IL 12p75 Antibody

Interleukin-12 p75, Rat Anti-Mouse
Shipped with Ice Packs
In Stock

Description

Immune Regulation

  • T Cell-Dependent IL-12p75 Production: Antigen-activated T cells induce dendritic cells (DCs) to secrete IL-12p75 via CD40 ligand (CD40L), IL-4, and GM-CSF synergism, independent of IFN-γ .

  • Autoimmunity: IL-12p75 restoration in IL-12p40-deficient mice reinstates myocarditis susceptibility, confirming its role in Th1-mediated pathology .

Therapeutic Applications

  • Cancer Immunotherapy: IL-12p75 antibodies modulate tumor microenvironments by reducing immunosuppressive factors (e.g., IL-10, TGF-β) and enhancing CD8+ T cell cytotoxicity .

  • Inflammation Control: Anti-IL-12p75 antibodies mitigate autoimmune responses in models of sepsis and delayed-type hypersensitivity .

Table 1: IL-12p75 Antibody Efficacy in Disease Models

Study ModelAntibody CloneOutcomeReference
Autoimmune MyocarditisR2-9A5Reduced lesion severity (0 vs. 1.8 mean grade in WT)
Tumor MicroenvironmentR2-9A5Decreased MDSC infiltration, improved survival
TLR-Stimulated DCsMM121Blocked IL-12p75 secretion post-poly(I:C)/R848 synergy

Technical Considerations for Antibody Use

  • Storage: Lyophilized antibodies remain stable at 4°C; reconstituted solutions avoid freeze-thaw cycles .

  • Endotoxin Levels: Commercial clones exhibit ≤10 EU/mg, ensuring minimal experimental interference .

  • Cross-Reactivity: No reactivity with human IL-12p70 confirmed for murine-specific clones .

Emerging Applications and Future Directions

  • Imaging: Anti-IL-12p75 antibodies conjugated with radiotracers enable positron emission tomography (PET) to visualize inflammatory microenvironments in tumors .

  • Combination Therapies: Pairing IL-12p75 blockade with checkpoint inhibitors (e.g., anti-PD-1) shows promise in enhancing antitumor immunity .

Product Specs

Introduction
Interleukin-12 (IL-12) is a heterodimeric cytokine with a crucial role in cell-mediated immunity. It acts as a potent inducer of interferon-gamma (IFN-γ) production by T cells and natural killer (NK) cells, key players in the immune response against intracellular pathogens and tumor cells. Moreover, IL-12 drives the differentiation of naive T helper cells into the Th1 subset, which are characterized by their ability to secrete IFN-γ and promote cellular immune responses.
Formulation
The antibody is provided at a concentration of 1 milligram per milliliter (mg/ml) in phosphate-buffered saline (PBS) after reconstitution.
Shipping Conditions
To ensure stability during transportation, the antibody is shipped in lyophilized form at ambient temperature.
Storage Procedures
For long-term storage, it is recommended to keep the lyophilized antibody at 4 degrees Celsius in a dry environment. After reconstitution, if not used within one month, aliquot the antibody solution and store it at -20 degrees Celsius to preserve its activity.
Solubility
To reconstitute the antibody, add sterile water (H2O) to the vial. Gently mix the solution by swirling or inverting the vial. Rinse the sides of the vial to ensure complete dissolution. Allow 30-60 seconds for the antibody to fully reconstitute before use.
Applications
This antibody is suitable for a variety of applications, including direct enzyme-linked immunosorbent assay (ELISA), Western blotting, and immunoprecipitation.
Titer
In a direct ELISA, a 1:5,000 dilution of the antibody will result in an optical density (O.D.) of 0.5 when using an alkaline phosphatase-conjugated rabbit anti-mouse immunoglobulin G (IgG) secondary antibody from Jackson Laboratories.
Note
This antibody exhibits specificity for the intact IL-12 molecule. It does not bind to denatured forms of the p40 or p35 subunits individually.
Purification Method
Ion exchange.
Type
Rat Anti Mouse Monoclonal.
Clone
NYRmIL-12p75.
Immunogen
r.Murine IL-12 p75.
Ig Subclass
Rat IgG2b.

Q&A

What is the structural and functional difference between IL-12p75 and IL-12p40?

IL-12p75 (also known as IL-12p70) is a heterodimeric cytokine composed of two different gene products: the p35 and p40 subunits . While researchers often use the terms IL-12 and IL-12p40 interchangeably, this is scientifically inaccurate. The p40 subunit can exist independently and is typically secreted earlier and in higher quantities than the complete p75 heterodimer . The p40 subunit appears to be induced as a T-independent response by antigen-presenting cells during early host-pathogen interactions, while the complete IL-12p75 heterodimer is produced later and requires T-dependent signals . Functionally, IL-12p75 plays a critical role in promoting IFN-γ production by T cells and NK cells, enhancing cell-mediated immunity against intracellular pathogens and tumors .

How does the production of IL-12p75 differ between naïve and antigen-activated T cell interactions with dendritic cells?

Research demonstrates a significant difference in IL-12p75 induction between naïve and antigen-activated T cells. When dendritic cells (DCs) are cocultured with antigen-activated T cells, substantial IL-12p75 production occurs, whereas naïve T cells induce minimal IL-12p75 . This distinction is critical for experimental design. In experimental systems, antigen-activated but not naïve T cells provide necessary signals for IL-12p75 production from DCs . These findings challenge the conventional paradigm that suggests naïve T cell interactions with DCs lead to IL-12p75 secretion as an initiating event in TH1 differentiation .

What are the optimal detection methods for distinguishing between IL-12p75 and free p40 subunits?

Distinguishing between IL-12p75 heterodimer and free p40 subunits requires careful antibody selection and assay design. The most reliable approach uses a sandwich ELISA with a capture antibody targeting one subunit (typically p35) and a detection antibody recognizing the other (p40) . Commercial IL-12p70 monoclonal antibodies like clone C17-8 have been validated for specific detection of the heterodimer in techniques such as ELISA and Western blot . When designing experiments, researchers should:

Detection MethodAdvantagesLimitationsBest Practices
Sandwich ELISAHigh specificity for heterodimerLower sensitivity for detecting small amountsUse validated antibody pairs specific for p35-p40 complex
Flow CytometrySingle-cell resolution of productionCannot distinguish secreted formsInclude intracellular staining controls
Western BlotDistinguishes by molecular weightLess quantitativeRun under non-reducing conditions to maintain heterodimer
Bioassay (IFN-γ induction)Confirms biological activityOther cytokines may influence resultsInclude neutralizing antibodies as controls

It's critical to remember that measurements of p40 should never be interpreted as measurements of the intact p75 heterodimer, as p40 is typically produced in excess and has distinct biological functions .

How should experiments be designed to study the temporal relationship between IL-12p75 and IFN-γ production?

To properly investigate the temporal relationship between IL-12p75 and IFN-γ production, researchers should employ time-course experiments with multiple controls. Research has revealed that during cognate interactions between antigen-activated T cells and DCs, IL-12p75 production follows specific kinetics, typically peaking at 6-24 hours post-coculture and declining thereafter . In contrast, p40 production follows different kinetics, remaining elevated throughout the experimental period .

Experimental design should include:

  • Time-course sampling from 0-120 hours post-stimulation

  • Parallel measurement of both IL-12p75 and IFN-γ from the same samples

  • Inclusion of both wild-type and IFN-γKO T cells to determine IFN-γ dependency

  • Comparison between "resting" and LPS-activated DCs

  • Controls with and without cognate peptide antigen

  • Measurement of both p40 and p75 to track their relative kinetics

This approach will help resolve the apparent paradox regarding whether IL-12p75 drives IFN-γ production or vice versa. Evidence suggests that during cognate interactions between antigen-activated T cells and DCs, IL-12p75 can be produced through IFN-γ-independent pathways, challenging the conventional model .

What controls are essential when using IL-12p75 antibodies in immunological assays?

When using IL-12p75 antibodies in research, proper controls are essential for valid interpretation. Critical controls include:

  • Specificity controls: Include recombinant IL-12p75, p40 alone, and p35 alone to confirm antibody specificity for the heterodimer

  • Genetic validation: When possible, include samples from IL-12p35-/- or IL-12p40-/- mice to validate antibody specificity

  • Cell type controls: Compare IL-12p75 detection between cell types known to produce (DCs, macrophages) and not produce (most lymphocytes) the cytokine

  • Stimulation controls: Include positive controls (LPS-activated DCs cocultured with activated T cells) and negative controls (unstimulated cells)

  • Isotype controls: Include matched isotype antibodies to control for non-specific binding

  • Biological activity validation: Confirm that detected IL-12p75 correlates with expected biological activity (e.g., induction of IFN-γ production)

These controls are particularly important given the heterodimeric nature of IL-12p75 and the potential for cross-reactivity with other IL-12 family members that share subunits.

How do I interpret contradictory results regarding IL-12p75 dependency on IFN-γ?

Contradictory results regarding the relationship between IL-12p75 and IFN-γ often reflect the complex regulatory networks and context-dependent production of these cytokines. Research demonstrates that while IFN-γ can enhance IL-12p75 production in response to TLR agonists like LPS, IL-12p75 can also be produced through IFN-γ-independent pathways during cognate interactions between antigen-bearing DCs and antigen-specific T cells .

When interpreting seemingly contradictory data:

  • Consider the activation state of T cells: Activated T cells, but not naïve T cells, can induce IL-12p75 from DCs even in the absence of IFN-γ

  • Examine the temporal relationship: Secretion of IL-12p75 follows specific kinetics, typically peaking at 6-24 hours post-coculture with antigen-activated T cells

  • Analyze the stimulation conditions: TLR-mediated signals alone (e.g., LPS) are typically inadequate to induce robust IL-12p75 secretion without IFN-γ priming, while T cell interactions can provide IFN-γ-independent signals

  • Consider experimental methodology: Different detection methods may have varying sensitivity for IL-12p75 versus p40

Research suggests these contradictions may reflect different pathways of IL-12p75 production: an early IFN-γ-dependent pathway involving innate stimuli, and a later IFN-γ-independent pathway involving antigen-specific T cell interactions .

Why might I observe high levels of IL-12p40 but low or undetectable IL-12p75 in my experiments?

This common observation reflects the differential regulation of p40 and p35 subunits. Several factors may explain this pattern:

  • Differential gene regulation: The genes encoding p40 and p35 are differentially regulated, with p40 typically produced in excess of p35

  • Timing: p40 secretion precedes p75 production in response to pathogens, so early sampling may detect p40 but not p75

  • Stimulus requirements: TLR agonists alone strongly induce p40 but require additional signals (often from activated T cells) for robust p75 production

  • Cell type differences: Some cells may primarily produce p40 without substantial p35 production

  • T cell dependency: IL-12p75 production often requires signals from antigen-activated (not naïve) T cells, while p40 production is less dependent on these signals

To troubleshoot this issue, researchers can:

  • Extend the time course of experiments to capture later p75 production

  • Co-culture with antigen-activated T cells to provide necessary signals

  • Add recombinant IFN-γ to enhance p75 production

  • Examine p35 mRNA expression to determine if transcriptional regulation is limiting

Understanding this differential regulation is essential for experimental design and data interpretation .

How can I differentiate technical issues from biological findings when IL-12p75 detection is inconsistent?

Inconsistent IL-12p75 detection can stem from either technical limitations or genuine biological variability. To differentiate between these possibilities:

  • Technical validation:

    • Test multiple IL-12p75-specific antibody clones or detection kits

    • Include positive controls (recombinant IL-12p75) at known concentrations

    • Verify antibody specificity using samples from IL-12p35-/- or IL-12p40-/- mice

    • Compare different detection methods (ELISA vs. bioassay vs. flow cytometry)

  • Biological considerations:

    • IL-12p75 production is highly regulated and context-dependent

    • Production requires both p35 and p40 expression in either the same cell or through the two-cell model

    • IL-12p75 production often requires specific signals from activated T cells

    • Production follows distinct kinetics, typically peaking at 6-24 hours post-coculture

    • IL-12p75 may be consumed rapidly in biological systems through receptor binding

  • Experimental design improvements:

    • Include time-course analysis to capture peak production

    • Use multiple detection methods in parallel

    • Include appropriate positive controls (LPS-activated DCs cocultured with activated T cells)

    • Measure both subunits individually alongside the heterodimer

Distinguishing technical from biological factors requires systematic validation and controls that account for the complex biology of IL-12p75 .

How can I design experiments to investigate the immunological synapse-dependent IL-12p75 production?

Investigating immunological synapse-dependent IL-12p75 production requires sophisticated experimental approaches:

  • Co-culture systems: Establish DC-T cell co-cultures with:

    • Antigen-specific T cells (e.g., TCR transgenic 5C7 T cells)

    • Antigen-bearing DCs with defined activation states (resting vs. LPS-activated)

    • Specific peptide concentrations (e.g., 0.1μM MCC peptide)

  • Synapse visualization and manipulation:

    • Use live-cell imaging to visualize synapse formation

    • Apply microfabricated surfaces with defined ligand patterns

    • Employ super-resolution microscopy to examine protein localization

  • Genetic approaches:

    • Compare wild-type T cells with signaling mutants affecting synapse formation

    • Use IFN-γKO T cells to differentiate IFN-γ-dependent from synapse-dependent signals

    • Generate reporter systems for real-time monitoring of IL-12 subunit expression

  • Pharmacological interventions:

    • Apply inhibitors of cytoskeletal rearrangement to disrupt synapse formation

    • Use specific signaling pathway inhibitors to identify critical nodes

  • Temporal analysis:

    • Conduct detailed time-course experiments (6-120 hours)

    • Compare kinetics between different experimental conditions

    • Simultaneously measure IL-12p75, p40, and IFN-γ from the same samples

This multifaceted approach will help elucidate how immunological synapse formation contributes to IL-12p75 production independent of soluble mediators like IFN-γ.

What approaches can be used to study the functional differences between canonical and two-cell model-derived IL-12p75?

To investigate functional differences between canonical (single-cell) and two-cell model-derived IL-12p75, researchers can employ these sophisticated approaches:

  • Genetic systems:

    • Generate chimeric mice containing mixtures of cells that express either IL-12p40 or IL-12p35, but not both

    • Create conditional knockout models to selectively delete p35 or p40 in specific cell populations

  • In vitro models:

    • Establish transwell co-culture systems separating p40-producing and p35-producing cells

    • Use CRISPR-engineered cell lines expressing only one subunit

    • Apply recombinant p40 to p35-expressing cells to reconstitute the two-cell model

  • Functional readouts:

    • Compare T cell differentiation (flow cytometry for IFN-γ, T-bet)

    • Measure proliferation through tritiated thymidine incorporation or CFSE dilution

    • Assess pathogen control in infection models

  • Molecular characterization:

    • Analyze receptor binding kinetics and signaling pathway activation

    • Perform structural studies to identify potential conformational differences

    • Examine stability and half-life of the heterodimer from different sources

  • In vivo validation:

    • Test protective immunity against model pathogens like Leishmania major

    • Compare local versus systemic immune responses

    • Assess trafficking of immune cells to infection sites

These approaches will help determine whether IL-12p75 formed through different mechanisms exhibits distinct functional properties, which has important implications for therapeutic targeting .

How can IL-12p75 antibodies be used to investigate IFN-γ-independent pathways in T cell differentiation?

IL-12p75 antibodies are valuable tools for investigating IFN-γ-independent pathways in T cell differentiation. Advanced experimental approaches include:

  • Genetic models with antibody validation:

    • Compare wild-type and IFN-γKO T cells in co-culture systems with DCs

    • Use IL-12p75-specific antibodies to neutralize or detect the cytokine

    • Track T cell differentiation markers (T-bet, STAT4 phosphorylation)

  • Temporal dissection:

    • Conduct detailed time-course experiments (6-120 hours)

    • Add neutralizing antibodies at different time points to determine critical windows

    • Compare kinetics of IL-12p75 production between WT and IFN-γKO systems

  • Cell-specific approaches:

    • Sort DC populations based on IL-12p75 production capacity

    • Use adoptive transfer of IL-12R-deficient T cells with IL-12p75 detection

    • Apply single-cell analysis techniques with antibody-based detection

  • Pathway analysis:

    • Combine IL-12p75 antibodies with inhibitors of candidate signaling pathways

    • Use phospho-flow cytometry to track signaling events downstream of TCR/CD28

    • Perform transcriptomic analysis of T cells under IL-12 stimulation with/without IFN-γ

Research shows that antigen-activated T cells induce IL-12p75 from DCs even in the absence of IFN-γ, challenging the conventional model of IFN-γ dependency . IL-12p75 antibodies enable precise detection and neutralization to dissect these complex regulatory networks.

What research questions remain regarding the relationship between IL-12p75 and IFN-γ in immune regulation?

Despite decades of research, several critical questions remain about IL-12p75 and IFN-γ regulation:

  • Temporal paradox: If IL-12p75 induces IFN-γ production but also requires IFN-γ for its own production, what initiates this cycle in vivo? Research suggests antigen-activated T cells may provide IFN-γ-independent signals for IL-12p75 production, but the molecular nature of these signals remains incompletely understood .

  • Spatial regulation: How is the two-cell model of IL-12 formation regulated in complex tissue microenvironments? The discovery that p40 from hematopoietic cells can combine with p35 from non-hematopoietic cells raises questions about how this process is spatially organized and regulated .

  • Subunit regulation: What factors control the differential expression of p40 and p35 subunits? While p40 is often produced in excess, the limiting factors for p35 expression remain unclear .

  • T cell heterogeneity: How do different T cell subsets (naïve, effector, memory) distinctly regulate IL-12p75 production from APCs? Research indicates activated but not naïve T cells induce IL-12p75, but the molecular basis for this difference requires further investigation .

  • Clinical translation: How can understanding of IL-12p75 biology inform better therapeutic interventions for autoimmune diseases, infections, and cancer?

Addressing these questions will require integrating advanced technologies with careful experimental design to dissect these complex regulatory networks.

What are the emerging technologies for studying IL-12p75 in tissue microenvironments?

Cutting-edge technologies are transforming our ability to study IL-12p75 in complex tissue contexts:

  • Spatial multi-omics:

    • Spatial transcriptomics to map p35 and p40 expression patterns within tissues

    • Multiplexed ion beam imaging (MIBI) to simultaneously visualize multiple proteins

    • Digital spatial profiling for quantitative protein measurement with spatial context

  • Advanced microscopy:

    • Intravital two-photon microscopy with fluorescent IL-12p75 antibodies

    • Expansion microscopy for nanoscale resolution of cytokine-producing cells

    • Lattice light-sheet microscopy for rapid 3D imaging with minimal phototoxicity

  • Single-cell technologies:

    • Single-cell RNA-seq with protein detection (CITE-seq) including IL-12p75

    • Mass cytometry (CyTOF) with IL-12 subunit-specific antibodies

    • Single-cell secretion assays (e.g., microfluidic platforms)

  • In situ detection:

    • Proximity ligation assays to visualize p35-p40 interactions in tissue sections

    • Highly multiplexed immunofluorescence for simultaneous detection of IL-12p75, cellular sources, and responding cells

    • RNA-protein correlation with combined RNAscope and immunofluorescence

  • Engineered reporter systems:

    • Dual-reporter mice for simultaneous visualization of p35 and p40 expression

    • Optogenetic systems for controlled expression of IL-12 subunits

These technologies will help resolve how the recently discovered two-cell model of IL-12 formation operates in vivo and advance our understanding of spatial cytokine regulation .

How might the discovery of the two-cell model of IL-12 formation change therapeutic approaches?

The paradigm-shifting discovery of the two-cell model of IL-12 formation has profound implications for therapeutic interventions:

  • Targeted therapy refinement:

    • Current therapeutics targeting p40 (e.g., ustekinumab) affect both single-cell and two-cell IL-12 formation

    • New approaches could selectively target cell type-specific production of individual subunits

    • Tissue-specific targeting might block p35 in non-hematopoietic cells while preserving protective immunity

  • Enhanced vaccine strategies:

    • Understanding the two-cell mechanism could improve adjuvant design

    • Vaccines might be engineered to optimize spatial distribution of p40 and p35-producing cells

    • Controlled delivery of individual subunits could enhance desired immune responses

  • Novel biomarkers:

    • Measuring the ratio of canonical versus two-cell IL-12 might better predict disease progression or treatment response

    • Tissue-specific analysis of subunit expression could identify new disease endotypes

  • Infectious disease applications:

    • The two-cell mechanism appears particularly important for limiting pathogen dissemination to tissues distant from primary infection sites

    • This could inform new approaches to prevent systemic spread of localized infections

  • Autoimmunity interventions:

    • Dysregulation of the two-cell model might contribute to specific autoimmune pathologies

    • Cell type-specific therapeutic targeting could provide more selective immunomodulation

This evolving understanding challenges conventional approaches to IL-12 targeting and opens new avenues for precision immunotherapy that considers the spatial organization of cytokine production .

Product Science Overview

Role and Function

IL-12 plays a crucial role in the regulation of cell-mediated immune responses. It is a potent inducer of Th1 responses, leading to the production of interferon-gamma (IFN-γ) and interleukin-2 (IL-2) . These responses are essential for the body’s defense against intracellular pathogens and for the development of long-lasting immunity.

Rat Anti-Mouse IL-12 p75 Antibody

The Rat Anti-Mouse IL-12 p75 antibody, specifically the R2-9A5 monoclonal antibody, is designed to react with mouse IL-12 . This antibody has been shown to neutralize the biological effects of IL-12 when administered in vivo . It is commonly used in research to study the immune response and to investigate the role of IL-12 in various diseases.

Applications

The Rat Anti-Mouse IL-12 p75 antibody is used in various applications, including:

  • In vivo IL-12p75 neutralization: This involves administering the antibody to live animals to study the effects of IL-12 neutralization on immune responses .
  • ELISA: The antibody can be used in enzyme-linked immunosorbent assays to detect and quantify IL-12 levels in biological samples .
Preparation and Purification

The antibody is purified from cell culture supernatant in an animal-free facility using Protein G affinity chromatography . It is formulated in phosphate-buffered saline (PBS) at pH 8.0 and contains no stabilizers or preservatives . The endotoxin level is kept below 2 EU/mg, as determined by the Limulus Amebocyte Lysate (LAL) gel clotting assay .

Storage

The antibody solution should be stored at the stock concentration at 4°C and should not be frozen to maintain its stability and efficacy .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2024 Thebiotek. All Rights Reserved.