Expression System: Produced in HEK 293 cells, which enable post-translational modifications mimicking native human IL-7 .
Formulation: Lyophilized from phosphate-buffered saline (PBS) and typically reconstituted in PBS containing 0.1% human serum albumin (HSA) or bovine serum albumin (BSA) .
Storage: Stable for ≥1 year at -20°C; reconstituted aliquots retain activity for up to 3 months at -20°C . Repeated freeze-thaw cycles degrade efficacy .
IL-7 Human, His binds the IL-7 receptor (IL-7Rα/γc), activating downstream pathways (e.g., JAK-STAT5, PI3K/Akt/mTOR) to:
Promote survival and expansion of naïve and memory T cells .
Enhance B cell progenitor differentiation and V(D)J recombination during TCRβ development .
Cancer Immunotherapy: In phase I trials, recombinant IL-7 increased CD4⁺/CD8⁺ T cell counts in refractory cancer patients, peaking at 3 weeks post-administration .
Infectious Diseases: Improved lymphocyte recovery in HIV patients on HAART and reduced JC viral load in progressive multifocal leukoencephalopathy .
COVID-19: IL-7 therapy doubled lymphocyte counts in critically ill patients with severe lymphopenia .
In Vitro Studies: Used to stimulate proliferation of murine 2E8 cells (specific activity: 3.3–5 million units/mg) .
Immune Reconstitution: Investigated for reversing lymphopenia post-chemotherapy or bone marrow transplantation .
Autoimmunity Research: Dysregulated IL-7 signaling is implicated in multiple sclerosis and rheumatoid arthritis, making this reagent valuable for mechanistic studies .
IL-7 exerts its biological effects through the IL-7 receptor (IL-7R), which is a heterodimeric complex consisting of the IL-7Rα chain (CD127) and the common gamma chain (γc, CD132). The γc chain is shared by several cytokine receptors including IL-2, IL-4, IL-9, IL-15, and IL-21. Notably, the IL-7Rα chain is also shared with thymic stromal lymphopoietin (TSLP), creating potential for signaling crossover . When IL-7 binds to its receptor, it initiates signaling cascades that promote lymphocyte development, proliferation, and survival. This signaling is particularly crucial for T-cell development and homeostasis in humans, as demonstrated by the severe T-cell deficiencies observed in patients with mutations in the IL-7Rα chain .
Phase I clinical trials of rhIL-7 have demonstrated dose-dependent biological effects in humans. In a study where subjects with incurable malignancy received rhIL-7 subcutaneously at doses ranging from 3 to 60 μg/kg every other day for two weeks, the following effects were observed:
At doses ≥10 μg/kg, there was a marked increase in peripheral CD3+, CD4+, and CD8+ lymphocytes in a dose-dependent but age-independent manner
Preferential expansion of CD4+ naive and central memory T cells as well as CD8+ naive T cells
Statistically significant broadening of T-cell repertoire diversity in both CD4+ and CD8+ T cells
Restoration of a rejuvenated circulating T-cell profile resembling that seen earlier in life
Less proliferation of CD4+/FoxP3+ regulatory T cells, resulting in a decreased proportion of circulating regulatory T cells
In some subjects, induction of a marked but transient polyclonal proliferation of bone marrow pre-B cells and an increase in circulating transitional B cells
These findings highlight the potent immunomodulatory effects of rhIL-7 at specific dosages, providing crucial information for designing research protocols involving this cytokine.
Based on clinical trial data, researchers working with rhIL-7 should monitor several safety parameters:
Researchers should consider these parameters when designing safety monitoring protocols for experiments involving rhIL-7 administration.
Recent research on the IL-7/IL7Rα axis has identified several single nucleotide polymorphisms (SNPs) in the IL7R gene that can influence IL-7 signaling and potentially affect experimental outcomes:
rs3194051GG genotype was found to be more frequent in non-survivors than in survivors of community-acquired pneumonia (CAP)
rs987106TT was more frequent and rs3194051AA less frequent in patients requiring intensive care
Specific genotypes (rs6897932CC, rs987106AA, and rs3194051GG) were associated with the highest levels of soluble IL-7 receptor (sIL7Rα), while rs6897932TT showed the lowest sIL7Rα levels
These genetic variations can impact IL-7 signaling efficiency and alter biological responses to IL-7. Researchers should consider genotyping experimental subjects or cell lines for these IL7R SNPs when conducting experiments with IL-7, as genetic variability may explain inconsistent results between different experimental systems.
To maintain optimal bioactivity of recombinant human IL-7 with His-tag, the following storage and handling protocols are recommended:
Store lyophilized protein at -20°C to -80°C
After reconstitution, store working aliquots at -20°C to -80°C to avoid repeated freeze-thaw cycles
For short-term use (less than one week), storage at 4°C is acceptable
Reconstitute in sterile, buffered solutions (typically PBS) at a concentration of 0.1-1.0 mg/ml
Avoid repeated freeze-thaw cycles as these can degrade the protein and diminish bioactivity
Use low-protein binding tubes and pipette tips to minimize protein loss during handling
Include carrier proteins (such as BSA) at 0.1-0.5% in working solutions to prevent loss due to adsorption to plastic surfaces when working with dilute solutions
These practices help maintain the structural integrity and biological activity of rhIL-7 for research applications.
Several functional assays can be employed to validate the bioactivity of rhIL-7 preparations:
Proliferation assays: Using IL-7-dependent cell lines or primary T cells to measure proliferative responses through methods such as tritiated thymidine incorporation or CFSE dilution
Phospho-flow cytometry: Measuring phosphorylation of STAT5, a downstream signaling molecule in the IL-7 pathway, in responsive cells following IL-7 stimulation
Survival assays: Assessing the anti-apoptotic effects of IL-7 on lymphocytes by measuring markers of apoptosis (Annexin V, caspase activation) after cytokine withdrawal and IL-7 rescue
T-cell receptor excision circle (TREC) analysis: Quantifying thymic output in response to IL-7 treatment in vivo
CD127 (IL-7Rα) receptor modulation: Monitoring the downregulation of surface IL-7Rα expression following IL-7 exposure, which indicates functional receptor engagement
Gene expression analysis: Measuring upregulation of IL-7-responsive genes such as Bcl-2 following treatment
These assays provide complementary information about the biological activity of IL-7 preparations and can help ensure experimental reproducibility.
Based on clinical and preclinical studies, researchers can optimize rhIL-7 dosing regimens using the following approach:
Dose-response assessment: In clinical studies, biological effects were observed at doses ≥10 μg/kg, with a dose-dependent increase in effects up to 60 μg/kg . Start with a dose-ranging study in your model system to establish the minimal effective dose.
Administration schedule: Clinical trials used an every-other-day administration for 2 weeks . Consider that continuous IL-7 exposure leads to receptor downregulation, potentially limiting efficacy with daily dosing.
Route of administration: Subcutaneous administration was effective in clinical trials , but intravenous, intraperitoneal, or local administration may be appropriate depending on the experimental model.
Monitoring receptor downregulation: The sharp decrease in cell cycling observed during the second week of treatment despite sustained IL-7 serum levels suggests IL-7Rα downregulation plays a role in the self-limited effects . Consider measuring IL-7Rα expression to optimize dosing intervals.
Species differences: Be aware that humans appear approximately 10-30 fold more sensitive to rhIL-7 than non-human primates, with biological activity observed at the lowest dose tested (3 μg/kg) in human trials .
This systematic approach can help establish optimal dosing regimens for specific research applications and experimental models.
The soluble IL-7 receptor (sIL7R) plays a complex role in modulating IL-7 activity and should be considered in experimental design:
Biomarker potential: In community-acquired pneumonia patients, elevated plasma sIL7Rα levels were associated with increased mortality (AUC of 0.71 for predicting 30-day mortality) and disease severity .
Genetic influence: Specific IL7R SNPs (rs6897932CC, rs987106AA, and rs3194051GG) were associated with higher sIL7Rα levels, while rs6897932TT carriers showed the lowest levels . This genetic component should be considered when interpreting experimental results.
Functional significance: While traditionally viewed as an inhibitor that competes with membrane-bound receptor, some studies suggest sIL7R may extend IL-7 half-life under certain conditions by protecting it from consumption, potentially acting as both an antagonist and an agonist depending on the context.
Experimental considerations: Researchers should consider measuring sIL7R levels in experimental systems, particularly in scenarios involving inflammatory conditions or when using primary human cells with unknown IL7R genotypes.
The complex relationship between IL-7 and sIL7R highlights the importance of considering this interaction when designing and interpreting experiments involving IL-7 signaling.
The IL-7/HGFβ hybrid cytokine represents an intriguing area for advanced research applications:
Enhanced proliferative capacity: When IL-7 associates with hepatocyte growth factor beta (HGFβ), it forms a hybrid cytokine (IL-7/HGFβ) that induces greater proliferation of CFU-S, SLPs, and pre–pro-B cells than does native IL-7 .
Dual receptor signaling: The hybrid cytokine signals through both the IL-7 receptor (IL-7Rα plus γc) and c-Met (the receptor for HGF), potentially activating multiple signaling cascades simultaneously .
Research applications: This hybrid cytokine may have applications in:
Enhancing hematopoietic stem cell expansion ex vivo
Improving immune reconstitution protocols
Developing novel immunotherapeutic approaches
Experimental considerations: When using IL-7 in complex biological systems where HGFβ may be present (such as bone marrow cultures or hepatic microenvironments), researchers should consider the potential formation of this hybrid cytokine and its impact on experimental outcomes.
This represents an advanced area of IL-7 biology that may offer new therapeutic and research opportunities beyond conventional IL-7 applications.
Recombinant human IL-7 has distinct effects on different T-cell subsets, which has important implications for immunotherapy research:
Differential expansion of T-cell subsets:
Increased T-cell repertoire diversity:
Implications for immunotherapy research:
The preferential expansion of naive and central memory T cells over regulatory T cells creates a favorable ratio for immunotherapy approaches
The broadening of T-cell repertoire diversity may enhance responses to tumor antigens or infectious agents
The preserved or increased in vitro responsiveness to anti-CD3 stimulation suggests functional competence of the expanded T cells
These effects position rhIL-7 as a potential adjunct to cancer immunotherapy, particularly in scenarios requiring expansion of the T-cell compartment while maintaining or enhancing T-cell receptor diversity.
Research on the IL-7/IL7Rα axis in community-acquired pneumonia (CAP) provides insights into potential applications for addressing infection-associated lymphocytopenia:
Association with disease severity:
Genetic susceptibility factors:
Potential therapeutic applications:
Supplementation with rhIL-7 might help restore lymphocyte counts and function in severe infections associated with lymphocytopenia
The well-tolerated dose range established in cancer patients (10-60 μg/kg) provides a starting point for investigating therapeutic applications in infectious disease settings
Research considerations:
Patient selection might be guided by IL7R genotyping and plasma IL-7/sIL7R levels
Timing of intervention may be critical, with earlier intervention potentially producing better outcomes
These findings suggest that IL-7 supplementation could represent a novel approach to address infection-associated immunosuppression, particularly in genetically susceptible individuals.
The human recombinant IL-7 with a His tag is a non-glycosylated polypeptide chain containing 152 amino acids, with a molecular mass of approximately 21.97 kDa . The His tag, which consists of a sequence of histidine residues, is added to the N-terminus of the protein to facilitate purification through affinity chromatography . This recombinant protein is typically produced in Escherichia coli (E. coli) expression systems .
IL-7 is essential for the development of lymphoid progenitors and the proliferation of T and B cells . It acts as a growth factor for these cells and has anti-apoptotic properties, which help in the survival of naive and memory T cells . Additionally, IL-7 has been shown to have biological effects on natural killer (NK) cells and myeloid lineages .
Recombinant IL-7 is widely used in research and clinical applications. Some of its key applications include:
The recombinant IL-7 with a His tag is supplied as a sterile filtered clear solution, typically in phosphate-buffered saline (PBS) with 50% glycerol . For short-term storage, it can be kept at 4°C if used within 2-4 weeks. For long-term storage, it should be frozen at -20°C to maintain its stability .