IL 12p40 Antibody

Interleukin-12 p40, Rat Anti-Mouse
Shipped with Ice Packs
In Stock

Product Specs

Introduction
Active IL-12, a p70 disulphide-linked dimer comprising p35 and p40 subunits, is a pleiotropic cytokine mainly produced by antigen-presenting cells. It exhibits diverse effects on T lymphocytes and natural killer cells, encompassing the stimulation of cytotoxicity, proliferation, cytokine production, and differentiation into the Th1 subset.
Formulation
This product is provided lyophilized.
Shipping Conditions
The antibody is shipped in lyophilized form at room temperature.
Storage Procedures
For long-term storage, keep the lyophilized antibody at 4°C in a dry environment. Once reconstituted, aliquot and store at -20°C if not intended for use within one month.
Solubility
To reconstitute, add sterile water to the vial. Mix gently by swirling, ensuring the solution reaches the sides of the vial. Allow 30-60 seconds for complete reconstitution before use.
Applications
This antibody is suitable for use in Direct ELISA, Western Blot, and Immunoprecipitation assays.
Titer
In a direct ELISA, a 1:5,000 dilution of the antibody will produce an optical density (O.D.) of 0.5 when using an alkaline phosphatase-conjugated rabbit anti-mouse IgG secondary antibody (available from Jackson Laboratories).
Synonyms
NKSF2, CTL maturation factor (TCMF), Cytotoxic lymphocyte maturation factor 40 kDa subunit (CLMF p40), TSF, Edodekin-alpha, IL-12 p40, IL-12B, IL-12 subunit p40, NK cell stimulatory factor chain 2.
Purification Method
Ion exchange.
Type
Rat Anti Mouse Monoclonal.
Clone
NYRmIL-12p40.
Immunogen
r.Murine IL-12 p40.
Ig Subclass
Rat IgG2a.

Q&A

What is IL-12p40 and why is it important for immunological research?

IL-12p40 is a protein subunit common to both IL-12 and IL-23 cytokines. IL-12 is composed of p40 and p35 subunits, while IL-23 contains p40 paired with p19 . This shared subunit plays a crucial role in immune regulation, particularly in pro-inflammatory responses. IL-12p40 binds to the IL-12Rβ1 receptor, which is the first step in the signaling cascade for both cytokines .

The importance of IL-12p40 in immunological research stems from its dual role in mediating IL-12 and IL-23 signaling pathways. IL-12 primarily drives the Th1 immune response and IFN-γ production, while IL-23 supports Th17 responses and IL-17 production . Notably, IL-12p40 can also exist as a monomer or homodimer (IL-12p80), functioning as natural antagonists to IL-12/IL-23 signaling by competing for binding to IL-12Rβ1 .

Genetically, IL-12p40 deficiency leads to Mendelian susceptibility to mycobacterial disease (MSMD), highlighting its physiological importance in antimicrobial immunity . Understanding IL-12p40 is therefore essential for researching inflammatory conditions, autoimmune diseases, and infection responses.

How does flow cytometric analysis with anti-IL-12p40 antibodies work?

Flow cytometric analysis using anti-IL-12p40 antibodies enables the identification and enumeration of IL-12 producing cells within mixed cell populations. The methodology involves:

  • Cell preparation: Cells must be fixed with paraformaldehyde and permeabilized with saponin to allow antibody access to intracellular IL-12p40.

  • Antibody titration: For optimal results, antibodies should be titrated, typically using ≤0.5 μg mAb per million cells .

  • Staining protocol: Permeabilized cells are incubated with fluorochrome-conjugated anti-IL-12p40 antibodies (such as PE- or APC-conjugated C15.6 clone) .

  • Controls: Two recommended specificity controls include:

    • Pre-blocking the labeled antibody with excess ligand prior to staining

    • Pre-blocking the permeabilized cells with unlabeled IL-12p40 antibody before adding the labeled antibody

  • Isotype control: Using a matched isotype control (e.g., rat IgG1 for the C15.6 clone) at equivalent concentrations to assess background staining levels .

This approach is particularly valuable for studying the kinetics of IL-12 production, identifying specific IL-12-producing cell populations, and evaluating the effects of stimulatory or inhibitory conditions on IL-12 expression.

What are the differences between antibodies targeting different epitopes of IL-12p40?

Anti-IL-12p40 antibodies can target different epitopes, resulting in distinct functional consequences:

Epitope RegionExample AntibodyMechanism of ActionFunctional EffectClinical Implications
Domain 1 (aa 1-88)UstekinumabBlocks IL-12/23 binding to IL-12Rβ1Inhibits both IL-12/23 signaling AND natural antagonism by IL-12p40/p80Effective for psoriasis; less effective for Crohn's disease and MS
Domain 3 (aa 253-286)6F6Blocks binding to IL-12Rβ2 and IL-23R, but not IL-12Rβ1Inhibits IL-12/23 signaling while preserving natural antagonism by IL-12p40/p80Potentially enhanced efficacy through dual antagonistic system
Other epitopesVarious research antibodiesVarious mechanismsDepends on specific binding characteristicsResearch tools for different applications

Antibodies like 6F6 represent a novel class that can neutralize IL-12/IL-23 bioactivity by preventing their interaction with signaling receptors (IL-12Rβ2/IL-23R) while allowing the natural antagonists (monomeric IL-12p40 and IL-12p80) to continue competing for IL-12Rβ1 binding . This mechanism creates a dual antagonistic system that may offer therapeutic advantages.

How can I validate the specificity of an IL-12p40 antibody?

Validating IL-12p40 antibody specificity requires multiple complementary approaches:

  • Cross-reactivity testing: Test antibody binding against:

    • Recombinant IL-12p40 monomer

    • IL-12p70 heterodimer (p40+p35)

    • IL-23 heterodimer (p40+p19)

    • IL-12p80 homodimer

    • IL-12p35 and IL-23p19 alone as negative controls

  • Receptor-neutralization assays: Determine if the antibody blocks specific receptor interactions:

    • IL-12 binding to IL-12Rβ1

    • IL-12 binding to IL-12Rβ2

    • IL-23 binding to IL-12Rβ1

    • IL-23 binding to IL-23R

  • Functional assays:

    • IFN-γ production assays (for IL-12 neutralization)

    • IL-17 production assays (for IL-23 neutralization)

  • Blocking controls for flow cytometry:

    • Pre-incubation with unlabeled antibody

    • Pre-incubation with recombinant IL-12p40

  • Genetic controls:

    • Testing on cells from IL-12p40 knockout models

    • Testing on cells from IL-12p40-deficient patients

This comprehensive validation ensures that the antibody specifically recognizes IL-12p40 and clarifies its precise mechanism of action in experimental settings.

How do regulatory mechanisms for IL-12p40 differ from those governing IL-12p35, and how does this affect antibody-based research?

The regulation of IL-12p40 and IL-12p35 gene expression differs significantly, with important implications for antibody-based research:

IL-12p40 regulation:

  • Highly inducible, with low basal expression in most cells

  • Expression primarily in antigen-presenting cells (macrophages, dendritic cells)

  • Regulated by multiple transcription factors including NF-κB, PU.1, C/EBP, and IRFs

  • Often produced in excess of IL-12p70, leading to free p40 monomers and homodimers

IL-12p35 regulation:

  • More restrictive expression pattern

  • Often the limiting factor in IL-12p70 production

  • Specifically requires IRF-1 for proper expression in macrophages

  • Has post-transcriptional regulatory mechanisms

Research implications:

  • When using anti-IL-12p40 antibodies, researchers must consider whether they are detecting free p40, IL-12p70, IL-23, or some combination.

  • Stimulation conditions may differently affect p40 vs. p35 expression, creating scenarios where:

    • High p40 but low p35 leads to predominantly antagonistic effects

    • Balanced p40/p35 production leads to IL-12p70 signaling

    • High p40 with high p19 leads to IL-23 signaling

  • Methodological considerations:

    • Use antibodies specific to IL-12p70 heterodimer when studying IL-12 specifically

    • Consider measuring both p40 and p35/p19 subunits independently

    • Perform functional assays to distinguish IL-12 vs. IL-23 bioactivity

Understanding these differential regulatory mechanisms enables more precise experimental design and interpretation when working with IL-12p40 antibodies.

What are the considerations for using IL-12p40 antibodies in animal models to study human diseases?

Using IL-12p40 antibodies in animal models requires careful consideration of several factors:

Species cross-reactivity and homology:

  • Human and mouse IL-12p40 share approximately 70% amino acid identity

  • Most antibodies are species-specific and don't cross-react

  • Some epitopes may be conserved, allowing cross-species reactivity for select antibodies

Functional considerations:

  • Human IL-12 is only weakly active in murine systems

  • Heterochimeric IL-12 (human p40/murine p35) can induce robust IFN-γ responses in mice, making it useful for testing human p40-specific antibodies in vivo

  • Testing in murine systems often requires monitoring IFN-γ production as a surrogate marker for IL-12 activity

Experimental design challenges:

  • Dosing optimization:

    • Antibody half-life varies by isotype and species

    • Tissue penetration may differ between animal models

    • Mouse studies typically use 0.1-10 mg/kg dosing regimens

  • Readout selection:

    • Direct measurements: cytokine levels (IFN-γ, IL-17)

    • Indirect assessments: T-cell differentiation (Th1, Th17)

    • Disease-specific endpoints: inflammation scores, autoantibody levels

  • Model selection table:

Disease TargetRecommended ModelKey IL-12p40 InvolvementAppropriate Readouts
Inflammatory bowel diseaseDSS-induced colitis, T-cell transferIL-23 > IL-12Colon histology, IL-17, IL-22
PsoriasisImiquimod-inducedIL-23 > IL-12Skin thickness, IL-17, IL-22
Multiple sclerosisEAEBoth IL-12 and IL-23Clinical scores, IFN-γ and IL-17
Mycobacterial infectionBCG challengeIL-12 > IL-23Bacterial burden, IFN-γ, granuloma formation
  • Translation considerations:

    • Differences in receptor distribution between species

    • Variation in downstream signaling pathway components

    • Potential differences in antagonist (p40/p80) functions across species

Rigorous control experiments and awareness of species differences are essential for meaningful translation of animal findings to human disease applications.

How can epitope mapping inform the development of more selective IL-12p40 antibodies?

Epitope mapping of IL-12p40 provides critical insights for developing antibodies with selective mechanisms:

Key structural domains of IL-12p40:

  • Domain 1 (D1): Involved in IL-12Rβ1 binding

  • Domain 2 (D2): Forms part of the interface with p35/p19

  • Domain 3 (D3): Contains regions critical for signaling receptor interactions

Epitope mapping approaches:

  • Alanine scanning mutagenesis: Systematic mutation of residues to identify critical binding sites

    • Example: D265 mutation in domain 3 completely abrogated 6F6 antibody binding

  • Domain-swapping experiments: Creating chimeric proteins with exchanged domains between human and mouse IL-12p40

  • Hydrogen-deuterium exchange mass spectrometry: Identifies regions protected from exchange upon antibody binding

  • X-ray crystallography: Provides high-resolution structural information on antibody-antigen complexes

Strategic epitope targeting:

Target EpitopeFunctional EffectPotential Applications
D1 (IL-12Rβ1 binding site)Blocks both IL-12/23 and natural antagonistsGeneral immunosuppression; psoriasis
D3 (signaling receptor interface)Blocks signaling while preserving natural antagonismEnhanced efficacy through dual antagonism
p40-p35 interfaceSelective for IL-12 over IL-23Targeting Th1-mediated diseases specifically
p40-p19 interfaceSelective for IL-23 over IL-12Targeting Th17-mediated diseases specifically

Case study: 6F6 antibody
The 6F6 antibody binds to domain 3 of IL-12p40, specifically residues Q253-C286, with D265 being critical. This epitope is positioned such that 6F6 binding prevents interaction with signaling receptors without affecting IL-12Rβ1 binding . This creates a unique mechanism where:

  • The antibody directly neutralizes IL-12/IL-23 signaling

  • Natural antagonists (IL-12p40/p80) can still compete for IL-12Rβ1

  • A dual antagonistic system potentially enhances efficacy

This level of epitope characterization enables rational design of antibodies with precise functional properties for specific disease applications.

What methodological approaches can resolve contradictory data in IL-12p40 antibody research?

Researching IL-12p40 antibodies can yield seemingly contradictory results due to the complex biology of the IL-12/IL-23 system. Here are methodological approaches to resolve such contradictions:

Sources of contradictory data:

  • Differential effects on IL-12 vs. IL-23 pathways

  • Context-dependent roles of monomeric IL-12p40 and IL-12p80

  • Varying concentrations of p40 relative to heterodimers (5-500 fold excess)

  • Different experimental readouts measuring distinct aspects of the system

Methodological approaches to resolve contradictions:

  • Comprehensive antibody characterization:

    • Determine exact epitope and binding mechanism

    • Test effects on all receptor interactions (IL-12Rβ1, IL-12Rβ2, IL-23R)

    • Evaluate effects on natural antagonist function

  • Multi-parameter analysis:

    • Measure multiple cytokines simultaneously (IFN-γ, IL-17, IL-22)

    • Assess effects on different cell populations (T cells, NK cells, innate lymphoid cells)

    • Determine relative levels of all relevant proteins (p40, p35, p19, p80)

  • Kinetic studies:

    • Examine temporal relationships between cytokine production and receptor expression

    • Monitor dynamics of signaling pathway activation

    • Track cell population changes over time

  • Integrated experimental approach:

Level of AnalysisTechniquesResolution of Contradictions
MolecularSPR, ELISA, receptor binding assaysClarify binding mechanisms and affinities
CellularFlow cytometry, signaling assays (STAT phosphorylation)Determine cell-specific responses
FunctionalT-cell differentiation, cytokine productionAssess biological outcomes
In vivoAnimal models with specific genetic modificationsValidate relevance in complex systems
  • Genetic approaches:

    • Compare antibody effects in wild-type vs. IL-12p40-deficient systems

    • Use cells from patients with genetic deficiencies in IL-12/23 pathway components

    • Create reporter systems for specific pathway activation

When faced with contradictory data, researchers should systematically evaluate whether differences stem from experimental conditions, biological complexity, or antibody-specific properties. This multi-faceted approach allows reconciliation of seemingly contradictory results within a coherent framework of IL-12/IL-23 biology.

How can researchers optimize sandwich ELISA protocols for IL-12p40 detection in complex biological samples?

Optimizing sandwich ELISA for IL-12p40 detection in complex biological samples requires addressing several technical challenges:

Key considerations for IL-12p40 sandwich ELISA optimization:

  • Antibody pair selection:

    • Capture antibody: Purified C15.6 antibody is recommended for capturing mouse IL-12p40

    • Detection antibody: Biotinylated C17.8 antibody pairs effectively with C15.6

    • These antibodies should recognize non-overlapping epitopes to avoid competition

  • Specificity considerations:

    • Determine whether the assay detects:
      a) Total IL-12p40 (free p40, IL-12p70, and IL-23)
      b) Only IL-12p70 heterodimer
      c) Only free IL-12p40

    • Use recombinant standards to verify specificity

  • Protocol optimization table:

ParameterOptimization StrategyTroubleshooting
Coating concentrationTitrate capture antibody (typically 1-5 μg/ml)Poor sensitivity may require higher coating density
Blocking bufferCompare BSA, casein, and commercial blockersHigh background suggests inadequate blocking
Sample dilutionPrepare serial dilutions to find optimal rangeMatrix effects may require sample-specific optimization
Incubation timesOptimize for sensitivity vs. throughputLonger incubations may increase sensitivity but introduce drift
Detection systemCompare direct vs. amplified detection systemsStreptavidin-HRP systems offer good sensitivity for biotin-labeled detection antibodies
  • Handling complex biological samples:

    • Pre-clear serum/plasma samples by centrifugation

    • Consider adding blocking agents specific to the sample type (e.g., heterophilic antibody blockers for human serum)

    • Include sample-matched matrix in standard curves

    • For tissue homogenates, optimize extraction buffers to preserve cytokine structure

  • Validation approaches:

    • Spike-and-recovery experiments with recombinant IL-12p40

    • Linearity-of-dilution tests to verify absence of matrix effects

    • Comparison with other quantification methods (e.g., multiplex bead assays)

    • Analysis of samples from IL-12p40-deficient sources as negative controls

  • Special considerations for IL-12p40 measurement:

    • Account for potential interference from IL-12p80 homodimers

    • Consider the high ratio of free p40 to heterodimeric forms (often 5-500 fold)

    • For human samples from treated patients, test for interference from therapeutic antibodies

For the most reliable results in complex samples like serum or plasma, commercial IL-12p40 ELISA sets (e.g., OptEIA™ mouse IL-12 ELISA set) are recommended as they are specifically validated for these applications .

How does IL-12p40 antibody research inform our understanding of Mendelian susceptibility to mycobacterial disease (MSMD)?

IL-12p40 antibody research has been instrumental in elucidating the pathophysiology of Mendelian susceptibility to mycobacterial disease (MSMD), a rare immunodeficiency:

Genetic basis of IL-12p40 deficiency:

  • Autosomal recessive inheritance pattern

  • Nine different identified mutant alleles of IL12B gene causing IL-12p40 deficiency

  • Most mutations cause frameshift and premature stop codons

  • Four recurrent variants affect 25/30 kindreds, suggesting founder effects in certain populations

Clinical features revealed through IL-12p40 research:

Clinical ManifestationFrequencyNotes
BCG disease97.5% of vaccinated patientsNearly universal after BCG vaccination
SalmonellosisCommonRecurrence rate 36.4%
Mycobacterial disease recurrence25%Lower than salmonellosis recurrence
Multiple mycobacterial infectionsRare (3 patients)Limited susceptibility to diverse mycobacteria
Combined mycobacteriosis + salmonellosis9 patientsSuggests shared protective mechanisms
Other infections (candidiasis, nocardiosis)UncommonShows broader immune defects

Immunological findings enabled by IL-12p40 antibodies:

  • Complete absence of IL-12p40 and IL-12p70 in patient samples

  • Reduced IFN-γ production in response to stimuli

  • Defects in both IL-12 and IL-23 pathways, explaining susceptibility to diverse pathogens

  • Impaired IL-17 responses potentially explaining susceptibility to Candida infections

Therapeutic implications:

  • IFN-γ therapy rather than IL-12 replacement is the logical therapeutic approach

  • Antibody-based research helps distinguish IL-12p40 deficiency from other genetic causes of MSMD

  • Understanding the IL-12/IFN-γ axis enables targeted treatment of infections

Research methodology:
Researchers use anti-IL-12p40 antibodies to assess IL-12 production in patient cells, comparing with controls to diagnose IL-12p40 deficiency. This diagnostic approach requires sensitive ELISA protocols and functional assays measuring IFN-γ production in response to stimuli .

IL-12p40 deficiency has a high but incomplete clinical penetrance, with 33.3% of genetically affected relatives showing no symptoms . This observation, revealed through IL-12p40 antibody screening of asymptomatic family members, suggests the existence of compensatory mechanisms warranting further investigation.

What are the methodological considerations for studying IL-12p40 in autoimmune disease models?

Studying IL-12p40 in autoimmune disease models requires careful methodological planning:

Model selection based on IL-12/IL-23 pathway involvement:

Autoimmune DiseaseRelevant Animal ModelsDominant PathwayKey Readouts
PsoriasisImiquimod-induced, IL-23 injectionIL-23/Th17Skin thickness, IL-17, IL-22
Inflammatory bowel diseaseDSS colitis, TNBS colitis, T-cell transferIL-23/Th17Histology, weight loss, IL-17
Multiple sclerosisEAEBoth IL-12/Th1 and IL-23/Th17Clinical scores, IFN-γ, IL-17
Rheumatoid arthritisCollagen-induced arthritisBoth pathwaysJoint swelling, bone erosion

Intervention timing considerations:

  • Preventive protocols: Administer antibodies before disease induction to assess role in disease initiation

  • Therapeutic protocols: Administer after disease onset to assess role in disease progression

  • Resolution protocols: Administer during recovery phase to assess role in resolution/relapse

Antibody selection strategies:

  • Select antibodies based on specific mechanism:

    • Those blocking IL-12Rβ1 binding (like ustekinumab) affect both IL-12/23 signaling and natural antagonism

    • Those blocking signaling receptor binding (like 6F6) preserve natural antagonism

  • Consider control antibodies:

    • Isotype-matched controls

    • Anti-IL-12p35 (IL-12-specific) antibodies

    • Anti-IL-23p19 (IL-23-specific) antibodies

    • These help distinguish IL-12 vs. IL-23 effects

Methodological challenges and solutions:

ChallengeSolutionTechnical Considerations
Distinguishing IL-12 vs. IL-23 effectsUse pathway-specific readouts (T-bet/IFN-γ for IL-12; RORγt/IL-17 for IL-23)Flow cytometry with transcription factor staining
Systemic vs. local effectsCompare systemic vs. site-specific antibody administrationLocal injection techniques, tissue-specific analysis
Compensatory mechanismsAnalyze multiple cytokines simultaneouslyMultiplex assays, comprehensive tissue analysis
Timing of interventionInclude multiple intervention timepointsRequires larger cohorts, careful planning

Data interpretation framework:

  • Distinguish direct effects on target cells from indirect effects via altered cytokine networks

  • Consider potential roles of monomeric IL-12p40 and IL-12p80 as natural antagonists

  • Acknowledge that genetic deficiency models may differ from antibody blockade due to developmental effects

  • Evaluate specificity by comparing with other cytokine-targeting approaches

These methodological considerations enable researchers to accurately assess IL-12p40's role in autoimmune disease pathogenesis and evaluate the therapeutic potential of targeting this pathway in specific conditions.

How might next-generation IL-12p40 antibodies improve upon current therapeutic approaches?

Next-generation IL-12p40 antibodies could address limitations of current approaches through several innovative strategies:

Current limitations to overcome:

  • Inadequate efficacy in certain conditions (Crohn's disease, multiple sclerosis)

  • Inability to distinguish between IL-12 and IL-23 blockade

  • Interference with natural antagonism by monomeric IL-12p40 and IL-12p80

  • Limited tissue penetration or tissue-specific targeting

Innovative antibody engineering approaches:

ApproachMechanismPotential Advantages
Epitope-selective antibodiesTarget domain 3 epitopes that allow natural antagonism while blocking signaling receptor engagement (like 6F6) Dual antagonistic system enhancing efficacy
Bispecific antibodiesOne arm targeting IL-12p40, another targeting tissue-specific markerTissue-focused delivery reducing systemic effects
pH-dependent bindingStronger binding in inflammatory microenvironmentsContext-dependent activity with reduced systemic effects
Subunit-interface antibodiesTarget epitopes at p35/p40 or p19/p40 interfacesSelective inhibition of IL-12 or IL-23 specifically
Engineered fragmentsSmaller antibody fragments with enhanced tissue penetrationImproved efficacy in certain tissues like CNS

Mechanistic innovations:

  • Dual antagonist system engineering: Building on the 6F6 example, developing antibodies that preserve or even enhance the natural antagonism of IL-12p40/p80 while blocking signaling receptor engagement .

  • Conditional activation: Creating antibodies that become neutralizing only in specific microenvironments (low pH, high protease activity) characteristic of inflammatory tissues.

  • Affinity modulation: Engineering antibodies with differential binding to monomeric vs. heterodimeric forms to selectively target specific configurations.

Delivery and formulation innovations:

  • Site-specific delivery systems for localized treatment

  • Extended half-life modifications for reduced dosing frequency

  • Subcutaneous formulations with improved patient convenience

Combination approaches:

  • IL-12p40 antibodies paired with other immunomodulatory agents

  • Sequential or alternating therapeutic regimens

  • Precision medicine approaches using biomarkers to guide therapy

The development of 6F6-like antibodies that create a dual antagonistic system represents a promising direction, potentially providing enhanced efficacy in conditions where current IL-12p40 antibodies have shown limitations . This strategy leverages the body's natural regulatory mechanisms rather than simply blocking all IL-12p40 activities.

What are the most promising experimental techniques for studying IL-12p40 antibody mechanisms in complex immune microenvironments?

Advanced experimental techniques are revolutionizing our understanding of IL-12p40 antibody mechanisms in complex immune microenvironments:

Single-cell analysis techniques:

  • Single-cell RNA sequencing (scRNA-seq):

    • Reveals heterogeneity in cellular responses to IL-12p40 blockade

    • Identifies previously unrecognized target cell populations

    • Methodology: Isolate cells from tissues before/after antibody treatment, perform scRNA-seq, and analyze differential gene expression

  • Cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq):

    • Simultaneously measures protein and mRNA levels at single-cell resolution

    • Can track IL-12 receptor components and downstream signaling molecules

    • Particularly valuable for correlating receptor expression with response to antibody treatment

Advanced imaging techniques:

  • Multiplex immunohistochemistry:

    • Simultaneously visualizes multiple markers in tissue sections

    • Reveals spatial relationships between IL-12-producing and responding cells

    • Can track antibody penetration and target engagement in tissues

  • Intravital microscopy:

    • Real-time visualization of cellular interactions in living tissues

    • Tracks dynamics of IL-12 signaling and antibody effects

    • Particularly valuable for understanding kinetics of antibody action

Functional genomics approaches:

  • CRISPR screening:

    • Identifies genes required for IL-12 response or resistance to IL-12p40 blockade

    • Can discover novel components of IL-12/IL-23 signaling pathways

    • Methodology: CRISPR knockout libraries in relevant cell types, followed by selection for altered response to IL-12 or IL-12p40 antibodies

  • Genetic reporter systems:

    • Fluorescent or luminescent reporters for IL-12/IL-23 pathway activation

    • Enables real-time monitoring of signaling dynamics

    • Can be combined with intravital imaging for in vivo studies

Systems biology and computational approaches:

  • Network analysis:

    • Models complex interactions between IL-12/IL-23 and other cytokine pathways

    • Predicts system-level effects of IL-12p40 blockade

    • Helps identify optimal combination therapy strategies

  • Machine learning algorithms:

    • Identify patterns in complex datasets that predict response to IL-12p40 blockade

    • Develop biomarker signatures for patient stratification

    • Optimize therapeutic regimens based on multiple parameters

Experimental design considerations:

  • Implement time-course studies to capture dynamic responses

  • Include multiple tissue compartments to assess systemic vs. local effects

  • Combine multiple analytical platforms for integrated data analysis

  • Develop appropriate analytical pipelines for high-dimensional data

These advanced techniques enable researchers to move beyond simplistic models of IL-12p40 antibody action to understand their effects in the complex, dynamic environments of inflamed tissues. This knowledge will inform more precise therapeutic strategies and potentially identify novel applications for IL-12p40-targeting approaches.

Product Science Overview

Introduction

Interleukin-12 (IL-12) is a cytokine that plays a crucial role in the immune response by promoting the differentiation of naive T cells into Th1 cells. It is composed of two subunits: p35 and p40. The p40 subunit can form homodimers (p80) or heterodimers with p35 (p70), which is the biologically active form of IL-12. The rat anti-mouse IL-12 p40 antibody is a monoclonal antibody that specifically binds to the p40 subunit of mouse IL-12, both in its free form and when complexed as p80 or p70 .

Production and Purification

The rat anti-mouse IL-12 p40 antibody is typically produced using hybridoma technology. This involves immunizing rats with recombinant mouse IL-12 p70 protein, followed by the fusion of the rat spleen cells with myeloma cells to create hybridomas. These hybridomas are then screened for the production of antibodies that specifically bind to the p40 subunit of IL-12. The monoclonal antibody is purified from the tissue culture supernatant or ascites fluid using affinity chromatography .

Applications

The rat anti-mouse IL-12 p40 antibody is widely used in various immunological assays, including:

  1. Flow Cytometry: This antibody is commonly used for intracellular staining to identify and enumerate IL-12 producing cells within mixed cell populations. It is conjugated with fluorophores such as PE (phycoerythrin) or APC (allophycocyanin) to facilitate detection by flow cytometry .
  2. ELISA: The antibody can be used as a capture antibody in sandwich ELISA assays to quantify IL-12 levels in biological samples. It is often paired with a biotinylated detection antibody .
  3. Immunohistochemistry: The antibody is also used in immunohistochemical staining to visualize IL-12 expression in tissue sections .
Specificity and Cross-Reactivity

The C15.6 monoclonal antibody specifically binds to both free and complexed forms of the p40 subunit of mouse IL-12. It does not cross-react with other cytokines, ensuring high specificity in immunological assays .

Storage and Handling

The rat anti-mouse IL-12 p40 antibody should be stored undiluted at 4°C and protected from prolonged exposure to light. It should not be frozen, as this can affect the antibody’s stability and binding affinity .

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.