IRF 2 Human

IFN Regulatory Factor-2 Human Recombinant
Shipped with Ice Packs
In Stock

Description

Gene and Protein Structure

  • Chromosomal Location: The IRF2 gene spans 86,822 bp on chromosome 4q35.1, containing 9 exons and 8 introns .

  • Protein Composition: The full-length isoform (IRF2 isoform 1) comprises 349 amino acids with a molecular weight of ~45 kDa .

  • Domains:

    • DNA-Binding Domain (DBD): A conserved N-terminal region (120 amino acids) enabling recognition of interferon-stimulated response elements (ISREs) .

    • Interferon Association Domain 2 (IAD2): A C-terminal region facilitating interactions with other transcription factors .

Mechanisms of Immune Regulation

IRF2 modulates immune responses through transcriptional regulation:

  • Antagonism of IRF1: Competes with IRF1 for ISRE binding, suppressing interferon-β (IFN-β) and IFN-stimulated gene activation .

  • NK Cell Development: Essential for human natural killer (NK) cell maturation. IRF2 knockdown reduces NK cell proliferation and cytotoxicity, while overexpression has minimal effects .

  • T Cell Exhaustion: Sustained IFN signaling in tumors induces IRF2, which promotes CD8+ T cell exhaustion. Deletion of IRF2 enhances antitumor responses and improves checkpoint therapy efficacy .

Inflammatory and Autoimmune Disorders

  • Atopic Dermatitis (AD): Genetic variants in IRF2 (e.g., rs793814, rs3756094) are linked to AD risk in European Americans (P = 0.007–0.037) and African Americans (P = 0.016) .

  • Inflammasome Regulation: IRF2 controls caspase-4 levels in human macrophages, critical for responses to Gram-negative bacteria .

Recombinant IRF2 Protein

ParameterDetails
SourceRecombinant protein produced in E. coli with a His-tag .
Molecular Weight15 kDa (133 amino acids: residues 1–113 + 20 His-tag residues) .
StabilityStore at -20°C; avoid freeze-thaw cycles .

Therapeutic Targeting

  • Cancer Immunotherapy: IRF2-deficient CD8+ T cells exhibit prolonged antitumor activity, suggesting IRF2 inhibition as a strategy to enhance checkpoint blockade .

  • Stem Cell Engineering: IRF2 suppression in keratinocytes improves epidermal regeneration, relevant for wound healing .

Key Research Gaps

  • Tissue-Specific Roles: Conflicting data in CRC vs. ovarian cancer warrant further mechanistic studies.

  • Ethnic Variability: IRF2 SNP associations differ between European and African ancestries , necessitating population-specific analyses.

Product Specs

Introduction
IFN regulatory factor 2 (IRF2) belongs to the IFN regulatory transcription factor (IRF) family. IRF2 acts as a competitive inhibitor of IRF1-mediated transcriptional activation of IFNs alpha and beta, and likely other genes that rely on IRF1 for activation. Conversely, IRF2 also functions as a transcriptional activator of histone H4. It binds to the upstream regulatory region of type-1 IFN and IFN-inducible MHC class-1 genes (the IFN consensus sequence (ics)) and represses their expression.
Description
Recombinant Human IFN Regulatory Factor-2, expressed in E. coli, is a single, non-glycosylated polypeptide chain containing 133 amino acids (residues 1-113) and a 20 amino acid His Tag. It has a molecular weight of 15 kDa. The IRF2 protein undergoes purification using proprietary chromatographic techniques.
Physical Appearance
Sterile Filtered colorless solution
Formulation
Supplied as a 1 mg/ml solution in 20mM Tris buffer pH 8.0, 1mM DTT, and 10% Glycerol.
Stability
For short-term storage (2-4 weeks), store at 4°C. For extended storage, freeze at -20°C. The addition of a carrier protein (0.1% HSA or BSA) is recommended for long-term storage. Avoid repeated freeze-thaw cycles.
Purity
Purity is determined to be greater than 95.0% by SDS-PAGE analysis.
Synonyms

IRF-2, IRF2, MAR, DKFZp686F0244, IFN regulatory factor 2.

Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MPVERMRMRP WLEEQINSNT IPGLKWLNKE KKIFQIPWMHAARHGWDVEK DAPLFRNWAI HTGKHQPGVD KPDPKTWKAN FRCAMNSLPD IEEVKDKSIKKGNNAFRVYR MLP.

Q&A

What is IRF2 and what is its role in the human immune system?

IRF2 is a member of the Interferon Regulatory Factor family, which includes nine transcription factors (IRF1-IRF9) in humans. It functions primarily as a transcriptional regulator that shapes immune cell development, differentiation, and function .

IRF2 acts predominantly as a negative regulator by antagonizing IRF1 through competition for the same promoter elements of interferon-inducible genes and by inhibiting nuclear translocation of IRF1 . While generally repressive, IRF2 can activate gene transcription in certain contexts, such as cooperating with IRF1 to induce TLR3 expression .

How does IRF2 differ functionally from other IRF family members?

The IRF family can be divided into four subfamilies based on phylogenetic analysis of their DNA binding domains: IRF1/IRF2, IRF3/IRF7, IRF8/IRF9, and others . While several IRF family members participate in NK cell biology, they have distinct roles:

  • IRF1: Regulates IL-15 expression in bone marrow stromal cells, essential for NK cell development

  • IRF2: Acts cell-intrinsically to regulate NK cell development and maturation

  • IRF8: Required for human NK cell development, functional maturation, and proliferative expansion after viral infection

Unlike IRF1, which acts extrinsically, IRF2 functions in a cell-intrinsic manner, as demonstrated by transplantation experiments showing that IRF2-deficient bone marrow cells result in decreased NK cell numbers even when transferred to wild-type recipients .

What are the known expression patterns of IRF2 across human immune cell types?

IRF2 is constitutively expressed in many immune cells and is upregulated in response to both type I and type II interferons . Mass cytometry (CyTOF) analysis of tumor-infiltrating immune cells shows IRF2 is widely expressed across immune populations in both mouse models and human tumors .

Key observations on expression patterns include:

  • CD4+ and CD8+ tumor-infiltrating T lymphocytes show significant IRF2 upregulation compared to their splenic counterparts

  • B cells, macrophages, and dendritic cells maintain relatively stable IRF2 expression levels between tumor and spleen

  • Human melanoma tumor-infiltrating immune cells exhibit a similar broad IRF2 expression pattern, indicating conservation across species

What experimental approaches are most effective for studying IRF2's role in human NK cell development?

Based on the literature, the most effective experimental approach involves:

  • Isolation of hematopoietic progenitor cells (HPCs) from umbilical cord blood

  • Lentiviral transduction for stable IRF2 knockdown or overexpression

  • In vitro NK cell differentiation cultures

  • Assessment of developmental progression, proliferation, apoptosis, and functional capacity

This methodology allows researchers to study the cell-intrinsic effects of IRF2 on human NK cell development, avoiding the limitations of murine models that may not accurately reflect human biology due to interspecies differences .

How does IRF2 knockdown specifically affect human NK cell maturation stages?

IRF2 knockdown has stage-specific effects on human NK cell development:

  • Early stages: IRF2 deficiency causes reduced cell numbers in all early differentiation stages, resulting in dramatically decreased mature NK cell numbers

  • Proliferation: The primary mechanism for reduced NK cell numbers is decreased proliferation rather than increased apoptosis

  • Lineage commitment: IRF2 knockdown leads to a strong trend in reduction of natural killer cell progenitors (NKPs) and ILC3 cell numbers, suggesting a role in NK cell lineage commitment and potentially innate lymphoid cell (ILC) development

  • Functional maturation: The remaining NK cells after IRF2 silencing display:

    • Less mature phenotype

    • Decreased cytotoxic potential against tumor targets

    • Greatly reduced cytokine secretion

What are the differences between IRF2 knockdown and IRF2 overexpression effects on human NK cells?

The research indicates an asymmetrical impact between knockdown and overexpression:

  • IRF2 knockdown: Causes severe developmental defects including reduced cell numbers across developmental stages, impaired proliferation, and compromised functional maturation

  • IRF2 overexpression: Shows limited effects on NK cell development and maturation, suggesting that endogenous IRF2 expression levels are sufficient for these processes

This asymmetry implies that IRF2 functions in a threshold-dependent manner rather than in a dose-dependent fashion for human NK cell development.

How does IRF2 regulate CD8+ T cell function in the tumor microenvironment?

IRF2 functions as a critical regulator of CD8+ T cell responses within the tumor microenvironment:

  • T cell exhaustion: CD8+ T cell-specific deletion of IRF2 prevents acquisition of the T cell exhaustion program within tumors

  • Effector function: IRF2-deficient CD8+ T cells maintain sustained effector functions that promote long-term tumor control

  • Therapy response: IRF2 deficiency in CD8+ T cells increases responsiveness to immune-checkpoint and adoptive cell therapies

  • Interferon signaling: The enhanced tumor control by IRF2-deficient CD8+ T cells requires continuous integration of both type I and type II interferon signals

These findings identify IRF2 as a foundational feedback molecule that redirects interferon signals to suppress T cell responses in the tumor microenvironment.

What experimental models best demonstrate IRF2's impact on anti-tumor immunity?

Multiple tumor models have demonstrated the impact of IRF2 on anti-tumor immunity:

  • MC38 colorectal adenocarcinoma: While tumors grew similarly through day 11 in both wild-type and IRF2-deficient mice, IRF2-deficient mice exhibited superior tumor control with some mice showing no detectable tumors and all surviving the 50-day experimental period (compared to all wild-type mice reaching endpoint by day 25)

  • B16-F10 melanoma: IRF2-deficient mice showed prolonged survival and enhanced tumor control of this minimally immunogenic model

  • Orthotopic polyoma middle T antigen (PyMT) breast tumor: Similar enhanced control was observed in IRF2-deficient mice, indicating the effect extends across diverse tumor types

These models provide a platform for studying the mechanisms by which IRF2 regulates anti-tumor immune responses.

What are the methodological considerations for investigating IRF2 expression in human cancer tissues?

Based on the breast cancer study methodology, key considerations include:

  • Tissue preparation: Use of formalin-fixed paraffin-embedded human archival tissue specimens

  • Control selection: Adjacent areas of normal breast tissue provide crucial comparative controls

  • Detection method: Immunohistochemistry using polyclonal IRF-1 and IRF-2 antibodies with an avidin-biotin-peroxidase complex technique after epitope retrieval

  • Comparison groups: Include both pre-invasive (DCIS) and invasive cancer specimens to assess expression changes across cancer progression

The methods must account for potential heterogeneity in expression across different regions of the tumor and adjacent normal tissue.

What protein interactions mediate IRF2's transcriptional regulatory functions?

IRF2 exerts its regulatory functions through interactions with multiple transcription factors and signaling molecules:

  • IRF1: IRF2 antagonizes IRF1 by competing for binding to the same promoter elements of IFN-I and IFN-II-inducible genes and by inhibiting nuclear translocation of IRF1

  • Other key interaction partners include:

    • NF-κB: IRF2 interacts with NF-κB, affecting inflammatory gene expression

    • STAT1: IRF2 influences interferon signal transduction through STAT1 interaction

    • IRF8: IRF2 modulates immune cell development through cooperation with IRF8

    • IRF9: IRF2 interacts with IRF9, affecting interferon-stimulated gene expression

These protein-protein interactions contribute to the context-dependent activity of IRF2 as either a transcriptional repressor or activator.

How does IRF2 differentially regulate type I versus type II interferon responses?

IRF2 plays a nuanced role in balancing responses to different interferon types:

Type I Interferons (IFN-α/β):

  • Signal through dimeric IFNAR1/IFNAR2 receptor

  • Activate JAK1 and Tyk2 to initiate STAT1/STAT2 phosphorylation

  • Induce expression of hundreds of interferon-stimulated genes (ISGs), including IRFs

  • IRF2 is upregulated by type I IFNs and subsequently limits their signaling

Type II Interferon (IFN-γ):

  • Signals through IFNγR (composed of IFNγR1 and IFNγR2)

  • Activates JAK1 and JAK2 to phosphorylate STAT1

  • STAT1 homodimerizes to form gamma-activated factors that induce ISG expression

  • IRF2 is also upregulated by IFN-γ and opposes IFN-γ-induced gene expression

By differentially inducing and antagonizing various IRFs, IRF2 helps balance the pro-inflammatory and regulatory aspects of interferon signaling.

What genomic alterations of IRF2 have been identified in human diseases?

  • In human breast cancer, expression of IRF1 and IRF2 is altered compared to normal adjacent tissue

  • Some tumor types show increased IRF2 expression, which correlates with cancer development and progression, potentially through repressing cancer cell-intrinsic interferon signaling

  • Other tumor types downregulate IRF2 to evade immune targeting, as IRF2 directly represses PDL1 expression and activates components of the MHC-I pathway

Further research is needed to characterize specific genomic alterations (mutations, copy number variations, etc.) affecting IRF2 in human diseases.

How might single-cell technologies advance our understanding of IRF2's cell type-specific functions?

Single-cell technologies could significantly advance IRF2 research by:

  • Resolving heterogeneity: Identifying cell subpopulations with distinct IRF2 expression levels and correlating with functional states

  • Developmental trajectories: Mapping IRF2 expression changes during immune cell development and differentiation

  • Spatial context: Using spatial transcriptomics to understand how IRF2 expression varies based on cellular location within tissues (e.g., tumor margin vs. core)

  • Multi-omic integration: Combining single-cell transcriptomics with proteomics or epigenomics to understand how IRF2 regulates gene expression programs

  • Response dynamics: Tracking temporal changes in IRF2 activity following stimulation with different cytokines or in disease states

These approaches would provide a more nuanced understanding of IRF2's context-dependent functions across different immune cell types and states.

What are the challenges in developing therapeutic strategies targeting IRF2 in human diseases?

Developing IRF2-targeted therapeutics presents several challenges:

  • Pleiotropic effects: IRF2 functions across multiple immune cell types, so targeting could have widespread and potentially unpredictable consequences

  • Opposing roles: IRF2 can act as both an oncogene and tumor suppressor depending on the cancer context

  • Intracellular target: As a transcription factor, IRF2 is difficult to target directly with small molecules or antibodies

  • Cell type specificity: Achieving selective targeting of IRF2 in specific cell populations (e.g., tumor-infiltrating T cells) remains technically challenging

  • Compensatory mechanisms: Other IRF family members may compensate for IRF2 inhibition

  • Timing considerations: The appropriate therapeutic window for IRF2 modulation may depend on disease stage and immune context

Despite these challenges, the pronounced effect of IRF2 deficiency on tumor control in mouse models suggests potential therapeutic value in targeting this pathway.

How does post-translational modification regulate IRF2 activity in different cellular contexts?

  • Phosphorylation: Likely regulates IRF2's DNA binding ability, protein-protein interactions, and subcellular localization

  • Ubiquitination: May control IRF2 protein stability and turnover in response to different stimuli

  • SUMOylation: Could modulate IRF2's interaction with transcriptional co-factors

  • Acetylation: Might affect IRF2's chromatin binding properties

  • Methylation: Potentially regulates IRF2's activity as a transcriptional repressor or activator

Research into these modifications would help explain how IRF2 activity is finely tuned in different cellular contexts and in response to various stimuli.

How can IRF2 expression be leveraged as a biomarker in cancer prognostication?

Based on the research presented, IRF2 shows potential as a prognostic biomarker in several ways:

  • Expression profiling: Analyzing IRF2 expression levels in tumor tissue compared to adjacent normal tissue

  • Cell type-specific expression: Assessing IRF2 levels specifically in tumor-infiltrating lymphocytes versus the tumor cells themselves

  • Expression ratio: Examining the balance of IRF1 to IRF2 expression, which may be more informative than absolute levels of either factor alone

  • Combined immunoprofiling: Integrating IRF2 expression with other immune markers to create comprehensive immunoscores

The prognostic value likely varies by cancer type, as increased IRF2 expression correlates with worse outcomes in some cancers (esophageal), while in others (melanoma), it may indicate better immunogenicity through enhanced MHC-I expression and reduced PDL1.

What strategies show promise for enhancing NK cell-based immunotherapies through IRF2 modulation?

From the research findings, several strategies emerge for enhancing NK cell-based immunotherapies:

  • Transient IRF2 modulation: Since complete IRF2 knockdown severely impairs NK cell development, temporary or partial inhibition during specific maturation stages might optimize NK cell numbers and functionality

  • Ex vivo engineering: For adoptive NK cell therapies, manipulating IRF2 expression in donor-derived NK cells or NK cell precursors could enhance:

    • Proliferative capacity

    • Cytotoxic potential against tumor targets

    • Cytokine secretion profiles

  • Combinatorial approaches: Pairing IRF2 modulation with other immunotherapeutic strategies such as checkpoint inhibition or cytokine therapy could enhance efficacy

  • Timing considerations: Optimal IRF2 modulation likely differs between NK cell development, expansion, and effector phases

These strategies require careful balancing, as IRF2 plays important roles in both NK cell development and functional maturation.

How might the interplay between IRF2 and T cell exhaustion inform next-generation immunotherapies?

The research showing that IRF2 deletion prevents T cell exhaustion suggests several therapeutic directions:

  • Targeted inhibition: Developing strategies to transiently inhibit IRF2 in tumor-infiltrating CD8+ T cells could prevent exhaustion and maintain anti-tumor activity

  • Checkpoint therapy enhancement: IRF2 inhibition increased responsiveness to immune checkpoint therapies, suggesting IRF2 as a novel checkpoint target or as a predictive biomarker for conventional checkpoint inhibitor response

  • Adoptive cell therapy: Engineering T cells with modified IRF2 function prior to adoptive transfer could improve persistence and activity within the tumor microenvironment

  • Interferon modulation: Since IRF2-deficient CD8+ T cells require continuous integration of both type I and type II interferon signals for tumor control, combinatorial approaches that maintain beneficial interferon signaling while blocking IRF2-mediated negative feedback could be effective

These approaches address a fundamental mechanism of T cell dysfunction in the tumor microenvironment and could potentially overcome resistance to existing immunotherapies.

Product Science Overview

Structure and Function

IRF-2 is a transcription factor that consists of 349 amino acids. It has a DNA-binding domain (DBD) at the N-terminal and a repression domain (RD) at the C-terminus . The DNA-binding domain allows IRF-2 to bind to specific DNA sequences, known as interferon-stimulated response elements (ISREs), which are found in the promoters of many virus and interferon-inducible mammalian genes .

IRF-2 is known to act as a repressor of transcription of IRF-1 inducible genes. However, it can also function as an activator of certain genes, such as the cell-cycle regulated H4 (histone) gene, VCAM-1 (Vascular Cell Adhesion Molecule-1), and TLR9 (Toll-like Receptor-9) gene . Moreover, both IRF-2 and IRF-1 are required for the expression of MHC II class molecules through the CIITA type IV promoter .

Biological Role

IRF-2 plays a significant role in cell growth regulation and has been shown to be a potential oncogene . It is involved in various biological processes, including host defense, oncogenesis, apoptosis, viral latency, and immune responses . The ability of IRF-2 to bind to DNA and regulate gene expression makes it a critical player in the cellular response to viral infections and other stress signals.

Recombinant IRF-2

Recombinant human IRF-2 is expressed in Escherichia coli (E. coli) and purified using conventional chromatography techniques . The recombinant protein is biologically active and retains its DNA-binding activity, making it a valuable tool for research and therapeutic applications .

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.