls (knockout samples or cell lines lacking target expression) to establish recognition parameters .
Determining the optimal working concentration for LCR63 Antibody requires systematic titration experiments across different applications. Similar to validation approaches used for other mAbs, researchers should:
Begin with a broad concentration range (typically 0.1-10 μg/mL for most applications)
Perform parallel experiments with serial dilutions
Analyze signal-to-noise ratio at each concentration
Determine the minimum concentration that yields reproducible specific signal
The table below provides starting concentration guidelines based on similar monoclonal antibody optimization protocols:
Application | Starting Concentration | Typical Working Range | Key Optimization Parameters |
---|---|---|---|
Western Blot | 1 μg/mL | 0.1-5 μg/mL | Blocking agent, incubation time |
ELISA | 2 μg/mL | 0.5-5 μg/mL | Coating buffer pH, detection system |
Immunofluorescence | 5 μg/mL | 1-10 μg/mL | Fixation method, permeabilization |
Flow Cytometry | 10 μg/mL | 1-20 μg/mL | Cell preparation, buffer composition |
Immunoprecipitation | 5 μg per 500 μg lysate | 1-10 μg | Bead type, pre-clearing protocol |
A well-designed titration experiment should include both positive and negative controls for each concentration tested, enabling quantitative determination of specificity at different concentrations .
Rigorous validation of LCR63 Antibody is essential for generating reliable research data. Based on established protocols for monoclonal antibody validation, researchers should implement the following critical steps:
Specificity Testing: Confirm target specificity using multiple approaches:
Western blotting with recombinant protein and cell/tissue lysates
Immunoprecipitation followed by mass spectrometry identification
Cross-reactivity assessment against related proteins
Testing in knockout/knockdown systems when available
Multi-Application Performance Assessment: Validate performance across intended applications:
Compare results between Western blot, immunofluorescence, and ELISA
Document application-specific optimization parameters
Establish concordance between different detection methods
Lot-to-Lot Consistency Evaluation: For reproducible research:
Test multiple antibody lots using standardized samples
Establish acceptance criteria for new lots
Document key performance metrics for reference
Controls Implementation: Design comprehensive control systems:
Positive controls (samples known to express target)
Negative controls (samples lacking target expression)
Isotype controls (non-specific antibodies of same isotype)
Secondary antibody-only controls
These validation steps should be performed systematically and documented thoroughly to ensure reliability of subsequent experimental results .
Confirming epitope specificity of LCR63 Antibody requires a multi-faceted approach similar to that used for characterizing other monoclonal antibodies. Based on established epitope mapping techniques, researchers should:
Peptide Array Analysis: Test binding against a library of overlapping peptides spanning the target protein (typically 25 μg/mL peptide concentration in ELISA format) . This approach is particularly valuable for identifying linear epitopes.
Competitive Binding Assays: Assess whether LCR63 Antibody competes with other well-characterized antibodies targeting the same protein. This approach requires:
Mutagenesis Studies: Introduce point mutations or deletions in the suspected epitope region and test antibody binding to mutant proteins.
Surface Plasmon Resonance (SPR): Use SPR to measure binding kinetics with:
Full-length protein
Protein fragments
Mutated variants
The epitope specificity determination should include both positive controls (antibodies with known epitopes) and negative controls (isotype-matched irrelevant antibodies) to establish unambiguous recognition patterns .
For optimal immunofluorescence results with LCR63 Antibody, researchers should follow these methodological recommendations based on established protocols for monoclonal antibody applications:
Sample Preparation Optimization:
Test multiple fixation methods (4% paraformaldehyde, methanol, acetone)
Evaluate different permeabilization reagents (0.1-0.5% Triton X-100, 0.1-0.5% saponin)
Optimize fixation duration (10-30 minutes) and temperature (room temperature vs. 4°C)
Blocking and Antibody Incubation:
Use 5-10% normal serum from the same species as the secondary antibody
Include 0.1-0.3% BSA and 0.1% Tween-20 in blocking buffer
Incubate with primary antibody (LCR63) at 5 μg/mL initially, then optimize
Perform primary antibody incubation overnight at 4°C for optimal results
Use fluorophore-conjugated secondary antibodies at 1:500-1:2000 dilution
Controls and Counterstaining:
Include positive control samples (known to express target)
Include negative control samples (known to lack target expression)
Include secondary antibody-only controls
Use DAPI (1 μg/mL) for nuclear counterstaining
Image Acquisition and Analysis:
Capture images using consistent exposure settings across samples
Analyze subcellular localization patterns
Perform quantitative analysis of signal intensity when appropriate
This protocol framework has been successful for detecting various cellular proteins in both cultured cells and tissue sections, as demonstrated with other well-characterized monoclonal antibodies .
For successful co-immunoprecipitation experiments with LCR63 Antibody, researchers should implement the following methodological approach:
Lysate Preparation:
Harvest cells at 80-90% confluence
Lyse cells in non-denaturing buffer (typically 25 mM Tris-HCl pH 7.4, 150 mM NaCl, 1 mM EDTA, 1% NP-40, 5% glycerol)
Include protease and phosphatase inhibitor cocktails
Clear lysate by centrifugation (14,000g, 10 minutes, 4°C)
Pre-clear with Protein G beads to reduce non-specific binding
Antibody-Bead Complex Formation:
Conjugate LCR63 Antibody (5 μg per 500 μg of lysate) to Protein G magnetic beads
Incubate for 1 hour at room temperature with gentle rotation
Wash to remove unbound antibody
Immunoprecipitation:
Add pre-cleared lysate to antibody-bead complex
Incubate overnight at 4°C with gentle rotation
Wash 4-5 times with lysis buffer
Elute proteins using either:
a) Low pH buffer (0.1 M glycine, pH 2.5-3.0)
b) SDS sample buffer for direct SDS-PAGE analysis
Controls and Validation:
Include isotype control antibody immunoprecipitation
Include input sample (typically 5% of lysate used for IP)
Perform reciprocal co-IP when possible
Validate results with alternative techniques (proximity ligation assay, FRET)
Analysis:
Analyze co-precipitated proteins by Western blotting or mass spectrometry
Quantify relative amounts of co-precipitated proteins
Assess specificity by comparing to control IPs
This protocol framework draws on established co-immunoprecipitation approaches that have been successful with various monoclonal antibodies in protein interaction studies .
Measuring binding affinity and avidity of LCR63 Antibody requires sophisticated biophysical techniques. Based on approaches used for characterizing other monoclonal antibodies, researchers should implement:
Surface Plasmon Resonance (SPR) Analysis:
Immobilize anti-mouse Fc γ antibody on a CM5 sensor chip (~10,000 RU)
Capture LCR63 Antibody (~300 RU)
Pass varying concentrations of purified target antigen (1 nM to 1.5 μM)
Measure association and dissociation phases
Analyze binding curves to determine:
Association rate constant (ka)
Dissociation rate constant (kd)
Equilibrium dissociation constant (KD = kd/ka)
Bio-Layer Interferometry (BLI):
Load LCR63 Antibody onto protein G sensors
Expose to varying concentrations of target antigen
Record real-time binding curves
Derive kinetic parameters through curve fitting
Enzyme-Linked Immunosorbent Assay (ELISA) for Avidity Measurement:
Perform standard ELISA with varying antibody concentrations
Include chaotropic agent wash steps (e.g., urea or sodium thiocyanate)
Compare retention of binding in presence of chaotropic agents
Calculate avidity index as ratio of binding with/without chaotropic agent
Isothermal Titration Calorimetry (ITC):
Measure heat changes during antibody-antigen binding
Determine thermodynamic parameters (ΔH, ΔS, ΔG)
Calculate binding stoichiometry
The relationship between binding characteristics and functional properties should be evaluated, as high affinity does not always correlate with optimal functional performance in research applications .
Reconciling contradictory results with LCR63 Antibody requires systematic investigation of multiple technical and biological variables. Based on approaches used to resolve discrepancies with other antibodies, researchers should:
Assess Target Protein Conformation:
Different applications expose different epitopes
Native vs. denatured conditions affect epitope accessibility
Fixation methods may alter epitope structure
Solution: Test antibody performance under varying conditions that preserve or modify protein structure
Evaluate Antibody-Specific Variables:
Lot-to-lot variations in antibody preparations
Storage conditions affecting antibody stability
Working concentration differences across applications
Solution: Standardize antibody aliquoting, storage, and application-specific concentrations
Analyze Sample Preparation Impact:
Create a matrix of sample preparation variables:
Variable | Western Blot | Immunofluorescence | Flow Cytometry | ELISA |
---|---|---|---|---|
Fixation | N/A | 4% PFA, methanol | 2% PFA | N/A |
Lysis buffer | RIPA, NP-40 | N/A | N/A | Variable |
Blocking | 5% milk, 5% BSA | 10% serum | 1% BSA | 1-5% BSA |
Antigen retrieval | N/A | Heat, enzymatic | N/A | N/A |
Implement Control Systems:
Use alternative antibodies against the same target
Include genetic knockdown/knockout controls
Test in multiple cell lines/tissue types
Solution: Establish minimum criteria for result acceptance across platforms
Biological Context Considerations:
Protein expression levels in different systems
Post-translational modifications affecting epitope accessibility
Protein-protein interactions masking binding sites
Solution: Characterize target protein biology comprehensively
By systematically addressing these variables, researchers can identify the source of contradictory results and establish reliable protocols for consistent LCR63 Antibody performance across experimental platforms .
High background signal is a common challenge when working with monoclonal antibodies. For LCR63 Antibody, the following systematic troubleshooting approach should be implemented:
Antibody Concentration Issues:
Problem: Excessive antibody concentration leading to non-specific binding
Solution: Perform titration experiments (0.1-10 μg/mL) to determine optimal concentration
Validation: Compare signal-to-noise ratio across concentration gradient
Blocking Inefficiency:
Problem: Inadequate blocking allowing non-specific binding
Solution: Test multiple blocking agents systematically:
Blocking Agent | Concentration | Incubation Time | Applications |
---|---|---|---|
BSA | 1-5% | 30-60 min | WB, ELISA, IF |
Non-fat milk | 3-5% | 30-60 min | WB |
Normal serum | 5-10% | 30-60 min | IF, IHC |
Casein | 0.5-2% | 30-60 min | ELISA |
Commercial blockers | As directed | As directed | Multiple |
Cross-Reactivity Issues:
Problem: Antibody binding to proteins with similar epitopes
Solution: Pre-absorb antibody with related antigens or test on knockout samples
Validation: Compare staining patterns before and after pre-absorption
Secondary Antibody Problems:
Problem: Non-specific binding of secondary antibody
Solution: Include secondary-only controls; try alternative secondary antibodies
Validation: Evaluate background in absence of primary antibody
Sample-Specific Factors:
Problem: Endogenous enzymes (peroxidases, phosphatases) causing background
Solution: Include quenching steps (3% H₂O₂ for peroxidases)
Validation: Compare signal with and without quenching steps
Protocol Optimization:
Problem: Suboptimal washing procedures
Solution: Increase wash duration and volume; add detergent (0.05-0.1% Tween-20)
Validation: Compare background after standard vs. extended washing
Each troubleshooting intervention should be tested systematically while keeping other variables constant to identify the specific cause of high background .
Enhancing sensitivity for detecting low-abundance targets with LCR63 Antibody requires optimizing multiple experimental parameters. Based on approaches used for other monoclonal antibodies, researchers should implement the following strategies:
Signal Amplification Systems:
Employ tyramide signal amplification (TSA) for immunohistochemistry and immunofluorescence
Use polymer-based detection systems (e.g., EnVision™) for enhanced sensitivity
Implement biotin-streptavidin amplification with careful control for endogenous biotin
Sample Preparation Optimization:
Concentrate proteins through immunoprecipitation before analysis
Optimize antigen retrieval methods (heat-induced vs. enzymatic)
Use membrane fractionation to enrich target proteins when appropriate
Detection System Enhancement:
Switch to high-sensitivity substrates (e.g., SuperSignal™ West Femto for Western blotting)
Use fluorophores with higher quantum yield for immunofluorescence
Implement nanoparticle-conjugated secondary antibodies for enhanced signal
Instrument and Acquisition Parameters:
Optimize exposure settings on imaging systems
Use confocal microscopy with spectral unmixing for immunofluorescence
Implement advanced detector systems for flow cytometry
Sensitivity Comparison Matrix:
Enhancement Approach | Fold Improvement | Best Applications | Limitations |
---|---|---|---|
TSA amplification | 10-100x | IF, IHC | Increased background possible |
Polymer detection | 5-10x | IHC | Cost |
High-sensitivity substrate | 10-50x | WB | Short signal duration |
Nanoparticle conjugates | 5-20x | Multiple | Complex preparation |
Signal accumulation (longer exposure) | 2-5x | WB, IF | Increased background |
Protocol Modifications:
Extend primary antibody incubation (overnight at 4°C)
Reduce washing stringency slightly while maintaining specificity
Optimize antibody diluent composition (add 0.1% gelatin or 0.5% BSA)
Each sensitivity enhancement strategy should be validated with appropriate controls to ensure specific signal amplification rather than increased background .
Quantitative analysis of LCR63 Antibody staining patterns requires rigorous methodology and appropriate statistical approaches. Based on established practices for immunohistochemistry and immunofluorescence quantification, researchers should:
Image Acquisition Standardization:
Use consistent microscope settings across all samples
Capture multiple fields per sample (minimum 5-10 representative regions)
Include scale bars in all images
Use identical exposure settings for comparative analyses
Quantification Methods Selection:
For membrane/cytoplasmic staining: Consider H-score approach (intensity × percentage)
For nuclear staining: Quantify percentage of positive nuclei
For punctate patterns: Analyze number, size, and intensity of discrete structures
Analysis Software Implementation:
Use dedicated image analysis software (ImageJ/FIJI, CellProfiler, QuPath)
Develop consistent thresholding parameters for signal detection
Implement batch processing with identical parameters across samples
Scoring System Development:
Staining Intensity | Score | Definition | Quantitative Threshold |
---|---|---|---|
Negative | 0 | No detectable signal | <10% of background |
Weak | 1 | Barely perceptible | 10-50% above background |
Moderate | 2 | Clearly visible | 50-200% above background |
Strong | 3 | Intense signal | >200% above background |
Statistical Analysis Selection:
For comparing two groups: t-test or Mann-Whitney U test
For multiple groups: ANOVA with appropriate post-hoc tests
For correlation with clinical parameters: Spearman or Pearson correlation
For survival analysis: Kaplan-Meier with log-rank test
Validation and Quality Control:
Assess inter-observer and intra-observer variability
Confirm automated quantification with manual scoring on subset
Compare results using alternative antibodies against same target
This methodological framework provides a standardized approach for generating reproducible quantitative data from LCR63 Antibody staining experiments, ensuring statistical rigor and interpretability .
Evaluating cross-reactivity of LCR63 Antibody requires a multi-faceted approach to establish specificity boundaries. Based on methodologies used for characterizing other monoclonal antibodies, researchers should implement:
Sequence Homology Analysis:
Identify proteins with sequence similarity to the target epitope
Assess structural homology in potential cross-reactive regions
Prioritize testing of proteins with highest similarity scores
Recombinant Protein Panel Testing:
Express recombinant variants of related proteins
Test LCR63 Antibody binding via Western blot and ELISA
Quantify relative binding affinity to each potential cross-reactant
Knockout/Knockdown Validation:
Generate CRISPR knockout or siRNA knockdown models
Test for persistence of LCR63 Antibody signal after target depletion
Quantify signal reduction compared to control samples
Peptide Competition Assays:
Synthesize peptides corresponding to the target epitope
Pre-incubate LCR63 Antibody with epitope peptide
Test binding inhibition across applications
Cross-Reactivity Assessment Matrix:
Validation Approach | Sensitivity | Specificity | Technical Complexity | Best Applications |
---|---|---|---|---|
Sequence analysis | Low | Low | Low | Initial screening |
Recombinant proteins | High | High | Medium | Direct binding assessment |
Knockout validation | High | High | High | Definitive evaluation |
Peptide competition | Medium | Medium | Low | Epitope confirmation |
Mass spectrometry | High | High | High | Unbiased identification |
Mass Spectrometry Validation:
Perform immunoprecipitation with LCR63 Antibody
Analyze precipitated proteins by mass spectrometry
Identify all proteins captured by the antibody
Compare to expected target profile
This systematic approach provides comprehensive characterization of LCR63 Antibody specificity and cross-reactivity profile, enabling researchers to interpret experimental results with appropriate confidence and identify potential confounding factors .
Incorporating LCR63 Antibody into multiplexed imaging systems requires strategic optimization of multiple technical parameters. Based on advanced immunofluorescence and imaging methodologies, researchers should:
Antibody Panel Design:
Assess compatibility of LCR63 Antibody with other primary antibodies
Select antibodies from different host species to minimize cross-reactivity
Test sequential staining protocols when same-species antibodies must be used
Evaluate antibody performance after fluorophore conjugation if direct labeling is planned
Multiplexing Technology Selection:
Cyclic immunofluorescence (CycIF): Multiple rounds of staining/imaging/quenching
Mass cytometry imaging (MIBI/IMC): Metal-tagged antibodies with spatial resolution
Spectral imaging: Simultaneous detection of spectrally overlapping fluorophores
Tyramide signal amplification multiplexing: Sequential TSA labeling with antibody stripping
Protocol Optimization Parameters:
Multiplexing Method | Key Parameters for LCR63 Integration | Maximum Markers | Resolution Limit |
---|---|---|---|
Standard IF | Fluorophore selection, crosstalk minimization | 4-5 | ~200 nm |
CycIF | Antibody stripping efficiency, epitope stability | 20-40 | ~200 nm |
MIBI/IMC | Metal conjugation efficiency, sensitivity | 30-40 | ~500 nm |
Spectral imaging | Unmixing algorithm optimization | 6-10 | ~200 nm |
TSA multiplexing | Heat-induced epitope retrieval between cycles | 7-10 | ~200 nm |
Image Acquisition and Analysis:
Develop standardized acquisition settings for each marker
Implement automated cell/tissue segmentation algorithms
Utilize machine learning approaches for pattern recognition
Create spatial relationship maps between markers
Validation Strategies:
Compare multiplex results with single-marker controls
Assess epitope persistence through staining/stripping cycles
Evaluate potential interactions between detection systems
Confirm cell type identification with orthogonal markers
The successful integration of LCR63 Antibody into multiplexed imaging workflows enables sophisticated analysis of cellular relationships and tissue architecture not possible with conventional single-marker approaches, advancing understanding of complex biological systems .
LCR63 Antibody can be strategically integrated into cutting-edge single-cell analysis technologies, enabling high-resolution protein expression studies. Based on advanced immunological techniques, researchers should consider the following emerging applications:
Single-Cell Proteomics Integration:
Incorporate LCR63 Antibody into mass cytometry (CyTOF) panels
Optimize metal conjugation without compromising binding properties
Develop compensation strategies for signal spillover
Combine with lineage markers for heterogeneity assessment
Spatial Transcriptomics Coupling:
Combine LCR63 immunostaining with in situ hybridization techniques
Develop sequential protein-RNA detection protocols
Implement computational approaches for multi-omic data integration
Compare protein expression with mRNA levels at single-cell resolution
Microfluidic Applications:
Adapt LCR63 Antibody for microfluidic-based single-cell Western blotting
Optimize antibody concentration for reduced-volume applications
Validate detection sensitivity in nanoliter-scale reactions
Develop protocols for combined phenotypic and functional analysis
Advanced Flow Cytometry Applications:
Technology | LCR63 Application | Special Considerations | Resolution |
---|---|---|---|
Spectral flow | Multi-parameter analysis | Fluorophore selection, unmixing | 35+ parameters |
Imaging flow | Protein localization | Fixation optimization | Subcellular |
CyTOF | High-dimensional phenotyping | Metal conjugation | 40+ parameters |
Single-cell proteomics | Absolute quantification | Antibody titration | Protein copies/cell |
Advanced Imaging Applications:
Super-resolution microscopy (STED, STORM, PALM)
Live-cell imaging with fluorescently-tagged Fab fragments
Correlative light-electron microscopy (CLEM)
3D tissue clearing and imaging (CLARITY, iDISCO)
Validation Requirements:
Compare sensitivity across platforms
Establish minimum detectable protein levels
Develop spike-in controls for quantification
Create reference datasets across multiple cell types
These emerging applications position LCR63 Antibody as a valuable tool for detailed characterization of cellular heterogeneity and function at unprecedented resolution, enabling researchers to address complex biological questions previously inaccessible with bulk analysis methods .
Maintaining consistent LCR63 Antibody performance across longitudinal studies requires implementation of comprehensive quality control systems. Based on established antibody validation frameworks, researchers should:
Reference Standard Development:
Create frozen aliquots of characterized positive control samples
Generate standard curves with recombinant target protein
Establish acceptance criteria for each experimental application
Maintain detailed records of antibody performance metrics
Lot-to-Lot Validation Protocol:
Test each new antibody lot against reference standards
Compare staining patterns across multiple applications
Determine correction factors if necessary for quantitative studies
Document validation results in laboratory records
Longitudinal Monitoring System:
Include standard controls in each experimental run
Monitor signal intensity and background over time
Track antibody usage, storage conditions, and freeze-thaw cycles
Implement statistical process control methods to detect performance drift
Comprehensive Documentation Requirements:
Documentation Element | Essential Information | Update Frequency | Purpose |
---|---|---|---|
Antibody inventory | Lot numbers, dates, storage location | Each new lot | Traceability |
Validation reports | Performance metrics, control results | Each new lot | Quality assurance |
Protocol modifications | Parameter changes, rationale | As needed | Methodology tracking |
Control performance | Signal/noise, consistency metrics | Each experiment | Drift detection |
Troubleshooting records | Issues encountered, resolutions | As needed | Knowledge management |
Inter-laboratory Standardization:
Develop standard operating procedures (SOPs)
Share reference samples between collaborating laboratories
Implement proficiency testing when multiple operators are involved
Calibrate equipment regularly according to manufacturer specifications
By implementing these comprehensive quality control measures, researchers can minimize variability in LCR63 Antibody performance across longitudinal studies, ensuring data comparability and scientific reproducibility over extended research timelines .
Ensuring reproducibility of research findings generated with LCR63 Antibody requires comprehensive reporting of methodological details in scientific publications. Based on best practices for antibody-based research, authors should include:
Antibody Identification and Sourcing:
Complete antibody identifier information (clone, catalog number, lot number)
Manufacturer/source details
RRID (Research Resource Identifier) when available
Concentration of antibody as received and working dilution used
Validation Documentation:
Specificity validation methods employed
Controls used (positive, negative, isotype)
Knockout/knockdown validation if performed
Cross-reactivity assessment results
Detailed Methodology:
Complete protocol with buffer compositions
Incubation times and temperatures
Detection system specifications
Image acquisition parameters
Reproducibility Demonstration:
Number of experimental replicates
Inter-assay and intra-assay variation assessment
Representative images showing full range of results
Quantification methods with statistical analysis
Reporting Checklist for Publications:
Reporting Element | Essential Details | Purpose | Common Omissions to Avoid |
---|---|---|---|
Antibody identity | Clone, catalog #, lot #, RRID | Traceability | Missing lot numbers |
Validation evidence | Methods, results, controls | Specificity confirmation | Assuming validation not needed |
Protocol details | Complete methods, concentrations | Reproducibility | Incomplete buffer descriptions |
Image acquisition | Equipment, settings, processing | Data quality | Undisclosed image manipulations |
Quantification | Methods, software, parameters | Analysis transparency | Selective quantification |
Data and Material Sharing:
Raw image data availability statement
Repository information for large datasets
Sharing protocol details via protocols.io or similar platforms
Willingness to share critical reagents and control samples