Recombinant Mouse IL-7 binds to the IL-7 receptor (IL-7Rα/γc complex), activating downstream pathways:
Key functional outcomes include:
5x increase in IL-17–competent γδ T-cells after in vivo administration .
70% reduction in γδ27+ T-cell numbers within 4 days of exposure .
3–4x expansion of naïve CD8+ T-cells in lymphopenic conditions .
Parameter | Baseline | Post-IL-7 Treatment | Change |
---|---|---|---|
IL-17+ γδ T-cells (LN) | 30% | 70% | +133% |
IFN-γ+ γδ T-cells (LN) | 60% | 65% | +8% |
CD44hi CD69+ γδ27− cells | 30% | 70% | +133% |
Data from PMC and BioLegend |
Administration of 5 μg recombinant IL-7 over 7 days:
B-Cell Development: Required for pro-B to pre-B cell transition via EBF transcription factor activation .
T-Cell Expansion: ED50 = 0.15–0.3 ng/mL in PHA-activated lymphocyte assays .
Immune Reconstitution: Clinical trials show efficacy in reversing chemotherapy-induced lymphopenia .
Recombinant mouse IL-7 is a full-length protein spanning amino acids 26 to 154, with a molecular weight of approximately 17 kDa as determined by SDS-PAGE under reducing conditions . The protein features three disulfide bonds and belongs to the IL-7/IL-9 family . The primary sequence is: ECHIKDKEGKAYESVLMISIDELDKMTGTDSNCPNNEPNFFRKHVCDDTKEAAFLNRAARKLKQFLKMNISEEFNVHLLTVSQGTQTLVNCTSKEEEKNVKEQKKNDACFLKRLLREIKTCWNKILKGSI . When comparing across species, mouse IL-7 shares approximately 88% amino acid sequence identity with rat IL-7 and 58-60% with human, equine, bovine, ovine, porcine, feline, and canine IL-7 .
Mouse IL-7 functions as a hematopoietic cytokine that plays essential roles in the development, expansion, and survival of naive and memory T-cells and B-cells . It regulates the number of mature lymphocytes and maintains lymphoid homeostasis . Mechanistically, IL-7 exerts its biological effects through a receptor composed of the IL7RA subunit and the cytokine receptor common subunit gamma (CSF2RG) . This receptor binding activates various kinases including JAK1 or JAK3, depending on the cell type, subsequently propagating signals through several downstream pathways including the PI3K/Akt/mTOR or the JAK-STAT5 pathways . Additionally, IL-7 selectively promotes IL-17-producing γδ cells in both mice and humans .
Recombinant mouse IL-7 can be produced using different expression systems. Common production methods include expression in HEK 293 cells, which yields protein with >95% purity suitable for various applications including SDS-PAGE, functional studies, mass spectrometry, and HPLC . Alternative production methods include expression in E. coli systems . The choice of expression system may affect post-translational modifications and biological activity, with mammalian expression systems generally providing more physiologically relevant modifications. Upon production, the protein is typically formulated as a lyophilized powder from a 0.2 μm filtered solution in PBS, either with or without bovine serum albumin (BSA) as a carrier protein .
For optimal reconstitution of lyophilized recombinant mouse IL-7, researchers should follow specific protocols depending on the formulation. For preparations containing BSA as a carrier protein, reconstitution at 50 μg/mL in sterile PBS containing at least 0.1% human or bovine serum albumin is recommended . For carrier-free preparations, reconstitution at 50 μg/mL (for 5 μg vials) or 100 μg/mL (for 25 μg or larger vials) in sterile PBS is advised .
For storage, use a manual defrost freezer and avoid repeated freeze-thaw cycles to maintain protein integrity . While short-term storage at 2-8°C is acceptable for reconstituted protein, aliquoting and storing at -20°C to -80°C is recommended for long-term preservation of activity. Working dilutions should be prepared fresh for each experiment to ensure consistent biological activity.
The effective dose of recombinant mouse IL-7 varies by application and cell type. For stimulation of cell proliferation in PHA-activated human peripheral blood lymphocytes, the ED50 (effective dose for 50% maximum response) is 0.15-0.3 ng/mL . In mouse models studying IL-17-producing γδ cells, concentrations around 10 ng/mL have been used for in vitro cultures . For in vivo applications such as those studying glioblastoma models, higher doses (10 mg/kg) of long-acting IL-7 analogs like NT-I7 have been employed . Researchers should establish dose-response relationships specific to their experimental system, as sensitivity to IL-7 varies significantly between different cell populations.
IL-7 primarily affects lymphoid cell populations. The most responsive cells include:
T lymphocytes, particularly:
B lymphocytes in developmental stages
Lymphoid tissues showing significant responses include:
Notably, IL-7 treatment leads to differential expansion of cell subsets, with CD44hi γδ 27− cells showing much greater proliferation (>90% Ki67 positive after 4 days in IL-7) compared to γδ 27+ cells (only ~30% Ki67 positive) . This selective expansion correlates with the capacity for IL-17 production, with expanding cells accounting for almost all IL-17 production upon stimulation .
Recombinant mouse IL-7 has distinct effects on different T-cell subpopulations, demonstrating remarkable specificity in its actions. When applied to lymphocyte cultures, IL-7 selectively promotes the expansion of CD44hi γδ 27− T-cells while CD44lo γδ 27+ T-cells show minimal proliferation . This selective expansion results in a three- to fourfold increase in absolute numbers of γδ 27− cells over 4 days, while γδ 27+ cell numbers decline by approximately 70% .
The expanded γδ 27− population shows enrichment for cells with IL-17–producing capacity, increasing from ~30% to ~70% of the γδ 27− subset . This correlates with increased expression of the transcription factor RORγt, a primary regulator of IL-17 production . In contrast, T-bet expression (associated with IFN-γ production) is not significantly affected by IL-7 treatment .
In vivo administration of recombinant IL-7 similarly increases absolute numbers of IL-17-competent cells in lymph nodes by >fivefold, compared with only two- to threefold increases in IFN-γ–competent cells . These findings demonstrate IL-7's potential for selective modulation of T-cell functional subsets.
Mouse IL-7 activates several key signaling pathways that can be monitored using various experimental approaches:
JAK-STAT Pathway:
PI3K/Akt/mTOR Pathway:
Cell Cycle Progression:
The differential activation of these pathways explains the selective effects of IL-7 on different cell populations. For example, γδ 27− cells express very low levels of SOCS3 (a STAT3 suppressor) compared to CD44lo γδ 27+ cells, which correlates with their selective response to IL-7 .
Research with long-acting IL-7 (NT-I7) in glioblastoma (GBM) mouse models demonstrates significant impacts on the tumor microenvironment. Treatment with NT-I7 (10 mg/kg) in combination with radiotherapy (RT) leads to:
Enhanced anti-tumor immune responses:
Systemic immune modulation:
Improved survival outcomes:
These findings suggest that IL-7-based therapies may enhance anti-tumor immunity through multiple mechanisms, potentially overcoming immunosuppression in the tumor microenvironment.
Successful experiments with recombinant mouse IL-7 require careful consideration of several critical factors:
Protein formulation selection:
Cell responsiveness assessment:
Dose optimization:
Establish dose-response curves for each experimental system
Standard range for in vitro applications: 0.1-10 ng/mL
Higher doses may be required for in vivo applications
Timing considerations:
Combinatorial approaches:
Distinguishing direct from indirect effects of IL-7 requires methodical experimental approaches:
Cell isolation and purification:
Receptor expression analysis:
Quantify IL-7R expression on different cell populations
Cells lacking IL-7R are unlikely to respond directly to IL-7
Signaling pathway inhibition:
Genetic approaches:
Timing analysis:
Direct effects typically occur rapidly (minutes to hours for signaling, 1-4 days for proliferation)
Indirect effects may require longer time periods
These approaches help researchers delineate the precise mechanisms through which IL-7 exerts its biological effects in complex systems.
While mouse and human IL-7 share only 58-60% amino acid sequence identity, they demonstrate significant cross-species activity , making mouse models valuable for translational research. Key translational aspects include:
Functional conservation:
Clinical development:
Differences requiring consideration:
Dose adjustments may be necessary between species
Human lymphocyte subsets may display different sensitivity thresholds
Mouse studies should be validated in human cell systems before clinical translation
A thorough understanding of both the similarities and differences between mouse and human IL-7 biology is essential for effective translational research.
Research with mouse models has identified several promising therapeutic applications for recombinant IL-7:
Cancer immunotherapy:
Infectious disease:
Restoration of T-cell numbers and function during chronic viral infections
Enhancement of vaccine efficacy through improved T-cell responses
Autoimmune regulation:
Selective modulation of specific T-cell subsets (e.g., promoting IL-17-producing cells)
Potential applications in autoimmune conditions requiring immune reconstitution
Hematopoietic stem cell transplantation:
Acceleration of immune reconstitution post-transplantation
Reduction of opportunistic infection risk during recovery
These applications are supported by findings from mouse models, such as the significant survival improvement observed in glioblastoma models treated with NT-I7 in combination with radiotherapy .
These controls ensure that observed effects are specific to IL-7 activity and help distinguish direct from indirect mechanisms of action.
Quantitative assessment of recombinant mouse IL-7 biological activity can be performed using several complementary approaches:
Proliferation assays:
STAT phosphorylation:
Flow cytometry-based detection of p-STAT5 or p-STAT3 in responsive cells
Western blot analysis of phosphorylated signaling proteins
Expected timing: 15-30 minutes post-stimulation
Survival assessment:
Measurement of anti-apoptotic protein induction (Bcl-2)
Annexin V/PI staining to quantify viable cells
Cell counting over time course (typically 4-7 days)
Functional readouts:
In vivo activity:
Lymphocyte counts in blood, lymph nodes, spleen after administration
Phenotypic changes in T-cell subsets (CD44/CD69 upregulation)
Functional capacity (cytokine production upon ex vivo stimulation)
These assays provide comprehensive assessment of biological activity across multiple parameters, ensuring the quality and consistency of recombinant IL-7 preparations.
Despite extensive research, several important questions about mouse IL-7 remain unanswered:
Molecular mechanism of selectivity:
How does IL-7 selectively expand certain lymphocyte subsets (e.g., γδ 27− cells) while having minimal effects on others?
What are the transcriptional networks that determine IL-7 responsiveness beyond SOCS3 expression?
Tissue microenvironment effects:
How does local production of IL-7 in specific tissue microenvironments influence resident and infiltrating immune cells?
What is the interplay between IL-7 and other tissue-derived factors?
Memory formation:
What is the precise role of IL-7 in generating and maintaining memory T cells of different subsets?
How does IL-7 interact with antigen persistence in shaping memory responses?
Developmental programming:
What epigenetic changes are induced by IL-7 signaling during lymphocyte development?
How do these changes influence long-term cell fate decisions?
Non-lymphoid effects:
Does IL-7 have significant direct effects on non-lymphoid cells that express IL-7R?
What are the consequences of IL-7 therapy on non-immune tissues?
Addressing these questions will require innovative experimental approaches and may reveal new applications for IL-7-based therapeutics.
Emerging technologies offer exciting possibilities for advancing IL-7 research:
Single-cell analysis:
Single-cell RNA sequencing to identify heterogeneity in IL-7 responsive populations
Single-cell proteomics to map signaling network activation at individual cell level
Spatial transcriptomics to understand IL-7 effects in tissue context
Advanced protein engineering:
Structure-guided modification of IL-7 to enhance stability or specificity
Development of cell subset-specific IL-7 variants through directed evolution
Creating bifunctional molecules combining IL-7 with other cytokines or targeting moieties
In vivo imaging:
Real-time tracking of IL-7-responsive cells using reporter mice
Intravital microscopy to visualize cellular interactions following IL-7 treatment
PET imaging with labeled IL-7 to map tissue distribution and receptor occupancy
Systems biology approaches:
Computational modeling of IL-7 signaling networks
Integration of multi-omics data to understand global effects of IL-7
Prediction of optimal combination therapies based on network analysis
CRISPR-based screening:
Genome-wide screens to identify novel regulators of IL-7 responsiveness
Precise genetic manipulation to test mechanistic hypotheses
In vivo CRISPR screens to identify resistance mechanisms
These technological advances promise to deepen our understanding of IL-7 biology and accelerate the development of IL-7-based therapeutic strategies.
Based on the available evidence, several experimental systems have demonstrated particular robustness for investigating IL-7 biology:
For in vitro studies:
For in vivo applications:
For mechanistic investigations:
These systems provide reliable platforms for studying the diverse biological activities of IL-7 and allow for rigorous testing of hypotheses regarding its mechanisms of action.
To further explore and develop the therapeutic potential of recombinant mouse IL-7, researchers should consider the following methodological approaches:
Combination therapy optimization:
Systematic testing of IL-7 with immune checkpoint inhibitors
Evaluation of sequential vs. concurrent administration with conventional therapies
Identification of synergistic drug combinations through high-throughput screening
Delivery system development:
Predictive biomarker identification:
Correlation of baseline immune parameters with IL-7 responsiveness
Development of ex vivo assays to predict in vivo efficacy
Identification of genetic markers associated with optimal response
Mechanism-based combination strategies:
Combining IL-7 with agents targeting complementary pathways
Sequential modulation of immune microenvironment before IL-7 treatment
Rational combinations based on systems biology approaches
Translational model development:
Humanized mouse models for more accurate prediction of human responses
Patient-derived xenograft models for personalized therapy assessment
Ex vivo human tissue systems for rapid screening