Mouse OX40L is a 35 kDa type II transmembrane glycoprotein encoded by the Tnfsf4 gene . The recombinant form used in studies consists of 157 amino acids (residues 51-198) with a C-terminal His tag, produced in Sf9 Baculovirus cells . It exhibits:
Cellular expression: Activated B cells, dendritic cells, macrophages, and vascular endothelial cells
Receptor specificity: Binds exclusively to OX40 (CD134) on activated CD4+/CD8+ T cells
OX40L-OX40 interactions mediate crucial immunomodulatory effects:
Promotes germinal center formation through T follicular helper (Tfh) cell differentiation
Essential for secondary antibody responses and affinity maturation
Parameter | InVivoMAb anti-OX40L (BE0033-1) |
---|---|
Clone | RM134L |
Isotype | Rat IgG2b |
Applications | - In vivo signaling blockade - T cell proliferation inhibition |
Efficacy in Asthma | Reduces BALF cytokines by 40-65% |
OX40L neutralization suppresses pathogenic Th2/Th17 responses in asthma
Agonist anti-OX40 antibodies exacerbate lupus nephritis (3-fold increase in proteinuria)
OX40:Fc fusion protein delays lupus onset (survival improvement: 60% → 90%)
Dose-dependent effects: High OX40L concentrations paradoxically enhance T cell apoptosis in chronic inflammation
Strain variability: C57BL/6 OX40L-Tg mice develop autoimmunity, while BALB/c counterparts remain resistant
Temporal regulation: Early vs late OX40L blockade shows opposing outcomes in memory T cell development
OX40 Ligand, a member of the tumor necrosis factor (TNF) receptor family, plays a crucial role in immune response. Its receptor, OX40, is primarily found on activated CD4+ T cells. OX40 Ligand is expressed on various antigen-presenting cells, including macrophages, DC2s, and activated B lymphocytes. The expression of OX40 is transient and occurs after T cell receptor engagement. The interaction between OX40 and OX40L stimulates the proliferation and differentiation of activated B cells, enhances immunoglobulin secretion, and provides a survival signal for T cells, ultimately contributing to the development of memory T cells.
Recombinant Mouse OX40L, produced in Sf9 Baculovirus cells, is a single, glycosylated polypeptide chain. It comprises 157 amino acids (51-198 aa), resulting in a molecular mass of 17.7 kDa. The protein is fused to a 6 amino acid His tag at the C-terminus and purified using proprietary chromatographic techniques.
Sterile filtered, colorless solution.
The OX40L solution is provided at a concentration of 1 mg/ml and contains 10% Glycerol and Phosphate-Buffered Saline (pH 7.4).
For short-term storage (2-4 weeks), store the vial at 4°C. For extended periods, store frozen at -20°C. Adding a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Greater than 90.0% purity as determined by SDS-PAGE analysis.
Biological activity is determined by its binding ability in a functional ELISA using Human TNFRSF4 (Catalog # cyt-1039).
Tumor necrosis factor ligand superfamily member 4, OX40 ligand, OX40L, OX-40L, CD252, Tnfsf4, Ox40l, Txgp1l, gp3, OX4, Ath-1, Ath1, CD134L, gp34, Tnlg2b, Txgp1l
Sf9, Baculovirus cells.
ADPSSSPAKD PPIQRLRGAV TRCEDGQLFI SSYKNEYQTM EVQNNSVVIK CDGLYIIYLK GSFFQEVKID LHFREDHNPI SIPMLNDGRR IVFTVVASLA FKDKVYLTVN APDTLCEHLQ INDGELIVVQ LTPGYCAPEG SYHSTVNQVP LHHHHHH
Mouse OX40L (OX40 Ligand) is a type II transmembrane protein belonging to the TNF superfamily that serves as the ligand for the OX40 receptor (CD134). It plays critical roles in multiple immunological processes:
T cell activation and expansion: OX40L expressed on antigen-presenting cells (APCs) provides costimulatory signals to OX40-expressing activated T cells, promoting their survival, proliferation, and cytokine production .
Memory T cell development: OX40-OX40L interactions significantly influence the generation of CD4+ memory T cells, with studies in OX40-deficient mice showing greatly reduced memory CD4+ T cell populations .
Regulation of inflammatory responses: This pathway mediates both pro-inflammatory and regulatory functions depending on the context, with critical roles in autoimmunity, infection, and tumor immunity .
Studies with OX40L knockout mice have demonstrated that these interactions are essential for efficient clonal expansion of naive T cells and effective primary CD4+ T cell responses .
OX40L expression shows distinctive patterns across different immune cell populations:
Dendritic cells (DCs): Expression is induced following activation with stimuli like LPS. Bone marrow-derived DCs upregulate OX40L in response to LPS treatment .
B cells: Freshly isolated lymphocyte populations typically don't express detectable OX40L, but B cells can strongly express OX40L after stimulation with anti-CD40 plus anti-IgM antibodies for approximately 3 days .
Macrophages and PMNs: OX40L is upregulated on peritoneal macrophages and polymorphonuclear cells (PMNs) in response to inflammatory stimuli, as demonstrated in cecal ligation and puncture (CLP) models of polymicrobial sepsis .
Expression is typically undetectable in resting immune cells and requires activation signals, making it an important marker for immune cell activation status in research models .
OX40L-deficient (OX40L−/−) mice display several notable characteristics under non-challenged conditions:
Normal lymphoid organ development: These mice do not show any apparent abnormalities in the formation of lymphoid organ structures .
Normal baseline lymphocyte populations: The numbers of T and B cells in these mice are comparable to wild-type counterparts under steady-state conditions .
Gene targeting strategy: The OX40L mutation was created by inserting a neomycin resistance gene cassette into the first exon through homologous recombination in embryonic stem cells .
Flow cytometric analysis confirms the absence of OX40L expression on activated B cells from homozygous OX40L-deficient mice, which would normally express OX40L strongly following stimulation with anti-CD40 plus anti-IgM antibodies .
Generation of OX40L knockout mice involves several key steps:
Vector construction: The first exon of the OX40L gene is replaced with a neomycin resistance cassette (PGKNeo). A diphtheria toxin A subunit (DT-A) gene is incorporated into the 3′ end of the vector to select against random integration .
ES cell targeting: The linearized vector is electroporated into embryonic stem (ES) cells, followed by G418 selection to identify clones with the disrupted OX40L gene .
Confirmation of homologous recombination: Southern blotting using flanking probes identifies ES clones heterozygous for OX40L disruption. In validated clones, 10.0-kb and 3.0-kb bands correspond to mutated and wild-type alleles, respectively .
Chimeric mouse production: Validated ES clones are injected into blastocysts to generate chimeric mice, which are bred with wild-type mice to establish heterozygous offspring .
Functional validation: Complete absence of OX40L expression is confirmed by flow cytometry on activated B cells (stimulated with anti-CD40 plus anti-IgM for 3 days), which would normally express high levels of OX40L .
For immunological analyses, OX40L knockout mice are typically backcrossed at least six generations onto the C57BL/6 background to ensure genetic homogeneity .
Humanized OX40/OX40L mouse models are valuable tools for evaluating potential therapeutic antibodies targeting this pathway:
B-hOX40/hOX40L mice: These mice express human versions of both OX40 and OX40L instead of their mouse counterparts. Flow cytometry analysis confirms human-specific expression patterns:
Validation: The functionality of these humanized models is assessed through:
Expression analysis using species-specific antibodies that can distinguish human from mouse proteins .
Flow cytometric analysis of spleen leukocyte subpopulations to ensure normal immune compartment development .
Experimental systems such as induction of autoimmune disease models to test the efficacy of anti-human OX40L antibodies .
These models enable preclinical testing of human-specific therapeutic antibodies targeting the OX40/OX40L pathway in a physiologically relevant in vivo setting.
The genetic background of mice significantly impacts the manifestation of OX40L deficiency in various disease models:
Autoimmune disease models: In experimental autoimmune encephalomyelitis (EAE), OX40L−/− mice on the C57BL/6 background show reduced clinical manifestations due to abortive T cell priming, with decreased production of IFN-γ, IL-2, and IL-6 .
Lupus models: In NZB/W F1 mice (a model for systemic lupus erythematosus), disruption of the OX40/OX40L pathway using antagonists significantly delays the onset of severe proteinuria and improves survival in type I IFN-accelerated lupus .
Sepsis models: OX40L−/− mice show dramatically improved survival compared to wild-type mice after cecal ligation and puncture (CLP), associated with decreased levels of inflammatory cytokines (IL-6, IL-10, IL-12) and reduced remote organ injury .
These variations highlight the importance of selecting appropriate genetic backgrounds when designing experiments and interpreting results from OX40L knockout studies across different disease contexts.
OX40L provides critical signals that shape CD4+ T cell fate and memory development:
Effector T cell generation: OX40 knockout animals generate significantly fewer primary effector CD4+ T cells after immunization. Conversely, treatment with agonist OX40 antibodies enhances antigen-specific effector T cell populations and prevents T cell tolerance induction .
Memory CD4+ T cell formation: Work with OX40-deficient mice has clearly demonstrated that OX40 signaling substantially impacts memory CD4+ T cell generation. In vivo treatment with agonist anti-OX40 strongly promotes more memory CD4+ T cells to develop .
T helper cell polarization: OX40L expressed on dendritic cells promotes Th1 differentiation while inhibiting regulatory T cell (Treg) development. This is supported by altered expression of co-inhibitory receptors like PD-L1 on the DCs .
The timing of OX40-OX40L interactions appears flexible - recent two-photon imaging studies show that naïve CD4+ T cells may interact with dendritic cells for 24-48 hours after antigen priming, but OX40 and OX40L expression can occur beyond this timeframe, suggesting multiple opportunities for this pathway to influence T cell fate .
OX40L contributes significantly to inflammatory disease development through multiple mechanisms:
Experimental autoimmune encephalomyelitis (EAE): OX40L−/− mice show impaired T cell priming that greatly reduces clinical manifestations of actively induced EAE. Interestingly, adoptive transfer experiments reveal that OX40L−/− mice can still support disease when receiving wild-type donor T cells, but OX40L−/− donor T cells fail to transfer disease to wild-type recipients .
Systemic lupus erythematosus (SLE): The OX40/OX40L pathway drives both cellular and humoral autoimmune responses during lupus nephritis. Treatment with agonist anti-OX40 mAbs exacerbates renal disease in NZB/W F1 mice by:
Sepsis: OX40L upregulation on innate immune cells promotes inflammation in polymicrobial sepsis. OX40L−/− mice show dramatic improvement in survival after cecal ligation and puncture, with reduced cytokine production and decreased remote organ injury (pulmonary capillary leak, hepatic NF-κB induction, and hepatic PMN accumulation) .
These findings demonstrate that OX40L signaling can be pathogenic in multiple inflammatory contexts, making it a potential therapeutic target.
OX40L significantly enhances dendritic cell (DC) immunogenicity through several mechanisms:
Antigen presentation efficiency: OX40L is required for efficient presentation, but not uptake, of antigens by DCs to stimulate both CD4+ and CD8+ T cells in vivo .
T cell differentiation modulation:
Enhanced CD8+ T cell cytotoxicity: CD8+ T cells demonstrate enhanced cytotoxicity toward target cells in response to OX40L expression on transferred DCs in vivo .
Anti-tumor immunity: OX40L expression on DCs leads to better tumor metastasis inhibition in DC-mediated tumor immunity models .
These findings highlight OX40L as a potential target in DC-based vaccine strategies for enhanced anti-tumor efficacy in vivo and suggest that modulating OX40L expression could be used to fine-tune DC immunogenicity for therapeutic applications.
For reliable OX40L expression analysis in mouse cells, follow these methodological approaches:
Flow Cytometry Protocol:
Cell isolation: Harvest target cells (e.g., splenic B cells, bone marrow-derived DCs, peritoneal macrophages) using appropriate tissue-specific protocols.
Stimulation conditions:
For B cells: Stimulate with anti-CD40 plus anti-IgM antibodies for 3 days (fresh lymphocytes typically lack detectable OX40L) .
For DCs: Differentiate bone marrow cells with appropriate cytokines (e.g., GM-CSF), then stimulate with LPS .
For peritoneal cells in sepsis models: Harvest cells 18 hours after cecal ligation and puncture (CLP) .
Antibody staining: Use fluorochrome-conjugated anti-mouse OX40L antibodies with appropriate isotype controls.
Analysis: Gate on relevant cell populations and quantify OX40L expression level.
Immunoblotting Protocol:
Confirm PMN expression of OX40L via immunoblot as an additional validation method, particularly when expression on rare populations is being assessed .
Important considerations:
Always include both positive and negative controls (e.g., wild-type vs. OX40L knockout cells) to confirm antibody specificity.
When studying humanized models, use species-specific antibodies that can distinguish human from mouse OX40L expression .
For stimulated cells, perform time-course experiments to determine optimal timing for OX40L detection, as expression kinetics may vary by cell type.
To investigate OX40L-dependent T cell responses in vivo, researchers can employ several complementary approaches:
Agonist/Antagonist Treatment Models:
Agonist approaches: Administer agonist anti-OX40 mAbs to wild-type mice to potently enhance the generation of antigen-specific effector T cells or memory CD4+ T cells .
Antagonist approaches: Use antagonist OX40:Fc fusion proteins to block the pathway, especially effective in disease models like type I IFN-accelerated lupus .
Adoptive Transfer Experiments:
Transfer wild-type T cells to OX40L−/− recipients: This approach tests whether OX40L expression in the recipient environment is necessary for effector responses .
Transfer OX40L−/− T cells to wild-type recipients: This tests whether OX40L expression on donor T cells is required for disease transfer .
Disease Induction Models:
EAE model: Actively induce EAE in wild-type versus OX40L−/− mice to assess disease progression, clinical scores, and T cell responses in vitro (cytokine production) .
Sepsis model: Use cecal ligation and puncture (CLP) to study OX40L contributions to innate immune responses and inflammation .
Tumor models: Employ DC-mediated tumor immunity models to evaluate OX40L effects on tumor metastasis inhibition .
Analysis Endpoints:
T cell expansion (absolute numbers)
Cytokine production (IFN-γ, IL-2, IL-6, IL-10, IL-12)
Cell activation markers
Disease-specific parameters (e.g., proteinuria, survival, clinical scores)
Tissue inflammation markers (e.g., pulmonary capillary leak, hepatic NF-κB induction)
These approaches allow comprehensive assessment of how OX40L signals influence T cell priming, expansion, differentiation, and effector functions across various immunological contexts.
Researchers can modulate OX40L expression or function using several techniques:
Genetic Approaches:
Gene knockout models: OX40L knockout mice (OX40L−/−) serve as complete loss-of-function models, generated by replacing the first exon with a neomycin resistance cassette .
Transgenic overexpression: OX40L-transgenic mice (OX40L-Tg) provide gain-of-function models with markedly enhanced T cell responses to protein antigens .
Humanized models: B-hOX40/hOX40L mice with human OX40 and OX40L replacing mouse counterparts enable testing of human-specific therapeutics .
Antibody-Mediated Approaches:
Blocking antibodies: Anti-OX40L antibodies block the interaction between OX40L and OX40, reducing T cell expansion and functional priming .
Agonistic antibodies: Anti-OX40 antibodies stimulate the receptor in a manner similar to OX40L binding, enhancing T cell responses .
Fusion proteins: OX40:Fc fusion proteins act as antagonists by binding to OX40L and preventing its interaction with OX40 .
Cell-Based Approaches:
Dendritic cell manipulation: Modify OX40L expression on DCs ex vivo before adoptive transfer to study effects on T cell stimulation and differentiation .
Adoptive transfer: Transfer OX40L-sufficient or OX40L-deficient cells to appropriate recipients to study cell-specific contributions of OX40L .
Experimental Disease Settings:
EAE induction: Compare wild-type, OX40L−/−, and OX40L-Tg mice in EAE models to assess how OX40L modulation affects disease progression .
Sepsis models: Use CLP in OX40L−/− mice or anti-OX40L antibody treatment to evaluate OX40L's role in inflammation and mortality .
Tumor models: Employ DC-mediated tumor immunity models to assess how OX40L modulation affects anti-tumor responses .
These complementary approaches provide researchers with multiple tools to investigate OX40L biology across different experimental contexts.
OX40L targeting offers several promising strategies for cancer immunotherapy:
DC-Based Vaccine Enhancement:
OX40L expression on dendritic cells significantly enhances their ability to stimulate anti-tumor T cell responses. Studies show that OX40L promotes DC-mediated tumor metastasis inhibition in vivo .
OX40L enhances both CD4+ and CD8+ T cell activation by DCs, with CD8+ T cells showing increased cytotoxicity toward target cells when stimulated by OX40L-expressing DCs .
Mechanism of Action:
OX40L on DCs drives CD4+ T cells toward Th1 differentiation while inhibiting regulatory T cell development, creating a more pro-inflammatory tumor microenvironment .
This effect is partly mediated through altered expression of co-inhibitory receptors like PD-L1 on DCs .
Enhanced CD8+ T cell cytotoxicity contributes directly to improved tumor cell killing .
Implementation Strategies:
Ex vivo DC modification: Genetic approaches to overexpress OX40L on DCs before adoptive transfer into tumor-bearing hosts.
Combination therapy: Pairing OX40/OX40L pathway agonists with checkpoint inhibitors (anti-PD-1/PD-L1) to overcome immunosuppression.
Targeted delivery: Developing tumor-targeting OX40 agonists to activate tumor-infiltrating T cells specifically.
These approaches leverage OX40L's strong immunogenic properties to enhance anti-tumor immune responses while potentially limiting systemic immune activation and associated toxicities.
OX40L exerts distinct effects on innate and adaptive immune components:
Innate Immune Effects:
PMNs and Macrophages: In sepsis models, OX40L is upregulated on peritoneal macrophages and PMNs after cecal ligation and puncture. PMNs express both OX40L and OX40, suggesting potential autocrine signaling .
Inflammatory Regulation: OX40L−/− mice show dramatically improved survival in sepsis models, with decreased levels of inflammatory cytokines (IL-6, IL-10, IL-12) in plasma, bronchoalveolar lavage, and peritoneal fluid .
Tissue Damage Reduction: OX40L deficiency reduces remote organ injury as evidenced by decreased pulmonary capillary leak, hepatic NF-κB induction, and hepatic PMN accumulation .
Adaptive Immune Effects:
T Cell Priming: OX40L is critical for efficient T cell priming, with OX40L−/− mice showing impaired APC capacity and reduced T cell responses in models like EAE .
Memory Formation: OX40-OX40L interactions are essential for generating high frequencies of memory CD4+ T cells .
Humoral Responses: Treatment with agonist anti-OX40 mAbs increases follicular helper T cells and plasmablasts in the spleen, leading to elevated serum IgM levels and enhanced renal glomerular IgM deposition in lupus models .
Comparative Analysis:
In innate immunity, OX40L primarily drives inflammatory responses and tissue damage.
In adaptive immunity, OX40L shapes T cell differentiation, memory formation, and subsequent B cell responses.
OX40L serves as a bridge between innate and adaptive immunity, with dendritic cells using OX40L to translate innate immune activation into effective adaptive responses.
Understanding these differential effects is crucial for designing targeted therapeutic interventions that modulate specific aspects of immunity while minimizing unwanted effects.
OX40L functions within a complex network of costimulatory and coinhibitory pathways:
Interactions with CD28/CD80-CD86 Pathway:
OX40L-transgenic mice (OX40L-Tg) that also lack CD28 (OX40L-Tg/CD28−/−) fail to develop experimental autoimmune encephalomyelitis (EAE), demonstrating a requirement for CD28 costimulation .
This suggests that OX40L signals cannot substitute for, but rather complement, the classical CD28 costimulatory pathway in T cell activation.
Interactions with CD40/CD40L Pathway:
OX40L-Tg/CD40−/− mice also fail to develop EAE, indicating that CD40 signals are essential even in the presence of enhanced OX40L signaling .
This highlights the sequential nature of costimulatory requirements, with CD40-CD40L interactions potentially preceding and enabling effective OX40-OX40L signaling.
Relationship with PD-1/PD-L1 Pathway:
OX40L expression on dendritic cells alters their expression of the coinhibitory receptor PD-L1, which influences their ability to promote Th1 versus Treg differentiation .
This suggests cross-regulation between costimulatory (OX40L) and coinhibitory (PD-L1) pathways on antigen-presenting cells.
Integrated Model:
Initial TCR engagement with peptide-MHC complex occurs
CD28-CD80/CD86 interactions provide primary costimulation
CD40-CD40L interactions enhance APC activation and function
OX40-OX40L interactions subsequently provide extended signals for T cell survival, proliferation, and differentiation
PD-1/PD-L1 and other inhibitory pathways eventually contain the response
This hierarchical and temporally regulated network of interactions ensures appropriate immune response magnitude and duration, with OX40L serving as a critical amplifier that operates within the constraints established by other pathways.
When confronted with seemingly contradictory findings about OX40L function, consider these methodological factors:
Model-Specific Differences:
Disease context: OX40L may be protective in some infections but pathogenic in autoimmunity. In EAE, OX40L deficiency reduces disease severity , while in sepsis models, OX40L deficiency improves survival .
Genetic background: Mouse strain backgrounds significantly impact OX40L function. Always consider the genetic background (e.g., C57BL/6, NZB/W F1) when comparing studies .
Timing Considerations:
Expression kinetics: OX40L expression occurs at specific timepoints after immune activation. Interventions at different timepoints may yield opposing results .
Disease phase: OX40L blockade may be beneficial during initiation phases but ineffective or harmful during established disease.
Technical Variables:
Antibody specificity: Confirm antibody specificity using appropriate controls (e.g., OX40L knockout cells) .
Redundancy and compensation: Other costimulatory pathways may compensate for OX40L deficiency in different models, masking its importance .
Integrated Analysis Framework:
Compare experimental systems methodically: Catalog key variables (strain, disease model, intervention timing, readouts) across studies.
Consider cell-specific roles: OX40L might have different functions depending on which cell type expresses it (DCs vs. B cells vs. PMNs).
Examine combined pathway interventions: Studies testing OX40L-Tg/CD28−/− or OX40L-Tg/CD40−/− mice reveal interaction requirements that help resolve apparent contradictions .
Both gain-of-function (OX40L-Tg, agonist antibodies) and loss-of-function (OX40L−/−, blocking antibodies) studies
Both adoptive transfer and direct induction models
Both in vitro and in vivo readouts
Researchers should be aware of these common challenges in OX40L studies:
Expression Analysis Challenges:
Baseline expression: OX40L is typically undetectable on resting cells, requiring proper stimulation protocols before analysis .
Antibody validation: Always confirm antibody specificity using OX40L knockout controls to avoid false positives .
Timing considerations: OX40L expression is transient; perform time-course experiments to identify optimal detection windows.
Knockout Model Considerations:
Compensatory mechanisms: Long-term genetic deletion may trigger compensatory upregulation of other costimulatory pathways.
Background effects: Ensure sufficient backcrossing (at least 6 generations) to appropriate genetic backgrounds .
Mixed chimeras: Consider generating bone marrow chimeras to distinguish cell-intrinsic versus environmental effects of OX40L deficiency.
Functional Assay Pitfalls:
Secondary effects: OX40L deficiency on APCs affects T cell responses, which then affect B cell responses - distinguish direct versus indirect effects.
Context dependency: OX40L effects vary dramatically between homeostatic, inflammatory, and autoimmune contexts.
Dose considerations: Agonist antibody dose significantly impacts outcomes - titrate carefully.
Experimental Design Recommendations:
Include both sexes in studies and analyze data by sex, as immune responses can differ significantly.
Use multiple time points for analysis, especially in disease progression models.
Employ comprehensive immune phenotyping beyond the primary cell type of interest.
Consider conditional knockout models (e.g., DC-specific OX40L deletion) to overcome global knockout limitations.
Use complementary approaches (genetic models, antibody blockade, adoptive transfer) to validate findings.
By addressing these potential pitfalls proactively, researchers can generate more robust and reproducible data in OX40L studies.
For translational research bridging mouse and human OX40L biology:
Comparative Expression Analysis:
Cell distribution: Compare OX40L expression patterns across equivalent cell populations in mice and humans under matched stimulation conditions.
Kinetics: Analyze temporal expression patterns following activation to identify potential species differences.
Pathway regulation: Compare transcriptional control mechanisms regulating OX40L expression.
Functional Conservation Assessment:
Signal transduction: Determine whether downstream signaling pathways triggered by OX40-OX40L interactions are conserved.
Biological outcomes: Compare effects on T cell expansion, differentiation, and memory formation between species.
Disease relevance: Assess whether OX40L participates in similar pathological processes in mouse models and human diseases.
Humanized Mouse Models:
B-hOX40/hOX40L mice: Use mice expressing human OX40 and OX40L to test human-specific therapeutics .
Validation approaches: Confirm proper expression using species-specific antibodies and flow cytometry .
Functional testing: Verify that humanized mice recapitulate expected immune responses in appropriate disease models .
Cross-Species Reactivity Assessment:
Test whether mouse OX40L can stimulate human OX40 and vice versa.
Evaluate species-specific antibodies for potential cross-reactivity.
Compare amino acid sequences and structural features to identify conserved binding domains.
Translational Framework:
Establish proof-of-concept in mouse models using mouse-specific reagents.
Validate key findings in humanized mouse models using human-specific reagents.
Correlate with human ex vivo systems (peripheral blood mononuclear cells, tissue explants).
Design early clinical studies based on converging evidence from multiple systems.
This systematic approach helps identify which aspects of OX40L biology translate reliably between species, enabling more effective clinical development of therapeutic strategies targeting this pathway.
The murine OX40L cDNA encodes a protein consisting of 198 amino acids, which includes a 28 amino acid N-terminal cytoplasmic domain, a 20 amino acid transmembrane segment, and a 150 amino acid C-terminal extracellular domain . OX40L is primarily expressed on antigen-presenting cells (APCs) such as activated B cells, dendritic cells, and macrophages . It can also be found on activated endothelial cells and CD4+ T cells during antigen stimulation .
OX40L binds to its receptor, OX40 (CD134), which is expressed on activated T cells. This interaction is pivotal for T cell activation and immune response modulation. When OX40 on T cells binds to OX40L on APCs, it leads to enhanced T cell survival, proliferation, and cytotoxicity . Additionally, it promotes the formation of memory T cells and reduces the immunosuppressive functions of regulatory T cells (Tregs) .
Recombinant Mouse OX40 Ligand is produced using a mouse myeloma cell line, NS0-derived mouse OX40L protein, which includes an N-terminal 10-His tag . The recombinant protein is typically purified to a high degree of purity (>95%) and is used in various research applications, including immunology and oncology studies .
OX40L has been extensively studied for its role in immune regulation and its potential as a therapeutic target in cancer immunotherapy. OX40 agonists are being tested in clinical trials, both as monotherapy and in combination with other immunotherapy agents, particularly immune checkpoint inhibitors . The goal is to harness the immune-stimulatory properties of OX40/OX40L interactions to enhance anti-tumor responses .