Structure
Recombinant human IL-7 is a disulfide-linked monomeric protein comprising 153 amino acids (26–177 aa) with a molecular weight of 17–17.5 kDa . It is structurally homologous to murine IL-7 (60% sequence identity) and belongs to the IL-7/IL-9 cytokine family .
Supplier | Expression System | Purity | Biological Activity | Endotoxin Level |
---|---|---|---|---|
BioLegend | HEK 293 | ≥95% | N/A | ≤0.05 EU/µg |
BPS Bioscience | E. coli | ≥97% | ≤0.5 ng/mL (ED₅₀) | <0.1 EU/µg |
BD Biosciences | E. coli | >95% | >2 × 10⁶ units/mg | ≤0.1 ng/µg |
Abcam | HEK 293 | ≥95% | Suitable for cell culture | ≤0.05 EU/µg |
Key notes: HEK 293-expressed IL-7 retains glycosylation, while E. coli-derived versions are non-glycosylated but retain functional activity .
IL-7 exerts its effects via the IL-7 receptor (IL-7R), a heterodimer of IL-7Rα (CD127) and the common γ-chain (CD132) . Its primary roles include:
Thymic Development: Critical for T-cell progenitor survival and differentiation into mature T cells .
Peripheral Homeostasis: Maintains naive and memory T-cell populations by upregulating anti-apoptotic proteins (e.g., Bcl-2, Mcl-1) .
Immune Reconstitution: Enhances recovery of CD4⁺ and CD8⁺ T cells post-chemotherapy or hematopoietic stem cell transplantation (HSCT) .
Early Development: Required for pro-B and pre-B cell proliferation in mice, but not in humans .
Survival Signals: Modulates B-cell survival via Bcl-2 family proteins, though mature B cells lack IL-7Rα .
Dendritic Cells: Supports thymic dendritic cell accumulation and T-cell priming .
Neutrophils/Eosinophils: Indirectly influences granulopoiesis via T-cell-derived cytokines (e.g., IL-17) .
Critical Observations:
T-Cell Rejuvenation: RhIL-7 restores a "youthful" T-cell profile in elderly patients .
B-Cell Effects: Polyclonal pre-B cell proliferation observed in some patients, though transient .
Signaling Pathways: Activates JAK/STAT5 and PI3K/Akt/mTOR pathways, promoting cell survival and proliferation .
Immune Reconstitution: Accelerates recovery of CMV-specific T cells post-allo-HSCT, reducing infection risk .
Combination Therapies: Synergistic use with checkpoint inhibitors (e.g., PD-1/PD-L1 blockers) to enhance anti-tumor immunity.
Biomarker-Driven Trials: Identifying IL-7Rα⁺ patient subsets for personalized dosing.
Infectious Disease Focus: Evaluating efficacy in sepsis and viral immunosuppression (e.g., COVID-19, PML) .
Interleukin-7 (IL-7) is a cytokine originally identified for its growth-promoting effects on progenitors of B cells. It plays vital roles in health maintenance and disease prevention, with congenital deficiency of IL-7 signaling leading to profound immunodeficiency. IL-7 primarily contributes to host defense by regulating the development and homeostasis of immune cells, including T lymphocytes, B lymphocytes, and natural killer (NK) cells . IL-7 signaling is essential for normal thymopoiesis and has been shown to maintain and restore T-cell numbers, increase T-cell diversity, boost T-cell function, and inhibit T-cell apoptosis .
Cells | Effects of IL-7 Deficiency | Effects of IL-7 Treatment |
---|---|---|
Thymus | Decreased thymic cell count; Thymic involution | Increased thymic cell count; Recovery of thymic function |
T cells | Inhibition of glucose metabolism; Cell atrophy; Impaired T-cell functions; Severe impairment of T lymphopoiesis; T-cell apoptosis | Restored T-cell numbers; Increased T-cell diversity; Enhanced T-cell function; Inhibited T-cell apoptosis; Promoted glucose metabolism; Prevention of T-cell atrophy |
B cells | Blocked transition to pro-B cells; Impaired B differentiation potential; Impaired early B lymphopoiesis; B-cell apoptosis | Increased B-cell numbers; Facilitated transition of pro-B cells; Promoted B-cell survival; Increased antibody production |
IL-7 has distinct effects on different immune cell populations, regulating their development, survival, and function. In T cells, IL-7 promotes survival of both naïve and memory populations, while in B cells, it influences multiple developmental stages including commitment, survival, differentiation, and proliferation . IL-7 also regulates the recruitment of other leukocytes such as neutrophils and monocytes .
Recombinant human IL-7 (rhIL-7) can be effectively produced using bacterial expression systems, with Escherichia coli HMS174 (DE3) pLysS under the control of a T7 promoter being well-documented . The expression typically results in insoluble inclusion bodies that require several purification steps. The production process involves separating inclusion bodies from cellular debris by cross-flow filtration, followed by solubilization using 6 M guanidine HCl . This approach yields substantial quantities of protein that can be subsequently refolded and purified.
Purification of biologically active rhIL-7 requires multiple chromatographic steps. First, denatured, monomeric rhIL-7 should be purified by size-exclusion chromatography using Prep-Grade Pharmacia Superdex 200 prior to refolding . Attempting to refold rhIL-7 from solubilized inclusion bodies without this prior purification step has proven unsuccessful . Correctly folded rhIL-7 monomer is then generated by statically refolding the denatured protein at a final concentration of 80-100 μg/ml in 100 mM Tris, 2 mM EDTA, 500 mM L-arginine, pH 9.0 buffer with 0.55 g/l oxidized glutathione at 2-8°C for at least 48 hours . The refolded protein is subsequently purified through a combination of hydrophobic interaction, cation-exchange, and size-exclusion chromatography, resulting in a final product with >95% purity .
The biological activity of purified rhIL-7 should be assessed using validated IL-7-dependent bioassays. A well-established method involves the pre-B-cell bioassay, where IL-7-dependent pre-B cells are used to measure the proliferative response to the purified cytokine . For rigorous quality control, multiple analytical methods should be employed, including SDS-PAGE with Coomassie brilliant blue staining, high-pressure size-exclusion chromatography (SEC-HPLC), and reverse-phase HPLC . Additionally, endotoxin levels should be measured to ensure they remain below 0.05 EU/mg for research involving primary cells or in vivo applications .
While specific storage conditions aren't detailed in the provided search results, based on standard practices for recombinant proteins, purified IL-7 should typically be stored at -80°C for long-term stability, with aliquoting recommended to avoid freeze-thaw cycles. Working solutions can be maintained at 2-8°C for limited periods, similar to the temperature range used during the refolding process . Stability studies should be conducted to determine the specific shelf-life under various storage conditions.
IL-7 exhibits potent pro-immune functions during viral infections, particularly in overcoming inhibitory immune pathways that characterize chronic viral infections. In experimental models using lymphocytic choriomeningitis virus (LCMV) clone 13 infection, therapeutic administration of IL-7 enabled viral clearance from spleen, liver, and other viral reservoirs after 3 weeks of treatment, while control animals remained chronically infected . IL-7 promotes antiviral immunity through multiple mechanisms, including increasing the numbers of virus-specific CD8+ T cells and enhancing their effector functions . While IL-7 treatment expands multiple immune cell populations, including B cells and non-virus-specific T cells, depletion experiments demonstrated that CD4+ and CD8+ T cells are specifically essential for IL-7-mediated viral clearance .
When designing experiments to investigate IL-7's role in viral infections, researchers should consider:
Appropriate viral model selection (e.g., LCMV clone 13 for chronic infection studies)
Timing of IL-7 administration (therapeutic versus prophylactic)
Duration of treatment (3 weeks was effective in LCMV studies)
Inclusion of proper controls (PBS-treated animals)
Comprehensive immune cell analysis, including:
Cell depletion experiments are valuable for determining which immune cell populations are necessary for IL-7-mediated effects, as demonstrated in studies showing the essential role of both CD4+ and CD8+ T cells in viral clearance .
Genetic variations in IL-7 have been associated with the risk of immune-related adverse events (irAEs) in cancer patients receiving immune checkpoint blockade (ICB) therapy. In a study of 214 melanoma patients receiving ICB, researchers observed increased risk of severe irAEs in carriers of the minor allele of rs16906115, which is intronic to the IL-7 gene . This genetic variation was associated with altered B cell subset distributions, particularly an increase in high IL-7-expressing unswitched memory (USM) B cells and a trend toward fewer naive B cells . These findings suggest that IL-7 genetic variants may influence clinical outcomes and toxicity of immunotherapies, potentially through modulation of B cell populations and function.
IL-7 influences B cell development through several mechanisms. Gene expression analysis has shown that IL-7 expression is anti-correlated with the naive immunoglobulin IGHD but is associated with the percentage of immunoglobulin chains carrying secondary divergent mutations, as observed in somatic hypermutation . Single-cell RNA sequencing of B cells from melanoma patients and healthy controls revealed that IL-7 expression varies across B cell subsets, being highest in unswitched memory (USM) and switched memory (SM) cells, intermediate in naive cells, and lowest in antibody-secreting cells (ASCs) . Interestingly, patient B cells showed significantly higher IL-7 expression compared to those from healthy controls across all subsets except ASCs . Carriers of the rs16906115 risk allele demonstrated increased IL-7 expression across all B cells and had significantly increased counts of high IL-7-expressing USM cells , suggesting that IL-7 genetic variations may affect B cell subset distribution and function.
IL-7 can be strategically combined with other immunomodulators to enhance therapeutic efficacy. Research has demonstrated that when IL-7 is administered in the presence of IL-12, the diversity of intratumoral CD8+ T cells increases, and IL-12's function is augmented to promote clonality . This synergistic effect suggests that combination therapies involving IL-7 and other cytokines or immune modulators may provide enhanced therapeutic benefits compared to monotherapies. Researchers investigating such combinations should design experiments that carefully evaluate potential synergistic or antagonistic effects on different immune cell populations and functions.
When studying IL-7's role in immune reconstitution, researchers should consider:
Model selection (aging, lymphopenia, immunodeficiency)
Dosing regimens and duration of treatment
Comprehensive immune phenotyping, including:
T cell subset analysis (naive vs. memory)
T cell receptor (TCR) diversity assessment
B cell developmental stages
Thymic output measurements
Functional immune assays
Research has shown that while IL-7 therapy alone in aged mice could not completely reverse thymic involution, it can accelerate recovery of thymic function . The multiple functions of IL-7 in T-cell lymphopoiesis have encouraged its application as a therapy for recovering T-cell numbers . Methodologically sound research in this area requires careful consideration of baseline immune status, treatment protocols, and comprehensive outcome measures.
One notable contradiction in IL-7 research concerns the species-specific differences in IL-7 dependency. While IL-7 is essential for murine B-cell development, human B cells appear to develop normally in the absence of IL-7 . This is evidenced by the different presentations of genetic immunodeficiencies affecting the IL-7 pathway. Gene mutations at the human γc locus lead to X-linked severe combined immunodeficiency with distinct B cell presentations compared to murine models . Researchers must carefully consider these species-specific differences when translating findings from mouse models to human applications, as the developmental requirements and responses to IL-7 may vary significantly.
Critical methodological challenges in IL-7 research include:
Producing consistently active recombinant protein (requires specific refolding conditions and purification steps)
Distinguishing direct versus indirect effects of IL-7 on different cell populations
Translating findings between species models (mouse vs. human differences)
Addressing variability in IL-7 receptor expression across immune cell populations
Developing standardized assays for IL-7 bioactivity
These challenges can be addressed through rigorous experimental design, careful validation of reagents, use of multiple complementary methodologies, and where possible, parallel studies in both human and mouse systems to identify species-specific differences. The purification protocol involving sequential chromatographic techniques has been shown to produce highly pure, biologically active rhIL-7 suitable for research applications .