CD80 antibodies are engineered monoclonal antibodies (mAbs) that bind to the CD80 glycoprotein, a member of the B7 immunoglobulin superfamily. CD80 is expressed on antigen-presenting cells (APCs) such as dendritic cells, activated B-cells, and macrophages . Structurally, CD80 comprises:
Extracellular domains: A single IgV-like domain and a single IgC-like domain critical for ligand binding .
Transmembrane region: Anchors the protein to the cell membrane.
Cytoplasmic tail: Short, with regulatory signaling functions .
CD80 antibodies typically target the IgV-like domain to block interactions with CD28 and CTLA-4 on T-cells .
CD80 antibodies modulate immune responses through two primary pathways:
For example, clone MEM-233 binds CD80’s IgV domain, blocking both CD28 and CTLA-4 interactions .
Corneal Transplantation: Anti-CD80/86 antibodies significantly prolonged graft survival in murine models by suppressing IFN-γ-producing CD4+ T-cells and pro-inflammatory cytokines (IL-1β, TNF-α) .
Systemic Lupus Erythematosus (SLE): CD80 upregulation on T-cells correlates with disease activity, making it a therapeutic target .
Lymphoma: Phase I/II trials of IDEC-114 (anti-CD80 mAb) combined with Rituximab showed tolerability in relapsed follicular lymphoma .
Solid Tumors: CD80 fusion proteins (e.g., FPT155, ALPN-202) enhance CD28 signaling while blocking CTLA-4, showing durable anti-tumor responses in early trials .
Mixed Lymphocyte Reaction (MLR): Anti-CD80/86 antibodies reduced T-cell proliferation by 36% and suppressed IFN-γ, IL-2, and TNF-α production .
Tumor Microenvironment: CD80 expression on tumor cells can either enhance (e.g., lung adenocarcinoma) or worsen (e.g., breast cancer) prognosis, influencing therapeutic strategies .
Dual Role in Cancer: CD80’s context-dependent effects complicate therapeutic targeting. For instance, high CD80 in lung adenocarcinoma improves survival but predicts poor outcomes in breast cancer .
Resistance Mechanisms: Tumor cells evade immunity via PD-L1/CD80 interactions, requiring combination therapies (e.g., anti-PD-1 agents) .
CD80 (also known as B7-1, B7, BB1) is a 60 kDa single chain type I glycoprotein belonging to the immunoglobulin superfamily. It plays a critical role in immune regulation through its interactions with receptor proteins on T cells. CD80 is expressed primarily by activated B cells, macrophages, dendritic cells, and can also be expressed by activated T cells .
CD80 has high affinity for binding to two T cell surface antigens: CD28 and CD152 (CTLA-4). The interaction with CD28 provides a costimulatory signal for T cell activation, while its interaction with CTLA-4 provides an inhibitory signal . This dual binding capability makes CD80 crucial in maintaining immune homeostasis through balancing activation and suppression signals.
Recent research has revealed that CD80 also interacts with PD-L1, forming cis-heterodimers on the same cell surface. This interaction can prevent PD-L1 from binding to PD-1 on T cells, thereby promoting T cell activation and enhancing anti-tumor immune responses . This complex network of interactions positions CD80 as a central regulator of adaptive immunity.
Research-grade CD80 antibodies are available in several formats to accommodate different experimental applications:
Monoclonal antibodies: Clones such as 2D10.4 are widely used for their high specificity to human CD80 . These recognize specific epitopes and are useful for applications requiring consistent detection of particular CD80 domains.
Polyclonal antibodies: These recognize multiple epitopes on CD80 (e.g., rabbit polyclonal antibodies) and can provide enhanced sensitivity, particularly in applications like Western blotting .
Conjugated antibodies: These include:
Functional grade antibodies: These are specially purified to remove endotoxins and other contaminants (containing <0.001 ng/μg endotoxin) for use in functional assays where cell viability and physiological responses are essential .
The choice of antibody depends on the specific application, with considerations for species reactivity, application compatibility, and detection strategy.
CD80 and CD86 (B7-2) are both ligands for CD28 and CTLA-4, but they exhibit important differences in expression, binding properties, and function:
| Feature | CD80 (B7-1) | CD86 (B7-2) |
|---|---|---|
| Molecular Weight | 60 kDa | 70 kDa |
| Expression Kinetics | Slower induction, more stable | Rapid induction, more transient |
| Basal Expression | Low/absent on resting APCs | Low but detectable on resting APCs |
| Binding Affinity to CTLA-4 | Higher | Lower |
| Binding Affinity to CD28 | Lower | Higher |
| T cell Response | Often associated with Th1 responses | Often associated with Th2 responses |
CD80 is rapidly induced on activated B cells, Epstein-Barr Virus (EBV) transformed B cell lines, Burkitt's lymphoma cell lines, freshly isolated follicular B lymphoma cells, T cells, and monocytes . Both CD80 and CD86 are essential for T cell activation and can substitute for each other in this process, but their different expression patterns and binding affinities suggest specialized roles in orchestrating immune responses .
Thorough validation of CD80 antibodies is essential for reliable research outcomes. A comprehensive validation approach should include:
Positive and negative controls:
Use cell lines with known CD80 expression (activated B cells, dendritic cells as positive controls; resting T cells as negative controls)
Include CD80 knockout or knockdown cells when available
Test CD80-transfected cells versus non-transfected parent cells
Cross-reactivity testing:
Test against related proteins (particularly CD86, which shares structural similarities)
Validate across relevant species if cross-reactivity is claimed
Functional validation:
Blocking assays: Confirm that the antibody blocks CD80-CD28 or CD80-CTLA-4 interactions in mixed lymphocyte reactions
T cell activation assays: Verify that the antibody affects T cell proliferation or cytokine production in co-culture experiments
Application-specific validation:
For flow cytometry: Compare with isotype controls and determine the optimal titration (typically ≤1 μg per test)
For Western blot: Confirm band size (approximately 60 kDa for glycosylated form, 33 kDa for non-glycosylated)
For immunohistochemistry: Compare with known expression patterns in lymphoid tissues
Whenever possible, use multiple antibody clones targeting different epitopes to confirm results, particularly for novel findings or contradictory observations.
For optimal flow cytometry results when studying CD80 expression:
Sample preparation:
Staining protocol:
Controls:
Include isotype control antibodies matched for species, isotype, and fluorochrome
Use fluorescence-minus-one (FMO) controls for accurate gating
Include positive controls (activated B cells or dendritic cells)
Analysis considerations:
Gate on live, single cells before analyzing CD80 expression
Consider CD80 expression level (median fluorescence intensity) rather than just percent positive
Co-stain with lineage markers (CD19 for B cells, CD11c for dendritic cells) to identify specific CD80+ cell populations
For specific clones such as 2D10.4, the antibody has been tested by flow cytometric analysis of normal human peripheral blood cells and shows reliable detection of CD80+ populations .
When facing inconsistent CD80 antibody staining in flow cytometry experiments, consider these systematic troubleshooting approaches:
Sample-related issues:
CD80 expression varies with cell activation status—ensure consistent activation protocols
Cell viability affects staining—include viability dye and gate on live cells only
Sample age and storage conditions can impact results—use freshly prepared samples when possible
Antibody-related factors:
Antibody concentration: Perform titration to determine optimal concentration for your specific cells
Clone selection: Different clones recognize different epitopes—some may be affected by glycosylation or conformational changes
Conjugate deterioration: Check fluorochrome integrity with rainbow beads and store antibodies according to manufacturer recommendations
Protocol adjustments:
Fixation sensitivity: Some epitopes are fixation-sensitive—test different fixation methods or use unfixed cells
Blocking conditions: Insufficient blocking leads to high background—optimize blocking reagent and duration
Washing stringency: Inadequate washing causes non-specific binding—increase wash volume and number of washes
Instrument considerations:
PMT voltage optimization: Set voltages to place negative populations in the first decade
Detector sensitivity: Use more sensitive detectors for dim signals
Daily quality control: Run calibration beads to ensure consistent instrument performance
When studying CD80 on primary cells, stimulation time is critical as peak expression occurs at different timepoints depending on cell type—typically 24-48 hours for B cells and 48-72 hours for dendritic cells . Document all protocol variables meticulously to ensure reproducibility across experiments.
CD80 plays multifaceted roles in tumor immunity that vary by cancer type and microenvironment:
These findings highlight the context-dependent role of CD80 in cancer and suggest that CD80-targeting strategies must be tailored to specific tumor types and microenvironments.
CD80 antibodies offer versatile applications in cancer immunotherapy research:
Targeting approaches:
Blocking antibodies: Can be used to modulate CD80 interactions with CD28, CTLA-4, or PD-L1
Anti-CD80 immunotoxins: Constructs containing anti-CD80 monoclonal antibodies and toxic components (e.g., saponin) have shown strong cytotoxicity against CD80+ B cell lines like Raji cells, Reed-Sternberg cells, and CD80-transfected epithelial cell lines
Combination therapy research:
CD80 antibodies can be combined with PD-1/PD-L1 inhibitors to potentially enhance T cell responses
Integration with CAR-T therapy: CD80 antibodies may enhance CAR-T efficacy by modulating the immunosuppressive microenvironment
Biomarker applications:
Monitoring CD80 expression in tumors before and after treatment
Using CD80 expression patterns to stratify patients for immunotherapy trials
Tracking CD80+ immune cell populations in peripheral blood during immunotherapy
Mechanistic studies:
Investigating the impact of modulating specific CD80 interactions on anti-tumor immunity
Studying how CD80 expression in different tumor compartments affects immune cell infiltration and function
Research in glioblastoma has shown that low CD80 expression in tumor stem cells may inhibit activation of the CD28 molecule on T cells, suggesting a mechanism for immune evasion . Similarly, in pancreatic cancer, TGF-β treatment upregulates CD80 expression, which is required for migration and invasion of tumor cells . These findings highlight the potential of CD80-targeted approaches in addressing tumor-specific immune evasion mechanisms.
CD80 plays significant roles in various autoimmune conditions, offering opportunities for therapeutic intervention and research:
Multiple Sclerosis (MS):
CD80+ lymphocytes increase significantly during MS exacerbation
Following IFN-β treatment, CD80+ lymphocyte numbers decrease substantially, suggesting CD80+ cells may serve as indicators of treatment efficacy
CD80+ B cells have been identified as potential therapeutic targets for both HTLV-1-related myelopathy/tropical spastic paraparesis and multiple sclerosis
Glomerular Diseases:
Systemic Lupus Erythematosus (SLE):
Elevated CD80 expression contributes to T cell hyperactivation and autoantibody production
CD80 upregulation is associated with disease activity and tissue damage
CD80 antibodies facilitate autoimmune disease research through:
Mechanistic studies:
Flow cytometric analysis of CD80+ cell populations in patient samples compared to healthy controls
Assessment of CD80 blockade effects on T cell activation in ex vivo assays
Investigation of CD80-dependent pathways in animal models of autoimmunity
Biomarker development:
Monitoring CD80+ cells in peripheral blood as disease activity markers
Assessing urinary CD80 levels in kidney diseases
Correlating CD80 expression patterns with clinical outcomes
Therapeutic development:
Preclinical testing of CD80-targeting approaches in animal models
Investigation of combination therapies targeting multiple costimulatory pathways
Development of CD80 modulation strategies with reduced systemic immunosuppression
The therapeutic efficacy of CD80 pathway inhibition in autoimmune conditions underscores its central role in immune dysregulation and its potential as a target for precision medicine approaches in autoimmunity.
CD80 undergoes several post-translational modifications that significantly impact its function and detection:
Glycosylation:
CD80 contains multiple N-linked glycosylation sites that increase its apparent molecular weight from the predicted 33 kDa to the observed 60 kDa
Glycosylation patterns affect protein stability, half-life, and binding properties
Different glycoforms may present different epitopes, affecting antibody recognition
Tissue-specific glycosylation can result in variable antibody binding efficiency across sample types
Phosphorylation:
The cytoplasmic domain of CD80 contains potential phosphorylation sites
Phosphorylation status can affect subcellular localization and signaling
Activated immune cells may exhibit different phosphorylation patterns than resting cells
Implications for antibody-based studies:
Western blot analysis may reveal multiple bands representing different glycoforms
Treatment with glycosidases may be necessary to confirm CD80 identity in complex samples
Fixation methods for flow cytometry or immunohistochemistry may differentially affect epitope exposure
Experimental considerations:
Expression systems matter: bacterial systems lack glycosylation machinery, while mammalian systems provide more physiologically relevant modifications
Cell activation state influences post-translational modification patterns
Sample preparation methods may alter modifications and affect antibody binding
When selecting CD80 antibodies for specific applications, researchers should consider which epitopes are targeted and whether these epitopes might be affected by post-translational modifications in their experimental system. Testing multiple antibody clones that recognize different domains can help overcome detection issues related to post-translational modifications.
Contradictory findings in CD80 research often stem from methodological differences and biological complexity. Systematic approaches to resolve these contradictions include:
Standardization of experimental systems:
Cell types: Results from different cell types (primary cells vs. cell lines vs. transfected cells) may not be directly comparable
Species considerations: Human and mouse CD80 have structural and functional differences
Activation states: CD80 expression and function differ dramatically between resting and activated cells
Context-specific analysis:
Tumor microenvironment: CD80 function in tumors may differ from its role in normal tissues
Disease context: CD80 may have opposing roles in different diseases
Cell-specific effects: CD80 on dendritic cells vs. B cells vs. tumor cells may have distinct functions
Contradictory tumor prognosis associations:
Methodological standardization:
Use multiple antibody clones and detection methods to confirm findings
Implement genetic approaches (CRISPR knockout/knockin) to validate antibody specificity
Apply dose-response studies to identify threshold effects
Enhanced reporting standards:
Document antibody clones, concentrations, and incubation conditions in detail
Clearly describe cell activation status and culture conditions
Include comprehensive controls and statistical analyses
For example, the contradictory findings regarding CD80's role in different cancer types suggest context-dependent functions rather than truly contradictory mechanisms. In some cancers, CD80's interaction with CD28 predominates, enhancing anti-tumor immunity, while in others, its interaction with CTLA-4 or effects on tumor cell properties may be more significant .
Detecting low levels of CD80 expression requires specialized approaches to enhance sensitivity without sacrificing specificity:
Flow cytometry optimization:
Signal enhancement:
Instrument settings:
Optimize PMT voltages for maximum resolution of dim populations
Use instruments with high sensitivity detectors
Analysis approaches:
Employ fluorescence-minus-one controls for accurate gating
Consider median fluorescence intensity rather than percent positive
Use biexponential scaling for better visualization of dim signals
Sample preparation enhancements:
Enrichment techniques:
Magnetic separation of target cell populations to increase frequency
Density gradient centrifugation to remove irrelevant cells
Ex vivo stimulation:
Short-term culture with CD40L, IL-4, or LPS to upregulate CD80 on B cells
Treatment with GM-CSF and IL-4 for dendritic cells
Western blot sensitivity improvements:
Protein concentration methods:
Immunoprecipitation before Western blotting
TCA precipitation to concentrate proteins
Detection enhancements:
Highly sensitive ECL substrates
Longer exposure times with digital imaging systems
Immunohistochemistry enhancements:
Signal amplification:
Polymer-based detection systems
Tyramide signal amplification
Extended primary antibody incubation (overnight at 4°C)
Background reduction:
Extensive blocking with appropriate sera
Optimization of antibody concentration through careful titration
Including detergents in wash buffers to reduce non-specific binding
When studying rare cell populations or samples with low CD80 expression, combining multiple optimization approaches provides the best strategy for reliable detection while maintaining specificity.
Translating CD80 antibody research into therapeutic development requires careful consideration of several factors:
Target specificity and biology:
CD80 interactions with multiple binding partners (CD28, CTLA-4, PD-L1) create complex signaling networks
Therapeutic antibodies must be designed with specific blocking or agonistic properties
Disease-specific expression patterns must inform targeting strategies (e.g., CD80 is upregulated in B cell lymphomas and certain solid tumors)
Epitope selection:
Antibodies targeting different CD80 domains can produce distinct functional outcomes
Epitopes at CD28/CTLA-4 binding interfaces may block both stimulatory and inhibitory signals
Epitopes distant from binding interfaces can be used for diagnostic or cell-depleting strategies
Format considerations:
Full IgG antibodies provide extended half-life but limited tissue penetration
Fab and scFv fragments offer better tissue penetration but shorter half-life
Bispecific formats can engage CD80 and another target simultaneously
Antibody-drug conjugates can deliver cytotoxic payloads to CD80+ cells
Preclinical testing:
In vitro functional assays must assess impact on:
T cell activation and cytokine production
Cancer cell killing by immune effector cells
Regulatory T cell function
Animal models should recapitulate human CD80 biology
Toxicity assessment must consider expression on normal immune cells
Biomarker development:
CD80 expression levels may predict response to CD80-targeted therapy
Monitoring CD80+ cell populations during treatment can provide pharmacodynamic information
Soluble CD80 levels in serum or other fluids may serve as accessible biomarkers
Anti-CD80 immunotoxins containing monoclonal antibodies and toxic components (e.g., saponin) have demonstrated strong cytotoxicity against CD80+ cell lines including B-cell lymphoma Raji cells and CD80-transfected epithelial cell lines . The efficacy of CD80 inhibitors (abatacept) in treating certain autoimmune conditions further supports the therapeutic potential of CD80-targeted approaches .
B7-1, also known as CD80, is a protein that plays a crucial role in the immune system. It is a member of the immunoglobulin superfamily and is primarily expressed on the surface of activated B cells, dendritic cells, and monocytes. The interaction between B7-1 and its receptors is essential for the activation and regulation of T cells, which are vital for the adaptive immune response.
B7-1 is a 60 kDa glycoprotein that consists of an extracellular domain, a transmembrane domain, and a cytoplasmic tail. The extracellular domain is responsible for binding to its receptors, CD28 and CTLA-4. B7-1 is expressed on the surface of antigen-presenting cells (APCs) such as dendritic cells, macrophages, and activated B cells. Its expression is upregulated in response to inflammatory signals and during immune responses.
The primary function of B7-1 is to provide costimulatory signals necessary for T cell activation and survival. When an APC presents an antigen to a T cell, the interaction between the T cell receptor (TCR) and the antigen-MHC complex is not sufficient for full T cell activation. The binding of B7-1 to CD28 on the T cell provides a crucial second signal that promotes T cell proliferation, cytokine production, and survival. Conversely, the interaction between B7-1 and CTLA-4 delivers an inhibitory signal that downregulates T cell responses, maintaining immune homeostasis and preventing autoimmunity.
Rat anti-mouse B7-1 (CD80) antibodies are monoclonal antibodies developed in rats that specifically target the mouse B7-1 protein. These antibodies are widely used in research to study the role of B7-1 in immune responses, as well as in various immunological assays such as flow cytometry, immunohistochemistry, and Western blotting.