The ATL1 antibody targets Atlastin-1, a GTPase critical for endoplasmic reticulum (ER) membrane fusion and structure. Dysregulation of ATL1 is linked to hereditary spastic paraplegia and neurodegenerative disorders.
This antibody is validated for detecting ATL1 in ER dynamics studies, with implications for neurodegeneration research .
CD81 (TAPA1) is a tetraspanin protein involved in immune cell adhesion, signal transduction, and hepatitis C virus (HCV) entry.
Parameter | Details | Source |
---|---|---|
Host Species | Mouse | |
Conjugate | Atlantic Blue™ Fluorescent Dye | |
Applications | Flow cytometry (FC) | |
Reactivity | Human | |
Gene ID | 975 |
CD81 antibodies are critical for studying HCV infection mechanisms and immune cell interactions .
The linker for activation of T-cells (LAT) is a transmembrane protein essential for T-cell receptor signaling.
LAT antibodies are used in immunocytochemistry and protein array validations .
ATL is caused by human T-cell leukemia virus type 1 (HTLV-1). Key antibodies in ATL studies include:
Anti-ATLA (ATLV-associated antigen): Detected in 26% of healthy adults in ATL-endemic areas and 100% of ATL patients .
Anti-M2e Antibodies: Target influenza matrix protein 2, with cross-reactive binding to multiple viral strains (e.g., H1N1, H5N1) .
ATL81 Antibody belongs to the category of research antibodies designed for protein detection in experimental settings. While specific information about ATL81's target is not provided in the search results, understanding antibody specificity is critical for research validity. Antibody specificity refers to the ability of an antibody to bind exclusively to its intended target protein without cross-reactivity with other proteins.
To determine the specificity of ATL81 Antibody, researchers should perform validation experiments using positive and negative controls. A recommended approach is to use knockout (KO) cell lines where the target protein is absent, which enables clear determination of antibody specificity. According to recent standardized characterization protocols, antibodies should be evaluated across key applications such as immunoblotting, immunoprecipitation, and immunofluorescence to establish their specificity profile .
Validation of ATL81 Antibody should follow a comprehensive approach that addresses both specificity and reproducibility concerns. Based on current best practices in antibody characterization, researchers should implement the following validation protocol:
Knockout Validation: Test the antibody in wild-type vs. target protein knockout samples to verify specific binding
Multi-Application Testing: Validate across all intended applications (immunoblotting, immunoprecipitation, immunofluorescence)
Cross-Reference Validation: Compare results with alternative antibodies targeting the same protein
Concentration Series: Perform titration experiments to determine optimal working concentration
Positive and Negative Control Tissues/Cells: Test in samples with known expression levels of the target protein
The Structural Genomics Consortium researchers at The Neuro have developed standardized protocols for antibody characterization that can be adapted for ATL81 validation. Their platform evaluates antibodies through side-by-side testing against the same protein targets, providing a robust framework for validation .
Optimal storage conditions are essential for preserving antibody functionality and ensuring experimental reproducibility. While specific storage recommendations for ATL81 Antibody would need to be obtained from the manufacturer, research antibodies generally require:
Temperature Maintenance: Store at -20°C for long-term storage or at 4°C for antibodies in frequent use
Aliquoting Strategy: Divide into small, single-use aliquots to prevent repeated freeze-thaw cycles
Buffer Composition: Store in appropriate buffer with stabilizers (typically PBS with carrier proteins)
Protection from Contamination: Use sterile techniques when handling
Researchers should document storage conditions and antibody lot numbers in their experimental protocols, as these factors can significantly impact experimental outcomes and reproducibility. When transferring results between laboratories, sharing detailed antibody information including storage history is considered best practice.
Evaluating cross-reactivity of ATL81 Antibody in complex biological systems requires sophisticated approaches that go beyond standard validation methods. A comprehensive cross-reactivity assessment should include:
Immunoprecipitation-Mass Spectrometry (IP-MS): Perform pull-down experiments followed by mass spectrometry to identify all proteins captured by the antibody
Epitope Mapping: Determine the specific peptide sequence recognized by ATL81 to predict potential cross-reactivity based on sequence homology
Competitive Binding Assays: Use purified proteins or peptides to compete for antibody binding
Multiple Cell Line Testing: Evaluate specificity across diverse cell types with different protein expression profiles
Orthogonal Technique Validation: Compare results with alternative detection methods that don't rely on antibodies
Recent advances in biophysics-informed modeling allow researchers to predict and validate antibody binding modes. This approach associates distinct binding modes with each potential ligand, enabling the identification of specific variants with customized binding profiles. Such computational methods can help determine whether ATL81 might exhibit unintended cross-reactivity with structurally similar proteins .
Computational prediction of antibody specificity has advanced significantly, allowing researchers to model binding behaviors before conducting wet-lab experiments. For analyzing ATL81 Antibody specificity:
Biophysics-Informed Modeling: This approach can disentangle multiple binding modes associated with specific ligands. Using training data from phage display experiments, models can predict outcomes for closely related epitopes
Binding Mode Identification: Computational models can identify distinct binding modes for each potential ligand, enabling prediction of specificity profiles beyond experimental observations
Neural Network Parametrization: Advanced models parametrize binding energies using shallow dense neural networks, optimizing parameters globally to capture antibody population evolution across experiments
Custom Simulation: Once trained, models can simulate experiments with selected/unselected modes to predict enrichment of sequence variants
These computational approaches are particularly valuable when working with antibodies like ATL81 that may need to discriminate between very similar epitopes. Researchers can apply these methods to predict potential cross-reactivity issues or to optimize experimental conditions for maximum specificity.
Batch-to-batch variability represents a significant challenge in antibody-based research, potentially undermining experimental reproducibility. For ATL81 Antibody, researchers should implement:
Lot-Specific Validation: Each new batch should undergo full validation testing against reference standards
Internal Control Samples: Maintain a set of validated positive and negative control samples to test each new lot
Quantitative Metrics: Establish quantifiable parameters for acceptable performance (signal-to-noise ratio, EC50 values)
Documentation System: Create a comprehensive record of lot numbers used in experiments for traceability
The impact of batch variability is significant enough that researchers at The Neuro have collaborated with antibody manufacturers (representing approximately 80% of global renewable antibody production) to develop standardized characterization protocols. This initiative has tested approximately 1,200 antibodies against 120 protein targets to address reproducibility challenges .
Integrating ATL81 Antibody into multi-omics research requires careful experimental design that maximizes data quality while minimizing artifacts. An optimal design includes:
Sequential Sampling Strategy: Design sample collection to accommodate multiple analytical techniques without compromising sample integrity
Parallel Validation: Simultaneously validate antibody performance across all intended applications (immunohistochemistry, flow cytometry, etc.)
Standardized Sample Processing: Develop consistent protocols for sample preparation that are compatible with multiple downstream applications
Integrated Controls: Implement controls that enable cross-validation between different data types
Data Integration Framework: Establish computational methods for integrating antibody-generated data with other omics datasets
When designing experiments using ATL81 Antibody across multiple platforms, researchers should consider how variations in sample preparation might affect epitope accessibility. For example, fixation methods used for immunofluorescence might alter epitope conformation compared to conditions used for immunoblotting, potentially affecting antibody binding efficiency.
Validating ATL81 Antibody for high-throughput applications requires specialized approaches that ensure consistency across large sample sets:
Gradient Concentration Testing: Determine optimal antibody concentration using a dilution series across representative tissue sections
Automated Staining Validation: Verify consistent performance in automated staining platforms compared to manual protocols
Spatial Heterogeneity Assessment: Evaluate staining consistency across different regions of the same tissue to account for microenvironmental variations
Temporal Stability Testing: Assess staining intensity over time to determine how long samples can be stored before analysis
Multi-Parameter Algorithm Development: Create image analysis algorithms specific to ATL81 staining patterns for quantitative assessment
For high-throughput screening applications, researchers should implement quality control measures at each step of the workflow. This includes regular testing of the antibody against reference standards and inclusion of control samples in each experimental batch to monitor inter-assay variability.
Statistical analysis of ATL81 Antibody data requires approaches that account for both technical and biological variability:
Normalization Strategies:
Within-plate normalization for microplate-based assays
Between-batch correction using reference standards
Intensity normalization relative to housekeeping proteins
Variability Assessment:
Coefficient of variation (CV) calculation for technical replicates
Intraclass correlation coefficient (ICC) for assessing reproducibility
Bland-Altman plots for comparing different detection methods
Significance Testing:
Account for multiple comparisons when analyzing large datasets
Consider non-parametric tests when distribution assumptions aren't met
Implement mixed-effects models to account for nested experimental designs
Data Integration Methods:
Develop correlation matrices between antibody-based and orthogonal measurements
Apply dimension reduction techniques for visualizing complex relationships
Implement Bayesian approaches for integrating prior knowledge with experimental data
Researchers should consider that different applications of ATL81 Antibody might require distinct statistical approaches. For example, quantitative analysis of immunoblots might benefit from regression-based methods, while spatial analysis of immunofluorescence might require specialized image analysis algorithms.
Non-specific binding represents a significant challenge in antibody-based research and can lead to misleading results. Common causes and mitigation strategies include:
Blocking Optimization:
Insufficient blocking is a primary cause of non-specific binding
Test multiple blocking agents (BSA, casein, normal serum) to identify optimal conditions
Extend blocking time in challenging samples with high background
Buffer Composition:
Adjust salt concentration to modify electrostatic interactions
Add detergents (Tween-20, Triton X-100) at appropriate concentrations
Consider adding carrier proteins to reduce non-specific interactions
Antibody Concentration:
Perform titration experiments to identify the optimal concentration that maximizes signal-to-noise ratio
Remember that higher concentrations often increase both specific and non-specific binding
Cross-Adsorption:
Pre-adsorb the antibody with proteins/tissues known to cause cross-reactivity
Consider using knockout cell lysates for pre-adsorption
Validation Controls:
Always include isotype controls and secondary-only controls
Use knockout or knockdown samples as negative controls
The specificity of antibodies is a critical challenge in biomedical research, with many commercially available antibodies lacking adequate specificity. This results in off-target effects that compromise research validity .
When faced with contradictory results using ATL81 Antibody across different applications, researchers should implement a systematic reconciliation approach:
Condition Mapping:
Create a detailed comparison of experimental conditions between contradictory results
Identify all variables that differ between applications (buffers, fixation, incubation time)
Epitope Accessibility Analysis:
Determine if sample preparation methods might differentially affect epitope exposure
Consider native versus denatured conditions and their impact on antibody binding
Orthogonal Validation:
Employ alternative detection methods that don't rely on antibodies
Use genetic approaches (siRNA, CRISPR) to manipulate target protein levels
Compare results with other antibodies targeting different epitopes of the same protein
Context-Dependent Expression:
Investigate whether the target protein undergoes post-translational modifications that might affect antibody binding
Consider potential protein-protein interactions that might mask the epitope in specific applications
Quantitative Comparison:
Develop standardized metrics to quantitatively compare results across applications
Establish threshold criteria for determining significant discrepancies
Recent initiatives like YCharOS test antibodies across multiple applications (immunoblotting, immunoprecipitation, and immunofluorescence) to provide comprehensive characterization and help researchers understand application-specific performance variations .
Optimizing ATL81 Antibody performance for challenging samples requires advanced techniques beyond standard protocols:
Sample Pre-treatment Options:
Antigen retrieval method optimization (heat-induced vs. enzymatic)
Extended deparaffinization for FFPE tissues
Controlled fixation time to balance structural preservation and epitope accessibility
Signal Amplification Techniques:
Tyramide signal amplification for low-abundance targets
Multi-layer detection systems for enhanced sensitivity
Polymer-based detection systems to reduce background
Protocol Modifications:
Extended incubation times at lower temperatures
Pulsed antibody incubation with washing steps
Specialized permeabilization protocols for difficult-to-access targets
Buffer Optimization:
pH gradient testing to identify optimal binding conditions
Additives such as polyethylene glycol or dextran sulfate to reduce non-specific binding
Specialized detergents for membrane protein extraction
Combinatorial Approach:
Systematic testing of multiple variables simultaneously using design of experiments (DoE) methodology
Development of sample-specific protocols based on tissue/cell characteristics
Researchers working with challenging samples should consider implementing the standardized characterization processes developed by initiatives like YCharOS, which has tested approximately 1,200 antibodies against 120 protein targets to establish robust protocols .
Integrating ATL81 Antibody with computational methods represents an advanced approach to target validation:
Biophysics-Informed Modeling:
Epitope Mapping Integration:
Compare experimental epitope mapping results with computational predictions
Use structural biology data to refine computational models
Predict potential cross-reactivity based on structural similarity to target epitope
Machine Learning Applications:
Systems Biology Integration:
Map antibody-generated data onto protein interaction networks
Identify potential off-target effects through network analysis
Validate findings through targeted experiments
Recent advances in computational antibody design demonstrate that biophysics-informed models can successfully disentangle multiple binding modes associated with specific ligands, enabling the prediction and generation of specific variants beyond those observed in experiments .
Spatially-resolved proteomics represents a frontier in biomedical research, with antibodies playing a crucial role. Recent methodological advances applicable to ATL81 include:
Multiplexed Imaging Techniques:
Cyclic immunofluorescence (CycIF) for sequential staining with multiple antibodies
Mass cytometry imaging (IMC) for highly multiplexed protein detection
Co-detection by indexing (CODEX) for high-parameter tissue imaging
Spatial Transcriptomics Integration:
Combined antibody and RNA detection in the same tissue section
Correlation of protein localization with gene expression patterns
Multi-modal data integration frameworks
Advanced Image Analysis:
Cell segmentation algorithms for single-cell protein quantification
Spatial statistics for analyzing protein distribution patterns
Machine learning approaches for feature extraction and pattern recognition
Microenvironment Characterization:
Analysis of protein expression in the context of tissue architecture
Quantification of cell-cell interactions based on protein expression
Spatial mapping of signaling pathway activities
These advanced applications require rigorous validation of antibody specificity. Standardized characterization approaches, such as those developed by YCharOS, are essential for ensuring reliable results in these complex experimental systems .