OAS1 (2'-5'-oligoadenylate synthase 1) is an interferon-inducible enzyme critical for antiviral responses and immune regulation in humans . It detects viral double-stranded RNA, activates RNase L to degrade viral RNA, and modulates cell proliferation, apoptosis, and immune checkpoint pathways . In contrast, OASA1 (O-acetylserine(thiol)lyase A1) refers to a plant-specific enzyme involved in cysteine biosynthesis in Arabidopsis thaliana, with no direct homology to human OAS1 . This article focuses on OAS1 antibodies—reagents essential for studying OAS1's roles in cancer, infectious diseases, and therapeutic development.
Commercial OAS1 antibodies are designed for specificity across applications, including Western blot (WB), immunohistochemistry (IHC), and ELISA . Key features include:
| Antibody | Host/Isotype | Target Species | Applications | Molecular Weight |
|---|---|---|---|---|
| Goat anti-OAS1 (Bio-Rad) | Polyclonal IgG | Human | WB, ELISA | 46 kDa (predicted) |
| Rabbit anti-OAS1 (Proteintech) | Recombinant IgG | Human, Mouse | WB, IHC, Indirect ELISA | 46 kDa (predicted) |
| Rabbit anti-OAS1 (Thermo Fisher) | Polyclonal IgG | Human | WB, Functional assays | 46 kDa (observed: 38 kDa) |
These antibodies recognize isoforms (e.g., p42, p46) and are validated for specificity using siRNA silencing or recombinant protein assays . For example, Proteintech’s antibody detects OAS1 at 38 kDa due to post-translational modifications .
Pan-Cancer Analysis: High OAS1 expression correlates with poor prognosis in lung adenocarcinoma (LUAD) and prostate cancer (PRAD), linked to cell proliferation and migration . Silencing OAS1 with siRNA reduces tumor growth and enhances chemotherapy-induced apoptosis .
Immune Modulation: OAS1 is associated with tumor mutational burden (TMB), immune checkpoint genes (e.g., PD-L1), and immune subtypes in 12 cancers, suggesting its role in tumor microenvironment regulation .
Antiviral Activity: OAS1 detects SARS-CoV-2 RNA, and higher pre-infection OAS1 levels reduce COVID-19 severity . A Neanderthal-derived OAS1 isoform (p46) shows enhanced antiviral activity compared to the ancestral p42 isoform .
Therapeutic Targeting: Antibodies enable isoform-specific detection, critical for studies on OAS1’s genetic variants (e.g., rs10774671, rs1131454) that influence COVID-19 outcomes .
OAS1 (2'-5'-oligoadenylate synthase 1), also known as E18/E16 and p46/p42 OAS, belongs to the OAS family of genes induced by interferons (IFN) and viruses. It plays a crucial role in regulating signaling pathways during viral infection . OAS1's antiviral function is primarily attributed to its ability to detect short double-stranded RNAs (dsRNAs) .
OAS1 shares structural and functional homology with cyclic GMP-AMP (cGAMP) synthetase (cGAS), and both proteins initiate antiviral immune responses by recognizing cytoplasmic foreign nucleic acids — OAS1 detects viral double-stranded RNA (dsRNA) while cGAS recognizes viral double-stranded DNA (dsDNA) . Upon activation, OAS1 produces 2'-5'-oligoadenylate (2-5A) second messengers that activate RNase L, which subsequently degrades viral and cellular RNA to interfere with viral propagation .
Beyond viral infections, OAS1 has been implicated in autoimmune diseases and cancer processes. Research indicates it can affect cell migration, and high OAS1 mRNA expression has been linked to worse prognosis in breast cancer patients .
When selecting an OAS1 antibody for research applications, consider:
Antibody specificity: Verify the antibody recognizes your target OAS1 isoform (p46/p42) and doesn't cross-react with other OAS family members. Anti-OAS1 antibodies should be validated against specific OAS1 protein variants .
Host species and format: For OAS1, goat-derived polyclonal antibodies are commonly available, often as purified IgG in liquid form . Consider experimental compatibility with your secondary detection system.
Validated applications: Confirm the antibody has been validated for your specific application (Western blot, immunohistochemistry, etc.).
Target species reactivity: Ensure the antibody recognizes OAS1 from your experimental species (human, mouse, etc.) .
Buffer and preservative compatibility: Standard preparations typically use TRIS-buffered saline with 0.02% sodium azide as a preservative .
OAS1 gain-of-function variants have been identified in patients with polymorphic autoinflammatory immunodeficiency characterized by recurrent fever, dermatitis, inflammatory bowel disease, pulmonary alveolar proteinosis, and hypogammaglobulinemia . To study these variants:
Comparative expression analysis: Use OAS1 antibodies to compare protein expression levels between normal and patient-derived samples through Western blotting or immunohistochemistry.
Functional studies in cell models: Apply OAS1 antibodies in interferon-stimulated cellular systems to observe:
Mechanistic investigations: OAS1 antibodies can help assess how gain-of-function variants impact:
Therapeutic response monitoring: Track changes in OAS1 variant protein expression and downstream effects during experimental treatments (e.g., RNase L-inhibition with curcumin or allogeneic hematopoietic cell transplantation) .
To investigate OAS1's role in cancer progression using OAS1 antibodies:
Expression correlation studies:
Migration and invasion assays:
Apply OAS1 antibodies in immunofluorescence studies to track protein localization during cell migration
Use neutralizing OAS1 antibodies to block function in migration assays to determine causative relationships
Signaling pathway analysis:
Employ OAS1 antibodies in co-immunoprecipitation experiments to identify cancer-relevant binding partners
Perform phospho-specific Western blots to determine activation states in cancer vs. normal cells
In vivo tumor models:
Use OAS1 antibodies for immunohistochemical analysis of tumor sections
Correlate OAS1 expression with markers of tumor aggressiveness and metastatic potential
Multi-step validation process for OAS1 antibodies:
Initial specificity testing:
Western blot using recombinant OAS1 protein alongside cell lysates
Include positive controls (interferon-stimulated cells) and negative controls
Test for cross-reactivity with other OAS family members
Application-specific optimization:
Titration experiments:
Knockout/knockdown validation:
Test antibody in OAS1 knockout or siRNA knockdown systems
Compare staining patterns in wild-type vs. depleted samples to confirm specificity
Mass spectrometry (MS)-based proteomics can complement and enhance OAS1 antibody research through:
Database expansion for antibody validation:
Confirmation of OAS1 antibody specificity:
Detection of post-translational modifications:
MS can reveal modifications on OAS1 not detectable by standard antibody methods
Combine antibody enrichment with MS analysis for comprehensive PTM mapping
Quantitative comparison across sample types:
Proportions of antibody peptides detected across sample types:
| Sample Type | UniProt Peptides | OAS Peptides |
|---|---|---|
| Blood Plasma | 5-15% | 1-11% |
| Depleted Plasma | 2-7% | 0.1-2.5% |
| Brain Cortex | ~0.8% | ~0.1% |
This data confirms the validity of newly detected peptides as genuine antibody components .
When encountering non-specific binding with OAS1 antibodies:
Optimize blocking conditions:
Test different blocking agents (BSA, normal serum, commercial blockers)
Extend blocking time to reduce background
Consider adding 0.1-0.3% Triton X-100 to blocking buffer to reduce hydrophobic interactions
Antibody dilution optimization:
Pre-absorption controls:
Incubate antibody with recombinant OAS1 protein prior to application
Compare pre-absorbed antibody with standard antibody to identify non-specific signals
Cross-reactivity testing:
Test antibody on samples known to lack OAS1 expression
Verify specificity against other OAS family members
Confirm results with a second OAS1 antibody targeting a different epitope
Buffer optimization:
Essential controls for OAS1 antibody experiments:
Positive controls:
Interferon-stimulated cells with confirmed OAS1 upregulation
Recombinant OAS1 protein for Western blot ladder verification
Tissues known to express OAS1 (e.g., immune cells after viral stimulation)
Negative controls:
Specificity controls:
Competitive binding with excess recombinant OAS1
Comparison with alternative OAS1 antibody clones
Verification across multiple detection methods (Western blot, IHC, flow cytometry)
Technical controls:
Loading controls for Western blots (housekeeping proteins)
Tissue processing controls for IHC (known antibodies with consistent performance)
Fluorophore controls for confocal microscopy (spectral overlap assessment)
OAS1 antibodies are advancing viral immunity research, particularly for SARS-CoV-2, through:
Antiviral mechanism elucidation:
Proteomics integration:
The Observed Antibody Space (OAS) database contains 30 million heavy antibody sequences from 146 SARS-CoV-2 patients, enabling expansion of proteomics databases
Database searches using this expanded repertoire allow detection of previously unidentified antibody peptides in SARS-CoV-2 plasma samples
The newly discovered antibody peptides show diagnostic potential, distinguishing diseased from healthy samples
Host response characterization:
Monitoring OAS1 expression in different patient populations
Correlating OAS1 levels with disease severity and outcomes
Investigating potential genetic variants affecting OAS1 function during infection
Therapeutic development implications:
Using OAS1 antibodies to screen compounds that modulate its activity
Identifying patients who might benefit from RNase L pathway modulation
Studying OAS1-targeting therapeutic antibodies for antiviral applications
Recent advances in antibody engineering relevant to OAS1 research include:
Phage display selection techniques:
Computational modeling for specificity design:
Validation through comparative testing:
Application to OAS1 research: