IFNGR1 Antibody

Shipped with Ice Packs
In Stock

Description

Definition and Biological Context

IFNGR1 antibodies target the ligand-binding alpha subunit of the heterodimeric interferon-gamma (IFN-γ) receptor, encoded by the IFNGR1 gene (CD119). This receptor is critical for initiating JAK-STAT signaling upon IFN-γ binding, driving antimicrobial, antiviral, and antitumor immune responses . Deficiencies in IFNGR1 are associated with Mendelian susceptibility to mycobacterial diseases (MSMD) .

Immune Regulation

  • Myeloid Cell Sensitivity: IFN-γ downregulates IFNGR1 surface expression in myeloid cells via transcriptional silencing, independent of type I interferons. This modulation affects antibacterial responses .

  • Cancer Biology: Reduced IFNGR1 expression in lung adenocarcinoma correlates with impaired IFN-γ signaling, promoting tumor progression .

Diagnostic Use

  • Flow Cytometry: Antibodies like ab275700 enable detection of IFNGR1 on immune cells (e.g., monocytes, T cells) .

  • Western Blotting: Antibody 10808-1-AP detects IFNGR1 at ~90 kDa in human tissues, aiding in protein quantification .

Therapeutic Insights

  • Blocking Antibodies: GR-20 inhibits IFN-γ binding, useful for studying receptor-ligand interactions in murine models .

  • Knockout Validation: ab134070 confirms IFNGR1 absence in CRISPR-edited HeLa and HEK293 cells .

Clinical Relevance

  • Genetic Disorders:

    • Complete IFNGR1 deficiency (e.g., 131delC mutation) abolishes receptor expression, causing severe mycobacterial infections .

    • Partial deficiencies (e.g., 818del4/WT) reduce STAT1 activation, leading to atypical immune responses .

Technical Considerations

  • Cross-Reactivity: Most antibodies are human-specific, but 10808-1-AP also reacts with mouse samples .

  • Buffer Compatibility: Storage in PBS with 0.02% sodium azide and 50% glycerol ensures antibody stability .

Product Specs

Buffer
PBS with 0.1% Sodium Azide, 50% Glycerol, pH 7.3. Store at -20°C. Avoid freeze/thaw cycles.
Lead Time
Typically, we can ship your order within 1-3 business days of receiving it. Delivery time may vary depending on your location and preferred shipping method. Please consult your local distributor for specific delivery timelines.
Synonyms
IFNGR1; Interferon gamma receptor 1; IFN-gamma receptor 1; IFN-gamma-R1; CDw119; Interferon gamma receptor alpha-chain; IFN-gamma-R-alpha; CD antigen CD119
Target Names
Uniprot No.

Target Background

Function
IFNGR1 is a receptor subunit for interferon gamma (IFN-γ). It plays critical roles in antimicrobial, antiviral, and antitumor immune responses by activating effector immune cells and enhancing antigen presentation. It associates with the transmembrane accessory factor IFNGR2 to form a functional receptor. Upon ligand binding, the intracellular domain of IFNGR1 undergoes a conformational change, enabling the association of downstream signaling components JAK1 and JAK2. Activated JAK1 then phosphorylates IFNGR1, creating a docking site for STAT1. Subsequent phosphorylation of STAT1 leads to dimerization, translocation to the nucleus, and stimulation of target gene transcription. STAT3 can also be activated in a similar manner, although its activation appears to be weaker. The phosphorylation of IFNGR1's intracellular domain also provides a docking site for SOCS1, which regulates the JAK-STAT pathway by competing with STAT1 binding to IFNGR1.
Gene References Into Functions
  1. A rare variant (c.G40A) in interferon gamma receptor 1 has been implicated in the pathogenesis of myasthenia gravis. PMID: 29441481
  2. Positive reactions in interferon-gamma release tests are not associated with IFNGR1 single nucleotide polymorphisms (SNPs). PMID: 29209098
  3. All known mutations, along with 287 other variations, have been deposited in the online IFNGR1 variation database. This article reviews the function of IFN-γR1 and the molecular genetics of human IFNGR1. PMID: 28744922
  4. Studies have revealed a significant correlation between IFNGR1- T-56CSNP and Nontuberculous mycobacteria infection in studied populations. PMID: 28719321
  5. Type 1 IFN receptor signaling in B cells accelerates, but is not required for, lupus development. PMID: 27069113
  6. A meta-analysis did not find sufficient evidence to establish a significant association between IFNGR1 -56C/T polymorphism and tuberculosis susceptibility. PMID: 25382336
  7. All patients tested positive for mycobacteria; one was heterozygous for the IFNGR1 exon 5 single-nucleotide-missense substitution. PMID: 27356097
  8. Deletion of IFNGR1 results in complete IFN-γR1 deficiency. While the deletion ends close to the IL22RA2 gene, it does not appear to affect IL22RA2 transcription. PMID: 26931784
  9. A significant association of IFN-γR1 and P2X7 gene polymorphisms with the risk of developing TB in the Iranian population has been observed. PMID: 27020872
  10. Mendelian susceptibility to mycobacteria can arise due to a partial dominant mutation of the interferon gamma receptor 1 gene. PMID: 26251056
  11. Targeted deep sequencing has identified rare loss-of-function variants in IFNGR1 associated with the risk of atopic dermatitis complicated by eczema herpeticum. PMID: 26343451
  12. Statistical analyses showed that four genetic variants in IFNGR1 were marginally associated with the risk of Tuberculosis (P = 0.02-0.04), while other single nucleotide polymorphisms in IFNGR1 and IFNGR2 did not exhibit any associations. PMID: 25815589
  13. FcgammaRIIa cross-talk with TLRs, IL-1R, and IFNgammaR selectively modulates cytokine production in human myeloid cells. PMID: 25108563
  14. In an African-American population, a significant difference in IFNGR1 expression was observed between patients with RA and controls. However, IFNGR1 expression levels were not significantly associated with erosion status or other radiographic outcomes. PMID: 25708927
  15. Intact IFN-γR1 expression and function distinguishes Langerhans cell histiocytosis from mendelian susceptibility to mycobacterial disease. PMID: 24254535
  16. Genetic polymorphisms in the IFNGR1 gene are involved in the risk of tuberculosis in the Chinese population. PMID: 24680779
  17. Research has investigated single nucleotide polymorphisms (SNPs) of IFNGR1, GSTT1, and GSTP1 genes in gastric cancer samples. PMID: 24453034
  18. A novel heterozygous frameshift mutation (805delT) encoding the IFN-γ receptor 1 (IFNGR1) was identified in a case of Mycobacterium intracellulare infection. PMID: 24220318
  19. An association study of functional polymorphic genes, including IFNGR-1, was conducted to investigate their relationship with disease progression, aspartate aminotransferase, alanine aminotransferase, and viral load in chronic hepatitis B and C. PMID: 23040881
  20. Interaction of IFNgammaR1 with TRAF6 regulates NF-κB activation and IFNlambdaR1 stability. PMID: 22644879
  21. A novel endocytosis motif, sharing characteristics of tyrosine-based and dileucine-based internalization sequences, is highly conserved in IFN-γ receptors across species. PMID: 22595141
  22. IFNGR1 is a modifier gene of cystic fibrosis disease. PMID: 21731057
  23. Japanese patients with a genetic mutation in the IFN-γ-R1 gene were found to be more susceptible to developing recurrent disseminated mycobacterial infections. PMID: 21221749
  24. Single nucleotide polymorphism in the IFNGR1 gene is associated with rectal cancer. PMID: 21859832
  25. Overexpression of the CD74 gene in TEC can increase IFN-γR mRNA expression. PMID: 21722521
  26. An autosomal recessive disorder caused by a single mutation in interferon-γ receptor-1 (IFNGR1) at position -56 was found to be associated with susceptibility to leprosy in children of the same family. PMID: 21460021
  27. IL-29 up-regulated, whereas IFNα down-regulated, the surface expression of the IFNγ receptor 1 chain on macrophages, leading to differential responsiveness of TLR-challenged macrophages to IFNγ. PMID: 21190998
  28. Research has not shown an implication of IFNGR1 gene polymorphisms in the susceptibility to or clinical expression of giant cell arteritis. PMID: 20412699
  29. Clinical trial of gene-disease association. (HuGE Navigator) PMID: 20399512
  30. A study revealed a positive association between the -56C/C genotype of IFNGR1 (OR = 1.7; 95% CI = 1.1-2.7) and pre-eclampsia. PMID: 20070287
  31. Functional analysis of naturally occurring amino acid substitutions in human IFN-γR1 has been conducted. PMID: 20015550
  32. A case-control association analysis did not detect significant association between IFNGR1 polymorphisms and cerebral malaria in the Thai population. PMID: 19712753
  33. Partial IFNGR1 mutations have been observed in Japanese patients with BCG osteomyelitis. PMID: 11865431
  34. IFNGR1 gene promoter polymorphisms may be associated with susceptibility to cerebral malaria. PMID: 12023780
  35. Mutations in interferon-gamma receptor 1. PMID: 12027427
  36. Research has identified a further role of IFN-γ on IL-4 responses, including reduced IL-4R surface expression by human monocytes. PMID: 12034035
  37. Lipid microdomains are required sites for the selective endocytosis and nuclear translocation of IFN-γ receptor-1. PMID: 12165521
  38. Partial deficiency of IFN-γ receptor 1 leads to the abrogation of IFN-γ-induced killing of Salmonella typhimurium and Toxoplasma gondii due to IFN-γ unresponsiveness of patients' cells of the monocyte/macrophage lineage. PMID: 12244188
  39. Förster resonance energy transfer (FRET) has been used to demonstrate that the IFNGR chains are preassembled on the cell membrane. PMID: 12438563
  40. IFNGR1 expression is suppressed by 2- to 3-fold in B-cell chronic lymphocytic leukemia cells, which is expected to increase CLL cell survival. PMID: 12454749
  41. Genome analysis has identified a polymorphism in the human interferon gamma receptor that affects Helicobacter pylori infection. PMID: 12516030
  42. Mutations in the IFNGR1 gene have not been associated with susceptibility to lepromatous leprosy in the Korean population. PMID: 12743658
  43. Unidentified allelic variations in the IFNGR1 gene may elevate or decrease the risk of tuberculosis in the Croatian population, as part of the multigenic predisposition to tuberculosis. PMID: 12753505
  44. While IFN-γ production in allergic patients with L467P was equivalent to that in non-allergic subjects, their serum IgE levels were high and they had allergic diseases. PMID: 12851715
  45. IFN-γ receptor deficiency alters the epitope hierarchy of the pool of lymphocytic choriomeningitis virus-specific memory CD8 T cells without significantly affecting the immunodominance of the primary CD8 T cell response in an acute infection. PMID: 14734726
  46. Disease susceptibility in Schistosoma mansoni infection to hepatic fibrosis is linked to a SNP in the interferon gamma receptor locus (P=0.000001). PMID: 15756299
  47. The IFN-GammaR2 Arg64/Arg64 genotype does not determine susceptibility to SLE in Chinese people, and the combination of IFN-Gamma R2 Arg64/Arg64 genotype and IFN-Gamma R1 Val14/Val14 genotype does not either. PMID: 15952126
  48. The relationship between polymorphisms at IFNGR1 and susceptibility to pulmonary tuberculosis has been reported in Iranian patients. PMID: 16233916
  49. IFNGR1 does not contribute to susceptibility to rheumatoid arthritis in Caucasians, despite the existence of single nucleotide polymorphisms in this disease. PMID: 16563189
  50. A novel tuberculosis association was found with the 56CC genotype of the IFNGR1 promotor. PMID: 16690980

Show More

Hide All

Database Links

HGNC: 5439

OMIM: 107470

KEGG: hsa:3459

STRING: 9606.ENSP00000356713

UniGene: Hs.520414

Involvement In Disease
Immunodeficiency 27A (IMD27A); Immunodeficiency 27B (IMD27B)
Protein Families
Type II cytokine receptor family
Subcellular Location
Cell membrane; Single-pass type I membrane protein.

Q&A

What is the biological function of IFNGR1?

IFNGR1 serves as a receptor subunit for interferon gamma (IFN-γ) that plays essential roles in antimicrobial, antiviral, and antitumor responses by activating effector immune cells and enhancing antigen presentation . It associates with the transmembrane accessory factor IFNGR2 to form a functional receptor complex . Upon ligand binding, the intracellular domain of IFNGR1 undergoes conformational changes to allow association of downstream signaling components JAK1 and JAK2 . Activated JAK1 phosphorylates IFNGR1, creating a docking site for STAT1, which subsequently undergoes phosphorylation, dimerization, and nuclear translocation to stimulate target gene transcription . Additionally, STAT3 can be activated in a similar manner, though this activation appears weaker .

What applications are IFNGR1 antibodies suitable for?

IFNGR1 antibodies have been validated for multiple experimental applications in research settings. Depending on the specific antibody clone and formulation, they can be effectively utilized in:

ApplicationValidated AntibodiesSample TypesNotes
Western Blot (WB)ab134070, ab154400Human cell lysatesBand typically observed at 70-75 kDa
Immunohistochemistry (IHC-P)ab134070, ab154400Human tissue (paraffin-embedded)Heat-mediated antigen retrieval recommended
Immunocytochemistry (ICC/IF)ab134070Human cell lines1/100 dilution typically effective
Flow Cytometry (Intracellular)ab134070Fixed/permeabilized cells>5000 events collected using 488nm laser

These applications have been validated through rigorous testing, with antibody specificity confirmed using knockout cell lines as negative controls .

How should I validate IFNGR1 antibody specificity in my experiments?

Validating antibody specificity is crucial for generating reliable research data. For IFNGR1 antibodies, a multi-faceted approach is recommended:

  • Knockout validation: Compare staining patterns between wild-type cells and IFNGR1 knockout cells. The antibody should show specific binding in wild-type cells with signal loss in knockout cells . This has been demonstrated with antibodies like ab134070 in HeLa and HEK293 knockout cell lines.

  • Multiple detection methods: Confirm expression using at least two different techniques (e.g., Western blot and immunofluorescence).

  • Positive controls: Include known IFNGR1-expressing tissues or cell lines such as MCF7 cells or human tonsil tissue .

  • Negative controls: Include isotype controls at equivalent concentrations to rule out non-specific binding.

  • Molecular weight verification: In Western blots, verify that the detected band appears at the expected molecular weight (approximately 70-75 kDa for IFNGR1) .

How do surface dynamics of IFNGR1 affect antibody-antigen interactions?

The interfacial dynamics between IFNGR1 and anti-IFNGR1 antibodies are critical for optimal binding and recognition. Recent research employing THz spectroscopy and molecular dynamics simulations has revealed that sample preparation methods significantly influence the reorganization of surface residue dynamics at the solvent-protein interface . This reorganization leads to both structural and kinetic heterogeneous dynamics that ultimately enhance the binding probability at the antigen binding site.

Specifically, modified interfacial dynamics of anti-IFNGR1 are directly associated with alterations in the complementarity-determining regions (CDRs) that designate both antigen-antibody affinity and recognition . These modifications can be influenced by:

  • Hydrogen-bonding networks at the interface

  • Van der Waals interactions between complementary surfaces

  • Hydration-related entropic and enthalpic contributions

  • Conformational adaptations upon binding

Understanding these dynamics is particularly important when developing highly specific antibodies for therapeutic and diagnostic applications targeting the IFNGR1 receptor.

What methodological considerations are important when studying IFNGR1 in disease models?

When investigating IFNGR1 in disease models, particularly in the context of immunodeficiency, several methodological considerations are crucial:

  • Model selection: Mouse models such as C57BL/6 Ifngr1^-/-^ have been established for studying IFNGR1 deficiency and should be selected based on research questions .

  • IFN-γ levels: IFNGR1-deficient subjects typically exhibit elevated IFN-γ levels which can influence experimental outcomes. Consider measuring and potentially neutralizing circulating IFN-γ when studying transplantation or cell-based therapies .

  • Mycobacterial challenge: Since IFNGR1 deficiency predisposes to mycobacterial infections, incorporating mycobacterial challenge (e.g., with M. bovis BCG) can provide clinically relevant insights .

  • Granuloma formation analysis: Evaluate granuloma formation capacity as this process is impaired in IFNGR1 deficiency and represents a critical endpoint in antimycobacterial immunity .

  • Stem cell transplantation studies: When conducting hematopoietic stem cell transplantation (HSCT) studies in IFNGR1-deficient models, account for the unusually high rejection rates that characterize this condition .

How can I optimize IFNGR1 antibody use in multiplexed immunoassays?

Optimizing IFNGR1 antibody performance in multiplexed immunoassays requires careful consideration of several technical factors:

  • Antibody pair selection: For sandwich-based assays, select non-competing antibody pairs that recognize different epitopes on IFNGR1. Monoclonal antibodies like EPR7866 (ab134070) can be paired with polyclonal antibodies for optimal detection .

  • Cross-reactivity testing: Prior to multiplexing, test each antibody individually and in combination to identify potential cross-reactivity with other targets or secondary reagents.

  • Signal optimization: Titrate antibody concentrations to determine optimal signal-to-noise ratios. For fluorescence-based detection, starting dilutions of 1:100-1:1000 are typically effective for primary antibodies .

  • Blocking optimization: Determine the most effective blocking reagent (e.g., 3% milk in TBS-0.1% Tween) to minimize background without compromising specific signal detection .

  • Sequential incubation strategy: For challenging multiplexed assays, consider sequential rather than simultaneous incubation of primary antibodies to minimize potential interference.

  • Validation controls: Include positive controls (IFNGR1-expressing cells), negative controls (IFNGR1 knockout cells), and technical controls (secondary-only staining) in each assay .

Why might I observe inconsistent IFNGR1 staining patterns in tissue sections?

Inconsistent staining patterns with IFNGR1 antibodies in tissue sections can result from several factors:

  • Antigen retrieval efficiency: IFNGR1 detection in paraffin-embedded tissues typically requires heat-mediated antigen retrieval . Insufficient or excessive retrieval can lead to variable staining patterns. Systematically optimize temperature, duration, and buffer composition for your specific tissue type.

  • Fixation variability: Differences in fixation protocols (duration, fixative type, post-fixation processing) can affect epitope accessibility. Standardize fixation conditions and consider testing different antibody clones if variability persists.

  • IFNGR1 expression heterogeneity: Natural biological variation in IFNGR1 expression levels between different cell types within a tissue section can be misinterpreted as technical inconsistency . Include positive control tissues with known IFNGR1 expression patterns (e.g., human tonsil) to establish expected staining patterns.

  • Antibody concentration: Titrate antibody concentrations (1:50-1:200 dilutions are common starting points for IHC-P) to determine optimal signal-to-noise ratio for each specific tissue type .

  • Detection system sensitivity: Different detection methods (fluorescent vs. chromogenic) have varying sensitivity and dynamic range. For low-abundance IFNGR1 expression, amplification systems or more sensitive detection methods may be required.

What strategies can resolve false negative results in IFNGR1 Western blots?

False negative results in IFNGR1 Western blots can be addressed through systematic troubleshooting:

  • Sample preparation optimization:

    • Ensure complete cell lysis using appropriate buffers containing protease inhibitors

    • Avoid excessive heating of samples which may cause protein aggregation

    • Consider non-reducing conditions if the epitope involves disulfide bonds

  • Loading control verification:

    • Always include housekeeping protein controls (e.g., GAPDH, alpha-Tubulin) to confirm successful protein transfer

    • Consider Ponceau S staining of membranes to verify protein transfer efficiency

  • Antibody selection and concentration:

    • Test multiple IFNGR1 antibodies targeting different epitopes

    • Increase antibody concentration (1:500-1:1000 dilutions are typical starting points)

    • Extend primary antibody incubation time (overnight at 4°C often improves signal)

  • Signal enhancement:

    • Implement more sensitive detection systems (chemiluminescent vs. colorimetric)

    • Consider signal amplification methods for low-abundance targets

    • Optimize exposure times for digital imaging systems

  • Membrane blocking optimization:

    • Test different blocking reagents (BSA vs. milk) as some may mask specific epitopes

    • Ensure appropriate blocking duration (typically 1 hour at room temperature)

How does IFNGR1 deficiency impact hematopoietic stem cell transplantation outcomes?

IFNGR1 deficiency presents unique challenges in hematopoietic stem cell transplantation (HSCT), the only curative treatment available for this condition. Research has revealed several critical factors affecting transplantation outcomes:

  • Elevated IFN-γ levels: Patients with IFNGR1 deficiency exhibit abnormally high circulating IFN-γ levels, which have been directly implicated in graft rejection . Experimental evidence from mouse models demonstrates that neutralization of IFN-γ significantly improves engraftment rates.

  • Unusual rejection mechanisms: Unlike other primary immunodeficiencies, IFNGR1 deficiency is associated with an extraordinarily high rate of graft rejection, even with HLA-identical intra-familial transplants . This phenomenon is unique to this condition and suggests specific immunological mechanisms.

  • Therapeutic approaches: Research indicates that pre-transplant IFN-γ neutralization strategies may improve outcomes. In mouse models, antibodies that remove circulating IFN-γ permitted successful engraftment even in infected IFNGR1-deficient recipients .

  • Mycobacterial infection impact: Active mycobacterial infection at the time of transplantation significantly worsens outcomes, likely through further elevation of IFN-γ levels . Optimal infection control prior to transplantation is therefore critical.

  • Conditioning regimen considerations: Standard conditioning regimens may be insufficient to overcome the unique barriers to engraftment in IFNGR1 deficiency, necessitating modified approaches that specifically address the elevated IFN-γ environment.

What role does IFNGR1 signaling play in tumor immunology and cancer therapy development?

IFNGR1 signaling has emerged as a critical component in tumor immunology with significant implications for cancer therapy development:

  • Anti-tumor immune responses: IFNGR1-mediated signaling plays crucial roles in anti-tumor responses by activating effector immune cells and enhancing antigen presentation . This pathway is essential for effective immune surveillance against developing malignancies.

  • JAK-STAT pathway regulation: Upon IFN-γ binding, IFNGR1 initiates a signaling cascade involving JAK1/JAK2 activation and subsequent STAT1 phosphorylation, dimerization, and nuclear translocation . This pathway regulates genes involved in antigen processing and presentation, cell cycle control, and apoptosis.

  • SOCS1 negative regulation: IFNGR1 intracellular domain phosphorylation provides a docking site for SOCS1, which regulates the JAK-STAT pathway by competing with STAT1 binding to IFNGR1 . This negative feedback mechanism represents a potential therapeutic target.

  • Therapeutic antibody development: Antibodies targeting IFNGR1 or its signaling components may offer novel approaches for modulating immune responses in cancer. Understanding the structural and kinetic dynamics of IFNGR1-antibody interactions is essential for this development .

  • Biomarker potential: IFNGR1 expression levels and signaling pathway integrity may serve as biomarkers for predicting response to immunotherapies, particularly immune checkpoint inhibitors that rely on functional IFN-γ signaling for efficacy.

How might advances in structural biology improve IFNGR1 antibody development?

Recent advances in structural biology offer promising avenues for enhancing IFNGR1 antibody development:

  • Cryo-electron microscopy applications: High-resolution structural analysis of the IFNGR1-antibody complex using cryo-EM can reveal precise epitope-paratope interactions, facilitating rational antibody engineering with improved specificity and affinity.

  • Molecular dynamics simulations: Computational approaches can provide insights into the reorganization of sample surface residue dynamics at the solvent-protein interface, which directly influences antibody binding . These simulations can guide the optimization of antibody complementarity-determining regions (CDRs).

  • Hydrogen-bonding network analysis: Detailed mapping of hydrogen-bonding networks between IFNGR1 and antibodies can identify critical interaction points that stabilize the complex . This information can inform site-directed mutagenesis strategies to enhance binding properties.

  • Hydration shell characterization: The role of solvent molecules in the hydration shell is essential for stabilizing antigen-antibody complexes . Advanced spectroscopic techniques combined with computational modeling can define these contributions and guide antibody design.

  • Protein engineering approaches: Structure-guided protein engineering can develop antibodies with enhanced specificity, reduced cross-reactivity, and improved stability for both research and therapeutic applications targeting IFNGR1.

What are the implications of IFNGR1 dynamics for personalized medicine approaches?

Understanding IFNGR1 dynamics has significant implications for developing personalized medicine approaches:

  • Genetic variation impact: Different mutations in the IFNGR1 gene lead to varying degrees of receptor dysfunction, from complete to partial deficiency . Characterizing these variations can guide personalized treatment strategies for patients with IFNGR1-related immunodeficiencies.

  • Biomarker development: IFNGR1 expression patterns and signaling pathway integrity could serve as biomarkers for predicting response to immunotherapies across multiple disease contexts, enabling patient stratification for optimal treatment selection.

  • Tailored transplantation protocols: For patients with IFNGR1 deficiency requiring hematopoietic stem cell transplantation, personalized conditioning regimens that account for individual IFN-γ levels could significantly improve engraftment success rates .

  • Antibody-based therapeutic targeting: Patient-specific antibody therapies could be developed to address particular IFNGR1 variants or expression patterns, offering precision approaches for modulating immune responses in autoimmunity, cancer, and infectious diseases.

  • Combination therapy optimization: Understanding individual differences in IFNGR1 signaling can inform optimal combinations of immunomodulatory agents, particularly in cancer immunotherapy where IFN-γ pathway integrity is critical for response to checkpoint inhibitors.

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.