OAS3 antibodies are polyclonal or monoclonal reagents designed to detect and quantify the OAS3 protein (UniProt ID: Q9Y6K5) in experimental settings. These antibodies are pivotal for investigating OAS3's roles in viral defense, tumor microenvironments (TME), and immune regulation .
OAS3 belongs to the oligoadenylate synthetase family, which synthesizes 2'-5'-linked oligoadenylates (2-5A) to activate RNase L, an enzyme that degrades viral and cellular RNA during infections . Key features include:
Antiviral Activity: OAS3 binds dsRNA with higher affinity than OAS1/OAS2, making it essential for RNase L activation against viruses like SARS-CoV-2 .
Cancer Relevance: OAS3 overexpression correlates with poor prognosis in pancreatic, lung, and renal cancers by promoting immunosuppressive TMEs .
CRISPR-KO studies show OAS3 is indispensable for RNase L activation, unlike OAS1/OAS2 .
OAS3 synthesizes long 2-5A chains (>20 units) necessary for sustained RNase L activity .
Tumor Progression: High OAS3 expression in tumors associates with immunosuppressive M2d macrophages, T-cell exhaustion, and reduced CD8+ T-cell infiltration .
Therapeutic Target: Knockdown of OAS3 in pancreatic cancer models restores CD8+ T-cell activity and synergizes with anti-PD-L1 therapy .
Pan-cancer analyses reveal OAS3 overexpression in 29/33 TCGA cancer types, linked to advanced staging and metastasis .
Cross-Reactivity Note: Some OAS3 antibodies may weakly cross-react with OAS1 due to structural homology .
OAS3 is an interferon-induced, dsRNA-activated antiviral enzyme that plays a critical role in cellular innate antiviral responses. It belongs to the oligoadenylate synthetase family, which includes OAS1, OAS2, OAS3, and OASL. OAS3 functions by synthesizing 2'-5'-oligoadenylates (2-5A) from ATP in the presence of double-stranded RNA, which then bind to and activate ribonuclease L (RNase L). This activation leads to degradation of both cellular and viral RNA, inhibiting protein synthesis and terminating viral replication. Beyond its classical RNase L-dependent pathway, OAS3 can also mediate antiviral effects through alternative mechanisms and has demonstrated activity against various viruses including Chikungunya virus, Dengue virus, Sindbis virus, and Semliki forest virus .
OAS3 antibodies are versatile tools that can be employed in multiple experimental techniques:
Application | Recommended Dilution | Sample Types |
---|---|---|
Western Blot (WB) | 1:1000-1:4000 | Cell lysates, tissue extracts |
Immunohistochemistry (IHC) | 1:20-1:200 | FFPE tissues |
Immunofluorescence (IF) | 1:200-1:800 | Fixed cells, tissue sections |
Immunoprecipitation (IP) | Varies by antibody | Cell lysates |
ELISA | Varies by antibody | Purified protein, serum |
Researchers should always validate antibodies in their specific experimental system, as performance can vary between different antibody clones and across applications .
For robust OAS3 antibody experiments, incorporate the following controls:
Positive control: Use samples known to express OAS3, such as interferon-stimulated cells or tissues (A375 cells, HeLa cells, and human placenta tissue have been validated) .
Negative control: Include samples where OAS3 expression is minimal or absent.
Isotype control: Use an irrelevant antibody of the same isotype to assess non-specific binding.
Blocking peptide control: Pre-incubate the antibody with its specific immunogen to confirm specificity.
siRNA knockdown: Reduce OAS3 expression in cells to validate antibody specificity.
For western blotting, the predicted band size for human OAS3 is approximately 121 kDa . Discrepancies in band size may indicate post-translational modifications or alternative splicing variants.
OAS3 has emerged as a significant co-immune biomarker associated with tumor progression and immune infiltration. Research has demonstrated that OAS3 is aberrantly expressed across almost all TCGA cancer types and subtypes, with expression levels correlating with tumor staging, metastasis, and prognostic deterioration in various cancers .
The relationship between OAS3 and tumor immune microenvironment (TME) is particularly noteworthy. OAS3 expression positively correlates with the infiltration of immunosuppressive cells, suggesting a potential role in immune evasion mechanisms. When investigating this relationship, researchers should consider:
Analyzing OAS3 expression alongside ImmuneScore, StromalScore, and ESTIMATEScore using the "ESTIMATE" R package
Utilizing platforms like TIMER2, Xcell, CIBERSORT, and ImmuCellAI to correlate OAS3 expression with specific immune cell infiltration
Examining the association between OAS3 expression and immune checkpoint-related genes
Investigating correlations between OAS3 expression and neoantigen load
This multi-dimensional analysis can provide insights into OAS3's role in cancer immunobiology and potential therapeutic implications.
Investigating OAS3's antiviral functions requires specialized approaches:
Viral Infection Models: Establish cell culture systems infected with OAS3-sensitive viruses (CHIKV, Dengue, SINV, SFV) to assess the impact of OAS3 on viral replication.
Pathway Analysis: Differentiate between RNase L-dependent and RNase L-independent antiviral mechanisms by:
Measuring 2-5A synthesis using HPLC or specialized assays
Assessing RNase L activation and RNA degradation patterns
Using RNase L knockout cells to identify alternative pathways
Structure-Function Studies: Utilize antibodies targeting specific domains of OAS3 to elucidate the functional significance of its three OAS1-like domains.
Metal Ion Dependency: Investigate how different metal ions (copper, iron, zinc, manganese) affect OAS3 enzymatic activity through in vitro enzymatic assays .
These approaches can provide comprehensive insights into OAS3's diverse antiviral mechanisms and potential therapeutic applications.
OAS3 functions within a complex network of interferon-stimulated genes (ISGs). To understand its position in this network:
Temporal Expression Analysis: Monitor OAS3 induction kinetics following interferon stimulation using qPCR and western blotting with validated antibodies.
Interactome Mapping: Employ co-immunoprecipitation with OAS3 antibodies followed by mass spectrometry to identify protein-protein interactions.
Signaling Pathway Integration: Use phospho-specific antibodies to examine how OAS3 influences or is influenced by key signaling nodes in interferon pathways.
Transcriptomic Analysis: Perform RNA-seq in OAS3-overexpressing or OAS3-knockout models to identify downstream effectors and feedback mechanisms.
This systems biology approach can reveal how OAS3 coordinates with other ISGs to establish an antiviral state and influence cancer progression .
For successful OAS3 detection by Western blot:
Sample Preparation:
Gel Electrophoresis:
Transfer and Blocking:
Perform wet transfer for large proteins like OAS3 (>100 kDa)
Block with 5% non-fat milk or BSA in TBST for 1 hour at room temperature
Antibody Incubation:
Primary antibody: Use at 1:1000-1:4000 dilution overnight at 4°C
Secondary antibody: HRP-conjugated at 1:5000-1:10000 for 1 hour at room temperature
Detection:
For optimal OAS3 detection in tissue sections:
Tissue Processing:
Fixation: 10% neutral buffered formalin for 24-48 hours
Paraffin embedding following standard protocols
Section thickness: 4-5 μm
Antigen Retrieval:
Antibody Selection and Dilution:
Detection Systems:
DAB (3,3'-diaminobenzidine) for brightfield microscopy
Fluorophore-conjugated secondary antibodies for fluorescence imaging
Validated Positive Control Tissues:
Researchers should optimize conditions based on their specific tissue type and fixation method.
Antibody batch variation can significantly impact experimental reproducibility. To address this:
Validation Strategy:
Test each new lot against a reference sample with known OAS3 expression
Compare titration curves between lots to establish equivalent working dilutions
Document lot numbers and validation data for publication
Internal Standards:
Maintain aliquots of a standard positive control sample for lot comparison
Use recombinant OAS3 protein as a standardized control
Multi-Antibody Approach:
When possible, confirm key findings with antibodies from different vendors or clones
Consider using antibodies targeting different epitopes of OAS3
Detailed Record-Keeping:
Maintain a database of antibody performance across different experiments
Note any variations in detection sensitivity or specificity between lots
This systematic approach ensures experimental consistency despite inevitable batch variations.
Discrepancies in OAS3 detection can arise from multiple factors:
Epitope Differences:
Isoform Specificity:
OAS3 may exist in multiple isoforms or splice variants
Some antibodies may detect all isoforms while others are isoform-specific
Cross-Reactivity:
Antibodies may cross-react with other OAS family members (OAS1, OAS2, OASL) due to sequence homology
Secondary cross-reactivity with unrelated proteins with similar epitopes
Technical Variables:
Different fixation methods can affect epitope exposure
Sample preparation techniques may preserve or destroy certain epitopes
When discrepancies occur, researchers should validate findings with multiple detection methods and consider using genetic approaches (siRNA, CRISPR) to confirm specificity.
Background signal in OAS3 immunofluorescence can be minimized through these approaches:
Antibody Optimization:
Blocking Enhancements:
Use image-specific blocking agents (normal serum from secondary antibody species)
Add 0.1-0.3% Triton X-100 for better antibody penetration
Include 0.1-1% BSA to reduce non-specific binding
Fixation Considerations:
Compare paraformaldehyde, methanol, and acetone fixation to determine optimal epitope preservation
Adjust fixation time to minimize autofluorescence while preserving antigenicity
Controls and Countermeasures:
Include a negative control without primary antibody
Use cells with OAS3 knockdown as a specificity control
Apply Sudan Black B (0.1-0.3%) to reduce autofluorescence
Consider spectral unmixing for multi-color experiments
These strategies have proven effective for OAS3 detection in A549 cells and other validated cell lines .
OAS3 expression is highly regulated and can be influenced by:
Cytokine Stimulation:
Type I interferons (IFN-α, IFN-β) strongly induce OAS3 expression
Type II interferons (IFN-γ) may have cell type-specific effects
Pro-inflammatory cytokines can synergize with interferons
Viral Infection:
Various RNA and DNA viruses trigger OAS3 upregulation
Virus-encoded antagonists may suppress OAS3 induction in certain infections
Cell Type Differences:
Technical Considerations:
Cell culture conditions (confluency, passage number, serum factors)
RNA/protein extraction methods can affect yield and quality
Time-dependent expression patterns after stimulation
Researchers should standardize these variables and include appropriate time-course analyses when studying OAS3 regulation.
Contextualizing OAS3 within immune networks requires:
Multi-omics Integration:
Correlate OAS3 protein levels (antibody-based detection) with transcript data (RNA-seq)
Connect OAS3 expression with upstream regulators and downstream effectors
Pathway Analysis Tools:
Functional Validation:
Confirm computational predictions with knockout/knockdown experiments
Use OAS3 antibodies to monitor protein changes in response to pathway perturbations
Therapeutic Relevance:
This integrated approach provides a comprehensive understanding of OAS3's role in immune regulation and potential therapeutic applications.
OAS3 antibodies have enabled significant discoveries about antiviral immunity:
Alternative Antiviral Pathways: Beyond the classical RNase L pathway, OAS3 has been found to mediate antiviral effects through RNase L-independent mechanisms, expanding our understanding of innate immune diversity .
Virus-Specific Responses: OAS3 demonstrates varying efficacy against different viruses (CHIKV, Dengue, SINV, SFV), suggesting virus-specific interaction mechanisms that can be further explored using epitope-specific antibodies .
Structural Insights: Antibodies targeting specific domains of OAS3 have helped elucidate how its three OAS1-like domains contribute to its unique antiviral properties compared to other OAS family members.
Regulatory Networks: Immunoprecipitation studies with OAS3 antibodies have revealed previously unknown protein interactions that regulate OAS3 activity and localization.
These advances highlight the critical role of high-quality antibodies in deciphering complex antiviral mechanisms.
OAS3's emerging role in cancer biology opens new research avenues:
Biomarker Potential: OAS3 expression correlates with tumor staging, metastasis, and prognostic deterioration across multiple cancer types, positioning it as a potential prognostic biomarker .
Immune Checkpoint Interactions: OAS3 expression shows significant correlation with immune checkpoint genes, suggesting potential involvement in immunotherapy response mechanisms .
Therapeutic Response Prediction: OAS3 expression patterns may help predict sensitivity to various chemotherapeutics and immunotherapies, as suggested by correlations with IC50 values in drug databases .
Tumor Microenvironment Modulation: OAS3's association with immunosuppressive cell infiltration suggests it may influence the tumor immune microenvironment, affecting treatment outcomes .
Researchers investigating these aspects should employ validated antibodies in combination with genomic and transcriptomic approaches to comprehensively characterize OAS3's role in cancer immunity.