HLA-C Antibody, Biotin conjugated

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
We typically dispatch orders within 1-3 business days of receipt. Delivery times may vary depending on the shipping method and destination. Please contact your local distributor for specific delivery information.
Synonyms
HLA class I histocompatibility antigen, Cw-6 alpha chain (MHC class I antigen Cw*6), HLA-C, HLAC
Target Names
Uniprot No.

Q&A

What is HLA-C and why are antibodies against it important in research?

HLA-C is a component of the major histocompatibility complex class I (MHC I) family that plays a critical role in immune response regulation. The human version of HLA-C has a canonical amino acid length of 366 residues and a protein mass of 40.6 kilodaltons, with two identified isoforms. It is primarily localized in the endoplasmic reticulum and cell membrane . Unlike other MHC I molecules, HLA-C has unique properties in immune regulation, particularly in natural killer (NK) cell function and viral defense mechanisms.

HLA-C antibodies enable researchers to detect, quantify, and characterize HLA-C expression in various biological contexts. These antibodies are instrumental in understanding immune regulation, transplantation biology, viral immune evasion, and disease associations. The biotin conjugation provides amplification of detection signals through the strong biotin-streptavidin interaction, enabling greater sensitivity in applications such as flow cytometry and immunohistochemistry .

How do W6/32 clone antibodies differ from HLA-C specific antibodies?

The W6/32 clone represents a pan-HLA class I antibody that recognizes a conformational epitope formed by the association of the HLA-A, HLA-B, or HLA-C heavy chain with β2-microglobulin. This distinguishes it from antibodies that recognize only HLA-C specifically . The key differences include:

FeatureW6/32 CloneHLA-C Specific Antibodies
RecognitionConformational epitope on HLA-A, B, C when associated with β2-microglobulinLinear or conformational epitopes unique to HLA-C
ApplicationsBroad assessment of MHC I expressionSpecific analysis of HLA-C contribution
Research UseGeneral immune function, transplantationNK cell interactions, viral studies specific to HLA-C
Epitope DependencyLost upon denaturationMay be retained depending on antibody
Cross-reactivityRecognizes multiple HLA allelesLimited to HLA-C alleles with varying specificity

The choice between W6/32 and HLA-C specific antibodies depends on research objectives. W6/32 is ideal for general MHC I expression studies, while HLA-C specific antibodies are essential when distinguishing HLA-C from other class I molecules is critical .

What are optimal sample preparation protocols for biotin-conjugated HLA-C antibody applications?

Sample preparation significantly impacts the quality of results when using biotin-conjugated HLA-C antibodies. For optimal results, researchers should follow these methodological guidelines:

For flow cytometry:

  • Harvest cells in log-phase growth (for cultured cells) or process fresh clinical samples within 24 hours of collection

  • Wash cells twice in cold PBS containing 2% fetal bovine serum to reduce background

  • Block Fc receptors using appropriate blocking reagent for 15 minutes at 4°C

  • For the standard protocol, use ≤1.0 μg of biotin-conjugated HLA-C antibody per 10^6 cells in 100 μL volume or per 100 μL of whole blood

  • Incubate for 30 minutes at 4°C protected from light

  • Wash twice with PBS/2% FBS

  • Add streptavidin-conjugated fluorophore at optimized concentration

  • Incubate for 20 minutes at 4°C protected from light

  • Wash twice and analyze immediately or fix in 1-2% paraformaldehyde for later analysis

For preserved tissues, antigen retrieval methods significantly affect epitope accessibility. Heat-induced epitope retrieval using citrate buffer (pH 6.0) typically yields better results than proteolytic enzyme-based methods for HLA-C detection in formalin-fixed, paraffin-embedded tissues.

What controls should be incorporated when working with biotin-conjugated HLA-C antibodies?

Appropriate controls are essential for reliable interpretation of results when using biotin-conjugated HLA-C antibodies:

Control TypePurposeImplementation
Isotype ControlAssesses non-specific bindingUse biotin-conjugated mouse IgG2a, κ (for W6/32 clone)
Unstained ControlEstablishes autofluorescence baselineProcess cells without any antibody
FMO ControlDetermines boundary for positive populationsInclude all fluorochromes except the one conjugated to streptavidin
Positive ControlConfirms staining protocol efficacyUse cell line with known high HLA-C expression (e.g., lymphoblastoid cell lines)
Negative ControlValidates specificityUse β2-microglobulin-deficient cells or HLA-null cell lines
Blocking ControlVerifies specificityPre-incubate cells with excess unconjugated antibody
Secondary Only ControlDetects non-specific streptavidin bindingOmit primary antibody, add only streptavidin conjugate

These controls should be processed identically to experimental samples. For quantitative studies, calibration beads should be included to standardize fluorescence intensity measurements across experiments and instruments.

How can biotin-conjugated HLA-C antibodies be optimized for multiparameter flow cytometry?

Optimizing biotin-conjugated HLA-C antibodies for multiparameter flow cytometry requires attention to several methodological considerations:

  • Panel Design: Place the streptavidin-conjugated fluorophore in an appropriate detection channel based on expression level. For typically high-expression HLA molecules, dimmer fluorophores like Pacific Blue or PE-Cy5 are suitable, while preserving brighter fluorophores like PE or APC for lower-expression markers.

  • Titration: Perform a titration series (typically 0.1-5.0 μg/10^6 cells) to determine the optimal concentration where the positive signal is maximized while background remains minimal. Calculate the staining index for each concentration:

    Staining Index = (MFI positive - MFI negative) / (2 × SD of negative)

  • Signal Amplification Options: For enhanced sensitivity, consider:

Amplification MethodAdvantagesLimitations
Streptavidin-PE5-10× signal increasePotential background increase
Tyramide Signal Amplification10-50× signal increaseRequires enzymatic step
Sequential Multilayer EnhancementUp to 100× signal increaseTime-consuming, may increase background
  • Reducing Background: Implement the following strategies:

    • Pre-block endogenous biotin with avidin/biotin blocking kit

    • Include 10% normal mouse serum to reduce non-specific binding

    • Use optimized washing buffer (PBS with 0.5% BSA and 0.02% sodium azide)

    • Consider adding 1 mM EDTA to prevent cell aggregation

  • Compensation: Due to the brightness of streptavidin conjugates, prepare separate single-color controls specifically for the streptavidin channel to avoid compensation errors in multiparameter analyses.

  • Order of Addition: For multimarker panels that include biotin-conjugated antibodies, apply the biotin-conjugated antibody first, followed by the streptavidin conjugate before adding directly labeled antibodies to minimize interference.

What are the methodological considerations for using HLA-C antibodies in transplantation research?

Transplantation research requires particularly careful application of HLA-C antibodies, considering:

  • Allele-Specific Recognition: HLA-C exhibits significant polymorphism. The W6/32 clone recognizes a conserved epitope, but analysis of specific HLA-C alleles may require allele-specific antibodies. Document the HLA typing of samples and consider how antibody epitope specificity relates to the alleles present .

  • Donor-Recipient Matching Assessment:

    • For flow crossmatch procedures, use donor lymphocytes incubated with recipient serum followed by biotin-conjugated anti-HLA-C detection

    • Implement fluorescence ratio calculations: (Mean channel fluorescence with test serum) ÷ (Mean channel fluorescence with negative control serum)

    • A ratio >1.5 typically indicates presence of donor-specific antibodies

  • Post-Transplant Monitoring Protocol:

    • Establish baseline HLA-C expression levels pre-transplant

    • Monitor for changes in expression levels that might indicate rejection

    • Correlate with other immune activation markers

    • Compare peripheral blood with biopsy samples when available

  • Analysis of Antibody-Mediated Rejection:

    • Combine HLA-C detection with C4d staining

    • Implement multiparameter assessment including NK cell activation markers

    • Correlate with donor-specific antibody titers in serum

  • Accommodation and Tolerance Studies:

    • Monitor HLA-C expression changes during accommodation

    • Assess regulatory T-cell interactions with HLA-C expressing cells

    • Implement time-course studies to capture dynamic changes

When integrating biotin-conjugated HLA-C antibodies into transplantation research workflows, researchers should validate the specificity against the particular HLA-C alleles relevant to their donor-recipient pairs to avoid misinterpretation of results.

How do fixation and permeabilization protocols affect HLA-C epitope detection?

Fixation and permeabilization can significantly impact the detection of HLA-C epitopes, requiring careful methodological consideration:

Fixation MethodImpact on HLA-C EpitopesRecommended Applications
Paraformaldehyde (1-4%)Preserves conformational epitopes recognized by W6/32; may reduce signal intensity at concentrations >2%Flow cytometry, immunohistochemistry for cell surface HLA-C
MethanolDisrupts conformational epitopes; enhances detection of linear epitopesInappropriate for W6/32 detection; may work for some HLA-C specific antibodies
AcetoneDisrupts membrane structure; may expose intracellular epitopesGenerally not recommended for HLA class I detection
GlutaraldehydeOver-fixes epitopes; generally reduces antibody bindingAvoid for HLA detection
EthanolPartially preserves conformational epitopesAcceptable for some applications with reduced sensitivity

For permeabilization protocols when intracellular HLA-C detection is required:

  • Saponin-based permeabilization (0.1-0.5%): Gentle and reversible; maintains most conformational epitopes recognized by W6/32 while allowing access to intracellular compartments. Optimal for detecting HLA-C in the endoplasmic reticulum.

  • Triton X-100 permeabilization: More aggressive; may disrupt conformational epitopes. Use at low concentrations (0.1%) and short incubation times if necessary.

  • Commercial kits comparison: In a comparative study of permeabilization methods for HLA detection:

Permeabilization MethodRelative Signal Preservation (%)Background LevelRecommended Use
BD Cytofix/Cytoperm85-90%LowGeneral-purpose detection
eBioscience Foxp3 Buffer Set60-70%LowWhen combining with transcription factor staining
Methanol-based methods40-50%ModerateNot recommended for W6/32
0.1% Saponin custom80-85%Very lowPreferred for quantitative studies

For optimal results with the W6/32 clone, gentle fixation with 1-2% paraformaldehyde followed by minimal or no permeabilization is recommended for cell surface HLA-C detection, as the conformational epitope is sensitive to harsh treatments .

What are the implications of HLA-C polymorphisms for antibody binding and specificity?

HLA-C is highly polymorphic with over 5,000 known alleles, creating significant challenges for antibody selection and data interpretation:

  • Epitope Conservation Analysis:

    • The W6/32 clone recognizes a conserved epitope formed by the α3 domain interaction with β2-microglobulin, making it largely insensitive to HLA polymorphisms

    • HLA-C specific antibodies typically target polymorphic regions and may show allele-dependent binding variation

  • C1/C2 Epitope Groups:
    HLA-C alleles are classified into C1 and C2 groups based on amino acid positions 77-80, which interact with different KIR receptors:

GroupKey ResiduesTypical AllelesKIR Interaction
C1Ser77, Asn80C01, C03, C07, C08KIR2DL2/3
C2Asn77, Lys80C02, C04, C05, C06KIR2DL1

Antibodies targeting these regions may show group-specific rather than pan-HLA-C reactivity.

  • Cross-Reactivity Assessment:
    When selecting HLA-C antibodies, researchers should consider potential cross-reactivity with:

    • Other HLA class I molecules (particularly HLA-B)

    • Non-classical HLA molecules (HLA-E, HLA-F, HLA-G)

    • MHC-like molecules (MICA, MICB)

  • Validation Methodology:

    • Test antibodies against cell lines with known HLA types

    • Use HLA-C knockout models as negative controls

    • Confirm specificity with competitive binding assays

    • Validate across multiple detection platforms

  • Experimental Design Considerations:

    • Document HLA typing of all experimental samples

    • Include allele representation from both C1 and C2 groups

    • Report antibody binding patterns in relation to HLA-C alleles present

    • Consider population-specific HLA-C frequency distributions in study design

Researchers must be aware that even well-characterized antibodies like W6/32 may show subtle variations in binding affinity across different HLA-C alleles, potentially introducing bias when comparing samples with different HLA types .

How can biotin-conjugated HLA-C antibodies be used to investigate immune evasion in viral infections and cancer?

Biotin-conjugated HLA-C antibodies offer powerful tools for investigating immune evasion mechanisms in both viral infections and cancer, with several methodological approaches:

  • Quantitative Expression Analysis:
    Using calibrated flow cytometry:

    • Measure HLA-C surface density on infected vs. uninfected cells

    • Compare HLA-C expression between tumor and matched normal tissues

    • Calculate molecules of equivalent soluble fluorochrome (MESF) values using standardization beads

    • Implement time-course studies to capture dynamic expression changes

  • Intracellular Trafficking Studies:

    • Combine biotin-conjugated HLA-C surface staining with permeabilization and intracellular staining

    • Quantify surface-to-intracellular ratios to assess retention/internalization

    • Implement confocal microscopy with biotin-streptavidin detection systems for high-resolution localization

  • Immune Synapse Formation Analysis:

    • Use biotin-conjugated HLA-C antibodies in conjugate formation assays

    • Visualize immune synapse organization using structured illumination microscopy

    • Quantify HLA-C clustering at NK-cell contact sites

  • Viral Protein Interference Assessment:
    Methodological approach for viral interference studies:

StepTechniqueMeasurement
1Flow cytometryBaseline HLA-C expression
2Viral infection/transfectionIntroduction of viral protein
3Time-course HLA-C stainingDynamic expression changes
4Co-immunoprecipitationPhysical interactions with viral proteins
5Peptide-MHC complex detectionFunctional impact on antigen presentation
  • Cancer Immunotherapy Applications:

    • Monitor HLA-C expression changes following immune checkpoint inhibitor treatment

    • Correlate expression levels with treatment response

    • Identify tumor subpopulations with differential HLA-C expression

    • Develop companion diagnostics for immunotherapy selection

  • CRISPR-Cas9 Screening Integration:

    • Use biotin-conjugated HLA-C antibodies as readouts in genetic screens

    • Identify novel regulators of HLA-C expression

    • Sort cells based on HLA-C expression for next-generation sequencing

    • Validate hits with targeted knockout/knockin approaches

For these applications, researchers should employ the recommended antibody concentration of ≤1.0 μg per 10^6 cells and optimize titration for each specific application context to maximize signal-to-noise ratio .

What are common pitfalls when using biotin-conjugated HLA-C antibodies and how can they be resolved?

Researchers frequently encounter several challenges when working with biotin-conjugated HLA-C antibodies. Here are methodological solutions:

  • High Background Signal:

    • Cause: Endogenous biotin or non-specific binding

    • Solution: Implement avidin/biotin blocking kit prior to antibody staining; increase wash steps; add 2% BSA or 5-10% serum from the same species as secondary reagent

  • Weak or Absent Signal:

    • Cause: Epitope masking, low expression, or antibody inactivation

    • Solution: Optimize fixation protocol; try antigen retrieval; increase antibody concentration; ensure proper storage at 4°C and avoid freeze-thaw cycles

  • Inconsistent Results Between Experiments:

    • Cause: Variations in cell preparation, antibody lots, or instrument settings

    • Solution: Standardize protocols; use calibration beads; include standard samples across experiments; document lot numbers

  • Poor Discrimination Between Positive and Negative Populations:

    • Cause: Suboptimal antibody concentration or fluorophore selection

    • Solution: Perform titration experiments to determine optimal concentration; switch to brighter fluorophore if needed

  • Loss of Signal During Multicolor Staining:

    • Cause: Antibody competition or fluorescence quenching

    • Solution: Optimize staining sequence; separate potentially competing antibodies into different panels; use fluorophores with minimal spectral overlap

  • Selective Loss of Certain HLA-C Alleles:

    • Cause: Epitope polymorphism affecting antibody binding

    • Solution: Verify antibody specificity against cells with known HLA-C types; consider using alternative clones or a combination of antibodies

Table of common issues and quantitative improvements with optimization:

IssueStandard ProtocolOptimized ProtocolImprovement Factor
Background signalDirect stainingAvidin/biotin blocking + 2% BSA3-5× reduction in background
Signal strengthFixed concentrationOptimized titration1.5-3× increase in staining index
Experiment variationBasic protocolWith calibration standardsCV reduced from 25-30% to 5-10%
Population resolutionStandard settingsOptimized PMT voltage2× improvement in resolution

How can biotin-conjugated HLA-C antibodies be used for quantitative analyses in research settings?

For quantitative applications, biotin-conjugated HLA-C antibodies require specific methodological refinements:

  • Absolute Quantification Protocol:

    • Use calibrated beads with known quantities of biotin-binding sites

    • Establish a calibration curve relating fluorescence intensity to molecule numbers

    • Apply conversion factor to convert sample fluorescence to molecules per cell

    • For W6/32 detection, ≤1.0 μg per 10^6 cells provides reliable quantitative data when properly calibrated

  • Signal Linearity Assessment:
    Before quantitative studies, verify signal linearity across the expected expression range:

    • Test cell lines with different expression levels

    • Plot measured values against expected expression

    • Calculate R² value (should exceed 0.95 for quantitative applications)

    • Adjust protocol if non-linearity is observed

  • Multi-Platform Standardization:
    For consistent quantification across different instruments or labs:

    • Use ABC (Antibody Binding Capacity) beads

    • Implement MESF (Molecules of Equivalent Soluble Fluorochrome) standards

    • Document instrument settings and standardization procedures

    • Participate in proficiency testing programs

  • Comparative Expression Analysis:
    When comparing HLA-C expression between experimental conditions:

    • Report fold-change relative to controls

    • Calculate statistical significance using appropriate tests

    • Present data as both raw values and normalized results

    • Consider biological relevance thresholds (typically >1.5-fold change)

  • Kinetic Measurements:
    For time-course studies of HLA-C expression:

    • Establish consistent harvesting and processing times

    • Use automated systems when possible to reduce time-dependent variables

    • Include time-matched controls

    • Consider both rate of change and absolute differences in expression

By implementing these quantitative approaches, researchers can obtain reliable, reproducible measurements of HLA-C expression levels suitable for comparative studies and publication-quality data.

How are biotin-conjugated HLA-C antibodies being integrated with single-cell technologies?

The integration of biotin-conjugated HLA-C antibodies with single-cell technologies represents an emerging frontier with several methodological approaches:

  • CITE-seq Applications:

    • Conjugate HLA-C antibodies to DNA barcodes rather than biotin

    • Capture surface protein expression and transcriptome simultaneously

    • Correlate HLA-C protein levels with gene expression profiles

    • Identify cell states associated with altered HLA-C expression

  • Imaging Mass Cytometry:

    • Use metal-tagged streptavidin to detect biotin-conjugated HLA-C antibodies

    • Achieve subcellular resolution of HLA-C localization

    • Multiplex with >40 other markers simultaneously

    • Analyze spatial relationships between HLA-C+ cells and immune populations

  • Single-Cell Functional Assays:

    • Sort cells based on HLA-C expression using biotin-conjugated antibodies

    • Perform downstream functional assays on sorted populations

    • Implement microfluidic systems for single-cell cytotoxicity assays

    • Correlate HLA-C expression with functional outcomes at single-cell resolution

  • Spatial Transcriptomics Integration:

    • Combine immunofluorescence using biotin-conjugated HLA-C antibodies with spatial transcriptomics

    • Map HLA-C protein expression onto tissue geography

    • Correlate with local immune infiltration and activation states

    • Identify tissue microenvironments with altered HLA-C expression

These emerging applications require careful validation and optimization of biotin-conjugated HLA-C antibodies for each specific technological platform, with particular attention to signal-to-noise ratio and specificity in multiplexed systems.

What are the latest methodological advances in using HLA-C antibodies for immunotherapy research?

Recent methodological advances have expanded the utility of HLA-C antibodies in immunotherapy research:

  • CAR-T Cell Engineering Applications:

    • Use biotin-conjugated HLA-C antibodies to assess target antigen density

    • Monitor HLA-C expression changes following immunotherapy

    • Screen for potential off-target binding of therapeutic antibodies

    • Evaluate HLA-C expression in treatment-resistant populations

  • Checkpoint Inhibitor Research:

    • Correlate HLA-C expression with response to checkpoint blockade

    • Examine HLA-C peptide presentation changes following treatment

    • Assess co-localization of HLA-C with PD-L1 and other checkpoint molecules

    • Develop companion diagnostics based on HLA-C expression patterns

  • NK Cell-Based Immunotherapies:
    HLA-C interacts with KIR receptors on NK cells, making it particularly relevant for NK-based therapies:

NK Cell Therapy ApproachHLA-C Assessment MethodClinical Relevance
Adoptive NK transferHLA-C typing and expression quantificationPredict inhibitory KIR engagement
KIR-transgenic NK cellsBinding assays with target HLA-C allelesOptimize engineering strategy
Checkpoint-blocked NK cellsHLA-C/KIR interaction analysisPredict responsiveness
NK-CAR cellsHLA-C expression on off-target tissuesSafety assessment
  • Bispecific Antibody Development:

    • Screen for potential cross-reactivity with HLA-C alleles

    • Assess impact of bispecific engagement on HLA-C expression

    • Optimize dosing based on HLA-C expression levels

    • Develop HLA-C targeting therapeutic strategies

These advanced applications typically require more sophisticated analysis than standard detection, often combining biotin-conjugated HLA-C antibodies with other detection systems in multiparameter analyses for comprehensive assessment of the immunotherapy landscape.

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.