IRF9 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Function

The IRF9 Antibody is a specialized research tool designed to detect and analyze the Interferon Regulatory Factor 9 (IRF9) protein, a critical transcription factor in type I interferon (IFN) signaling. IRF9 forms part of the ISGF3 complex, which regulates the expression of interferon-stimulated genes (ISGs) during antiviral immune responses. The antibody facilitates the study of IRF9’s role in immune regulation, inflammation, and disease progression .

Applications in Research

The IRF9 Antibody is utilized across multiple experimental platforms:

ApplicationDescriptionSources
Western Blot (WB)Detects IRF9 protein in lysates (e.g., A549, THP-1 cells)
Immunoprecipitation (IP)Isolates IRF9 for downstream analysis (e.g., mouse heart tissue)
Immunofluorescence (IF)Visualizes IRF9 localization in cells (e.g., HepG2 cells)
Immunohistochemistry (IHC)Identifies IRF9 in tissue sections (e.g., human cervical cancer)
Chromatin Immunoprecipitation (ChIP)Maps IRF9 binding to DNA in ISGs

Antibody Structure and Features

Commercially available IRF9 Antibodies vary in specificity and reactivity:

ProductClone/IsotypeReactivityMolecular Weight
Cell Signaling #28492Rabbit IgG (D8G7H)Human48 kDa
Cell Signaling #76684Rabbit IgG (D2T8M)Human48 kDa
Proteintech 14167-1-APRabbit IgGHuman, Mouse, Rat44–48 kDa
Abcam ab271043Rabbit IgG (EPR24260-55)Human48 kDa

IRF9 Regulation and Disease Relevance

IRF9’s activity is modulated by post-translational modifications and microRNAs:

  • Phosphorylation: IRF9 lacks autoinhibitory domains, but its interaction with STAT2 is critical for ISGF3 formation .

  • Acetylation: CREB-binding protein (CBP) acetylates IRF9 at Lys81, enhancing DNA binding .

  • miRNA Regulation: miR-93 and miR-302d inhibit IRF9 expression, impacting angiogenesis and lupus-like autoimmunity .

Disease Implications:

  • Systemic Lupus Erythematosus (SLE): IRF9 drives IgG autoantibody production and TLR7 activation in B cells .

  • COVID-19: SARS-CoV-2 Spike protein suppresses IRF9 via exosomal miRNAs, modulating pro-inflammatory pathways .

  • Cardiovascular Disease: IRF9 overexpression promotes neointima formation in injured arteries .

Research Findings and Clinical Implications

  • Antiviral Defense: IRF9 is antagonized by viral proteins (e.g., HPV16 E7, SVV ORF63), highlighting its role in immune evasion .

  • Therapeutic Targeting: Inhibiting IRF9 may reduce autoimmune inflammation while preserving antiviral functions .

Product Specs

Buffer
Phosphate-buffered saline (PBS) with 0.02% sodium azide, 50% glycerol, pH 7.3. Store at -20°C. Avoid repeated freeze-thaw cycles.
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your orders. Delivery times may vary depending on the purchase method or location. Please consult your local distributors for specific delivery timeframes.
Synonyms
IFN alpha responsive transcription factor subunit antibody; IFN-alpha-responsive transcription factor subunit antibody; Interferon regulatory factor 9 antibody; interferon stimulated transcription factor 3 antibody; Interferon-stimulated gene factor 3 gamma antibody; interferon-stimulated transcription factor 3, gamma 48kDa antibody; IRF 9 antibody; IRF-9 antibody; Irf9 antibody; IRF9_HUMAN antibody; ISGF 3 gamma antibody; ISGF-3 gamma antibody; ISGF3 antibody; ISGF3 p48 subunit antibody; ISGF3G antibody; OTTHUMP00000164692 antibody; OTTHUMP00000164693 antibody; p48 antibody; Transcriptional regulator ISGF3 subunit gamma antibody
Target Names
Uniprot No.

Target Background

Function
IRF9 is a transcription factor that plays a vital role in antiviral immunity. It mediates signaling by type I interferons (IFN-α and IFN-β). Upon type I IFN binding to cell surface receptors, Jak kinases (TYK2 and JAK1) are activated, leading to tyrosine phosphorylation of STAT1 and STAT2. IRF9/ISGF3G associates with the phosphorylated STAT1:STAT2 dimer, forming a complex termed ISGF3 transcription factor, which then translocates to the nucleus. ISGF3 binds to the interferon-stimulated response element (ISRE) to activate the transcription of interferon-stimulated genes, ultimately driving the cell into an antiviral state.
Gene References Into Functions
  1. Priming cells with IFNβ synergistically enhances IL6 induction in response to treatments that activate NF-κB, in a process dependent on the recruitment of STAT2 and IRF9. PMID: 29581268
  2. Surface features in the interacting domains of IRF9 and STAT2 have diverged to enable specific interaction between these family members and facilitate the antiviral response. PMID: 29317535
  3. Recent research has revealed a unique role for IRF9 as a conductor of cellular responses to IFN-Is. Interestingly, novel functions for IRF9 beyond the antiviral response are also being identified. PMID: 26987614
  4. These findings highlight miR-302d as a key regulator of type I IFN-driven gene expression through its ability to target IRF9 and regulate ISG expression, emphasizing the significance of non-coding RNA in modulating the IFN pathway in systemic lupus erythematosus (SLE). PMID: 28318807
  5. Decreased IRF9 expression was associated with increased replication of hepatitis C virus and hepatitis E virus. PMID: 28442624
  6. PKV VP3 associated with STAT2 and IRF9, interfering with the formation of the STAT2-IRF9 and STAT2-STAT2 complex. PMID: 28441586
  7. Interferon-stimulated gene factor 3 (ISGF3) complex, composed of STAT1, STAT2, and IRF9, is necessary for the induction of SAMHD1 expression by IFN-α in SMMC-7721 cells. PMID: 26397446
  8. U-ISGF3 induced by IFN-λs and IFN-β drives prolonged expression of a set of IFN-stimulated genes during HCV infection. PMID: 26216956
  9. IRF9 is a vascular injury-response molecule that promotes vascular smooth muscle cell proliferation. IRF9 expression is upregulated during neointima formation. PMID: 25319116
  10. DC-SIGN-induced ISGF3 by fucose-based pathogen-associated molecular patterns (PAMPs) plays a crucial role in driving IL-27 production and subsequent T follicular helper (TFH) polarization, which could be harnessed for vaccine design. PMID: 25278262
  11. IRF9 mediated myocardial reperfusion injury. PMID: 25150882
  12. STAT2 and IRF9 overexpression is sufficient to drive interferon-related DNA damage signature expression upon cell crowding. PMID: 25156627
  13. IL6 is an inducer of IRF9 expression in prostate cancer and a sensitizer for the antiproliferative effects of IFNα2. PMID: 23913484
  14. The hepatitis C virus (HCV) non-structural 5A (NS5A) protein, known to modulate the IFN response, competes with IRF9 for cyclophilin A (CypA) binding and can prevent the formation of IRF9-CypA complexes. PMID: 22902549
  15. Histone deacetylases 1 and 2 (HDAC1 and HDAC2) differentially modulate STAT activity in response to IFNα2: while they are required for the induction of ISGF3-responsive genes, they impair the transcription of STAT3-dependent genes. PMID: 21957129
  16. Western blot and electrophoretic mobility-shift assays identified the interferon-stimulated gene factor-3 (ISGF-3) components STAT1 and IRF-9 as the proximal targets of human herpesvirus 8 vIRF-2 activity. PMID: 21697347
  17. STAT2 may interact with IRF-9 in a STAT1-independent manner. The complex STAT2/IRF-9 is the key factor mediating the expression of RIG-G gene regulated by IFN-α. PMID: 20403236
  18. Signals induced by IFN-α and IL-4 induce cytoplasmic sequestration of IL-4-activated STAT6 and IFN-α-activated STAT2:p48 in B cells through the formation of pY-STAT6:pY-STAT2:p48 complex. PMID: 21268015
  19. Analysis of interferon-stimulated response elements (ISREs) that bind to both the IFN-stimulated gene factor 3 (ISGF3) as well as to IFN response factor 7 (IRF7). PMID: 20943654
  20. Results suggest that the amount of cellular IRF9 is a crucial determinant for amplification of early dynamics of IFNα-mediated signal transduction. PMID: 20964804
  21. A key factor for eliciting the antiproliferative activity of IFN-α in tumors. PMID: 19752753
  22. NOD1 can activate the ISGF3 signaling pathway, typically associated with protection against viral infection, to provide robust type I IFN-mediated protection from *H. pylori* and possibly other mucosal infections. PMID: 20389019
  23. The IFN-activated ISGF3 transcription factor regulates transcription through contact with DRIP150. PMID: 12509459
  24. IRF9 functions to recruit RNA polymerase II to the promoter of interferon-stimulated genes and requires histone deacetylases. PMID: 15194680
  25. The conserved DNA-binding domain of STAT2 has a role specific to the activity of ISGF3-independent STAT2-containing complexes. PMID: 15668228
  26. Defects in ISGF3 can cause resistance to IFN-α(2a) treatment. PMID: 15714000
  27. GBF1 is recruited to the endogenous IRF-9 promoter and interacts with C/EBP-β, IL-1, and IL-6. PMID: 16318580
  28. Pretreatment of Huh-7 cells with 0.5-1 mM H2O2 resulted in the suppression of the IFN-α-induced antiviral protein MxA and of IRF-9 mRNA expression. PMID: 16595158
  29. Data reveal the existence of a collection of GAS-regulated target genes whose expression is interferon-inducible and independent of ISGF3 but highly dependent on the STAT2 DNA binding domain. PMID: 16689942
  30. Results identify Viperin as a tightly regulated ISGF3 target gene, which is counter-regulated by PRDI-BF1. PMID: 16849320
  31. Suggest that the JAK-STAT pathway may play a major role in mediating the effects of IFN-α against hepatitis B virus, and that ISGF3 might be a key factor. PMID: 17559358
  32. These data define the role of the ISGF3 members in IFN-β inhibitory signaling. PMID: 18370868
  33. The data suggest that liberation of the IFNaR2-ICD by regulated proteolysis could trigger a novel mechanism for moving the transcription factor Stat2 to the nucleus. PMID: 18456457

Show More

Hide All

Database Links

HGNC: 6131

OMIM: 147574

KEGG: hsa:10379

STRING: 9606.ENSP00000380073

UniGene: Hs.1706

Protein Families
IRF family
Subcellular Location
Cytoplasm. Nucleus. Note=Translocated into the nucleus upon activation by IFN-alpha/beta.

Q&A

What is IRF9 and what are its alternative names in the literature?

IRF9 (Interferon regulatory factor 9) functions as a critical transcription factor in the interferon signaling pathway. In the scientific literature, it appears under several alternative names which can complicate literature searches and data interpretation:

  • IRF-9

  • ISGF3 (Interferon-stimulated gene factor 3)

  • ISGF3G

  • p48

  • IFN-alpha-responsive transcription factor subunit

  • ISGF-3 gamma

The protein has a molecular weight of approximately 43.7 kilodaltons and can be found across multiple species with orthologs in canine, porcine, monkey, mouse, and rat models . When designing experiments or searching literature, it's essential to account for all these nomenclature variations to ensure comprehensive data collection.

What are the common applications for IRF9 antibodies in research?

IRF9 antibodies are employed across numerous experimental techniques in immunology, cell biology, and cancer research. Based on currently available commercial antibodies, the primary applications include:

ApplicationCommon UsageSample PreparationTypical Dilution Ranges
Western Blot (WB)Protein expression quantificationCell/tissue lysate1:500-1:2000
Immunohistochemistry (IHC)Tissue localization studiesFFPE or frozen sections1:100-1:500
Immunofluorescence (IF)Subcellular localizationFixed cells1:100-1:400
Flow Cytometry (FCM)Cellular expression levelsSingle-cell suspensions1:50-1:200
ELISAQuantitative detectionSerum, plasma, culture media1:1000-1:5000

When selecting an IRF9 antibody, it's crucial to validate its reactivity with your species of interest, as antibodies demonstrate varying cross-reactivity across human, mouse, and other models . For optimal results, antibody validation using positive and negative controls is strongly recommended.

How do I optimize Western blot protocols for IRF9 detection?

Optimizing Western blot protocols for IRF9 detection requires attention to several critical factors:

  • Sample Preparation: For optimal IRF9 detection, lyse cells in RIPA buffer supplemented with protease inhibitors. Type I IFN stimulation (1000 U/ml for 12-24 hours) can serve as a positive control by upregulating IRF9 expression.

  • Gel Selection: Use 10-12% polyacrylamide gels for optimal resolution of the 43.7 kDa IRF9 protein.

  • Transfer Conditions: Semi-dry transfer at 15V for 30 minutes or wet transfer at 100V for 1 hour typically yields good results for IRF9.

  • Blocking and Antibody Incubation:

    • Block with 5% non-fat milk in TBST for 1 hour at room temperature

    • Primary antibody dilution: typically 1:1000 in blocking buffer (overnight at 4°C)

    • Secondary antibody dilution: typically 1:5000 for 1 hour at room temperature

  • Troubleshooting:

    • High background: Increase blocking time or use 5% BSA instead of milk

    • Weak signal: Increase primary antibody concentration or extend incubation time

    • Non-specific bands: Verify antibody specificity using IRF9 knockout cells or IRF9-depleted lysates as negative controls

Remember that phosphorylation status can affect IRF9 mobility on SDS-PAGE, potentially resulting in bands at approximately 48-50 kDa rather than the predicted 43.7 kDa.

What controls should I include when using IRF9 antibodies in my experiments?

Proper experimental controls are essential for valid interpretation of results when working with IRF9 antibodies:

Positive Controls:

  • IFN-α/β treated cells (1000 U/ml for 12-24 hours) to upregulate IRF9 expression

  • Cell lines known to express high levels of IRF9 (e.g., certain breast cancer cell lines like MCF-7)

  • Recombinant IRF9 protein (for calibration curves or as Western blot standards)

Negative Controls:

  • IRF9 knockout or knockdown cells (using CRISPR-Cas9 or siRNA)

  • Cell lines with naturally low IRF9 expression

  • Secondary antibody-only controls to assess non-specific binding

  • Isotype controls for flow cytometry or immunohistochemistry applications

Specificity Controls:

  • Antibody neutralization with blocking peptides

  • Multiple antibodies targeting different IRF9 epitopes to confirm specificity

  • Cross-validation using different detection techniques (e.g., confirm Western blot findings with immunofluorescence)

Implementing these controls helps ensure that observed signals genuinely represent IRF9 rather than experimental artifacts or cross-reactivity with related IRF family proteins.

How can I distinguish between different IRF family members when studying signaling pathways?

Distinguishing between IRF family members presents a significant challenge due to structural similarities and shared DNA-binding domains. To accurately characterize IRF9-specific functions:

  • Antibody Selection: Choose antibodies raised against unique regions of IRF9 rather than conserved domains. Antibodies targeting the C-terminal region of IRF9 typically offer higher specificity compared to those targeting the N-terminal DNA-binding domain shared with other IRF family members.

  • Sequential Immunoprecipitation: For co-immunoprecipitation studies, use sequential immunoprecipitation with antibodies against different components of the ISGF3 complex (IRF9, STAT1, STAT2) to confirm specific interactions.

  • Chromatin Binding Profiles: IRF9-dominant regions contain both ISRE and GAS motifs, often without other motifs typically associated with IRF3 or IRF5. This distinctive binding pattern can differentiate IRF9 activity from other IRF proteins .

  • Functional Validation: IRF9 uniquely forms the ISGF3 complex with STAT1 and STAT2 in response to type I IFNs. Detecting ISGF3 formation through co-immunoprecipitation or proximity ligation assays can distinguish IRF9-specific signaling from other IRF pathways.

  • Genetic Approaches: Use IRF9-specific knockouts or knockdowns while monitoring the expression of unique downstream targets (see comparative table below).

IRF MemberKey Target GenesAssociated StimuliComplex Formation
IRF9ISG15, IFIT1, MX1Type I IFNsISGF3 (with STAT1/STAT2)
IRF3IFN-β, CXCL10Viral infection, TLR3/4IRF3 homodimers
IRF5IL-6, TNF-α, IL-12TLR7/8/9, virusIRF5 homodimers

This integrated approach enables the specific attribution of signaling events to IRF9 rather than other IRF family members in complex experimental systems .

What role does IRF9 play in autoimmune disease models, and how can IRF9 antibodies help study these mechanisms?

IRF9 plays a critical role in autoimmune disease pathogenesis, particularly in systemic lupus erythematosus (SLE). Strategic application of IRF9 antibodies can elucidate these mechanisms:

  • Autoantibody Production: Studies using the pristane-induced mouse model of SLE demonstrate that IRF9 is required for IgG autoantibody production against nucleic acid-associated antigens. IRF9-deficient mice develop high titers of IgM autoantibodies but significantly reduced IgG autoantibodies, suggesting IRF9's critical role in isotype switching .

  • B Cell Activation: IRF9-deficient B cells show impaired activation through TLR7, a key receptor recognizing single-stranded RNA. This defect links IRF9 to B cell activation in autoimmunity, which can be studied using IRF9 antibodies in conjunction with B cell activation markers .

  • Analysis Techniques Using IRF9 Antibodies:

    • ChIP-seq to map IRF9 binding sites on autoimmunity-related gene promoters

    • Co-immunoprecipitation to identify IRF9 interaction partners in autoimmune models

    • Immunohistochemistry to examine IRF9 expression in lymphoid organs during disease progression

    • Phospho-specific antibodies to track IRF9 activation status

  • IFN Signaling Integration: IRF9 appears to function upstream of TLR signaling in autoimmune B cell activation, positioning the IFN-I pathway as a critical mediator in autoimmunity. Multi-parameter flow cytometry using antibodies against IRF9, STAT1, and TLR7 can map this signaling cascade in various immune cell populations .

The table below summarizes key findings from pristane-induced SLE model comparing wild-type and IRF9-deficient mice:

ParameterWild-type MiceIRF9^(-/-) MiceSignificance
IgG autoantibodies (Sm/RNP)High levelsSignificantly reducedp < 0.001
IgM autoantibodies (Sm/RNP)Moderate levelsSignificantly increasedp < 0.01
TLR7 expression after IFN-αUpregulatedGreatly reducedp < 0.001
B cell activation via TLR7NormalDefectivep < 0.001

These findings highlight IRF9 as a potential therapeutic target in SLE and other autoimmune diseases characterized by excessive type I IFN signaling .

How does IRF9 overexpression influence drug resistance in cancer, and what experimental approaches can investigate this phenomenon?

IRF9 overexpression has been implicated in drug resistance, particularly to antimicrotubule agents in breast cancer. This emerging role can be investigated through several experimental approaches:

  • Expression Correlation Studies: Approximately half of breast and uterine tumors show IRF9 overexpression, suggesting its importance in these cancer types. Immunohistochemistry with IRF9 antibodies on tissue microarrays can establish correlations between IRF9 expression levels and clinical outcomes, including drug response patterns .

  • Mechanistic Analysis: Transient overexpression of IRF9 in breast adenocarcinoma cells (MCF-7) induces resistance to paclitaxel (13-fold) and vinblastine (3.3-fold), but not to doxorubicin. This resistance occurs through an IFN-independent mechanism, as neither Stat1/Stat2 overexpression nor IFNα treatment reproduces this effect .

  • Downstream Pathway Identification: IRF9 overexpression induces expression of several IFN-responsive genes, including Stat1, Stat2, and MHC class I, but not IRF1. This selective gene activation pattern differs from classical IFN responses and can be mapped using:

    • RT-qPCR for transcript analysis

    • Western blotting with antibodies against pathway components

    • ChIP-seq to identify direct IRF9 binding targets in resistant cells

  • Experimental Models: Several approaches can investigate IRF9-mediated drug resistance:

Experimental ApproachKey MethodsExpected OutcomesTechnical Considerations
Transient IRF9 overexpressionTransfection with IRF9 expression vectorDrug resistance, selective gene inductionUse appropriate vector controls
Stable IRF9 knockdownshRNA/CRISPR in resistant cellsRestored drug sensitivityConfirm knockdown efficiency
Pharmacological inhibitionSmall molecule inhibitors of IRF9 or downstream targetsReversal of resistance phenotypeValidate target specificity
Clinical sample correlationTissue microarrays, IHC for IRF9Correlation with treatment responseUse standardized scoring systems
  • Resistance Measurements: To quantify IRF9-mediated resistance, researchers can compare IC50 values between IRF9-overexpressing and control cells. For example, paclitaxel resistance increased 13-fold in IRF9-transfected MCF-7 cells compared to vector controls .

These findings reveal a novel role for IRF9 as a potential biomarker for predicting response to antimicrotubule agents in breast cancer and suggest targeting IRF9 as a strategy to overcome drug resistance.

What are the optimal approaches for studying IRF9 binding to DNA and chromatin in different cellular contexts?

Studying IRF9 binding to DNA and chromatin requires specialized techniques that can capture both direct binding events and contextual influences:

  • ChIP-seq Analysis for IRF9 Binding Patterns:

    • Use high-quality ChIP-grade IRF9 antibodies (validate specificity with IRF9-deficient cells)

    • Optimal crosslinking: 1% formaldehyde for 10 minutes at room temperature

    • Sonication conditions: typically 10-12 cycles (30s ON/30s OFF) to achieve 200-500bp fragments

    • Include input controls and IgG controls to filter non-specific binding

    • For unbiased analysis, integrate with ATAC-seq to correlate binding with chromatin accessibility

  • Motif Analysis for IRF9 Binding Specificity:

    • IRF9-dominant regions frequently contain both ISRE (Interferon-Stimulated Response Element) and GAS (Gamma-Activated Sequence) motifs

    • The consensus sequence 5′-WBVGGAAANNGAAACT-3′ and its variants are enriched in IRF9-dominant clusters

    • GAS motif enrichment suggests that STAT1 homodimers activated by type I IFN signaling are important determinants for IRF9-dominant binding

  • Integrated Analysis Approaches:

    • Combine ChIP-seq with RNA-seq to correlate binding events with transcriptional outcomes

    • Use sequential ChIP (Re-ChIP) to identify regions where IRF9 co-binds with STAT1 and STAT2

    • Employ CRISPR-based techniques to functionally validate the importance of identified binding sites

  • Differential Binding Analysis:

    • Compare IRF9 binding profiles across different:

      • Cell types (immune cells vs. cancer cells)

      • Stimulation conditions (IFN-treated vs. untreated)

      • Disease states (normal vs. autoimmune or cancer models)

  • Advanced Technologies:

    • Cut&Run or CUT&Tag for higher resolution and lower background compared to traditional ChIP

    • HiChIP to identify long-range interactions between IRF9-bound enhancers and target promoters

    • Single-cell approaches to resolve cell-to-cell variability in IRF9 binding

These approaches enable comprehensive characterization of IRF9's genomic binding landscape and its context-dependent functions in different cellular states and disease models .

How can I design experiments to differentiate between IFN-dependent and IFN-independent functions of IRF9?

Differentiating between IFN-dependent and IFN-independent functions of IRF9 requires strategic experimental design:

  • Cell System Selection:

    • Use IFNAR1/2 knockout cells to eliminate type I IFN receptor signaling

    • Compare with IRF9 knockout cells to identify differential phenotypes

    • Consider U5A cells (IFNAR2-deficient) as established research tools for IFN-independent studies

  • IRF9 Mutant Analysis:

    • Generate IRF9 constructs with mutations in domains required for STAT interaction

    • Create IRF9 variants with altered nuclear localization signals

    • Test these constructs in rescue experiments with IRF9-deficient cells

  • Temporal Analysis:

    • Compare acute vs. chronic effects of IRF9 expression

    • In cancer drug resistance studies, IRF9 overexpression confers resistance to antimicrotubule agents through mechanisms distinct from those activated by IFNα treatment

  • Pathway Validation:

    • Monitor expression of downstream genes using RT-qPCR or RNA-seq

    • Compare IRF9-induced gene expression profiles with those induced by IFN treatment

    • Use statistical approaches (principal component analysis, hierarchical clustering) to identify unique gene signatures

  • Combined Interventions:

    • Compare IRF9 overexpression alone, IFN treatment alone, and combined conditions

    • Test if IFN neutralizing antibodies block IRF9-dependent phenotypes

The table below highlights differences observed between IFN-dependent and IFN-independent effects in a breast cancer model:

ParameterIFNα TreatmentIRF9 OverexpressionStat1/Stat2 Overexpression
MHC Class I expressionIncreasedIncreasedModerately increased
IRF1 expressionIncreasedUnchangedNot determined
Paclitaxel resistanceNo effect13-fold resistanceNo effect
Vinblastine resistanceNo effect3.3-fold resistanceNo effect
Doxorubicin resistanceNo effectNo effectNo effect

This comparative approach clearly demonstrates that IRF9 can mediate distinct cellular effects independent of the classical IFN signaling pathway, particularly in the context of cancer drug resistance .

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.