IL 7 Mouse

Interleukin-7 Mouse Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure and Production

IL-7 Mouse is a 15–17.4 kDa non-glycosylated protein encoded by a 154-amino-acid precursor containing a 25-residue signal peptide . Recombinant forms are produced in E. coli or mammalian systems, with cross-species activity between mouse and human IL-7 (65% sequence homology) .

B Cell Development

  • IL-7 is indispensable for murine B cell maturation:

    • IL-7-deficient mice lack pre-B and mature B cells .

    • Upregulates early B cell factor (EBF) to drive pro-B cell transitions .

T Cell Homeostasis

  • Sustains survival of naïve/memory CD4+/CD8+ T cells via anti-apoptotic proteins (Bcl-2, Bcl-xL) .

  • Regulates CD8+ T cell lineage specification through Runx3 induction .

γδ T Cells and IL-17 Production

  • IL-7 selectively expands IL-17–producing γδ T cells in vivo:

    • In mice, IL-7 administration increases IL-17–competent γδ T cells 5-fold in lymph nodes .

    • Human neonatal γδ T cells similarly exhibit IL-7–dependent IL-17 skewing .

Signaling Mechanisms

IL-7 binds to its receptor (IL-7Rα/γc), activating JAK1/JAK3 and downstream STAT5 phosphorylation . Key pathways include:

  • PI3K-Akt: Regulates T cell size and glucose metabolism .

  • STAT5: Promotes survival and proliferation .

  • STAT3: Required for IL-17 skewing in γδ T cells .

Cross-Species Activity:

Mouse IL-7 stimulates human peripheral blood mononuclear cells (PBMCs) with an ED₅₀ of 1 ng/mL .

Autoimmunity

  • Soluble IL-7Rα (sIL7Rα) potentiates IL-7 bioactivity:

    • In murine models, sIL7Rα + IL-7 exacerbates experimental autoimmune encephalomyelitis (EAE) severity .

    • Enhances IL-7–mediated T cell survival by reducing cytokine consumption .

Lymphopenia and Therapeutics

  • Circulating IL-7 increases during lymphopenia, promoting T cell reconstitution .

  • Clinical trials highlight IL-7’s potential in sepsis and COVID-19 lymphopenia .

Table 1: Recombinant IL-7 Mouse Products

SourceMolecular MassPurityBiological Activity (ED₅₀)
R&D Systems 17.4 kDa≥95%0.2–0.4 µg/mL (neutralization)
Irvine Scientific 15 kDa≥95%≤1 ng/mL (proliferation)
STEMCELL 17.4 kDaNot specifiedSupports T cell survival

Table 2: In Vivo Effects of IL-7 Mouse

Study ModelKey FindingReference
Imiquimod-induced psoriasis (Mouse)Anti–IL-7R antibody blocks IL-17–driven lesions
Neonatal human γδ T cellsIL-7 + TCR agonists expand IL-17 producers 20-fold
EAE (Mouse)IL-7 + sIL7Rα accelerates disease progression

Product Specs

Introduction
Interleukin-7 (IL-7) is a cytokine crucial for the development of lymphoid cells, primarily secreted by stromal cells. It supports the growth and differentiation of precursor B cells and early T cells. Additionally, IL-7 exhibits biological effects on cells of the Natural Killer (NK) and myeloid lineages.
Description
Recombinant Mouse Interleukin-7, produced in E. coli, is a single, non-glycosylated polypeptide chain. It comprises 129 amino acids and has a molecular weight of 14.9 kDa.
Physical Appearance
Sterile white powder obtained by lyophilization (freeze-drying).
Formulation
The product is filtered through a 0.2µm filter and lyophilized. The initial solution for lyophilization is a 1mg/ml solution in phosphate-buffered saline (PBS) at pH 7.4 with 2% trehalose.
Solubility
To reconstitute the lyophilized Interleukin-7, it is recommended to dissolve it in sterile 18 MΩ-cm H2O at a concentration of at least 100 µg/ml. This solution can be further diluted in other aqueous solutions.
Stability
Lyophilized Interleukin-7 remains stable for 3 weeks at room temperature. However, for long-term storage, it is recommended to store it in a dry environment below -18°C. After reconstitution, store IL-7 at 4°C for 2-7 days. For future use, store below -18°C. To ensure stability during long-term storage, adding a carrier protein (0.1% HSA or BSA) is recommended. Avoid repeated freeze-thaw cycles.
Purity
The purity of the protein is greater than 96.0%, as determined by: (a) Reverse-Phase High-Performance Liquid Chromatography (RP-HPLC) analysis and (b) Sodium Dodecyl Sulfate-Polyacrylamide Gel Electrophoresis (SDS-PAGE) analysis.
Biological Activity
The ED50, defined as the concentration that induces 50% of the maximum proliferation response, is determined by the dose-dependent stimulation of murine 2E8 cell proliferation. The ED50 is less than 0.2 ng/ml, corresponding to a specific activity greater than 5.0 x 106 IU/mg.
Synonyms
Lymphopoietin 1 (LP-1), pre-B cell factor, IL-7.
Source
Escherichia Coli.
Amino Acid Sequence
ECHIKDKEGK AYESVLMISI DELDKMTGTD SNCPNNEPNF FRKHVCDDTK EAAFLNRAAR KLKQFLKMNI SEEFNVHLLT VSQGTQTLVN CTSKEEKNVK EQKKNDACFL KRLLREIKTC WNKILKGSI.

Q&A

What is IL-7 and what is its function in the mouse immune system?

IL-7 is a critical cytokine in the mouse immune system that plays essential roles in lymphocyte development, survival, and homeostasis. It signals through the IL-7 receptor (IL-7R), which consists of the IL-7Rα chain (CD127) and the common gamma chain (γc). In mice, IL-7 is particularly important for the development of B and T lymphocytes, maintaining naive T cell survival, and promoting T cell expansion during lymphopenia. IL-7 signaling activates multiple pathways, notably the JAK-STAT pathway, particularly STAT5 phosphorylation, which induces proliferation of responsive cells . IL-7 also promotes TH1 cell responses and can prime and activate autoreactive T cells in various mouse models of autoimmune diseases .

How does murine IL-7 differ from human IL-7 in experimental settings?

Murine and human IL-7 share significant functional similarities but have distinct species-specific properties. Both can activate STAT5 signaling and induce cell proliferation, though with different potencies across species. Interestingly, murine IL-7 can signal through the human IL-7 receptor, making it relevant in xenogeneic experimental systems. As demonstrated in HSC/MS-5 xenogeneic cultures, murine IL-7 produced by mouse stromal cells can effectively stimulate human B-lineage cells expressing IL-7R . This cross-species reactivity is important when designing experiments using human cells in mouse environments. The sensitivity of detection also differs, with human IL-7 ELISA having a sensitivity of approximately 0.1 pg/ml, while murine IL-7 ELISA typically has a sensitivity threshold of around 10 pg/ml .

What mouse strains are commonly used for IL-7 research?

Several mouse strains are utilized in IL-7 research, each offering specific advantages for different experimental questions. C57BL/6 (B6) mice are commonly used as wild-type controls. The Jackson Laboratory offers IL-7 transgenic mice (IL-7tg+) maintained on a B6.CD90.1+ background . For studying IL-7 receptor signaling mechanisms, FcRn-/- mice and various Fcγ receptor knockout strains (Fcγ-/-, FcγRIIb-/-, FcγRI-/-) are employed . C3H/HeJ mice are particularly useful for studying IL-7's role in autoimmune conditions like alopecia areata . When investigating IL-7's effects on specific cell populations, bone marrow chimeras can be generated to restrict IL-7 receptor expression to either hematopoietic or non-hematopoietic compartments .

How should IL-7 activity be measured in mouse models?

Measuring IL-7 activity in mouse models requires multiple complementary approaches. The most direct method involves quantifying IL-7 concentration using murine-specific ELISA, with typical sensitivity around 10 pg/ml. A robust detection system employs a sandwich ELISA using polyclonal goat anti-mouse IL-7 capture antibody (5 μg/ml in PBS), followed by biotinylated polyclonal goat anti-mouse IL-7 sandwich antibody (300 ng/ml) and HRP-labeled streptavidin for color development .

Beyond concentration measurement, IL-7 activity should be assessed functionally through:

  • Measurement of STAT5 phosphorylation in target cells (typically CD8+ or CD4+ T cells) following IL-7 exposure

  • Proliferation assays using IL-7R+ target cells cultured with recombinant IL-7 or serum from test mice

  • In vivo expansion of adoptively transferred CFSE-labeled T cells in response to endogenous or exogenous IL-7

  • Flow cytometric analysis of IL-7-responsive cell populations (e.g., CD19+/IL-7R+ cells) for changes in number, activation markers, and cytokine production

What is the recommended protocol for administering IL-7 in mouse models?

When administering IL-7 in mouse models, the preparation, dosage, and delivery route must be carefully optimized for experimental objectives. For enhanced potency, IL-7 should be administered as a complex with anti-IL-7 neutralizing monoclonal antibody (clone M25), which dramatically improves its in vivo potency compared to IL-7 alone . These IL-7/anti-IL-7 mAb complexes (IL-7c) can be prepared by combining recombinant murine IL-7 with the M25 antibody.

A typical administration protocol involves:

  • Preparation of IL-7 complexes by mixing recombinant murine IL-7 (typically 10 ng/ml concentration) with M25 antibody at an appropriate molar ratio

  • Intraperitoneal injection of the complex, with dosing intervals of approximately 7 days (total of 2-3 injections depending on the experimental timeline)

  • Control groups should receive PBS injections following the same schedule

  • For studies of autoimmune conditions, such as alopecia areata in C3H/HeJ mice, two injections at 7-day intervals have been shown to accelerate disease development

When studying specific populations, consider that IL-7c substantially increases total lymphocyte numbers in lymphoid organs and particularly enhances CXCR3+CD8+ T cells, T-bet+CD8+ T cells, and IFN-γ-producing CD8+ T cells in skin-draining lymph nodes .

How should researchers isolate and culture IL-7-responsive cells from mice?

Isolation and culture of IL-7-responsive cells from mice requires precise techniques to maintain cell viability and responsiveness. For B-lineage cells, perform positive or negative selection of CD19+ cells and plate them on confluent monolayers of MS-5 stromal cells in 96-well flat-bottom plates using MEM supplemented with 10% FBS . For T cells, isolation can be performed using magnetic bead purification followed by stimulation with anti-CD3/CD28 Dynabeads at a 2:1 bead-to-cell ratio .

The culture protocol should include:

  • Addition of recombinant murine IL-7 at concentrations ranging from 0.01-100 ng/ml (dose-response studies) or at a fixed concentration of 10 ng/ml for standard applications

  • For T cell cultures focused on TH1 or Tc1 induction, stimulate with anti-CD3/CD28 beads in the presence of IL-7 (10 ng/ml) for 4 days

  • For detecting intracellular cytokines, restimulate cells with Cell Stimulation Cocktail for 5 hours plus Protein Transport Inhibitor Cocktail

  • For accurate quantification of cell numbers, add a known quantity of polystyrene microspheres (e.g., 6-μm Polybeads) during flow cytometric analysis

It's critical to note that IL-7R expression creates functionally distinct populations, with CD19+/IL-7R+ cells showing different light scatter profiles and enhanced survival compared to CD19+/IL-7R- cells. The former are larger with greater heterogeneity in forward scatter and respond to picogram concentrations of IL-7 .

How does the IL-7/anti-IL-7 mAb complex mechanism enhance cytokine potency?

The dramatic enhancement of IL-7 potency when administered as a complex with anti-IL-7 neutralizing monoclonal antibody (clone M25) represents a sophisticated approach to cytokine delivery. This mechanism involves several coordinated processes:

  • The neonatal Fc receptor (FcRn) plays a crucial role in the enhanced potency of IL-7/M25 complexes. In FcRn-/- mice, the potency of IL-7/M25 is significantly reduced compared to wild-type mice, while responses to other forms of IL-7 stimuli remain normal .

  • FcRn extends the serum half-life of antibodies, including M25. In FcRn-/- mice, M25 shows greatly abbreviated serum persistence compared to wild-type mice, suggesting that prolonged cytokine presence is a key factor in the enhanced activity .

  • The antibody portion of the complex appears critical, as IL-7 complexes generated with M25 Fab fragments drive significantly reduced CD8+ T-cell proliferation compared to those with complete antibody molecules .

  • The enhancement is specific to IL-7/M25 complexes and not due to generalized effects on IL-7 responsiveness, as confirmed by comparing the response to endogenous IL-7 in irradiated hosts or high-dose exogenous IL-7 treatment between FcRn-/- and wild-type mice .

This mechanism represents an important paradigm for enhancing cytokine function in research and potentially therapeutic applications, providing a model for studying controlled immune activation.

What role does IL-7 play in mouse models of autoimmune diseases?

IL-7 serves as a critical mediator in mouse models of autoimmune diseases, functioning at multiple levels of immune dysregulation. In models such as experimental autoimmune encephalomyelitis (EAE), nonobese diabetic (NOD), and alopecia areata (AA), IL-7 promotes pathogenic immune responses through several mechanisms:

  • Enhancement of TH1 and Tc1 development: IL-7 significantly increases the percentages of IFN-γ+CD4+ T cells and IFN-γ+CD8+ T cells upon stimulation, driving the pro-inflammatory response characteristic of many autoimmune conditions .

  • Acceleration of disease development: In C3H/HeJ mice susceptible to AA, IL-7 complex (IL-7c) treatment leads to faster and more severe hair loss compared to PBS-treated controls, with increased frequencies of skin-infiltrating CD8+ T cells and IFN-γ-producing CD8+ T cells .

  • Promotion of cytotoxic T cell function: IL-7 enhances the release of cytotoxic mediators, including granzymes and perforin-1, which constitute the cytotoxic T lymphocyte (CTL) gene signature defined in AA lesional skin from both human AA and C3H/HeJ AA mice .

  • Expansion of pathogenic T cell subsets: IL-7c robustly increases the frequency of CXCR3+CD8+ T cells, T-bet+CD8+ T cells, and IFN-γ-producing CD8+ T cells in skin-draining lymph nodes, all of which contribute to tissue destruction in autoimmune conditions .

  • Potential reduction in regulatory T cells: IL-7c treatment may decrease the frequency of CD4+ regulatory T cells due to their low response to IL-7 stimulation, further skewing the immune balance toward inflammation .

These findings suggest that IL-7 blockade could be a therapeutic strategy for autoimmune diseases. Indeed, anti-IL-7Rα treatment suppresses effector T cell function, reducing IFN-γ and IL-2 production by CD8+ T cells and preventing the transfer of autoimmunity in adoptive transfer models .

How do researchers distinguish between effects of endogenous and exogenous IL-7 in mouse studies?

Distinguishing between the effects of endogenous and exogenous IL-7 in mouse studies requires sophisticated experimental designs and controls. Researchers employ several methodological approaches to differentiate these sources:

  • Species-specific neutralization: Using species-specific neutralizing antibodies allows selective blockade of either endogenous (murine) or exogenous (e.g., human) IL-7. For example, goat anti-mouse IL-7 can neutralize endogenous murine IL-7 while leaving exogenously administered human IL-7 unaffected .

  • Species-specific detection assays: Employing highly specific ELISA systems with minimal cross-reactivity enables researchers to quantify murine and human IL-7 separately, even in the same experimental system. Human-specific IL-7 ELISA kits with sensitivity of 0.1 pg/ml and murine-specific sandwich ELISAs with 10 pg/ml sensitivity allow precise source determination .

  • CFSE-labeled T cell transfer: Adoptively transferring CFSE-labeled T cells into hosts allows tracking of proliferative responses to endogenous IL-7 (e.g., in irradiated hosts where IL-7 levels naturally increase) versus responses to exogenously administered IL-7 or IL-7 complexes .

  • Genetic models: Using IL-7 transgenic mice or conditional knockout models permits controlled alteration of endogenous IL-7 levels, providing a baseline against which exogenous IL-7 effects can be compared .

  • Bone marrow chimeras: Generating chimeric mice with FcRn expression restricted to either bone marrow-derived or non-hematopoietic cells helps distinguish between cellular sources of IL-7 and cellular targets of IL-7 complexes .

In xenogeneic models, such as human HSC in mouse stromal (MS-5) cultures, researchers have demonstrated that endogenous murine IL-7 (14-38 pg/ml) can significantly influence human cell development, with goat anti-murine IL-7 antibody treatment reducing CD19+ cell emergence by 67±15% after 4 weeks .

What are the key considerations for measuring IL-7 levels in mouse serum and tissues?

Accurate measurement of IL-7 in mouse serum and tissues presents several technical challenges requiring careful methodological consideration:

  • Selection of appropriate detection method: For murine IL-7, a sandwich ELISA using specific reagents is recommended. The protocol should include coating plates with polyclonal goat anti-mouse IL-7 capture antibody (5 μg/ml in PBS), followed by biotinylated polyclonal goat anti-mouse IL-7 sandwich antibody (300 ng/ml) and HRP-labeled streptavidin for detection . Commercial high-sensitivity kits are available for human IL-7 with detection limits as low as 0.1 pg/ml.

  • Sensitivity threshold considerations: Standard murine IL-7 ELISA has a sensitivity threshold of approximately 10 pg/ml, which may be insufficient for some physiological conditions. Since IL-7 can be biologically active at picogram concentrations, bioassays measuring STAT5 phosphorylation or proliferation of IL-7-dependent cells may provide functional evidence of IL-7 presence below ELISA detection limits .

  • Compartment-specific sampling: IL-7 concentrations vary significantly between compartments. In HSC/MS-5 culture systems, murine IL-7 concentrations range from 14-38 pg/ml in supernatants, while serum levels may differ. Tissue-specific extraction protocols should be optimized to preserve cytokine integrity, with immediate processing or storage at -80°C .

  • Accounting for IL-7 binding to extracellular matrix: Optimal IL-7 signaling depends on IL-7 binding to heparan sulfate proteoglycans. IL-7 synthesized by stromal cells may be presented to responsive cells by these matrix components, potentially making soluble IL-7 measurements underestimate the total bioactive IL-7 in a tissue .

  • Control for cross-reactivity: When performing experiments with multiple species (e.g., human cells in mouse environments), it's essential to verify the species specificity of the detection method. Include controls by testing the detection system against known quantities of both murine and human IL-7 .

How should researchers design IL-7 receptor blocking experiments in mice?

Designing effective IL-7 receptor blocking experiments requires careful consideration of antibody selection, dosing regimen, and appropriate controls:

  • Antibody selection: Anti-IL-7Rα antibodies are the primary tool for blocking IL-7 signaling. The specific clone and isotype should be selected based on the research question, with consideration for factors such as half-life, Fc receptor binding capabilities, and potential depletion of IL-7Rα-expressing cells versus pure signaling blockade .

  • Control groups: Essential controls include:

    • Isotype-matched control antibody at equivalent concentration

    • PBS-treated control group

    • If studying autoimmune models, both disease-induction and disease-treatment paradigms should be explored

  • Dosing regimen: Anti-IL-7Rα treatment typically follows a specific schedule, with intraperitoneal injections often administered at key timepoints relative to disease induction or experimental intervention. The dosage and frequency must be optimized for the specific antibody and experimental system .

  • Validation of receptor blockade: Experimental verification of IL-7Rα blockade should include:

    • Flow cytometric assessment of IL-7Rα expression (noting that some anti-IL-7Rα antibodies may interfere with detection by other anti-IL-7Rα antibodies)

    • Functional validation by measuring STAT5 phosphorylation following ex vivo IL-7 stimulation

    • Assessment of biological effects, such as changes in lymphocyte counts and subset distribution

  • Mechanistic analysis: To understand the mechanism of IL-7R blockade effects, researchers should examine:

    • Changes in effector cell functions (cytokine production, proliferation)

    • Alterations in regulatory cell populations (proportions, suppressive capacity)

    • Effects on specific disease-relevant cell types (e.g., CD8+ T cells in alopecia areata models)

Properly designed IL-7R blocking studies have revealed that anti-IL-7Rα treatment can significantly reduce IFN-γ and IL-2 production by CD8+ T cells, prevent disease induction in adoptive transfer models, and reduce total numbers of lymphocytes across multiple compartments .

What are common pitfalls in IL-7 mouse experiments and how can they be avoided?

Several critical pitfalls can compromise the validity and reproducibility of IL-7 mouse experiments. Being aware of these challenges and implementing appropriate countermeasures is essential:

By anticipating these common pitfalls and implementing appropriate experimental designs and controls, researchers can ensure more robust and reproducible results in IL-7 mouse experiments.

How might single-cell analysis techniques advance our understanding of IL-7 signaling in mouse models?

Single-cell analysis techniques offer unprecedented opportunities to dissect the heterogeneity in IL-7 signaling across immune cell populations in mouse models. These approaches can reveal nuanced aspects of IL-7 biology that bulk population studies may obscure:

  • Identifying rare IL-7-responsive populations: Single-cell RNA sequencing (scRNA-seq) can identify previously uncharacterized IL-7-responsive cell populations that may be numerically minor but functionally significant in immune regulation or pathology. This is particularly relevant given the observed differences between IL-7R+ and IL-7R- subpopulations within the same lineage .

  • Mapping signaling pathway heterogeneity: Single-cell phospho-flow cytometry enables quantification of STAT5 phosphorylation and other signaling intermediates at the individual cell level, revealing the spectrum of signaling intensities within nominally homogeneous populations following IL-7 stimulation. This approach can identify threshold effects and binary versus graded responses to IL-7 .

  • Correlating IL-7R expression with functional outcomes: Combinatorial indexing-based methods allow simultaneous assessment of surface IL-7Rα expression, signaling pathway activation, and functional outputs such as cytokine production or proliferation within individual cells, enabling construction of comprehensive response maps.

  • Spatial context of IL-7 signaling: Emerging spatial transcriptomics and imaging mass cytometry techniques can reveal the microanatomical context of IL-7 signaling, potentially identifying niches where IL-7 availability and receptor expression create functionally distinct microenvironments within lymphoid tissues.

  • Temporal dynamics of IL-7 responses: Single-cell trajectory analysis can reconstruct the developmental and activation pathways influenced by IL-7, revealing how IL-7 signaling shapes cell fate decisions and immune cell differentiation over time.

These single-cell approaches would particularly benefit studies of autoimmune disease models where IL-7 influences pathogenesis, potentially revealing specific cellular subsets that drive pathology and might represent targets for selective intervention without compromising beneficial IL-7 functions .

What emerging genetic tools are being developed for IL-7 research in mouse models?

Emerging genetic tools are dramatically expanding the capabilities for IL-7 research in mouse models, enabling more precise manipulation and monitoring of IL-7 biology:

  • Conditional and inducible IL-7 and IL-7Rα models: Advanced Cre-lox systems with tissue-specific and temporally controlled expression allow researchers to manipulate IL-7 or IL-7Rα expression with unprecedented specificity. These systems can restrict IL-7 production or responsiveness to particular cell types or developmental stages, avoiding the confounding effects of germline modifications on immune system development .

  • Reporter mice: Fluorescent reporter constructs knocked into the IL-7 or IL-7Rα locus enable real-time visualization of expression patterns without disrupting function. These tools are particularly valuable for identifying IL-7-producing cells in tissues, which has been technically challenging with traditional methods.

  • CRISPR/Cas9-based approaches: The development of in vivo CRISPR/Cas9 delivery systems permits rapid generation of targeted mutations in IL-7 pathway components without the need for lengthy breeding of germline-modified animals. This approach facilitates efficient screening of potential regulatory elements and interacting partners.

  • Humanized mouse models: Advanced humanized mouse models with reconstituted human immune systems provide platforms to study human-specific aspects of IL-7 biology in vivo, building on observations from xenogeneic culture systems where murine IL-7 influences human cell development .

  • IL-7 signaling biosensors: Genetically encoded biosensors that report on IL-7R engagement and downstream signaling events can be integrated into mouse models, allowing real-time monitoring of IL-7 activity in living animals through intravital imaging techniques.

These emerging genetic tools will enable researchers to address sophisticated questions about IL-7 biology, such as the contribution of different cellular sources of IL-7 to immune homeostasis and the cell type-specific requirements for IL-7 signaling in various physiological and pathological contexts. The capacity to manipulate IL-7 pathway components with greater precision will also accelerate the development of targeted therapeutic approaches for conditions where IL-7 signaling plays a pathogenic role .

How might findings from IL-7 mouse research translate to human therapeutic applications?

Findings from IL-7 mouse research offer several promising translational pathways to human therapeutic applications, though with important considerations for species differences and clinical implementation:

  • IL-7/anti-IL-7 complex therapy: The dramatic enhancement of IL-7 potency when complexed with anti-IL-7 monoclonal antibody M25 in mice suggests a potential approach for enhancing IL-7 efficacy in human applications. This strategy could allow lower doses of IL-7 while achieving greater biological effects, potentially reducing side effects and costs in clinical applications aimed at immune reconstitution or vaccination enhancement .

  • IL-7 receptor blockade for autoimmunity: The effectiveness of anti-IL-7Rα treatment in suppressing effector T cell function and preventing autoimmunity in mouse models indicates a promising therapeutic avenue for human autoimmune diseases. Mouse studies showing reduced IFN-γ and IL-2 production following IL-7Rα blockade provide a mechanistic foundation for human trials in conditions like rheumatoid arthritis, multiple sclerosis, and alopecia areata .

  • Species-bridging experimental systems: The observation that murine IL-7 can signal through human IL-7 receptors provides a valuable platform for testing IL-7-targeted approaches in xenogeneic systems before moving to clinical trials. HSC/MS-5 cultures where human B cell development depends on murine IL-7 illustrate the preservation of core signaling mechanisms across species .

  • Targeted delivery approaches: Insights from mouse studies regarding IL-7 presentation by heparan sulfate proteoglycans and the microanatomical distribution of IL-7 responsiveness could inform the development of targeted delivery systems that concentrate IL-7 activity in desired tissues or cell populations .

  • Biomarkers of IL-7 response: Mouse research identifying distinct patterns of IL-7 responsiveness among seemingly homogeneous populations (e.g., CD19+/IL-7R+ versus CD19+/IL-7R- cells) suggests the importance of developing biomarkers to identify likely responders to IL-7-targeted therapies in human patients .

Translational efforts must account for important species differences, including subtle variations in signaling pathway components and tissue distribution of IL-7 and IL-7R. Additionally, the complex interplay between IL-7 and other cytokines in human immune regulation may differ from murine patterns. Nevertheless, the fundamental conservation of IL-7 biology across mammals supports the translational value of findings from sophisticated mouse models to human therapeutic development.

Product Science Overview

Structure and Production

Mouse IL-7 is a recombinant protein produced in Escherichia coli (E. coli) and optimized for use in cell culture, differentiation studies, and functional assays . The recombinant form of mouse IL-7 typically includes a Met on the amino terminus and has a calculated molecular weight of approximately 15 kDa . The protein is purified to a high degree, with purities exceeding 95% and endotoxin levels below 0.01 ng/μg .

Biological Activity

IL-7 is primarily produced by non-hematopoietic stromal cells in the bone marrow, thymus, and lymphoid organs and tissues . It acts as both a T cell growth factor and a critical anti-apoptotic survival factor . The primary targets of IL-7 are T cells, B cells, and dendritic cells . It signals through a heterodimeric receptor composed of the common cytokine signaling γ-chain and IL-7 receptor α-chain .

Applications

Mouse IL-7 is widely used in various research applications, including:

  • In vitro T cell expansion: It is used to expand naive T cells in culture .
  • In vitro differentiation of T cells: It aids in the differentiation of invariant natural killer T (iNKT) cells .
  • Investigation of IL-7 mediated signaling pathways: Researchers study the signaling pathways activated by IL-7 to understand its role in immune regulation .
Storage and Stability

Recombinant mouse IL-7 is stable in its lyophilized state at -20°C for up to one year . Once reconstituted with a carrier protein, it remains stable at 4°C for two months and at -20°C for six months . It is essential to avoid repeated freeze-thaw cycles to maintain its biological activity .

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.