IRF3 Antibody

Shipped with Ice Packs
In Stock

Description

Applications in Research

The IRF3 antibody has been employed in diverse experimental contexts:

  • Western Blotting: Detects phosphorylated IRF3 (e.g., at Serine 385/386) to study activation during viral infections or inflammation .

  • Immunohistochemistry: Localizes IRF3 in tissue sections, aiding in the analysis of its nuclear translocation post-activation .

  • Immunofluorescence: Visualizes IRF3 dynamics in live or fixed cells, such as its redistribution during antiviral responses .

Viral Infection and Antiviral Defense

IRF3 is central to type-I interferon (IFN-α/β) production following viral detection. Studies using the IRF3 antibody have revealed:

  • Activation Mechanism: Phosphorylation of IRF3 at conserved motifs (e.g., Serine 173/175) by TBK1 kinase is critical for nuclear translocation and IFN gene transcription .

  • Apoptotic Pathway: A non-transcriptional role of IRF3 involves polyubiquitination-mediated apoptosis of infected cells .

Inflammation Modulation

IRF3 exhibits dual roles in immune regulation:

  • Anti-inflammatory: Binds NF-κB–p65 to prevent nuclear translocation, mitigating excessive inflammation .

  • Pro-inflammatory: Drives IFN production, which can exacerbate conditions like sepsis or autoimmune diseases .

Cardiovascular and Oncological Insights

  • Myocardial Infarction: IRF3 knockout models show reduced infarct size, suggesting its role in ischemic damage .

  • Tumor Growth: IRF3-deficient mice exhibit impaired antiviral responses but reduced tumor growth, highlighting complex immune-tumor interactions .

Data Table: Key Research Insights

Study FocusKey FindingsCitation
IRF3-NF-κB InteractionIRF3 sequesters NF-κB–p65 in cytoplasm, inhibiting inflammation
Viral Apoptosis PathwayIRF3 induces apoptosis via polyubiquitination, limiting viral spread
Cardiovascular RoleIRF3 deletion protects against myocardial infarction
Tumor DynamicsIRF3 deficiency correlates with reduced tumor growth in vivo

Product Specs

Buffer
Phosphate Buffered Saline (PBS) with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze / thaw cycles.
Lead Time
Typically, we can dispatch the products within 1-3 business days after receiving your orders. Delivery times may vary depending on the purchasing method or location. For specific delivery times, please consult your local distributors.
Synonyms
IIAE7 antibody; Interferon regulatory factor 3 antibody; IRF 3 antibody; IRF-3 antibody; IRF3 antibody; IRF3_HUMAN antibody; MGC94729 antibody
Target Names
Uniprot No.

Target Background

Function
IRF3 (Interferon Regulatory Factor 3) is a key transcriptional regulator of type I interferon (IFN)-dependent immune responses, playing a critical role in the innate immune response against DNA and RNA viruses. It 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. IRF3 acts as a more potent activator of the IFN-beta (IFNB) gene compared to the IFN-alpha (IFNA) gene and plays a critical role in both the early and late phases of the IFNA/B gene induction. In uninfected cells, IRF3 exists in an inactive form in the cytoplasm. Upon viral infection, double-stranded RNA (dsRNA), or toll-like receptor (TLR) signaling, IRF3 is phosphorylated by IKBKE and TBK1 kinases. This phosphorylation induces a conformational change, leading to dimerization and nuclear localization of IRF3. In the nucleus, IRF3 associates with CREB binding protein (CREBBP) to form dsRNA-activated factor 1 (DRAF1), a complex that activates the transcription of type I IFN and ISG genes. IRF3 can activate distinct gene expression programs in macrophages and can induce significant apoptosis in primary macrophages. In response to Sendai virus infection, IRF3 is recruited by TOMM70:HSP90AA1 to the mitochondrion and forms an apoptosis complex TOMM70:HSP90AA1:IRF3:BAX, inducing apoptosis. IRF3 is a key transcription factor regulating the IFN response during SARS-CoV-2 infection.
Gene References Into Functions
  1. Reduced GATA-1 could be responsible for the upregulation of IRF-3 in lung adenocarcinoma cells through binding with a specific domain of IRF-3 promoter. PMID: 28566697
  2. The STING-IRF3 pathway promotes hepatocyte injury and dysfunction by inducing inflammation and apoptosis, and by disturbing glucose and lipid metabolism. PMID: 29106945
  3. beta-catenin interacts with IRF3 and blocks its nuclear translocation. PMID: 30004146
  4. Upregulation of endogenous SAMHD1 expression is attributed to the phosphorylation and nuclear translocation of IRF3. PMID: 27411355
  5. This research found that differences in type 1 interferon production by T1 and T3 reoviruses correlate with differential IRF3 activation. PMID: 29437975
  6. Proteins 8b and 8ab of severe acute respiratory syndrome coronavirus physically interact with IRF3 and induce degradation of IRF3 in a ubiquitin-proteasome-dependent manner. PMID: 29294448
  7. These findings highlight the importance of IRF3 and type-I IFNs signaling for the pro-apoptotic effects induced by RA and synthetic dsRNA in breast cancer cells. PMID: 28409399
  8. This study investigates the role of MITA (Mediator of IRF3 Activation), a regulator of innate immunity, in the regulation of autophagy and its implication in cell death of breast cancer cells. It reports that MITA inhibits the fusion of autophagosome with lysosome as evident from different autophagy flux assays. PMID: 28366813
  9. New research suggests that altering a subset of extracellular matrix factors, including interferon regulatory factor (IRF)3 and casein kinase (CK)2, may decrease the migratory potential of aggressive tumors. PMID: 28774478
  10. IRF-3 gene polymorphisms were associated with the susceptibility and prognosis of CLL, and can be used as an auxiliary index for clinical detection of CLL. PMID: 27348780
  11. Clarithromycin acts as a crucial modulator of the innate immune response, particularly IFN production, by modulating IRF-3 dimerization and subsequent translocation to the nucleus of airway epithelial cells. PMID: 27468646
  12. c-Cbl negatively regulates IFN-beta signaling and cellular antiviral response by promoting IRF3 ubiquitination and degradation. PMID: 27503123
  13. This research describes an unappreciated role for EAP30 in IRF3-dependent innate antiviral response in the nucleus. PMID: 29084253
  14. IRF-3 is an important regulator of ORMDL3 induction following RSV infection by binding directly to the promoter of ORMDL3. PMID: 28336364
  15. These findings suggest that ATG5-ATG12 positively regulate anti-viral NF-kappaB and IRF3 signaling during FMDV infection, thereby limiting FMDV proliferation. FMDV has evolved mechanisms to counteract the antiviral function of ATG5-ATG12, via degradation of them by viral protein 3C(pro). PMID: 28102839
  16. NEMO-IKKbeta are essential for IRF3 and NF-kappaB activation in the cGAS-STING pathway. PMID: 28939760
  17. This research suggests that HNSs, an antagonist of host innate immunity, interacts with TBK1 and thereby hinders the association of TBK1 with its substrate IRF3, thus blocking IRF3 activation and transcriptional induction of the cellular antiviral responses. PMID: 28848048
  18. IRF3 is a major transcriptional regulator of adipose inflammation and is involved in maintaining systemic glucose and energy homeostasis. PMID: 27400129
  19. This study shows that IRF-3-mediated apoptosis of virus-infected cells could be an effective antiviral mechanism, without expression of the interferon-stimulated genes. PMID: 27178468
  20. Data in this study show that cFLIPL inhibits IFN regulatory factor 3 (IRF3), a transcription factor central for IFN-beta and IFN-stimulated gene expression. PMID: 27342840
  21. 1,8-cineole potentiates the antiviral activity of IRF3 in addition to its inhibitory effect on proinflammatory NF-kappaB signaling in an ex vivo model of rhinosinusitis. PMID: 27129189
  22. TBK1 complexes required for the phosphorylation of IRF3 and the production of interferon-beta have been identified. PMID: 28159912
  23. RIG-I-like receptor-induced IRF3 mediated pathway of apoptosis (RIPA): a new antiviral pathway. PMID: 27815826
  24. cGAs recognizes bacterial/viral DNA, and is a strong activator of STING that can further activate IRF3 and subsequent type I interferon production. (Review) PMID: 27696330
  25. IRF3 overexpression in Acute myeloid leukemia (AML) promotes cell growth and survival, and miR-155 is involved, indicating that IRF3 may be a potential new biomarker and therapeutic target for AML. PMID: 27530922
  26. Down-regulation of IRF3 inhibited the proliferation and extracellular matrix expression in keloid fibroblasts. PMID: 28192879
  27. Rotavirus NSP1 (nonstructural protein 1) employs a pLxIS motif to target IRF-3 for degradation, but phosphorylation of NSP1 is not required for its activity. These results suggest a concerted mechanism for the recruitment and activation of IRF-3 that can be subverted by viral proteins to evade innate immune responses. PMID: 27302953
  28. Highly pathogenic Porcine reproductive and respiratory syndrome virus modulates Interferon-beta expression mainly through attenuating IRF-3 phosphorylation. PMID: 27314873
  29. Data suggest that molecular chaperone GRP78 contributes to toll-like receptor-3 (TLR3)-mediated, interferon regulatory factor 3 protein (IRF3)-dependent innate immune response to hepatitis C virus (HCV) in hepatocytes. PMID: 27129228
  30. Findings suggest a common and conserved mechanism through which highly pathogenic MERS-CoV and SARS-CoV harness their M proteins to suppress type I IFN expression at the level of TBK1-dependent phosphorylation and activation of IRF3, resulting in evasion of the host innate antiviral response. PMID: 27094905
  31. Observations suggest IRF3 may function as a novel regulator to modulate TGF-beta1-induced LX-2 proliferation, at least in part, via AKT signaling pathway. PMID: 26611114
  32. FAF1 plays a novel role in negatively regulating virus-induced IFN-beta production and the antiviral response by inhibiting the translocation of active, phosphorylated IRF3 from the cytosol to the nucleus. PMID: 26811330
  33. The LxxLL motifs of IRF3 bind within the hydrophobic pocket of E6, precluding Ser-patch phosphorylation, necessary for IRF3 activation and interferon induction. PMID: 26289783
  34. The suppression of type I IFN production by HTLV-1 Tax through interaction with and inhibition of TBK1 kinase that phosphorylates IRF3. PMID: 26819312
  35. This research found that Ca(2+) signaling associated with membrane perturbation and recognition of incoming viral genomes by cytosolic nucleic acid receptors are required to activate IRF3 in response to Sendai virus and human cytomegalovirus. PMID: 26719279
  36. This study identifies crosstalk between PTEN and IRF3 in tumor suppression and innate immunity. PMID: 26692175
  37. Viral infection induced DAPK1-IRF7 and DAPK1-IRF3 interactions and overexpression of DAPK1 enhanced virus-induced activation of the interferon-stimulated response element (ISRE) and IFN-beta promoters and the expression of the IFNB1 gene. PMID: 24531619
  38. TEL-AML1 fusion protein blocks B-cell differentiation and downregulates the IRF3-IFNalpha/beta pathway by modulating expression and phosphorylation of IRF3 in human primary hematopoietic precursor cells. PMID: 25893288
  39. The expression levels of IRF3 were not different between CHB patients and healthy controls. PMID: 26058929
  40. Results revealed a new paradigm in which the antiviral host factor, IRF3, plays a cell-intrinsic pro-parasitic role. PMID: 25811886
  41. The results of this study suggested that IRF3 is likely a risk gene for schizophrenia, at least in Caucasians. PMID: 25843157
  42. Induction of miR-576-3p by IRF3 triggers a feedback mechanism to reduce interferon expression and set an antiviral response threshold to likely avoid excessive inflammation. PMID: 25232931
  43. The present study indicated that HSPD1 interacted with IRF3 and it contributed to the induction of IFN-beta. PMID: 25506707
  44. The identification of IRF3 deficiency in HSE provides the first description of a defect in an IFN-regulating transcription factor conferring increased susceptibility to a viral infection in the CNS in humans. PMID: 26216125
  45. The data show that SARS coronavirus PLpro also inhibits IRF3 activation at a step after phosphorylation and that this inhibition is dependent on the de-ubiquitination (DUB) activity of PLpro. PMID: 25481026
  46. The presence of TLR3 and IRF-3 in both human normal and PCa clinical samples, potentially envisaging poly I:C-based therapy for PCa. PMID: 25444175
  47. This study demonstrates HSV-2 US1 inhibits production of IFN-beta by suppressing activation of IFN-beta promoter by interfering with the association of nuclear IRF-3 with IRF-3-responsive domain of the the IFN-beta promoter. PMID: 25712217
  48. These results demonstrate for the first time that IRF3 and IRF7 are both involved in inducing TLR4-dependent IFN-beta expression in response to HSV-2 in its primary infected genital epithelial cells. PMID: 24722640
  49. Collectively, this study characterizes a novel protein complex, Tom70/Hsp90/IRF3/Bax, that is important for Sendai virus-induced apoptosis. PMID: 25609812
  50. Stable activation during respiratory syncytial virus infection requires both RIG-1 and MDA5. PMID: 24800889

Show More

Hide All

Database Links

HGNC: 6118

OMIM: 603734

KEGG: hsa:3661

STRING: 9606.ENSP00000310127

UniGene: Hs.289052

Involvement In Disease
Herpes simplex encephalitis 7 (HSE7)
Protein Families
IRF family
Subcellular Location
Cytoplasm. Nucleus. Mitochondrion.
Tissue Specificity
Expressed constitutively in a variety of tissues.

Q&A

What is IRF3 and why is it important in immunological research?

IRF3 is a critical transcription factor (~55 kDa) involved in the innate immune response, particularly in the detection of viral infections and the activation of type I interferons such as IFN-β. Structurally, IRF3 consists of several domains, including a DNA-binding domain, an IRF association domain, and a regulatory C-terminal domain . It remains inactive in the cytoplasm until phosphorylated following viral detection, after which it dimerizes, translocates to the nucleus, and initiates the transcription of antiviral genes . This makes IRF3 vital for studying host-pathogen interactions and antiviral immunity mechanisms.

How do I select the appropriate IRF3 antibody for my experiment?

Selection criteria should include:

  • Specificity: Verify specificity through knockout validation or comparison with isotype controls. For instance, the D-3 monoclonal antibody targets amino acids 389-427 near the C-terminus of human IRF3 , while clone 3F10 recognizes amino acids 108-166 .

  • Application compatibility: Ensure the antibody is validated for your specific application:

    • Western blotting (dilutions typically 1/1000-1/3000)

    • Immunoprecipitation

    • Immunofluorescence

    • Flow cytometry

    • ELISA

  • Cross-reactivity profile: Some antibodies show cross-species reactivity (human, mouse, rat), while others are species-specific. For example, AF4019 shows approximately 20% cross-reactivity with recombinant mouse IRF3 in direct ELISAs .

  • Conjugation status: Available as non-conjugated or conjugated with agarose, HRP, PE, FITC, or various Alexa Fluor conjugates for specialized applications .

What are the common troubleshooting issues when using IRF3 antibodies in Western blotting?

IssuePotential CauseRecommended Solution
Multiple bandsPhosphorylation statesUse phospho-specific antibodies or lambda phosphatase treatment
No signalLow expression levelConcentrate protein or use enrichment (IP before WB)
Non-specific bandsCross-reactivityInclude proper negative controls (IRF3 knockout cells)
Inconsistent resultsProtein degradationAdd protease and phosphatase inhibitors
Weak signalInsufficient transferOptimize transfer conditions for high molecular weight proteins

For optimal results, use freshly prepared lysates with phosphatase inhibitors to preserve IRF3 phosphorylation states. Include IRF3 knockout cell lines as negative controls to confirm band specificity, as demonstrated with HeLa parental and IRF3 knockout cell lines .

How should I optimize fixation protocols for IRF3 immunofluorescence staining?

Effective IRF3 immunofluorescence depends greatly on fixation methods:

  • Fixative selection: For phosphorylated IRF3 detection, 4% paraformaldehyde is recommended (10-15 minutes at room temperature), as it better preserves protein phosphorylation compared to methanol fixation.

  • Permeabilization: Use 0.1-0.5% Triton X-100 or saponin for nuclear transcription factor accessibility4. For flow cytometry applications, fixation buffers like Flow Cytometry Fixation Buffer followed by permeabilization with Flow Cytometry Permeabilization/Wash Buffer I have been validated .

  • Antibody concentration: For immunofluorescence, optimal concentration ranges (e.g., 15 µg/mL for 3 hours at room temperature) should be determined experimentally .

  • Signal validation: Always include appropriate controls:

    • Primary antibody omission

    • IRF3-deficient cells

    • Competing peptide blockade

Counterstaining with DAPI helps confirm nuclear translocation during IRF3 activation, while cytoplasmic staining predominates in resting cells .

What are effective strategies for detecting IRF3 activation status?

IRF3 activation can be monitored through:

  • Phosphorylation-specific antibodies: Target key phosphorylation sites (Ser385/386 and Ser396), with Ser385/386 being particularly critical for IRF3 activation .

  • Nuclear translocation: Use fractionation or immunofluorescence to track movement from cytoplasm to nucleus. The shift is dramatic upon viral infection or poly(I:C) stimulation.

  • Dimerization assays: Non-denaturing gel electrophoresis can detect IRF3 dimers.

  • Proximity ligation assay (PLA): This technique can detect IRF3 interactions with co-factors like TBK1. PLA has been used to show how Ebola virus reduces TBK1:IRF3 complexes during infection .

  • Chromatin immunoprecipitation (ChIP): Quantifies IRF3 binding to target gene promoters, allowing measurement of binding intensity at sites like IFN-β promoter.

How can I validate the specificity of IRF3 antibodies for my research application?

Rigorous validation approaches include:

  • Genetic controls:

    • Use IRF3 knockout cell lines (as demonstrated in Western blot validations with HeLa parental and IRF3 knockout cell lines)

    • siRNA/shRNA knockdown samples

  • Peptide competition: Pre-incubate antibody with immunizing peptide to confirm binding specificity.

  • Cross-validation: Test multiple antibodies targeting different IRF3 epitopes.

  • Stimulation experiments: Verify increased signal upon appropriate stimulation (viral infection, poly(I:C)).

  • Multiple techniques: Confirm findings using complementary methods (IF, WB, IP).

How can IRF3 antibodies be used to investigate cross-talk between different IRF family members?

IRF family cross-talk investigation requires specialized approaches:

  • Sequential ChIP (ChIP-reChIP): This technique helps identify co-occupancy of IRF3 with other IRFs (IRF5, IRF9) at the same genomic region. Studies have shown that IRF3, IRF5, and IRF9 regulate overlapping but distinct sets of target genes .

  • Co-immunoprecipitation with specificity controls: When investigating interactions, use antibodies targeting unique regions to prevent cross-reactivity between family members. This is critical as IRFs share significant homology in DNA-binding domains.

  • Isoform-specific detection: Ensure antibody specificity for detecting the alpha isoform without cross-reactivity to beta isoforms, as demonstrated with GSK3 isoform-specific antibodies4.

  • Combined stimulation experiments: Challenge cells with multiple stimuli to observe competition or cooperation between IRF pathways. Research has shown that different ligand combinations can either inhibit or enhance IRF3, IRF5, and IRF9 binding to their respective genomic regions .

What are the best practices for using IRF3 antibodies to study viral evasion mechanisms?

Viral evasion studies require specialized experimental approaches:

  • Subcellular fractionation: Determine if viruses alter normal IRF3 distribution patterns. For example, Ebola virus has been shown to sequester IRF3 in viral inclusion bodies (IBs) .

  • Time-course experiments: Track IRF3 phosphorylation, dimerization, and nuclear translocation kinetics during infection.

  • Co-localization studies: Use dual-labeling with viral proteins and IRF3. This approach revealed how EBOV traps IRF3 in specific cellular compartments.

  • Proximity ligation assay (PLA): Detect interactions between IRF3 and viral proteins or usual binding partners. PLA experiments demonstrated that EBOV infection significantly reduces TBK1:IRF3 complexes that normally form after poly(I:C) stimulation .

  • Functional rescue experiments: Determine if supplementing IRF3 activity can overcome viral evasion strategies.

How can phospho-specific IRF3 antibodies be leveraged to dissect signaling pathways?

Phospho-specific antibodies enable precise pathway interrogation:

  • Site-specific analysis: Different phosphorylation sites have distinct functional impacts. Research has demonstrated that mutations at Ser-385/386 abolish promotion of IFN-β, ISG15, and IFITM3 expression, while Ser-396 mutations only moderately affect these pathways .

  • Temporal regulation: Track phosphorylation sequence during activation using site-specific antibodies.

  • Pathway inhibitor studies: Combined with kinase inhibitors to map phosphorylation dependencies.

  • Quantitative analysis: Use phospho-to-total IRF3 ratios for accurate activation measurement.

  • Mutant complementation: Compare antibody reactivity in cells expressing wild-type versus phosphorylation site mutants of IRF3. Studies confirmed that both S385/386A and S396A mutations strongly diminished ISG15 and IFITM3 protein expression .

How should IRF3 antibodies be stored and handled to maintain optimal performance?

Proper handling ensures antibody longevity and consistent results:

  • Storage temperature: Most IRF3 antibodies should be stored at 2-8°C and should not be frozen . For long-term storage, small aliquots at -20°C may be recommended to prevent freeze-thaw cycles.

  • Buffer considerations: Many IRF3 antibodies come in phosphate-buffered saline (PBS) with preservatives like 0.02% sodium azide and 10% glycerol .

  • Light sensitivity: For fluorophore-conjugated antibodies (e.g., Alexa Fluor 488-conjugated IRF3 antibodies), protection from light is essential .

  • Shelf life: Typical guarantee period is 12 months from date of despatch when stored properly .

  • Working solution preparation: Dilute only the amount needed for immediate use in appropriate buffer.

What are the key differences between using IRF3 antibodies in flow cytometry versus immunofluorescence microscopy?

ParameterFlow CytometryImmunofluorescence Microscopy
Cell stateSuspensionAdherent/fixed on slides
Fixation protocolQuick fixation (10 min)May require longer fixation (15-20 min)
PermeabilizationMore critical (must reach all cells)Can be visualized for efficiency
Antibody dilutionTypically higher concentrationCan use lower concentrations with longer incubation
ControlsIsotype controls criticalPrimary antibody omission often sufficient
Data outputQuantitative (MFI)Qualitative and semi-quantitative
Subcellular localizationLimited resolutionHigh resolution of subcellular compartments

For flow cytometry, successful detection of IRF3 has been demonstrated using Alexa Fluor 488-conjugated monoclonal antibodies with specific fixation and permeabilization buffers in cell lines like Daudi human Burkitt's lymphoma . For immunofluorescence, NorthernLights 557-conjugated secondary antibodies have been used successfully with primary IRF3 antibodies at 15 µg/mL concentration for detailed subcellular localization .

How can IRF3 antibodies be effectively used in ChIP-seq experiments to study genome-wide binding patterns?

For successful IRF3 ChIP-seq:

  • Crosslinking optimization: Typically 1% formaldehyde for 10 minutes, but may require titration.

  • Antibody selection: Choose ChIP-validated antibodies targeting the DNA-binding domain or C-terminal region. Avoid phospho-specific antibodies unless specifically studying activated IRF3.

  • Chromatin fragmentation: Aim for 200-500bp fragments for optimal resolution of binding sites.

  • Controls: Include:

    • Input chromatin

    • IgG control

    • IRF3-deficient cells

    • Stimulated vs. unstimulated samples to capture activation-dependent binding

  • Data analysis: Focus on motif analysis to identify IRF3 binding elements (IBEs) and co-occurring transcription factor motifs. Research has shown that IRF3 binding co-occurs frequently with RelA binding sites .

  • Validation: Confirm key peaks with ChIP-qPCR, especially at known IRF3 target genes like IFN-β, CCL5, and CXCL10 .

How can IRF3 antibodies be used to study viral pathogenesis mechanisms?

IRF3 antibodies provide critical insights into viral pathogenesis:

  • Evasion mechanism profiling: Different viruses employ distinct strategies to counteract IRF3. For example, Ebola virus sequesters IRF3 in viral inclusion bodies to prevent it from activating interferon responses .

  • Comparative analysis: Study how various viral proteins (NS1, NS3/4A, VP35) target different aspects of IRF3 activation.

  • Time-course studies: Track IRF3 phosphorylation, dimerization, and nuclear translocation during different phases of viral infection.

  • Mutant virus comparisons: Compare wild-type viruses with mutants lacking specific immune evasion genes.

  • Host-range determination: Investigate how viral antagonism of IRF3 varies across host species, particularly at host-species barriers.

What approaches can be used to study IRF3 signaling in primary immune cells versus cell lines?

Primary cell research requires specific considerations:

  • Antibody validation: Revalidate antibodies in primary cells, as expression levels and isoforms may differ from cell lines:

    Cell TypeRecommended Antibody Dilution (WB)Optimization Notes
    Cell lines (HeLa, U937)1:1000-3000Standard protocols usually effective
    Primary human MDMs1:500-1000May require longer exposure times
    Mouse BMDMs1:500-1000May show species-specific differences
    Human PBMCs1:500-1000Higher background common
  • Transfection challenges: For primary cells resistant to transfection, antibody-based detection becomes even more critical for studying endogenous IRF3.

  • Activation kinetics: Primary cells often show different IRF3 activation kinetics than immortalized lines. Design time-course experiments accordingly.

  • Cell-type specific interactions: Investigate co-binding with lineage-specific transcription factors, as IRF3 frequently collaborates with other transcription factors like AP-1 and NF-κB .

  • Ex vivo validation: Confirm key findings from cell cultures in tissue samples using IHC with IRF3 antibodies.

How are IRF3 antibodies being used to investigate the interplay between IRF3 and co-regulatory proteins?

Recent research focuses on complex interactions:

  • Co-immunoprecipitation coupled with mass spectrometry: Identify novel IRF3-interacting proteins using antibodies that don't disrupt protein complexes.

  • Proximity labeling techniques: BioID or APEX2 fusions with IRF3 coupled with specific antibody detection.

  • Sequential ChIP (ChIP-reChIP): Investigate co-occupancy of IRF3 with other transcription factors at genomic loci, revealing extensive collaboration between IRF3 and RelA in antiviral responses .

  • FRET/BRET analysis: Study real-time interactions between IRF3 and partners using antibody-based detection systems.

  • PBLD interactions: Recent findings show that phenazine biosynthesis-like domain-containing protein (PBLD) enhances type I interferon expression through IRF3, suggesting new regulatory mechanisms .

What are the latest techniques for studying post-translational modifications of IRF3 using specific antibodies?

Advanced PTM research approaches include:

  • Phospho-specific antibodies: Critical for distinguishing between various activated forms. Research has shown differential importance of phosphorylation sites, with Ser-385/386 being more critical than Ser-396 for certain pathways .

  • Mass spectrometry validation: Confirm antibody-detected modifications with MS analysis of immunoprecipitated IRF3.

  • High-resolution imaging: Study spatiotemporal dynamics of IRF3 modifications in different cellular compartments.

  • Single-cell analysis: Detect heterogeneity in IRF3 activation states within populations using phospho-flow cytometry.

  • Orthogonal labeling strategies: Combine antibody detection with chemical labeling of specific modifications.

How can IRF3 antibodies contribute to therapeutic research targeting innate immune pathways?

Therapeutic applications leverage IRF3 antibodies in multiple ways:

  • Compound screening: Use phospho-IRF3 antibodies to screen for modulators of innate immune signaling.

  • Target validation: Confirm mechanism of action for compounds designed to modulate IRF3 activity.

  • Biomarker development: Explore IRF3 activation status as a predictive or pharmacodynamic biomarker.

  • Safety assessment: Evaluate potential off-target effects of therapeutics on innate immune pathways.

  • Antibody-based therapies: Development of antibodies that could potentially deliver cargoes to cells with aberrant IRF3 activity.

  • Viral evasion countermeasures: Research into preventing viral sequestration of IRF3, such as that seen with Ebola virus, could lead to novel broad-spectrum antiviral approaches .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.