IL 12 Mouse

Interleukin-12 Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to IL-12 (Mouse)

Interleukin-12 (IL-12) is a heterodimeric cytokine critical for bridging innate and adaptive immunity. The murine variant, IL-12 Mouse, consists of two covalently linked subunits: p35 (35 kDa) and p40 (40 kDa), forming the bioactive p70 heterodimer . Produced primarily by macrophages, dendritic cells, and B lymphocytes, it drives Th1 differentiation, enhances cytotoxic lymphocyte activity, and induces interferon-gamma (IFN-γ) . Unlike human IL-12, the mouse variant is cross-reactive with human cells, a property determined by structural features of its p35 subunit .

Subunit Composition

SubunitAccession NumberMolecular WeightKey Features
p35NP_03237727.5 kDaShares 60% homology with human p35; critical for species specificity
p40P4343234.7 kDaHomologous to IL-6 receptor; binds IL-12Rβ1

Immune Modulation

  • IFN-γ Induction: IL-12 Mouse synergizes with IL-18 to amplify IFN-γ production in T and NK cells, pivotal for antiviral and antitumor responses .

  • Th1 Polarization: Drives naïve T cells toward Th1 lineage via STAT4 activation, suppressing Th2 cytokines (e.g., IL-4, IL-10) .

  • Cytotoxic Activity: Enhances NK cell cytotoxicity and CD8+ T cell proliferation through perforin and granzyme upregulation .

Experimental Autoimmune Encephalomyelitis (EAE)

Mouse StrainEAE SusceptibilityCytokine Profile (Spleen)
IL-12p35⁻/⁻SusceptibleReduced IFN-γ, IL-2
IL-12p40⁻/⁻ResistantElevated IL-4, IL-10

IL-12p40 deficiency completely abrogates EAE, while p35-deficient mice develop pathology akin to wild-type, implicating IL-23 (p40/p19) in disease progression .

Recombinant IL-12 Mouse

  • Source: Expressed in Spodoptera frugiperda (Sf21) insect cells .

  • Activity: Stimulates proliferation of PHA-activated splenocytes (ED₅₀: 0.01–0.1 ng/mL) .

Detection Tools

ProductCatalog NumberRange/SensitivityApplication
Recombinant Protein419-ML0.01–0.1 ng/mL (ED₅₀)Cell assays
ELISA KitCL042215.6–1,000 pg/mLQuantification
Neutralizing AntibodyAF-419-NAND₅₀: 0.01–0.06 µg/mLFunctional studies

Neuroprotection in Neuroinflammation

  • IL-12 signaling in neurons activates neuroprotective pathways, preserving CNS integrity during EAE by sustaining trophic factor release .

  • Il12rb2⁻/⁻ mice exhibit exacerbated EAE severity, underscoring IL-12’s role in mitigating neurodegeneration .

Toxicity and Adaptive Resistance

  • Acute Toxicity: Single-dose IL-12 Mouse triggers transient leukopenia and anemia, mediated by IFN-γ .

  • Cytokine Exhaustion: Prolonged IL-12 exposure diminishes IFN-γ and IL-10 production in lymph node cells, suggesting receptor desensitization .

Clinical and Therapeutic Implications

  • Autoimmunity: IL-12 blockade (e.g., anti-p40 antibodies) ameliorates collagen-induced arthritis and diabetes in murine models .

  • Cancer Immunotherapy: IL-12 adjuvant therapy enhances tumor-specific CD8+ T cell responses, though dose-limiting toxicity remains a challenge .

Product Specs

Introduction
Interleukin-12 (IL-12) is a heterodimeric cytokine that plays a critical role in cell-mediated immunity. It acts on T cells and natural killer (NK) cells, promoting the production of interferon gamma (IFN-γ) and driving the differentiation of T helper 1 (Th1) cells.
Description
Recombinant Mouse Interleukin-12 (IL-12) is a glycosylated heterodimer produced in HEK 293 cells. It consists of two disulfide-linked subunits, p35 (35 kDa) and p40 (40 kDa), with a total molecular weight of approximately 75 kDa. The protein is purified using proprietary chromatographic methods.
Physical Appearance
Sterile, white, lyophilized powder.
Formulation
The protein is lyophilized in a solution containing 0.5x PBS at a pH of 7.5.
Solubility
To reconstitute lyophilized murine IL-12, it is recommended to dissolve it in sterile, pyrogen-free water at a concentration of 100 µg/ml. This solution can be further diluted in other aqueous solutions as needed.
Stability
Lyophilized murine IL-12 is stable at room temperature for up to 3 weeks; however, for long-term storage, it is recommended to store the lyophilized powder below -18°C. After reconstitution, the IL-12 solution should be stored at 4°C for up to 7 days. For extended storage, freeze the solution below -18°C, but avoid repeated freeze-thaw cycles. Adding a carrier protein, such as 0.1% HSA or BSA, to the solution before freezing is recommended for long-term storage.
Purity
The purity of this product is determined to be greater than 95% as assessed by SDS-PAGE analysis.
Biological Activity
Mouse IL-12 exhibits full biological activity compared to established standards. The ED50, which represents the concentration required to induce 50% of the maximal response, is determined by measuring the dose-dependent expression of CD25 on CD8+ mouse T cells. The ED50 is in the range of 10-20 ng/ml, corresponding to a specific activity of 50,000-100,000 units/mg.
Synonyms
NKSF, CTL maturation factor (TCMF), Cytotoxic lymphocyte maturation factor (CLMF), TSF, Edodekin-alpha, IL-12.
Source
HEK-293 Cells.
Amino Acid Sequence
p35 Subunit: RVIPVSGPAR CLSQSRNLLK TTDDMVKTAR EKLKHYSCTA EDIDHEDITR DQTSTLKTCL PLELHKNESC LATRETSSTT RGSCLPPQKT SLMMTLCLGS IYEDLKMYQT EFQAINAALQ NHNHQQIILD KGMLVAIDEL MQSLNHNGET LRQKPPVGEA DPYRVKMKLC ILLHAFSTRV VTINRVMGYL SSA.

p40 Subunit: MWELEKDVYV VEVDWTPDAP GETVNLTCDT PEEDDITWTS DQRHGVIGSG KTLTITVKEF LDAGQYTCHK GGETLSHSHL LLHKKENGIW STEILKNFKN KTFLKCEAPN YSGRFTCSWL VQRNMDLKFN IKSSSSSPDS RAVTCGMASL SAEKVTLDQR DYEKYSVSCQ EDVTCPTAEE TLPIELALEA RQQNKYENYS TSFFIRDIIK PDPPKNLQMK PLKNSQVEVS
WEYPDSWSTP HSYFSLKFFV RIQRKKEKMK ETEEGCNQKG AFLVEKTSTE VQCKGGNVCV QAQDRYYNSS CSKWACVPCR VRS.

Q&A

What is the molecular structure and composition of mouse IL-12?

Mouse IL-12 is a heterodimeric cytokine composed of two subunits: p35 (21.7 kDa) and p40 (35.8 kDa), which together form a complex of approximately 57.5 kDa. The p35 subunit consists of 193 amino acids while the p40 subunit contains 313 amino acids, creating a complete protein of 506 amino acids . The heterodimeric structure is essential for biological activity, as neither subunit alone can fully replicate the complete cytokine's functions. The amino acid sequences for both subunits have been fully characterized and are available in protein databases under accession numbers P43432 (p35) and P43431 (p40) . This heterodimeric configuration enables IL-12 to bind to its receptor complex and initiate downstream signaling cascades crucial for its diverse biological functions in research contexts.

What cellular sources produce IL-12 in mouse models during steady state and inflammation?

In mice, IL-12 is primarily produced by dendritic cells, macrophages, and B cells following antigen stimulation under normal physiological conditions . During neuroinflammation, the cellular sources shift significantly, with infiltrating myeloid-derived cells (MdCs) becoming the dominant producers of IL-12 in the inflamed central nervous system (CNS), generating substantially higher amounts compared to resident microglia . This differential production pattern becomes particularly evident during experimental autoimmune encephalomyelitis (EAE), where IL-12 concentrations remain almost undetectable for approximately one week post-immunization, then begin increasing around the time of clinical disease onset (day 10 post-immunization), coinciding with the influx of MdCs . This temporal and cell-specific production pattern has significant implications for experimental design when studying IL-12's roles in neuroinflammatory conditions.

How does the IL-12 receptor complex function in mouse tissues?

The mouse IL-12 receptor complex consists of two essential subunits: IL-12Rβ1 (encoded by Il12rb1) and IL-12Rβ2 (encoded by Il12rb2), both of which are required for effective IL-12 signaling. Research using conditional knockout mouse strains (Il12rb2 fl/fl) has demonstrated that complete deletion of Il12rb2 across all tissues results in hypersusceptibility to myelin oligodendrocyte glycoprotein (MOG)-induced experimental autoimmune encephalomyelitis (EAE) . Interestingly, while IL-12 receptors were traditionally thought to function primarily on immune cells, recent studies have revealed their expression on neuroectodermal cells, particularly neurons and oligodendrocytes, even under steady-state conditions . This discovery significantly expands our understanding of IL-12's biological roles beyond immune modulation. The receptor complex initiates signaling through the JAK-STAT pathway, with IL-12 stimulation inducing phosphorylation of STAT4, which can be experimentally measured to confirm functional receptor activity .

How can researchers distinguish between IL-12 and other related cytokines in mouse experimental systems?

Distinguishing IL-12 from related cytokines, particularly those sharing the p40 subunit (such as IL-23), requires specific methodological approaches in mouse experimental systems. Researchers should employ antibodies that recognize the complete heterodimeric IL-12 (p70) rather than just individual subunits to avoid cross-reactivity. When measuring biological activities, IL-12-specific functional assays such as the induction of secreted embryonic alkaline phosphatase (SEAP) in HEK-Blue IL-12 reporter cells provide specificity, with an effective dose (ED50) typically ≤15 ng/mL for recombinant mouse IL-12 . Genetic approaches using conditional knockouts of IL-12-specific receptor components (particularly Il12rb2) rather than shared components can help delineate IL-12-specific effects from those of related cytokines . Additionally, downstream effects such as STAT4 phosphorylation and IFN-γ induction provide relatively specific indicators of IL-12 activity, while analyzing transcriptional signatures through RNA sequencing can further distinguish IL-12-specific responses from those induced by related cytokines in various tissue and cell types .

What is the role of neuronal IL-12 signaling in neuroprotection during mouse models of neuroinflammation?

Recent research has revealed an unexpected neuroprotective role for IL-12 signaling in neurons during neuroinflammatory conditions. Studies using conditional knockout mouse models have demonstrated that IL-12 receptor signaling in neuroectodermal cells, rather than immune cells, provides critical protection during experimental autoimmune encephalomyelitis (EAE) . Mice lacking IL-12 receptors across all tissues (Il12rb2 del/del) develop significantly more severe EAE with higher maximal and cumulative clinical scores compared to wild-type animals . Surprisingly, selective deletion of IL-12 receptors in T cells (using Cd4 Cre/+ Il12rb2 fl/fl mice) or all hematopoietic cells (using Vav1 Cre/+ Il12rb2 fl/fl mice) did not recapitulate this exacerbated disease phenotype, indicating that non-immune cells mediate IL-12's protective effects .

Single-nucleus RNA sequencing analysis revealed that IL-12 receptor signaling in neuronal populations, particularly granule cells, maintains the expression of genes involved in neuronal survival and neuroprotection during inflammatory challenges . This IL-12-induced neuroprotective tissue adaptation prevents early neurodegeneration and sustains trophic factor release, thereby preserving CNS integrity during inflammation . The identification of this neuronal IL-12 sensing mechanism represents a paradigm shift in our understanding of IL-12 biology, expanding its role beyond immune modulation to include direct neuroprotective functions through neuron-intrinsic signaling pathways.

How does IL-12 delivery method affect outcomes in mouse cancer immunotherapy models?

The delivery method of IL-12 significantly impacts therapeutic outcomes in mouse cancer immunotherapy models. Research using the 70Z/3 murine pre-B cell leukemia model has demonstrated that the route of administration and ability to achieve sustained local cytokine concentrations are critical factors determining efficacy . While the 70Z/3-L variant of this leukemia is typically lethal in syngeneic mice, IL-12 administration can potentially initiate protective immune responses against these otherwise non-immunogenic leukemia cells .

Comparative studies of systemic versus localized IL-12 delivery have revealed important differences in therapeutic outcomes. Systemic administration, while reaching all tissues, may lead to suboptimal concentrations at tumor sites and potential systemic toxicity . In contrast, localized delivery methods, such as using lentiviral vectors to transduce tumor cells for continuous IL-12 expression, can achieve higher sustained local concentrations while minimizing systemic exposure .

The molecular engineering approach using lentiviral vectors (e.g., pHR-cPPT-EF1α-muIL-12-WPRE) to express IL-12 directly within tumor cells represents an advanced strategy for localized delivery . This approach involves placing the IL-12 transgene under control of a strong, constitutive promoter like elongation factor 1 alpha (EF1α) to ensure robust expression . The effectiveness of this strategy depends on multiple factors including vector design, transduction efficiency, and stable expression levels, highlighting the complexity of optimizing IL-12 delivery for cancer immunotherapy applications.

What are the cell type-specific transcriptional responses to IL-12 signaling in mouse central nervous system?

Single-nucleus RNA sequencing has revealed remarkably diverse cell type-specific transcriptional responses to IL-12 signaling in the mouse central nervous system. Studies comparing neuroectoderm-specific IL-12 receptor knockout mice (Nestin Cre/+ Il12rb2 fl/fl) with control animals during experimental autoimmune encephalomyelitis (EAE) have identified distinct transcriptional alterations across multiple cell populations . The most profound changes occur in granule cells, excitatory neurons, oligodendrocyte populations (MOL1 and MOL2), cholinergic neurons, microglia, and molecular layer interneurons (MLIs), indicating cell type-specific IL-12 responsiveness .

In neuronal populations, particularly granule cells (the most abundant and most affected neuronal subtype), IL-12 receptor deficiency leads to reduced expression of genes involved in neuronal survival and neuroprotection during inflammatory challenges . This results in increased vulnerability to neurodegeneration and compromised trophic support during neuroinflammation . The transcriptional alterations in oligodendrocytes suggest IL-12 also influences myelination processes, which may have implications for demyelinating disorders.

The selective expression pattern of IL-12 receptor components further contributes to these cell type-specific responses, with Il12rb2 mRNA transcripts being most abundant in neuronal populations, while Il12rb1 transcripts are enriched within oligodendrocyte subsets . This differential receptor component expression may underlie the unique transcriptional signatures observed across CNS cell types in response to IL-12 signaling.

How do genetic backgrounds influence mouse IL-12 responses in different disease models?

Genetic background significantly impacts IL-12 responses in mouse disease models through multiple mechanisms affecting both cytokine production and receptor signaling. Different mouse strains exhibit variable baseline immune characteristics that can profoundly influence responses to IL-12 in experimental settings . These strain-specific differences may manifest as variations in IL-12 receptor expression levels, downstream signaling efficiency, or the balance of pro-inflammatory versus regulatory immune components.

In neuroinflammatory models like experimental autoimmune encephalomyelitis (EAE), genetic background affects susceptibility to disease induction and the severity of clinical manifestations, with strains such as C57BL/6 being highly susceptible while others show relative resistance . These background-dependent differences confound interpretation of IL-12's roles unless rigorously controlled through appropriate experimental design.

For cancer immunotherapy studies using models such as the 70Z/3 murine pre-B cell leukemia, strain-specific immune responses can determine whether IL-12 treatment effectively initiates protective immunity against otherwise non-immunogenic tumor variants . The 70Z/3-L leukemia variant is typically lethal in syngeneic mice, while the 70Z/3-NL variant elicits protective immune responses, highlighting how genetic factors influence immunogenicity and IL-12 responsiveness .

To address these genetic influences, researchers should standardize experiments using mice with consistent genetic backgrounds, include multiple strains with appropriate controls when possible, and explicitly acknowledge genetic background as a potential confounding variable when interpreting experimental outcomes across different studies.

What are the optimal protocols for reconstitution and storage of recombinant mouse IL-12?

Optimal handling of recombinant mouse IL-12 requires specific reconstitution and storage protocols to maintain biological activity. Prior to reconstitution, the vial should be centrifuged to ensure the lyophilized product is collected at the bottom . For reconstitution, sterile water should be gently pipetted down the sides of the vial to achieve a final concentration of 0.1 mg/mL, avoiding direct pipetting onto the lyophilized powder . Importantly, vortexing should be strictly avoided as it can denature the protein and compromise activity . Complete reconstitution typically requires several minutes of gentle handling to ensure the protein fully dissolves without denaturation .

For long-term storage, reconstituted IL-12 should be diluted into working aliquots in a 0.1% bovine serum albumin (BSA) solution, which helps maintain protein stability and prevent adsorption to storage vessel surfaces . These aliquots should be stored at -80°C to preserve biological activity . Repeated freeze-thaw cycles must be avoided as they significantly reduce cytokine potency; each aliquot should ideally be thawed only once immediately before use .

Quality control for reconstituted IL-12 should include verification of purity (≥95% as determined by reducing and non-reducing SDS-PAGE) and endotoxin content (≤0.1 EU/μg using the kinetic LAL method) . Adherence to these handling protocols ensures consistent and reliable experimental results when using recombinant mouse IL-12 in research applications.

What experimental approaches can assess IL-12 receptor expression in different mouse tissue types?

Multiple complementary experimental approaches can comprehensively assess IL-12 receptor expression across mouse tissue types. Genetic reporter systems using knock-in mouse strains such as Il12rb2 LacZ/LacZ enable visualization of receptor expression through β-galactosidase activity detection . When combined with immunostaining for cell type-specific markers (e.g., NeuN for neurons), this approach can identify specific cell populations expressing IL-12 receptors within complex tissues .

For mRNA detection, multiplexed RNA fluorescence in situ hybridization (RNAscope) allows simultaneous visualization of IL-12 receptor subunit transcripts (Il12rb1 and Il12rb2) alongside cell-specific markers within intact tissue sections, providing crucial spatial information about receptor expression patterns . This can be particularly valuable for heterogeneous tissues like the central nervous system, where cell type-specific expression patterns might otherwise be obscured in bulk analyses.

Cell-specific expression can be quantitatively assessed using fluorescence-activated cell sorting (FACS) of dissociated tissue cells or fluorescence-activated nuclei sorting (FANS) followed by quantitative PCR (qPCR) . These approaches enable precise measurement of receptor expression levels in specifically isolated cell populations.

For comprehensive transcriptomic profiling, single-nucleus RNA sequencing (snRNA-seq) can identify IL-12 receptor expression patterns across the full spectrum of cell types within a tissue . This technique is particularly valuable for analyzing fragile neuroectodermal cells that may be underrepresented in conventional single-cell approaches . By integrating these complementary methods, researchers can develop a comprehensive understanding of IL-12 receptor distribution across tissues and cell types in mouse models.

How should researchers design experiments to evaluate IL-12-mediated effects in conditional knockout mouse models?

Designing rigorous experiments to evaluate IL-12-mediated effects in conditional knockout mouse models requires careful consideration of several critical factors. First, appropriate Cre-driver lines must be selected based on the specific cell populations of interest. For investigating neuronal IL-12 signaling, Nestin-Cre effectively targets neuroectodermal cells, while Cd4-Cre and Vav1-Cre target T cells and all hematopoietic cells, respectively . These conditional approaches allow precise dissection of cell type-specific IL-12 functions that might be obscured in global knockout models.

Comprehensive validation of receptor deletion in target tissues is essential. This should include multiple complementary approaches such as qPCR analysis of sorted cell populations, STAT4 phosphorylation assays following IL-12 stimulation, and immunohistochemical verification when possible . For example, validation of T cell-specific deletion can be performed by analyzing IL-12-induced STAT4 phosphorylation in isolated T cells .

Experimental designs should include all relevant controls: conditional knockout mice (Cre/+ Il12rb2 fl/fl), Cre-negative littermates (Il12rb2 fl/fl), global knockouts (Il12rb2 del/del), and wild-type animals . Having this complete set of controls allows researchers to distinguish cell type-specific effects from global IL-12 deficiency and background strain effects.

Disease models should be carefully selected based on known IL-12 involvement. For neuroinflammation, the MOG-induced EAE model has proven valuable for revealing IL-12's neuroprotective effects . Disease progression should be monitored using both clinical parameters (e.g., EAE scoring) and molecular/cellular analyses (e.g., histopathology, transcriptomics) to capture the full spectrum of phenotypic changes in conditional knockout models.

What are the technical considerations for generating IL-12-expressing lentiviral vectors for mouse studies?

Generating IL-12-expressing lentiviral vectors for mouse studies involves several technical considerations to ensure optimal expression and experimental outcomes. The molecular cloning process should begin with selecting appropriate plasmids containing the mouse IL-12 coding sequence, such as pORF-mIL12 (IL-12elasti(p35::p40)) . Strategic modification of these plasmids to create suitable restriction enzyme sites (e.g., EcoRI and BamHI) flanking the IL-12 gene facilitates efficient subcloning into lentiviral vector backbones .

Promoter selection significantly impacts expression levels and patterns. The elongation factor 1 alpha (EF1α) promoter provides strong, constitutive expression suitable for most applications . The resulting construct (e.g., pHR-cPPT-EF1α-muIL-12-WPRE) should undergo thorough verification through diagnostic restriction enzyme digestion and DNA sequencing before proceeding to virus production .

Lentiviral vector production requires optimization of the transient triple-transfection method using 293T cell monolayers . Vector titers should be carefully estimated using parallel production of reporter viruses (e.g., LV/enGFP) tested on naïve 293T cells to ensure consistent transduction efficiency across experiments .

For target cell transduction, the multiplicity of infection (MOI) must be optimized; an MOI of approximately 20 has been effective for murine pre-B leukemia cell lines like 70Z3-L . Following transduction, establishing stable IL-12-expressing cell lines through single-cell cloning by limiting dilution (at densities below 0.3 cells/well) ensures homogeneous expression . Comprehensive experimental controls should include vector-only transduced cells, untransduced cells, and appropriate in vivo controls such as PBS-injected and untreated groups .

What methods enable quantitative assessment of IL-12 activity in mouse tissue samples?

Quantitative assessment of IL-12 activity in mouse tissue samples requires multi-faceted approaches that extend beyond simple protein detection. For baseline measurement, enzyme-linked immunosorbent assays (ELISAs) specific for the biologically active IL-12p70 heterodimer (rather than individual subunits) can quantify cytokine levels in tissue lysates, with detection becoming reliable around disease onset in neuroinflammatory conditions (approximately 10 days post-immunization) .

Functional assessment of IL-12 bioactivity can be performed using reporter cell systems such as HEK-Blue IL-12 cells, which secrete embryonic alkaline phosphatase (SEAP) in response to IL-12 stimulation . The effective dose (ED50) for this response is typically ≤15 ng/mL for bioactive recombinant mouse IL-12, providing a quantitative benchmark for activity assessment .

Analysis of downstream signaling events offers another quantitative approach, particularly phosphorylation of STAT4, a key transcription factor in the IL-12 signaling pathway . Phospho-specific flow cytometry or western blotting can measure STAT4 activation in isolated cells or tissue lysates following IL-12 exposure, providing a proximal readout of receptor functionality.

For comprehensive assessment of IL-12's biological effects, measurement of secondary cytokine production (particularly IFN-γ and TNF-α) in tissue culture supernatants or by intracellular cytokine staining of tissue-derived cells provides functional readouts of IL-12 activity . In neuroinflammation models, single-nucleus RNA sequencing can quantify transcriptional changes in neuroprotective genes across multiple cell types, revealing the functional consequences of IL-12 signaling in complex tissues .

How can researchers reconcile IL-12's pro-inflammatory versus neuroprotective roles in mouse disease models?

This apparent paradox can be resolved by understanding that IL-12 acts on distinct cell types with different downstream consequences. In immune cells, IL-12 promotes inflammatory cytokine production and Th1 polarization . Conversely, in neurons, IL-12 receptor signaling maintains the expression of genes involved in neuronal survival and neuroprotection during inflammatory challenges .

Experimental evidence supporting this reconciliation comes from conditional knockout studies demonstrating that while global IL-12 receptor deficiency exacerbates experimental autoimmune encephalomyelitis (EAE), selective deletion in T cells or all hematopoietic cells does not worsen disease outcomes . This indicates that IL-12's protective effects are mediated through non-immune, neuroectodermal cells rather than through modulation of the inflammatory response itself.

The temporal dynamics of IL-12 signaling add another layer of complexity—early IL-12 signaling in neurons may establish neuroprotective programs that prepare the CNS for subsequent inflammatory challenges, representing an adaptive response rather than contradicting its pro-inflammatory functions in immune cells .

What factors contribute to variability in IL-12-based experimental outcomes in mouse models?

Multiple factors contribute to variability in IL-12-based experimental outcomes in mouse models, requiring careful experimental design and interpretation. Genetic background represents a major source of variability, as different mouse strains exhibit distinct immune characteristics and susceptibilities to IL-12-responsive disease models . This genetic influence necessitates consistent strain usage across experiments and appropriate littermate controls.

The quality and preparation of recombinant IL-12 significantly impacts experimental consistency. Variations in protein purity (which should be ≥95%), endotoxin contamination (which should be ≤0.1 EU/μg), and biological activity (ED50 ≤15 ng/mL) can substantially affect outcomes . Researchers should adhere to standardized reconstitution protocols that avoid vortexing and minimize freeze-thaw cycles .

The timing, duration, and route of IL-12 administration relative to disease progression represent critical variables. In neuroinflammation models, endogenous IL-12 levels begin to increase around clinical disease onset (approximately 10 days post-immunization), suggesting that intervention timing should consider this natural kinetic . Similarly, systemic versus localized delivery methods may produce dramatically different outcomes in cancer immunotherapy models .

Environmental factors including housing conditions, microbiome composition, and stress levels can modulate baseline immune status and subsequent IL-12 responsiveness. Standardizing these conditions across experimental cohorts helps reduce unexplained variability.

Technical approaches for measuring outcomes also contribute to apparent variability. Using multiple complementary readouts—combining functional assessments, transcriptional analysis, and protein-level measurements—provides a more robust evaluation of IL-12's effects than relying on single outcome measures.

How should single-cell transcriptomic data be analyzed to understand IL-12's cell type-specific effects in complex tissues?

Analyzing single-cell transcriptomic data to understand IL-12's cell type-specific effects requires sophisticated computational approaches that account for cellular heterogeneity and complex signaling networks. When examining single-nucleus RNA sequencing (snRNA-seq) datasets from IL-12 receptor-deficient versus control tissues, researchers should first ensure rigorous quality control, removing low-quality nuclei, adjusting for batch effects, and normalizing data appropriately before proceeding to cell type identification .

Cell clustering should employ unbiased methods that identify populations based on their transcriptional signatures rather than predefined markers. This approach has revealed that IL-12 receptor signaling affects diverse cell populations differently, with pronounced effects in granule cells, excitatory neurons, oligodendrocytes, cholinergic neurons, and molecular layer interneurons . After identifying cell clusters, differential expression analysis between genotypes should be performed within each cell type separately to capture population-specific responses to IL-12 signaling .

Pathway enrichment analysis rather than individual gene analysis provides more robust biological insights. In neuronal populations, IL-12 receptor deficiency has been associated with downregulation of pathways involved in neuronal survival and neuroprotection . These pathway-level analyses help identify coherent functional changes that might be missed when focusing solely on top differentially expressed genes.

Integration of transcriptomic data with other experimental approaches strengthens interpretation. Validating key findings from snRNA-seq using techniques such as multiplexed RNA fluorescence in situ hybridization (RNAscope) or quantitative PCR on sorted cell populations confirms transcriptional changes and provides spatial context that might be lost in dissociated single-cell approaches .

What emerging technologies could advance understanding of IL-12 biology in mouse models?

Emerging technologies poised to advance IL-12 biology research include spatial transcriptomics methods that preserve tissue architecture while providing transcriptome-wide information about IL-12 and its receptor expression patterns. Unlike traditional single-cell approaches that lose spatial information, techniques such as Slide-seq, Visium, and MERFISH can map IL-12 signaling networks within their native tissue contexts, particularly valuable for understanding IL-12's neuroprotective effects in specific brain regions .

CRISPR-based genetic screening approaches in vivo offer unprecedented opportunities to systematically identify genes involved in IL-12 signaling pathways across different cell types. This could reveal novel components of neuron-specific IL-12 response pathways distinct from those in immune cells, potentially explaining the dichotomous functions of this cytokine .

Advanced protein-protein interaction mapping technologies such as proximity labeling (BioID, APEX) applied in vivo could elucidate cell type-specific IL-12 receptor complexes and downstream signaling components. This might identify neuron-specific signaling partners that mediate protective rather than inflammatory responses .

Intravital imaging approaches using fluorescent IL-12 reporter mice combined with two-photon microscopy could visualize IL-12 production and signaling dynamics in real-time during neuroinflammation or cancer progression, providing insights into the temporal aspects of IL-12 biology that are currently challenging to capture .

Advancements in delivery technologies, including nanoparticle-based approaches and engineered cell therapies expressing IL-12 under specific conditional promoters, could enable precise spatiotemporal control of IL-12 activity in mouse models, facilitating more nuanced studies of its context-dependent functions .

Product Science Overview

Structure and Composition

Mouse recombinant IL-12 is a heterodimer consisting of two subunits: p35 and p40. The p40 subunit is a 40 kDa protein, while the p35 subunit is a 35 kDa protein. Together, they form the active IL-12p70 heterodimer, which has a total molecular mass of approximately 70 kDa . The recombinant form of IL-12 is produced using various expression systems, such as HEK293 cells, to ensure high purity and biological activity .

Biological Functions

IL-12 is a key cytokine in the immune response, with several important functions:

  1. Activation of NK Cells: IL-12 stimulates NK cells to produce IFN-γ, which enhances their cytotoxic activity against infected and malignant cells .
  2. T Cell Differentiation: IL-12 promotes the differentiation of naive CD4+ T cells into Th1 cells, which are essential for cell-mediated immunity .
  3. Cytokine Production: IL-12 induces the production of various cytokines, including IFN-γ, by T cells and NK cells .
  4. Enhancement of Cytotoxic Activity: IL-12 enhances the cytotoxic activity of CD8+ T cells and NK cells, contributing to the elimination of intracellular pathogens and tumor cells .
Applications in Research

Recombinant mouse IL-12 is widely used in immunological research to study its effects on various immune cells and to investigate its potential therapeutic applications. Some common applications include:

  • Cell Culture: IL-12 is used to stimulate and differentiate immune cells in vitro, providing insights into its role in immune responses .
  • Functional Assays: IL-12 is employed in functional assays to assess its effects on cytokine production, cell proliferation, and cytotoxic activity .
  • Animal Models: Recombinant IL-12 is used in animal models to study its therapeutic potential in diseases such as cancer, infectious diseases, and autoimmune disorders .
Therapeutic Potential

IL-12 has shown promise as a therapeutic agent due to its ability to enhance immune responses. It has been investigated in various clinical trials for its potential use in treating cancer, infectious diseases, and autoimmune disorders. However, its clinical application is limited by its potential side effects, such as systemic inflammation and toxicity .

In conclusion, Interleukin-12 (Mouse Recombinant) is a vital cytokine with significant roles in the immune system. Its recombinant form is a valuable tool in immunological research, providing insights into its functions and potential therapeutic applications.

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.