Interleukin-12 (IL-12) is a heterodimeric glycoprotein composed of α and β subunits linked by a disulfide bond and is a potent anticancer agent . The protein, also known as IL12RB1(CD212), functions as an interleukin receptor that binds interleukin-12 with low affinity and participates in IL-12 transduction . It is a type I transmembrane protein belonging to the hemopoietin receptor superfamily . IL12RB1(CD212) associates with IL23R to form the interleukin-23 receptor, which functions in IL-23 signal transduction, likely through activation of the Jak-Stat signaling cascade .
Recombinant proteins, such as Recombinant Lama Glama Interleukin-4 (IL4), are produced using expression systems like E. coli . These recombinant proteins often include tags like N-6His to aid in purification and detection .
| Description | Details |
|---|---|
| Purity | Greater than 90% as determined by SDS-PAGE |
| Uniprotkb | Q865X5 |
| Target Symbol | IL4 |
| Species | Lama glama (Llama) |
| Expression System | E.coli |
| Tag | N-6His |
| Target Protein Sequence | HKCDITLQEIIKTLNTLTARKNSCMELTVADVFAAPKNTTEKETFCKAATALRHIYRHHNCLSKHLSGLDRNLSGLANTTCSVNDSKKSTLRDFLERLKKIMKEKYSKC |
| Expression Range | 25-133aa |
| Protein Length | Full Length of Mature Protein |
| Mol. Weight | 18.4 kDa |
| Research Area | Cancer |
| Form | Liquid or Lyophilized powder |
| Buffer | Liquid form: default storage buffer is Tris/PBS-based buffer, 5%-50% glycerol. Lyophilized powder form: the buffer before lyophilization is Tris/PBS-based buffer, 6% Trehalose, pH 8.0. |
| Reconstitution | Briefly centrifuged the vial prior to opening to bring the contents to the bottom. Reconstitute protein in deionized sterile water to a concentration of 0.1-1.0 mg/mL. It is recommended to add 5-50% of glycerol (final concentration) and aliquot for long-term storage at -20°C/-80°C. The default final concentration of glycerol is 50%. |
| Storage | 1. Store at -20°C/-80°C upon receipt, aliquoting is necessary for mutiple use. 2. Avoid repeated freeze-thaw cycles. 3. Store working aliquots at 4°C for up to one week. 4. In general, protein in liquid form is stable for up to 6 months at -20°C/-80°C. Protein in lyophilized powder form is stable for up to 12 months at -20°C/-80°C. |
IL-12 family members play a role in cardiovascular diseases . Elevated serum IL-12 levels are observed in atherosclerosis, and increased IL-12 levels are associated with the progression of atherosclerosis . Studies show that treatment with exogenous recombinant murine IL-12 significantly aggravates the progression of atherosclerosis and increases aortic atherosclerotic plaque areas in both ApoE-knockout mice and LDL receptor-deficient mice .
IL-12 expression is increased in both plasma and heart tissue of coxsackievirus B3-induced viral myocarditis in mice . Elevated IL-12R levels were also found in heart tissue of mice with viral myocarditis . IL-12 expression was found to be elevated in patients with autoimmune cardiomyopathy or alcoholic cardiomyopathy .
Recombinant human interleukin-12 (rhIL-12) can prevent radiation damage, improve hematopoietic function, regulate immunity, and reduce the side effects of radiotherapy . Clinical studies explore therapeutic strategies using IL-12 in cancer therapy and its protective role in hematological recovery in chemoradiotherapy . In patients with melanoma given a combination of IL-12 and PD-1-targeted therapy, the disease control rate reached 59%, and 41% of patients had a complete response .
IL-12R gene polymorphisms, including IL-12B 3' UTR C and IL-12B 3' UTR CC, result in significantly higher gene expression and may increase the incidence of Chagas cardiomyopathy . Individuals with IL-12, IL-23R polymorphisms are more susceptible to dilated cardiomyopathy among the Chinese Han population .
Interleukin-12 subunit beta (IL12B), also known as natural killer cell stimulatory factor 2, cytotoxic lymphocyte maturation factor p40, or interleukin-12 subunit p40, is a protein subunit encoded by the IL12B gene. This 40 kDa cytokine receptor-like subunit forms a disulfide-linked heterodimer with the 35 kDa subunit encoded by IL12A to create the biologically active IL-12 cytokine . IL12B serves as a common subunit for both interleukin-12 and interleukin-23, playing crucial roles in immune regulation .
Functionally, IL12B is primarily expressed by activated macrophages and serves as an essential inducer of T helper 1 (Th1) cell development. The cytokine acts on T cells and natural killer (NK) cells, exerting a broad array of biological activities . Its primary mechanism of action involves binding to the IL-12 receptor complex, which activates the JAK-STAT signaling pathway, particularly through JAK2/STAT4, promoting Th1 cell differentiation and enhancing the production of interferon-gamma (IFN-γ) by T cells and NK cells .
IL12B plays a key role in maintaining sufficient numbers of memory/effector Th1 cells to mediate long-term protection against intracellular pathogens and tumor cells . This cytokine is essential for establishing cell-mediated immune responses and orchestrating Th1-type immune reactions, making it a critical component of the immune system's defense mechanisms.
While the search results don't provide specific structural information about Lama glama IL12B, human IL12B has been well characterized. In humans, IL12B forms a heterodimer with IL12A through disulfide linkages to create the biologically active IL-12 cytokine .
For researchers investigating Lama glama IL12B, a comparative analysis approach would be recommended. This would typically involve sequence alignment with human IL12B and other mammalian orthologs to identify conserved domains and species-specific variations. The methodology for such analysis would include:
Obtaining the genomic or cDNA sequence of Lama glama IL12B
Performing multiple sequence alignments with known IL12B sequences from humans and other mammals
Identifying conserved functional domains, signal peptides, and post-translational modification sites
Constructing phylogenetic trees to understand evolutionary relationships
Using protein structure prediction software to model the tertiary structure of Lama glama IL12B
Given the evolutionary conservation of cytokine functions across mammals, researchers should expect substantial homology in functional domains while recognizing potential species-specific adaptations in regulatory regions and surface epitopes.
For expression of recombinant Lama glama IL12B, researchers should consider expression systems that have proven successful for other mammalian cytokines. Based on general recombinant protein methodologies, the following systems would be recommended:
Mammalian expression systems: CHO or HEK293 cells often provide proper folding and post-translational modifications critical for cytokine functionality. These systems are particularly valuable when glycosylation patterns are important for biological activity.
Insect cell expression systems: Baculovirus-infected insect cells (Sf9, Sf21, or High Five) offer advantages for expression of complex mammalian proteins while maintaining proper folding.
Yeast expression systems: Pichia pastoris can be suitable for secreted proteins like IL12B and offers high yields with eukaryotic processing.
For experimental methodology, researchers should:
Clone the Lama glama IL12B coding sequence into appropriate expression vectors
Optimize codons if necessary for the chosen expression system
Include appropriate tags (His, FLAG, etc.) for purification while ensuring they don't interfere with biological activity
Establish stable cell lines or optimize transient expression conditions
Develop purification protocols using affinity chromatography followed by size exclusion or ion exchange steps
The choice of expression system should be influenced by the intended experimental applications, required protein yield, and the importance of post-translational modifications for the specific research objectives.
While specific data on Lama glama IL12B polymorphisms is not available in the search results, research approaches can be extrapolated from human IL12B studies. In humans, promoter polymorphisms of the IL12B gene have been associated with varying severity of atopic and non-atopic asthma in children .
For researchers investigating IL12B polymorphisms in Lama glama, the following methodological approach would be recommended:
Polymorphism identification:
Sequence the IL12B gene and promoter regions from multiple Lama glama individuals
Compare with established databases to identify single nucleotide polymorphisms (SNPs), insertions/deletions, and copy number variations
Focus particularly on promoter regions, as these may affect expression levels
Functional analysis:
Develop reporter gene assays using different promoter variants
Measure expression levels of IL12B variants in response to various stimuli
Assess the impact of coding region polymorphisms on protein structure and function
Correlation with immune phenotypes:
Design challenge studies using relevant pathogens
Measure cytokine production, T cell differentiation, and pathogen clearance
Correlate immune responses with specific IL12B genotypes
This research would be particularly relevant for understanding Lama glama's unique adaptations to high-altitude environments and resistance to specific pathogens endemic to their natural habitat.
To investigate IL12B-mediated signaling in Lama glama immune cells, researchers should adapt techniques used for studying JAK-STAT signaling in other species. Based on established knowledge about IL12B signaling in humans, the following methodological approaches would be effective:
Phospho-flow cytometry:
Isolate peripheral blood mononuclear cells (PBMCs) from Lama glama
Stimulate cells with recombinant IL12B
Use phospho-specific antibodies to detect STAT4 phosphorylation
Analyze by flow cytometry to identify responding cell populations
Western blotting and immunoprecipitation:
Treat Lama glama immune cells with recombinant IL12B
Harvest cells at various time points
Perform Western blots with antibodies against phosphorylated forms of JAK2 and STAT4
Use immunoprecipitation to identify interacting proteins in the signaling complex
Transcriptomic analysis:
Stimulate Lama glama immune cells with IL12B
Perform RNA-seq to identify genes regulated by IL12B signaling
Compare with known IL12B-responsive genes in humans and other species
Use pathway analysis to identify potentially unique aspects of IL12B signaling in Lama glama
Inhibitor studies:
Use specific JAK-STAT pathway inhibitors to block IL12B signaling
Measure the effect on downstream functions including IFN-γ production
Determine if alternative signaling pathways exist in Lama glama
These approaches would provide comprehensive data on IL12B signaling in Lama glama and allow comparison with other species to identify conserved and divergent aspects of this important immune pathway.
For researchers investigating the cross-species bioactivity of Lama glama IL12B compared to human IL12B, the following experimental approach would be recommended:
Production of recombinant proteins:
Express and purify both Lama glama and human IL12B using identical expression systems
Confirm proper folding and heterodimer formation with respective IL12A subunits
Validate purity by SDS-PAGE and identity by mass spectrometry
Receptor binding assays:
Perform comparative binding studies using surface plasmon resonance
Measure binding kinetics to IL12 receptors from different species
Compare binding affinities to determine species specificity
Cellular response assays:
Test biological activity on immune cells from multiple species
Measure:
STAT4 phosphorylation
IFN-γ production
Proliferation of activated T cells
NK cell cytotoxicity
In vivo comparative studies:
Evaluate immune responses in appropriate animal models
Compare potency in inducing Th1 polarization across species
Assess species-specific effects on disease models
Evidence from human IL12R beta 2 studies suggests that human and mouse IL12R beta 2 do not bind cross-species IL12 , indicating potential species-specific interactions. Researchers should expect similar species restrictions with Lama glama IL12B and design experiments to characterize these limitations.
While specific information about IL12B in Lama glama autoimmune conditions is not available in the search results, researchers can develop investigations based on known associations in humans. In humans, overexpression of IL12B has been observed in the central nervous system of multiple sclerosis patients, suggesting a role in disease pathogenesis .
To investigate potential roles of IL12B in Lama glama autoimmune conditions:
Baseline expression studies:
Establish normal expression patterns of IL12B in healthy Lama glama tissues
Compare expression levels across different immune cell populations
Develop Lama glama-specific assays (ELISA, qPCR) for IL12B detection
Clinical studies:
Identify Lama glama with suspected autoimmune conditions
Measure IL12B expression in relevant tissues and peripheral blood
Correlate IL12B levels with clinical manifestations and disease progression
Genetic association studies:
Screen for IL12B polymorphisms in Lama glama populations
Analyze association between specific variants and disease susceptibility
Investigate epigenetic modifications affecting IL12B expression
Intervention studies:
Develop neutralizing antibodies against Lama glama IL12B
Test therapeutic potential in Lama glama with autoimmune conditions
Assess changes in inflammatory markers and clinical improvement
This research would not only advance our understanding of Lama glama immunology but could also provide comparative insights relevant to human autoimmune conditions, potentially identifying camelid-specific immune mechanisms with translational value.
For researchers developing purification protocols for recombinant Lama glama IL12B, the following comprehensive methodology is recommended:
Expression system selection:
Mammalian expression systems (preferably CHO or HEK293) are recommended for proper folding and post-translational modifications
Expression vector should include appropriate secretion signal and affinity tag (preferably His-tag or Fc fusion)
Purification strategy:
Initial capture: Affinity chromatography using Ni-NTA for His-tagged protein or Protein A/G for Fc-fusion proteins
Intermediate purification: Ion exchange chromatography (typically anion exchange at pH 8.0)
Polishing: Size exclusion chromatography to remove aggregates and ensure homogeneity
Buffer optimization:
Quality control:
SDS-PAGE (both reducing and non-reducing) to confirm purity and heterodimer formation
Western blot with specific antibodies
Mass spectrometry to confirm identity
Endotoxin testing (<0.1 EU/μg protein)
Functional assays measuring STAT4 phosphorylation or IFN-γ induction
The purified protein should be stored at -20°C in single-use aliquots to avoid freeze-thaw cycles that may compromise activity . For shipping, gel packs should be used to maintain temperature during transport.
Developing specific antibodies against Lama glama IL12B requires a strategic approach to overcome potential cross-reactivity issues and ensure high specificity. The following methodology is recommended:
Antigen design and preparation:
Option 1: Use purified recombinant full-length Lama glama IL12B
Option 2: Design synthetic peptides from unique regions of Lama glama IL12B
Option 3: Use recombinant fragments containing specific domains
All antigens should be validated for purity by SDS-PAGE and mass spectrometry
Immunization strategies:
For polyclonal antibodies:
Immunize rabbits or goats with purified antigen
Use appropriate adjuvants to enhance immunogenicity
Collect serum at multiple time points to monitor antibody development
For monoclonal antibodies:
Immunize mice or rats with purified antigen
Isolate B cells from spleen or lymph nodes
Perform cell fusion with myeloma cells
Screen hybridomas for specific antibody production
Antibody purification and characterization:
Purify antibodies using Protein A/G affinity chromatography
Test specificity by ELISA, Western blot, and immunoprecipitation
Evaluate cross-reactivity with human and other species' IL12B
Perform epitope mapping to identify binding regions
Validation for specific applications:
Flow cytometry: Test on Lama glama PBMCs with appropriate controls
Immunohistochemistry: Validate on fixed tissue sections
Neutralization assays: Confirm ability to block IL12B biological activity
ELISA development: Establish sandwich ELISA for IL12B quantification
For research requiring the highest specificity, recombinant antibody approaches using phage display or yeast display technologies could be employed to select antibodies with optimal binding characteristics.
To effectively study IL12B gene regulation in Lama glama, researchers should implement a multi-faceted approach combining genomic, transcriptomic, and functional analyses:
Promoter and regulatory region characterization:
Transcription factor binding studies:
Perform chromatin immunoprecipitation (ChIP) assays to identify bound transcription factors
Use electrophoretic mobility shift assays (EMSA) to confirm specific binding
Validate with reporter gene assays using luciferase constructs containing promoter fragments
Expression analysis under various stimuli:
Isolate monocytes/macrophages from Lama glama blood
Stimulate with various TLR ligands, cytokines, and pathogens
Measure IL12B mRNA expression by qRT-PCR
Compare expression kinetics with other species
Epigenetic regulation studies:
Analyze DNA methylation patterns in the IL12B locus using bisulfite sequencing
Perform ChIP assays for histone modifications associated with active/repressed chromatin
Investigate the effects of epigenetic modifiers on IL12B expression
CRISPR-based approaches:
Develop CRISPR/Cas9 systems to modify specific regulatory elements
Establish Lama glama cell lines with reporter constructs integrated at the IL12B locus
Use CRISPRa/CRISPRi to activate or repress the endogenous IL12B gene
This comprehensive approach would provide valuable insights into the unique aspects of IL12B regulation in Lama glama and allow comparison with regulatory mechanisms in other species.
Recombinant Lama glama IL12B has significant potential for immunotherapeutic applications, leveraging the unique properties of camelid proteins. The following methodological approaches would be valuable for researchers in this area:
Cancer immunotherapy applications:
Develop fusion proteins combining Lama glama IL12B with:
Tumor-targeting antibodies or antibody fragments
Camelid single-domain antibodies (nanobodies) recognizing tumor antigens
Evaluate anti-tumor activity in appropriate models
Compare efficacy and toxicity profiles with human IL12B-based immunotherapies
Vaccine adjuvant development:
Incorporate recombinant Lama glama IL12B into vaccine formulations
Assess enhancement of antigen-specific immune responses
Evaluate Th1 polarization and memory cell generation
Compare adjuvant properties with human IL12B
Anti-infectious disease applications:
Test efficacy in enhancing immunity against intracellular pathogens
Develop delivery systems for targeted IL12B administration
Evaluate combinatorial approaches with other immunomodulators
Xenogeneic applications:
Investigate if Lama glama IL12B has advantages over human IL12B in terms of:
Reduced immunogenicity in heterologous systems
Extended half-life in circulation
Novel receptor binding properties
Superior stability under various conditions
This research would not only advance our understanding of camelid cytokine biology but could potentially lead to novel immunotherapeutic agents with advantages over existing human IL12B-based approaches.
To investigate evolutionary adaptations in Lama glama IL12B, researchers should employ comprehensive comparative genomic and molecular evolutionary analyses:
Sequence acquisition and alignment:
Obtain IL12B sequences from Lama glama and other camelids
Include sequences from diverse mammalian taxa for comprehensive comparison
Perform multiple sequence alignments using tools like MUSCLE or MAFFT
Identify conserved domains and variable regions
Phylogenetic analysis:
Construct phylogenetic trees using maximum likelihood or Bayesian approaches
Determine the relationship of Lama glama IL12B to other mammalian orthologs
Identify lineage-specific clustering patterns
Selection pressure analysis:
Calculate dN/dS ratios to identify sites under positive or purifying selection
Use branch-site models to detect episodic selection in the camelid lineage
Identify selection signatures potentially related to adaptation to high-altitude environments
Structural biology approaches:
Predict 3D structures of Lama glama IL12B using homology modeling
Compare with crystal structures of human IL12B
Identify structural adaptations that might affect:
Receptor binding interfaces
Heterodimer formation
Protein stability under environmental stressors
Regulatory region evolution:
Compare promoter and enhancer sequences across species
Identify conserved and divergent transcription factor binding sites
Correlate with known differences in expression patterns or inducibility
This research would provide insights into how evolutionary pressures have shaped IL12B function in camelids and potentially identify adaptive features related to their unique ecological niches and immune challenges.
Researchers working with recombinant Lama glama IL12B may encounter solubility and stability challenges. The following methodological approaches can help address these issues:
Expression optimization:
Test multiple secretion signal sequences to improve folding
Co-express with IL12A to promote proper heterodimer formation
Evaluate different cell lines (HEK293, CHO, Expi293) for optimal expression
Optimize culture conditions (temperature, media composition, induction timing)
Protein engineering approaches:
Introduce stabilizing mutations based on computational predictions
Create fusion proteins with solubility-enhancing partners (SUMO, thioredoxin)
Consider removing hydrophobic regions that might promote aggregation
Evaluate the impact of glycosylation site modifications
Buffer optimization:
Systematic screening of buffer conditions:
pH range (6.0-8.0)
Salt concentrations (0-500 mM NaCl)
Stabilizing additives (glycerol, sucrose, arginine, trehalose)
Detergents (Tween-20, Tween-80) at low concentrations
Use differential scanning fluorimetry to assess thermal stability
Storage and handling:
Analytical methods to monitor stability:
Size exclusion chromatography to detect aggregation
Dynamic light scattering to assess particle size distribution
Circular dichroism to monitor secondary structure changes
Activity assays to confirm biological function maintenance
By systematically addressing these aspects, researchers can develop robust protocols for producing and maintaining stable, active recombinant Lama glama IL12B for various research applications.
Developing specific detection assays for Lama glama IL12B presents several challenges, particularly regarding cross-reactivity with homologous proteins from other species. The following methodological approaches can help researchers overcome these challenges:
Epitope mapping and antibody development:
Identify unique epitopes in Lama glama IL12B through sequence analysis
Design peptides representing these unique regions
Generate and screen antibodies for specificity against:
Recombinant Lama glama IL12B
Human IL12B
Other mammalian IL12B orthologs
Select antibody pairs recognizing distinct epitopes for sandwich assays
ELISA development strategy:
For sandwich ELISA:
Use a capture antibody targeting a conserved epitope
Employ a detection antibody against a Lama glama-specific region
Optimize antibody concentrations and incubation conditions
Validate with recombinant standards and native samples
For competitive ELISA:
Develop with Lama glama-specific detection antibodies
Use biotinylated or enzyme-linked Lama glama IL12B as competitor
Validation approaches:
Test assay performance with:
Recombinant Lama glama IL12B spiked into various matrices
Lama glama biological samples (serum, cell culture supernatants)
Samples containing potential cross-reactive proteins
Establish standard curves and determine:
Lower and upper limits of quantification
Precision (intra- and inter-assay CV%)
Accuracy (recovery of spiked standards)
Specificity (cross-reactivity with related proteins)
Alternative detection technologies:
Develop aptamer-based detection systems for increased specificity
Consider surface plasmon resonance for label-free detection
Explore proximity ligation assays for improved sensitivity and specificity
Implement multiplexed bead-based assays to detect IL12B alongside other cytokines
By employing these strategies, researchers can develop robust, specific assays for detecting and quantifying Lama glama IL12B in various experimental and clinical samples.