Inducers: Viral infections (e.g., influenza, HCV), double-stranded RNA, LPS .
Producers: Dendritic cells, macrophages, Th17 cells, and mast cells .
Activates JAK1 and TYK2, leading to phosphorylation of STAT1/2 and formation of ISGF3 (STAT1/2 + IRF9) .
Downstream effects: Upregulation of MHC class I, antiviral proteins (e.g., ISG15), and suppression of viral replication .
Macrophages: Enhances TLR-induced IL-12p40 and TNF-α production .
B Cells: Amplifies TLR7/8-driven IgG/IgM and IL-6 secretion .
Th2 Polarization: Suppresses GATA3 and Th2 cytokines (IL-4, IL-5) .
Rheumatoid Arthritis (RA): Elevated IL-29 correlates with synovial inflammation .
Systemic Lupus Erythematosus (SLE): Dysregulated IL-29 contributes to IFN signature .
Obesity: Promotes adipose tissue inflammation, insulin resistance, and macrophage M1 polarization .
HCV Infection: IL-29 expression is elevated in untreated chronic HCV patients and declines post-treatment .
Cancer Immunotherapy: Enhances TRAILR1 and p18/p27, potentiating apoptosis in cervical cancer .
Study Focus | Key Outcome | Citation |
---|---|---|
Obesity/Diabetes | IL-29 ↑ MCP-1, ↓ insulin sensitivity | |
Viral Hepatitis | IL-29 synergizes with IFN-α for HCV clearance | |
Autoimmunity | IL-29 blockade reduces IL-12 in RA |
Interleukin-29 (IL-29), also known as interferon-lambda 1 (IFN-λ1), belongs to the type III interferon family. While distantly related to type I interferons (IFNs) and the IL-10 family, IL-29 shares functional similarities with type I IFNs. Its expression is induced by viral infection, and it binds to a heterodimeric receptor composed of IL-10 receptor beta (IL10RB) and IL-28 receptor alpha. IL-29 exhibits antiviral, antiproliferative, and antitumor activities. Though similar to type I IFNs, IL-29 generally exhibits less potency and affects a narrower range of cell lines. Notably, the genes encoding IFN-lambda 1, IFN-lambda 2, and IFN-lambda3 are clustered on human chromosome 19. IL-29 uniquely induces the expression of CXC chemokine mRNAs lacking the ELR motif (ELR(-)) in human peripheral blood mononuclear cells, independent of IFN-gamma. Additionally, IL-29 can generate tolerogenic dendritic cells (DCs), potentially counteracting IFN-beta functions. Produced in response to viral infection, IL-29 activates both monocytes and macrophages, leading to the production of a specific set of cytokines. This suggests a key role for IL-29 in initiating innate immune responses at the site of viral infection. The antiviral and antiproliferative functions of IFN-Lambda 1 rely on specific tyrosine residues within the IFN-Lambda 2 receptor.
Interleukin-29, IL-29, IFN-Lambda 1, IFN-Lambda 1, Cytokine ZCYTO21, IL29, IFNL1, ZCYTO21.
IL-29 (interferon-lambda 1) belongs to the helical cytokine family and is classified as a type III interferon. It was discovered in 2003 alongside IL-28A and IL-28B (IFN-lambda 2 and IFN-lambda 3, respectively) . While IL-28A and IL-28B are structurally nearly identical to each other, IL-29 is more structurally unique . Despite possessing only weak homology to IFN-alpha, IL-29 induces the expression of genes normally activated by IFN-alpha/beta . IL-29 is encoded by the IL29 gene located on chromosome 19 in humans .
Unlike the universal expression of IFN-alpha receptors, the IL-29 receptor distribution is more restricted, suggesting evolution of specialized functions. When designing experiments to study IL-29's role within the interferon family, researchers should consider using comparative transcriptomics to distinguish IL-29-specific gene signatures from broader type I interferon responses.
IL-29 signals through a distinct receptor composed of IL-28R1 and IL-10R2 subunits . This distinguishes it from type I interferons, which bind to the IFN-alpha/beta receptor. Despite this difference in receptor utilization, IL-29 activates similar downstream pathways, inducing STAT phosphorylation that leads to antiviral gene expression .
The IL-29 receptor demonstrates tissue-specific distribution patterns, being widely expressed on nonhematopoietic cells but having limited expression on leukocytes . This receptor distribution explains the targeted effects of IL-29 on epithelial cells and hepatocytes while sparing many immune cell populations.
When designing receptor-targeting experiments, researchers should consider using receptor-blocking antibodies specific to IL-28R1 rather than IL-10R2, as the latter is shared with IL-10 family cytokines and would confound results by affecting multiple signaling pathways.
IL-29 exhibits significant antiviral activity, particularly in hepatocellular carcinoma cells infected with viruses such as encephalomyocarditis virus, with an effective dose (ED50) ranging from 0.500-6.00 ng/mL . The cytokine primarily targets epithelial cells, with high expression during infections of gastrointestinal and respiratory tracts, and mucosal regions .
Methodologically, when studying IL-29's antiviral effects, researchers should establish tissue-specific models. For respiratory infections, human bronchial epithelial cell cultures are preferred, while intestinal organoids better represent enteric virus responses. Hepatocytes (like HepG2 cells) serve as excellent models for hepatotropic viruses . Time-course experiments are essential as IL-29 production peaks approximately 24-48 hours post-infection in most experimental systems.
Experimentally validated target cells include:
Keratinocytes and melanocytes (responsive)
Hepatocytes (highly responsive)
Epithelial cells (primary targets)
Endothelial cells, subcutaneous adipocytes, and fibroblasts (non-responsive)
Recent research has established IL-29's involvement in obesity-induced inflammation and insulin resistance . When designing studies in this area, researchers should consider both in vitro and in vivo approaches:
For in vitro studies:
Human Simpson-Golabi-Behmel syndrome (SGBS) adipocytes represent an excellent model for studying IL-29's effects on adipocyte metabolism
Measure inflammatory cytokine expression (IL-1β, IL-8, MCP-1) using qPCR and ELISA
Assess insulin sensitivity via glucose uptake assays and Western blotting for GLUT4 expression and AKT phosphorylation
Implement macrophage-adipocyte co-culture systems to mimic the obese microenvironment
For in vivo studies:
High-fat diet (HFD)-induced obese mice provide a physiologically relevant model
Key parameters to measure include:
The finding that serum IL-29 levels are significantly elevated in obese patients compared to non-obese controls provides clinical relevance to these investigations . Researchers should carefully control for confounding variables such as concurrent viral infections that may independently elevate IL-29.
IL-29 demonstrates complex effects in cancer biology, either promoting or inhibiting tumor growth depending on the cancer cell type . This dichotomy necessitates careful experimental design:
For cancer studies, researchers should:
Begin with cell-type specific screening to determine IL-29 receptor expression levels across different cancer cell lines
Conduct dose-response experiments (typically 0.12-0.6 ng/mL range) to identify optimal concentrations for each cell type
Implement both proliferation assays (MTT, BrdU) and apoptosis assays (Annexin V, TUNEL) to comprehensively assess IL-29's effects
Examine changes in tumor microenvironment using co-culture systems with cancer cells and relevant stromal or immune cells
Validate in vitro findings using xenograft models with IL-29 treatment or genetic manipulation of the IL-29 signaling pathway
When interpreting results, researchers should consider the specific molecular subtype of cancer being studied, as this appears to influence responsiveness to IL-29 treatment.
A significant methodological challenge in IL-29 research is that IL-29 exists only as a pseudogene in mice, limiting the use of standard murine models . To overcome this limitation:
Use humanized mouse models where human IL-29 is expressed under tissue-specific promoters
Consider alternative animal models that express functional IL-29 orthologs
Implement in vitro human cell systems that better recapitulate human IL-29 biology
For infections studies, use chimeric viruses or transgenic mice expressing human IL-29 receptors
Employ ex vivo human tissue explants for more physiologically relevant IL-29 research
When using recombinant IL-29 in experimental systems, researchers should be aware of different formulations:
E. coli-derived IL-29 (non-glycosylated) is suitable for structural studies
HEK293-expressed IL-29 (glycosylated, 27-31 kDa) better represents the natural form for functional studies
For accurate quantification and functional assessment of IL-29:
Expression Analysis:
qRT-PCR for mRNA expression (preferred for early response detection)
ELISA for protein quantification in serum or culture supernatants (detection range typically corresponds to 0.12-0.6 ng/mL activity range)
Western blotting for protein detection (expect bands at 25-31 kDa, with variation due to glycosylation)
Immunohistochemistry for tissue localization studies
Functional Assays:
Antiviral activity measurement using reporter cell lines
Specific activity determination (approximately 8.00 x 10^5 IU/mg for recombinant preparations)
STAT phosphorylation assays (Western blot or flow cytometry)
Reporter gene assays using IFN-stimulated response element (ISRE) constructs
When studying IL-29 in complex biological samples, researchers should account for potential interfering factors:
Presence of soluble receptor splice variants that can bind IL-29
Cross-reactivity with other type III interferons
Stability issues (avoid repeated freeze-thaw cycles for experimental samples)
Proper handling of recombinant IL-29 is crucial for experimental reproducibility. Recommended procedures include:
Storage Condition | Product Form | Temperature | Maximum Storage Duration |
---|---|---|---|
Lyophilized | -20°C to -80°C | Until expiry date | |
Lyophilized | Room temperature | 2 weeks | |
Reconstituted | -20°C to -80°C | 6 months | |
Reconstituted | 4°C | 1 week |
Reconstitution protocol:
Briefly centrifuge the vial before opening
Reconstitute to 0.2 mg/mL in sterile 1x PBS (pH 7.4)
For optimal stability, include 0.1% endotoxin-free recombinant human serum albumin (HSA)
Researchers should avoid repeated freeze-thaw cycles as this significantly reduces protein activity. For long-term experiments, prepare single-use aliquots upon initial reconstitution.
To distinguish direct IL-29 effects from secondary responses:
Compare immediate (0-6 hours) versus delayed (24-72 hours) responses
Use receptor blocking antibodies against IL-28R1
Implement CRISPR/Cas9 knockout of IL-28R1 in target cells
Conduct transcriptomics at multiple time points to identify primary response genes
Use metabolic inhibitors of protein synthesis (cycloheximide) to distinguish between primary and secondary gene induction
Compare responses in IL-28R1-positive versus IL-28R1-negative cell populations within the same tissue context
For complex in vivo studies, conditional cell-type specific knockout models of IL-28R1 provide the most definitive approach to identifying direct versus indirect IL-29 effects.
IL-29 functions cooperatively with other interferons to induce antiviral responses . To investigate these interactions:
Design factorial experimental layouts testing IL-29 in combination with IFN-alpha or IFN-gamma at various concentrations
Analyze synergistic, additive, or antagonistic effects using appropriate statistical models (e.g., Bliss independence model)
Employ phospho-proteomics to map convergent and divergent signaling nodes
Use single-cell RNA sequencing to identify cell populations responsive to IL-29 alone versus combination treatments
Implement CRISPR screens to identify genes essential for IL-29/IFN cooperative functions
Special consideration should be given to temporal aspects, as sequential versus simultaneous cytokine exposure may yield different outcomes. Time-course experiments are essential for identifying the optimal treatment sequence for maximal antiviral effects.
When studying IL-29's therapeutic applications:
Begin with dose-optimization studies (typical range: 0.500-6.00 ng/mL for in vitro work)
For antiviral studies, assess both prophylactic (pre-infection) and therapeutic (post-infection) administration protocols
In models of inflammatory conditions, carefully monitor both pro- and anti-inflammatory effects
Consider cell-type specific delivery systems to target IL-29 to relevant tissues while minimizing off-target effects
For chronic conditions, implement extended treatment protocols with careful assessment of potential compensatory mechanisms and receptor downregulation
When interpreting therapeutic potential, researchers should be aware that IL-29's specific activity (approximately 8.00 x 10^5 IU/mg) can vary between preparations, necessitating standardization against international reference preparations before making comparative efficacy assessments.
IL-29 was discovered as part of the interferon lambda family, which includes IL-28A, IL-28B, and IL-29. These cytokines are structurally related to both type I interferons and the interleukin-10 family . IL-29 is produced by various cells, including monocytes and dendritic cells, in response to viral infections and stimulation by toll-like receptor ligands .
IL-29 exerts its biological effects through a receptor complex composed of IL-28Rα and IL-10Rβ. This receptor is expressed on most non-hematopoietic cells . The primary function of IL-29 is to induce antiviral responses. It upregulates the expression of major histocompatibility complex (MHC) class I molecules on the cell surface and stimulates the production of antiviral proteins such as protein kinase R (PKR), myxovirus resistance protein A (MxA), and 2’,5’-oligoadenylate synthetase (2’,5’-OAS) .
Recombinant human IL-29 is produced using various expression systems, including human embryonic kidney (HEK) 293 cells . The recombinant protein is typically purified to high levels of purity, often exceeding 95% . It is used in research to study its antiviral properties and potential therapeutic applications.
IL-29 has shown promise in various research areas, particularly in antiviral therapies. Its ability to induce a potent antiviral state in cells makes it a valuable tool for studying viral infections and developing new treatments . Additionally, IL-29’s role in modulating immune responses has implications for autoimmune diseases and cancer research .