IFNG Antibody Pair

Shipped with Ice Packs
In Stock

Product Specs

Buffer
**Capture Buffer:** 50% Glycerol, 0.01M PBS, pH 7.4
**Detection Buffer:** 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
We typically ship products within 1-3 business days of receiving your order. Delivery times may vary depending on the shipping method and destination. Please contact your local distributor for specific delivery time estimates.
Notes
We recommend using the capture antibody at a concentration of 0.2 µg/mL and the detection antibody at a concentration of 0.25 µg/mL. Optimal dilutions should be determined experimentally by the researcher.
Synonyms
IFN-gamma,Immune interferon,IFNG,
Target Names

Q&A

What is an IFNG antibody pair and how does it function in immunoassays?

An IFNG antibody pair consists of two carefully matched antibodies (capture and detector) designed to bind different epitopes on the interferon gamma protein. In immunoassays such as ELISA, the capture antibody is immobilized on a solid surface (typically a microplate), while the detector antibody is used to form a sandwich complex with the target analyte. This principle enables specific and sensitive quantification of IFNG in biological samples.

The assay follows a sequential process:

  • A target-specific capture antibody is coated to the microplate wells (typically an overnight process)

  • Samples, standards, or controls are added and bind to the immobilized antibody

  • The detector antibody is added, forming a sandwich with the bound IFNG

  • A substrate solution reacts with the enzyme-antibody-target complex to produce measurable signal

  • Signal intensity correlates directly with IFNG concentration in the original sample

This sandwich format significantly enhances specificity compared to direct detection methods, as the target must be recognized by both antibodies to generate signal.

What biological roles does IFNG play that make it an important research target?

IFNG (Interferon gamma) serves as a critical immunoregulatory cytokine with pleiotropic effects that make it an important research target for multiple fields:

  • It functions as a macrophage activation factor produced primarily by T-lymphocytes and natural killer cells in response to antigens, mitogens, and other cytokines

  • IFNG plays crucial roles in antimicrobial, antiviral, and antitumor responses by activating effector immune cells and enhancing antigen presentation

  • It primarily signals through the JAK-STAT pathway after interaction with IFNGR1, leading to STAT1 activation, nuclear translocation, and transcription of IFNG-regulated genes

  • IFNG induces replacement of catalytic proteasome subunits with immunoproteasome subunits, increasing the quantity, quality, and repertoire of peptides for class I MHC loading

  • It upregulates MHC II complexes on cell surfaces by promoting expression of key molecules like cathepsins B/CTSB, H/CTSH, and L/CTSL

  • IFNG participates in hematopoietic stem cell regulation during development and homeostasis

Research targeting IFNG enables investigation of these pathways and their role in infectious disease, autoimmunity, and cancer.

How do researchers choose between ELISA, ELISPOT, and cytometric bead array for IFNG detection?

The selection between these methodologies depends on specific research questions and experimental constraints:

MethodPrimary ApplicationKey AdvantagesLimitationsSample Requirements
ELISAQuantitative measurement of IFNG in solutionWell-standardized, high throughput, quantitativeCannot identify cellular sourceSerum, plasma, culture supernatants
ELISPOTDetection of individual cytokine-producing cellsSingle-cell resolution, detection of rare populationsLimited quantification of cytokine amountSingle cell suspensions
Cytometric Bead ArrayMultiplex cytokine analysisSimultaneous detection of multiple cytokines, low sample volumeRequires flow cytometer, more complex analysisSerum, plasma, culture supernatants

For instance, ELISPOT is particularly valuable for measuring cytokine-producing cells in a single cell suspension with minimal in-vitro manipulation, making it ideal for ex-vivo analysis of antigen-specific T cells . Cytometric bead arrays provide an efficient alternative when sample volume is limited and multiple cytokines need to be analyzed simultaneously .

What are the critical parameters for optimizing IFNG antibody pair assays?

Optimizing IFNG antibody pair assays requires careful attention to several parameters:

  • Antibody Selection and Concentration: Screening of antibody pairs is essential as demonstrated in rhinoceros studies where commercial ELISA antibody pairs were evaluated for cross-reactivity . Optimal capture antibody concentration (typically 2 μg/mL) must be determined empirically .

  • Incubation Temperature: Studies have shown significant improvement in assay performance at 37°C compared to room temperature. For example, rhinoceros IFN-γ ELISA showed higher reproducibility with optimal recovery when incubated at 37°C .

  • Blocking Conditions: Proper blocking (e.g., 0.1% bovine serum albumin in PBS with 0.05% Tween 20) is critical to minimize nonspecific binding .

  • Sample Dilution: Serial dilutions should be performed to ensure measurements fall within the linear range of the assay (typically 7.8–125 pg/mL for human IFNG) .

  • Assay Linearity: Regression analysis should be performed to ensure linear response to both recombinant and endogenous IFNG. The correlation coefficient (R²) value should be evaluated to characterize assay linearity .

  • Parallelism Testing: Comparison of regression slopes for recombinant and endogenous IFNG is necessary to confirm that the assay recognizes both forms equivalently .

Systematic optimization of these parameters ensures reliable and reproducible results across experiments.

How can researchers troubleshoot low signal or high background issues in IFNG immunoassays?

When encountering performance issues with IFNG immunoassays, a systematic troubleshooting approach is recommended:

Low Signal Issues:

  • Antibody Functionality: Ensure antibodies retain binding activity; improper storage can lead to degradation. Test with positive controls known to contain high IFNG levels.

  • Cytokine Induction: For cell stimulation experiments, verify stimulation efficacy using positive controls like PMA/Ionomycin with Monensin as used in flow cytometry protocols .

  • Matrix Effects: Sample matrix can interfere with antibody binding. Test for matrix effects by spiking known concentrations of recombinant IFNG into sample matrix and comparing recovery to standard diluent .

  • Detection System: Ensure substrate and detection reagents are fresh and properly prepared.

High Background Issues:

  • Insufficient Blocking: Increase blocking buffer concentration or incubation time (≥1 hour at room temperature).

  • Cross-Reactivity: Test for cross-reactivity with related cytokines, particularly when using the assay in non-validated species .

  • Washing Steps: Increase number and volume of washes (≥4 times with 300 µL/well) .

  • Contamination: Cross-contamination between wells can occur; use proper technique and clean equipment.

A methodical approach that systematically modifies one parameter at a time allows identification of specific issues affecting assay performance.

What strategies are effective for extending the dynamic range of IFNG detection?

Researchers can implement several strategies to extend the dynamic range of IFNG detection:

  • Optimized Antibody Pairs: Selecting antibody pairs with complementary binding characteristics can inherently improve dynamic range. For example, the Human IFN gamma Matched Antibody Pair from commercial sources demonstrates a linear response in the range of 7.8–125 pg/mL .

  • Signal Amplification Systems: Employing streptavidin-HRP systems with optimized detector antibody concentrations can enhance signal without increasing background, effectively extending the lower limit of detection .

  • Sample Dilution Strategy: Implementing a multi-dilution approach where samples are tested at several dilutions allows measurement of samples with widely varying IFNG concentrations. This approach requires establishing parallelism between dilution curves and standards .

  • Alternative Detection Technologies: For samples outside the traditional ELISA range, cytometric bead array methods offer complementary approaches with different dynamic ranges .

  • Data Analysis Optimization: Using five-parameter logistic curve fitting rather than traditional four-parameter logistic models can improve accuracy at the extremes of the standard curve.

Implementation of these approaches can extend the functional dynamic range from the typical 7.8–125 pg/mL to potentially 1–1000 pg/mL depending on the specific antibody pair and detection system used.

How do researchers verify cross-species reactivity of IFNG antibody pairs?

Verifying cross-species reactivity requires a systematic approach:

  • Sequence Homology Analysis: Before experimental validation, researchers should examine sequence homology between species. Human IFNG shows approximately 40% sequence homology with mouse IFNG and 90% with rhesus IFNG . These differences explain why human IFNG antibodies typically don't cross-react with mouse but may work with non-human primates.

  • Parallelism Testing: Researchers should perform dilution series with both recombinant protein of the target species and endogenous samples (e.g., stimulated blood). Regression slopes should be compared statistically using methods such as F-tests to confirm parallel response .

  • Positive Control Validation: When testing in non-validated species, researchers should include robust positive controls known to induce IFNG, such as Phytohemagglutinin (PHA), Phorbol 12-myristate 13-acetate (PMA) with Ionomycin, or Pokeweed Mitogen (PWM) .

  • Recombinant Protein Spike Recovery: Spiking known quantities of recombinant protein into sample matrix helps establish recovery rates in the new species context .

A successful cross-species application was demonstrated with equine IFN-γ antibody pairs used for white rhinoceros samples, where linear response to both recombinant equine and endogenous rhinoceros IFN-γ was established .

What structural features of IFNG explain species-specificity constraints in antibody detection?

IFNG's structural characteristics significantly influence species-specificity in antibody detection:

  • Dimeric Structure: Functional IFNG exists as a homodimer of approximately 45 kDa, composed of two 146 amino acid subunits . This dimeric conformation creates complex epitopes that may not be conserved across species.

  • Glycosylation Patterns: IFNG is subject to differential glycosylation, appearing as a combination of 25, 20, and minor 15.5 kDa bands on SDS-PAGE . Glycosylation sites at positions like Asn25 and Asn97 in human IFNG are critical for protease resistance but may vary between species .

  • Receptor Binding Domains: The biological activity of the IFNG homodimer is highly species-specific because key regions involved in receptor binding are not well conserved. This explains why human IFNG antibodies targeting receptor-binding epitopes show poor cross-reactivity with mouse IFNG .

  • Sequence Divergence in Key Regions: While human IFNG shows approximately 40% sequence homology with mouse IFNG, specific epitope regions may have even lower conservation . Antibodies targeting these divergent regions will show strict species specificity.

These structural factors necessitate careful validation when applying IFNG antibody pairs across species and explain why species-specific antibody pairs are often required for optimal detection.

What role do neutralizing anti-IFNG antibodies play in therapeutic research and disease models?

Neutralizing anti-IFNG antibodies serve critical functions in therapeutic research and disease models:

  • Modulation of Immune Responses: Administration of anti-IFNG antibodies can regulate immune responses by blocking IFN-γ activity. Studies have demonstrated that treatment with clone XMG1.2 (rat anti-mouse IFN-γ IgG1) at doses of 200 μg significantly altered immune responses in mouse models .

  • Enhanced Vaccine Immunogenicity: Paradoxically, anti-IFNG antibodies can enhance the immunogenicity of recombinant adenoviral vectors. Research has shown that HIV-1 envelope-specific CTL responses were higher in IFN-γ-knockout mice than in wild-type mice following immunization with recombinant adenovirus. Administration of anti-IFN-γ antibodies augmented vaccine-elicited CTL responses in vivo .

  • Prevention of Experimental Cerebral Malaria: Monoclonal antibodies against interferon gamma have been shown to prevent experimental cerebral malaria and its associated overproduction of tumor necrosis factor .

  • Therapeutic Target in Autoimmune Conditions: Anti-IFNG antibody treatment has demonstrated efficacy in various experimental disease models, suggesting potential therapeutic applications .

  • Investigation of Anti-IFNG Autoantibodies in Human Disease: A rare cause of adult-onset immunodeficiency is associated with anti-interferon gamma autoantibodies (AIGA), leading to severe disseminated opportunistic infections. Studies have investigated the neutralizing mechanisms of these antibodies, finding they target a major epitope on free IFNγ crucial for receptor activation, thereby inhibiting IFNγ-induced pSTAT-1 phosphorylation and cytokine production .

The paradoxical enhancement of immune responses through anti-IFNG antibody treatment highlights the complex immunomodulatory role of this cytokine.

How can researchers detect and characterize anti-IFNG autoantibodies in clinical samples?

Detection and characterization of anti-IFNG autoantibodies require sophisticated methodological approaches:

  • Functional Neutralization Assays: The gold standard for detecting clinically relevant anti-IFNG autoantibodies involves measuring their ability to neutralize IFNγ activity. This can be assessed by evaluating the inhibition of IFNγ-dependent augmentation of LPS-induced TNFα production or by measuring suppression of IFNγ-induced pSTAT-1 phosphorylation .

  • Epitope Mapping: Advanced epitope mapping techniques can identify the specific regions on IFNγ targeted by autoantibodies. Studies have shown that pathogenic autoantibodies target epitopes crucial for IFNγ receptor activation . Techniques include:

    • Peptide arrays covering the entire IFNγ sequence

    • Competition binding assays with monoclonal antibodies of known epitope specificity

    • Hydrogen-deuterium exchange mass spectrometry to identify binding interfaces

  • Immunoglobulin Subclass Analysis: Determining the IgG subclass of anti-IFNG autoantibodies provides insight into their potential pathogenicity. This can be accomplished through subclass-specific secondary antibodies in ELISA formats .

  • Longitudinal Sample Analysis: Serial serum collection and analysis can track the development and persistence of autoantibodies. Research has shown that anti-IFNG antibodies may appear 18 months before clinical onset of associated disorders, suggesting their acquisition precedes disease manifestation .

  • Genetic Association Studies: HLA typing of patients with anti-IFNG autoantibodies has revealed associations with specific alleles including DRB116:02, DRB115:02, DQB105:02, and DQB105:01, suggesting genetic predisposition to their development .

These methodologies have contributed to our understanding of anti-IFNG autoantibody-associated immunodeficiency, which has been predominantly reported in Southeast Asian populations or Asian immigrants to other countries .

How are IFNG antibody pairs being engineered for enhanced selectivity in complex biological systems?

Recent advances in antibody engineering are revolutionizing the selectivity of IFNG detection and therapeutic applications:

  • Logic-Gated Antibody Pairs: Researchers have developed novel approaches like HexElect®, which creates IgG antibody pairs that only induce activation of oligomerization-dependent functions if both antibodies bind the same target cell. This technology effectively creates Boolean logic AND gates that integrate two antibody-binding signals, providing unprecedented selectivity .

  • Hexamerization-Enhancing Mutations: Engineering antibodies with hexamerization-enhancing mutations (like E430G) creates antibodies with unique functional properties. For example, when applied to anti-CD52 and anti-CD20 antibodies, these mutations enabled mutually dependent activation only when both targets were present on the same cell .

  • Recombinant Antibody Technology: The transition from hybridoma-derived to recombinant antibody production has enabled precise engineering of binding domains. Recombinant production provides unrivalled batch-to-batch consistency, easy scale-up, and future security of supply for IFNG antibody pairs .

  • Carrier-Free Formulations: Antibody pairs are now available in carrier-free formulations (BSA and azide-free), allowing researchers to perform custom conjugations for specialized applications without interference from carrier proteins .

These engineering approaches significantly expand the potential applications of IFNG antibody pairs beyond traditional detection methods, enabling precise targeting of specific cell populations and enhanced control over immune activation.

What methodological approaches allow researchers to assess IFNG production at the single-cell level?

Several sophisticated methodologies enable single-cell resolution analysis of IFNG production:

  • ELISPOT with Image Analysis: Enhanced ELISPOT techniques combined with automated spot counting and size analysis allow researchers to quantify not only the number of IFNG-producing cells but also the amount produced per cell based on spot size and intensity. This method requires minimal in-vitro manipulation, making it ideal for ex-vivo analysis .

  • Intracellular Cytokine Staining Flow Cytometry: Flow cytometry protocols using optimized fixation and permeabilization buffers (like FlowX FoxP3 Fixation & Permeabilization Buffer Kit) allow detection of intracellular IFNG in combination with surface markers. This approach has been successfully implemented for human PBMCs treated with PMA, Ionomycin, and Monensin, enabling identification of specific cellular sources of IFNG .

  • Single-Cell RNA Sequencing: This approach provides comprehensive analysis of IFNG gene expression alongside the entire transcriptome at single-cell resolution, allowing identification of novel cell populations and regulatory networks.

  • Mass Cytometry (CyTOF): Metal-tagged antibodies against IFNG combined with dozens of other cellular markers enable high-dimensional analysis of IFNG-producing cells with precise phenotypic characterization.

  • Functional Live Cell Imaging: Using fluorescent reporter systems linked to IFNG promoter activity allows real-time visualization of IFNG production in living cells, providing temporal information not available through fixed-cell approaches.

These methodologies collectively provide unprecedented insight into the cellular sources, regulation, and heterogeneity of IFNG production in complex biological systems.

How do emerging single-domain antibody technologies compare with traditional antibody pairs for IFNG detection?

Single-domain antibody technologies represent an emerging alternative to traditional antibody pairs for IFNG detection:

ParameterTraditional Antibody PairsSingle-Domain Antibodies
Size~150 kDa (full IgG)~12-15 kDa
StabilityModerate; sensitive to temperatureHigh thermal and pH stability
Tissue PenetrationLimited by sizeEnhanced due to smaller size
ProductionComplex mammalian cell systemsSimpler bacterial expression systems
MultiplexingRequires labeled secondary antibodiesDirect labeling more efficient
Target AccessLimited to exposed epitopesCan access cryptic epitopes
CostHigher production costsPotentially lower production costs

Key advantages of single-domain antibodies for IFNG detection include:

  • Improved Signal-to-Noise Ratio: The smaller size reduces steric hindrance, potentially allowing higher density of capture molecules and improved target binding kinetics.

  • Enhanced Stability: Single-domain antibodies typically show greater resistance to harsh conditions, enabling more robust assay formats.

  • Novel Epitope Recognition: Their unique binding properties may allow recognition of IFNG epitopes inaccessible to conventional antibodies, potentially improving specificity or cross-species reactivity.

  • Simplified Multiplexing: The compact nature of single-domain antibodies facilitates creation of fusion proteins and multi-specific constructs for advanced detection platforms.

While promising, these technologies are still emerging in the IFNG detection field, and extensive validation against gold-standard antibody pairs is needed before widespread adoption in critical research applications.

What are the key considerations for selecting the optimal IFNG detection system for specific research questions?

Selecting the optimal IFNG detection system requires thoughtful evaluation of several parameters:

  • Research Objective Alignment: Match the detection method to the specific question:

    • For quantifying IFNG in biological fluids: Sandwich ELISA or cytometric bead array

    • For enumerating IFNG-producing cells: ELISPOT or flow cytometry

    • For identifying cellular sources in tissue context: Immunohistochemistry or multiplexed imaging

  • Sample Type Compatibility: Consider the nature of your experimental samples:

    • Serum/plasma: Standard sandwich ELISA or multiplexed cytokine assays

    • Cell culture supernatants: High-sensitivity ELISA or cytometric bead arrays

    • Single cell suspensions: ELISPOT or intracellular cytokine staining

    • Tissue sections: Immunohistochemistry or multiplexed imaging

  • Sensitivity Requirements: Determine required detection limits:

    • Standard ELISA typically offers detection ranges of 7.8-125 pg/mL

    • Enhanced sensitivity may require amplification systems or alternative platforms

  • Species Considerations: Ensure antibody cross-reactivity with target species:

    • Human IFNG does not show cross-reactivity with mouse IFNG

    • Human IFNG shares ~40% sequence homology with mouse IFNG but ~90% with rhesus IFNG

  • Multiplexing Needs: If analyzing multiple cytokines, consider integrated systems:

    • Cytometric bead arrays allow simultaneous detection of multiple cytokines

    • Multiplex ELISA platforms offer higher throughput but may compromise sensitivity

  • Available Instrumentation: Match detection method to available equipment:

    • Standard ELISA requires only a plate reader

    • Flow cytometry and imaging approaches require specialized instrumentation

By systematically evaluating these parameters, researchers can select detection systems that optimize data quality while efficiently utilizing valuable biological samples.

What emerging technologies are likely to revolutionize IFNG detection and analysis in the next decade?

Several transformative technologies are poised to revolutionize IFNG detection and analysis:

  • CRISPR-Based Biosensors: CRISPR-Cas systems are being adapted as programmable biosensors for protein detection. These systems may offer unprecedented sensitivity for IFNG detection without requiring antibodies, potentially overcoming species-specificity limitations.

  • Single-Molecule Array (Simoa) Technology: Ultra-sensitive digital detection platforms can detect IFNG at femtomolar concentrations, enabling analysis of samples previously below detection limits. This could transform understanding of low-level IFNG signaling in homeostasis.

  • Spatial Transcriptomics and Proteomics: Integration of IFNG detection with spatial transcriptomics and proteomics will provide comprehensive understanding of IFNG's cellular sources and downstream effects within the tissue microenvironment context.

  • Machine Learning Integration: Advanced computational approaches will enable more sophisticated analysis of complex datasets, revealing previously unrecognized patterns in IFNG expression and regulation across diverse physiological and pathological conditions.

  • Microfluidic and Organ-on-Chip Systems: Integration of IFNG detection into microfluidic platforms will enable real-time monitoring of IFNG production and signaling in physiologically relevant tissue models, bridging the gap between in vitro and in vivo research.

  • Wearable Biosensors: Development of minimally invasive continuous monitoring systems for IFNG and other cytokines could transform clinical research by enabling longitudinal cytokine profiling in response to treatments or environmental changes.

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.