IRF3 antibodies are protein-specific reagents designed to bind IRF3, a 47–55 kDa protein involved in regulating interferon production during viral infections . They are available in polyclonal and monoclonal formats, with reactivity across human, mouse, rat, and other species . Key applications include:
Phosphorylation-Specific Antibodies: Antibodies targeting phosphorylated residues (e.g., Ser386, Ser396, Ser398) distinguish between inactive (monomeric) and activated (dimerized) IRF3 forms . For example:
Native-PAGE Analysis: IRF3 dimerization, a hallmark of activation, is detected using anti-IRF3-NES antibodies .
IRF3−/− Mice: Show impaired IFN-β production and increased susceptibility to viral infections (e.g., EMCV) .
Myeloid-Specific IRF3 Ablation: Reduces endotoxin-induced shock severity, highlighting IRF3's pro-inflammatory role .
NF-κB Inhibition: Activated IRF3 binds NF-κB–p65, preventing its nuclear translocation and curbing excessive inflammation during viral infections .
Post-Translational Modifications (PTMs): IRF3 undergoes phosphorylation, ubiquitination, and sumoylation at residues like T3, K70, and S396, which regulate its activity .
Knockout Validation: Antibodies like AF4019 (R&D Systems) show no cross-reactivity in IRF3-knockout HeLa cells .
Species Cross-Reactivity: Predictions for pig, bovine, and dog IRF3 are available based on sequence alignment scores (>80 = high confidence) .
Multiplex Staining: Alexa Fluor® 488-conjugated antibodies (e.g., IC4019G) enable simultaneous detection of IRF3 and other intracellular markers .
IRF3 is a key transcriptional regulator of type I interferon (IFN)-dependent immune responses that plays a critical role in the innate immune response against DNA and RNA viruses. It functions as a ~55-60 kDa constitutively expressed member of the IRF family . IRF3 regulates the transcription of type I IFN genes (IFN-alpha and IFN-beta) and IFN-stimulated genes (ISG) by binding to an interferon-stimulated response element (ISRE) in their promoters . It acts as a more potent activator of the IFN-beta gene than the IFN-alpha gene and plays a critical role in both early and late phases of IFN gene induction . Its significance extends to SARS-CoV-2 infection response, making it a crucial target for immunological and virological research .
IRF3 contains several functional domains that determine its activity and regulation:
The protein exists in an inactive form in the cytoplasm of uninfected cells. Following viral infection, double-stranded RNA (dsRNA), or toll-like receptor (TLR) signaling, IRF3 becomes phosphorylated by IKBKE and TBK1 kinases . This phosphorylation induces a conformational change, leading to dimerization and nuclear translocation, where it associates with CREB binding protein (CREBBP) to form dsRNA-activated factor 1 (DRAF1) . This complex then activates the transcription of type I IFN and ISG genes, orchestrating the antiviral response .
IRF3 antibodies have been validated for multiple research applications:
Selection should be based on the specific experimental requirements and the validated applications of each antibody clone .
When selecting an IRF3 antibody, consider these key factors:
Antibody Format: Choose between:
Target Species: Verify cross-reactivity with your experimental model:
Application Compatibility: Ensure validation for your technique:
Epitope Location: Select based on your research question:
Conjugation Requirements: Choose between:
Match the antibody specifications to your experimental design for optimal results .
Implementing proper controls is essential for reliable IRF3 antibody experiments:
Positive Controls:
Negative Controls:
Phosphorylation-State Controls:
Subcellular Localization Controls:
Application-Specific Controls:
Incorporating these controls ensures specificity, validates responses, and allows accurate interpretation of experimental results .
Differentiating between inactive and phosphorylated IRF3 requires specific techniques:
Phospho-specific antibodies: Though not mentioned in the search results, commercial phospho-specific antibodies that recognize Ser396, Ser386, and other phosphorylation sites are available and essential for direct detection of activated IRF3.
Mobility shift detection: Phosphorylated IRF3 migrates more slowly in SDS-PAGE, appearing as higher molecular weight bands compared to inactive IRF3 . Use lower percentage gels (7-8%) and longer running times to improve separation.
Nuclear/cytoplasmic fractionation: Since phosphorylated IRF3 translocates to the nucleus, comparing nuclear vs. cytoplasmic fractions can indirectly indicate activation state . Use subcellular fractionation followed by western blotting with IRF3 antibodies.
Dimerization analysis: Activated IRF3 forms dimers. Use non-denaturing gel electrophoresis or chemical crosslinking followed by SDS-PAGE to detect dimeric forms .
Immunofluorescence microscopy: Monitor IRF3 nuclear translocation as a proxy for activation. Inactive IRF3 appears predominantly cytoplasmic, while activated IRF3 shows nuclear accumulation .
Co-immunoprecipitation with CBP/p300: Active IRF3 associates with CREB binding protein to form DRAF1 complex. Co-IP experiments can detect this interaction using antibodies like D-3 that work in IP applications .
Combining these approaches provides comprehensive analysis of IRF3 activation status in experimental systems .
For successful IRF3 western blotting, follow these optimized protocols:
Sample Preparation:
Lyse cells in RIPA or NP-40 buffer with protease and phosphatase inhibitors
For detecting phosphorylated forms, add phosphatase inhibitors (sodium orthovanadate, sodium fluoride, β-glycerophosphate)
Use fresh samples when possible or snap-freeze and store at -80°C
Gel Electrophoresis:
Transfer and Blocking:
Antibody Incubation:
Detection:
Troubleshooting Tips:
Following these protocols will produce reliable western blot results for IRF3 detection .
Studying IRF3 nuclear translocation effectively requires these methodological approaches:
Immunofluorescence Microscopy:
Fix cells with 4% paraformaldehyde (10-15 minutes at room temperature)
Permeabilize with 0.1-0.5% Triton X-100 (5-10 minutes)
Block with 1-5% BSA or normal serum
Incubate with IRF3 antibodies validated for IF applications (e.g., D-3, IRF35I218)
Co-stain with DAPI for nuclear visualization
Use confocal microscopy for highest resolution of nuclear/cytoplasmic distinction
Subcellular Fractionation:
Separate nuclear and cytoplasmic fractions using commercial kits or established protocols
Analyze fractions by western blotting using antibodies like D-3, 3F10, or IRF35I218
Include markers for nuclear (e.g., lamin, histone) and cytoplasmic (e.g., GAPDH, tubulin) fractions
Quantify the nuclear/cytoplasmic ratio of IRF3
Time-Course Experiments:
Stimulate cells with appropriate triggers (viral infection, poly(I:C), LPS)
Collect samples at multiple timepoints (0, 30, 60, 90, 120, 240 minutes)
Process for IF or fractionation to capture the dynamics of translocation
Consider phospho-IRF3 antibodies to correlate phosphorylation with translocation
Live-Cell Imaging:
Create IRF3-fluorescent protein fusions (e.g., GFP-IRF3)
Perform time-lapse imaging after stimulation
Quantify nuclear accumulation over time
Flow Cytometry:
Image Analysis:
Use software to quantify nuclear/cytoplasmic fluorescence intensity ratios
Analyze at least 50-100 cells per condition for statistical significance
Plot translocation kinetics to determine rate and extent of nuclear accumulation
These approaches can be used individually or in combination for comprehensive analysis of IRF3 nuclear translocation dynamics .
IRF3 antibodies are valuable tools for investigating virus-host interactions:
Viral Evasion Mechanisms:
Type I Interferon Response Dynamics:
Monitor IRF3 nuclear translocation kinetics during infection using immunofluorescence
Correlate IRF3 activation with interferon production using paired samples
Study how different viral strains affect IRF3 activation patterns
SARS-CoV-2 Research:
Cell-Type Specific Responses:
Apoptosis Pathway Investigation:
Therapeutic Intervention Assessment:
Evaluate how antivirals or immunomodulators affect IRF3 activation during infection
Screen for compounds that enhance or inhibit IRF3 signaling as potential therapeutic approaches
Use phospho-IRF3 detection to measure pathway modulation
These applications demonstrate how IRF3 antibodies serve as critical reagents for understanding virus-host interactions and developing interventions for viral diseases .
Investigating IRF3-mediated gene regulation requires specialized techniques:
Chromatin Immunoprecipitation (ChIP):
Electrophoretic Mobility Shift Assay (EMSA):
Detect IRF3 binding to DNA probes containing ISRE motifs
Use IRF3 antibodies for supershift assays to confirm specificity
Compare nuclear extracts from stimulated vs. unstimulated cells
Reporter Gene Assays:
Co-Immunoprecipitation with Transcriptional Co-factors:
RNA Analysis:
Perform RT-qPCR to measure expression of IRF3 target genes after stimulation
Compare with IRF3 activation status detected by western blotting
Consider RNA-seq for genome-wide analysis of IRF3-dependent gene expression
Proteomics Approaches:
Use mass spectrometry to identify IRF3-interacting proteins after immunoprecipitation
Study post-translational modifications of IRF3 that affect its transcriptional activity
Compare protein interaction networks in different activation states
CRISPR/Cas9 Gene Editing:
Generate IRF3 knockout or mutant cell lines
Use IRF3 antibodies to confirm knockout efficiency
Compare transcriptional responses between wild-type and modified cells
These methodologies provide comprehensive insights into the mechanisms and targets of IRF3-mediated gene regulation in diverse biological contexts .
Multiple bands in IRF3 western blots can provide valuable biological information when properly interpreted:
Phosphorylation States:
Higher molecular weight bands (above the main 55-60 kDa band) often represent phosphorylated forms
Increasing intensity of these upper bands after stimulation indicates IRF3 activation
The pattern of multiple closely spaced higher bands may reflect different phosphorylation combinations on serine/threonine residues
Splice Variants:
Degradation Products:
Bands below the main IRF3 band might represent proteolytic fragments
Increasing lower bands during viral infection could indicate viral-induced degradation
Include protease inhibitors in lysis buffers to minimize artifactual degradation
Antibody-Specific Patterns:
Analytical Approach:
Use phosphatase treatment controls to identify phosphorylation-dependent bands
Compare patterns across different cell types and stimulation conditions
Consider using multiple antibodies recognizing different epitopes
For critical experiments, confirm with knockout/knockdown controls
Common Band Patterns:
Understanding these patterns enables accurate interpretation of IRF3 biology in experimental systems .
Quantifying IRF3 activation requires careful attention to several methodological considerations:
Western Blot Quantification:
Calculate the ratio of phosphorylated to total IRF3
Use densitometry software to measure band intensities
Normalize to appropriate loading controls (β-actin, GAPDH)
Present data as fold-change relative to unstimulated conditions
Consider the dynamic range limitations of western blotting
Nuclear Translocation Measurement:
In immunofluorescence: Calculate nuclear/cytoplasmic intensity ratios
In subcellular fractionation: Measure IRF3 band intensity in nuclear vs. cytoplasmic fractions
Account for background signal and normalize to fraction-specific markers
Analyze sufficient cell numbers (>50-100) for statistical significance
Functional Readouts:
Measure downstream gene expression (IFN-β, ISGs) as proxy for IRF3 activity
Correlate with direct IRF3 measurements for comprehensive assessment
Consider luciferase reporter assays with ISRE-containing promoters
Time-Course Considerations:
IRF3 activation is dynamic with distinct phases
Early activation (30-60 minutes): Initial phosphorylation
Peak activation (1-4 hours): Maximum nuclear translocation and target gene induction
Resolution phase: Dephosphorylation or degradation
Design sampling timepoints accordingly
Statistical Analysis:
Perform experiments in biological triplicates minimum
Use appropriate statistical tests (t-test, ANOVA) with post-hoc corrections
Report both statistical significance and effect size
Consider variability between cell types and stimulation conditions
Validation Approaches:
Verify with multiple detection methods (western blot, IF, reporter assays)
Include positive controls (known IRF3 activators like poly(I:C))
Use IRF3 inhibitors or knockdown as negative controls
Consider alternative measurement techniques for confirmation
These considerations ensure robust and reproducible quantification of IRF3 activation across different experimental paradigms .
IRF3 antibodies provide valuable tools for investigating SARS-CoV-2 and emerging viral pathogenesis:
Virus-Host Interaction Mechanisms:
Cell-Type Specific Responses:
Examine IRF3 activation in different respiratory cell types (epithelial cells, alveolar macrophages)
Use immunohistochemistry with IRF3 antibodies on COVID-19 patient samples
Compare with other respiratory viruses to identify unique signatures
Therapeutic Development:
Screen compounds that modulate IRF3 signaling as potential antivirals
Use IRF3 antibodies to monitor pathway activation in drug screening assays
Evaluate how repurposed drugs affect IRF3-dependent antiviral responses
Vaccine Research:
Study how vaccine adjuvants activate IRF3 signaling
Compare innate immune signatures between different vaccine platforms
Use IRF3 antibodies to characterize the initial immune response to vaccination
Long COVID Investigations:
Examine persistent alterations in IRF3 signaling in long COVID patients
Compare tissue samples from acute vs. long COVID cases using IHC
Investigate potential autoimmune aspects involving IRF3 pathway dysregulation
Emerging Pathogen Preparedness:
Develop standardized IRF3 activation assays for rapid evaluation of novel pathogens
Create biosensor cell lines with IRF3-dependent reporters for pathogen screening
Establish IRF3 antibody-based diagnostic approaches for viral infections
IRF3 antibodies thus serve as critical reagents for understanding the pathogenesis of SARS-CoV-2 and other emerging viruses, with direct implications for therapeutic and preventive strategies .
Beyond viral infections, IRF3 plays important roles in various physiological and pathological processes:
Autoimmune Diseases:
IRF3 contributes to inflammatory pathways in autoimmune conditions
Use IRF3 antibodies to assess activation status in patient samples
Compare phosphorylation patterns and nuclear localization in affected tissues
Investigate potential dysregulation in type I interferon-mediated pathologies
Cancer Biology:
IRF3 can influence apoptotic pathways and potentially affect tumor development
The apoptosis complex (TOMM70:HSP90AA1:IRF3:BAX) identified in search results suggests cancer relevance
Use immunohistochemistry with IRF3 antibodies to evaluate expression in tumor tissues
Investigate correlations between IRF3 activation status and cancer progression
Sterile Inflammation:
IRF3 responds to damage-associated molecular patterns (DAMPs) in non-infectious inflammation
Study IRF3 activation in models of ischemia-reperfusion injury or trauma
Use western blotting and immunofluorescence to track IRF3 in inflammatory conditions
Metabolic Disorders:
Emerging evidence suggests IRF3 involvement in metabolic regulation
Investigate IRF3 activation in adipose tissue, liver, and muscle in metabolic disease models
Use IRF3 antibodies to study tissue-specific activation patterns
Neurological Conditions:
IRF3 signaling occurs in microglia and affects neuroinflammation
Examine IRF3 activation in neurodegenerative disease models
Use immunofluorescence with IRF3 antibodies to study microglial activation
Research Methodologies:
Immunoprecipitation to identify novel IRF3 interaction partners in different disease contexts
ChIP-seq to map IRF3 binding sites in different cell types and pathological states
Tissue microarrays with IRF3 staining to screen multiple patient samples
These applications demonstrate how IRF3 antibodies facilitate investigation of diverse pathological processes beyond viral infections, potentially identifying new therapeutic targets and biomarkers .
Interferon Regulatory Factor-3 (IRF3) is a critical transcription factor involved in the innate immune response, particularly in the induction of antiviral immunity. It plays a pivotal role in the regulation of type I interferon (IFN) genes and other interferon-stimulated genes (ISGs). The mouse anti-human IRF3 antibody is a monoclonal antibody used in various research applications to study the expression and function of IRF3 in human cells.
IRF3 is a member of the interferon regulatory factor family, which consists of several transcription factors involved in the regulation of the immune response. IRF3 is constitutively expressed in most cells and remains inactive in the cytoplasm of uninfected cells. Upon viral infection or other stimuli, IRF3 undergoes phosphorylation, leading to its dimerization and translocation to the nucleus. In the nucleus, IRF3 binds to specific DNA sequences in the promoters of target genes, initiating the transcription of type I IFNs and ISGs .
IRF3 is essential for the induction of type I IFNs, which are crucial for the antiviral response. Type I IFNs, including IFN-α and IFN-β, are cytokines that play a key role in the defense against viral infections. They activate the expression of ISGs, which inhibit various stages of the viral life cycle. The activation of IRF3 is triggered by the recognition of viral nucleic acids by pattern recognition receptors (PRRs) such as Toll-like receptors (TLRs) and RIG-I-like receptors (RLRs) .
The mouse anti-human IRF3 antibody is a monoclonal antibody that specifically recognizes human IRF3. It is widely used in research to study the expression, localization, and function of IRF3 in human cells. This antibody is suitable for various applications, including Western blotting, immunohistochemistry, immunofluorescence, and flow cytometry .