IFIT3 Antibody

Shipped with Ice Packs
In Stock

Description

What is IFIT3 Antibody?

IFIT3 antibodies are immunodetection reagents specifically targeting the IFIT3 protein, a 56–65 kDa interferon-stimulated gene (ISG) product. Structurally, IFIT3 contains eight tetratricopeptide repeat (TPR) motifs that mediate protein-protein interactions critical for its antiviral and immunoregulatory roles . These antibodies are widely used in techniques such as Western blot (WB), immunoprecipitation (IP), immunofluorescence (IF), and immunohistochemistry (IHC) .

Applications of IFIT3 Antibodies

IFIT3 antibodies are utilized in diverse experimental contexts:

  • Western Blot: Detects endogenous IFIT3 (~60–65 kDa) in human cell lines (e.g., HEK293, HepG2) .

  • Immunofluorescence: Localizes IFIT3 to mitochondria and cytosol during viral infection .

  • Immunohistochemistry: Identifies IFIT3 expression in clinical samples (e.g., COVID-19 patient neutrophils) .

  • Functional Studies: Knockdown/overexpression experiments reveal IFIT3’s role in restricting viruses like influenza A, Ebola, and HBV .

Antiviral Mechanisms

  • SARS-CoV-2: Elevated IFIT3 expression in neutrophils and monocytes correlates with reduced viral load in COVID-19 patients .

  • HBV: IFIT3 knockdown increases HBsAg/HBeAg levels, while overexpression enhances IFN-α’s suppression of HBV DNA .

  • Adenovirus: IFIT3 inhibits viral replication by activating STING and MAVS pathways independently of pathogen-associated molecular patterns (PAMPs) .

Cell Cycle Regulation

IFIT3 upregulates CDKN1A/p21 and CDKN1B/p27, arresting cell proliferation to limit viral spread .

Neutrophil-Specific Activity

Single-cell RNA sequencing identifies IFIT3 as a key ISG in neutrophils, highlighting its role in early antiviral defense .

Key Studies and Citations

  1. MAVS-TBK1-IRF3 Activation: IFIT3 bridges MAVS and TBK1, boosting IRF3 phosphorylation and IFN-β production .

  2. STAT2 Dependency: IFIT3 enhances JAK-STAT signaling, critical for IFN-α’s antiviral effects against HBV .

  3. Broad-Spectrum Restriction: IFIT3 suppresses diverse viruses (e.g., dengue, Ebola) by sequestering viral RNA and enhancing IFN responses .

Limitations and Future Directions

While IFIT3 antibodies have proven invaluable, unresolved questions remain:

  • Mechanistic Details: How IFIT3 differentially restricts enveloped vs. non-enveloped viruses .

  • Therapeutic Potential: Engineering IFIT3-based therapies requires deeper insights into its immunomodulatory networks .

Product Specs

Buffer
PBS with 0.02% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze-thaw cycles.
Lead Time
Typically, we can dispatch your order within 1-3 business days of receiving it. Delivery times may vary depending on the purchasing method and location. For specific delivery times, please consult your local distributor.
Synonyms
CIG 49 antibody; CIG49 antibody; GARG 49 antibody; IFI-60K antibody; IFI60 antibody; IFI60K antibody; IFIT-3 antibody; IFIT-4 antibody; Ifit3 antibody; IFIT3_HUMAN antibody; IFIT4 antibody; Interferon induced 60 kDa protein antibody; Interferon induced protein 60 antibody; Interferon induced protein with tetratricopeptide repeats 3 antibody; Interferon-induced 60 kDa protein antibody; Interferon-induced protein with tetratricopeptide repeats 3 antibody; Interferon-induced protein with tetratricopeptide repeats 4 antibody; IRG2 antibody; ISG-60 antibody; ISG60 antibody; P60 antibody; Retinoic acid induced gene G protein antibody; Retinoic acid-induced gene G protein antibody; RIG G antibody; RIG-G antibody; RIGG antibody; RP11-149I23.4 antibody
Target Names
IFIT3
Uniprot No.

Target Background

Function
IFIT3 is an IFN-induced antiviral protein that inhibits cellular and viral processes, including cell migration, proliferation, signaling, and viral replication. It enhances MAVS-mediated host antiviral responses by acting as an adapter that bridges TBK1 to MAVS, leading to the activation of TBK1 and phosphorylation of IRF3. Phosphorylated IRF3 translocates into the nucleus to promote antiviral gene transcription. IFIT3 exhibits antiproliferative activity by upregulating cell cycle negative regulators CDKN1A/p21 and CDKN1B/p27. Normally, CDKN1B/p27 turnover is regulated by COPS5, which binds CDKN1B/p27 in the nucleus and exports it to the cytoplasm for ubiquitin-dependent degradation. IFIT3 sequesters COPS5 in the cytoplasm, thereby increasing nuclear CDKN1B/p27 protein levels. It upregulates CDKN1A/p21 by downregulating MYC, a repressor of CDKN1A/p21. IFIT3 can negatively regulate the apoptotic effects of IFIT2.
Gene References Into Functions
  1. Our research demonstrated that IFIT3 is upregulated during hepatic ischemia-reperfusion injury (IRI) and that knockdown of IFIT3 exerts potent protective effects against hepatic IRI in vitro and in vivo. These findings not only revealed the mechanism for IFIT3-regulated hepatic IRI but also proposed potential clinical significance of treatment targeting IFIT3 for patients undergoing liver resection and liver transplantation. PMID: 29734133
  2. These findings indicated that hepatitis B virus-induced miR146a attenuates cell-intrinsic anti-viral innate immunity through targeting RIG-I and RIG-G. PMID: 27210312
  3. Low RIG-G expression is associated with lung cancer. PMID: 27602766
  4. Biomarker expression in pancreatic ductal adenocarcinoma (PDAC) of CXCR4, SMAD4, SOX9 and IFIT3 will be prospectively assessed by immunohistochemistry and verified by rt.-PCR from tumor and adjacent healthy pancreatic tissue of surgical specimen. PMID: 28356064
  5. These data suggest that postpartum, the normalization of the physiological rheostat controlling IFN signaling depends on IFNL3 genotype. PMID: 27601663
  6. Higher expression of IFIT3 enhances anti-apoptotic activity and chemotherapy resistance of pancreatic ductal adenocarcinoma cells. High expression of IFIT3 was independently correlated to shorter patients' survival and may serve as a prognostic marker. PMID: 28210844
  7. Among the associated variants were two in regions previously unreported for COPD; a low frequency non-synonymous SNP in MOCS3 (rs7269297, pdiscovery=3.08x10(-6), preplication=0.019) and a rare SNP in IFIT3, which emerged in the meta-analysis (rs140549288, pmeta=8.56x10(-6)). PMID: 26917578
  8. HSV-1 was shown for the first time to evade the antiviral function of IFIT3 via UL41. PMID: 27681138
  9. Results indicated that RIG-G level was high in maturated cells and low in blast cells, and suggested that RIG-G might play a role in the differentiation of bone marrow hemocytes in vivo. PMID: 26686474
  10. The transcription factor SOX9, which is linked to regulation of hypoxia-related genes, was identified as a key mediator of upregulation of the oncogene IFIT3 and thereby sustaining a "pseudoinflammatory" cellular condition in pancreatic tumors. PMID: 25650658
  11. Reovirus T3D infection induced STAT-1, ISG-15, IFIT-1, Mx1 and IFIT-3 expression. PMID: 25905045
  12. In cell models of dengue virus 2 infection, authors found that IFITM3 contributed to both the baseline and interferon-induced inhibition of virus entry. PMID: 25131332
  13. Protective roles of IFIT3 following IFN-alpha production in DV infection of human lung epithelial cells. PMID: 24223959
  14. These findings reveal for the first time the negative regulation of Rig-G on SCF-E3 ligase activities through disrupting CSN complex, not only contributing to further investigation on biological functions of Rig-G, but also leading to better understanding of the CSN complex as a potential target in tumor diagnosis and treatment. PMID: 23415865
  15. RIG-G gene expression is closely correlated with the cross-talk between all-trans retinoic acid and IFN-alpha-induced signaling pathways in NB4 tumor cells. PMID: 22490698
  16. Our study characterizes IFIT3 as an important modulator in innate immunity. PMID: 21813773
  17. All-trans retinoic acid upregulated RIG-G in NB4 cells by upregulating IRF1, IRF9 and STAT2. PMID: 20137113
  18. The complex STAT2/IRF-9 is the key factor mediating the expression of RIG-G gene regulated by IFN-alpha. PMID: 20403236
  19. We concluded that the expression of RIG-G was independent on the classical JAK-STAT pathway, but could be greatly increased by it. PMID: 21056555
  20. Identification of alpha interferon-induced genes associated with antiviral activity in Daudi cells and characterization of IFIT3 as a novel antiviral gene. PMID: 20686046
  21. Transcription factors for the reporter gene. CONCLUSION: Both ISRE I and ISRE II on the RIG-G promoter are the binding sites for the complex of transcription factors. They are required for RIG-G expression, and ISRE I has a preferential role over ISRE II. PMID: 20533260
  22. The induction of IFIT4 transcription by IFN-alpha depends upon sequential activation of PKCdelta, JNK and STAT1, and the influence of PKCdelta or JNK on IFN-alpha-mediated induction of IFIT4 is dependent upon the phosphorylation of STAT1 at Ser-727. PMID: 17933493
  23. IFIT4 may play roles in promoting monocyte differentiation into dendritic cell-like cells (DCLCs) and in directing DCLCs to modulate T-helper-1 cell differentiation, thus contributing to the autoimmunity and pathogenesis of systemic lupus erythematosus. PMID: 18706081
  24. Induction of RIG-G by retinoic acid in NB4 cells resulted, to some extent, from an IFNalpha autocrine pathway, a finding that suggests a novel mechanism for the signal cross-talk between IFNalpha and retinoic acid. PMID: 19351818
Show More
Hide All
Database Links

HGNC: 5411

OMIM: 604650

KEGG: hsa:3437

STRING: 9606.ENSP00000360876

UniGene: Hs.47338

Protein Families
IFIT family
Subcellular Location
Cytoplasm. Mitochondrion.
Tissue Specificity
Expression significantly higher in peripheral blood mononuclear cells (PBMCs) and monocytes from systemic lupus erythematosus (SLE) patients than in those from healthy individuals (at protein level). Spleen, lung, leukocytes, lymph nodes, placenta, bone m

Customer Reviews

Overall Rating 5.0 Out Of 5
,
B.A
By Anonymous
★★★★★

Applications : IHC staining

Sample type: human cells

Review: The protein contents and distribution of IFIT1 and IFIT3 were examined in nonmetastatic HCC tissues, metastatic HCC tissues, and para-carcinoma tissues by IHC staining.

Q&A

How do I select the appropriate IFIT3 antibody for my research application?

When selecting an IFIT3 antibody, consider the specific epitope recognition pattern and intended application. IFIT3 contains conserved RNA-binding domains and interacts with multiple signaling proteins, so antibodies targeting different epitopes may yield varied results. For applications requiring detection of specific IFIT3 domains (such as the RNA-binding sites at C239, C283, or S360/H361), select antibodies raised against these specific regions . For general IFIT3 detection, polyclonal antibodies recognizing multiple epitopes provide robust signal across applications.

For immunoprecipitation studies investigating IFIT3's interactions with binding partners like IFIT2, TBK1, or MAVS, choose antibodies validated for IP applications that don't interfere with protein-protein interaction sites. Western blot applications typically work well with most IFIT3 antibodies, though sensitivity may vary by manufacturer .

What validation steps are essential before using a new IFIT3 antibody?

Proper validation of IFIT3 antibodies is crucial due to the protein's complex biology and potential cross-reactivity with other IFIT family members. Essential validation steps include:

  • Specificity testing using positive controls (IFN-stimulated cells) versus negative controls (IFIT3 knockout cells)

  • Western blot analysis confirming a single band at approximately 60 kDa (the expected molecular weight of IFIT3)

  • Confirmation of antibody specificity via siRNA knockdown or CRISPR knockout cells

  • Cross-reactivity testing with other IFIT family members, particularly IFIT2, which shares structural similarities and can heterodimerize with IFIT3

  • Validation in the specific experimental system and cell types to be used in your research

Remember that IFIT3 expression is typically low in unstimulated cells but increases significantly after interferon treatment, so include appropriate stimulated positive controls .

What are the key differences between monoclonal and polyclonal IFIT3 antibodies in research applications?

Monoclonal and polyclonal IFIT3 antibodies present distinct advantages and limitations for research applications:

Monoclonal antibodies:

  • Provide consistent lot-to-lot reproducibility

  • Offer high specificity for a single epitope

  • May have reduced sensitivity for detecting IFIT3 under different conformational states

  • Particularly useful for quantitative applications and where batch consistency is crucial

  • May be less effective if the epitope is masked by protein-protein interactions

Polyclonal antibodies:

  • Recognize multiple epitopes, increasing detection sensitivity

  • Better at detecting denatured proteins in applications like Western blotting

  • May exhibit batch-to-batch variation

  • Provide more robust detection of IFIT3 across different experimental conditions

  • Potentially higher risk of cross-reactivity with other IFIT family members

For studies examining IFIT3's interactions with viral RNA or protein binding partners, consider how antibody binding might affect these interactions. In co-immunoprecipitation experiments investigating IFIT3-IFIT2 heterodimers, carefully select antibodies that don't interfere with the dimerization interface .

How should I design experiments to distinguish between IFIT3 homo- and heterodimers?

Distinguishing between IFIT3 homo- and heterodimers requires careful experimental design due to their similar biochemical properties. Based on research findings, IFIT3 forms both homodimers and heterodimers with IFIT2, with the heterodimer being thermodynamically preferred and potentially more active in promoting viral gene expression .

Methodological approach:

  • Sequential immunoprecipitation: First immunoprecipitate with an IFIT3-specific antibody, then probe the precipitate with an IFIT2-specific antibody to identify heterodimers.

  • Size exclusion chromatography: Separate protein complexes by size before immunoblotting to distinguish between monomers, homodimers, and heterodimers.

  • Fluorescence resonance energy transfer (FRET): Tag IFIT2 and IFIT3 with appropriate fluorophores to detect heterodimer formation in live cells.

  • Co-expression systems: As demonstrated in the research, transfect cells with tagged versions of IFIT2 and IFIT3 (e.g., IFIT3-V5 and IFIT3-eGFP) and use tag-specific antibodies for co-immunoprecipitation analyses .

For control experiments, use IFIT2-/- and IFIT3-/- knockout cells to validate the specificity of your detection methods. When interpreting results, remember that co-expression of both proteins yields the highest viral gene expression enhancement, suggesting a synergistic effect of the heterodimer .

What are the optimal conditions for detecting IFIT3 expression changes during viral infection?

Detecting IFIT3 expression changes during viral infection requires careful timing and appropriate controls due to the dynamic nature of interferon responses and IFIT3's dual role in viral immunity.

Optimal experimental design should include:

  • Temporal considerations: Monitor IFIT3 expression at multiple timepoints (2, 6, 12, 24, and 48 hours post-infection) to capture both early interferon-induced upregulation and potential viral-mediated regulation.

  • Appropriate controls:

    • Uninfected cells with and without interferon stimulation

    • Cells treated with UV-inactivated virus to distinguish between active viral replication and innate immune activation

    • For influenza studies, include both wild-type virus and strains with compromised interferon antagonism

  • Detection methods:

    • Western blotting with validated antibodies (1:800 dilution recommended for IFIT3 detection)

    • qRT-PCR to measure transcript levels alongside protein expression

    • Immunofluorescence to assess subcellular localization changes

  • Cell-type considerations: IFIT3 expression and function may vary between cell types, with lung epithelial cells and macrophages being particularly relevant for respiratory viruses like influenza .

When interpreting results, remember that IFIT3 plays a dual role—it is both an interferon-stimulated gene with antiviral properties and can be exploited by certain viruses (like influenza A) to enhance replication .

How can I effectively study IFIT3's RNA-binding activity in relation to viral infection?

Studying IFIT3's RNA-binding activity in viral infection contexts requires specialized techniques that can capture these interactions while maintaining their biological relevance. Based on recent research findings, IFIT3 binds viral mRNAs through specific RNA-binding sites, which contributes to its pro-viral activity during influenza A virus infection .

Recommended methodological approaches:

  • RNA-binding site identification (RBS-ID):

    • UV crosslink cells to capture RNA-protein interactions

    • Purify IFIT3 from cell lysates using immunoprecipitation

    • Digest RNA and protein, then perform mass spectrometry to identify crosslinked residues

    • This approach identified three high-confidence RNA-protein crosslink sites in IFIT3: C239, C283, and S360/H361

  • RNA immunoprecipitation (RIP):

    • Crosslink RNA-protein complexes in virus-infected cells

    • Immunoprecipitate IFIT3 using specific antibodies

    • Extract and analyze bound RNA through sequencing or qRT-PCR

    • Include appropriate controls: IgG precipitation, RNA from total lysate

  • Mutagenesis studies:

    • Generate IFIT3 constructs with mutations at identified RNA-binding sites

    • Express these constructs in IFIT3-knockout cells

    • Assess effects on viral gene expression and replication

    • Compare wild-type and mutant IFIT3 binding to viral RNAs

  • Structural analysis:

    • Use structural prediction models based on related proteins (e.g., IFIT2)

    • Map conserved basic patches that may form RNA-binding sites

    • Correlate structural features with functional effects of mutations

When designing these experiments, consider that IFIT3's RNA-binding activity appears to enhance translation of viral proteins, potentially through interactions with specific viral mRNA structures or sequences .

How do I interpret contradictory data regarding IFIT3's antiviral versus proviral functions?

The dual role of IFIT3 as both an antiviral and proviral factor presents a significant challenge for data interpretation. Research has revealed that IFIT3's function can vary depending on the virus type, cell context, and experimental conditions. When encountering contradictory data, consider the following analytical framework:

When designing experiments to resolve contradictions, include appropriate controls targeting each of these variables. Use IFIT3 knockout cells complemented with wild-type or mutant IFIT3 to isolate specific functions, and compare results across multiple viral systems to identify virus-specific versus general mechanisms .

What methodological approaches are most effective for studying IFIT3's role in tumor immunity?

Studying IFIT3's complex role in tumor immunity requires multidimensional approaches that capture both molecular mechanisms and physiological relevance. Recent research suggests that IFIT3 plays a dual role in tumor progression, influencing both anti-tumor immune responses and potentially promoting tumor growth through inflammatory pathways .

Recommended methodological framework:

  • In vitro tumor cell models:

    • Generate stable IFIT3 knockdown and overexpression in tumor cell lines

    • Assess changes in proliferation, migration, invasion, and cytokine production

    • Evaluate NF-κB pathway activation using reporter assays

    • Measure changes in expression of immune checkpoint molecules (PD-L1, etc.)

  • Immune cell co-culture systems:

    • Co-culture IFIT3-modified tumor cells with immune cells (T cells, NK cells)

    • Measure immune cell activation, cytotoxicity, and cytokine production

    • Analyze immune checkpoint receptor-ligand interactions

    • Use transwell systems to distinguish between contact-dependent and soluble factor-mediated effects

  • In vivo tumor models:

    • Implant IFIT3-modified tumor cells in immunocompetent and immunodeficient mice

    • Monitor tumor growth, metastasis, and survival

    • Analyze tumor-infiltrating immune cell populations by flow cytometry

    • Evaluate responses to immunotherapy (checkpoint inhibitors) and conventional therapies

  • Clinical correlation studies:

    • Assess IFIT3 expression in patient tumor samples by immunohistochemistry

    • Correlate expression with immune cell infiltration, prognosis, and treatment response

    • Analyze IFIT3 expression in public cancer databases in relation to immune signatures

    • Examine potential correlation with inflammatory biomarkers

For data analysis, integrate findings across these approaches to distinguish between IFIT3's direct effects on tumor cells and indirect effects mediated through immune modulation. Remember that IFIT3's function may vary significantly between cancer types and immune microenvironments .

How can I design experiments to investigate the interaction between IFIT3 and the RIG-I-like receptor signaling pathway?

Investigating IFIT3's interaction with the RIG-I-like receptor (RLR) signaling pathway requires carefully designed experiments that capture both physical interactions and functional consequences. According to the research, IFIT3 enhances RLR signaling and functions as an adapter bridging TBK1 to MAVS, leading to IRF3 activation and antiviral gene transcription .

Comprehensive experimental design should include:

  • Protein-protein interaction studies:

    • Co-immunoprecipitation of IFIT3 with RLR pathway components (RIG-I, MDA5, MAVS, TBK1)

    • Proximity ligation assays to visualize interactions in intact cells

    • Domain mapping using truncated constructs to identify interaction interfaces

    • FRET or BRET assays for real-time interaction monitoring in live cells

  • Functional signaling assays:

    • IRF3 phosphorylation and nuclear translocation assays

    • IFN-β promoter luciferase reporter assays in control vs. IFIT3-deficient cells

    • Quantification of downstream ISG expression by qRT-PCR and Western blotting

    • Analysis of signaling kinetics with time-course experiments

  • Structure-function analysis:

    • Generate IFIT3 mutants affecting potential interaction sites with RLR components

    • Assess both binding capacity and signaling function of mutants

    • Utilize structure prediction models to guide mutation design

    • Evaluate if RNA-binding activity (C239, C283, S360/H361 sites) affects RLR interaction

  • Viral infection models:

    • Compare RLR pathway activation in wild-type vs. IFIT3-deficient cells during infection

    • Use RNA viruses known to trigger RLR signaling (not just influenza)

    • Include stimulation with synthetic RLR ligands (poly(I:C), 5'ppp-RNA) as controls

    • Measure both early signaling events and downstream antiviral effects

When analyzing results, consider that IFIT3's effect on RLR signaling may differ depending on the virus type and infection stage. The seemingly contradictory roles of IFIT3 in antiviral immunity and enhancement of influenza replication may involve distinct mechanisms and interaction partners .

What are the optimal conditions for using IFIT3 antibodies in immunoprecipitation experiments?

Successful immunoprecipitation (IP) of IFIT3 requires optimization of several parameters to maintain protein interactions while ensuring specificity. Based on research protocols, the following conditions have proven effective:

  • Lysis buffer composition:

    • Use mild lysis conditions: 50 mM Tris pH 7.4, 150 mM NaCl, and 0.5% NP40

    • Include protease inhibitors to prevent degradation

    • For studying RNA-protein interactions, add RNase inhibitors

    • Consider adding phosphatase inhibitors when studying signaling interactions

  • Antibody selection and application:

    • For co-IP of natural complexes, use antibodies targeting regions distant from interaction interfaces

    • For tagged IFIT3 constructs, GFP-trap resin has shown high efficiency

    • Typical antibody concentration: 2-5 μg antibody per 500 μg of total protein

    • Pre-clear lysates with protein A/G beads to reduce non-specific binding

  • Incubation conditions:

    • Optimal incubation: overnight at 4°C with gentle rotation

    • For detecting transient interactions, consider crosslinking approaches

    • Multiple wash steps (4 times) with co-IP buffer to reduce background

  • Controls and validation:

    • Include IgG control immunoprecipitations

    • Use IFIT3 knockout cells as negative controls

    • For heterodimerization studies, include single transfection controls

    • Confirm successful IP by Western blot with IFIT3 antibody (1:800 dilution)

  • Special considerations for IFIT3-specific IPs:

    • When studying IFIT3-IFIT2 heterodimers, consider sequential IP

    • For RNA-binding studies, UV crosslinking prior to lysis preserves interactions

    • When investigating interaction with signaling proteins (TBK1, MAVS), include stimulation conditions that activate these pathways

For troubleshooting, if IP efficiency is low, interferon stimulation prior to lysis can increase IFIT3 expression levels, enhancing detection. If studying interactions disrupted by detergents, consider formaldehyde crosslinking before cell lysis .

How can I optimize Western blotting protocols for reliable IFIT3 detection?

Optimizing Western blotting for IFIT3 detection requires attention to several key parameters to achieve consistent and specific results. Based on published research protocols, the following optimizations are recommended:

  • Sample preparation:

    • Lyse cells in buffer containing 50 mM Tris pH 7.4, 150 mM NaCl, and 0.5% NP40

    • Include protease inhibitors to prevent degradation

    • Denature samples completely before loading

    • For comparing expression levels, normalize loading by total protein rather than housekeeping genes, as these may change during interferon responses

  • Gel and transfer conditions:

    • Use 10% SDS-PAGE gels for optimal separation (IFIT3 MW: ~60 kDa)

    • Transfer to nitrocellulose membranes (preferred over PVDF for IFIT3)

    • Use wet transfer methods for more consistent results with this protein

  • Blocking and antibody incubation:

    • Block membranes in a 1X solution of BSA (preferred over milk for IFIT3)

    • Primary antibody dilutions:

      • Anti-IFIT3 (Proteintech 15201-1-AP): 1:800 dilution

      • Anti-V5 tag (for tagged constructs): 1:5000 dilution

      • Anti-GFP (for GFP-fusion proteins): 1:1000 dilution

    • Secondary antibody recommendations: IR-compatible antibodies at 1:10,000 dilution for quantitative analysis

  • Detection methods:

    • IR-based imaging systems (such as Odyssey Fc) provide quantitative results and superior linearity

    • For enhanced sensitivity with chemiluminescence, use extended exposure times

    • For multiplex detection of IFIT3 with binding partners, use differently labeled secondary antibodies

  • Controls and validation:

    • Include positive controls (interferon-treated cells)

    • Use IFIT3 knockout cells as negative controls

    • For induction studies, include a time course of interferon treatment

    • When comparing viral effects, include both infected and uninfected samples

If background is high, increase washing stringency and optimize antibody dilutions. For weak signals, consider concentrating samples or using signal enhancement systems compatible with your detection method .

What are the most effective approaches for studying IFIT3 localization and trafficking during infection?

Studying IFIT3 localization and trafficking during viral infection requires techniques that can capture dynamic changes while maintaining biological relevance. The following approaches have proven effective in research settings:

  • Immunofluorescence microscopy:

    • Fix cells at multiple timepoints post-infection (2, 6, 12, 24 hours)

    • Use paraformaldehyde fixation (4%) followed by permeabilization

    • Block with BSA solution to reduce background

    • Co-stain for viral markers and potential interaction partners (MAVS, TBK1)

    • Include markers for subcellular compartments (ER, Golgi, mitochondria)

    • Analyze colocalization using appropriate statistical methods

  • Live-cell imaging with fluorescent fusion proteins:

    • Generate IFIT3-GFP fusion constructs (verify functionality)

    • Perform time-lapse imaging during infection

    • Consider photoactivatable or photoconvertible tags for pulse-chase experiments

    • Use fluorescently labeled viral components to track co-trafficking

    • Quantify movement parameters (velocity, directionality)

  • Biochemical fractionation:

    • Separate cellular compartments (cytosolic, membrane, nuclear, mitochondrial)

    • Analyze IFIT3 distribution by Western blotting

    • Track changes in distribution during infection progression

    • Co-fractionate potential interaction partners (IFIT2, TBK1, MAVS)

  • Proximity labeling approaches:

    • Generate IFIT3 fusions with BioID or APEX2

    • Identify proximal proteins in different cellular compartments

    • Compare proximity interactomes between uninfected and infected states

    • Validate key interactions by co-immunoprecipitation

  • Correlative light and electron microscopy (CLEM):

    • For high-resolution localization at specific infection stages

    • Particularly useful for examining associations with cellular membranes

    • Can reveal virus-induced membrane reorganization and IFIT3 localization

When interpreting results, consider that IFIT3 functions as an adapter linking TBK1 to MAVS, which suggests dynamic trafficking between cytosolic and mitochondrial locations . Additionally, its interaction with IFIT2 and role in viral RNA binding implies potential localization to sites of viral replication and protein synthesis . The seemingly contradictory roles of IFIT3 in both antiviral signaling and proviral enhancement of influenza replication may involve distinct localization patterns that should be carefully documented throughout the infection cycle .

How does IFIT3 expression pattern differ between various viral infections?

IFIT3 expression patterns show significant variations across different viral infections, reflecting both common interferon-stimulated gene (ISG) induction pathways and virus-specific manipulation mechanisms. Understanding these differences is crucial for interpreting experimental results.

Pattern comparison across viral infections:

  • Influenza A virus (IAV):

    • Rapid induction of IFIT3 expression via type I IFN signaling

    • Expression peaks approximately 12-24 hours post-infection

    • IAV appears to exploit IFIT3's RNA-binding activity to enhance viral mRNA translation

    • IFIT3 forms heterodimers with IFIT2 that further promote viral gene expression

  • RNA viruses triggering RIG-I pathways:

    • Strong, sustained IFIT3 induction via both IFN-dependent and IFN-independent mechanisms

    • IFIT3 enhances MAVS-mediated antiviral responses, creating a positive feedback loop

    • Functions as an adapter bridging TBK1 to MAVS, leading to IRF3 activation

    • Predominantly displays antiviral activity through signaling enhancement

  • DNA viruses:

    • Generally slower IFIT3 induction compared to RNA viruses

    • Expression primarily depends on cGAS-STING-mediated interferon production

    • Some DNA viruses actively suppress IFIT3 expression through viral antagonists

    • IFIT3's role may be more restricted to interferon signaling amplification

  • Persistent viral infections:

    • Chronic, low-level IFIT3 expression often observed

    • May contribute to inflammatory environments supporting tumor development

    • Potential role in regulating immune exhaustion via sustained activation

When designing experiments to study IFIT3 in different viral contexts, include appropriate timepoints that capture the expected expression dynamics for each virus type. Additionally, consider how virus-encoded interferon antagonists might influence IFIT3 expression patterns, and include viral mutants lacking these antagonists as controls when possible .

What experimental approaches best demonstrate IFIT3's proviral function in influenza infection?

Demonstrating IFIT3's proviral function in influenza A virus (IAV) infection requires multifaceted experimental approaches that capture both molecular mechanisms and functional outcomes. Based on recent research, the following methodological framework effectively reveals IFIT3's enhancement of IAV replication:

  • Genetic manipulation approaches:

    • Generate IFIT3 knockout cell lines using CRISPR-Cas9

    • Create IFIT3/IFIT2 double knockouts to study potential synergy

    • Complement knockout cells with wild-type or mutant IFIT3 constructs

    • This approach revealed reduced viral gene expression in cells lacking IFIT3

  • Reporter virus systems:

    • Utilize influenza reporter viruses expressing nanoluciferase

    • Compare reporter activity in wild-type versus IFIT3-deficient cells

    • Measure effects of IFIT3 overexpression on reporter activity

    • Determine the impact of IFIT3 RNA-binding site mutations

  • RNA-binding analysis:

    • Perform RNA-binding site identification (RBS-ID) to locate critical binding residues

    • Generate IFIT3 mutants with altered RNA-binding properties

    • Compare viral gene expression enhancement between wild-type and mutant IFIT3

    • This approach identified key residues (C239, C283, S360/H361) essential for IFIT3's proviral activity

  • Viral replication assessment:

    • Measure viral titers at multiple timepoints post-infection

    • Perform multi-step growth curves in various cell types

    • Compare replication kinetics between wild-type and IFIT3-deficient cells

    • These assays demonstrated increased viral yields when both IFIT2 and IFIT3 were present

  • Translational enhancement analysis:

    • Conduct polysome profiling to assess translation efficiency of viral mRNAs

    • Compare translation rates between IFIT3-sufficient and -deficient cells

    • Perform pulse-labeling experiments to measure viral protein synthesis rates

    • Examine association of IFIT3 with translation initiation factors

For experimental design, include appropriate controls for each assay, such as cells expressing IFIT3 mutants that retain protein stability but lack RNA-binding activity. Statistical analysis should involve multiple independent biological replicates (at least three) with technical triplicates for each condition, as performed in the referenced study .

How can I design experiments to investigate the mechanisms behind IFIT3's dual role in viral infections?

Investigating the seemingly contradictory dual role of IFIT3 in viral infections—both antiviral and proviral functions—requires carefully designed experiments that can distinguish between different mechanisms and contexts. Based on current research, IFIT3 appears to enhance antiviral signaling through the RIG-I pathway while also promoting influenza virus replication through RNA-binding activities .

Comprehensive experimental design strategy:

  • Domain-specific mutant analysis:

    • Generate an IFIT3 mutant panel targeting different functional domains:

      • RNA-binding site mutants (C239A, C283A, S360A/H361A)

      • Signaling interface mutants affecting TBK1 or MAVS interactions

      • IFIT2 interaction interface mutants

    • Express these mutants in IFIT3-knockout cells

    • Test each mutant's effect on both viral replication and interferon signaling

    • This approach can separate functions mediated by different protein domains

  • Virus-comparative studies:

    • Compare IFIT3's effect across multiple virus types:

      • Influenza A virus (reportedly enhanced by IFIT3)

      • VSV or Sendai virus (typically restricted by interferon signaling)

      • DNA viruses (different activation pathways)

    • Measure both viral replication and interferon response parameters

    • This can reveal virus-specific versus general mechanisms

  • Temporal dynamics investigation:

    • Perform time-course experiments with inducible IFIT3 expression

    • Introduce IFIT3 at different stages of infection

    • Monitor effects on viral replication and interferon signaling

    • This approach can identify timing-dependent functions

  • Interactome analysis:

    • Conduct immunoprecipitation-mass spectrometry at different infection stages

    • Compare IFIT3-interacting partners between antiviral and proviral contexts

    • Validate key interactions through co-immunoprecipitation and functional assays

    • This reveals context-specific protein complexes that may explain dual functions

  • RNA-binding specificity assessment:

    • Perform RNA immunoprecipitation followed by sequencing (RIP-seq)

    • Compare bound RNAs between different viral infections

    • Identify RNA features that correlate with proviral versus antiviral functions

    • Test if specific RNA structures determine IFIT3's functional outcome

For data analysis, integrate findings across these approaches to develop a comprehensive model of how IFIT3's functions are regulated. Consider factors such as protein concentration, cellular localization, post-translational modifications, and the presence of competing binding partners as potential switches between antiviral and proviral functions .

What methods are most effective for analyzing IFIT3's impact on tumor microenvironment?

Analyzing IFIT3's impact on the tumor microenvironment requires multidimensional approaches that capture both cellular interactions and molecular mechanisms. Based on research into IFIT3's role in tumor immunity, the following methodological framework provides comprehensive assessment:

  • Spatial transcriptomics and multiplex immunohistochemistry:

    • Apply multiplex immunofluorescence staining for IFIT3 alongside immune cell markers (CD4, CD8, CD68, etc.)

    • Perform spatial transcriptomics to correlate IFIT3 expression with immune signatures

    • Quantify distances between IFIT3-expressing cells and immune cell populations

    • Analyze tissue organization patterns in relation to IFIT3 expression gradients

  • Single-cell RNA sequencing of tumor tissues:

    • Dissociate tumor tissues from models with altered IFIT3 expression

    • Perform scRNA-seq to identify cell-type-specific effects of IFIT3

    • Analyze immune cell populations, activation states, and exhaustion markers

    • Construct cellular interaction networks based on receptor-ligand expression patterns

  • 3D co-culture systems:

    • Develop spheroid co-cultures containing tumor cells, fibroblasts, and immune cells

    • Compare systems with IFIT3-modified versus control tumor cells

    • Assess immune cell infiltration, cytokine production, and tumor cell death

    • Monitor changes in immune checkpoint molecule expression in response to IFIT3 alterations

  • In vivo imaging approaches:

    • Generate tumor models with fluorescently tagged immune cells and IFIT3-modified tumors

    • Perform intravital microscopy to track immune cell dynamics and interactions

    • Measure immune cell motility, dwell time, and contact duration with tumor cells

    • Correlate with therapeutic response to immunotherapy agents

  • Secretome analysis:

    • Collect conditioned media from IFIT3-modified versus control tumor cells

    • Perform multiplex cytokine/chemokine profiling

    • Assess impact on immune cell migration and polarization

    • Identify soluble factors mediating IFIT3's effects on the microenvironment

When designing these experiments, include appropriate controls such as IFIT3 knockout cells reconstituted with wild-type or mutant IFIT3. Consider that IFIT3's effects may vary based on tumor type, immune context, and inflammatory state, so validate findings across multiple model systems .

How should researchers design experiments to evaluate IFIT3 as a potential immunotherapy target?

Evaluating IFIT3 as a potential immunotherapy target requires systematic experiments that assess both efficacy and safety across multiple models. Given IFIT3's dual role in tumor immunity and innate immune responses, a comprehensive evaluation framework should include:

  • Target validation studies:

    • Analyze IFIT3 expression across cancer types using tissue microarrays and public databases

    • Correlate expression with patient survival and response to existing immunotherapies

    • Perform immunohistochemistry to assess IFIT3 expression in tumor versus normal tissues

    • Evaluate association with immune infiltration patterns and inflammatory markers

  • Therapeutic strategy development:

    • Design approaches for IFIT3 modulation:

      • Small molecule inhibitors targeting RNA-binding function

      • Peptide inhibitors disrupting protein-protein interactions

      • siRNA/shRNA for targeted knockdown

      • PROTAC-based degradation

    • Test effects on both tumor and immune cell functions in vitro

    • Evaluate specificity by comparing effects on other IFIT family members

  • Preclinical efficacy assessment:

    • Test in immunocompetent syngeneic mouse models

    • Evaluate using patient-derived xenografts in humanized mouse models

    • Combine with existing immunotherapies (checkpoint inhibitors, CAR-T)

    • Monitor tumor growth, survival, and immune infiltration

    • Assess mechanisms through ex vivo analysis of tumor and immune populations

  • Safety and toxicity evaluation:

    • Assess effects on immune response to concurrent infections

    • Evaluate impact on wound healing and inflammatory conditions

    • Monitor for autoimmune-like adverse events

    • Test reversibility of effects after treatment cessation

    • Consider inducible/conditional knockdown systems to evaluate temporal factors

  • Biomarker development:

    • Identify predictive biomarkers for response to IFIT3-targeted therapy

    • Develop companion diagnostics to assess IFIT3 pathway activation

    • Establish pharmacodynamic markers to confirm target engagement

    • Design monitoring protocols for potential immune-related adverse events

When designing these experiments, carefully consider IFIT3's complex biology: it functions as a modulator of interferon signaling, RIG-I-like receptor pathways, and NF-κB signaling . Therapeutic targeting should aim to disrupt tumor-promoting functions while preserving beneficial antiviral and anti-tumor immune surveillance functions. Additionally, given IFIT3's role in antiviral immunity, assessment should include potential risks of increased susceptibility to viral infections .

What are the most informative approaches for studying IFIT3's influence on immune checkpoint regulation?

IFIT3's emerging role in immune checkpoint regulation requires sophisticated experimental approaches that capture both direct and indirect mechanisms. Recent research suggests IFIT3 may influence immune checkpoint molecules and tumor-immune interactions , necessitating multifaceted investigation strategies:

  • Expression correlation analyses in clinical samples:

    • Perform multiplex immunohistochemistry for IFIT3 and checkpoint molecules (PD-L1, PD-1, CTLA-4)

    • Conduct transcriptomic analysis correlating IFIT3 with checkpoint gene expression

    • Stratify by cancer type, immune infiltration patterns, and inflammatory status

    • Analyze public datasets (TCGA, GEO) for broader correlation patterns

  • Mechanistic studies of checkpoint regulation:

    • Generate IFIT3 knockdown and overexpression in tumor cell lines

    • Measure changes in checkpoint molecule expression at mRNA and protein levels

    • Analyze promoter activity using reporter assays

    • Perform ChIP-seq to identify transcription factors affected by IFIT3

    • Investigate post-transcriptional regulation through RNA-binding activities

  • Signaling pathway analysis:

    • Assess NF-κB pathway activation in IFIT3-modified cells

    • Measure STAT1/STAT3 phosphorylation and nuclear translocation

    • Evaluate IRF1/IRF3 activation, which can regulate PD-L1 expression

    • Use pathway inhibitors to determine which signals mediate checkpoint regulation

    • Investigate MAVS-TBK1-IRF3 axis involvement

  • Functional immune interaction assays:

    • Co-culture IFIT3-modified tumor cells with T cells or NK cells

    • Measure T cell activation, proliferation, and cytotoxic function

    • Assess checkpoint receptor-ligand interactions using blocking antibodies

    • Evaluate cytokine production profiles in the co-culture system

    • Compare results between checkpoint inhibitor-sensitive and resistant models

  • In vivo checkpoint blockade response studies:

    • Create tumor models with IFIT3 modulation (knockout or overexpression)

    • Treat with checkpoint inhibitors (anti-PD-1, anti-CTLA-4)

    • Monitor tumor growth, survival, and immune infiltration

    • Perform ex vivo analysis of tumor-infiltrating lymphocytes for exhaustion markers

    • Assess memory T cell formation and durability of responses

When analyzing data, consider that IFIT3's influence may vary between cancer types and immune contexts. The protein's dual role in promoting interferon signaling while potentially supporting tumor growth suggests context-dependent functions that should be carefully delineated . Importantly, IFIT3's interaction with other IFIT family members, particularly IFIT2, may affect its impact on immune checkpoint regulation, warranting investigation of these protein complexes in relation to checkpoint expression .

How should experiments be designed to distinguish between functions of different IFIT family members?

Distinguishing between the functions of different IFIT family members requires careful experimental design due to their structural similarities, potential for heterodimerization, and overlapping but distinct roles. Based on current research, particularly regarding IFIT2 and IFIT3, the following approaches are recommended:

  • Sequential gene knockout strategy:

    • Generate single knockouts for each IFIT family member

    • Create double and triple knockouts in relevant combinations

    • Complement knockout cells with individual IFIT proteins

    • Test functional outcomes in various contexts (viral infection, immune stimulation)

    • This approach revealed that IFIT2 functions in the absence of IFIT3, and IFIT3 functions in the absence of IFIT2, but they have synergistic effects when co-expressed

  • Domain swapping experiments:

    • Create chimeric proteins exchanging domains between IFIT family members

    • Focus on RNA-binding domains and protein interaction interfaces

    • Test functional outcomes to map domain-specific activities

    • This can reveal which protein features determine specific functions

  • Heterodimer-specific analysis:

    • Use proximity-based protein labeling (BioID, APEX) to identify interaction partners

    • Compare interactomes of different IFIT proteins and their heterodimers

    • Develop heterodimer-specific antibodies or biosensors

    • Create forced heterodimers using chemical inducers of dimerization

    • Research shows that IFIT2-IFIT3 heterodimers are thermodynamically preferred and potentially more active than homodimers

  • Comparative RNA-binding studies:

    • Perform RNA immunoprecipitation followed by sequencing for different IFITs

    • Compare binding motifs and RNA targets between family members

    • Analyze the impact of RNA secondary structures on binding specificity

    • RBS-ID identified specific RNA-binding sites in IFIT3 (C239, C283, S360/H361)

  • Temporal expression analysis:

    • Monitor expression kinetics of different IFIT family members after stimulation

    • Analyze protein stability and turnover rates

    • Develop inducible expression systems for temporal control

    • Determine if certain family members prime cells for expression of others

When interpreting results, consider that IFIT proteins form a network of interactions rather than functioning as isolated entities. Their relative expression levels, potential for heterodimerization, and competition for binding partners all influence their functional outcomes . Statistical analysis should include multiple independent biological replicates with appropriate controls for each IFIT family member manipulation.

What experimental methods best reveal the structural basis for IFIT3's functional differences from other IFIT proteins?

Understanding the structural basis for IFIT3's unique functions compared to other IFIT family members requires specialized methods that can elucidate protein structure-function relationships. Current research has relied on structural modeling of IFIT3 based on related proteins, but more direct approaches are needed .

Recommended methodological approaches:

  • Structural determination techniques:

    • X-ray crystallography of IFIT3 alone and in complex with binding partners

    • Cryo-electron microscopy for larger complexes (IFIT3-IFIT2 heterodimers)

    • NMR spectroscopy for dynamic regions and RNA interactions

    • Hydrogen-deuterium exchange mass spectrometry to map flexible regions

    • These approaches would provide direct structural information, surpassing the current reliance on homology modeling using IFIT2 structures

  • Comparative structural analysis:

    • Superimpose IFIT3 structures with other family members

    • Focus on RNA-binding pockets and protein interaction interfaces

    • Map conservation and divergence of key residues

    • Current models predict IFIT3 has a positively charged surface similar to but less extensive than IFIT2

  • Structure-guided mutagenesis:

    • Generate point mutations at structurally significant positions

    • Focus on the conserved RNA-binding sites (C239, C283, S360/H361)

    • Target residues in the predicted basic patch (K249, R252/R253, K286)

    • Create chimeric proteins exchanging structural elements between IFITs

    • Test functional consequences in relevant biological assays

  • Molecular dynamics simulations:

    • Perform comparative simulations of IFIT family members

    • Analyze protein flexibility, conformational changes, and potential energy landscapes

    • Model interactions with RNA and protein partners

    • Identify allosteric communication networks within the protein structure

  • In situ structural analysis:

    • Employ FRET sensors to detect conformational changes in living cells

    • Use protein complementation assays to map interaction interfaces

    • Apply proximity labeling in different cellular compartments

    • These approaches can capture structural dynamics in physiological contexts

When analyzing results, particular attention should be paid to the positively charged regions that likely form RNA-binding surfaces and to the structural features that determine specific protein-protein interactions, especially those mediating IFIT3-IFIT2 heterodimerization . Also important is understanding how RNA binding might induce conformational changes that affect protein function, potentially explaining IFIT3's dual roles in antiviral signaling and viral replication enhancement .

How can I design experiments to investigate potential compensatory mechanisms among IFIT family members?

Investigating compensatory mechanisms among IFIT family members requires sophisticated experimental approaches that can detect functional redundancy, altered expression patterns, and pathway adaptations. Given the structural similarities and partially overlapping functions of IFIT proteins, particularly IFIT2 and IFIT3 , the following experimental design framework is recommended:

  • Combinatorial knockout/knockdown strategy:

    • Generate single, double, and triple IFIT knockouts using CRISPR-Cas9

    • Create inducible knockdown systems for temporal control

    • Compare phenotypes across the knockout series for additive, synergistic, or compensatory effects

    • Measure both expression changes in remaining IFIT members and functional outcomes

    • This approach can reveal if removal of one IFIT leads to upregulation of others

  • Temporal expression profiling:

    • Perform time-course analysis after immune stimulation in various knockout backgrounds

    • Use qRT-PCR, Western blotting, and proteomics to track expression kinetics

    • Compare induction rates, peak expression levels, and protein stability

    • Analyze if expression patterns of remaining IFITs change in knockout contexts

    • This can identify altered regulation that may indicate compensation

  • Interactome rewiring analysis:

    • Conduct immunoprecipitation-mass spectrometry of remaining IFIT members in single knockouts

    • Compare protein interaction networks between wild-type and knockout conditions

    • Identify new interactions that emerge when specific IFITs are absent

    • This approach can reveal how protein complexes reorganize to maintain function

  • Functional rescue experiments:

    • Express individual IFITs in multiple-knockout backgrounds

    • Test which functions can be rescued by which family members

    • Create chimeric proteins combining domains from different IFITs

    • Identify minimal domain requirements for functional compensation

    • Research shows IFIT2 functions in the absence of IFIT3 and vice versa, but with different efficiencies

  • Pathway adaptation analysis:

    • Perform phosphoproteomics and transcriptomics in IFIT knockout backgrounds

    • Identify signaling pathways that show altered activation in response to IFIT depletion

    • Test if alternative pathways become upregulated to compensate for lost IFIT functions

    • Focus on RIG-I, JAK-STAT, and NF-κB pathways known to interact with IFITs

When analyzing data, consider that compensation may be context-dependent, varying between cell types and stimulation conditions. It's also important to distinguish between true functional compensation (where another protein assumes the role of the missing IFIT) and pathway rewiring (where alternative mechanisms achieve similar outcomes through different means). Statistical analysis should include multiple biological replicates and appropriate adjustments for multiple comparisons when screening across pathways .

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.