MHC Class I, H-2K Antibody, FITC

MHC Class I, (H-2K) Mouse Antibody, FITC
Shipped with Ice Packs
In Stock

Description

Structure and Specificity

MHC class I molecules are heterodimers composed of a polymorphic α chain noncovalently linked to β2-microglobulin. The H-2K Antibody, FITC specifically binds to the α3 domain of the H-2Kb haplotype in mice .

Flow Cytometry

  • Detection Sensitivity: Optimized for staining 10⁵–10⁸ cells/test in 100 µL volume .

  • Protocol: Use ≤1 µg antibody per test; titrate for optimal signal-to-noise ratios .

Immunological Research

  • Antigen Presentation: Facilitates studies on CD8+ T cell activation via MHC class I-peptide complexes .

  • In Vivo Models: Administration of anti-H-2Kb antibodies in murine cardiac allograft models induces antibody-mediated rejection (AMR) with microvascular damage and C4d deposition .

Antibody-Mediated Rejection (AMR)

  • Mechanism: Anti-MHC class I antibodies trigger phosphorylation of Akt, mTOR, and S6K, promoting endothelial survival and proliferation pathways .

  • Pathology: Cardiac allografts treated with anti-H-2Kd antibodies show intravascular macrophage accumulation and complement activation .

Antigenic Modulation

  • PdCl₂ Exposure: Reduces MHC class I/peptide complex stability, as shown by diminished 25D1.16 antibody binding to H-2Kb/SIINFEKL complexes in murine splenocytes .

Comparative Analysis of Clones

FeatureAF6-88.5.5.3 Clone (H-2Kb)34-1-2S Clone (H-2Kd/Dd)
Cross-ReactivityNoneH-2Kb, s, r, q, p
ImmunogenC57BL/6 mouse splenocytes BDF1 mouse splenocytes
Key ApplicationsIn vivo T cell depletion, flow cytometryBlocking T cell adhesion, immunoprecipitation

References in Peer-Reviewed Studies

  1. AMR Pathogenesis: Passive transfer of anti-H-2Kd antibodies in B6.RAG1 KO mice replicates human AMR histopathology .

  2. Therapeutic Modulation: Anti-MHC class I antibodies block CD8+ T cell priming against viral peptides, suggesting therapeutic potential .

  3. Antigenic Peptide Loading: PdCl₂ alters MHC class I-peptide complexes, reducing antigenicity .

Product Specs

Introduction
MHC Class I is a transmembrane protein complex crucial for the adaptive immune response. It comprises two polypeptide chains: a larger alpha chain and a smaller beta-2 microglobulin. The alpha chain folds into three domains (Alpha-1, Alpha-2, Alpha-3) with the Alpha-3 domain anchoring the complex to the cell membrane. The peptide-binding groove, situated between the Alpha-1 and Alpha-2 domains, accommodates short peptide fragments derived from intracellular proteins. This peptide-MHC complex is presented on the cell surface for surveillance by cytotoxic T lymphocytes (CTLs). The diverse repertoire of peptides presented by MHC Class I molecules ensures broad antigen recognition. The interaction between the peptide-MHC complex and the T cell receptor (TCR) on CTLs is characterized by a balance between specificity and affinity, essential for effective immune surveillance.
Formulation
Supplied as a 1 mg/ml solution in phosphate-buffered saline (PBS).
Storage Procedures
The lyophilized antibody should be stored at 4 degrees Celsius. After reconstitution, it is recommended to use the antibody promptly. For long-term storage, aliquot the reconstituted antibody and store at -20 degrees Celsius to preserve its activity.
Solubility
To reconstitute the lyophilized antibody, add deionized water. Gently mix the solution to ensure complete dissolution. Briefly centrifuge the vial to collect any droplets on the sides. Allow the reconstituted antibody to stand at room temperature for 30-60 seconds before use.
Applications
This antibody has applications in cytotoxicity assays and cell staining. For staining applications, use 10 microliters of antibody per 1 million cells. The appropriate titer for cytotoxicity assays needs to be empirically determined by the researcher.
Available Conjugates
In addition to the FITC-conjugated version, this antibody is also available in unconjugated and biotinylated formats.
Purification Method
Ion exchange column.
Specificity
Recognizes most mouse H-2K haplotypes (k,p,q,r,s). Does NOT react with H-2kd.
Type
Mouse Antibody Monoclonal.
Clone
NYRmH-2K.
Immunogen
Purified mouse LN cells (C57Bl/6 anti-BALB/c) .
Ig Subclass
Mouse IgG2b.

Q&A

What is MHC Class I H-2K and what role does it play in immune response?

MHC Class I H-2K molecules are cell surface glycoproteins involved in antigen presentation to T cells expressing CD3/TCR and CD8. They present peptide antigens from intracellular proteins to CD8+ T cells, playing a critical role in the body's defense against intracellular pathogens and malignant transformation. The H-2K locus in mice produces multiple alloantigens (like H-2Kb and H-2Kd) that are strain-specific and homologous to human HLA class I molecules . MHC Class I molecules consist of a heavy chain with three domains (α1, α2, and α3) and associate with β2-microglobulin. The α1 and α2 domains form the peptide-binding groove, while the α3 domain interacts with CD8 on T cells .

How do different H-2K antibody clones vary in their specificity and application?

H-2K antibody clones demonstrate distinct specificity profiles based on their epitope recognition and reactivity patterns:

  • The AF6-88.5.5.3 clone specifically reacts with the H-2Kb MHC class I alloantigen and shows no cross-reactivity with other haplotypes (d, f, j, k, p, q, r, s, u, and v) .

  • The SF1-1.1 antibody clone binds to the α3 domain of the H-2K[d] MHC class I alloantigen and similarly shows no reactivity with other haplotypes like b, j, k, p, q, s, and v .

These specificity differences make clone selection critical for experimental design, especially when working with different mouse strains. For instance, the AF6-88.5.5.3 clone is appropriate for C57BL/6 mice (H-2Kb haplotype), while the SF1-1.1 clone should be used with BALB/c mice (H-2Kd haplotype) .

What considerations should researchers make when using FITC-conjugated MHC Class I antibodies?

When using FITC-conjugated MHC Class I antibodies, researchers should consider several technical aspects:

  • Fluorescence properties: FITC has an excitation maximum around 488 nm and emission maximum around 520 nm, requiring use with a blue laser in flow cytometry .

  • Storage conditions: These antibodies should be stored at 4°C protected from light exposure, and should not be frozen to maintain conjugate stability .

  • Buffer compatibility: The antibody formulation contains protein stabilizers and ≤0.09% sodium azide, which may interfere with certain fixation protocols or enzymatic assays .

  • Signal strength: FITC is susceptible to photobleaching and has relatively lower brightness compared to newer fluorochromes, potentially affecting detection of low-abundance antigens.

  • Titration requirement: Optimal antibody concentration must be determined empirically, with recommendations starting at ≤1 μg per test (defined as the amount needed to stain a sample of 10^5 to 10^8 cells in 100 μL) .

How does MHC Class I expression regulation impact cancer immunobiology?

MHC Class I expression regulation plays a multifaceted role in cancer immunobiology through several mechanisms:

  • Immune evasion: Deranged expression of MHC class I glycoproteins is characteristic of various malignancies and contributes to cancer's ability to avoid T cell-mediated immune destruction. Studies with B16 melanoma models demonstrate that MHC class I-deficient clones show enhanced metastatic capacity compared to MHC class I-expressing clones .

  • Immunocyte death induction: Class I-deficient melanoma cells (BL9 and BL12 clones) demonstrate greater potency in inducing apoptosis in splenic lymphocytes compared to class I-expressing (CL8-2) clones. This suggests that absence of MHC class I glycoproteins helps malignant cells prevent the elimination of potential effector immunocytes .

  • Non-FAS-mediated mechanisms: Research indicates that production of FAS-ligand may not be the only mechanism by which malignant cells induce apoptosis in immunocytes. MHC class I-deficient tumors appear to utilize alternative pathways to induce T cell death .

  • Differential impact on T cell subsets: MHC class I deficiency affects CD4+ and CD8+ T cell populations differently in the tumor microenvironment, with proportions of these cells being lower when exposed to class I-deficient tumor clones .

These findings suggest that therapeutic strategies targeting MHC class I expression restoration in tumors may enhance anti-tumor immunity through multiple mechanisms beyond simple antigen presentation.

What mechanisms underlie palladium-induced MHC Class I internalization and how does this affect antigen presentation?

Palladium-induced MHC Class I internalization represents a significant mechanism affecting antigen presentation dynamics:

Palladium treatment (PdCl2) causes a temporal internalization of MHC Class I molecules, which critically influences antigenicity generation. In experimental models, mice injected with ovalbumin protein followed by PdCl2 treatment showed reduced binding of the 25D1.16 antibody (which recognizes MHC class I H-2Kb loading the SIINFEKL peptide from ovalbumin) compared to mice receiving ovalbumin alone .

This internalization process affects antigen presentation through differential peptide loading on MHC class I molecules. Notably, while the binding of SIINFEKL-loaded H-2Kb complexes decreased with PdCl2 treatment, the total H-2Kb levels remained comparable regardless of treatment, suggesting selective effects on specific peptide-MHC combinations rather than global MHC reduction .

This mechanism provides insights into how environmental factors and chemical exposures might modulate immune responses by altering the peptide repertoire presented by MHC class I molecules, with potential implications for understanding metal hypersensitivity, autoimmunity, and tumor immunosurveillance.

How can researchers optimize flow cytometry protocols for detecting subtle changes in MHC Class I expression?

Optimizing flow cytometry protocols for detecting subtle changes in MHC Class I expression requires several methodological refinements:

  • Antibody titration: Perform comprehensive titration experiments with each MHC Class I antibody clone to determine the optimal concentration that provides maximum signal-to-noise ratio. The optimal concentration is typically ≤1 μg per test (10^5-10^8 cells in 100 μL), but must be determined empirically for each application .

  • Surface marker preservation: Carefully select cell isolation and preparation methods that preserve surface MHC expression. Avoid harsh enzymatic digestion methods that might cleave surface proteins.

  • Appropriate controls:

    • Include isotype controls matched for immunoglobulin class, concentration, and fluorophore (e.g., FITC Mouse IgG2a, κ Isotype Control for H-2Kd antibodies) .

    • Include biological reference controls (e.g., cells known to express high or low levels of the target).

    • Consider fluorescence-minus-one (FMO) controls for multicolor panels.

  • Buffer optimization:

    • Use specialized staining buffers containing protein (BSA or FBS) to reduce non-specific binding (e.g., Stain Buffer BSA or Stain Buffer FBS) .

    • Maintain consistent buffer composition throughout all experimental conditions.

  • Data analysis approaches:

    • Analyze data using both percentage of positive cells and mean/median fluorescence intensity.

    • Consider using standardized metrics like Molecules of Equivalent Soluble Fluorochrome (MESF) or antibody binding capacity (ABC) for quantitative comparisons across experiments.

    • Apply appropriate statistical tests sensitive to small differences in expression.

Implementation of these optimization strategies enables detection of subtle changes in MHC Class I expression that might be biologically significant in contexts such as early malignant transformation or initial responses to immunomodulatory treatments.

What are the critical sample preparation steps for optimal MHC Class I detection?

Critical sample preparation steps for optimal MHC Class I detection include:

  • Cell isolation: Gentle isolation techniques that preserve membrane integrity are essential. For splenic lymphocytes, mechanical dissociation methods are preferred over enzymatic digestion to avoid cleaving surface MHC molecules .

  • Viability maintenance: Process samples rapidly and maintain them at 4°C throughout preparation to minimize internalization of surface MHC molecules and preserve epitope integrity.

  • Red blood cell lysis: When preparing splenocytes, use ammonium chloride-based RBC lysis buffers rather than hypotonic solutions to better preserve lymphocyte membrane proteins .

  • Cell concentration standardization: Adjust cell concentrations to 10^5-10^8 cells per 100 μL test volume, with optimal density being application-specific and requiring empirical determination .

  • Blocking procedure: Incorporate blocking of Fc receptors using anti-CD16/CD32 antibodies prior to staining to minimize non-specific binding, particularly important when analyzing myeloid cell populations.

  • Washing protocol: Use multiple gentle washing steps with cold buffer containing protein stabilizers and low concentration sodium azide (≤0.09%) to reduce background while preserving antibody binding .

  • Fixation considerations: If fixation is necessary, use paraformaldehyde at low concentrations (0.5-1%) as higher concentrations may alter MHC epitopes. Validate that fixation doesn't affect antibody binding to the specific epitope being studied .

Consistent application of these preparation steps across experimental conditions is critical for generating reproducible and comparable results in MHC Class I expression studies.

How should researchers troubleshoot inconsistent MHC Class I staining results?

When encountering inconsistent MHC Class I staining results, researchers should systematically evaluate and troubleshoot using this approach:

  • Antibody validation:

    • Verify antibody specificity using positive and negative control samples (appropriate haplotype strains).

    • Check for antibody degradation by comparing with new lots/vials.

    • Confirm the antibody clone is appropriate for the mouse strain being studied (e.g., AF6-88.5.5.3 for H-2Kb vs. SF1-1.1 for H-2Kd) .

  • Technical variables assessment:

    • Review staining temperature and duration (maintain at 4°C to prevent internalization).

    • Evaluate buffer composition (protein content and pH can affect binding).

    • Assess flow cytometer performance using standardization beads.

  • Biological variables investigation:

    • Consider cell activation status (activated cells may modulate MHC expression).

    • Check for treatments that might induce MHC internalization (like PdCl2) .

    • Evaluate sample viability (dead/dying cells often show altered autofluorescence and non-specific binding).

  • Systematic optimization:

    • Perform side-by-side comparisons of varied protocols.

    • Create a standardized checklist for sample preparation.

    • Document all procedural details to identify inconsistency sources.

  • Common pitfalls to address:

    • Over-fixation may mask epitopes.

    • Cell clumping can create false negative populations.

    • Excessive compensation in multicolor panels may distort FITC signals.

    • Photobleaching of FITC during extended light exposure.

By systematically addressing these aspects, researchers can identify the source of inconsistencies and establish reliable protocols for MHC Class I detection across experiments.

What experimental controls are essential when analyzing MHC Class I expression in different treatment conditions?

Essential experimental controls for analyzing MHC Class I expression across different treatment conditions include:

  • Technical controls:

    • Isotype control: Include a FITC-conjugated isotype-matched control antibody (e.g., FITC Mouse IgG2a, κ) at the same concentration as the experimental antibody to establish background staining thresholds .

    • Single-color controls: For multicolor panels, include single-stained samples for each fluorochrome to establish proper compensation.

    • Unstained controls: Include completely unstained samples to establish autofluorescence levels.

  • Biological reference controls:

    • Haplotype controls: Include cell samples from mouse strains with known positive and negative expression of the specific MHC haplotype (e.g., C57BL/6 for H-2Kb or BALB/c for H-2Kd) .

    • Negative biological controls: Include cell types known to express minimal levels of MHC Class I for background reference.

    • Positive biological controls: Include samples known to express high levels of the specific MHC Class I molecule.

  • Treatment-specific controls:

    • Vehicle controls: Samples treated with all vehicles/solvents used in experimental conditions without the active compound.

    • Time-matched controls: Samples processed at identical time points to control for time-dependent changes in MHC expression.

    • Dose-response controls: Include multiple concentrations of treatment agents to establish dose-dependency relationships (e.g., titration series of PdCl2) .

  • Experiment validation controls:

    • Biological response controls: Include conditions known to modulate MHC expression (e.g., IFN-γ treatment to upregulate MHC Class I).

    • Cross-validation controls: Analyze MHC expression using alternative techniques (e.g., Western blot, qPCR) to confirm flow cytometry results.

Implementation of this comprehensive control strategy ensures reliable interpretation of treatment effects on MHC Class I expression by distinguishing specific biological responses from technical artifacts or non-specific effects.

How can researchers quantitatively compare MHC Class I expression levels across different experimental conditions?

Researchers can employ several quantitative approaches to compare MHC Class I expression levels across experimental conditions:

  • Fluorescence intensity metrics:

    • Mean Fluorescence Intensity (MFI): Calculate the arithmetic mean of fluorescence intensity for the positive population.

    • Median Fluorescence Intensity: Often more robust than mean for non-normally distributed data.

    • Geometric Mean: Appropriate for log-normally distributed flow cytometry data.

    • Integrated MFI: Product of percent positive cells and MFI, useful when both parameters change.

  • Standardization methods:

    • Molecules of Equivalent Soluble Fluorochrome (MESF): Convert arbitrary fluorescence units to standardized values using calibration beads.

    • Antibody Binding Capacity (ABC): Determine the number of antibody binding sites per cell.

    • Normalization to reference populations: Express results as fold-change relative to control samples.

  • Population analysis approaches:

    • Percent positive cells: Determine the proportion of cells above a threshold set using isotype controls.

    • Coefficient of Variation (CV): Measure the spread of expression within a population.

    • Subpopulation definition: Use clustering algorithms to identify distinct expression profiles.

  • Statistical analysis considerations:

    • For comparing two conditions: Paired t-tests (for matched samples) or Mann-Whitney U tests (for non-parametric data).

    • For multiple conditions: ANOVA with appropriate post-hoc tests (Tukey, Dunnett, etc.) or Kruskal-Wallis tests.

    • For complex experimental designs: Consider mixed-effects models to account for batch effects and repeated measures.

  • Visualization techniques:

    • Histogram overlays: Direct visual comparison of fluorescence distribution shifts.

    • Contour plots: Useful for analyzing co-expression with other markers.

    • Heat maps: Effective for visualizing expression patterns across multiple conditions.

Implementation of these approaches enables robust quantitative comparison of MHC Class I expression across experimental conditions while accounting for technical variability and biological heterogeneity .

How do changes in MHC Class I expression correlate with functional outcomes in immune response studies?

Changes in MHC Class I expression correlate with several functional outcomes in immune response studies:

  • Cytotoxic T lymphocyte (CTL) recognition:

    • Reduced MHC Class I expression correlates with decreased recognition by CD8+ T cells, as demonstrated in melanoma models where class I-deficient clones showed enhanced metastatic capacity compared to class I-expressing clones .

    • The relationship between MHC expression and CTL killing efficiency is not strictly linear - threshold levels of MHC-peptide complexes are required for effective immune synapse formation.

  • NK cell activity modulation:

    • Decreased MHC Class I expression can enhance susceptibility to NK cell-mediated killing through the "missing self" recognition mechanism.

    • The balance between activating and inhibitory signals in NK cells is influenced by the density of MHC Class I molecules on target cells.

  • Antigen presentation efficiency:

    • Changes in MHC Class I trafficking (such as those induced by palladium treatment) affect the efficiency of antigen presentation to T cells, as demonstrated by reduced binding of the 25D1.16 antibody (recognizing SIINFEKL-H-2Kb complexes) following PdCl2 treatment .

    • Alterations in MHC Class I recycling pathways can change the peptide repertoire presented to T cells.

  • Immunocyte survival:

    • MHC Class I expression levels on tumor cells inversely correlate with their ability to induce apoptosis in splenic lymphocytes, with class I-deficient melanoma cells (BL9 and BL12 clones) showing greater potency in inducing lymphocyte death compared to class I-expressing (CL8-2) clones .

    • This suggests that MHC Class I molecules provide protective signals that prevent elimination of potential effector immunocytes.

  • Correlation with disease progression:

    • In malignancy studies, progressive loss of MHC Class I expression often correlates with increased metastatic potential and poorer prognosis .

    • The pattern of MHC loss (total vs. selective allele loss) provides insights into tumor evolution and immune escape mechanisms.

Understanding these correlations is essential for interpreting the biological significance of observed changes in MHC Class I expression and for developing targeted immunotherapeutic strategies.

What are the methodological approaches for distinguishing between altered MHC Class I expression and modified peptide loading?

Distinguishing between altered MHC Class I expression and modified peptide loading requires specialized methodological approaches:

  • Comparative antibody analysis:

    • Utilize antibodies recognizing different epitopes: Compare staining with antibodies that recognize total MHC Class I (conformationally independent) versus those that recognize only properly folded/peptide-loaded MHC complexes.

    • For mouse H-2K molecules, compare staining patterns between antibodies recognizing structural domains (e.g., SF1-1.1 binding to the α3 domain of H-2Kd) versus conformation-dependent antibodies .

    • Use peptide-specific antibodies: Employ antibodies like 25D1.16 that specifically recognize MHC molecules loaded with particular peptides (e.g., H-2Kb-SIINFEKL complex) .

  • Biochemical approaches:

    • Perform thermal stability assays: Peptide-loaded MHC molecules show greater thermal stability than empty MHC molecules.

    • Conduct pulse-chase experiments with metabolic labeling to track MHC Class I biogenesis, peptide loading, and trafficking.

    • Use peptide stripping and reloading assays to assess MHC occupancy with endogenous peptides.

  • Combined phenotypic analysis:

    • Simultaneously analyze components of the peptide-loading complex (TAP, tapasin, calreticulin) alongside MHC Class I.

    • Examine subcellular localization of MHC molecules using imaging flow cytometry or confocal microscopy to detect retention in the ER (suggestive of peptide-loading defects).

  • Functional readouts:

    • Compare direct binding of soluble TCRs to cells under different conditions.

    • Assess T cell activation using reporter cells expressing cognate TCRs for specific MHC-peptide complexes.

    • Analyze peptide repertoire using mass spectrometry to directly identify MHC-bound peptides.

  • Genetic and pharmacological manipulation:

    • Use TAP inhibitors or TAP-deficient model systems as controls for peptide-loading defects.

    • Compare effects of treatments targeting different steps in MHC biosynthesis and peptide loading (e.g., proteasome inhibitors versus treatments affecting MHC trafficking).

This integrated approach revealed, for example, that palladium treatment affects peptide loading rather than total MHC expression, as demonstrated by reduced binding of the peptide-specific antibody 25D1.16 despite unchanged total H-2Kb levels .

How can MHC Class I antibodies be integrated into multi-parameter flow cytometry panels for comprehensive immune profiling?

Integration of MHC Class I antibodies into multi-parameter flow cytometry panels requires strategic planning:

  • Fluorochrome selection considerations:

    • Position FITC-conjugated MHC Class I antibodies appropriately in the panel based on target abundance. Since MHC Class I is typically highly expressed, FITC's moderate brightness is usually sufficient .

    • Consider spectral overlap: FITC has minimal overlap with PE and APC but may have significant overlap with CFSE, GFP, and other green fluorescent proteins.

    • For targets with lower expression, consider brighter fluorochromes (PE, APC) conjugated to MHC Class I antibodies instead of FITC.

  • Panel design strategy:

    • Complement MHC Class I staining with markers for:

      • T cell subsets (CD3, CD4, CD8) to correlate MHC expression with specific T cell interactions.

      • Activation markers (CD69, CD25) to assess functional relationships.

      • Exhaustion markers (PD-1, CTLA-4) in tumor immunology studies.

      • Antigen-presenting cell markers (CD11c, F4/80) when studying presentation dynamics.

    • Include viability dyes in channels with minimal spillover into FITC.

  • Technical optimization:

    • Perform comprehensive titration of all antibodies in the context of the full panel.

    • Establish compensation controls using cells rather than beads when possible.

    • Consider automated compensation and standardization across experiments using reference beads.

  • Analysis approaches:

    • Implement dimensionality reduction techniques (tSNE, UMAP) to visualize MHC Class I expression patterns across multiple cell populations.

    • Use clustering algorithms (FlowSOM, Phenograph) to identify cell subsets with distinct MHC Class I expression profiles.

    • Apply Boolean gating strategies to identify populations with specific combinations of markers.

  • Validation methods:

    • Include fluorescence-minus-one (FMO) controls for accurate gate setting.

    • Use biological controls with known expression patterns to validate panel performance.

    • Consider batch controls across experiments for longitudinal studies.

This strategic integration enables comprehensive characterization of MHC Class I expression in relation to immune cell phenotype and function, critical for understanding complex immunological processes in cancer, infection, and autoimmunity .

What novel insights have emerged from studying MHC Class I expression in cancer immunotherapy resistance?

Several novel insights have emerged from studying MHC Class I expression in cancer immunotherapy resistance:

  • Beyond simple immune evasion:

    • Research reveals that MHC Class I deficiency in cancer cells does more than prevent antigen recognition - it actively contributes to eliminating potential effector immune cells. Studies with B16 melanoma clones demonstrate that class I-deficient cells (BL9 and BL12 clones) are more potent inducers of apoptosis in splenic lymphocytes than class I-expressing cells (CL8-2 clone) .

    • This suggests a dual mechanism where cancer cells both hide from and actively suppress anti-tumor immunity through MHC Class I regulation.

  • Non-classical death pathways:

    • The mechanisms by which MHC Class I-deficient tumor cells induce immunocyte death appear to be independent of the classical FAS/FAS-L pathway. Research confirms that melanoma clones studied did not produce FAS-ligand, indicating alternative pathways for inducing lymphocyte apoptosis .

    • This insight points to novel targetable pathways for preventing tumor-induced immune suppression.

  • Differential impact on immune cell subsets:

    • MHC Class I expression status on tumor cells differentially affects immune cell subpopulations. When exposed to class I-deficient melanoma cells, the proportion of CD4+ and CD8+ cells among splenocytes was lower than when exposed to class I-expressing cells .

    • This finding suggests complex interactions between tumor MHC expression and the composition of the tumor immune microenvironment.

  • Mechanisms of MHC Class I downregulation:

    • Beyond genetic loss, research has identified dynamic mechanisms of MHC regulation, including internalization induced by environmental factors like palladium compounds .

    • This reveals how exposure to certain substances might modulate anti-tumor immunity through effects on MHC presentation.

  • Implications for immunotherapy:

    • These findings suggest that strategies solely focused on enhancing T cell activation may be insufficient in tumors with aberrant MHC Class I expression.

    • Combination approaches targeting both MHC Class I restoration and prevention of tumor-induced lymphocyte death may be required for optimal therapeutic efficacy.

These insights collectively reframe our understanding of the role of MHC Class I in tumor-immune interactions, suggesting more complex mechanisms than previously appreciated and pointing to new therapeutic opportunities .

How can researchers leverage MHC Class I antibodies for studying antigen presentation dynamics in real-time?

Researchers can leverage MHC Class I antibodies for studying antigen presentation dynamics in real-time through several innovative approaches:

  • Live-cell imaging techniques:

    • Utilize conjugated non-blocking MHC Class I antibody fragments (Fab) linked to pH-sensitive fluorophores to track MHC internalization and recycling pathways in live cells.

    • Combine with fluorescently tagged peptides to simultaneously visualize MHC trafficking and peptide loading events.

    • Implement FRET (Förster Resonance Energy Transfer) systems between fluorescently labeled MHC molecules and TCRs to visualize molecular interactions during antigen recognition.

  • Advanced flow cytometry applications:

    • Apply imaging flow cytometry using antibodies that distinguish between total MHC Class I (structure-specific) and properly folded/peptide-loaded MHC complexes (conformation-specific).

    • Implement kinetic flow cytometry to measure rates of MHC internalization and recycling following stimuli (similar to observations with palladium treatment) .

    • Use fluorescent peptide exchange assays combined with MHC antibody staining to monitor peptide loading in real-time.

  • Microfluidic approaches:

    • Develop microfluidic devices with immobilized T cells to detect MHC-peptide complex presentation by measuring calcium flux or other activation markers in response to antigen-presenting cells in real-time.

    • Create gradient systems to study how varying concentrations of inflammatory mediators affect MHC trafficking and peptide loading kinetics.

  • Advanced microscopy methods:

    • Apply super-resolution microscopy (STORM, PALM) with MHC Class I antibodies to visualize nanoscale organization of MHC molecules in the membrane during antigen presentation.

    • Use lattice light-sheet microscopy for long-term, low-phototoxicity imaging of MHC trafficking in 3D cellular environments.

    • Implement correlative light and electron microscopy (CLEM) to connect functional observations of MHC dynamics with ultrastructural context.

  • Reporter systems:

    • Engineer cells with fluorescent protein-tagged MHC molecules combined with surface-specific antibody quenching to distinguish between internalized and surface pools.

    • Develop split-fluorescent protein complementation systems to visualize specific interactions between MHC molecules and components of the antigen processing machinery.

These approaches enable researchers to move beyond static snapshots of MHC expression to understand the dynamic processes that regulate antigen presentation timing, essential for comprehending the kinetics of immune responses in contexts such as viral infection, cancer, and autoimmunity .

Product Science Overview

Introduction

MHC Class I molecules are essential components of the immune system, playing a crucial role in the presentation of intracellular antigens to cytotoxic T cells. The H-2K molecule is a specific type of MHC Class I molecule found in mice. The FITC-conjugated mouse antibody against H-2K is widely used in immunological research to study antigen presentation and immune responses.

MHC Class I Molecules

MHC Class I molecules are glycoproteins expressed on the surface of almost all nucleated cells. They present peptide fragments derived from intracellular proteins to CD8+ T cells. This process is vital for the immune system to recognize and eliminate infected or malignant cells. The MHC Class I molecule consists of a heavy chain, β2-microglobulin, and a peptide-binding groove that accommodates the antigenic peptide.

H-2K Molecule

The H-2K molecule is a specific MHC Class I molecule encoded by the H-2 complex in mice. It is involved in presenting endogenous antigens to CD8+ T cells, triggering an immune response. The H-2K molecule is polymorphic, meaning there are multiple alleles, each encoding a slightly different protein. This polymorphism is crucial for the immune system’s ability to recognize a wide range of antigens.

FITC Conjugation

Fluorescein isothiocyanate (FITC) is a fluorescent dye commonly used to label antibodies. FITC-conjugated antibodies are used in various applications, including flow cytometry and immunofluorescence microscopy, to detect specific antigens. The FITC label allows researchers to visualize and quantify the binding of the antibody to its target antigen.

Mouse Antibody Against H-2K

The mouse antibody against H-2K is a monoclonal antibody that specifically binds to the H-2K molecule. This antibody is used in research to study the expression and function of MHC Class I molecules in mice. The FITC-conjugated version of this antibody enables researchers to use fluorescence-based techniques to analyze H-2K expression.

Applications in Research

The FITC-conjugated mouse antibody against H-2K is used in various research applications, including:

  • Flow Cytometry: To analyze the expression of H-2K on the surface of cells.
  • Immunofluorescence Microscopy: To visualize the localization of H-2K in tissues or cells.
  • Immunohistochemistry: To study the distribution of H-2K in tissue sections.
  • Functional Assays: To investigate the role of H-2K in antigen presentation and immune responses.

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 2024 Thebiotek. All Rights Reserved.