SHE Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
**Preservative:** 0.03% Proclin 300
**Constituents:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship orders within 1-3 business days of receipt. Delivery timelines may vary based on the chosen purchasing method or location. For specific delivery estimates, we recommend reaching out to your local distributor.
Synonyms
RP11-350G8.8 antibody; SH2 domain-containing adapter protein E antibody; She antibody; SHE_HUMAN antibody; Src homology 2 domain containing E antibody
Target Names
SHE
Uniprot No.

Q&A

What constitutes proper antibody characterization in research applications?

Proper antibody characterization requires documentation of four essential elements: (1) confirmation that the antibody binds to the target protein; (2) verification that binding occurs within complex protein mixtures such as cell lysates or tissue sections; (3) demonstration of specificity with no cross-reactivity to non-target proteins; and (4) validation that the antibody performs as expected under the specific experimental conditions employed . Comprehensive characterization typically involves multiple complementary techniques including Western blotting, immunoprecipitation, immunofluorescence, and ELISA. Recombinant antibodies typically outperform both monoclonal and polyclonal antibodies across multiple assays, making them preferable when available .

What essential controls should be included when using antibodies in research?

The gold standard control for antibody experiments is the use of knockout cell lines, which have consistently proven superior to other control types, particularly for Western blots and immunofluorescence imaging . Additional recommended controls include:

  • Positive controls: Samples known to express the target protein at detectable levels

  • Negative controls: Samples known to lack the target protein

  • Isotype controls: Non-specific antibodies of the same isotype and concentration

  • Secondary antibody-only controls: To detect non-specific binding of secondary antibodies

  • Antigen competition assays: Pre-incubation with the target antigen should abolish specific signals

  • Dilution series: To establish the optimal antibody concentration for signal-to-noise ratio

Importantly, controls should match the experimental samples in terms of processing, fixation, and other relevant parameters to ensure valid comparisons .

How do monoclonal, polyclonal, and recombinant antibodies compare in research applications?

Each antibody type offers distinct advantages and limitations for research applications:

Antibody TypeSpecificityBatch ConsistencyRenewableProduction ComplexityBest Applications
MonoclonalHighHighYesModerateApplications requiring high specificity to a single epitope
PolyclonalVariable (recognizes multiple epitopes)LowNoLowDetection of denatured proteins, low abundance targets
RecombinantHighestHighestYesHighCritical research requiring maximum reproducibility

What strategies exist for resolving contradictory results when using antibodies across different experimental platforms?

Contradictory results across experimental platforms often stem from context-dependent antibody behavior. To resolve such discrepancies:

  • Cross-validate using multiple antibodies: Test independent antibodies targeting different epitopes of the same protein

  • Perform complementary assays: Confirm results using orthogonal methods (e.g., mass spectrometry)

  • Optimize conditions for each platform: Antibodies may require platform-specific optimization of concentration, incubation time, and buffers

  • Consider post-translational modifications: These can alter epitope accessibility in different experimental contexts

  • Evaluate fixation impact: Different fixation methods may differentially affect epitope availability

  • Implement knockout validation: Test antibody specificity in knockout models across all experimental platforms

Particularly challenging cases may require returning to basic validation steps in each experimental system, as antibodies that perform well in one application may fail in others due to differences in protein conformation, sample preparation, or detection methods .

How can researchers distinguish between specific binding and experimental artifacts when using antibodies?

Distinguishing specific binding from artifacts requires systematic validation approaches:

  • Signal patterns analysis: Compare observed patterns with known biological distribution of the target

  • Signal-to-noise ratio assessment: Evaluate background relative to specific signal

  • Concentration-dependent signal changes: Specific binding typically shows dose-dependent changes within a certain range

  • Knockout validation: Absence of signal in knockout samples confirms specificity

  • Multiple antibody concordance: Convergent results from antibodies targeting different epitopes support specificity

  • Peptide competition: Pre-incubation with the immunizing peptide should abolish specific signals

  • Application-specific controls: For immunohistochemistry, include tissue known to lack expression; for flow cytometry, include fluorescence-minus-one controls

The YCharOS initiative has established that knockout cell line testing provides the most definitive method for distinguishing specific from non-specific binding, particularly for Western blot and immunofluorescence applications .

What is the optimal workflow for validating antibodies in immunohistochemistry applications?

Validating antibodies for immunohistochemistry (IHC) requires a comprehensive workflow:

  • Literature and database review: Examine published characterization data and vendor specifications

  • Western blot pre-validation: Confirm antibody detects a band of the expected molecular weight

  • Titration optimization: Determine optimal antibody concentration using serial dilutions

  • Positive control tissues: Test on tissues known to express the target at varying levels

  • Negative control tissues: Confirm absence of signal in tissues known to lack the target

  • Knockout/knockdown validation: Test on tissues/cells with genetic modification of the target

  • Fixation optimization: Compare different fixation methods and durations

  • Antigen retrieval comparison: Evaluate different antigen retrieval methods

  • Multi-antibody concordance: Compare staining patterns with independent antibodies

  • Orthogonal validation: Correlate IHC results with RNA expression or other protein detection methods

Johns Hopkins researchers have documented widespread inconsistencies in IHC practices, emphasizing the need for rigorous validation to ensure reproducibility . Following standardized protocols similar to those developed by YCharOS can significantly improve reliability .

How should researchers approach antibody validation for complex applications like multiplex immunofluorescence?

Multiplex immunofluorescence presents unique validation challenges requiring additional considerations:

  • Individual validation first: Validate each antibody independently before combining

  • Cross-reactivity assessment: Test each primary antibody with all secondary antibodies to detect cross-reactivity

  • Spectral overlap evaluation: Ensure fluorophores have minimal spectral overlap or apply appropriate compensation

  • Sequential staining validation: Compare results from sequential and simultaneous staining approaches

  • Antibody order optimization: Test different staining sequences to minimize interference

  • Signal stability assessment: Confirm signal stability over time and under imaging conditions

  • Panel-specific controls: Include controls omitting one primary antibody at a time

  • Multiplexed positive controls: Use samples with known co-expression patterns

  • Biological reference patterns: Compare multiplex patterns to known biological relationships

  • Orthogonal validation: Correlate with other methods like single-cell RNA sequencing

Systematic antibody validation for multiplex applications is critical as interaction effects between antibodies can create misleading results even when individual antibodies perform well in single-staining applications .

What quantitative methods exist for comparing antibody performance across different batches or sources?

Quantitative antibody performance comparison requires standardized metrics:

  • Signal-to-noise ratio calculation: Quantify specific signal relative to background

  • Titration curve analysis: Compare EC50 values from serial dilution experiments

  • Limit of detection determination: Establish the minimum detectable amount of target

  • Dynamic range measurement: Quantify the range of target concentrations yielding proportional signals

  • Coefficient of variation analysis: Calculate intra- and inter-assay variability

  • ROC curve analysis: For diagnostic applications, compare sensitivity/specificity profiles

  • Cross-reactivity profiling: Quantify binding to related and unrelated proteins

  • Knockout signal ratio: Compare signal in wild-type vs. knockout samples (ideal ratio approaches infinity)

  • Epitope mapping concordance: Compare epitope recognition patterns

  • Interlaboratory reproducibility: Implement standardized protocols across multiple sites

The YCharOS initiative has developed consensus protocols that enable quantitative comparison of antibodies, revealing that approximately 20% of commercially available antibodies fail to recognize their targets and approximately 40% perform inconsistently across different applications .

How are recombinant antibody technologies changing validation requirements and research practices?

Recombinant antibody technologies are transforming validation practices through several mechanisms:

  • Defined molecular identity: Sequence-defined antibodies eliminate batch-to-batch variation

  • Enhanced reproducibility: Consistent production yields reliable performance across experiments

  • Engineered specificity: Rational design can improve target specificity and reduce cross-reactivity

  • Systematic validation: Standardized production enables comprehensive characterization

  • Public sequence availability: Sequences can be published, enabling independent verification

  • Format flexibility: Recombinant methods allow production of different antibody formats (Fab, scFv, etc.)

  • Reduced animal use: Expression systems eliminate ongoing animal immunization requirements

Large-scale studies have confirmed that recombinant antibodies consistently outperform both monoclonal and polyclonal antibodies across multiple applications . Projects like the Protein Capture Reagents Program (PCRP) and Affinomics have generated thousands of characterized recombinant antibodies targeting human proteins, although comprehensive proteome coverage remains challenging .

What role do knockout cell lines play in modern antibody validation strategies?

Knockout cell lines have emerged as the gold standard for antibody validation, offering several advantages:

  • Definitive specificity testing: Complete absence of the target protein provides the ultimate negative control

  • Application versatility: Effective for validating antibodies across multiple techniques

  • Quantitative assessment: Enables calculation of specific-to-nonspecific signal ratios

  • Isogenic comparison: Wild-type and knockout cells share genetic background except for the target

  • Endogenous protein levels: Tests antibody performance at physiologically relevant concentrations

  • Context-specific validation: Confirms specificity in the cellular context used for experiments

  • Detection of off-target binding: Reveals cross-reactivity with unexpected proteins

The YCharOS initiative demonstrated that knockout cell line validation is particularly critical for immunofluorescence applications, where traditional controls often fail to detect significant off-target binding . Importantly, their work found that approximately 12 publications per protein target included data from antibodies that did not recognize their intended targets, underscoring the value of rigorous knockout-based validation .

How can proteomics approaches complement traditional antibody validation methods?

Mass spectrometry-based proteomics offers powerful complementary approaches to antibody validation:

  • Immunoprecipitation-mass spectrometry: Identifies all proteins captured by an antibody

  • Targeted proteomics: Provides antibody-independent measurement of target proteins

  • Epitope mapping: Defines precise binding sites to predict potential cross-reactivity

  • Post-translational modification analysis: Identifies modifications that affect antibody binding

  • Protein complex characterization: Validates co-immunoprecipitation results

  • Absolute quantification: Establishes reference standards for antibody-based quantification

  • Cross-platform correlation: Correlates antibody-based measurements with MS-based quantification

  • Off-target binding profiling: Identifies proteins that cross-react with antibodies

These orthogonal approaches provide crucial verification of antibody specificity and can identify potential confounding factors in antibody-based experiments, particularly important when working with novel targets or in complex biological systems .

What information should researchers include when reporting antibody usage in publications?

Comprehensive reporting of antibody details is essential for reproducibility:

  • Vendor and catalog number: Exact source identification

  • Clone name/number: For monoclonal or recombinant antibodies

  • RRID (Research Resource Identifier): Unique identifier in the Antibody Registry

  • Lot number: Specific production batch used

  • Host species and isotype: Source organism and antibody class

  • Clonality: Monoclonal, polyclonal, or recombinant

  • Target epitope: Antigen region recognized (if known)

  • Dilution/concentration: Exact antibody amount used

  • Incubation conditions: Time, temperature, buffers

  • Validation performed: Specific validation steps conducted

  • Controls included: Detailed description of experimental controls

  • Pretreatment details: Antigen retrieval, blocking conditions

  • Detection method: Secondary antibody or detection system specifications

  • Known limitations: Any caveats or restrictions in interpretation

The lack of consistent reporting contributes significantly to reproducibility failures, as evidenced by the estimated 50% of commercial antibodies that fail to meet basic characterization standards .

How should researchers address batch-to-batch variability in long-term projects?

Managing batch-to-batch variability requires proactive strategies:

  • Advance planning: Purchase sufficient quantities for entire project when possible

  • Side-by-side testing: Validate new batches alongside previous batches before depletion

  • Reference sample banking: Maintain positive control samples throughout the project

  • Standard curve preservation: Establish and maintain standard curves for quantitative work

  • Performance metrics documentation: Record key metrics for each batch

  • Critical reagent bridging: Implement formal protocols for transitioning between batches

  • Recombinant preference: Use sequence-defined recombinant antibodies when available

  • Orthogonal method comparison: Verify key findings using antibody-independent methods

  • Multi-antibody approach: Use multiple antibodies targeting different epitopes

  • Internal reference inclusion: Include consistent internal controls in each experiment

Batch variability represents a particularly significant challenge with polyclonal antibodies, which demonstrated the poorest consistency in large-scale validation studies compared to monoclonal and recombinant alternatives .

What resources exist to help researchers identify validated antibodies for specific applications?

Several databases and resources can guide researchers to appropriately validated antibodies:

  • CiteAb: Database indexing over 14 million reagents with citation information

  • Antibodypedia: Repository of antibody validation data for research applications

  • The Antibody Registry: Provides RRIDs for antibody tracking across literature

  • YCharOS Reports: Independent characterization data for commercial antibodies

  • Developmental Studies Hybridoma Bank (DSHB): Repository distributing hybridoma-produced antibodies

  • Antibody Characterization Laboratory (ACL): Cancer-focused antibody characterization data

  • Structural Genomics Consortium (SGC): Data on antibodies for epigenetic and structural research

  • Protein Capture Reagents Program (PCRP): Collection of 1,406 monoclonal antibodies targeting 737 human proteins

  • Affinomics Database: Information on protein binding reagents developed in EU programs

  • Zenodo YCharOS Community: Repository of antibody characterization reports

The YCharOS initiative's systematic evaluation of 614 antibodies targeting 65 proteins demonstrated that commercial catalogs likely contain specific and renewable antibodies for more than half of the human proteome, but identifying these reliable reagents requires careful assessment of validation data .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.