Mouse IFN-γ (mIFN-γ) is a pleiotropic cytokine that functions as a macrophage-activating factor. It shares approximately 41% sequence identity with human IFN-γ (hIFN-γ) . Despite this relatively low homology, both variants perform similar immunoregulatory functions in their respective species. The active form of mouse IFN-γ exists as an antiparallel dimer that triggers the IFN-γ/JAK/STAT pathway . This dimerization is essential for receptor binding and subsequent biological activity.
The molecular weight of mouse IFN-γ is approximately 20 kDa, which may vary slightly when produced with a His-tag for purification purposes . Understanding these structural differences is crucial when designing cross-species experiments or interpreting results from mouse models in the context of human disease.
Mouse IFN-γ is produced by multiple immune cell populations including:
Interestingly, research has demonstrated that mouse peritoneal macrophages can constitutively express IFN-γ, suggesting an autocrine regulatory mechanism that may play an important role in macrophage function and immune response coordination . This finding challenges the traditional view that IFN-γ production is restricted to lymphoid cells and highlights the complex network of cytokine regulation in the immune system.
Research indicates that glycosylation of mouse IFN-γ does not affect its biological activity . This finding has significant implications for recombinant protein production, as it suggests that non-glycosylated recombinant IFN-γ (such as that produced in bacterial expression systems like E. coli) should maintain full biological functionality.
For His-tagged mouse IFN-γ specifically, the addition of the histidine tag generally does not interfere with the protein's biological activity. This makes His-tagged versions particularly useful for research applications as they combine ease of purification with preserved functionality.
Mouse IFN-γ initiates signaling by binding to its heterodimeric receptor composed of IFNGR1 and IFNGR2 subunits. This binding event triggers a signaling cascade primarily involving:
Janus kinase 1 (JAK1)
Janus kinase 2 (JAK2)
Upon receptor engagement, conformational changes in the receptor's intracellular domains activate JAK1 and JAK2, which subsequently phosphorylate STAT1. Phosphorylated STAT1 forms homodimers that translocate to the nucleus and bind to gamma-activated sequence (GAS) elements in the promoters of IFN-γ-responsive genes, inducing transcriptional changes that mediate IFN-γ's biological effects.
Yes, mouse IFN-γ can upregulate its own gene expression through an autocrine mechanism, particularly in peritoneal macrophages . When mouse peritoneal macrophages are treated with IFN-γ, a significant accumulation of IFN-γ mRNA occurs, followed by secretion of IFN-γ protein after 24-48 hours.
This self-regulatory capacity is not observed in mouse lymphocytes from mesenteric lymph nodes or in various mouse cell lines, indicating cell type specificity . The upregulation of IFN-γ expression can also be found in peritoneal macrophages from anti-asialo GM1-treated nude mice, further supporting an autocrine regulatory mechanism in these cells. This self-amplification loop may be critical for sustaining appropriate immune responses in specific tissue microenvironments.
Mouse IFN-γ orchestrates diverse biological functions related to host defense and immune regulation, including:
Antiviral and antiparasitic defense mechanisms
Apoptosis induction in target cells
Inflammatory response regulation
Innate and acquired immunity coordination
Macrophage activation and polarization
Cell proliferation inhibition (for both normal and transformed cells)
IFN-γ signaling recruits and activates various immune cell populations, including macrophages, natural killer cells, and cytotoxic T lymphocytes . This cytokine network creates a coordinated immune response against pathogens and tumors. Paradoxically, IFN-γ has also been implicated in resistance to NK cell and CTL responses and in immune escape mechanisms in certain cancer contexts .
For maximum stability and activity of His-tagged mouse IFN-γ, follow these guidelines:
Reconstitution Protocol:
Lyophilized protein should be reconstituted in sterile water or appropriate buffer (typically PBS with 0.1% BSA as a carrier protein)
Gently mix by swirling or pipetting; avoid vigorous vortexing that may cause protein denaturation
Allow complete dissolution before determining concentration
Storage Recommendations:
Long-term storage: -80°C in single-use aliquots to prevent freeze-thaw cycles
Medium-term storage: -20°C for up to 3 months
Working solution: 2-8°C for up to 1 week
Stability Considerations:
Avoid repeated freeze-thaw cycles as they significantly compromise protein activity
For His-tagged preparations specifically, monitor for potential aggregation as His-tags can sometimes promote protein-protein interactions
When designing dose-response experiments with recombinant mouse IFN-γ, consider these methodological recommendations:
Concentration Range:
For cell activation studies: 1-100 ng/mL, with typical effective concentrations around 5-20 ng/mL
For ELISA standard curves: serial dilutions starting from 2000 pg/mL as recommended for commercial kits
Experimental Timeline:
Acute responses (signaling activation): 15 minutes - 6 hours
Gene expression changes: 6-24 hours
Phenotypic alterations: 24-72 hours
Control Conditions:
Include appropriate vehicle controls matching the reconstitution buffer
Consider including a heat-inactivated IFN-γ control to confirm specificity
For His-tagged proteins, compare with non-tagged versions in critical experiments to rule out tag-mediated effects
Readout Selection:
Select appropriate readouts based on your research question (e.g., STAT1 phosphorylation for signaling, specific gene expression for transcriptional responses, functional assays for biological effects)
Several complementary approaches can be used to measure mouse IFN-γ:
Enzyme-Linked Immunosorbent Assay (ELISA):
Commercial kits available with detection ranges typically from 15-2000 pg/mL
Suitable for cell culture supernatants, serum, plasma, and tissue homogenates
Follow manufacturer protocols for sample dilution and preparation
Western Blotting:
Detects mouse IFN-γ protein at approximately 17-20 kDa
Useful for cell and tissue lysates
Can confirm protein integrity and detect potential degradation products
Flow Cytometry:
Intracellular staining for IFN-γ in specific cell populations
Requires cell permeabilization and fluorescently-labeled antibodies
Provides information about the cellular sources of IFN-γ in heterogeneous samples
Quantitative PCR (qPCR):
Measures IFN-γ mRNA expression levels
Requires appropriate reference genes for accurate normalization
Useful for studying transcriptional regulation when protein detection is challenging
When working with His-tagged mouse IFN-γ, researchers should consider these important factors:
Purification Advantages:
The His-tag facilitates protein purification using nickel affinity chromatography
Results in high-purity preparations suitable for sensitive immunological assays
Allows for stringent washing conditions to remove contaminants
Potential Interference Considerations:
While His-tags generally have minimal impact on protein function, they might:
Slightly alter protein folding or dimerization kinetics
Create non-specific interactions in certain experimental systems
Influence protein half-life in vivo
Validation Approaches:
Compare biological activity with non-tagged versions in key experiments
Consider tag removal using protease cleavage if a cleavage site is incorporated
Include appropriate controls for non-specific effects of the tag itself
Detection Advantages:
His-tagged preparations can be detected using anti-His antibodies, providing an additional detection method independent of IFN-γ-specific antibodies
Useful for tracking protein distribution or confirming expression in complex systems
Mouse IFN-γ has significant functions in neuroinflammatory disease contexts, as demonstrated in models of multiple system atrophy (MSA):
In a mouse model of MSA characterized by alpha-synuclein overexpression in oligodendrocytes, researchers observed:
Infiltration of CD4+ and CD8+ T cells into the brain
Increased CD4+ T-cells positive for the transcription factor T-bet
Significant production of IFN-γ by infiltrating immune cells
Further mechanistic studies using genetic and pharmacological approaches demonstrated that:
IFN-γ is produced primarily by infiltrating CD4+ T-cells
IFN-γ mediates key pathological mechanisms driving MSA progression
IFN-γ-neutralizing antibody treatment attenuated disease progression
These findings suggest IFN-γ represents a potential therapeutic target in MSA, with researchers noting that "IFNγ represents a potential future disease-modifying therapeutic target in multiple system atrophy" . This research highlights the importance of understanding IFN-γ's functions in specific disease contexts to develop targeted therapeutic approaches.
When designing experiments to target IFN-γ signaling in mouse models, consider these methodological approaches:
Neutralizing Antibodies:
Anti-IFN-γ monoclonal antibodies can effectively block IFN-γ activity in vivo
Timing is critical - treatment before disease onset tests preventive effects, while treatment after onset assesses therapeutic potential
Dosing frequency must account for antibody half-life and tissue distribution
Researchers have successfully used IFN-γ-neutralizing antibody treatment in mouse models of MSA both before and after disease onset
Genetic Approaches:
Complete IFN-γ knockout mice provide information about developmental and systemic effects
Conditional knockout systems using Cre-lox technology allow for tissue-specific or temporal control of gene deletion
CRISPR/Cas9-mediated editing enables precise modification of IFN-γ pathway components
Receptor Targeting:
Blocking antibodies against IFNGR1 or IFNGR2 can inhibit signaling
Small molecule inhibitors of downstream signaling components (JAK/STAT) provide alternative approaches
Soluble receptor domains can act as decoys to sequester IFN-γ
Readout Selection:
Include multiple readouts spanning signaling (STAT1 phosphorylation), transcriptional (IFN-γ-responsive genes), and functional (disease parameters) endpoints
Account for potential compensatory mechanisms in chronic models
The interaction between the microbiome and IFN-γ responses in mouse models represents an emerging research area with important implications:
Airway Microbiota Interactions:
Respiratory exposure to specific microbes can modulate airway inflammation and immune responses
Studies incorporating "wild" mouse microbes in laboratory settings better align mouse models with human disease responses
Natural members of the mouse microbiota, such as Bordetella pseudohinzii, can persist in the respiratory tract for months and influence immune parameters without causing overt illness
Experimental Approaches:
Gnotobiotic versus conventional mice comparisons reveal microbiome-dependent differences in IFN-γ production
Colonization with defined microbial communities allows for controlled studies of specific microbe-immune interactions
Antibiotic treatment can temporarily alter microbiota composition to study acute effects on IFN-γ responses
Observed Effects:
Airway colonization with B. pseudohinzii increased both effector T cells and T regulatory cells in mouse lungs
Interestingly, this colonization preferentially increased IL-17A-expressing T cells rather than IFN-γ-producing Th1 cells
This demonstrates how specific members of the microbiome can shape the balance between different T cell subsets and their cytokine production profiles
These findings highlight the importance of considering microbiome variables when studying IFN-γ responses in mouse models, particularly when translating results to human disease contexts.
Researchers now have access to sophisticated computational tools for predicting IFN-γ-inducing peptides:
IFNepitope2:
A host-specific technique for annotating IFN-γ inducing peptides
Updated version of the IFNepitope platform introduced by Dhanda et al.
Utilizes a dataset containing 7,983 experimentally validated IFN-γ inducing peptides specifically for mouse hosts
Methodological Approach:
Combines machine learning techniques with similarity-based methods (BLAST)
Employs compositional features, particularly dipeptide composition, which outperforms one-hot encoding or binary profile approaches
The hybrid model achieved an AUROC of 0.85 for mouse host predictions
Applications:
Design of peptide-based vaccines with enhanced immunogenicity
Development of immunomodulatory peptides for therapeutic applications
Screening potential epitopes from pathogen proteins for vaccine development
Rational design of experimental tools to study IFN-γ responses in specific contexts
This resource is available as both a web server (https://webs.iiitd.edu.in/raghava/ifnepitope2/) and as a standalone application or Python package (https://github.com/raghavagps/ifnepitope2)[6].
Mouse IFN-γ serves as a valuable tool in cancer immunotherapy research due to its pleiotropic effects on immune responses and tumor cells:
Direct Anti-tumor Effects:
Upregulates MHC class I expression on tumor cells, enhancing their recognition by cytotoxic T lymphocytes
Induces growth arrest and apoptosis in certain tumor cell lines
Inhibits angiogenesis, limiting tumor vascularization and growth
Immune Modulation Applications:
Enhances dendritic cell maturation and antigen presentation
Polarizes macrophages toward an M1 (anti-tumor) phenotype
Augments NK cell cytotoxicity against tumor targets
Paradoxical Effects:
IFN-γ is also implicated in resistance to NK cell and CTL responses
Can contribute to immune escape mechanisms in various cancer types
May induce PDL1 expression on tumor cells, potentially limiting anti-tumor immunity
Experimental Approaches:
Combination with checkpoint inhibitors to overcome potential immunosuppressive effects
Local delivery systems to achieve high intratumoral concentrations while limiting systemic toxicity
Genetic engineering of tumor-infiltrating lymphocytes or CAR-T cells to produce IFN-γ at tumor sites
Several cutting-edge approaches have emerged for studying IFN-γ-dependent pathways in mouse models:
Reporter Systems:
IFN-γ reporter mice expressing fluorescent proteins under the control of the IFN-γ promoter
Allow real-time visualization of IFN-γ expression patterns in vivo
Can be combined with intravital microscopy for longitudinal studies
Single-cell Technologies:
Single-cell RNA sequencing reveals cell-specific responses to IFN-γ stimulation
Mass cytometry (CyTOF) enables simultaneous detection of multiple signaling nodes
Spatial transcriptomics provides information about IFN-γ pathway activation in tissue context
CRISPR/Cas9 Applications:
Precise gene editing to create novel mouse models with specific mutations in IFN-γ pathway components
CRISPR screening approaches to identify new regulators of IFN-γ responses
In vivo CRISPR delivery for tissue-specific modification of pathway components
Optogenetic and Chemogenetic Tools:
Allow temporal and spatial control of IFN-γ signaling
Enable studies of the acute versus chronic effects of pathway activation
Provide unprecedented precision in manipulating specific aspects of the signaling cascade
Interferon-gamma (IFN-γ) is a critical cytokine belonging to the type II interferon family. It plays a pivotal role in the immune response, particularly in antiviral, immunoregulatory, and anti-tumor activities. The recombinant form of mouse IFN-γ, tagged with a polyhistidine (His) tag, is widely used in research to study its biological functions and interactions.
The recombinant mouse IFN-γ protein is typically produced in HEK293 cells, a human embryonic kidney cell line, which ensures proper folding and post-translational modifications. The protein consists of 144 amino acids and has a calculated molecular mass of approximately 17 kDa. Due to glycosylation, its apparent molecular mass is around 21 kDa when analyzed by SDS-PAGE under reducing conditions .
The DNA sequence encoding mouse IFN-γ (accession number P01580) is expressed with a polyhistidine tag at the C-terminus. This His tag facilitates the purification of the protein using immobilized metal affinity chromatography (IMAC), which exploits the affinity between histidine residues and metal ions like nickel or cobalt .
Recombinant mouse IFN-γ exhibits significant biological activity. It is measured in antiviral assays using L929 cells infected with vesicular stomatitis virus (VSV), where the effective dose (ED50) typically ranges from 0.2 to 1 ng/mL. Additionally, its binding ability is assessed through functional ELISA, demonstrating interaction with mouse IFN-γ receptor 1 (IFNGR1) fused with an Fc tag .
IFN-γ is predominantly produced by natural killer (NK) cells, natural killer T (NKT) cells, and effector T cells (CD4+ and CD8+). It is a key player in the innate and adaptive immune responses. IFN-γ activates macrophages, enhances antigen presentation, and promotes the differentiation of T helper 1 (Th1) cells. It also has direct antiviral effects and can inhibit the proliferation of certain tumor cells .
The recombinant mouse IFN-γ protein is extensively used in immunological research to: