IL-28A is encoded on human chromosome 19, adjacent to IL-28B and IL-29 . Key features include:
Protein Structure: Mature IL-28A is a 19.8–22.1 kDa protein , sharing 96% amino acid identity with IL-28B and 67% with IL-29 .
Receptor: Binds a heterodimeric receptor composed of IL-10Rβ and IL-28Rα, activating the JAK-STAT pathway .
Property | Details |
---|---|
Gene Location | Chromosome 19 (near IL-28B/IL-29) |
Molecular Weight | 19.8–22.1 kDa |
Receptor Complex | IL-10Rβ + IL-28Rα |
Homology | 96% with IL-28B; 67% with IL-29 |
Induces Mx proteins, 2',5'-oligoadenylate synthetase, and ISGF3G to establish an antiviral state .
Activates monocytes/macrophages, enhancing innate immune responses at infection sites .
Th1 Polarization: Promotes dendritic cell production of IL-12 p70 and IFN-γ, suppressing Th2/Th17 inflammation .
T Cell Activation: Augments IFN-γ release and CD8+ T cell cytotoxicity in vaccine adjuvants .
Crohn’s Disease (CD): Elevated IL-28A in active CD patients disrupts intestinal barriers via JAK-STAT1, reducing ZO-1 and E-cadherin .
Acute Myocardial Infarction (AMI): Serum IL-28A correlates with biomarkers (cTnI, BMI) and exacerbates cardiac dysfunction .
Cancer: Exhibits anti-tumor effects by enhancing CTL responses , but promotes migration in bladder cancer .
Vaccine Adjuvants: Enhances protection against influenza and HIV in preclinical models .
JAK Inhibitors: Mitigate IL-28A-driven epithelial damage in CD .
Biomarker Potential: Serum IL-28A levels correlate with disease severity in AMI and CD .
IL-28A is produced recombinantly in E. coli or human cells for research . Key specifications:
Interleukin 28A (IL-28A), a member of the type III interferon family, shares a distant relationship with type I interferons and the IL-10 family. Viral infections trigger the expression of IL-28A, which subsequently binds to a heterodimeric class II cytokine receptor composed of interleukin 10 receptor, beta (IL10RB) and interleukin 28 receptor, alpha. IL-28A exhibits functional similarities to type I IFNs, including antiviral, antiproliferative, and in vivo antitumor activities. However, its potency is generally lower, and its activity is observed in a more limited range of cell lines. The genes encoding IFN-lambda 1, IFN-lambda 2, and IFN-lambda 3 are located in close proximity on human chromosome 19. IL-28A uniquely induces the mRNA expression of ELR(-) CXC chemokines in human peripheral blood mononuclear cells, independent of IFN-gamma. Furthermore, IL-28A possesses the ability to generate tolerogenic dendritic cells (DCs), a function that could potentially counteract the effects of IFN-beta. Produced in response to viral infection, IL-28A activates both monocytes and macrophages, leading to the production of a specific set of cytokines. This highlights its significant role in initiating innate immune responses at the site of viral infection.
Interleukin-28A, IL-28A, IFN-Lambda 2, IFN-Lambda 2, Cytokine ZCYTO20, IL28A, IFNL2, ZCYTO20.
IL-28A signals through a heterodimeric receptor complex consisting of interleukin 10 receptor beta (IL10RB) and interleukin 28 receptor alpha (IL28RA). This specific receptor composition helps explain IL-28A's more targeted biological activities compared to the broadly expressed type I interferon receptors. The gene encoding human IL-28A is located at GeneID 282616, and the protein has several synonyms including Cytokine Zcyto20, IFNL2, and Interferon lambda 2 .
IL-28A plays several critical roles in the immune system, particularly in antiviral defense:
IL-28A exhibits potent antiviral activity by inducing antiviral protein production in target cells and upregulating MHC class I expression. This enhances the visibility of infected cells to the immune system. It activates the JAK-STAT signaling pathway similar to type I interferons, leading to the expression of interferon-stimulated genes (ISGs) that establish an antiviral state within cells .
Research demonstrates that IL-28A significantly contributes to antiviral immune defense in the intestinal epithelium . It can effectively suppress hepatitis C viral RNA replication , and recent studies suggest a role in COVID-19 pathogenesis, with lower expression potentially linked to more severe disease outcomes .
The protein has immunomodulatory effects, including the ability to induce HLA class I antigen expression in tumor cells, suggesting potential applications in cancer immunotherapy . This upregulation of MHC class I molecules may enhance the recognition of malignant cells by the adaptive immune system.
Upon binding to its heterodimeric receptor complex (IL-28RA/IL-10RB), IL-28A initiates a complex signaling cascade that resembles that of type I interferons but with important distinctions:
The signaling process begins with receptor dimerization, which activates receptor-associated Janus kinases (JAKs). These kinases phosphorylate Signal Transducer and Activator of Transcription (STAT) proteins, primarily STAT1 and STAT2. The phosphorylated STATs form complexes with IRF9 (Interferon Regulatory Factor 9) to create the ISGF3 (Interferon-Stimulated Gene Factor 3) complex .
This complex translocates to the nucleus where it binds to specific DNA sequences known as Interferon-Stimulated Response Elements (ISREs) to regulate gene expression. Treatment of human hepatoma cells with IL-28A activates this JAK-STAT pathway and induces expression of interferon-stimulated genes, establishing an antiviral state within the cell .
The signaling outcome includes upregulation of numerous antiviral proteins, including MxA, OAS, PKR, and ISG15. Additionally, IL-28A signaling leads to increased MHC class I expression, enhancing antigen presentation capabilities of affected cells .
Researchers have several options for detecting and quantifying IL-28A, each with distinct advantages:
For gene expression analysis, quantitative PCR (qPCR) offers a highly sensitive method for detecting IL-28A mRNA levels. Human IL-28A cDNA can be amplified using specific primers such as 5'-GGGTGACAGCCTCAGAGTG-3' and 5'-ATAGCGACTGGGTGGCAATA-3' . This approach is valuable for studying regulation of IL-28A expression in different cell types or in response to various stimuli.
Western blotting provides information about protein size and potential post-translational modifications. While less sensitive than ELISA for quantitative analysis, it can confirm specificity of detection in complex samples.
For optimal detection strategy in research, consider:
Combining protein detection (ELISA) with mRNA analysis (qPCR) for comprehensive assessment
Being aware that normal IL-28A levels in human plasma may be below detection limits of most assays
Validating antibody specificity to avoid cross-reactivity with related type III interferons
The choice of expression system significantly impacts the quality and functionality of recombinant IL-28A:
HEK293 mammalian cell expression represents the gold standard for producing research-grade IL-28A. This system yields human IL-28A with proper folding and native structure. The resulting protein is typically monomeric and non-glycosylated, with a molecular mass of 22 kDa in reduced form and 19-35 kDa in non-reduced form . Commercial recombinant human IL-28A produced in HEK293 cells shows excellent purity (>95%) and low endotoxin levels (<1 EU/μg) . This system allows for endotoxin-free and animal-component-free production, making it suitable for both functional studies and therapeutic research.
Mouse myeloma cell line NS0 expression provides another mammalian system used for IL-28A production. The expressed protein spans Val26-Val200 of the human IL-28A sequence (Accession # Q8IZJ0) . This system offers high-level expression of functional protein suitable for various applications including antibody production.
For laboratory-scale production, transient transfection of expression vectors (such as pEF6/V5-His-TOPO) into Huh7 or other mammalian cells can be used . A typical protocol involves transfecting cells using Lipofectin Reagent with 2 μg plasmid DNA per 1 × 10^5 cells in a 6-well plate format, followed by harvesting the expressed protein after 48 hours .
Designing robust experiments to investigate IL-28A signaling requires careful consideration of several factors:
Cell model selection is critical, as IL-28A receptor expression varies across cell types. Hepatocytes or hepatoma cell lines (e.g., Huh7) are excellent models due to their high receptor expression and well-characterized responses to IL-28A, including suppression of HCV replication . Intestinal epithelial cell lines also represent relevant models given IL-28A's role in intestinal antiviral defense .
For stimulation protocols, researchers should establish dose-response relationships. Commercial recombinant IL-28A typically shows activity at 0.01-0.06 ng/mL , but optimal concentrations should be determined for each experimental system. Both rapid (minutes to hours) and delayed (hours to days) responses should be monitored to capture the full spectrum of IL-28A effects.
JAK-STAT pathway activation can be assessed through Western blotting for phosphorylated STATs (particularly STAT1 and STAT2), immunofluorescence to track nuclear translocation, and reporter assays with ISRE-driven constructs. These approaches provide complementary information about signaling intensity and kinetics.
Downstream gene expression analysis using qPCR for known ISGs validates pathway activation, while RNA-seq offers comprehensive analysis of the IL-28A-induced transcriptome. Comparing IL-28A responses with type I interferon effects helps identify unique versus shared gene induction patterns.
Functional readouts should include relevant biological endpoints such as antiviral assays (using appropriate challenge viruses), flow cytometric analysis of MHC class I upregulation , and assessment of cell proliferation or survival effects.
IL-28A demonstrates significant antiviral activities across multiple viral infections:
In hepatitis C virus (HCV) infection, IL-28A has been shown to suppress viral RNA replication in cell culture models . The mechanism involves activation of the JAK-STAT pathway and induction of interferon-stimulated genes with antiviral properties. Genetic variations near the IL-28A locus strongly predict spontaneous HCV clearance and treatment response, highlighting its clinical relevance.
Recent research has revealed important connections between IL-28A and COVID-19 pathogenesis. A retrospective cohort study found that serum IL-28A/IFN-λ2 expression was significantly lower in patients with moderate to severe COVID-19 compared to those with mild to moderate disease . This suggests that IL-28A may play a protective role, with insufficient levels potentially contributing to disease progression. Notably, while IL-28A levels were decreased in severe COVID-19, inflammatory cytokines like IL-6 were significantly elevated, indicating an inverse relationship between protective antiviral responses and harmful inflammatory reactions .
The role of IL-28A in intestinal antiviral defense has been documented in multiple studies . The targeted expression of the IL-28A receptor on epithelial cells suggests an evolved function in protecting mucosal barriers from viral infection, complementing the broader activities of type I interferons.
IL-28A's immunomodulatory properties suggest several promising applications in cancer treatment:
The ability of IL-28A to induce HLA class I antigen expression in tumor cells represents a potentially valuable mechanism for cancer immunotherapy . By increasing MHC class I levels, IL-28A could enhance tumor cell recognition by cytotoxic T lymphocytes, potentially overcoming one mechanism of tumor immune evasion. This effect has been demonstrated in human hepatoma cells treated with IL-28A, which showed increased class I antigen expression as measured by flow cytometry .
Type III interferons, including IL-28A, have demonstrated direct anti-proliferative effects on certain cancer cell types. These growth-inhibitory properties could complement immune-mediated mechanisms for a multi-faceted anti-tumor approach.
The restricted receptor distribution of IL-28A offers a potential advantage for cancer therapy, as it may result in fewer systemic side effects compared to type I interferons . This more favorable toxicity profile could allow for higher or more sustained dosing regimens.
Combination therapy approaches represent a particularly promising direction. IL-28A might enhance the efficacy of checkpoint inhibitors by increasing tumor antigen presentation, improve responses to adoptive cell therapies through enhanced tumor recognition, or serve as an adjuvant for cancer vaccines.
Despite promising preclinical findings, several obstacles must be overcome for successful clinical translation:
Detection and quantification challenges present a significant hurdle. IL-28A concentration is typically very low in normal plasma and may fall below detection limits of many assays . Current ELISA methods have detection limits around 15 pg/mL, which may be insufficient for normal physiological levels . This complicates both research efforts and potential clinical monitoring of IL-28A-based therapies.
Production and formulation considerations include ensuring consistent, high-quality recombinant IL-28A manufacturing. While current production methods using HEK293 cells provide high purity (>95%) , scaling up for clinical applications may require additional optimization. Protein stability, formulation, and appropriate delivery methods must be carefully developed.
Pharmacokinetic and pharmacodynamic factors require thorough investigation. The limited natural concentration of IL-28A in circulation complicates understanding of its normal physiological roles. Determining optimal dosing regimens, tissue distribution, and elimination pathways will be essential for clinical development.
Identifying appropriate target populations represents another challenge. Genetic factors, including polymorphisms in the IL-28A gene or receptor, may influence response to IL-28A-based therapies. Developing reliable biomarkers to predict which patients might benefit most from such treatments will be critical for successful clinical application.
Several promising research directions may expand IL-28A's therapeutic applications:
The connection between IL-28A and COVID-19 severity opens new avenues for investigation. The finding that lower IL-28A levels correlate with more severe disease suggests potential therapeutic applications. Administration of recombinant IL-28A might help boost antiviral responses in early infection, while the targeted nature of IL-28A's effects on epithelial cells could provide antiviral benefits without exacerbating systemic inflammation.
Combination therapies leveraging IL-28A's unique properties offer significant promise. For viral hepatitis, combining IL-28A with direct-acting antivirals might enhance efficacy or reduce the emergence of viral resistance. In oncology, IL-28A could potentially sensitize tumors to immune checkpoint inhibitors by increasing antigen presentation.
Genetic associations between IL-28A polymorphisms and disease outcomes represent an important area for personalized medicine approaches. Further characterization of how genetic variations affect IL-28A expression or function may guide patient selection for IL-28A-based therapies across multiple disease contexts.
Advanced delivery systems targeting specific tissues could optimize IL-28A's therapeutic index. Site-specific delivery to affected tissues (such as the liver for hepatitis or the respiratory tract for COVID-19) might maximize efficacy while minimizing systemic exposure.
Several technological developments could significantly advance the field:
Improved detection methods with enhanced sensitivity are urgently needed. Current assays struggle to reliably detect physiological IL-28A levels in normal plasma . Next-generation immunoassays with lower detection limits and digital ELISA technologies could overcome this limitation, enabling better characterization of IL-28A biology.
Standardized activity assays would facilitate comparison across studies. Developing reporter cell systems specifically engineered to respond to IL-28A with minimal cross-reactivity to related cytokines would provide valuable tools for functional assessment.
Advanced genetic models, including conditional knockouts of IL-28A or its receptor components in specific tissues, would help delineate its tissue-specific functions. CRISPR/Cas9-based approaches allow for precise genetic manipulation to investigate IL-28A signaling components.
Single-cell analysis techniques applied to IL-28A research could reveal cell-specific responses that may be masked in bulk tissue studies. This approach would be particularly valuable for understanding the heterogeneity of responses in complex tissues like the liver or intestinal epithelium.
IL-28A is a protein that is expressed in human cells and has a molecular weight of approximately 21 kDa . The recombinant form of IL-28A is typically produced in human 293 cells . The protein is lyophilized from a filtered solution of PBS, pH 7.2, and can be stored at -20°C for long-term stability .
IL-28A exhibits several important biological activities:
IL-28A interacts with a heterodimeric receptor composed of the IL28-Rα and IL10-RI subunits . The expression of IL-28Rα is limited to a few cell types, including plasmacytoid dendritic cells and epithelial cells . This selective receptor distribution may result in fewer side effects compared to Type I Interferons .
Recombinant IL-28A is used in various research applications, including: