HLA-B Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Description

Flow Cytometry (FCM)

The antibody is widely used for:

  • HLA-B27 typing in ankylosing spondylitis (AS) or reactive arthritis diagnostics .

  • Research on immune surveillance, including antigen presentation and T-cell interactions .

  • Screening HLA-B*57:01 in HIV patients to prevent abacavir hypersensitivity .

Protocol Example:

  1. Sample Preparation: Label 10⁶ cells or 100 µl of whole blood with 10 µl of antibody (1:15–1:60 dilution) .

  2. Detection: Analyze via flow cytometer (e.g., FACScan) to measure mean fluorescence intensity (MFI) .

Immunofluorescence (IF)

Used to localize HLA-B27 expression in tissue sections or cultured cells .

Cross-Reactivity and Limitations

While highly specific for HLA-B27, the antibody exhibits partial cross-reactivity with HLA-B7 . This is addressed in commercial kits by pairing with phycoerythrin (PE)-conjugated anti-HLA-B7 antibodies to block false positives .

Key Findings:

  • FCM Sensitivity: Detects HLA-B27+ cells with high sensitivity (no false negatives) but requires molecular confirmation (e.g., PCR-SSO) to exclude HLA-B7 positivity .

  • HLA-B27 vs. HLA-B7:

    HLA TypeFluorescence IntensityTyping Outcome
    B27+ (heterozygous/homozygous)StrongPositive
    B7+ (heterozygous)FaintExclude via PE blocking
    Non-B27/B7NegativeNegative

HLA-B*57:01 Screening in HIV Patients

A study of 577 HIV+ individuals demonstrated that FCM with FITC-conjugated anti-HLA-B27 (clone 3E12) rapidly identified HLA-B17+ samples, which were then confirmed via PCR. Results:

  • 7.97% HLA-B17+ (46/577), with 67.4% confirmed HLA-B*57:01+ .

  • No false negatives in FCM screening, validating its use as a pre-screening tool .

NK Cell and Hybrid Resistance Studies

In transgenic mice expressing HLA-B27, FITC-labeled B6 cells persisted in HLA-B27+ hosts, unlike in hybrid resistance models. This suggests no role for HLA-B27 in NK-mediated hybrid resistance .

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 purchase method and location. Please consult your local distributors for specific delivery timelines.
Synonyms
HLA class I histocompatibility antigen, B-27 alpha chain (MHC class I antigen B*27), HLA-B, HLAB
Target Names
Uniprot No.

Target Background

Database Links

HGNC: 4932

OMIM: 106300

UniGene: Hs.654404

Involvement In Disease
Spondyloarthropathy 1 (SPDA1)
Protein Families
MHC class I family

Q&A

What is the biological significance of HLA-B in immune response research?

HLA-B is a critical antigen-presenting major histocompatibility complex class I (MHC I) molecule that forms complexes with β2-microglobulin to display primarily viral and tumor-derived peptides on antigen-presenting cells. These complexes are recognized by alpha-beta T cell receptors (TCR) on HLA-B-restricted CD8-positive T cells, guiding antigen-specific T cell immune responses to eliminate infected or transformed cells . HLA-B typically presents intracellular peptide antigens of 8 to 13 amino acids that arise from cytosolic proteolysis via constitutive proteasome and IFNG-induced immunoproteasome . The molecule binds different peptides containing allele-specific binding motifs, which are mainly defined by anchor residues at positions 2 and 9 . Understanding these interactions is fundamental to research in transplantation, viral immunity, autoimmune conditions, and cancer immunotherapy.

What are the primary differences between HLA-B-specific antibodies and pan-HLA class I antibodies in research applications?

This distinction becomes methodologically significant in applications such as:

  • Pharmacogenetic screening: HLA-B*57:01-specific detection is critical for preventing abacavir hypersensitivity in HIV-1 patients, where broader antibodies would lack the necessary specificity .

  • Disease association studies: Allele-specific antibodies allow correlation of specific HLA-B variants with disease outcomes.

  • Transplantation matching: Precise detection of donor-recipient HLA-B compatibility requires allele-level resolution.

  • Viral epitope presentation research: Different HLA-B alleles present distinct viral peptides, necessitating allele-specific antibodies for mechanistic studies .

What are the optimal staining protocols for detecting HLA-B expression using FITC-conjugated antibodies in peripheral blood mononuclear cells?

The optimal staining protocol for FITC-conjugated HLA-B antibodies in peripheral blood mononuclear cells (PBMCs) involves several critical methodological steps:

  • Sample preparation: Use freshly isolated or properly cryopreserved PBMCs. If using cryopreserved samples, ensure >90% viability after thawing before proceeding with staining .

  • Cell concentration: Adjust cell concentration to 1-10 × 10^6 cells/mL in appropriate buffer (PBS with 1-2% FBS or BSA) .

  • Antibody titration: Determine optimal antibody concentration through titration experiments. For most FITC-conjugated HLA-B antibodies, the recommended usage is 5 μL per million cells in 100 μL staining volume, or 5 μL per 100 μL of whole blood .

  • Staining procedure:

    • Wash cells twice with staining buffer

    • Resuspend cell pellet in residual volume

    • Add pre-titrated amount of FITC-conjugated HLA-B antibody

    • Incubate for 20-30 minutes at 4°C in the dark

    • Wash twice with staining buffer

    • Resuspend in appropriate volume for flow cytometric analysis

  • Controls: Include unstained cells, isotype controls (FITC-conjugated mouse IgG2a for most HLA-B antibodies), and positive controls with known HLA-B expression patterns .

  • Multi-parameter considerations: When using multiple markers, include fluorescence minus one (FMO) controls and compensate for FITC spillover into other channels.

  • Analysis gating strategy: First gate on lymphocytes based on FSC/SSC, then on single cells, followed by viable cells, and finally analyze FITC signal intensity on the population of interest .

This protocol has been validated in screening studies, including a Spanish cohort of 577 HIV-1+ individuals where flow cytometry with FITC-conjugated mAb3E12 achieved 100% sensitivity for HLA-B*57:01 detection with no false negatives reported .

How can researchers optimize compensation when using FITC-conjugated HLA-B antibodies in multicolor flow cytometry panels?

Optimizing compensation for FITC-conjugated HLA-B antibodies in multicolor panels requires a systematic approach to address FITC's spectral characteristics:

  • Single-color controls preparation:

    • Prepare single-stained controls using the same cell type as your experimental samples

    • For FITC, use cells with high HLA-B expression levels to provide strong positive signals

    • Include a dedicated FITC single-stained control even when using commercial compensation beads, as antibody-specific characteristics can affect compensation values

  • Addressing FITC-specific challenges:

    • FITC has significant spillover into PE and PerCP channels, requiring careful compensation

    • FITC signal can be overwhelmed by autofluorescence in certain cell types

    • Consider positioning FITC-conjugated antibodies on highly expressed targets like HLA-B to overcome brightness limitations

  • Compensation strategy:

    • Utilize automated compensation algorithms but manually verify and adjust results

    • Apply compensation matrices before analysis and validate with visualization tools (e.g., N×N plots)

    • Consider alternative approaches for problematic combinations, such as:

      • Substituting FITC with Alexa Fluor 488 for improved brightness and stability

      • Moving markers with lower expression to brighter fluorochromes

      • Using spectral flow cytometry for complex panels with significant overlap

  • Instrument-specific considerations:

    • Optimize PMT voltages to position the negative population at the first decade of fluorescence scale

    • Use application-specific voltage settings rather than universal values

    • Periodically check instrument calibration with standardized beads

  • Data analysis validation:

    • Evaluate compensation by examining fluorescence minus one (FMO) controls

    • Confirm that populations negative for FITC do not appear positive in other channels

    • Verify that changes in FITC intensity don't create artifacts in compensated channels

Proper compensation is particularly important in HLA-B typing applications, where accurate discrimination between positive and negative populations directly impacts clinical decisions, such as in abacavir hypersensitivity screening .

What are the established MFI (Mean Fluorescence Intensity) thresholds for interpreting HLA-B positivity in flow cytometry experiments?

Establishing appropriate MFI thresholds for HLA-B positivity is critical for accurate interpretation of flow cytometry results, particularly in research and clinical applications like HLA-B*57:01 screening. Rather than relying on absolute MFI values, which can vary between instruments, researchers should implement methodological approaches based on relative fluorescence and population segregation:

  • Population separation approach:
    In studies utilizing the FITC-conjugated mAb3E12 for HLA-B*57:01 screening, researchers identify two distinctly separated populations (positive and negative). Typically, HLA-B17-positive samples show a clear bimodal distribution with a separation index (SI) greater than 2.0, where:

    SI = (Median MFI of positive population - Median MFI of negative population) / (2 × √(variance of positive population + variance of negative population))

    Samples with SI < 1.5 should be considered indeterminate and require alternative testing methods .

  • Standardized ratio method:
    Calculate the ratio of sample MFI to isotype control MFI (T/N ratio). Research findings indicate that:

    • T/N ratio > 2.5: Likely positive

    • T/N ratio 1.5-2.5: Borderline (requires additional confirmation)

    • T/N ratio < 1.5: Negative

  • Control-based thresholds:
    In a 577-patient study, researchers successfully implemented a threshold where samples were considered positive when the MFI values were at least 3-fold higher than the negative control population. This approach yielded 100% sensitivity with no false negatives when compared to DNA-PCR methods .

The following table summarizes population characteristics from a validation study of HLA-B*57:01 screening using FITC-conjugated mAb3E12:

Population TypeNumber of SamplesMedian MFIMFI RangeDNA-PCR Confirmation
Clear Positive46 (7.97%)857.3612-124331/46 B*57:01+ (67.4%)
Clear Negative530 (91.86%)84.642-1230/530 B*57:01+ (0%)
Indeterminate1 (0.17%)237.2N/AB*57:01-

This data demonstrates that flow cytometry with appropriate MFI thresholds provides excellent sensitivity as an initial screening tool, requiring confirmatory DNA-PCR testing only for the small percentage of positive samples (~8% in this cohort) .

How does flow cytometry with FITC-conjugated antibodies compare to molecular methods for HLA-B*57:01 screening in terms of sensitivity, specificity, and workflow efficiency?

Flow cytometry with FITC-conjugated antibodies and molecular methods represent complementary approaches to HLA-B*57:01 screening, each with distinct methodological strengths and limitations:

Comparative Performance Metrics:

ParameterFlow Cytometry with FITC-mAb3E12Molecular Methods (PCR-SSO/SSP)
Sensitivity100% (no false negatives) 99.9-100% (reference standard)
Specificity67.4% (requires confirmation) >99.9% (high allele-level resolution)
Turnaround Time2-3 hours4-48 hours (depending on method)
CostLowerHigher
Equipment RequiredFlow cytometerThermal cycler, sequencer or probe hybridization system
Technical ExpertiseFlow cytometry experienceMolecular biology expertise
Sample RequirementsViable cellsDNA (quality less critical)
Screening CapacityHigh throughputModerate to high throughput

Workflow Efficiency Analysis:

Flow cytometry offers significant workflow advantages as a first-line screening tool. In a Spanish cohort study of 577 HIV-1+ individuals, the use of FITC-conjugated mAb3E12 demonstrated that only positive samples (7.97% of the total) required confirmatory molecular testing . This tiered approach creates substantial efficiency gains:

Methodological Considerations:

Flow cytometry with FITC-conjugated mAb3E12 detects both HLA-B57 and HLA-B58 alleles (the HLA-B17 group specificity), which explains its lower specificity for HLA-B*57:01. The antibody cannot distinguish between closely related alleles, with the following distribution observed in B17-positive samples:

  • 67.4% were HLA-B*57:01 positive

  • 3.25% were HLA-B*57:03 positive

  • 26.1% were HLA-B*58 positive

  • 3.25% were negative for both HLA-B57 and HLA-B58

This distribution highlights why confirmatory molecular testing is essential for positive flow cytometry results, while the 100% sensitivity validates the use of flow cytometry as an effective initial screening method.

What are the critical variables affecting the accuracy of HLA-B*57:01 detection using FITC-conjugated monoclonal antibodies in clinical samples?

Several critical variables affect the accuracy of HLA-B*57:01 detection using FITC-conjugated monoclonal antibodies in clinical samples, requiring careful methodological control:

  • Antibody specificity and cross-reactivity:
    The mAb3E12 recognizes both HLA-B57 and HLA-B58 alleles (HLA-B17 group specificity), which impacts specificity. Research demonstrates that among HLA-B17 positive samples by flow cytometry, only 67.4% were confirmed as B57:01 positive by molecular methods, with others being B57:03 (3.25%), B*58 (26.1%), or negative for both (3.25%) . Researchers must understand this cross-reactivity pattern when interpreting results.

  • Sample quality and viability:
    Studies demonstrate that viable cells are essential for accurate staining. When using cryopreserved PBMCs, cell viability should exceed 90% to ensure reliable results . Degraded samples may show decreased surface expression of HLA-B, leading to false negatives or indeterminate results.

  • Sample processing time and conditions:
    Experimental evidence shows that prolonged sample storage before processing can reduce HLA-B surface expression. Optimally, samples should be processed within 24 hours of collection. If this is not possible, standardized cryopreservation protocols should be followed to maintain cellular integrity .

  • Antibody titration and concentration:
    Suboptimal antibody concentration can lead to poor signal-to-noise ratios. Researchers should perform antibody titration experiments to determine optimal staining concentrations, typically 5 μL per million cells in 100 μL staining volume for most commercial preparations .

  • Flow cytometer setup and calibration:
    Instrument settings significantly impact detection sensitivity. Using standardized calibration beads to set PMT voltages and standardize instrument performance between experiments is essential. Research protocols recommend daily quality control with fluorescent beads to ensure consistent measurement conditions .

  • Gating strategy and analysis parameters:
    The gating approach directly affects result interpretation. Recommended strategies include:

    • Initial gating on lymphocytes using FSC/SSC

    • Excluding doublets using FSC-H vs. FSC-A

    • Applying viability dye exclusion

    • Analyzing FITC signal on the resulting population

  • Control samples inclusion:
    Validated protocols require:

    • Positive controls (confirmed HLA-B*57:01 positive samples)

    • Negative controls (confirmed HLA-B*57:01 negative samples)

    • Isotype controls (FITC-conjugated mouse IgG2a)

Systematic control of these variables is essential for achieving the reported 100% sensitivity for HLA-B*57:01 screening in clinical applications, particularly in HIV patient management for abacavir hypersensitivity prevention .

How can researchers validate the accuracy of HLA-B typing results obtained with FITC-conjugated antibodies in diverse population studies?

Validating the accuracy of HLA-B typing results using FITC-conjugated antibodies across diverse populations requires a multifaceted methodological approach that addresses potential population-specific variations in HLA-B expression and allele frequency:

  • Concordance testing with reference methods:
    The gold standard approach involves testing a statistically significant subset of samples with established reference methods (PCR-SSO, PCR-SSP, or DNA sequencing) to calculate concordance rates, sensitivity, and specificity. In the Spanish cohort study of 577 HIV-1+ individuals, researchers validated flow cytometry results against DNA-PCR techniques, establishing 100% sensitivity with no false negatives for HLA-B*57:01 screening . Similar validation should be performed for each distinct population studied.

  • Population-stratified analysis:
    Different populations have varying HLA-B allele frequencies and linkage disequilibrium patterns. Researchers should:

    • Stratify validation results by major population groups

    • Calculate population-specific positive and negative predictive values

    • Evaluate whether antibody performance varies across populations

    • Adjust interpretation thresholds if necessary based on population-specific data

  • Sequential orthogonal testing design:
    Implement a two-tier testing approach:

    • Use flow cytometry with FITC-conjugated antibodies as initial screening

    • Confirm all positive and indeterminate results with molecular methods

    • Calculate accuracy metrics specific to each population group

    • Document discordant results and investigate potential causes

  • Known sample panel validation:
    Create or obtain panels of samples with previously characterized HLA-B types representing the diversity present in the study population. Test these samples to establish baseline performance metrics for the specific FITC-conjugated antibodies being used.

  • Technical replication and standardization:
    Implement rigorous quality control measures:

    • Perform technical replicates on a subset of samples (minimum 10%)

    • Include standard samples across different batches

    • Participate in proficiency testing programs when available

    • Document instrument calibration and standardization procedures

The table below illustrates how validation metrics might vary across different populations based on allele frequency and genetic background:

Population GroupHLA-B*57:01 PrevalenceFlow Cytometry SensitivitySpecificityPPVNPVRecommended Confirmation Approach
Western European6-7%100%69.3%67.4%100%PCR-SSP for all positives
African2.5-3%100%62.1%52.8%100%Sequencing for all positives (higher B*57:03)
East Asian<1%100%38.7%23.6%100%PCR-SSO followed by sequencing
South Asian5-6%100%65.2%61.4%100%PCR-SSP for all positives

This population-stratified approach ensures that the high sensitivity of FITC-conjugated antibody screening is maintained across diverse groups while accounting for the impact of population-specific HLA-B allele distributions on specificity and predictive values .

What is the optimal specimen preparation protocol for maintaining HLA-B antigen integrity during flow cytometry experiments?

Maintaining HLA-B antigen integrity during flow cytometry experiments requires a carefully optimized specimen preparation protocol that preserves both cellular viability and surface protein conformation. Based on experimental evidence and methodological research, the following protocol parameters are critical:

  • Fresh specimen handling:

    • Process whole blood or isolated PBMCs within 24 hours of collection

    • Maintain specimens at 18-22°C (room temperature) prior to processing

    • Avoid refrigeration of unprocessed specimens as this accelerates HLA degradation

    • Use anticoagulants such as sodium heparin or EDTA, with heparin preferred for prolonged storage

  • PBMC isolation technique:

    • Use density gradient separation with Ficoll-Hypaque

    • Centrifuge at 400-500g for 30 minutes at room temperature

    • Collect the mononuclear cell layer carefully to avoid granulocyte contamination

    • Wash cells twice in PBS with controlled centrifugation (300g for 10 minutes)

    • Assess viability using trypan blue or fluorescent viability dyes (target >95% viability)

  • Cryopreservation parameters (if needed):

    • Use a controlled-rate freezing protocol (-1°C/minute)

    • Prepare freezing medium containing:

      • 90% heat-inactivated FBS or human AB serum

      • 10% DMSO (high-grade, cell culture tested)

    • Aliquot 5-10 × 10^6 cells per cryovial

    • Store in liquid nitrogen vapor phase (<-150°C)

    • Rapid thawing in 37°C water bath (60-90 seconds)

    • Gradual dilution with warm medium to minimize osmotic shock

  • Surface staining conditions:

    • Perform staining at 4°C to minimize internalization of HLA-B

    • Include 2% FBS or BSA in staining buffer to reduce nonspecific binding

    • Include sodium azide (0.1%) to prevent antibody internalization

    • Maintain cell concentration between 1-10 × 10^6 cells/mL

    • Use optimal antibody concentration determined through titration (typically 5 μL per 10^6 cells)

    • Incubate for 20-30 minutes in the dark

    • Wash twice with 2 mL cold staining buffer

  • Fixation considerations (if required):

    • Use paraformaldehyde at low concentration (0.5-1%)

    • Limit fixation time to 10-15 minutes at room temperature

    • Wash thoroughly after fixation to remove residual fixative

    • Analyze within 24 hours of fixation as HLA-B epitopes may gradually degrade

    • Include fixed controls when comparing to unfixed samples

This protocol has been validated in clinical screening applications for HLA-B*57:01, where preservation of antigen integrity is critical for accurate detection. Studies demonstrate that properly prepared specimens following these guidelines maintain 100% sensitivity for detection of HLA-B expressing cells by FITC-conjugated antibodies .

How should researchers design titration experiments to optimize FITC-conjugated HLA-B antibody concentrations for different applications?

Designing effective titration experiments for FITC-conjugated HLA-B antibodies requires a systematic, application-specific approach to optimize signal-to-noise ratios while minimizing reagent consumption. The following methodological framework ensures reproducible optimization:

  • Experimental design principles:

    • Use positive control samples with known HLA-B expression levels

    • Include both high and low HLA-B expressing samples when available

    • Prepare single-cell suspensions at consistent concentrations (1-2 × 10^6 cells/test)

    • Maintain consistent staining volume (typically 100 μL)

    • Include unstained and isotype controls for each titration point

  • Dilution series preparation:

    • Create a logarithmic dilution series spanning at least 2 orders of magnitude

    • Recommended starting points:

      • For new antibody assessment: 10, 5, 2.5, 1.25, 0.625, 0.313, 0.156 μL per test

      • For optimization of established antibodies: concentrate around manufacturer's suggestion

    • Prepare antibody dilutions in staining buffer immediately before use

    • Use calibrated pipettes for accuracy at low volumes

  • Application-specific considerations:

    a) For HLA-B*57:01 screening (clinical applications):

    • Prioritize sensitivity and clear population separation

    • Calculate separation index (SI) between positive and negative populations

    • Select concentration that maximizes SI while maintaining distinct population resolution

    • Validate with known HLA-B*57:01 positive and negative controls

    • Typical optimal concentration: 5 μL per million cells in 100 μL staining volume

    b) For quantitative expression analysis (research applications):

    • Prioritize linearity of fluorescence signal with antigen density

    • Use quantitative beads to establish standard curves

    • Select concentration at the upper end of the linear range without saturation

    • Ensure staining is performed at antibody excess to maintain proportionality

    c) For multicolor panel integration:

    • Consider FITC brightness relative to other fluorochromes

    • Titrate in the context of the full antibody panel to account for interactions

    • Evaluate spillover into other channels at each concentration

    • Optimize for balanced signal across all parameters

  • Data analysis for optimal concentration determination:

    • Calculate staining index (SI) for each concentration:
      SI = (MFI positive - MFI negative) / (2 × SD of negative)

    • Plot SI against antibody concentration to identify the inflection point

    • Select the minimum concentration that achieves ≥85% of maximum SI

    • Verify results with replicate experiments using different donor samples

  • Validation and implementation:

    • Test optimal concentration on 5-10 additional samples

    • Document batch-to-batch variation by retitrating new lots

    • Establish acceptable performance ranges for quality control

    • Implement standardized protocols for routine use

This structured titration approach ensures optimal signal detection while maintaining cost-effectiveness. For most FITC-conjugated HLA-B antibodies in flow cytometry applications, studies indicate that the optimal concentration typically falls between 2.5-5 μL per million cells in 100 μL staining volume, but this should be verified for each specific antibody clone and application .

What are the key considerations for designing multicolor flow cytometry panels that include FITC-conjugated HLA-B antibodies?

Designing effective multicolor flow cytometry panels that incorporate FITC-conjugated HLA-B antibodies requires strategic planning to optimize detection sensitivity, minimize spectral overlap, and ensure accurate data interpretation. The following methodological framework addresses key considerations:

  • Spectral characteristics and positioning:

    • FITC has moderate brightness (quantum yield ~0.9) and is excited by the 488nm laser

    • Peak emission occurs at ~520nm with significant spillover into PE and PerCP channels

    • Optimal panel position: Use FITC for abundantly expressed antigens like HLA-B

    • When monitoring HLA-B expression on rare subsets, consider brighter fluorochromes (e.g., PE or APC) for improved detection sensitivity

  • Fluorochrome selection and compatibility:
    The following table outlines compatible fluorochromes to pair with FITC in multicolor panels:

    Laser LineCompatible FluorochromesSpillover ConsiderationsRecommended Pairing Strategy
    488nmPE, PE-Cy5, PE-Cy7Significant FITC→PE spilloverPair FITC with dim PE markers
    405nmPacific Blue, BV421, BV510Minimal spectral overlapExcellent compatibility
    640nmAPC, APC-Cy7, APC-H7Minimal spectral overlapExcellent compatibility
    561nmPE-CF594, PE-Texas RedModerate FITC spilloverRequires careful compensation
  • Panel design hierarchy for HLA-B studies:

    • Backbone markers: Include markers for primary population identification (e.g., CD45, viability dye)

    • Lineage markers: Add markers to identify relevant immune cell populations (e.g., CD3, CD4, CD8, CD19)

    • Functional markers: Include markers relevant to HLA-B research questions (e.g., activation markers, cytokines)

    • HLA-B positioning: Place FITC-conjugated HLA-B antibody strategically based on expression level and research question

  • Antibody panel validation:

    • Perform antibody titration for each marker individually

    • Test for antibody interactions in combined staining

    • Evaluate the impact of fixation/permeabilization on FITC-HLA-B staining if intracellular markers are included

    • Use fluorescence minus one (FMO) controls to set accurate gates

    • Validate compensation settings with single-stained controls

  • Application-specific considerations:

    a) For HLA-B*57:01 screening panels:

    • Simplify panel to essential markers (typically 3-4 colors)

    • Include CD45 to identify lymphocytes

    • Add CD3 and CD19 to distinguish T cells from B cells (both express HLA-B)

    • Consider adding viability dye in non-FITC channel

    • Typical panel: FITC-HLA-B, APC-CD3, Pacific Blue-CD19, PerCP-CD45

    b) For research panels investigating HLA-B-restricted T cell responses:

    • Include CD8 T cell identification markers

    • Add activation markers (e.g., CD69, CD25)

    • Consider including peptide-MHC multimers for antigen-specific cells

    • Cytokine or effector molecule markers may require intracellular staining

  • Technical optimization:

    • Standardize staining protocols (temperature, incubation time, buffer composition)

    • Establish consistent voltage settings for reproducible results

    • Implement daily quality control with fluorescent beads

    • Consider instrument-specific characteristics (laser configuration, filter sets)

This framework has been applied successfully in clinical HLA-B57:01 screening studies, achieving 100% sensitivity for identifying HLA-B57:01-positive individuals when compared to molecular methods, while maintaining clear population separation and minimal interference from other markers in the panel .

What are the most common technical issues in flow cytometry experiments with FITC-conjugated HLA-B antibodies and their solutions?

Flow cytometry experiments with FITC-conjugated HLA-B antibodies may encounter several technical challenges that can compromise data quality and interpretation. The following table outlines common issues, their root causes, and evidence-based solutions:

Technical IssuePotential CausesDiagnostic SignsMethodological Solutions
Poor separation between positive and negative populationsSuboptimal antibody concentration; Antibody degradation; Low HLA-B expressionDiffuse populations; Low staining index; High background1. Perform antibody titration to identify optimal concentration
2. Use fresh antibody aliquots and minimize freeze-thaw cycles
3. Include known positive controls
4. Increase acquisition events (collect >10,000 cells)
High background fluorescenceNonspecific binding; Cell autofluorescence; Fc receptor bindingElevated MFI in negative controls; Population overlap1. Include 2% FBS/BSA in staining buffer
2. Add Fc receptor blocking reagent
3. Implement more stringent washing steps
4. Use appropriate isotype controls to set negative thresholds
Signal loss or degradationPhotobleaching; Prolonged sample storage; Inadequate fixationDecreasing signal over time; Inconsistent results1. Minimize light exposure during processing
2. Analyze samples within 24 hours of staining
3. If fixation is needed, use 0.5-1% paraformaldehyde and store at 4°C
4. Establish consistent time points for acquisition
Indeterminate resultsCell viability issues; Inadequate staining; Pre-analytical variablesUnclear population definition; SI < 1.5; Poor reproducibility1. Implement viability dye to exclude dead cells
2. Standardize sample processing timeframes
3. Use fresh samples when possible
4. Refer indeterminate samples for alternative testing methods
Compensation artifactsImproper compensation; Autofluorescence; Fluorochrome intensity differencesPopulation distortion; False positives/negatives in other channels1. Use single-stained controls for each fluorochrome
2. Apply automated compensation with manual verification
3. Use compensation beads as a supplement to cell controls
4. Consider alternative panel design if problems persist
Inconsistent results between experimentsInstrument variability; Reagent lot changes; Protocol driftShifting positive populations; Varying MFI values1. Implement standardized calibration beads
2. Document PMT voltages and instrument settings
3. Include standard samples across experiments
4. Compare new antibody lots with existing inventory

In HLA-B*57:01 screening applications, research has identified specific technical factors that significantly impact result reliability:

  • Sample viability effects: Studies demonstrate that cell viability below 90% can reduce HLA-B detection sensitivity by up to 15%. Implementing viability dyes and establishing minimum viability thresholds is critical .

  • Cryopreservation impact: Analysis of 577 HIV-1+ patient samples revealed that properly cryopreserved PBMCs maintain HLA-B epitope integrity when standardized freezing/thawing protocols are followed. The study found no significant difference in sensitivity between fresh and properly cryopreserved samples .

  • Antibody specificity considerations: The B-17 monoclonal antibody (mAb3E12) recognizes both HLA-B57 and HLA-B58 alleles, requiring researchers to understand this broader specificity when interpreting positive results. In the Spanish cohort study, only 67.4% of flow cytometry positive samples were confirmed as HLA-B*57:01 by molecular methods .

Implementing these evidence-based solutions has enabled researchers to achieve 100% sensitivity in HLA-B*57:01 screening, making flow cytometry with FITC-conjugated antibodies a reliable initial screening method prior to confirmatory molecular testing .

How can researchers distinguish between true HLA-B expression signals and artifacts in flow cytometry data?

Distinguishing between genuine HLA-B expression signals and technical artifacts requires a systematic analytical approach combined with appropriate controls. The following methodological framework enables reliable discrimination:

  • Control implementation strategy:
    A comprehensive control system is the foundation for accurate data interpretation:

    Control TypePurposeImplementation MethodInterpretation Guide
    Unstained cellsEstablish autofluorescence baselineSame cell preparation without antibodyDefines natural fluorescence boundaries
    Isotype controlEvaluate non-specific bindingMatched isotype FITC-conjugated antibody (typically mouse IgG2a)Sets background threshold; should overlay with unstained cells
    FMO (Fluorescence Minus One)Assess spillover effectsFull panel minus FITC-HLA-B antibodyDefines boundary between positive and negative when compensation is applied
    Known positive sampleVerify antibody performanceCell sample with confirmed HLA-B expressionEstablishes expected positive signal intensity
    Known negative sampleConfirm specificityCell sample lacking target HLA-B alleleConfirms background level in biologically negative cells
    Technical replicatesAssess reproducibilitySame sample stained multiple timesEvaluates technical variability
  • Pattern recognition for common artifacts:
    Certain signal patterns indicate specific technical issues rather than biological expression:

    • Spread distribution: When positive population spreads asymmetrically rather than forms a distinct peak, this indicates compensation artifacts

    • Diagonal smears: In bivariate plots, diagonal patterns suggest improper compensation between FITC and other fluorochromes

    • Time-dependent shifts: Signal decreasing during acquisition indicates photobleaching

    • Bimodal distribution in controls: May indicate antibody aggregation or non-specific binding

  • Analytical verification techniques:

    a) Statistical approaches:

    • Calculate separation index (SI) between positive and negative populations:
      SI = (MFI positive - MFI negative) / (2 × √(variance positive + variance negative))

    • Values >2.0 indicate reliable separation; <1.5 suggest potential artifacts

    b) Visual assessment methods:

    • Examine population distributions in multiple projections (histogram, density plots)

    • Verify population boundaries remain consistent across different display methods

    • Assess if positive signals correlate with expected biological parameters

    c) Replication and validation:

    • Compare results with alternative detection methods

    • Evaluate consistency across technical and biological replicates

    • Apply standardized gating strategies across experiments

  • Artifact identification and correction framework:

    a) Dead cell artifacts:
    Research demonstrates dead cells can bind antibodies non-specifically, creating false positives. Implement viability dye exclusion in a non-FITC channel to remove this artifact .

    b) Doublet contamination:
    Cell aggregates can present as false positives with artificially high MFI. Apply FSC-H vs. FSC-A gating to exclude doublets .

    c) Compensation artifacts:
    Improper compensation can distort FITC signal distribution. Verify compensation with visualization tools and manual adjustments when necessary .

    d) Fixation-induced artifacts:
    Fixation can alter epitope conformation and fluorochrome properties. If fixation is required, validate with parallel fixed/unfixed samples and adjust gating accordingly .

In clinical applications like HLA-B*57:01 screening, where decisions impact patient care, researchers should implement a tiered interpretation approach:

  • Clear positives (SI >2.0): Proceed to confirmatory testing

  • Clear negatives (overlay with negative controls): Report as negative

  • Indeterminate results (SI 1.5-2.0): Retest with fresh sample or proceed directly to molecular methods

This structured approach enabled researchers to achieve 100% sensitivity with no false negatives in a 577-patient HIV cohort study using FITC-conjugated mAb3E12 .

What statistical methods and analytical approaches are recommended for quantifying HLA-B expression levels from flow cytometry experiments?

Quantifying HLA-B expression levels from flow cytometry experiments requires rigorous statistical methods and analytical approaches to ensure data accuracy, reproducibility, and meaningful biological interpretation. The following framework provides a comprehensive methodology:

  • Fluorescence intensity measurement approaches:

    a) Relative quantification methods:

    • Median Fluorescence Intensity (MFI): Preferred over mean due to resistance to outliers

    • Staining Index (SI): (MFI positive - MFI negative) / (2 × SD of negative)

    • T/N ratio: Ratio of sample MFI to isotype control MFI

    • Percent positive: Requires careful gating based on controls

    b) Absolute quantification methods:

    • Molecules of Equivalent Soluble Fluorochrome (MESF): Converts fluorescence to standardized units

    • Antibody Binding Capacity (ABC): Estimates actual number of antibodies bound per cell

    • Quantitative Flow Cytometry (QFCM): Uses calibration beads with known numbers of fluorochromes

  • Statistical analysis framework for HLA-B expression:

    a) Descriptive statistics:

    • Report medians with interquartile ranges rather than means with standard deviations

    • Use coefficient of variation (CV) to assess measurement precision

    • Apply 95% confidence intervals for population estimates

    b) Comparative statistics:

    • Non-parametric tests (Mann-Whitney, Kruskal-Wallis) for non-normally distributed data

    • ANOVA with post-hoc tests for multiple group comparisons if normality is confirmed

    • Mixed-effects models for longitudinal data with repeated measures

    c) Correlation analysis:

    • Spearman's rank correlation for association with other parameters

    • Pearson correlation only if linear relationship and normality are confirmed

    • Regression analysis to identify predictors of expression levels

  • Advanced analytical approaches:

    Analytical MethodApplicationAdvantagesLimitations
    Standardized MFI (sMFI)Comparison between experimentsNormalizes to reference standardsRequires consistent standards
    Probability binningIdentifying subtle population shiftsObjective, automated detectionComputationally intensive
    Clustering algorithmsIdentifying HLA-B expression patternsUnbiased population identificationRequires optimization of parameters
    Machine learning classificationHLA typing applicationsHigh sensitivity and specificityRequires large training datasets
    Visualization tools (t-SNE, UMAP)Multiparameter analysisReveals complex relationshipsDimensional reduction may obscure some relationships
  • Methodological considerations for accurate quantification:

    a) Standardization requirements:

    • Use calibration beads to standardize fluorescence scales

    • Establish consistent PMT voltage settings between experiments

    • Include standard samples across experimental batches

    • Apply standard curves for converting to absolute units

    b) Batch effect correction:

    • Implement batch normalization algorithms

    • Use reference samples as internal controls

    • Apply statistical methods (e.g., ComBat) for batch correction

    • Document and report batch-specific variables

  • Reporting standards for reproducibility:

    • Document complete instrument configuration and settings

    • Report all preprocessing steps and transformations

    • Provide detailed gating strategy with justification

    • Include sample sizes, technical replicates, and statistical power calculations

    • Specify software and algorithms used for analysis with version numbers

Implementation of these methodological approaches has proven effective in quantitative HLA-B expression studies. For example, in the Spanish cohort study of HLA-B*57:01 screening, researchers successfully applied standardized MFI analysis with calculation of separation indices to achieve 100% sensitivity and clear discrimination between positive and negative populations . For research applications requiring more precise quantification, ABC determination using calibration beads provides superior resolution of expression differences between sample groups and correlates more directly with biological function.

How can FITC-conjugated HLA-B antibodies be integrated into single-cell technologies for immunophenotyping and functional studies?

Integrating FITC-conjugated HLA-B antibodies into single-cell technologies enables sophisticated immunophenotyping and functional characterization at unprecedented resolution. The following methodological framework outlines approaches for various single-cell platforms:

  • Integration with mass cytometry (CyTOF):
    While CyTOF typically uses metal-conjugated antibodies, FITC-conjugated HLA-B antibodies can be incorporated indirectly:

    • Metal labeling strategy: Use anti-FITC metal-conjugated secondary antibodies to detect FITC-HLA-B

    • Sequential staining protocol:

      1. Stain cells with FITC-conjugated HLA-B antibody

      2. Wash thoroughly to remove unbound antibody

      3. Apply metal-tagged anti-FITC secondary antibody

      4. Include additional metal-conjugated antibodies for comprehensive phenotyping

    • Methodological advantages: Enables simultaneous detection of >40 parameters including HLA-B

    • Research applications: Identification of HLA-B-restricted T cell subsets in heterogeneous samples

  • Single-cell RNA-sequencing with protein detection (CITE-seq):
    CITE-seq allows simultaneous measurement of transcriptome and protein expression:

    • Antibody modification requirements: FITC-HLA-B antibodies must be further conjugated with oligonucleotide barcodes

    • Protocol adaptation:

      1. Convert FITC-conjugated HLA-B antibodies to CITE-seq compatible reagents using commercial kits

      2. Optimize concentration through titration (typically 1:10 to 1:100 dilution of flow cytometry concentration)

      3. Include in antibody cocktail with other oligo-tagged antibodies

      4. Process using standard CITE-seq workflows

    • Analytical approach: Correlate HLA-B protein expression with gene expression patterns

    • Unique insights: Identify transcriptional signatures associated with specific HLA-B alleles

  • Spectral flow cytometry applications:
    Spectral flow cytometry offers advantages for complex panels including FITC-HLA-B antibodies:

    • Panel design considerations:

      • Utilize full emission spectrum rather than single PMT detection

      • Combine with fluorochromes that would be challenging in conventional flow cytometry

      • Expand beyond 30 parameters simultaneously

    • Optimization requirements:

      • Create spectral libraries that include FITC-HLA-B antibody alone

      • Perform unmixing algorithm optimization

      • Validate against conventional flow cytometry results

    • Research applications: Complex phenotyping of HLA-B-restricted T cell responses with minimal spillover issues

  • Imaging flow cytometry integration:
    Combining HLA-B detection with cellular localization provides insights into antigen presentation:

    • Protocol modifications:

      • Optimize FITC-HLA-B antibody concentration (typically 2× flow cytometry concentration)

      • Include membrane and nuclear markers for spatial reference

      • Validate signal-to-noise ratio at multiple magnifications

    • Analysis parameters:

      • Quantify HLA-B localization relative to cell membrane

      • Measure co-localization with other proteins of interest

      • Assess morphological features associated with HLA-B expression patterns

    • Research applications: Studying HLA-B trafficking and distribution during viral infection or tumor immune responses

  • Microfluidic single-cell functional assays:
    Microfluidic platforms enable detection of functional outputs from individual cells:

    • Integration approaches:

      • Combine FITC-HLA-B phenotyping with cytokine secretion assays

      • Integrate with T cell receptor signaling reporters

      • Link with cell proliferation or cytotoxicity assays

    • Technical considerations:

      • Optimize antibody concentration for confined microfluidic channels

      • Verify photobleaching rates in time-lapse experiments

      • Establish compensation protocols specific to microfluidic constraints

    • Research applications: Correlating HLA-B expression with functional capacity at single-cell resolution

The methodological flexibility of FITC-conjugated HLA-B antibodies makes them adaptable across these advanced platforms, though each requires specific optimization. In HIV immunology research, these integrated approaches have revealed how HLA-B*57:01 expression correlates with specific CD8+ T cell functional signatures associated with viral control, demonstrating the value of multimodal single-cell analysis incorporating HLA-B detection .

What are the current challenges and methodological advances in studying HLA-B-restricted T cell responses using FITC-conjugated antibodies?

Studying HLA-B-restricted T cell responses using FITC-conjugated antibodies presents several challenges while benefiting from recent methodological advances. This complex research area continues to evolve with innovative approaches to overcome technical limitations:

  • Challenge: Limited detection of antigen-specific T cells

    Current limitations:

    • FITC-conjugated HLA-B antibodies detect total HLA-B expression but not peptide-specific complexes

    • Low frequency of antigen-specific T cells (typically 0.01-0.1% of CD8+ T cells)

    • Conventional approaches lack functional characterization

    Methodological advances:

    • Peptide-MHC multimer technology: Combine FITC-HLA-B phenotyping with peptide-MHC multimers in complementary channels

    • Activation-induced marker assays: Detect antigen-responsive T cells via upregulation of CD137, CD69, and other activation markers alongside HLA-B typing

    • Cytokine capture technology: Link cytokine production to HLA-B expression at single-cell level

  • Challenge: Allelic variation and specificity

    Current limitations:

    • Most FITC-conjugated HLA-B antibodies recognize multiple alleles (e.g., mAb3E12 binds both HLA-B57 and HLA-B58)

    • Difficulty in discriminating closely related alleles with functional differences

    • Variable expression levels between alleles complicate interpretation

    Methodological advances:

    • Allele-specific monoclonal antibodies: Development of highly specific antibodies against polymorphic regions

    • Sequential staining strategies: Competitive binding approaches to distinguish closely related alleles

    • Computational deconvolution: Algorithms to resolve mixed HLA-B signals based on expression patterns

  • Challenge: Studying low-expression variants and conformational changes

    Current limitations:

    • FITC brightness limitations for detecting low expression variants

    • Inability to distinguish empty vs. peptide-loaded HLA-B molecules

    • Conformational epitopes may be affected by certain peptide interactions

    Methodological advances:

    • Signal amplification systems: Tyramide signal amplification and other enhancement methods for low-abundance detection

    • Conformation-specific antibodies: Development of reagents that distinguish peptide-loaded vs. empty HLA-B molecules

    • Super-resolution microscopy: Nanoscale visualization of HLA-B clustering and organization

  • Challenge: Functional correlation with expression levels

    Current limitations:

    • Difficulty linking HLA-B expression levels to functional T cell responses

    • Inter-individual variations in surface expression complicate standardization

    • Expression dynamics change during immune responses

    Methodological advances:

    ApproachMethodologyAdvantagesResearch Applications
    Multiparameter functional profilingCombine FITC-HLA-B detection with cytokine, degranulation, and transcription factor markersCorrelates expression with multiple functional outputsCharacterizing T cell polyfunctionality in HLA-B-restricted responses
    Live-cell imagingTime-lapse fluorescence microscopy with FITC-HLA-B and functional reportersCaptures dynamic interactions in real timeStudying immunological synapse formation
    Standardized quantificationCalibration with ABC (Antibody Binding Capacity) beadsEnables absolute quantification of HLA-B moleculesCross-study comparisons of expression thresholds
  • Challenge: Technical variability in longitudinal studies

    Current limitations:

    • FITC photobleaching affects long-term monitoring

    • Batch effects between antibody lots

    • Instrument variability over time

    Methodological advances:

    • Stabilized FITC derivatives: More photostable fluorophores with similar spectral properties

    • Internal standards: Inclusion of calibration beads and standard samples across batches

    • Normalized reporting: Development of standardized MFI (sMFI) approaches for cross-experiment comparison

These methodological advances are enabling more sophisticated research applications. For example, studies of HIV-specific immune responses have combined FITC-conjugated HLA-B antibodies with peptide-MHC tetramers to correlate HLA-B57:01 expression levels with the magnitude and functionality of virus-specific CD8+ T cell responses. This approach has revealed that higher surface expression of HLA-B57:01 correlates with enhanced recognition of Gag epitopes by CD8+ T cells, providing mechanistic insights into the protective effects of certain HLA-B alleles in HIV infection .

How are FITC-conjugated HLA-B antibodies being applied in translational research for personalized medicine approaches?

FITC-conjugated HLA-B antibodies are increasingly instrumental in translational research for personalized medicine, bridging laboratory discoveries with clinical applications. The following applications represent cutting-edge methodological approaches in this rapidly evolving field:

These translational applications demonstrate how FITC-conjugated HLA-B antibodies bridge research and clinical practice. The rapid flow cytometric screening approach for HLA-B*57:01 exemplifies this transition, with implementation studies demonstrating cost-effectiveness, workflow efficiency, and excellent clinical performance in real-world settings. The study of 577 HIV-1+ individuals validated this approach with 100% sensitivity compared to molecular methods, establishing a clinically viable methodology that balances accuracy, turnaround time, and resource utilization . As personalized medicine continues to evolve, these methodological approaches will expand to additional HLA-B-associated conditions, further enhancing the clinical impact of HLA-B research.

Key Methodological Insights

The integration of FITC-conjugated HLA-B antibodies into research and clinical workflows represents a significant advancement in immunological techniques. These reagents enable rapid, sensitive detection of HLA-B expression with applications ranging from pharmacogenetic screening to complex immunophenotyping. The evidence-based methodologies detailed in this FAQ collection demonstrate how researchers can optimize these tools for various applications while addressing common technical challenges .

Flow cytometry with FITC-conjugated HLA-B antibodies has proven particularly valuable as a first-line screening approach for HLA-B*57:01, achieving 100% sensitivity with significant workflow and cost advantages over universal molecular testing. While confirmatory testing remains necessary for positive results due to cross-reactivity with related alleles, the ability to rapidly classify over 90% of samples as negative has substantial practical implications for clinical implementation .

Future Directions

Emerging single-cell technologies and advanced analytical approaches are expanding the utility of HLA-B detection in research settings. The integration of FITC-conjugated antibodies with spectral cytometry, imaging platforms, and multi-omics approaches promises deeper insights into the functional significance of HLA-B expression in health and disease. As methodologies continue to evolve, the balance between assay simplicity, cost-effectiveness, and information depth will remain an important consideration for researchers selecting the optimal approach for their specific applications .

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.