The protein is synthesized in E. coli using codon-optimized expression systems. Post-expression, the His tag facilitates purification via immobilized metal affinity chromatography (IMAC). Final products undergo rigorous quality control:
IL-13 Human, His is critical for investigating airway hyperresponsiveness (AHR) and mucus hypersecretion. In murine models, IL-13 administration replicates asthma phenotypes, including:
Goblet Cell Hyperplasia: STAT6-dependent mucus overproduction .
Airway Remodeling: Subepithelial fibrosis and eosinophil infiltration .
Receptor Binding: Binds IL-13Rα1/IL-4Rα heterodimer with high affinity (K<sub>D</sub> = 22.5–34.4 nM) .
The protein activates JAK/STAT6 pathways, mimicking native IL-13 activity:
TF-1 Cell Proliferation: EC₅₀ values confirm bioequivalence to endogenous IL-13 .
MUC5AC Upregulation: Induces mucin production in human airway epithelial cells .
IL-13 is an immunoregulatory cytokine predominantly produced by activated Th2 cells, mast cells, and NK cells. It functions as a key regulator of immune responses, particularly in type 2 immunity. Targeted deletion studies in mice have demonstrated IL-13's essential role in Th2 cell development and gastrointestinal parasite expulsion. In human immune regulation, IL-13 exerts significant anti-inflammatory effects on monocytes and macrophages by inhibiting the expression of multiple inflammatory cytokines including IL-1beta, TNF-alpha, IL-6, and IL-8 .
IL-13 also enhances B cell proliferation and induces isotype switching that leads to increased IgE production, which is particularly relevant to allergic responses. The cytokine has demonstrated cross-species reactivity between human and mouse variants, making mouse models valuable for studying human IL-13 biology . Additionally, IL-13 has been identified as a critical factor in asthma pathophysiology, where blocking its activity inhibits disease manifestations .
Methodologically, researchers studying IL-13's basic functions often use cellular proliferation assays with TF-1 cells to assess bioactivity, with effective doses typically less than 15 ng/ml, corresponding to a specific activity exceeding 6.7×10^5 units/mg .
IL-13 significantly alters the phenotype and function of multiple immune cell types. In human monocytes, IL-13 treatment induces substantial changes in cell surface marker expression. Specifically, IL-13 enhances the expression of CD11b, CD11c, CD18, CD29, CD49e (VLA-5), class II MHC molecules, CD13, and CD23. Conversely, it downregulates the expression of CD64, CD32, CD16, and CD14 in a dose-dependent manner .
These phenotypic changes have functional consequences. IL-13 strongly inhibits antibody-dependent cellular cytotoxicity (ADCC) of human monocytes against anti-D coated Rh+ erythrocytes. This occurs even in the presence of IL-10 or IFN-gamma, which normally enhance ADCC activity, indicating that IL-13 profoundly impairs monocyte cytotoxic function .
At the cytokine production level, IL-13 inhibits the production of multiple inflammatory mediators in LPS-activated monocytes, including IL-1 alpha, IL-1 beta, IL-6, IL-8, IL-10, IL-12 p35, IL-12 p40, macrophage inflammatory protein-1 alpha, GM-CSF, G-CSF, IFN-alpha, and TNF alpha. Interestingly, IL-13 enhances the production of IL-1 receptor antagonist (IL-1ra), further contributing to its anti-inflammatory profile .
For researchers studying these effects, flow cytometry assessing surface marker expression, cytotoxicity assays, and cytokine production analysis after LPS challenge represent essential methodological approaches.
IL-13 interacts with two primary receptors: IL-13Rα1, which forms a complex with IL-4Rα to initiate signaling, and IL-13Rα2, which has been identified as having a significant role in IL-13 clearance. Studies with antibodies targeting different IL-13 epitopes have revealed that receptor interactions critically influence IL-13 clearance rates in humans .
Research using two antibodies with distinct epitope specificity—IMA-638 and IMA-026—has provided valuable insights into these mechanisms. IMA-638 allows IL-13 to interact with both IL-13Rα1 and IL-13Rα2 but blocks the recruitment of IL-4Rα to the IL-13/IL-13Rα1 complex. In contrast, IMA-026 competes with IL-13 for interaction with both IL-13Rα1 and IL-13Rα2. Clinical studies demonstrated approximately 10-fold higher circulating titers of captured IL-13 in subjects treated with IMA-026 compared to those receiving IMA-638 .
This difference is attributed to IL-13Rα2's role as a scavenger receptor. Cells with high IL-13Rα2 expression rapidly and efficiently deplete extracellular IL-13. This clearance pathway persists in the presence of IMA-638 but is inhibited by IMA-026 . Unlike mice, which express a soluble form of IL-13Rα2 that regulates IL-13 responses, humans utilize inducible cell surface IL-13Rα2 for IL-13 clearance .
Methodologically, researchers investigating IL-13 receptor interactions should consider both signaling outcomes and clearance kinetics when designing antagonist strategies, as epitope specificity can significantly impact clinical efficacy by altering IL-13 bioavailability.
IL-13 activates multiple signaling pathways in human airway epithelial cells, with the phosphoinositide 3-kinase (PI3K) pathway playing a particularly critical role. When primary human airway epithelial cells are exposed to IL-13, there is time-dependent induction of Akt phosphorylation, indicating PI3K pathway activation .
This PI3K activation is functionally significant, as it leads to critical downstream events, including c-Jun binding to the human IRAK-M gene promoter and subsequent IRAK-M upregulation. The importance of this pathway has been demonstrated experimentally using the PI3K inhibitor wortmannin, which significantly abolishes IL-13-induced IRAK-M expression in human primary airway epithelial cells .
The activation of the PI3K/Akt pathway by IL-13 ultimately leads to suppression of toll-like receptor (TLR) signaling in airway epithelial cells. Specifically, IL-13-induced IRAK-M inhibits TLR2 signaling and suppresses expression of antimicrobial molecules like human β-defensin 2, partly through inhibition of nuclear factor κB activation .
For researchers studying these pathways, key methodological approaches include:
Air-liquid interface (ALI) cultures of primary human airway epithelial cells
Western blotting for phospho-Akt, total Akt, and IRAK-M
Use of specific pathway inhibitors (e.g., wortmannin at 100 nmol/L)
Assessment of downstream functional consequences on TLR signaling and antimicrobial peptide expression
Single-cell RNA sequencing (scRNA-seq) has emerged as a powerful method to delineate IL-13 effects on specific cell populations within heterogeneous tissues like the airway epithelium. This approach reveals how IL-13 differentially regulates gene expression across distinct cell types, providing insights not possible with bulk RNA analysis .
Methodologically, researchers have successfully applied scRNA-seq to primary human bronchial epithelial cells (HBECs) grown in air-liquid interface (ALI) culture. In one comprehensive study, HBECs from four donors were cultured in ALI for 16 days, followed by IL-13 treatment for 7 days to induce mucus hyperplasia. Single-cell analysis yielded data for approximately 2,000 cells per condition, enabling robust statistical comparisons .
This approach allows identification of cell populations (basal, ciliated, and secretory cells) and quantification of cell type-selective effects of IL-13. By applying linear models with terms for cell type, cytokine effect, and their interaction, researchers can detect gene expression changes that are specific to particular cell types rather than uniform across the epithelium .
For researchers implementing this methodology, key considerations include:
Optimization of cell dissociation protocols to maintain viability
Sufficient sequencing depth to detect low-abundance transcripts
Computational approaches that account for donor-to-donor variability
Integration with other datasets (e.g., bulk RNA-seq, ChIP-seq) to validate findings
The power of this approach lies in its ability to reveal unexpected cell type-specific regulatory mechanisms and identify novel therapeutic targets that may be masked in bulk analyses.
IL-13 stimulation activates specific DNA regulatory elements in human cells, which can be identified through chromatin immunoprecipitation sequencing (ChIP-seq) targeting histone modifications associated with active regulatory regions. In human bronchial epithelial cells (HBECs), H3K27ac ChIP-seq has revealed IL-13-responsive regulatory elements across the genome .
A comprehensive analysis of HBECs from four donors grown in air-liquid interface (ALI) culture with or without IL-13 stimulation identified 19,629 H3K27ac peaks, with 602 (3.1%) significantly altered by IL-13 treatment. These included 387 enriched and 215 depleted regions (FDR < 0.1) .
Genomic annotation analysis revealed that approximately one-third of these IL-13-responsive peaks contain putative promoters. Regions enriched in H3K27ac upon IL-13 stimulation were preferentially located near transcription start sites of IL-13-induced genes, while regions depleted of H3K27ac were associated with genes showing decreased expression .
De novo motif analysis identified STAT6 binding sites as the most highly enriched motif in IL-13-induced H3K27ac regions, confirming the central role of STAT6 in IL-13 signaling .
For researchers studying IL-13-responsive regulatory elements, key methodological approaches include:
ChIP-seq targeting H3K27ac or other relevant histone modifications
Integration with transcriptomic data to correlate chromatin changes with gene expression
Motif analysis to identify transcription factor binding sites
Functional validation using reporter assays, such as Massively Parallel Reporter Assays (MPRA)
The identification of IL-13-responsive regulatory elements provides opportunities for mechanistic understanding and potential therapeutic targeting of IL-13-driven pathologies.
Assessing the functional impact of IL-13 on cellular responses requires a multi-faceted methodological approach that captures changes in signaling, gene expression, and cellular physiology. Several key methodologies have proven effective in IL-13 research:
Protein phosphorylation analysis: Western blotting for phosphorylated signaling proteins (e.g., phospho-Akt) at various time points after IL-13 stimulation reveals activation kinetics of key pathways. For example, IL-13 treatment of primary airway epithelial cells induces time-dependent Akt phosphorylation within 120 minutes .
Pathway inhibition studies: Selective inhibitors (e.g., wortmannin for PI3K inhibition) can validate the role of specific pathways in IL-13-induced effects. Pretreatment with 100 nmol/L wortmannin significantly abolishes IL-13-induced IRAK-M expression in human airway epithelial cells .
Cell surface marker analysis: Flow cytometry to assess changes in surface protein expression reveals IL-13's impact on cellular phenotype. IL-13 enhances expression of multiple markers (CD11b, CD11c, CD18, etc.) while decreasing others (CD64, CD32, CD16, CD14) in monocytes .
Functional assays: Antibody-dependent cellular cytotoxicity (ADCC) assays using anti-D coated Rh+ erythrocytes can measure IL-13's inhibitory effects on monocyte cytotoxic activity .
Cytokine production analysis: Measuring cytokine levels after stimulation (e.g., with LPS) in the presence or absence of IL-13 reveals its immunomodulatory effects. IL-13 inhibits production of multiple inflammatory cytokines while enhancing IL-1ra production .
Gene regulatory element analysis: Massively Parallel Reporter Assays (MPRA) can functionally validate IL-13-responsive regulatory elements identified through ChIP-seq. For example, testing candidate regulatory sequences (CRSs) from IL-13-enriched H3K27ac peaks has identified elements that show induced expression after IL-13 treatment .
These complementary approaches provide a comprehensive understanding of IL-13's functional effects across different cell types and biological processes.
Epitope specificity critically influences the efficacy of anti-IL-13 antibodies by altering IL-13 clearance kinetics and bioavailability. Clinical studies comparing antibodies targeting different IL-13 epitopes have revealed significant differences in their impact on circulating IL-13 levels and subsequently their therapeutic efficacy .
Research comparing two antibodies—IMA-638 and IMA-026—that target non-overlapping IL-13 epitopes has provided key insights into this phenomenon. Both antibodies block IL-13 responses mediated through the IL-13Rα1/IL-4Rα complex but do so through different mechanisms. IMA-638 allows IL-13 to bind IL-13Rα1 but blocks recruitment of IL-4Rα, while IMA-026 prevents the initial interaction between IL-13 and IL-13Rα1 .
Clinical studies in healthy subjects and mild asthmatics demonstrated approximately 10-fold higher circulating titers of captured IL-13 in subjects treated with IMA-026 compared to those receiving IMA-638. This difference is attributed to IMA-026's ability to inhibit IL-13 internalization through cell surface IL-13Rα2, which functions as a scavenger receptor in humans .
The mechanistic explanation lies in the preservation or disruption of IL-13's interaction with IL-13Rα2:
IMA-638 allows IL-13 to interact with IL-13Rα2, permitting continued clearance via this pathway
IMA-026 prevents IL-13 from binding IL-13Rα2, inhibiting this clearance mechanism and resulting in higher circulating IL-13 levels
For researchers developing IL-13-targeting therapeutics, these findings highlight the importance of considering not only signaling blockade but also the impact on clearance pathways. The choice of epitope can significantly influence pharmacokinetics and potentially the therapeutic window of anti-IL-13 interventions.
Genomic approaches offer powerful methods to identify novel therapeutic targets in IL-13-mediated diseases by characterizing the full spectrum of IL-13-responsive genes and regulatory elements. These approaches reveal cell type-specific effects and regulatory mechanisms that may not be apparent from traditional methods .
One comprehensive strategy combines single-cell RNA sequencing (scRNA-seq) with chromatin immunoprecipitation sequencing (ChIP-seq) and functional validation through massively parallel reporter assays (MPRA):
Cell type-specific gene identification: scRNA-seq of IL-13-treated human bronchial epithelial cells identifies genes regulated by IL-13 in specific cell populations (basal, ciliated, and secretory cells). This reveals cell type-selective effects of IL-13 that may represent more precise therapeutic targets .
Regulatory element mapping: H3K27ac ChIP-seq identifies IL-13-responsive regulatory elements genome-wide. This approach has identified hundreds of regions with altered H3K27ac levels after IL-13 stimulation, with approximately one-third containing putative promoters .
Transcription factor identification: De novo motif analysis of IL-13-responsive regulatory regions identifies key transcription factors, such as STAT6, that mediate IL-13 responses. These transcription factors and their binding sites represent potential therapeutic targets .
Functional validation: MPRA using candidate regulatory sequences (CRSs) from IL-13-enriched H3K27ac peaks can validate IL-13-responsive elements. In one study, 26 of 1,194 tested CRSs showed induced expression after IL-13 treatment, with fold changes up to 25 times baseline levels .
This integrated genomic approach has several advantages for therapeutic target discovery:
Identifies cell type-specific targets that may allow more precise intervention
Characterizes regulatory elements that could be targeted by novel therapeutic modalities
Reveals transcriptional networks and feedback mechanisms that influence disease progression
Provides a catalog of IL-13-responsive genes and elements for comprehensive pathway analysis
For researchers applying these approaches, key considerations include donor diversity, appropriate control conditions, and statistical methods for integrating multi-omic datasets.
IL-13 significantly impairs antimicrobial immunity in the airway epithelium through complex mechanisms centered on dysregulation of innate immune signaling pathways. A key mechanism involves IL-13-mediated upregulation of IL-1 receptor-associated kinase M (IRAK-M), which functions as a negative regulator of Toll-like receptor (TLR) signaling .
In asthmatic patients, IRAK-M protein levels are increased in the airway epithelium compared to normal epithelium. This upregulation can be recapitulated in vitro by IL-13 treatment of primary human airway epithelial cells . The functional consequence is suppression of TLR2 signaling and reduced expression of antimicrobial factors like human β-defensin 2 (hBD2) .
The molecular pathway involves several steps:
IL-13 stimulation activates the PI3K/Akt pathway in airway epithelial cells
PI3K activation leads to c-Jun binding to the human IRAK-M gene promoter
IRAK-M expression increases, inhibiting TLR2 signaling activation
Nuclear factor κB (NF-κB) activation is suppressed
Expression of antimicrobial factors like TLR2 and hBD2 is reduced
This mechanism helps explain the increased susceptibility to respiratory infections observed in patients with Th2-high asthma. By dampening innate immune responses in the airway epithelium, IL-13 compromises the first line of defense against inhaled pathogens .
For researchers investigating this phenomenon, air-liquid interface cultures of primary airway epithelial cells treated with IL-13 (10 ng/mL) for 6-7 days provide an excellent model system. Key readouts include IRAK-M expression, TLR2 signaling activation, and antimicrobial peptide production in response to microbial challenges .
IL-13 exerts distinct effects on different airway epithelial cell populations, with cell type-specific gene regulation patterns that contribute to asthma pathophysiology. Single-cell RNA sequencing (scRNA-seq) of IL-13-treated human bronchial epithelial cells has revealed these differential responses across basal, ciliated, and secretory cell populations .
To quantify cell type-selective effects, researchers have employed linear modeling approaches with terms for cell type, cytokine effect, and their interaction. This statistical framework allows identification of genes that respond to IL-13 differently across cell types, revealing nuanced regulatory patterns that would be obscured in bulk analysis .
Among the significant findings from this approach:
Cell type-specific IL-13 responses can be identified based on the interaction between IL-13 stimulation and cell type identity
Secretory cells show distinct response patterns compared to basal and ciliated cells
These differential responses likely contribute to the tissue-level changes observed in asthma, including mucus hyperplasia and airway remodeling
This cell type-specific regulation has important implications for therapeutic development, as targeting IL-13 effects in specific cell populations might provide more precise intervention with fewer side effects.
Methodologically, researchers studying these differential effects should consider:
Using air-liquid interface cultures that allow proper differentiation of multiple epithelial cell types
Employing single-cell technologies rather than bulk analysis
Designing statistical approaches that explicitly model cell type-specific responses
Validating key findings in primary cells from both healthy donors and patients with IL-13-mediated diseases
This advanced understanding of cell type-specific IL-13 responses provides a foundation for developing next-generation therapeutics that target specific cellular compartments within the airway epithelium.
Interleukin-13 (IL-13) is a cytokine that plays a crucial role in the immune system, particularly in the regulation of inflammatory and immune responses. It is produced primarily by T helper type 2 (Th2) cells, but also by other cell types such as CD4 cells, natural killer T cells, mast cells, basophils, eosinophils, and nuocytes . The recombinant form of IL-13, tagged with a histidine (His) tag, is widely used in research and therapeutic applications.
IL-13 is a protein encoded by the IL13 gene located on chromosome 5q31.1 . It has a molecular weight of approximately 13 kDa and folds into four alpha-helical bundles . The His tag, typically a sequence of six histidine residues, is added to the recombinant protein to facilitate purification through affinity chromatography .
The recombinant IL-13 (Human, His Tag) is often expressed in HEK293 cells, a human embryonic kidney cell line, which ensures proper protein folding and post-translational modifications . The protein is usually purified to a high degree of purity (>95%) and is available in both lyophilized and liquid forms .
IL-13 is involved in various stages of B-cell maturation and differentiation. It up-regulates CD23 and MHC class II expression and promotes IgE isotype switching of B cells . Additionally, IL-13 down-regulates macrophage activity, thereby inhibiting the production of pro-inflammatory cytokines and chemokines . This cytokine is critical in the pathogenesis of allergen-induced asthma and other allergic conditions .
IL-13 exerts its effects through a multi-subunit receptor complex that includes the alpha chain of the IL-4 receptor (IL-4Rα) and at least one of two known IL-13-specific binding chains, IL-13Rα1 and IL-13Rα2 . Upon binding to its receptor, IL-13 activates the JAK-STAT signaling pathway, leading to the transcription of target genes involved in immune regulation and inflammation .