IRF1 Human

IFN Regulatory Factor-1 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Introduction to IRF1

Interferon Regulatory Factor 1 (IRF1) is a transcription factor encoded by the IRF1 gene on chromosome 5q31.1. It plays pivotal roles in innate and adaptive immunity, tumor suppression, and cellular stress responses . As a member of the IRF family, IRF1 regulates interferon (IFN) and IFN-stimulated gene (ISG) expression by binding to IFN-stimulated response elements (ISREs) . Its dysregulation is implicated in cancers, immune disorders, and viral pathogenesis .

Genomic Features

  • Chromosomal Location: 5q31.1

  • Gene Length: 9,165 base pairs (10 exons, 9 introns) .

  • Splicing Variants: Constitutively expressed isoform 1 (325 amino acids; 45 kDa) is the predominant functional variant .

Protein Domains

  • DNA-Binding Domain (DBD): N-terminal helix-turn-helix motif (amino acids 1–120) for ISRE recognition .

  • Interferon Association Domain (IAD): C-terminal region (IAD2 subtype) mediating interactions with transcription factors .

Table 2: IRF1 in Cancer Biology

Cancer TypeIRF1 RoleKey Findings
Colorectal Cancer (CRC)Tumor suppressorDownregulated in tumors; upregulation inhibits proliferation, enhances radiosensitivity .
LeukemiaRegulator of HSC self-renewalIrf1−/− mice show impaired hematopoietic stem cell (HSC) function and leukemogenesis .
Gastric/Lung CancerPrognostic markerLow IRF1 correlates with poor survival; mutations linked to oncogenesis .

Immune Dysregulation

  • Viral Infections: IRF1 is critical for IFN-λ1 and IFN-β induction in HMPV-infected macrophages . Knockdown reduces antiviral responses by 80% .

  • Autoimmunity: Polymorphisms associate with susceptibility to autoimmune diseases via dysregulated IFN signaling .

Cancer Therapeutics

  • CRC Models: IRF1 overexpression in SW480/CCL244 cells reduces tumor growth by 60% in vivo and increases apoptosis via Bcl-2 downregulation .

  • Radiosensitization: IRF1 upregulation enhances CRC cell sensitivity to X-ray irradiation (2-fold increase in apoptosis) .

Table 3: IRF1-Modulated Genes in CRC

GeneFunctionFold Change (IRF1 Overexpression)
IFI6Apoptosis regulation+3.5
IFITM1Viral restriction+2.8
IFI35Innate immune signaling+2.6

Regulatory Networks and Interactions

  • Protein Partners: STAT1, NF-κB, p53, IRF8, and PCAF .

  • Post-Translational Regulation:

    • Phosphorylation by TBK1/IKKε enhances nuclear translocation .

    • SUMOylation at Lys275/299 and ubiquitination modulate stability .

Table 4: IRF1 Interaction Partners

InteractorRole in IRF1 Function
STAT1Amplifies ISG expression
p53Synergizes in apoptosis and DNA repair
TBK1Phosphorylates IRF1 for antiviral signaling

Diagnostic and Therapeutic Potential

  • Biomarker: High IRF1 expression in CRC correlates with 5-year survival rates (70% vs. 40% in low-IRF1 patients) .

  • Therapeutic Strategies: Small-molecule agonists (e.g., IFN-γ) or gene therapy to restore IRF1 in cancers .

Product Specs

Introduction
IRF1, short for IFN regulatory factor 1, plays a crucial role in regulating gene expression vital for immune response, hematopoiesis, and proliferation. As a transcription factor, IRF1 activates genes stimulated by IFN-A, IFN-B, and IFN-G. Additionally, it acts as a tumor suppressor, triggering apoptosis in tumorigenic cell lines.
Description
Recombinant human IRF1, produced in E. coli, is a single, non-glycosylated polypeptide chain comprising 134 amino acids (residues 1-114). It includes a 20-amino acid His tag and has a molecular weight of 15 kDa. The purification of IRF1 is achieved through proprietary chromatographic techniques.
Physical Appearance
Clear, colorless solution that has been sterilized by filtration.
Formulation
The product is supplied at a concentration of 1 mg/ml in a buffer consisting of 20mM Tris (pH 8) and 10% glycerol.
Stability
Liquid IRF1 remains stable at 10°C for up to one week; however, it is recommended to store it below -18°C for optimal long-term stability. Adding a carrier protein like 0.1% HSA or BSA is advised for extended storage. Avoid repeated freeze-thaw cycles.
Purity
The purity of the protein is greater than 90%, as determined by reverse-phase high-performance liquid chromatography (RP-HPLC) and sodium dodecyl-sulfate polyacrylamide gel electrophoresis (SDS-PAGE).
Synonyms
IRF-1, IRF1, MAR.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MPITRMRMRP WLEMQINSNQ IPGLIWINKE EMIFQIPWKHAAKHGWDINK DACLFRSWAI HTGRYKAGEK EPDPKTWKAN FRCAMNSLPD IEEVKDQSRN KGSSAVRVYR MLPP.

Q&A

What is IRF1 and what is its primary function in human cells?

IRF1 is a transcription factor first identified as a positive regulator of interferon-beta (IFN-β). It plays fundamental roles in immune response regulation, cell development, and function. In human cells, IRF1 primarily regulates the transcription of genes involved in antimicrobial defense, inflammasome activation, and antiviral responses . IRF1 contains a DNA-binding domain that recognizes specific interferon-stimulated response elements (ISREs) in target gene promoters, enabling transcriptional activation of various immune-related genes including inducible nitric oxide synthase (iNOS), immune-responsive gene 1 (IRG1), guanylate binding proteins (GBPs), and certain interleukins . In macrophages, baseline IRF1 nuclear localization occurs in approximately 20% of unstimulated cells, which increases significantly upon pathogen challenge .

How is IRF1 expression regulated at the transcriptional level?

IRF1 expression is tightly regulated at the transcriptional level through multiple mechanisms. It is potently induced by interferons (particularly IFN-γ) and other cytokines including TNF-α, IL-1β, and IL-6 . Additionally, pattern recognition receptors (PRRs) such as Toll-like receptors (TLRs) can trigger IRF1 expression in a cell- and context-dependent manner . The regulation involves a complex interplay of signaling cascades:

  • Cytokine-mediated induction: TNF-α and IL-6 autocrine/paracrine signaling contributes significantly to IRF1 activation

  • Pathogen-sensing pathways: TLR7/8 sensing of microbial components leads to IRF1 induction

  • Kinase pathways: IKKβ inhibition reduces IRF1 activation, demonstrating involvement of this pathway

In viral infections such as Human metapneumovirus (HMPV), IRF1 expression increases markedly from 1-3 hours post-infection and continues rising through 18 hours, correlating with viral replication kinetics .

What experimental methods should researchers use to measure IRF1 activation?

To comprehensively assess IRF1 activation, researchers should implement a multi-parameter approach:

  • Nuclear translocation assays: Since IRF1 functions primarily as a nuclear transcription factor, quantifying its nuclear localization by immunofluorescence microscopy provides direct evidence of activation. Approximately 20% of uninfected macrophages show nuclear IRF1, increasing significantly upon infection .

  • mRNA expression analysis: Quantitative PCR to measure IRF1 transcript levels, using appropriate housekeeping genes for normalization.

  • Protein expression analysis: Western blotting to detect total IRF1 protein levels and phosphorylated IRF1.

  • Chromatin immunoprecipitation (ChIP): To assess IRF1 binding to promoter regions of target genes.

  • Reporter assays: Using constructs containing IRF1-responsive elements linked to reporter genes.

For statistical robustness, researchers should perform 3-5 independent biological experiments and apply appropriate statistical tests (Student's t-test for single comparisons or two-way ANOVA with post hoc Tukey's test for multiple comparisons) .

How does IRF1 contribute to antimicrobial defense mechanisms?

IRF1 orchestrates multiple antimicrobial defense mechanisms in human cells, particularly in macrophages responding to pathogen challenge:

  • Regulation of IRG1 expression: IRF1 controls the expression of Immune-Responsive Gene 1 (IRG1), a mitochondrial enzyme that produces itaconate, which has both antimicrobial and immunoregulatory activities .

  • Antimicrobial metabolite production: Through IRG1 induction, IRF1 contributes to the production of itaconate, which can inhibit bacterial isocitrate lyases, methylcitrate lyase, or methylmalonyl-CoA mutase, thereby disrupting metabolic pathways required for intracellular pathogen growth .

  • Coordination of bystander cell responses: IRF1 activation occurs not only in directly infected cells but also in uninfected bystander cells, creating a reinforced defense network through paracrine signaling .

Experimental evidence demonstrates that siRNA-mediated knockdown of IRF1 in human primary macrophages results in a significant 45% increase in intracellular Mycobacterium avium burden compared to control cells . Similarly, silencing IRG1, which is downstream of IRF1, leads to a 70% increase in bacterial burden, highlighting the critical role of this pathway in controlling infection .

What is the relationship between IRF1 and cytokine signaling networks?

IRF1 has a bidirectional relationship with cytokine signaling networks:

  • Cytokine-mediated IRF1 activation: TNF-α and IL-6 signaling pathways are crucial for full activation of IRF1 in both directly infected macrophages and bystander cells . Treatment with recombinant TNF-α or IL-6 (25 nM) independently induces IRG1 expression, demonstrating that these cytokines can activate the IRF1-IRG1 axis .

  • IRF1-regulated cytokine production: IRF1 regulates the expression of various inflammatory cytokines, including IL-12, contributing to the amplification of immune responses.

The importance of this relationship is evidenced by experiments using cytokine blockade: treatment with infliximab (TNF-α blocker) or tocilizumab (IL-6R blocker) significantly reduces IRF1 activation and subsequent IRG1 expression by approximately 32% . This mechanism helps explain clinical observations that patients on anti-inflammatory treatments targeting these cytokines may have increased susceptibility to mycobacterial infections .

TreatmentEffect on IRF1 Nuclear LocalizationEffect on IRG1 ExpressionImpact on Pathogen Control
ControlBaseline (~20% in uninfected cells)BaselineBaseline
M. avium infectionSignificant increase45% reduction with IRF1 knockdownBaseline
TNF-α blockadeReduced activation~32% reductionCompromised
IL-6R blockadeReduced activation~32% reductionCompromised
IRF1 siRNA75% reduction45% decrease45% increase in bacterial burden
IRG1 siRNANot directly affected80% reduction70% increase in bacterial burden

How do viral infections specifically trigger IRF1 activation?

Viral infections trigger IRF1 activation through distinct mechanisms that differ from bacterial recognition pathways:

  • RIG-I-like receptor (RLR) engagement: Viruses like Human metapneumovirus (HMPV) are recognized by RLRs such as RIG-I and MDA5, which can initiate IRF1 induction .

  • Toll-like receptor activation: TLR3 (recognizing double-stranded RNA) and TLR7 (recognizing single-stranded RNA) contribute to IRF1 induction during viral infections .

  • TBK1-mediated signaling: TANK-Binding Kinase 1 (TBK1) plays a critical role in HMPV-stimulated induction of IRF1 in human monocyte-derived macrophages through a mechanism involving NF-κB activation and type I IFN-mediated STAT1 activation .

In HMPV infection of human macrophages, IRF1 induction correlates with viral replication kinetics, with expression detectable as early as 1-3 hours post-infection and increasing markedly up to 18 hours post-infection . Comparative analyses show that viral induction patterns differ from other stimuli: LPS induces IRF1 more potently than HMPV, while polyIC (a synthetic double-stranded RNA) induces IRF1 to similar levels as HMPV .

What is the role of IRF1 in regulating metabolic pathways during infection?

IRF1 plays a sophisticated role in modulating cellular metabolism during infection, particularly through its control of mitochondrial function and metabolite production:

The antimicrobial mechanisms of IRF1-regulated metabolites like itaconate operate through inhibition of bacterial enzymes critical for survival when intraphagosomal pathogens rely on fatty acids as carbon sources, including isocitrate lyases, methylcitrate lyase, and methylmalonyl-CoA mutase . This metabolic interference represents an evolutionarily conserved antimicrobial strategy that doesn't rely on direct toxicity.

How does IRF1 activation in bystander cells contribute to immune responses?

IRF1 activation in uninfected bystander cells represents a sophisticated amplification mechanism in the immune response:

  • Paracrine signaling: Uninfected bystander cells actively contribute to infection resolution by producing IL-6 and TNF-α, which activate the IRF1/IRG1 pathway in neighboring infected cells through paracrine signaling .

  • Enhanced antimicrobial activity: This bystander activation strengthens the antimicrobial activity of infected macrophages, creating a reinforced defense network .

  • Spatial distribution of resistance: During M. avium infection, approximately 30% of infected macrophages and 20% of bystander macrophages express IRG1 mRNA at 5 hours post-infection, demonstrating the distributed nature of this response .

This distributed response system helps explain why broad anti-inflammatory treatments that inhibit cytokine signaling (like tocilizumab or infliximab) may increase susceptibility to mycobacterial infections by disrupting the communication between infected and uninfected cells that amplifies antimicrobial mechanisms .

What contradictions exist in the literature regarding IRF1 function?

Several areas of contradiction or uncertainty exist in the current understanding of IRF1 function:

  • Cell-type specificity: The contribution of various pattern recognition receptors (PRRs) to IRF1 activation varies by cell type, leading to apparently contradictory findings when different experimental systems are used .

  • Species differences: While mouse models have demonstrated strong iNOS-driven production of nitric oxide as an important part of antimycobacterial defense dependent on IRF1, human macrophages do not produce this robust NO response in vitro, suggesting species-specific differences in IRF1 effector functions .

  • Itaconate levels: Despite significant IRG1 induction in M. avium-infected macrophages, only low levels of itaconate were detected, creating an apparent contradiction that may be explained by directed delivery mechanisms or rapid metabolism of the compound .

  • Interferon dependency: While IRF1 is strongly associated with interferon responses in many contexts, some clinical M. avium strains are poor inducers of interferons in human primary macrophages, suggesting alternative functions of IRF1 may be important for antimycobacterial defenses in these scenarios .

These contradictions highlight the complexity of IRF1 biology and the need for context-specific research approaches that consider cell type, species, pathogen strain, and experimental conditions.

What are optimal techniques for investigating IRF1-dependent gene regulation?

To comprehensively investigate IRF1-dependent gene regulation, researchers should employ a combination of complementary techniques:

  • Chromatin Immunoprecipitation sequencing (ChIP-seq): To identify genome-wide IRF1 binding sites and establish direct target genes.

  • RNA sequencing with IRF1 knockdown/knockout: Comparing transcriptomes between IRF1-sufficient and IRF1-deficient cells to identify IRF1-dependent gene expression patterns.

  • ATAC-seq (Assay for Transposase-Accessible Chromatin): Since IRF1 is involved in regulating chromatin accessibility, ATAC-seq can reveal IRF1-dependent changes in chromatin structure .

  • Fluorescence in situ hybridization: For cellular localization of IRF1-target transcripts, as demonstrated in studies examining IRG1 expression in infected versus bystander cells .

  • Promoter-reporter constructs: To validate direct regulation of specific genes by IRF1.

These approaches should be complemented with appropriate statistical analyses, including Student's t-test for single comparisons or two-way ANOVA with post hoc Tukey's test for multiple comparisons, with significance thresholds clearly defined (p ≤ 0.05 *, p ≤ 0.01 **, p ≤ 0.001 ***, p ≤ 0.0001 ****) .

How can researchers effectively silence IRF1 to study its function?

Effective silencing of IRF1 requires careful consideration of methodology to ensure specific knockdown with minimal off-target effects:

  • siRNA transfection: Small interfering RNA has been successfully used to achieve approximately 75% reduction in IRF1 expression in human primary macrophages, resulting in measurable phenotypic changes including a 45% increase in M. avium burden .

  • Optimization parameters:

    • Transfection timing: For studying infection responses, perform knockdown prior to pathogen challenge

    • Validation methods: Confirm knockdown efficiency at both mRNA (qPCR) and protein (Western blot) levels

    • Controls: Include non-targeting siRNA controls to account for transfection effects

    • Duration assessment: Measure knockdown persistence throughout the experimental timeframe

  • Alternative approaches:

    • CRISPR-Cas9 gene editing for complete knockout studies

    • Inducible shRNA systems for temporal control of IRF1 expression

    • Dominant negative IRF1 constructs to interfere with function rather than expression

When reporting IRF1 silencing experiments, researchers should clearly document knockdown efficiency (e.g., 75% reduction achieved in previous studies), phenotypic effects (e.g., 45% increase in pathogen burden), and appropriate statistical analyses to demonstrate significance (p = 0.001) .

What considerations are important when designing experiments to study IRF1 nuclear translocation?

Investigating IRF1 nuclear translocation requires careful experimental design:

  • Temporal considerations:

    • Include multiple timepoints (e.g., 4, 24, and 48 hours post-infection) to capture the dynamics of translocation

    • Include early timepoints (1-3 hours) to capture initial activation events

  • Spatial analysis:

    • Distinguish between infected and bystander populations in heterogeneous infection models

    • Quantify percentage of cells with nuclear IRF1 (baseline of approximately 20% in uninfected macrophages)

    • Use appropriate nuclear markers and high-resolution imaging

  • Signaling pathway dissection:

    • Include cytokine blocking antibodies (e.g., infliximab, tocilizumab) to assess contribution of autocrine/paracrine signaling

    • Use kinase inhibitors (e.g., IKKβ inhibitors) to dissect upstream signaling requirements

  • Quantification methods:

    • Automated image analysis for unbiased quantification

    • Clear definition of nuclear translocation threshold

    • Report both percentage of cells with nuclear IRF1 and intensity measurements

By incorporating these considerations, researchers can generate comprehensive data on IRF1 activation dynamics, distinguishing between direct pathogen effects and secondary autocrine/paracrine signaling mechanisms.

What are the technical challenges in studying IRF1-dependent metabolic reprogramming?

Investigating IRF1-dependent metabolic reprogramming presents several technical challenges:

To address these challenges, researchers should combine multiple approaches, including targeted metabolomics, isotope tracing, subcellular imaging of metabolite-producing enzymes (like IRG1), and functional readouts such as pathogen growth inhibition assays under various metabolic conditions.

What emerging technologies might advance our understanding of IRF1 biology?

Several emerging technologies hold promise for advancing IRF1 research:

  • Single-cell transcriptomics and proteomics: To dissect heterogeneity in IRF1 responses across individual cells within populations, particularly important for understanding infected versus bystander cell dynamics .

  • Live-cell imaging of IRF1 dynamics: Using fluorescently tagged IRF1 to track real-time activation, nuclear translocation, and binding site occupancy.

  • Spatial transcriptomics: To map IRF1-dependent gene expression patterns within tissues, providing insights into the spatial organization of immune responses.

  • CRISPR-based epigenome editing: To selectively modify the chromatin landscape at IRF1 target sites and assess the impact on gene regulation.

  • Organoid and tissue-on-chip technologies: To study IRF1 function in more physiologically relevant three-dimensional contexts that better recapitulate tissue microenvironments.

These technologies could help resolve existing contradictions in the literature and provide more nuanced understanding of context-dependent IRF1 functions across different cell types, infection scenarios, and disease states.

How might targeting IRF1 pathways impact clinical approaches to infectious and inflammatory diseases?

Understanding IRF1 pathways has significant clinical implications:

  • Refined anti-inflammatory approaches: Current anti-cytokine therapies (like tocilizumab and infliximab) may increase susceptibility to infections by disrupting IRF1-dependent antimicrobial mechanisms . More selective approaches targeting specific downstream pathways might maintain therapeutic benefits while preserving host defense.

  • Host-directed therapies: Enhancing IRF1-dependent antimicrobial mechanisms could provide alternative approaches to combat infections, particularly those caused by drug-resistant pathogens.

  • Biomarker development: IRF1 activation patterns could serve as biomarkers for infection status, inflammatory disease progression, or treatment response.

  • Vaccine adjuvant strategies: Modulating IRF1 pathways could enhance vaccine efficacy by optimizing innate immune activation.

The balance between IRF1's antimicrobial and inflammatory functions must be carefully considered in any therapeutic approach, as demonstrated by the observation that patients on anti-inflammatory treatments targeting TNF-α or IL-6 can be more susceptible to mycobacterial disease .

Product Science Overview

Introduction

Interferon Regulatory Factor-1 (IRF-1) is a crucial transcription factor in the regulation of interferon (IFN) genes and plays a significant role in the immune response. It is a member of the IRF family, which consists of several transcription factors involved in the regulation of the IFN system and other genes related to immune responses .

Discovery and Structure

IRF-1 was first identified as a transcription factor that binds to the IFN-stimulated response element (ISRE) in the promoters of IFN and IFN-stimulated genes . The IRF-1 protein comprises 325 amino acids and contains an N-terminal DNA-binding domain (DBD) characterized by a helix-turn-helix motif. This motif is highly conserved among all IRF proteins and is responsible for recognizing specific DNA sequences .

Functions and Mechanisms

IRF-1 is known for its role in various cellular processes, including:

  • Immune Response: IRF-1 activates the transcription of IFN-α and IFN-β genes in response to viral infections .
  • Tumor Suppression: It acts as a tumor suppressor by regulating genes involved in cell cycle control, apoptosis, and DNA damage response .
  • Cell Cycle Regulation: IRF-1 influences the expression of genes that control the cell cycle, thereby playing a role in cell proliferation and differentiation .
Recombinant IRF-1

Recombinant human IRF-1 is produced using genetic engineering techniques, typically in Escherichia coli (E. coli) cells. The gene encoding IRF-1 is cloned and expressed in E. coli, resulting in the production of the recombinant protein . This recombinant protein is used in various research applications to study the biological functions of IRF-1 and its role in immune responses and tumor suppression .

Applications in Research

Recombinant IRF-1 is valuable in research for several reasons:

  • Studying Immune Responses: It helps in understanding how IRF-1 regulates the expression of IFN genes and other immune-related genes .
  • Cancer Research: Researchers use recombinant IRF-1 to investigate its role in tumor suppression and its potential as a therapeutic target .
  • Drug Development: The protein is used in screening assays to identify compounds that modulate IRF-1 activity, which could lead to the development of new therapies for viral infections and cancer .

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 2024 Thebiotek. All Rights Reserved.