The IL6R Mouse is a genetically humanized rodent model where the endogenous interleukin-6 receptor alpha (Il6ra) gene is replaced with the human IL6R gene via knock-in technology. This humanization enables mice to express functional human IL6R, including both membrane-bound and soluble forms, mimicking human IL-6 signaling systems . Key structural features include:
Human IL6R expression: Replaces mouse Il6ra, preserving native regulatory elements for tissue-specific expression .
Soluble hIL6R levels: Plasma concentrations (15–30 ng/mL in homozygotes) align with human values, unlike earlier transgenic models with excessive soluble receptor levels .
IL6R Mice are critical for studying human-specific therapies and pathologies.
Tissue Distribution: Human IL6R expression mirrors endogenous mouse Il6ra patterns (e.g., liver, spleen) .
Signal Transduction: Requires gp130 (signal transducer) for IL-6-mediated responses, similar to humans .
Therapy | Mechanism | Model Validation |
---|---|---|
Tocilizumab | Neutralizes IL-6R, blocking IL-6 signaling . | Castleman’s, AAMR |
Sarilumab | IL-6R antagonist; efficacy in autoimmune diseases . | EAMG models |
Disease Model | Outcome |
---|---|
Experimental Autoimmune Myasthenia Gravis (EAMG) | IL-6R blockade reduces neuromuscular junction IgG deposition and muscle weakness . |
Hepatocyte-Specific IL6R Knockout: Demonstrates membrane-bound IL6R is critical for acute-phase responses (e.g., turpentine-induced SAA production) .
IL-6Rα Deficiency: Mimics IL-6 knockout in inflammatory resolution but not in wound healing, revealing IL-6Rα-independent roles .
This IL-6R protein solution has a concentration of 1 mg/ml and contains phosphate-buffered saline (pH 7.4) and 10% glycerol. |
IL-6 receptor subunit beta, IL-6R subunit beta, IL-6R-beta, IL-6RB, Interleukin-6 signal transducer, Membrane glycoprotein 130, gp130, Oncostatin-M receptor subunit alpha,CD130.
QLLEPCGYIY PEFPVVQRGS NFTAICVLKE ACLQHYYVNA SYIVWKTNHA AVPREQVTVI NRTTSSVTFT DVVLPSVQLT CNILSFGQIE QNVYGVTMLS GFPPDKPTNL TCIVNEGKNM LCQWDPGRET YLETNYTLKS EWATEKFPDC QSKHGTSCMV SYMPTYYVNI EVWVEAENAL GKVSSESINF DPVDKVKPTP PYNLSVTNSE ELSSILKLSW VSSGLGGLLD LKSDIQYRTK DASTWIQVPL EDTMSPRTSF TVQDLKPFTE YVFRIRSIKD SGKGYWSDWS EEASGTTYED RPSRPPSFWY KTNPSHGQEY RSVRLIWKAL PLSEANGKIL DYEVILTQSK SVSQTYTVTG TELTVNLTND RYVASLAARN KVGKSAAAVL TIPSPHVTAA YSVVNLKAFP KDNLLWVEWT PPPKPVSKYI LEWCVLSENA PCVEDWQQED ATVNRTHLRG RLLESKCYQI TVTPVFATGP GGSESLKAYL KQAAPARGPT VRTKKVGKNE AVLAWDQIPV DDQNGFIRNY SISYRTSVGK |
IL-6R (Interleukin-6 Receptor) in mice consists of the IL-6R α-chain (IL-6Rα, also known as CD126) and the signal transducer glycoprotein 130 (gp130). When IL-6 binds to IL-6Rα, this complex engages gp130 to initiate intracellular signaling, primarily through STAT3 activation. Recent research has demonstrated that IL-6Rα is the only biologically relevant receptor for IL-6 in mice, as IL-6Rα deficiency completely prevents downstream STAT3 activation in response to IL-6 .
IL-6 signaling occurs through two distinct mechanisms: classic signaling (via membrane-bound IL-6Rα) and trans-signaling (via soluble IL-6Rα). Trans-signaling has been implicated in various inflammatory and developmental processes, particularly in the brain where it can produce selective effects without triggering a general neuroinflammatory response .
Several key IL-6R mouse models have been developed for different research applications:
IL-6Rα knockout mice (Il6ra−/−): These mice completely lack IL-6Rα expression, preventing IL-6 signaling. Studies with these mice demonstrate that IL-6Rα ablation protects from IL-6-triggered inflammation and prevents downstream STAT3 activation .
IL-6Rα heterozygous mice (Il6ra+/−): These mice have reduced IL-6Rα expression, allowing for the study of dose-dependent effects of IL-6 signaling .
CD11c-Cre-dependent IL-6 overexpression model: In this model, murine IL-6 is overexpressed, leading to immune dysregulation characterized by increased neutrophils, monocytes/macrophages, T cell activation, and suppressed B-1a cell development .
Humanized IL-6R mouse model: This model has the endogenous mouse IL-6 receptor gene replaced with human IL6R, making it particularly valuable for evaluating human IL-6R-specific therapeutic agents .
Ischemia-reperfusion (I/R) injury model: Used to study the effects of IL-6R blockade on cardiac remodeling through administration of monoclonal antibodies like MR16-1 .
Understanding the differences between mouse and human IL-6R is critical for translational research. Many therapeutic monoclonal antibodies developed against human IL-6R have "no or low cross-reactivity to orthologous molecules of animals other than primates" . This limitation has driven the development of humanized IL-6R mouse models, where the endogenous IL-6 receptor gene is replaced by human IL6R.
These humanized models provide several advantages: they allow evaluation of human IL-6R-specific therapeutic agents in a smaller animal model, require less candidate agent material, and leverage the well-characterized mouse experimental system . For instance, humanized Castleman's disease mouse models have successfully demonstrated that plasma levels of human soluble IL-6R and human IL-6 were elevated after tocilizumab treatment, mirroring clinical observations in human patients .
For investigating IL-6 trans-signaling in the developing mouse brain, consider using Hyper IL-6 (HIL-6), a fusion protein of IL-6 bound to IL-6Rα by a short peptide chain. This chimeric protein has 100-fold higher receptor binding affinity than IL-6/IL-6Rα and provides enhanced and longer activation of STAT3-dependent and mitogen-activated protein kinase pathways .
Key experimental design considerations include:
Dose selection: Test multiple doses (e.g., 5ng and 10ng) to establish dose-responsive effects. In previous studies, 5ng was sufficient for qRT-PCR analysis of myelin-specific genes, while 10ng was used for less sensitive array procedures .
Administration timing: For developmental studies, expose the early postnatal brain to HIL-6, as this timing captures critical neurodevelopmental windows .
Control groups: Include vehicle-treated animals and, if applicable, animals receiving control IgG antibodies .
Endpoint selection: Measure changes in myelin-specific genes (MBP, PLP/DM20), assess complexity of MBP processes in the cortex by immunohistochemistry, and analyze the non-hydroxy cerebroside fraction of cerebral lipids .
Sample size determination: Use a single pup per litter per dose per endpoint to avoid litter effects .
Based on cardiac ischemia-reperfusion studies with the monoclonal antibody MR16-1, a comprehensive approach to evaluating IL-6R blockade efficacy should include:
Functional assessments:
Cardiac Magnetic Resonance Imaging (CMR) to measure left ventricular ejection fraction (LVEF), volumes (LVEDV, LVESV), stroke volume, and mass (LVM) .
Hemodynamic measurements including rate of LV pressure rise and fall (dP/dT min, dP/dT max), LV end diastolic pressures, and maximum aortic pressure .
Histological assessments:
Molecular assessments:
Dosing strategy:
A comprehensive data table should be maintained to record all measurements, as shown in this example from cardiac research:
Parameter | Control Group | IL-6R Blockade Group | Statistical Significance |
---|---|---|---|
LVEF (%) | 35 ± 6 | 28 ± 4 | p = 0.02 |
LVEDV (μl) | 75 ± 16 | 78 ± 10 | NS |
dP/dT max (mmHg/s) | 6506 (6065;6736) | 5797 (5440;6522) | NS |
% Fibrosis | Similar between groups | Similar between groups | NS |
Distinguishing between classical IL-6 signaling (via membrane-bound IL-6Rα) and trans-signaling (via soluble IL-6Rα) requires specific experimental approaches:
Use of specialized reagents:
Genetic approaches:
Cell-type specific analysis:
Determine which cell populations express membrane-bound IL-6Rα (capable of classical signaling).
Cells lacking membrane IL-6Rα but expressing gp130 can only respond to IL-6 through trans-signaling.
Target gene analysis:
Generating a humanized IL-6R mouse model involves replacing the endogenous mouse IL-6 receptor gene with its human counterpart. While detailed methodological steps weren't provided in the search results, the successful creation of such models has been reported .
Key validation steps should include:
Genetic confirmation:
PCR-based genotyping to confirm successful replacement of mouse Il6ra with human IL6R.
Sequencing to verify the integrity of the inserted human gene.
Expression analysis:
qRT-PCR to confirm human IL6R mRNA expression.
Flow cytometry to verify human IL-6R protein expression on relevant cell surfaces.
ELISA to measure soluble human IL-6R in plasma.
Functional validation:
Confirm the ability of human IL-6 to signal through the humanized receptor.
Demonstrate STAT3 activation in response to human IL-6 stimulation.
Show that human-specific IL-6R antibodies (e.g., tocilizumab) can block signaling in this model.
Pharmacological response:
Optimal dosing of IL-6R modulators depends on the specific research question, model system, and reagent used. From the available studies, we can extract the following guidance:
For Hyper IL-6 (HIL-6) in developmental brain studies:
For IL-6R blockade with monoclonal antibody MR16-1 in cardiac I/R models:
The experimental design should consider:
Target tissue accessibility (blood-brain barrier penetration for CNS studies)
Half-life of the modulator (approximately 2 hours for HIL-6)
Duration of desired effect (acute vs. chronic)
Potential compensatory mechanisms that may develop with long-term modulation
Pilot dose-finding studies are recommended when establishing new experimental paradigms to determine both efficacy and potential toxicity.
When faced with contradictory results in IL-6R mouse studies, consider these analytical approaches:
Evaluate model differences:
Examine experimental design variables:
Timing of intervention: Preventive vs. therapeutic approaches may have opposite effects.
Dosing regimens: Different doses may activate different pathways or compensatory mechanisms.
Age and sex of animals: IL-6 signaling may have different impacts at different developmental stages.
Consider signaling specificity:
Analyze unexpected findings systematically:
Validate key findings with multiple approaches:
Combine genetic models (knockout) with pharmacological interventions (antibody blockade).
Assess outcomes using complementary methods (e.g., both imaging and histology).
IL-6R mouse models provide valuable insights into neuroinflammation and neurodevelopmental processes:
Developmental neurotoxicity assessment:
IL-6 signaling affects the developing brain, particularly myelination processes .
HIL-6 exposure in early postnatal brain can decrease mRNA levels for myelin basic protein (MBP) and proteolipid protein (PLP/DM20) .
Immunohistochemistry reveals reduced complexity of MBP processes in the cortex following IL-6 exposure .
Selective effects without general neuroinflammation:
Behavioral outcomes:
Translational relevance:
Myelin composition analysis:
IL-6R mouse models have revealed critical insights into immune dysregulation mechanisms:
Cellular immune effects:
Receptor dependency:
Signaling mechanisms:
Therapeutic implications:
Organ-specific effects:
IL-6R mouse models provide critical platforms for evaluating therapeutic agents targeting IL-6 signaling:
Humanized models for human-specific therapies:
Humanized Castleman's disease mouse models with human IL6R allow direct testing of human-specific antibodies like tocilizumab .
These models overcome the limitation that many therapeutic monoclonal antibodies have "no or low cross-reactivity to orthologous molecules of animals other than primates" .
Comprehensive assessment of therapeutic effects:
Unexpected outcomes revealing complexity:
Biomarker identification and validation:
Dose optimization:
Interleukin-6 (IL-6) is a multifunctional cytokine that plays a crucial role in immune response, inflammation, and hematopoiesis. The IL-6 receptor (IL-6R) is composed of two subunits: the IL-6 receptor alpha (IL-6Rα) and the signal-transducing component gp130 (also known as IL-6Rβ or CD130). The IL-6 receptor beta (IL-6Rβ) is essential for IL-6 signal transduction and is involved in various physiological and pathological processes.
The IL-6 receptor beta (mouse recombinant) is a single non-glycosylated polypeptide chain containing 187 amino acids . It signals through a cell-surface type I cytokine receptor complex consisting of the ligand-binding IL-6Rα chain and the signal-transducing component gp130 . The recombinant form of IL-6Rβ is produced using E. coli expression systems and is purified to high levels of purity for research purposes .
IL-6Rβ is a critical component of the IL-6 signaling pathway. Upon binding of IL-6 to IL-6Rα, the complex associates with gp130, leading to the activation of intracellular signaling cascades such as the JAK/STAT, MAPK, and PI3K/Akt pathways . These signaling pathways regulate various cellular processes, including cell proliferation, differentiation, and survival.
Recombinant IL-6Rβ is widely used in immunological research to study the molecular mechanisms of IL-6 signaling and its role in immune modulation . It is also utilized in the development of therapeutic strategies for diseases associated with dysregulated IL-6 signaling, such as autoimmune disorders, chronic inflammatory diseases, and certain cancers .
The recombinant mouse IL-6Rβ protein is typically lyophilized from a 0.2 μm filtered solution in sodium acetate and EDTA with trehalose and BSA as a carrier protein . It is reconstituted at 100 μg/mL in sterile PBS containing at least 0.1% human or bovine serum albumin . The product is shipped at ambient temperature and should be stored at -20 to -70 °C to maintain stability and bioactivity .