CD80 modulates immune responses through ligand-specific interactions:
CD28 Interaction: Triggers NF-κB, MAPK, and calcium-calcineurin pathways, enhancing T-cell survival and effector functions .
CTLA-4 Interaction: Outcompetes CD28 due to higher avidity, inducing regulatory T-cell (Treg) suppression and peripheral tolerance .
PD-L1 Crosstalk: CD80-PD-L1 binding reduces T-cell activation, offering a mechanism for tumor immune evasion .
CD80 also facilitates dendritic cell licensing, enabling cytotoxic T-cell priming via MHC class II interactions .
Multiple Sclerosis: Elevated CD80+ lymphocytes correlate with disease exacerbation; IFN-β therapy reduces CD80 expression .
Systemic Lupus Erythematosus (SLE): Increased CD80 on CD4+ T-cells links to disease activity .
Minimal Change Nephropathy: Urinary CD80 levels predict relapse frequency .
CD80’s role in HIV progression involves viral spread through immune cell interactions .
CD80-Fc Fusion Protein: Combats PD-L1-mediated immunosuppression by dual mechanisms:
CTLA-4/CD80 Axis Modulation: Enhances Treg suppression in autoimmune diseases .
Treg-Specific CD80 Expression: CD80+ Tregs exhibit high FoxP3 and CTLA-4 levels, regulating self-tolerance .
mTOR Pathway Dependency: CD86+ effector T-cells rely on mTOR, while CD80+ Tregs are mTOR-independent .
Trans-endocytosis: Activated T-cells internalize CD80 from APCs, inducing apoptosis under prolonged stimulation .
CD80 Molecule, CD80 Antigen (CD28 Antigen Ligand 1, B7-1 Antigen), B-Lymphocyte Activation Antigen B7, CTLA-4 Counter-Receptor B7.1, Activation B7-1 Antigen, CD28LG1, CD28LG, LAB7, BB1, B7, Costimulatory Molecule Variant IgV-CD80, T-Lymphocyte Activation Antigen CD80, Costimulatory Factor CD80, CD80 Antigen, B7-1, B7.1.
Sf9, Insect cells.
VIHVTKEVKE VATLSCGHNV SVEELAQTRI YWQKEKKMVL TMMSGDMNIW PEYKNRTIFD ITNNLSIVIL ALRPSDEGTY ECVVLKYEKD AFKREHLAEV TLSVKADFPT PSISDFEIPT SNIRRIICST SGGFPEPHLS WLENGEELNA INTTVSQDPE TELYAVSSKL DFNMTTNHSF MCLIKYGHLR VNQTFNWNTT KQEHFPDNLE HHHHHH.
CD80 is a costimulatory molecule primarily expressed on antigen-presenting cells (APCs) including B cells and dendritic cells. It provides the critical second signal for T cell activation by binding to CD28 on T cells, while the first signal comes from the TCR-CD3 complex interaction with the antigen peptide-MHC complex . Without this CD80-CD28 interaction, T cells may undergo activation-induced cell death (AICD) . When CD80 binds to CD28, it enhances cytokine secretion (particularly IL-2), promotes CD4+ T cell proliferation, and increases the cytotoxic activity of both CD4+ and CD8+ T cells . This dual-signal system ensures proper T cell activation while maintaining immune tolerance.
Human CD80 has three main binding partners: CD28, CTLA-4 (CD152), and PD-L1 (B7-H1). The binding characteristics are critical for understanding CD80's function:
CD80-CTLA-4: Highest affinity interaction. CD80 binds CTLA-4 with significantly greater affinity than CD28, which enables immune suppression when CTLA-4 is upregulated .
CD80-CD28: Moderate affinity interaction. This binding delivers costimulatory signals that promote T cell activation, proliferation, and cytokine production .
CD80-PD-L1: CD80 can bind to PD-L1, forming cis-heterodimers when expressed on the same cell, or interact in trans when on different cells. This interaction prevents PD-L1 from binding to PD-1, thereby reducing immunosuppression .
To study these interactions, surface plasmon resonance (SPR) or bioluminescence resonance energy transfer (BRET) assays are typically used to measure binding kinetics and affinities. Competitive binding experiments can also help determine the relative strengths of these interactions in physiological contexts.
Detection of CD80 in human samples requires specific methodology depending on the sample type and research question:
Flow cytometry: For cell surface expression on fresh or cryopreserved cells, anti-CD80 antibodies conjugated to fluorochromes (such as FITC, PE, or APC) are commonly used. Clone L307.4 has been validated for human CD80 detection .
Immunohistochemistry: For tissue sections, appropriate antibodies with validated specificity for CD80 should be used with proper controls.
ELISA: For soluble CD80 in serum or other fluids. Elevated soluble CD80 (sCD80) has been associated with poor prognosis in some soft tissue tumors .
RT-PCR and qPCR: For mRNA expression analysis when protein detection is difficult.
Western blotting: For total protein expression analysis in cell or tissue lysates.
Transcriptomic analysis: For large-scale studies, as performed in analyses of CD80 expression in cancer databases like TCGA .
Each method requires proper positive and negative controls, and results should be validated using multiple detection techniques when possible.
The CD80-PD-L1 interaction represents a complex regulatory mechanism distinct from the canonical PD-L1-PD1 inhibitory pathway. When CD80 and PD-L1 are expressed on the same cell, they can form cis-heterodimers, which serve dual functions :
The CD80-PD-L1 cis-heterodimer maintains T cell activation by allowing CD80 to engage CD28.
Simultaneously, this interaction prevents PD-L1 from binding to PD-1 on T cells, thus blocking the inhibitory signal.
The CD80-PD-L1 interaction can also inhibit the T cell immunosuppressive CTLA-4 pathway .
Research has shown that introducing mutations in the interaction sites on either PD-L1 or CD80 significantly suppresses anti-tumor immune responses in mouse models . For studying this interaction, researchers should employ:
Co-immunoprecipitation experiments to detect the formation of CD80-PD-L1 complexes
FRET (Fluorescence Resonance Energy Transfer) analyses to visualize these interactions in living cells
Functional T cell assays comparing wild-type and mutant CD80 or PD-L1 proteins that cannot form heterodimers
In vivo tumor models with genetic modifications to disrupt specific interaction sites
The CD80-PD-L1 interaction may explain why CD80-Fc fusion proteins demonstrate superior efficacy compared to anti-PD-1 or anti-PD-L1 antibodies in restoring T cell activation in some experimental systems .
CD80-Fc fusion proteins demonstrate enhanced efficacy compared to anti-PD1/PDL1 antibodies through a dual mechanism of action :
Dual-activity mechanism: Unlike antibodies that only block one interaction, CD80-Fc simultaneously:
Downstream signaling amplification: CD80-Fc activates multiple T cell signaling pathways, including:
Selective receptor engagement: Importantly, despite CD80's higher affinity for CTLA-4 than CD28, studies show that soluble CD80 does not inhibit T cell function through CTLA-4 interaction. This suggests CTLA-4 may function as a decoy receptor for CD80 rather than transmitting inhibitory signals in this context .
To investigate these mechanisms, researchers should employ:
Phosphorylation studies of key signaling molecules
Transcriptome analysis of T cells following different treatments
In vitro T cell functional assays (proliferation, cytokine production, cytotoxicity)
Competitive binding studies between CD80-Fc, PD-L1, PD-1, CD28, and CTLA-4
These findings suggest that bispecific or multi-specific therapeutics targeting both inhibitory and costimulatory pathways may offer superior clinical outcomes compared to simple checkpoint blockade .
Trans-costimulation—where CD80 is provided on a different cell than the one presenting the antigen-specific signal—represents a promising approach for cancer immunotherapy that overcomes limitations of direct tumor cell modification. Optimizing this approach requires attention to several key parameters:
Cell ratio optimization: Research demonstrates a dose-response relationship consistent with two-hit kinetics when varying the ratio of cells expressing signal 1 (antigen) versus signal 2 (CD80). The optimal ratio ranges from 1:1 to 1:10, with efficiency comparable to cis-costimulation under defined conditions .
Expression level requirements: Importantly, the level of cell-surface CD86 (a related costimulatory molecule) required for effective trans-costimulation is equivalent to that constitutively expressed by human peripheral blood monocytes . This suggests physiological expression levels may be sufficient.
Cell type selection: Human fibroblasts transduced with CD80 or CD86 efficiently provide trans-costimulation, offering a practical approach since fibroblasts:
Spatial and temporal considerations: The physical proximity between cells providing signal 1 and signal 2 is critical. Optimization strategies include:
Co-encapsulation of both cell types in biomaterials
Engineering cell adhesion molecules to promote cell-cell contact
Using controlled release systems for synchronized delivery
To evaluate trans-costimulation efficacy, researchers should assess:
T cell proliferation using CFSE dilution assays
Cytokine production profiles
In vivo tumor growth inhibition
Memory T cell generation
This approach circumvents significant technical and logistical problems associated with direct gene modification of primary tumor cells while maintaining therapeutic efficacy .
The prognostic significance of CD80 expression varies markedly across different human tumor types, presenting a complex picture that researchers must carefully consider:
These divergent patterns suggest tumor-specific immune evasion mechanisms involving CD80. For research purposes, this means:
Comprehensive transcriptomic analysis should be performed to correlate CD80 expression with clinical outcomes in specific tumor types
Immune contexture analysis is crucial to understand how CD80 functions within the tumor microenvironment
Single-cell analyses may reveal which cells express CD80 and how they interact with other immune cells
Functional studies are needed to determine whether CD80 primarily engages CD28 (activating) or CTLA-4 (inhibitory) in different tumor contexts
These findings have important therapeutic implications: tumors where high CD80 expression correlates with poor prognosis may benefit from direct CD80 targeting, while those where high CD80 improves prognosis may respond better to checkpoint inhibitors targeting CTLA-4 or PD-1/PD-L1 .
CD80's involvement in various autoimmune diseases presents opportunities for therapeutic intervention, though approaches must be carefully tailored to each condition:
Multiple Sclerosis (MS):
CD80+ lymphocytes increase significantly during MS exacerbation
After interferon-β treatment, CD80+ lymphocyte numbers decrease significantly
CD80+ cell count may serve as a biomarker for IFN-β treatment efficacy
CD80+ B cells represent a potential therapeutic target for both HTLV-1-related myelopathy/tropical spastic paraparesis and MS
Minimal Change Nephropathy:
Systemic Lupus Erythematosus (SLE):
Methodological approaches for CD80-targeted autoimmune therapy include:
Blocking antibodies: Monoclonal antibodies that specifically target CD80 without affecting related molecules
Selective fusion proteins: Engineered proteins that interfere with CD80-CD28 interaction while preserving CD80-CTLA4 interaction to promote tolerance
Small molecule inhibitors: Compounds that modulate specific CD80 interactions
Cell-based therapies: Regulatory T cells expanded or engineered to express high levels of CTLA-4 to compete for CD80 binding
Targeted delivery systems: Nanoparticles or other carriers that deliver CD80-modulating agents specifically to sites of inflammation
Research should evaluate these approaches using:
Flow cytometry to monitor CD80+ cell populations in peripheral blood and affected tissues
Functional T cell assays to assess treatment effects on immune activation
Animal models of specific autoimmune diseases
Biomarker studies correlating CD80 expression with clinical parameters and treatment responses
Designing robust T cell activation assays to study CD80-mediated costimulation requires careful attention to multiple parameters:
Cell preparation and purification:
For CD8+ and CD4+ T lymphocytes, magnetic bead isolation or fluorescence-activated cell sorting provide high purity
T cells should be rested overnight in serum-free media before assays to minimize background activation
Cell viability should exceed 95% as assessed by trypan blue or flow cytometry
Signal optimization:
Signal 1 (TCR stimulation): Use anti-CD3 antibodies at titrated concentrations (typical range 0.1-1 μg/ml) or cognate peptide-MHC complexes
Signal 2 (CD80 costimulation): When studying trans-costimulation, the ratio of cells expressing signal 1 versus signal 2 should be systematically varied (optimal ranges from 1:10 to 10:1)
Expression level: For meaningful physiological results, CD80 expression should be comparable to levels on activated professional APCs
Experimental controls:
Positive control: Full T cell activation with PMA/ionomycin or anti-CD3/CD28 beads
Negative control: Signal 1 alone without costimulation
Specificity control: Blocking antibodies against CD80 or its binding partners
Readout parameters:
Proliferation: CFSE dilution or 3H-thymidine incorporation
Cytokine production: ELISA or intracellular cytokine staining for IL-2, IFN-γ, TNF-α
Activation markers: Flow cytometry for CD25, CD69, HLA-DR
Long-term functionality: Secondary stimulation assays to assess memory generation
Timeline considerations:
Early activation markers (CD69): 6-24 hours
Cytokine production: 24-72 hours
Proliferation: 3-5 days
Memory generation: 7-14 days
For trans-costimulation studies specifically, human fibroblasts transduced with CD80 can effectively provide costimulatory signals comparable to cis-costimulation, offering a clinically relevant model system .
Investigating CD80-PDL1 interactions at the molecular level requires sophisticated techniques that provide structural, kinetic, and functional insights:
Structural analysis techniques:
X-ray crystallography: To determine atomic-level structures of CD80-PDL1 complexes
Cryo-electron microscopy: For visualizing larger complexes involving CD80-PDL1 and other binding partners
NMR spectroscopy: To analyze dynamic binding interfaces and conformational changes
Binding and kinetic assays:
Surface plasmon resonance (SPR): Gold standard for determining kon/koff rates and binding affinities
Bioluminescence resonance energy transfer (BRET): For real-time analysis of protein interactions in living cells
Isothermal titration calorimetry (ITC): To measure thermodynamic parameters of binding
Microscale thermophoresis (MST): For analyzing interactions in solution with minimal sample consumption
Cellular interaction visualization:
Proximity ligation assay (PLA): Detects protein interactions within 40nm distance in fixed cells
Förster resonance energy transfer (FRET): For live-cell visualization of molecular proximity
Fluorescence lifetime imaging microscopy (FLIM): Provides spatial resolution of interactions
Functional validation approaches:
Site-directed mutagenesis: Creating point mutations at key interface residues identified through structural studies
Domain swapping: Exchanging domains between CD80 and related molecules to determine specificity determinants
Competitive binding assays: Using titrated concentrations of CD80, PDL1, and other binding partners (PD1, CD28, CTLA4)
Systems for studying cis versus trans interactions:
For cis-heterodimer analysis: Co-expression systems with differentially tagged proteins
For trans interactions: Co-culture systems with cells expressing either CD80 or PDL1
Membrane reconstitution systems: Synthetic membranes containing purified proteins
Research has shown that CD80-PDL1 cis-heterodimers play important roles in maintaining T cell activation while preventing PD1-PDL1 inhibitory signaling . Mutations in the interaction sites significantly suppress anti-tumor immune responses in mouse models, highlighting the physiological importance of these interactions .
Designing and optimizing CD80-Fc fusion proteins for therapeutic applications requires systematic consideration of multiple parameters:
Domain selection and orientation:
Extracellular domain selection: Include complete CD80 extracellular domain without transmembrane or intracellular portions
Fc domain selection: Human IgG1 Fc is commonly used for extended half-life, though other isotypes may be selected based on desired effector functions
Orientation: N-terminal CD80 fusion to Fc typically preserves function, but both orientations should be tested
Linker optimization:
Flexible linkers (e.g., (GGGGS)n): Allow proper folding and receptor engagement
Length optimization: Systematically vary linker length to identify optimal spacing
Protease-resistant linkers: Consider stability in physiological environments
Protein engineering considerations:
Glycosylation site analysis: Preserve natural glycosylation sites in CD80 for proper folding and function
Fc modification options:
LALA mutations to reduce unwanted ADCC/CDC effects if immunosuppression is not desired
Affinity-enhanced FcRn binding for extended half-life
Stability engineering: Introduce disulfide bonds or other stabilizing mutations if needed
Production and purification:
Expression system selection: CHO or HEK293 cells typically yield properly folded and glycosylated mammalian proteins
Optimized purification protocols: Protein A chromatography followed by size exclusion chromatography
Endotoxin removal: Critical for in vivo applications
Functional validation assays:
Binding studies: SPR analysis of interaction with PDL1, CD28, and CTLA4
Competition assays: Ability to block PDL1-PD1 interactions
T cell activation assays: Measurement of proliferation, cytokine production, and cytotoxicity
In vivo pharmacokinetics: Half-life determination in relevant animal models
Research has demonstrated that properly designed CD80-Fc fusion proteins can be more effective than antibodies to PD1 or PDL1 in preventing PDL1-PD1-mediated suppression and restoring T cell activation . This superior efficacy stems from the dual mechanism of blocking inhibitory PDL1-PD1 interactions while simultaneously providing costimulation through CD28 . Interestingly, despite CD80's higher affinity for CTLA4 than CD28, soluble CD80 does not inhibit T cell function through CTLA4 interaction, suggesting CTLA4 may function primarily as a decoy receptor in this context .
CD80 expression has emerging value as a biomarker in both cancer and autoimmune diseases, though with disease-specific implications:
Methodological approaches for CD80 biomarker assessment include:
Tissue analysis:
Immunohistochemistry with validated anti-CD80 antibodies
Multiplexed immunofluorescence to examine CD80 in relation to other immune markers
Digital pathology with quantitative image analysis for standardized assessment
Liquid biopsies:
Flow cytometry of peripheral blood for CD80+ cell enumeration
ELISA or multiplex assays for soluble CD80 in serum, plasma, or urine
Single-cell RNA sequencing for comprehensive immune profiling
Molecular analysis:
Transcriptomic analysis of CD80 expression in tumor specimens
Integration with other immune signatures for comprehensive immune profiling
For clinical implementation, standardized protocols with appropriate reference ranges and quality control measures are essential to ensure reproducible biomarker assessment across different laboratories and clinical settings .
Translating CD80-based therapies to clinical applications faces several significant challenges that researchers must address:
Dual nature of CD80 signaling:
CD80 interacts with both stimulatory (CD28) and inhibitory (CTLA-4) receptors
CTLA-4 has higher affinity for CD80 than CD28 (approximately 10-20 fold)
T cells upregulate CTLA-4 upon activation, potentially shifting the balance toward inhibitory signaling
Solution approaches: Engineered CD80 variants with altered receptor binding preferences or contextual delivery strategies
Delivery and pharmacokinetic challenges:
Soluble CD80-Fc may have limited tissue penetration, particularly in solid tumors
Potential rapid clearance through endogenous receptor-mediated mechanisms
Solution approaches: PEGylation, albumin fusion, or advanced formulation strategies to extend half-life
Context-dependent effects across diseases:
Manufacturing and quality control:
Ensuring consistent glycosylation and proper folding of CD80-Fc fusion proteins
Developing reliable potency assays that capture dual mechanisms of action
Solution approaches: Standardized cell expression systems and comprehensive analytical characterization
Clinical trial design challenges:
Patient selection strategies given the complex and context-dependent role of CD80
Appropriate clinical endpoints that reflect immunomodulatory mechanisms
Combination therapy approaches—determining optimal partners and sequencing
Solution approaches: Adaptive trial designs with integrated biomarker analysis
Safety considerations:
Risk of excessive immune activation leading to cytokine release syndrome
Potential for inducing or exacerbating autoimmunity
Unanticipated effects on diverse cell populations expressing CD80 receptors
Solution approaches: Careful dose escalation, robust safety monitoring, and potentially localized delivery approaches
CD80 offers significant potential for enhancing cancer vaccine efficacy through multiple mechanisms that address key limitations of current approaches:
Enhanced antigen presentation and T cell priming:
CD80-modified dendritic cells or other antigen-presenting cells can provide optimal costimulation for naïve T cell activation
Research shows CD80 co-expression significantly enhances the immunogenicity of cancer vaccines compared to unmodified vaccines
This approach may be particularly valuable for weak tumor antigens that typically induce suboptimal T cell responses
Combination with checkpoint blockade:
CD80-based vaccines can synergize with checkpoint inhibitors by:
Providing strong costimulation via CD28 engagement
Competitively inhibiting PD-L1-PD-1 interactions through CD80-PD-L1 binding
Creating more favorable TME for effector T cell function
Advanced vaccine design strategies:
Bifunctional fusion constructs: Linking CD80 directly to tumor antigens for targeted delivery and enhanced immunogenicity
Genetic modification approaches: Viral vectors or mRNA encoding both tumor antigens and CD80
Nanoparticle platforms: Co-delivery of antigens and CD80 (either as protein or encoding nucleic acids) for synchronized uptake by APCs
Cell-based vaccines: Tumor cells or artificial APCs engineered to express both tumor antigens and CD80
In situ vaccination approaches:
Local delivery of CD80-encoding vectors to convert the tumor into its own vaccine
Combination with ablative therapies (radiation, cryotherapy) to release tumor antigens while providing costimulation
Potential for abscopal effects through systemic immune activation
Memory formation enhancement:
CD80-CD28 signaling promotes development of long-lived memory T cells
This could address the challenge of transient responses seen with many current immunotherapies
Prime-boost strategies incorporating CD80 at different stages may optimize memory formation
Research implementing CD80 in vaccines has demonstrated enhanced anti-tumor efficacy compared to traditional approaches . For HIV vaccination, co-stimulation with CD80 enhances the acquisition of antigen-specific amplification and effector function in HIV-specific memory CD8+ T cells, representing a promising therapeutic vaccination strategy .
Methodologically, researchers should evaluate CD80-enhanced vaccines through:
Assessment of T cell quantity (expansion) and quality (polyfunctionality, memory phenotype)
In vivo tumor challenge models with long-term follow-up for relapse
Immune correlate studies to identify determinants of response
Spatial analysis of the tumor microenvironment before and after vaccination
While the search results don't specifically address CD80's role in COVID-19, we can use our understanding of CD80 biology to propose several significant areas of investigation:
Dysregulated costimulation in COVID-19 pathophysiology:
COVID-19 severity correlates with immune dysregulation, including T cell exhaustion and hyperinflammation
CD80 expression may be altered on antigen-presenting cells during SARS-CoV-2 infection
Methodological approach: Flow cytometric analysis of CD80 expression on monocytes, dendritic cells, and B cells from COVID-19 patients of varying disease severity compared to healthy controls
CD80 as a potential therapeutic target:
In severe COVID-19, modulating CD80 might help restore T cell function or control excessive inflammation
In mild/moderate disease, enhancing CD80-mediated costimulation might boost anti-viral immunity
Methodological approach: Ex vivo studies using patient samples to evaluate the effect of CD80 modulation on T cell responses to viral antigens
CD80's role in COVID-19 vaccine efficacy:
CD80-mediated costimulation is crucial for generating robust T cell responses
Variations in CD80 expression or function might contribute to differential vaccine responses
Methodological approach: Correlative studies measuring CD80 expression or polymorphisms against vaccine-induced immune responses
CD80-Fc as a potential therapeutic agent:
Similar to its application in cancer, CD80-Fc could potentially:
Block PD-L1-PD-1 interactions that may contribute to T cell exhaustion in COVID-19
Provide costimulation through CD28 to enhance anti-viral T cell responses
Methodological approach: Preclinical studies in appropriate animal models of COVID-19 testing CD80-Fc administration at different disease stages
CD80 in long COVID pathophysiology:
Chronic immune dysregulation is a hallmark of long COVID
Altered CD80 expression or function might contribute to persistent symptoms
Methodological approach: Longitudinal studies comparing CD80 expression profiles between patients who recover fully versus those who develop long COVID
Research methodologies should include:
Single-cell RNA sequencing to characterize CD80 expression in diverse immune populations
Spatial transcriptomics/proteomics to examine CD80 distribution in affected tissues
Functional assays evaluating T cell responses with and without CD80 modulation
Genetic association studies examining CD80 polymorphisms and COVID-19 outcomes
Given CD80's role in other viral infections and autoimmune conditions, understanding its function in COVID-19 could provide valuable insights for both therapeutic development and understanding disease pathophysiology.
CD80, also known as B7-1, is a type I membrane protein that belongs to the immunoglobulin superfamily. It plays a crucial role in the regulation of T-cell activation and immune responses. CD80 is primarily expressed on antigen-presenting cells (APCs) such as dendritic cells, activated B-cells, and macrophages .
CD80 is a transmembrane glycoprotein with an extracellular domain that includes an immunoglobulin constant-like domain and a variable-like domain required for receptor binding . It is closely related to CD86 (B7-2), and both molecules often work in tandem to regulate immune responses .
The primary function of CD80 is to provide co-stimulatory signals necessary for T-cell activation and survival. It achieves this by binding to receptors on T-cells, specifically CD28 and CTLA-4. The interaction between CD80 and CD28 provides a positive co-stimulatory signal that promotes T-cell activation and proliferation. Conversely, the interaction between CD80 and CTLA-4 delivers an inhibitory signal that downregulates T-cell responses .
Recombinant CD80 is produced using various expression systems, including human embryonic kidney (HEK293) cells. The recombinant protein is typically tagged with a polyhistidine (His) tag to facilitate purification and detection . The recombinant form of CD80 retains its ability to bind to its receptors, making it a valuable tool for research and therapeutic applications.
Recombinant CD80 is widely used in immunological research to study T-cell activation and co-stimulation. It is also employed in the development of immunotherapies for cancer and autoimmune diseases. By modulating the interactions between CD80 and its receptors, researchers can influence immune responses to achieve therapeutic benefits .