IL-15 signals via a unique trans-presentation mechanism:
IL-15/IL-15Rα complexes form on antigen-presenting cells (APCs) and bind to IL-2Rβ/γc receptors on effector cells (e.g., T, NK cells) .
Cis-presentation occurs when soluble IL-15 binds IL-15Rα on the same cell, activating intracellular STAT3/5/6 pathways .
Mechanism | Key Features | Target Cells |
---|---|---|
Trans-presentation | APC-dependent, immune synapse formation | T cells, NK cells |
Cis-presentation | Autocrine/paracrine, local activation | Monocytes, macrophages |
IL-15 drives proliferation and survival of multiple immune cells:
Amplifies neutrophil recruitment via NF-κB-dependent IL-8 production .
Linked to autoimmune pathologies (e.g., rheumatoid arthritis, psoriasis) .
System | Yield | Advantages | Reference |
---|---|---|---|
E. coli | 1–5 mg/L | Cost-effective, no glycosylation | |
HEK293 | 10–20 mg/L | Proper folding, post-translational modifications | |
Pichia pastoris | Up to 75 mg/L | High yield, scalable |
Preclinical Data: Synergizes with checkpoint inhibitors (anti-PD-1) to enhance tumor infiltration by CD8+ T/NK cells .
Clinical Trials: Phase I/II studies show IL-15 variants (e.g., hetIL-15) improve survival in metastatic melanoma and renal carcinoma .
Recombinant Human Interleukin-15 (IL-15) is produced by expressing a DNA sequence encoding amino acids 49-162 of human IL-15 in E. coli. SDS-PAGE and HPLC analyses demonstrate a purity exceeding 97% for the full-length mature protein. Biological activity is confirmed by a murine CTLL-2 cell proliferation assay (ED50 < 0.5 ng/ml, specific activity > 2.0 x 106 IU/mg). Endotoxin levels are less than 1.0 EU/µg, as determined by the LAL method. This tag-free IL-15 protein is available in stock; however, custom services are available for the addition of specific tags. Applications include the generation of anti-IL-15 antibodies and immunological research.
IL-15 is a pro-inflammatory cytokine crucial for the development, survival, and activation of NK, T, and B cells. Binding to the IL-2/IL-15Rβ/γc heterodimeric receptor activates the JAK-STAT, PI3K-Akt, and MAPK signaling pathways. JAK-STAT signaling stimulates transcriptional activation of genes such as bcl-2 and proto-oncogenes (c-myc, c-fos, and c-jun), while PI3K-Akt and MAPK pathways promote cell survival and activation. While IL-15 exhibits reported anti-tumor effects on the immune system, it can also initiate and promote various malignancies.
Interleukin-15 (IL-15) is a cytokine that stimulates T-lymphocyte proliferation. This stimulation requires interaction between IL-15 and components of the IL-2 receptor, including IL-2Rβ and likely IL-2Rγ, but not IL-2Rα. In neutrophils, IL-15 stimulates phagocytosis, potentially through signaling via the IL-15 receptor (IL-15Rα, IL-2Rβ, and IL-2Rγ subunits), leading to SYK kinase activation.
Recombinant human IL-15 is a 14-15 kDa non-glycosylated protein belonging to the 4-α helix bundle family of cytokines that includes IL-2, IL-4, IL-7, IL-9, and IL-21 . Despite having distinct primary sequences from IL-2, IL-15 folds into a similar 4-α helix bundle structure . The protein functions as a monomer and was discovered simultaneously by two independent research groups in 1994 . Expression systems like E. coli and P. pastoris can produce active recombinant human IL-15 with preserved structural integrity and biological function .
While IL-15 and IL-2 share receptor subunits (IL-2Rβ and common γ-chain), their biological functions are fundamentally different due to unique receptor biology . IL-15 utilizes a specific high-affinity IL-15Rα subunit rather than IL-2Rα . Unlike IL-2, IL-15 does not affect T-regulatory cells or activation-induced cell death (AICD) . The primary mode of IL-15 activity in vivo involves trans-presentation, where IL-15 bound to IL-15Rα on one cell is presented to neighboring cells expressing IL-2/15Rβγc receptors . This trans-presentation mechanism is critical for supporting NK cell development and homeostasis, distinguishing IL-15 from IL-2, which primarily acts through secretion and cis-binding to its receptor .
IL-15 primarily affects cytotoxic lymphocytes, with NK cells and CD8+ T cells being particularly responsive to IL-15 stimulation . These cells express the necessary IL-2/15Rβγc receptor components for IL-15 signaling . IL-15 also supports the development and function of NKT cells and γδT cells . When stimulated with IL-15, NK cells and CD8+ T cells show increased expression of Bcl-2 and Ki67, promoting their survival and proliferation . Additionally, IL-15 induces activated B-cells to produce immunoglobulins, establishing a connection between innate and adaptive immunity .
Several expression systems have proven effective for producing biologically active recombinant human IL-15. E. coli-based expression systems can generate non-glycosylated IL-15 monomers of approximately 13 kDa that maintain functional activity in immunological assays . The yeast Pichia pastoris has also demonstrated capacity for high-level expression of active recombinant human IL-15 . When selecting an expression system, researchers should consider that IL-15 translation is tightly regulated through multiple negative regulatory elements in natural settings, which may impact recombinant production strategies . Quality control testing should include binding assays with IL-15Rα and IL-2Rβ receptors, as shown in SPR and BLI assays with affinity constants in the nanomolar range (9.5-10.2 nM) .
Bioactivity assessment of recombinant human IL-15 should employ multiple complementary approaches. Receptor binding assays using surface plasmon resonance (SPR) or bio-layer interferometry (BLI) can quantify the binding affinity to IL-15Rα and IL-2Rβ with expected affinity constants around 10 nM . Functional assays should measure IL-15's ability to promote proliferation of IL-15-dependent cell lines or primary NK and CD8+ T cells . Researchers can also assess upregulation of Ki67 and Bcl-2 as markers of proliferation and survival, respectively, along with functional readouts such as increased production of perforin, granzyme B, and IFN-γ . When studying IL-15 in the context of immunosuppression, researchers can measure its ability to overcome Treg-mediated suppression of effector T cells, which is a distinguishing feature compared to IL-2 .
For research-grade recombinant human IL-15, long-term storage should be at -80°C, with aliquoting recommended to avoid repeated freeze-thaw cycles that can degrade protein structure and function . For working solutions, IL-15 can typically be stored at 4°C for up to one week, though specific formulation buffers may alter stability profiles. When designing experiments, researchers should account for IL-15's relatively short plasma half-life of approximately 2.5 hours following intravenous administration . To overcome stability limitations, researchers can explore IL-15/IL-15Rα complexes, which exhibit enhanced stability and activity compared to IL-15 monomers . When preparing working solutions, low-protein binding tubes and filters should be used to minimize protein loss through adsorption.
To study IL-15 trans-presentation, researchers can develop experimental models that recapitulate this unique signaling mechanism. Bone marrow chimera approaches using IL-15−/− and IL-15Rα−/− mice have provided clear evidence that IL-15 trans-presentation by IL-15Rα to NK cells expressing IL-2/15Rβγc is the major mode supporting NK cell development and homeostasis . For in vitro studies, researchers can generate cell lines expressing IL-15Rα to present IL-15 to responder cells expressing IL-2/15Rβγc. Co-culture systems can be established with fixed antigen-presenting cells pre-loaded with IL-15 to isolate trans-presentation effects. Alternatively, soluble IL-15/IL-15Rα complexes can be generated to mimic trans-presentation in solution . Sophisticated microscopy techniques, including single-molecule tracking and FRET analysis, can visualize the dynamics of IL-15/IL-15Rα interactions with IL-2/15Rβγc at cell-cell interfaces.
Researchers investigating IL-15's role in anti-tumor immunity can employ several sophisticated approaches. In vitro co-culture systems of IL-15-activated NK or CD8+ T cells with tumor cell lines can assess direct cytotoxicity, with flow cytometry-based killing assays or real-time impedance measurements providing quantitative readouts . For mechanistic studies, researchers should analyze the XCL1-cDC1 recruitment axis, as IL-15-activated tumor-infiltrating immune cells upregulate XCL1 secretion, which recruits conventional type I dendritic cells (cDC1) with cross-presentation capabilities . This creates a positive feedback immune surveillance loop through CXCL9/10 production and CXCR3+ lymphocyte recruitment . Syngeneic or humanized mouse tumor models comparing IL-15 monotherapy versus combination with checkpoint inhibitors or monoclonal antibodies can evaluate therapeutic potential, with endpoints including tumor growth kinetics, survival, and immune infiltrate characterization.
IL-15 plays a crucial role in the homeostatic survival and proliferative activation of memory T cells . To study this phenomenon, researchers can utilize adoptive transfer experiments with CFSE-labeled memory T cells into recipients treated with or without IL-15/IL-15Rα complexes, tracking proliferation, phenotypic markers, and functional responses to antigen re-challenge. Ex vivo culture systems of sorted memory T cell populations with IL-15 can assess survival pathway activation, with particular attention to Bcl-2 upregulation and resistance to apoptosis . Transcriptomic analysis of IL-15-stimulated versus unstimulated memory T cells can identify gene expression signatures associated with maintenance and functional potential. For long-term studies, investigators can employ inducible IL-15 or IL-15Rα knockout models to determine the temporal requirements for IL-15 signaling in memory T cell persistence across different tissue compartments.
When designing experiments to investigate IL-15 for cancer immunotherapy applications, researchers should consider several key factors. First, the short half-life of recombinant human IL-15 monomer (approximately 2.5 hours) necessitates frequent dosing or development of stabilized formulations like IL-15/IL-15Rα complexes . Dose escalation studies should be carefully designed, as clinical trials have demonstrated dose-dependent toxicities including fever, chills, thrombocytopenia, and hypotension at higher concentrations . Different administration routes (intravenous bolus versus subcutaneous) result in distinct pharmacokinetic profiles and toxicity patterns that should be systematically compared . Researchers should include appropriate biomarkers to assess IL-15 activity, including peripheral NK and CD8+ T cell counts, expression of Ki67 and Bcl-2, and functional readouts like perforin, granzyme B, and IFN-γ production .
To investigate IL-15's capacity to overcome immunosuppression, researchers should design experiments that directly compare IL-15 with IL-2 in suppressive environments. Co-culture systems incorporating purified regulatory T cells (Tregs) from peripheral blood or tumor tissues with effector T cells or NK cells can assess IL-15's ability to maintain effector cell function despite Treg presence . Key readouts should include proliferation (measured by CFSE dilution or Ki67 expression), cytokine production (IFN-γ, TNF-α), cytotoxic molecule expression (perforin, granzymes), and target cell killing capacity . Mechanistic studies should examine IL-15-mediated resistance to apoptosis in the presence of Tregs, with particular attention to Bcl-2 family protein regulation . The experimental design from the search results showed that IL-15, unlike IL-2, promoted proliferation, effector function, and apoptosis resistance of effector T cells and EBV-CTLs in the presence of Tregs isolated from healthy donors and Hodgkin lymphoma patients .
Researchers working with recombinant human IL-15 may encounter several technical challenges. The protein's short half-life (approximately 2.5 hours in vivo) can lead to inconsistent results in prolonged experiments . To address this, researchers should implement regular dosing schedules or utilize stabilized forms like IL-15/IL-15Rα complexes . Protein adsorption to laboratory plasticware can reduce effective concentrations; pre-coating tubes and pipette tips with carrier proteins or using low-binding materials can minimize this issue. Functional assays may show variability due to differences in receptor expression on target cells; researchers should characterize IL-2/15Rβγc expression on test cells and include positive controls with validated IL-15-responsive cell lines. The tight translational regulation of natural IL-15 can affect recombinant production efficiency . Expression systems like P. pastoris that have demonstrated high-level production capabilities should be considered when larger quantities are needed .
Distinguishing IL-15-specific effects from those of other common γ-chain cytokines requires careful experimental design. Receptor blocking experiments using antibodies specific to IL-15Rα can isolate IL-15-dependent effects while leaving other γ-chain cytokine pathways intact. Comparative studies should include matched concentrations of IL-2, IL-7, IL-9, and IL-21 alongside IL-15 to identify unique versus shared outcomes . Researchers can exploit IL-15's distinctive trans-presentation mechanism by using IL-15/IL-15Rα complexes, which specifically activate IL-15 signaling pathways . Gene expression profiling can identify IL-15-specific transcriptional signatures that differ from other γ-chain cytokines. For in vivo studies, cytokine-specific or receptor-specific knockout models provide definitive tools to delineate non-redundant functions. The unique capability of IL-15 to support NK cell development in the absence of T cells (unlike IL-2) offers another distinguishing feature that can be leveraged in experimental designs .
Significant advances in protein engineering have addressed several limitations of native IL-15. The development of IL-15/IL-15Rα complexes mimics physiological trans-presentation, resulting in enhanced stability and activity compared to IL-15 monomers . These complexes demonstrate improved pharmacokinetic properties and more potent biological effects. Several modified IL-15 agonists have progressed to clinical testing, including SO-C101 (SOT-101), hetIL-15 (NIZ985), P22339 (SHR-1501), NKTR-255, HCW9218, HCW9201, ALT-803 (N-803), BJ-001, and OBX-115 . Mutation strategies have been employed to enhance protein stability, optimize receptor binding, or modulate signaling pathway activation. Researchers investigating novel IL-15 variants should conduct comparative binding studies to IL-15Rα and IL-2/15Rβγc receptors, assess stability under physiological conditions, and evaluate functional potency through lymphocyte proliferation and activation assays .