Gene location: Chromosome 4 (mouse genome) with NCBI Gene ID 22163 .
Protein: Type I transmembrane glycoprotein (256 amino acids) containing:
Expressed on activated CD4⁺ and CD8⁺ T cells, regulatory T cells (Tregs), and lymphoid/non-lymphoid cells .
Upregulated 24–72 hours post-T cell activation, dependent on CD28 co-stimulation .
T cell survival: Sustains T cell proliferation beyond 72 hours by upregulating anti-apoptotic proteins (Bcl-2, Bcl-xL) and survivin .
Cytokine modulation: Enhances IL-4 secretion in Th2 responses and promotes memory T cell development .
Treg activity:
Atherosclerosis: Anti-OX40L antibody treatment in LDLr⁻/⁻ mice reduced lesion size by 53% via suppressed Th2-mediated IgG1 responses and elevated anti-oxLDL IgM .
Cancer:
Reagent | Application | Catalog |
---|---|---|
Anti-OX40 PE-conjugated Ab | Flow cytometry (e.g., activated splenocytes) | FAB1256P |
OX40L-Fc fusion protein | Co-stimulation in vaccine studies | N/A |
Tnfrsf4⁻/⁻ mice:
Agonistic antibodies: Enhance vaccine efficacy (e.g., OVA/alum, gp96-Ig) by costimulating CD8⁺ T cells .
OX40-Ig fusion protein: Reduces cytokine storms in viral infections (e.g., H5N1) while preserving antiviral immunity .
TNFRSF4, also known as TNF Receptor Superfamily Member 4, is a co-stimulatory molecule found on T cells. It belongs to the TNF receptor superfamily. TNFRSF4 works in concert with other co-stimulatory molecules like CD28, CD40, CD30, CD27, and 4-1BB to regulate immune responses. This receptor plays a crucial role in the expansion and survival of antigen-specific T cells. When the T cell receptor (TCR) encounters antigens presented by antigen-presenting cells, and with the help of co-stimulation from CD40-CD40 Ligand and CD28-B7 interactions, TNFRSF4 expression increases on CD4+ and CD8+ T cells. Furthermore, TNFRSF4 influences the production of cytokines from various immune cells, including T cells, antigen-presenting cells, and natural killer cells. It also plays a role in modulating cytokine receptor signaling.
Recombinant TNFRSF4 from mice, produced in HEK293 cells, is a single-chain polypeptide with glycosylation. It encompasses amino acids 20 to 211, resulting in a protein of 435 amino acids with a molecular weight of 48.6 kDa. The TNFRSF4 protein is fused with a 243 amino acid hIgG-His tag at its C-terminus. Purification is achieved through proprietary chromatographic methods.
The TNFRSF4 protein solution has a concentration of 0.25 mg/ml and is prepared in Phosphate-Buffered Saline (pH 7.4) with 10% glycerol.
The purity is determined to be greater than 95.0% using SDS-PAGE analysis.
The ED50, measured as its binding affinity to Mouse OX40 Ligand/TNFSF4 in a functional ELISA, is less than or equal to 0.4 ug/ml.
Tumor necrosis factor receptor superfamily member 4, OX40 antigen, OX40L receptor, Txgp1, Tnfrsf4, tax-transcriptionally activated glycoprotein 1 receptor, TXGP1L, ACT35, Txgp, Ly-70, ACT3, OX4, CD134.
HEK293 cells.
DGSMVTARRL NCVKHTYPSG HKCCRECQPG HGMVSRCDHT RDTLCHPCET GFYNEAVNYD TCKQCTQCNH RSGSELKQNC TPTQDTVCRC RPGTQPRQDS GYKLGVDCVP CPPGHFSPGN NQACKPWTNC TLSGKQTRHP ASDSLDAVCE DRSLLATLLW ETQRPTFRPT TVQSTTVWPR TSELPSPPTL VTPEGPLEPK SCDKTHTCPP CPAPELLGGP SVFLFPPKPK DTLMISRTPE VTCVVVDVSH EDPEVKFNWY VDGVEVHNAK TKPREEQYNS TYRVVSVLTV LHQDWLNGKE YKCKVSNKAL PAPIEKTISK AKGQPREPQV YTLPPSRDEL TKNQVSLTCL VKGFYPSDIA VEWESNGQPE NNYKTTPPVL DSDGSFFLYS KLTVDKSRWQ QGNVFSCSVM HEALHNHYTQ KSLSLSPGKH HHHHH
Mouse OX40/TNFRSF4 is a 256 amino acid type I transmembrane precursor protein with a distinctive structure consisting of:
A 19 amino acid signal peptide
A 192 amino acid extracellular domain containing 4 TNFR-cysteine rich repeats
A 25 amino acid transmembrane domain
A 36 amino acid cytoplasmic region
The protein undergoes significant post-translational modifications, particularly glycosylation, which increases its apparent molecular weight from the predicted 24.2 kDa to approximately 48-55 kDa when analyzed by Bis-Tris PAGE . The extracellular domain (Val20-Pro211) represents the functional portion commonly used in recombinant protein studies .
Mouse OX40/TNFRSF4 shares approximately 63% amino acid sequence identity with its human counterpart and 90% with its rat counterpart . This relatively high conservation between species, particularly with rat, makes mouse models valuable for studying OX40 biology, though researchers should be aware of the differences when translating findings to human applications. The mouse protein contains the same functional domains as the human version, including the cysteine-rich repeats in the extracellular domain and the cytoplasmic signaling region, but species-specific variations can affect ligand binding kinetics and downstream signaling efficiency .
TNFRSF4 is primarily expressed on activated CD4+ T cells, appearing 24-72 hours after activation, rather than on resting T cells . It is also expressed on activated CD8+ T cells, though typically at lower levels than on CD4+ T cells . Importantly, OX40 is constitutively expressed on regulatory T cells (Tregs), where it enhances sensitivity to IL-2 and promotes Treg proliferation, though it has been shown to decrease their immunosuppressive activity on effector T cells . The absence of expression on resting T cells makes OX40 an attractive target for specifically modulating activated immune responses without broadly affecting the entire T cell compartment .
Several validated methods are available for detecting mouse TNFRSF4 expression, each with specific applications:
Flow Cytometry: The gold standard for cellular expression analysis, using PE-conjugated antibodies such as Goat Anti-Mouse OX40/TNFRSF4. Researchers typically use activated mouse splenocytes stained with Rat Anti-Mouse CD3 APC-conjugated Monoclonal Antibody along with anti-OX40 antibodies to identify OX40-expressing T cell populations . This method allows for multi-parameter analysis to correlate OX40 expression with other markers.
ELISA: Sandwich ELISA kits provide quantitative measurement of mouse OX40 in various sample types including cell lysates, culture supernatants, plasma, serum, and tissue homogenates. These assays demonstrate high precision with coefficient of variation (CV) values typically between 3.4% and 6.9% .
Western Blot: Useful for protein size confirmation and semi-quantitative analysis, particularly with recombinant proteins. Western blotting can confirm the presence of both monomeric and potential multimeric forms of OX40 .
Evaluating OX40-OX40L interactions in mouse models can be approached through several complementary methods:
Binding Assays: Immobilized Mouse OX40 Ligand with hFc Tag (5μg/ml) can be used in plate-based assays to generate dose-response curves with biotinylated Mouse OX40 (His-tagged). The EC50 of this interaction has been measured at approximately 1.36 μg/ml by ELISA . Similarly, immobilized OX40 His at 2 μg/mL can bind Biotin-OX40L His with a linear range of 1.22-19.53 ng/mL .
Functional Assays: Researchers can use anti-OX40 antibodies with agonist activity to stimulate OX40 signaling. The ED50 for this effect typically ranges from 0.4-1.2 μg/mL . These assays can measure downstream effects such as T cell proliferation, cytokine production, or survival.
In vivo Models: OX40-OX40L interactions can be studied through knockout models or through blocking/stimulating antibodies administered to mice. These approaches allow for assessment of physiological relevance in immune responses, inflammation, or disease models .
When working with OX40/TNFRSF4 knockout (KO) mouse models, researchers should consider several important factors:
Phenotypic Characterization: TNFRSF4 KO mice show specific defects in CD4+ T cell responses, which should be thoroughly characterized before using these models for specific disease studies . This includes assessing basal T cell populations, activation status, and responses to various stimuli.
Compensatory Mechanisms: As with many immune receptor knockouts, compensatory upregulation of related costimulatory molecules may occur. Researchers should evaluate the expression of other TNFR family members and costimulatory molecules to account for potential compensatory effects that might confound experimental interpretation .
Strain Background: The genetic background of the knockout mice can significantly influence the phenotype. Researchers should document and consider the strain background when interpreting results and comparing data across studies .
Conditional vs. Global Knockouts: Consider whether a global or conditional knockout is more appropriate for your research question. Cell-specific deletion using Cre-loxP systems can help distinguish direct from indirect effects of OX40 deletion .
TNFRSF4 plays complex roles in mouse models of autoimmunity and inflammation:
Allergic Airway Inflammation: OX40-OX40L signaling has been implicated in the development and persistence of allergic airway inflammation in mouse models of asthma . Blocking this pathway can reduce inflammatory cell infiltration and cytokine production in the airways.
Autoimmune Diseases: Studies show OX40-OX40L interactions contribute to pathogenesis in multiple autoimmune disease models including experimental autoimmune encephalomyelitis (EAE, a multiple sclerosis model), collagen-induced arthritis, and inflammatory bowel disease models. In these contexts, OX40 signaling typically promotes the expansion and survival of autoreactive T cells .
Graft-versus-Host Disease (GVHD): The OX40-OX40L pathway contributes to GVHD pathogenesis in mouse models of allogeneic transplantation. Interrupting this pathway can reduce disease severity by limiting donor T cell expansion and inflammatory cytokine production .
Regulatory T Cell Function: Though OX40 is constitutively expressed on Tregs and enhances their sensitivity to IL-2 (promoting proliferation), it has also been shown to decrease their immunosuppressive activity on effector T cells . This dual effect creates a complex role in autoimmune regulation that depends on the specific disease context.
Research in mouse cancer models has revealed TNFRSF4 as a promising immunotherapeutic target:
Anti-tumor Immunity: OX40 agonists have been shown to augment anti-tumor immunity in several mouse cancer models by enhancing T cell expansion, survival, and effector functions . This approach helps overcome T cell exhaustion and promotes durable anti-tumor responses.
Combination Therapies: Studies indicate that combining OX40 agonists with other immune checkpoint modulators (such as PD-1/PD-L1 blockers) or with conventional therapies (radiation, chemotherapy) often produces synergistic effects in mouse tumor models . These combinations can convert immunologically "cold" tumors to "hot" tumors more susceptible to immune attack.
Memory T Cell Generation: OX40 stimulation enhances the generation of memory T cells in mouse models, which is critical for long-term tumor control and prevention of recurrence . This aspect is particularly important for developing durable cancer immunotherapies.
Tumor Microenvironment Modulation: Beyond direct T cell effects, OX40 engagement can reshape the tumor microenvironment by altering cytokine profiles and reducing immunosuppressive cell populations in mouse models .
OX40 agonists and antagonists demonstrate distinct immunomodulatory effects in mouse models:
Agonists:
Enhance T cell survival and expansion, particularly of antigen-specific CD4+ and CD8+ T cells
Increase cytokine production, especially Th1 and Th2 cytokines depending on the context
Promote memory T cell generation and long-term immunity
Typical ED50 for agonist antibodies ranges from 0.4-1.2 μg/mL in functional assays
Can diminish Treg suppressive function while potentially expanding Treg populations
Antagonists:
Reduce inflammatory responses in autoimmune and allergic disease models
Decrease T cell-mediated tissue damage in graft rejection models
Limit pathogenic T cell expansion and cytokine production
Particularly effective at preventing costimulatory signals during initial T cell activation
Help maintain tolerance in models of autoimmunity
The choice between agonist and antagonist approaches depends on the desired outcome: enhancing immunity (cancer, infections) or suppressing immunity (autoimmunity, transplantation) .
Mouse TNFRSF4 engagement activates several key signaling pathways:
TRAF-Mediated Signaling: Upon OX40L binding, OX40 recruits TNFR-associated factors (TRAFs), particularly TRAF2, TRAF3, and TRAF5, to form a TCR-independent signaling complex . This recruitment is mediated by the cytoplasmic QEE motif present in the mouse OX40 protein.
NF-κB Pathway: A primary downstream effect of OX40 engagement is activation of both canonical and non-canonical NF-κB pathways. One component of the signaling complex, PKCθ, is particularly important for NF-κB activation . This leads to expression of survival factors and inflammatory mediators.
PI3K/Akt Pathway: OX40 signaling enhances the PI3K/Akt pathway, which can directly augment TCR signaling. This pathway promotes T cell survival by upregulating anti-apoptotic molecules like Bcl-2 and Bcl-xL .
MAPK Pathways: OX40 activates multiple MAPK pathways including p38, ERK, and JNK, contributing to T cell proliferation, differentiation, and cytokine production.
Understanding these pathways is crucial for developing targeted immunomodulatory strategies and predicting potential off-target effects of OX40-directed therapies .
Mouse models have provided critical insights for therapeutic targeting of TNFRSF4 in human disease:
Translational Considerations: Despite the 63% sequence homology between mouse and human OX40 , researchers must carefully interpret mouse model results when translating to human applications. Differences in expression patterns, binding affinities, and downstream signaling can affect therapeutic outcomes.
Predictive Biomarkers: Studies in mouse models have helped identify potential biomarkers that predict response to OX40-targeted therapies, including baseline OX40 expression levels on tumor-infiltrating lymphocytes and the composition of the tumor microenvironment .
Toxicity Profiles: Mouse models have revealed potential on-target toxicities of OX40 modulation, particularly autoimmune-like phenomena. These observations have informed safety monitoring in human clinical trials of OX40-targeted agents .
Combination Strategies: Mouse studies demonstrating synergy between OX40 modulation and other immunotherapies (checkpoint inhibitors, cytokine therapies, conventional treatments) have directly informed the design of human clinical trials testing similar combinations .
Dosing and Scheduling: Mouse pharmacodynamic studies have provided insights into optimal dosing schedules for OX40-targeted therapies, revealing the importance of timing relative to antigen exposure and other treatments .
Researchers face several methodological challenges when studying TNFRSF4 in complex mouse disease models:
Temporal Expression Dynamics: Since OX40 is only expressed 24-72 hours after T cell activation , experimental timing is critical. Researchers must carefully design sampling timepoints to capture the relevant biology, particularly in acute disease models.
Distinguishing Cell-Type Specific Effects: OX40 is expressed on multiple T cell subsets including conventional CD4+, CD8+, and regulatory T cells . Cell-specific conditional knockout models or adoptive transfer approaches may be necessary to dissect the relative contribution of OX40 signaling in each population.
Ligand Availability: The regulated expression of OX40L on antigen-presenting cells creates another layer of complexity . Researchers must consider both receptor and ligand expression patterns when interpreting results from disease models.
Technical Detection Limitations: Flow cytometric detection of OX40 requires careful optimization of staining protocols and appropriate controls, as demonstrated in the protocols for staining membrane-associated proteins . Similarly, ELISA measurements should account for sample type-specific matrix effects, as indicated by the validation data showing different detection ranges depending on sample type .
Proper storage and handling of mouse TNFRSF4 reagents is critical for maintaining their functionality:
Recombinant Proteins: Lyophilized recombinant mouse OX40 proteins should be stored at -20°C to -80°C for up to 12 months from the date of receipt. After reconstitution, aliquoting is strongly recommended to minimize freeze-thaw cycles, with reconstituted protein stable at -80°C for approximately 3 months . Reconstitution to concentrations exceeding 100 μg/ml is typically recommended for optimal stability .
Antibodies: For anti-mouse OX40/TNFRSF4 antibodies, storage at -20°C to -70°C in a manual defrost freezer is recommended, with stability for up to 12 months from the date of receipt. Once reconstituted, antibodies maintain stability for approximately 1 month at 2-8°C under sterile conditions, or 6 months at -20°C to -70°C .
ELISA Kits: Components must be stored according to manufacturer instructions, typically with detection antibodies and standards requiring -20°C storage, while plate components and buffers can often be stored at 2-8°C .
Avoid Repeated Freeze-Thaw Cycles: This is particularly important for all protein reagents, as repeated freezing and thawing can lead to protein denaturation, aggregation, and loss of biological activity .
Thorough validation of mouse TNFRSF4 antibodies is essential for generating reliable research data:
Positive and Negative Controls: Always include appropriate positive controls (activated mouse T cells or recombinant OX40 protein) and negative controls (naive T cells or isotype-matched control antibodies). For flow cytometry, comparing staining patterns between activated splenocytes and appropriate isotype controls is essential, as demonstrated in the protocols using Goat Anti-Mouse OX40/TNFRSF4 PE-conjugated Antibody alongside Normal Goat IgG Phycoerythrin Control .
Cross-Reactivity Testing: Evaluate potential cross-reactivity with related TNFR family members. For example, some anti-mouse OX40 antibodies show less than 2% cross-reactivity with related proteins like mouse EDAR and mouse 4-1BB in direct ELISAs and Western blots .
Application-Specific Validation: Validate antibodies specifically for the intended application. An antibody that works well for Western blotting may not be suitable for flow cytometry or functional assays. For example, antibodies used for agonist activity should be validated specifically for this purpose, with typical ED50 values in the range of 0.4-1.2 μg/mL .
Titration Experiments: Perform antibody titrations to determine optimal concentrations for each specific application and cell type. This is particularly important for flow cytometry and functional assays where antibody concentration can significantly impact results .
Measuring soluble mouse TNFRSF4 in biological samples requires attention to several technical considerations:
Sample Type Variability: Different biological sample types (serum, plasma, cell culture supernatants, tissue homogenates) can affect assay performance. Validation data for mouse OX40 ELISA shows variation in detection parameters across sample types, with precision (CV) values ranging from 3.4% to 6.9% depending on the sample matrix .
Sample Collection and Processing: Standardize collection procedures, including anticoagulant use for plasma (EDTA or heparin), processing times, and storage conditions. For example, ELISA kits have been validated for both EDTA and heparin plasma samples .
Assay Sensitivity and Range: Choose methods with appropriate sensitivity for the expected concentration range in your samples. Mouse OX40 ELISA kits typically have detection ranges that can measure from approximately 224 pg/mL to 1463 pg/mL .
Interference Testing: Consider potential interference from heterophilic antibodies, rheumatoid factor, or other components in complex biological samples that might affect measurement accuracy .
Standard Curve Optimization: Prepare standards in the same matrix as samples when possible, or use appropriate diluents that mimic the sample matrix to minimize matrix effects .
Validation Across Sample Types: When transitioning to a new sample type, perform spike-and-recovery experiments to validate the assay in the specific matrix of interest .
TNFRSF4 is a type I transmembrane protein that consists of an extracellular domain, a transmembrane domain, and a cytoplasmic domain. The extracellular domain is responsible for ligand binding, while the cytoplasmic domain is involved in signal transduction. The primary ligand for TNFRSF4 is OX40L (OX40 Ligand), which is also a member of the TNF superfamily .
The interaction between TNFRSF4 and OX40L is essential for the activation and survival of T cells. This interaction promotes the proliferation and differentiation of T cells, enhances the production of cytokines, and supports the formation of memory T cells. Additionally, TNFRSF4 signaling has been shown to inhibit apoptosis by upregulating anti-apoptotic proteins such as BCL2 and BCL2L1 .
TNFRSF4 is predominantly expressed on activated T cells, particularly CD4+ T cells. It plays a critical role in the regulation of T cell-mediated immune responses. The activation of TNFRSF4 signaling enhances the survival and function of effector T cells, which are responsible for eliminating infected or malignant cells. Moreover, TNFRSF4 signaling is involved in the regulation of T cell-dependent B cell responses, including the production of antibodies .
Recombinant mouse TNFRSF4 is a laboratory-produced version of the protein that is used in various research applications. It is typically produced using recombinant DNA technology, where the gene encoding TNFRSF4 is inserted into an expression vector and introduced into a host cell system, such as bacteria or mammalian cells. The host cells then produce the TNFRSF4 protein, which can be purified and used for experimental purposes .
Recombinant mouse TNFRSF4 is used in studies to investigate the molecular mechanisms of TNFRSF4 signaling, its role in immune responses, and its potential therapeutic applications. For example, researchers use recombinant TNFRSF4 to study its effects on T cell activation, proliferation, and survival. Additionally, recombinant TNFRSF4 is used in the development of therapeutic strategies targeting TNFRSF4 signaling pathways for the treatment of autoimmune diseases, cancer, and other immune-related disorders .
The TNFRSF4-OX40L signaling axis has significant clinical implications. Modulating this pathway has the potential to enhance immune responses against tumors and infections, as well as to regulate autoimmune and inflammatory diseases. Therapeutic agents targeting TNFRSF4 or OX40L are being developed and tested in clinical trials for their efficacy in treating various conditions, including cancer and autoimmune diseases .