IFNGR1 Human Recombinant produced in Sf9 Baculovirus cells is a single, glycosylated polypeptide chain containing 234 amino acids (18-245a.a.) and having a molecular mass of 26.6kDa (Migrates at 28-40kDa on SDS-PAGE under reducing conditions).
IFNGR1 is expressed with a 6 amino acid His tag at C-Terminus and purified by proprietary chromatographic techniques.
IFNGR1, CD119, IFNGR, IMD27A, IMD27B, CDw119.
EMGTADLGPS SVPTPTNVTI ESYNMNPIVY WEYQIMPQVP VFTVEVKNYG VKNSEWIDAC INISHHYCNI SDHVGDPSNS LWVRVKARVG QKESAYAKSE EFAVCRDGKI GPPKLDIRKE EKQIMIDIFH PSVFVNGDEQ EVDYDPETTC YIRVYNVYVR MNGSEIQYKI LTQKEDDCDE IQCQLAIPVS SLNSQYCVSA EGVLHVWGVT TEKSKEVCIT IFNSSIKGHH HHHH.
IFNGR1 transcription is tightly regulated through multiple mechanisms, with type I interferons playing a particularly important role in its negative regulation. Research demonstrates that type I interferons (particularly IFNβ) rapidly silence ifngr1 transcription in myeloid cells through recruitment of repressive transcriptional complexes .
The silencing mechanism involves:
Recruitment of Early growth response factor 3 (Egr3) to the ifngr1 promoter
Subsequent recruitment of NGFI-A binding protein 1 (Nab1) corepressor
Reduction in activated RNA polymerase II at the promoter
Deacetylation of histones H3 and H4, creating a repressive chromatin environment
This transcriptional silencing occurs as rapidly and effectively as actinomycin D treatment, resulting in a significant reduction in ifngr1 transcript abundance. The proximal ifngr1 promoter contains a critical Egr-binding site that is essential for this repressive mechanism .
| Factor | Role | Detection Method | Effect When Inhibited |
|---|---|---|---|
| Egr3 | DNA-binding repressor | Chromatin immunoprecipitation | Prevents down-regulation |
| Nab1 | Corepressor | Chromatin immunoprecipitation | Prevents down-regulation and histone deacetylation |
| HDACs | Histone modification | Histone acetylation analysis | Prevents chromatin remodeling |
Type I and type II interferons participate in a complex regulatory network affecting IFNGR1 expression. While type II interferon (IFNγ) signals through IFNGR1, type I interferons (like IFNβ) can down-regulate IFNGR1 expression, creating a negative feedback loop that modulates IFNγ responsiveness .
Methodologically, this cross-regulation can be studied through:
Sequential treatment with different interferon types followed by flow cytometry analysis of surface IFNGR1
Quantitative PCR measurement of ifngr1 transcript levels using specific primers
Promoter-reporter assays using ifngr1-luciferase constructs
Chromatin immunoprecipitation to assess transcription factor binding
This cross-regulation has significant implications for immune response dynamics during infections, as type I interferon-mediated suppression of IFNGR1 can limit myeloid cell activation by IFNγ, potentially contributing to increased susceptibility to certain bacterial infections .
Accurate measurement of IFNGR1 requires complementary approaches at both protein and transcript levels:
Protein Level Detection:
Flow cytometry using biotinylated antibodies to IFNGR1/CD119 followed by streptavidin-APC secondary antibody
For human cells, CD3-FITC and CD14-PE antibodies can be used to simultaneously identify T cells and monocytes
Transcript Level Detection:
Real-time quantitative PCR using SYBR Green PCR Master Mix and specific primers for human ifngr1
Transcript abundance calculation using the equation: 2^(-1*((IFNGR1 Ct – Mean GAPDH Ct) – (Mean Unstimulated dCT)))
Transcript half-life calculation using: half-life=(elapsed time*log2)/(log (beginning amount/ending amount))
| Level | Method | Key Reagents | Applications |
|---|---|---|---|
| Protein | Flow Cytometry | Biotinylated anti-IFNGR1/CD119, streptavidin-APC | Cell surface expression |
| mRNA | qRT-PCR | Commercial primer sets for human ifngr1 | Transcript abundance |
| Promoter Activity | Luciferase Assay | ifngr1 promoter (-2320 to +1) in pGL3-Basic | Transcriptional regulation |
| Protein-DNA Interactions | ChIP | Antibodies to Egr3, Nab1, acetylated histones | Regulatory mechanisms |
Studying IFNGR1 promoter regulation requires multiple complementary approaches:
Reporter assays: The ifngr1 promoter can be cloned from position -2320 to +1 relative to the transcription start site using specific primers with KpnI and XhoI restriction sites for insertion into the pGL3-Basic vector
Site-directed mutagenesis: To validate the importance of specific binding sites, such as the Egr binding site that is crucial for IFNβ-mediated repression
Chromatin immunoprecipitation (ChIP): To detect protein binding and histone modifications at the endogenous ifngr1 promoter, enabling temporal analysis of regulatory events
Transcription factor knockdown: Using siRNA or shRNA approaches to deplete specific factors (e.g., Nab1) and assess their role in promoter regulation
When applying these methodologies, researchers should consider the cell type-specificity of IFNGR1 regulation, as mechanisms differ considerably between myeloid cells and T cells .
IFNGR1-mediated signaling plays a complex role in SARS-CoV-2 infection. Research using organoid models has revealed that IFNγ (which signals through IFNGR1) promotes cellular differentiation into ACE2-expressing enterocytes, thereby enhancing susceptibility to SARS-CoV-2 infection .
Key experimental findings include:
IFNγ treatment increases expression of ACE2 (the SARS-CoV-2 receptor) in differentiated human organoids
Organoids treated with IFNγ show higher SARS-CoV-2 infection rates and increased viral load
JAK/STAT inhibition using pyridone 6 (P6) prevents the IFNγ-induced increase in viral infection
| Culture Condition | ACE2 Expression | Virus Load | Effect of JAK Inhibitor (P6) |
|---|---|---|---|
| Basic Medium | Moderate | Moderate | Not tested |
| Basic Medium + IFNγ | Significantly increased | Significantly increased | Prevented increase |
| Full Medium | Low | Low | Not tested |
| Full Medium + IFNγ | Increased | Increased | Not tested |
This relationship suggests a potential mechanism for enhanced COVID-19 severity in patients with pre-existing inflammatory conditions, as chronic inflammation might increase IFNγ levels, drive ACE2 expression, and consequently enhance viral replication .
Type I interferons can increase susceptibility to certain bacterial infections by down-regulating myeloid cell IFNGR1 expression, thereby reducing responsiveness to IFNγ, which is essential for antimicrobial defense .
This mechanism has been demonstrated for intracellular bacterial pathogens including Mycobacterium tuberculosis and Listeria monocytogenes . The process involves:
Type I IFN binding to IFNAR on myeloid cells
Rapid silencing of ifngr1 transcription through the Egr3/Nab1 complex
Reduced surface IFNGR1 expression
Diminished myeloid cell activation in response to IFNγ
This mechanism provides insight into the paradoxical observation that type I interferons, typically considered antiviral, can sometimes be detrimental during bacterial infections.
Contradictory findings in IFNGR1 research often stem from contextual differences. A methodological framework to resolve such contradictions includes:
Cell type consideration: IFNGR1 regulation differs markedly between myeloid cells and T cells. The search results indicate that type I IFNs rapidly silence ifngr1 transcription in mouse and human macrophages but not T cells .
Temporal dynamics: IFNGR1 regulation is highly time-dependent. The recruitment of Egr3 to the ifngr1 promoter occurs rapidly after IFNβ stimulation, followed by Nab1 recruitment and histone deacetylation .
Signaling context: The presence of other cytokines can significantly alter IFNGR1 regulation. For example, in the context of SARS-CoV-2 infection, multiple cytokines may be present that affect IFNGR1 expression and function .
Experimental validation approach:
Use multiple complementary techniques (flow cytometry, qPCR, ChIP)
Include appropriate controls for each experiment
Perform time-course studies to capture dynamic changes
Validate findings in multiple cell types and/or donor samples
JAK/STAT signaling is integral to IFNGR1 function, and experimental designs to study this interplay should include:
Pharmacological inhibition: Using JAK inhibitors like pyridone 6 (P6) to block signaling downstream of IFNGR1. Research shows that P6 treatment prevents the IFNγ-induced increase in ACE2 expression and SARS-CoV-2 infection in organoids .
Genetic manipulation: Using siRNA or CRISPR-Cas9 to target specific components of the JAK/STAT pathway.
Phosphorylation analysis: Tracking STAT1 phosphorylation as a readout of IFNGR1 signaling activity.
Reciprocal regulation: Investigating how JAK/STAT activation through one cytokine receptor affects signaling through others. The search results demonstrate that type I IFN signaling (which uses JAK/STAT) negatively regulates IFNGR1 expression .
Reporter systems: Using STAT-responsive promoter-reporter constructs to quantify signaling output.
| Approach | Technique | Readout | Applications |
|---|---|---|---|
| Inhibition | JAK inhibitor (P6) | Downstream gene expression | Block IFNγ signaling effects |
| Phosphorylation | Western blot/flow cytometry | pSTAT1 levels | Signal transduction analysis |
| Gene expression | qPCR | JAK/STAT target genes | Functional consequences |
| Genetic | siRNA knockdown | Protein depletion | Mechanism validation |
Several promising research directions emerge from current IFNGR1 understanding:
Chronic inflammation and IFNGR1 dynamics: Investigating how persistent inflammatory states affect IFNGR1 expression and function. Evidence suggests that chronic inflammation in the lungs of smokers might increase risk for severe COVID-19, potentially through mechanisms involving IFNGR1-mediated signaling .
Age-related changes in IFNGR1 regulation: Research indicates that IFN responses change with aging, correlating with increased severity of COVID-19 in elderly patients . Studying how IFNGR1 expression and signaling change with age could provide insights into age-dependent disease susceptibility.
Tissue-specific IFNGR1 regulation: The search results show differential IFNGR1 regulation between cell types. Expanding this to understand tissue-specific regulation could explain organ-specific pathologies in inflammatory diseases.
Therapeutic targeting of IFNGR1 regulation: Developing approaches to modulate IFNGR1 expression or signaling for therapeutic benefit. JAK inhibitors like P6 that block signaling downstream of IFNGR1 show potential in preventing SARS-CoV-2 infection enhancement by IFNγ .
Single-cell technologies offer unprecedented opportunities to understand IFNGR1 biology:
Heterogeneity in IFNGR1 expression: Single-cell RNA sequencing can reveal cell-to-cell variation in IFNGR1 expression within seemingly homogeneous populations. The search results mention single-cell RNA sequencing data showing that IFN-γ receptors (INFGR1/2) are expressed in Krt20-expressing enterocytes .
Dynamic regulation: Single-cell approaches can capture transient states during IFNGR1 regulation that might be missed in bulk analyses.
Integration with spatial information: Spatial transcriptomics can relate IFNGR1 expression patterns to tissue microenvironments, providing context for functional studies.
Multi-modal analysis: Combining transcript and protein measurements at single-cell resolution can reveal post-transcriptional regulation of IFNGR1.
Trajectory analysis: Single-cell data can reconstruct developmental or activation trajectories, revealing how IFNGR1 expression changes during cellular differentiation processes, similar to the differentiation of organoids toward the enterocyte lineage observed with IFN-γ treatment .
IFNGR1 is the ligand-binding chain (alpha) of the gamma interferon receptor. The human interferon-gamma receptor is a heterodimer composed of IFNGR1 and IFNGR2 . When interferon-gamma (IFN-γ) binds to IFNGR1, it induces the rapid dimerization of the receptor chains, which is crucial for signal transduction and subsequent immune responses .
Recombinant human IFNGR1 is produced using advanced biotechnological methods. Typically, the gene encoding IFNGR1 is inserted into a suitable expression system, such as E. coli or a mammalian cell line. The recombinant protein is then purified using techniques like column chromatography to ensure high purity and activity .