Recombinant Human Interferon-gamma (IFNG), partial (Active) (GMP)

Shipped with Ice Packs
In Stock

Description

Mechanism of Action

IFN-γ binds to its heterodimeric receptor (IFNGR1/IFNGR2), activating the JAK-STAT signaling pathway. This induces:

  • Immunoproteasome upregulation, enhancing MHC class I antigen presentation .

  • MHC class II expression via cathepsin induction (CTSB, CTSH, CTSL) .

  • Transcriptional activation of IRF1 and STAT1, amplifying antiviral and antitumor responses .

Heparan sulfate binding at the C-terminal D1/D2 clusters modulates receptor interaction, potentially regulating activity duration .

Approved Uses

  • Chronic Granulomatous Disease (CGD): Reduces severe infections by enhancing phagocyte oxidative burst .

  • Osteopetrosis: Promotes osteoclast function to alleviate bone density abnormalities .

Investigational Applications

ConditionPhaseOutcomeSource
Idiopathic Pulmonary Fibrosis (IPF)II↓CXCL5, PDGFA; ↑CXCL11, collagen remodeling
Advanced HIVIII50% reduction in opportunistic infections
Cutaneous LymphomaI/IITumor regression via TG-1041 adenoviral vector

Manufacturing and Quality Control

GMP-grade IFN-γ undergoes:

  • Reverse-phase chromatography for >99% purity .

  • Refolding optimization in Tris buffer (50–100 μg/mL) to preserve dimeric structure .

  • Bioactivity validation using antiviral assays (e.g., HeLa/EMC virus ED₅₀ = 0.15-0.75 ng/mL) .

Research Advancements

  • Purification Yield: 40 mg per gram of E. coli cell mass achieved via improved RPC methods .

  • Stability: Lyophilized formulations maintain activity for 6 months at -80°C .

  • Novel Delivery: Adenovirus-vectored IFN-γ (TG-1042) shows promise in melanoma models .

Product Specs

Buffer
Lyophilized from a 0.2 µm filtered concentrated solution in PBS, pH 5.0, with 3% Trehalose.
Form
Lyophilized powder
Lead Time
Typically, we can ship your orders within 5-10 business days of receipt. Delivery timelines may vary depending on the chosen shipping method and destination. Please contact your local distributor for specific delivery times.
Notes
Avoid repeated freezing and thawing. Store working aliquots at 4°C for up to one week.
Reconstitution
We recommend centrifuging the vial briefly before opening to ensure all contents settle to the bottom. Reconstitute the protein in sterile, deionized water to a concentration of 0.1-1.0 mg/mL. We recommend adding 5-50% glycerol (final concentration) and aliquotting for long-term storage at -20°C/-80°C. Our standard glycerol concentration is 50%, which can serve as a reference.
Shelf Life
The shelf life of our product is influenced by various factors including storage conditions, buffer components, temperature, and the inherent stability of the protein. Generally, liquid form has a 6-month shelf life at -20°C/-80°C, while lyophilized form has a 12-month shelf life at -20°C/-80°C.
Storage Condition
Store at -20°C/-80°C upon receipt. Aliquoting is necessary for multiple uses. Avoid repeated freeze-thaw cycles.
Tag Info
Tag-Free
Synonyms
Immune interferon,IFN-gamma
Datasheet & Coa
Please contact us to get it.
Expression Region
24-166aa
Mol. Weight
16.9 kDa
Protein Length
Partial
Purity
> 98 % by SDS-PAGE and HPLC analyses.
Research Area
Immunology
Source
E.Coli
Species
Homo sapiens (Human)
Target Names
Uniprot No.

Target Background

Function
Interferon-gamma (IFN-γ), a type II interferon produced by immune cells like T-cells and NK cells, plays a crucial role in antimicrobial, antiviral, and antitumor responses. IFN-γ activates effector immune cells and enhances antigen presentation, primarily through the JAK-STAT pathway. It binds to its receptor, IFNGR1, triggering the intracellular domain to open and allow the association of downstream signaling components JAK2, JAK1, and STAT1. This interaction leads to STAT1 activation, nuclear translocation, and the transcription of IFN-γ-regulated genes. Several induced genes are transcription factors, such as IRF1, which further regulate downstream transcription. IFN-γ influences the class I antigen presentation pathway by inducing a replacement of catalytic proteasome subunits with immunoproteasome subunits, thereby increasing the quantity, quality, and repertoire of peptides for class I MHC loading. It also enhances peptide generation by inducing the expression of activator PA28, which associates with the proteasome and alters its proteolytic cleavage preference. Additionally, IFN-γ upregulates MHC II complexes on the cell surface by promoting the expression of key molecules like cathepsins B/CTSB, H/CTSH, and L/CTSL. IFN-γ participates in the regulation of hematopoietic stem cells during development and homeostasis by affecting their development, quiescence, and differentiation.
Gene References Into Functions
  1. Impaired IFNgamma-Signaling and Mycobacterial Clearance in IFNgammaR1-Deficient Human iPSC-Derived Macrophages. PMID: 29249666
  2. High expression of IFNG is associated with breast cancer. PMID: 30336781
  3. Results suggest that the interplay of pro-inflammatory cytokines IFN-gamma derived from CD4+T lymphocytes and TNF-alpha from CD14+ cells has no direct additive impact on parasite replication but induces IL-4 production. PMID: 29953494
  4. IFN-lambda may participate in local inflammation in the salivary glands of primary Sjogren's syndrome patients through direct and indirect regulations of the expressions of BAFF and CXCL10 in salivary gland epithelium. PMID: 28421993
  5. The expression of CXCL10 mRNA and IFN-gamma mRNA was significantly higher in non-lesional and perilesional skin of vitiligo and alopecia areata patients compared with the skin of healthy controls; however, the level of expression of CXCL10 and IFN-gamma in lesional skin was not different than that in healthy skin. PMID: 27863059
  6. High IFNG expression is associated with Chronic Periodontitis. PMID: 30051674
  7. Hypomethylation of the IFNG promoter is significantly related to the risk of essential hypertension. PMID: 29643275
  8. These results demonstrated that the IFNGinduced immunosuppressive properties of B7H1 in human BM and WJMSCs were mediated by STAT1 signaling, and not by PI3K/RACalpha serine/threonineprotein kinase signaling. PMID: 29901104
  9. Serum IP-10 level and the IFN-gamma/IL-4 ratio have great potential to predict significant fibrosis among chronic hepatitis B patients. PMID: 28067328
  10. IFN-gamma increases free ISG15 levels in the cytoplasm and ISGylation in the nucleus and cytoplasm, but in a manner distinct between MCF-7 and MDA-MB-231cells. PMID: 29626479
  11. The expression of IFN-gamma and IL-17 was also suppressed by IRAK1/4 inhibitor both in active Behcet's patients and in normal subjects. PMID: 28780618
  12. IFNgamma induces a rapid activation of aerobic glycolysis followed by a reduction in oxidative phosphorylation in M1 macrophages. PMID: 29463472
  13. Results provide evidence that rs2069707 locus SNPs of IFN-gamma is a risk factor for contracting tuberculous pericarditis. PMID: 30017738
  14. No correlation was observed between interferon gamma mRNA/protein levels and recurrent depressive disorders. PMID: 29367100
  15. Aberrant IFN-gamma promoter methylation may be involved in the process of tumorigenesis of oral cancer. PMID: 28091876
  16. This study shows that elevated levels of interferon-gamma are associated with high levels of Epstein-Barr virus reactivation in patients with the intestinal type of gastric cancer. PMID: 29349089
  17. This study contributes to clarification of the previously inconsistent prognostic performance of IFNgamma by providing the first prognostic evaluation with long follow-up, time-dependence assessment and absence of any chemotherapy influence. PMID: 29478965
  18. Association Between the Interferon Gamma 874 T/A Polymorphism and the Severity of Valvular Damage in Patients with Rheumatic Heart Disease. PMID: 29332266
  19. IFN-gamma can promote cancer immunoevasion. (Review) PMID: 29283429
  20. An electrophoretic mobility shift assay showed that signal transducers and activators of transcription 1 (STAT1) attach to the GAS motif on the human STING promoter region. This indicates that IFN-gamma/Janus kinases/STAT1 signaling is essential for the STING upregulation in human keratinocytes. PMID: 29143896
  21. We review the direct and indirect effects of IFN-gamma on hematopoiesis, as well as the underlying signaling mechanisms of how IFN-gamma modulates the self-renewal, cell cycle entry, and proliferation of hematopoietic stem cells. PMID: 28852997
  22. IFN-gamma +874T allele may increase the risk of ocular lesions in Toxoplasma infection. The principle of natural selection seems to also play a role. The less common TNF-308A allelic form could be protective against the development of Toxoplasma ocular infection. PMID: 27081842
  23. This study shows the age-related reductions in serum and PBMC IFN-gamma in healthy nonobese subjects. PMID: 28762199
  24. Phosphorylation of T-bet by RSK2 is required for IFNgamma expression for attenuation of colon cancer metastasis and growth. PMID: 29133416
  25. SNX8 mediates IFNG-triggered non-canonical signaling pathway and host defense against Listeria monocytogenes. PMID: 29180417
  26. The frequencies of IFNgamma and IL-17A(+) cells were increased in the antrum, particularly in patients with H. pylori induced gastric ulcers. PMID: 28683359
  27. rs2069718 in the IFNG gene was significantly associated with pulmonary tuberculosis but not spinal tuberculosis. PMID: 28867622
  28. IFN-gamma was associated with a cerebral volume reduction in systemic lupus erythematosus with central nervous system involvement. PMID: 28848179
  29. Data suggest that semen exhibits substantial individual variation over time in pro-inflammatory seminal fluid cytokines IFNG and CXCL8. (IFNG = interferon gamma; CXCL8 = C-X-C motif chemokine ligand 8) PMID: 28541460
  30. Dysregulation of the IFN-gamma-STAT1 signaling pathway in a cell line model of large granular lymphocyte leukemia. PMID: 29474442
  31. STAT1b plays a key role in enhancing the tumor suppressor function of STAT1a, in ESCC, in a manner that can be amplified by IFN-gamma. PMID: 28981100
  32. Epigenetic silencing by single CpG methylation determines differential IL18BP inducibility in monocytic versus epithelial cells. PMID: 29409936
  33. Systemic IFN activation is associated with higher activity only in the ESSDAI biological domain but not in other domains or the total score. Our data raise the possibility that the ESSDAI biological domain score may be a more sensitive endpoint for trials targeting either IFN pathway. PMID: 29474655
  34. These findings suggest that IFN-a can inhibit HCV replication through a STAT2-dependent but STAT1-independent pathway, whereas IFN-g induces ISG expression and inhibits HCV replication exclusively through a STAT1- and STAT2-dependent pathway. PMID: 27929099
  35. rs1861493 and rs2430561 polymorphisms were conformed to be in HWE in genotypes distribution of the control group (P>0.05 for both). However, only TT genotype and T allele of rs2430561 presented significantly higher frequencies in Ankylosing Spondylitis patients than in healthy controls. IFN-g rs2430561 polymorphism may contribute to the risk of IFN-g rs2430561 polymorphism may contributhrough influencing IFN-g expression. PMID: 28843049
  36. Interactions among polymorphisms of IFN-gamma+874 AA, IL-2-330 TT, IL-10-1082 AA, IL10--592 AC and IL-4-589 CC/CT significantly influenced the clinical progression of the subjects with hepatitis B virus and/or hepatitis C virus infection. PMID: 28838891
  37. Ex vivo interferon-gamma production is a useful biomarker for assessing disease activity and predicting poor clinical outcomes of systemic lupus erythematosus patients. PMID: 28841837
  38. This study demonstrates reduced IFN-gamma production in chronic brucellosis patients. PMID: 28919584
  39. These data suggest that the de novo expression of PDL1 on tumor cells is upregulated by IFN-g secreted from CD8+ TILs upon recognition of the tumor cells with an MHC class I molecule. PMID: 28791392
  40. H pylori expression of cgt reduces cholesterol levels in infected gastric epithelial cells and thereby blocks IFNG signaling, allowing the bacteria to escape the host inflammatory response. PMID: 29273450
  41. Strategies to block MICA-NKG2D interactions resulted in reductions in IFNgamma production. Depletion of monocytes in vivo resulted in decreased IFNgamma production by murine NK cells upon exposure to Ab-coated tumor cells. PMID: 28724544
  42. Study concluded that the IFN-gamma (874A/T) polymorphism is associated with the susceptibility to oral lichen planus. PMID: 27544215
  43. IFN gamma induced upregulation of BCL6 was dependent on the classical STAT1 signaling pathway, and affected both major BCL6 variants. Interestingly, although IFN alpha induced stronger STAT1 phosphorylation than IFN gamma, it only slightly upregulated BCL6 in multiple myeloma lines. PMID: 29510136
  44. IFN-gamma, CXCL16 and uPAR are promising as effective biomarkers of disease activity, renal damage, and the activity of pathological lesions in systemic lupus erythematosus. PMID: 28628472
  45. Serum levels of soluble FAS ligand (sFASL) and interferon gamma (IFN-gamma) were analyzed and correlated with sFGL2 levels in Hepatitis C Virus-Infected Patients and Hepatocellular Carcinoma Patients. PMID: 28609212
  46. IFN-gamma induces activated but insufficient autophagy and thus contributes to a degree to p62-dependent apoptosis of nasal epithelial cells in chronic rhinosinusitis with nasal polyps. PMID: 28258963
  47. Results suggested that IFN-gamma induces autophagy-associated apoptosis in CRC cells via inducing cPLA2-dependent mitochondrial ROS production. PMID: 29551681
  48. Activated interferon-gamma-producing CD56(bright) NK cells are positioned to play a key role in the fibrotic process and progression to chronic kidney disease. PMID: 28396119
  49. Posttransplant immune monitoring by donor-specific IFN-gamma ELISPOT can assess risk for developing subclinical T-cell mediated rejection and anti-donor HLA antibodies. PMID: 28274484
  50. Genetic polymorphism is not associated with increased susceptibility to chronic spontaneous urticaria in Iran. PMID: 28159384

Show More

Hide All

Database Links

HGNC: 5438

OMIM: 147570

KEGG: hsa:3458

STRING: 9606.ENSP00000229135

UniGene: Hs.856

Involvement In Disease
Aplastic anemia (AA)
Protein Families
Type II (or gamma) interferon family
Subcellular Location
Secreted.
Tissue Specificity
Released primarily from activated T lymphocytes.

Customer Reviews

Overall Rating 5.0 Out Of 5
,
B.A
By Anonymous
★★★★★

Applications : Media additive

Review: LPS-Preconditioned PDLSCs Accentuate the M1 Polarization of IFN-γ Treated Macrophages.

Q&A

What is the molecular structure of recombinant human IFN-gamma?

Recombinant human IFN-gamma (rhIFN-γ) is a dimerized protein consisting of the amino acid sequence Gln24-Gln166 with an N-terminal Met, derived from expression in E. coli systems . Structurally, functional rhIFN-γ exists as a homodimer with a molecular weight of approximately 38 kDa, as determined by gel filtration chromatography against protein standards including ovalbumin (43 kDa) and chymotrypsinogen (25 kDa) . The dimerization of rhIFN-γ is essential for its biological activity, with properly assembled dimers representing the physiologically active form of the protein. X-ray crystallography and other structural studies have revealed that each monomer adopts an alpha-helical fold, with the two subunits associating in an antiparallel fashion to form the functional dimer.

How should recombinant IFN-gamma be reconstituted and stored for optimal stability?

For optimal stability and activity retention, lyophilized rhIFN-γ should be reconstituted at a concentration of 0.2 mg/mL using sterile, deionized water . The reconstitution process should be performed carefully to avoid excessive agitation that might lead to protein denaturation. Following reconstitution, rhIFN-γ preparations should be stored in a manual defrost freezer, with particular attention to avoiding repeated freeze-thaw cycles that can significantly reduce biological activity . For long-term storage, aliquoting the reconstituted protein into single-use volumes is recommended to minimize freeze-thaw cycles. Storage temperature should be maintained at -20°C or lower for extended periods, though working solutions may be kept at 4°C for up to one week. The addition of carrier proteins like BSA (bovine serum albumin) enhances protein stability, increases shelf-life, and allows the recombinant protein to be stored at more dilute concentrations, making carrier-containing formulations preferable for general cell culture applications or as ELISA standards .

What are the key differences between carrier-free and BSA-containing IFN-gamma preparations?

The primary difference between carrier-free (CF) and BSA-containing rhIFN-γ preparations lies in their formulation and intended applications. The BSA-containing version (e.g., 285-IF) is lyophilized from a 0.2 μm filtered solution containing sodium succinate, mannitol, Tween® 80, and BSA as a carrier protein . This formulation provides enhanced stability, increased shelf-life, and allows for storage at more dilute concentrations.

In contrast, the carrier-free version (e.g., 285-IF/CF) contains the same components but without BSA . This formulation is specifically designed for applications where the presence of BSA might interfere with experimental outcomes, such as:

  • Protein conjugation procedures requiring pure rhIFN-γ

  • Mass spectrometry analyses

  • Applications involving antibodies against BSA

  • Experiments requiring precise protein quantification without carrier interference

  • Cell culture systems sensitive to bovine proteins

How is the biological activity of recombinant IFN-gamma measured?

The biological activity of rhIFN-γ is primarily quantified through cytopathic antiviral assays, which measure the protein's ability to protect susceptible cells from viral infection . This functional assessment is expressed as International Units (IU) per milligram of protein, with high-quality preparations typically exhibiting specific activities ranging from 2 × 10^7 to 4 × 10^7 IU/mg protein .

Additional methodologies for assessing rhIFN-γ activity include:

  • Cell-based assays: Measuring IFN-γ-induced upregulation of MHC class II molecules (particularly HLA-DR) on monocytes or other responsive cell types .

  • Reporter gene assays: Utilizing cell lines transfected with IFN-γ-responsive promoter elements linked to easily detectable reporter genes.

  • Immunological assays: Evaluating IFN-γ-induced activation of immune cells, including macrophage activation, NK cell potentiation, or T-cell polarization.

  • Molecular assays: Quantifying the expression of IFN-γ-responsive genes through RT-PCR, microarray analysis, or RNA sequencing.

  • Functional assays: Assessing anti-parasitic activity against organisms like Toxoplasma gondii in neuronal or other cell models .

The selection of an appropriate assay depends on the specific research question, with antiviral protection assays remaining the gold standard for determining IFN-γ potency in International Units.

What are the optimal conditions for refolding recombinant IFN-gamma to maximize dimer formation and biological activity?

The optimization of refolding conditions for rhIFN-γ is critical for maximizing the yield of biologically active dimers. Research indicates that protein concentration during the refolding process significantly impacts dimerization efficiency, with optimal refolding occurring at protein concentrations between 50-100 μg/ml . The refolding process should be conducted in an appropriate buffer system, such as 100 mM Tris pH 7.2 containing 0.2 mM EDTA, with gentle stirring during the dilution of denatured protein .

The refolding protocol that yields greater than 90% dimer formation includes the following key steps:

  • Dilution of purified monomeric rhIFN-γ into refolding buffer at a carefully controlled concentration (optimally 50-100 μg/ml)

  • Slow addition of the denatured protein into the refolding buffer with constant gentle stirring

  • Extended incubation period of 24-36 hours at 4°C to allow complete refolding and dimer formation

  • Concentration of the refolded solution by ultrafiltration to approximately 2-5 mg/ml

  • Purification via size exclusion chromatography (e.g., Superdex-75) to isolate properly formed dimers

Advanced techniques to enhance refolding efficiency include the use of hydrophobic chromatographic column matrices as templates or stabilizing surfaces during the refolding process, which helps avoid inactive aggregate formation often observed in solution-phase refolding . Additionally, molecular chaperones that facilitate proper protein folding in vivo have been explored for in vitro refolding of rhIFN-γ, potentially offering further improvements to yield and activity .

What purification strategy yields the highest purity and specific activity for recombinant IFN-gamma?

A high-yield, high-purity purification strategy for rhIFN-γ involves a combination of reversed phase chromatography (RPC) followed by refolding and size exclusion chromatography. This approach yields greater than 99% purity with specific activities of 2-4 × 10^7 IU/mg protein, representing a substantial improvement over earlier methods .

The optimized purification protocol consists of the following steps:

  • Initial capture: Solubilization of rhIFN-γ from E. coli inclusion bodies

  • RPC purification: Using a rigid, monosized, polystyrene/divinyl benzene reversed phase chromatography column (e.g., Source-30™ matrix)

  • Refolding: Controlled dilution of purified monomers in refolding buffer (100 mM Tris pH 7.2, 0.2 mM EDTA) at optimal protein concentration (50-100 μg/ml)

  • Concentration: Ultrafiltration to increase protein concentration to 2-5 mg/ml

  • Size exclusion chromatography: Purification on Superdex-75 column equilibrated with PBS buffer to isolate properly formed dimers

This method yields approximately 40 mg of purified rhIFN-γ per gram of cell mass, representing a nearly 3-fold enhancement in yield compared to conventional approaches while maintaining high specific activity . The purified product demonstrates extremely low DNA and endotoxin content per mg of protein, well below the limits established for therapeutic applications .

For analytical confirmation of purity and proper dimer formation, gel filtration chromatography against molecular weight standards (ovalbumin at 43 kDa, chymotrypsinogen at 25 kDa, and ribonuclease at 13.7 kDa) should be performed, with properly formed rhIFN-γ dimers eluting between ovalbumin and chymotrypsinogen, confirming the expected molecular weight of approximately 38 kDa .

How can researchers design experiments to investigate interferon-gamma's immunomodulatory effects in complex disease models?

Designing experiments to investigate rhIFN-γ's immunomodulatory effects in complex disease models requires careful consideration of dosing, timing, and readout parameters. Based on clinical and preclinical studies, the following experimental design principles should be considered:

  • Dose optimization: Titrate rhIFN-γ doses based on the specific model system. For human studies, doses ranging from 50-75 mcg/m² have shown biological activity with acceptable safety profiles . For in vitro studies, dose-response assessments should be performed, typically starting in the range of 0.15-0.75 ng/mL, which represents the ED50 for many cellular responses .

  • Temporal considerations: Design experiments with appropriate time points to capture both immediate and delayed effects of IFN-γ treatment. Clinical studies have demonstrated significant changes in immune cell phenotypes from pre-induction to cycle 1 day 1 (C1D1) and cycle 2 day 15 (C2D15) .

  • Combination approaches: Consider combining rhIFN-γ with other immunomodulatory agents, such as checkpoint inhibitors (e.g., nivolumab), based on the understanding that IFN-γ induces PD-L1 expression, which may counter its pro-inflammatory effects .

  • Comprehensive immune monitoring:

    • Phenotypic analysis of monocyte subpopulations (classical, intermediate, non-classical)

    • Measurement of HLA-DR expression on monocytes as a marker of IFN-γ activity

    • Assessment of PD-L1 expression on intermediate and non-classical monocytes

    • Analysis of chemokine production and T-cell responses

  • Disease-specific readouts: Include model-specific readouts relevant to the disease being studied. For example:

    • In infectious disease models: pathogen clearance, immune cell recruitment

    • In cancer models: tumor growth, infiltrating immune cell characterization

    • In autoimmune models: tissue damage, inflammatory markers

  • Control conditions: Include appropriate controls:

    • Vehicle-only control

    • Carrier protein control (if using BSA-containing preparations)

    • Heat-inactivated rhIFN-γ control to distinguish between specific and non-specific effects

Researchers should be aware that rhIFN-γ treatment can lead to significant increases in HLA-DR expression on monocytes and elevated PD-L1 expression on intermediate and non-classical monocytes , which may have implications for interpretation of experimental outcomes in immune-mediated disease models.

What strategies can improve the stability and half-life of recombinant IFN-gamma for in vivo applications?

Several strategies can be employed to improve the stability and half-life of rhIFN-γ for in vivo applications, addressing the challenges of rapid clearance and potential immunogenicity:

  • Formulation optimization:

    • Inclusion of stabilizing excipients like mannitol and Tween® 80, which are present in standard lyophilized preparations

    • Addition of carrier proteins (BSA) for enhanced stability

    • Use of buffer systems optimized for maintaining native protein conformation (e.g., sodium succinate buffer systems)

  • Chemical modification:

    • PEGylation (attachment of polyethylene glycol) to increase molecular size and reduce renal clearance

    • Site-specific PEGylation to maintain biological activity while extending half-life

    • Glycosylation engineering to mimic natural post-translational modifications

  • Advanced delivery systems:

    • Encapsulation in biodegradable microparticles or nanoparticles for sustained release

    • Liposomal formulations to protect against degradation

    • Hydrogel-based depot formulations for extended local delivery

  • Protein engineering approaches:

    • Development of rhIFN-γ mutant analogues with enhanced stability

    • Fusion to stabilizing protein domains (e.g., Fc fragment of immunoglobulin)

    • Introduction of disulfide bonds to stabilize the dimer structure

  • Storage and handling considerations:

    • Use of manual defrost freezers for storage to avoid temperature fluctuations

    • Strict avoidance of repeated freeze-thaw cycles

    • Aliquoting of reconstituted protein into single-use volumes

For in vivo applications, researchers should conduct preliminary pharmacokinetic studies to determine the optimal dosing regimen based on the chosen stabilization strategy. The selected approach should balance the need for extended half-life with maintenance of biological activity, as modifications to improve stability may potentially impact receptor binding or downstream signaling cascades.

How can researchers troubleshoot low biological activity in recombinant IFN-gamma preparations?

Low biological activity in rhIFN-γ preparations can stem from various factors throughout the production, purification, storage, and application processes. Systematic troubleshooting should address each potential issue:

  • Production and purification issues:

    • Incomplete refolding leading to monomeric or misfolded proteins instead of functional dimers

    • Suboptimal protein concentration during refolding (should be 50-100 μg/ml)

    • Insufficient incubation time for complete refolding (24-36 hours at 4°C is recommended)

    • Inadequate purification of dimeric forms (verify using size exclusion chromatography)

  • Storage and handling problems:

    • Repeated freeze-thaw cycles causing protein denaturation

    • Improper reconstitution techniques (e.g., excessive agitation)

    • Storage in inappropriate buffers or at suboptimal protein concentrations

    • Use of incorrect storage containers (protein may adhere to certain plastics)

  • Assay-specific considerations:

    • Cell lines or systems used for activity assessment may have reduced sensitivity

    • Improper positive controls in biological assays

    • Interfering substances in the experimental system

    • Incorrect dosing for the specific assay system (ED50 typically 0.15-0.75 ng/mL)

  • Verification approaches:

    • SDS-PAGE analysis under non-reducing conditions to assess dimer formation

    • Western blot using specific anti-IFN-γ antibodies

    • Size exclusion chromatography to determine the proportion of dimeric protein

    • Analytical techniques to verify structure (circular dichroism, dynamic light scattering)

If low activity persists despite addressing these factors, researchers should consider using fresh starting material and reviewing each step of the production and purification process, with particular attention to the refolding conditions. For commercial preparations, comparison with reference standards using the same assay system can help determine if the issue lies with the protein preparation or the activity measurement methodology.

What are the critical quality attributes that should be analyzed for recombinant IFN-gamma in GMP production?

For GMP (Good Manufacturing Practice) production of rhIFN-γ, several critical quality attributes (CQAs) must be rigorously analyzed to ensure consistency, safety, and efficacy:

  • Structural and physical attributes:

    • Protein concentration (typically determined by absorbance at 280 nm or BCA assay)

    • Dimer percentage (>90% dimeric form is desirable)

    • Molecular weight verification by mass spectrometry

    • Primary sequence confirmation via peptide mapping

    • Higher-order structure analysis by circular dichroism or Fourier-transform infrared spectroscopy

    • Thermal stability assessment

  • Purity and impurity profile:

    • Protein purity (≥99% by reversed-phase HPLC and size exclusion chromatography)

    • Host cell protein content (typically <100 ng per mg of product)

    • Residual DNA content (<10 ng per dose)

    • Endotoxin levels (<5 EU per kg body weight per hour)

    • Aggregates and fragments quantification

    • Process-related impurities (chromatography leachables, buffer components)

  • Biological activity assessment:

    • Specific activity (2-4 × 10^7 IU/mg protein by antiviral assay)

    • Cell-based bioassays measuring appropriate cellular responses

    • Receptor binding assays

    • Potency relative to international reference standard

  • Stability indicators:

    • Shelf-life determination under recommended storage conditions

    • Forced degradation studies to identify critical degradation pathways

    • Stability in formulation buffer after reconstitution

    • Photostability assessment

  • Formulation attributes:

    • pH and osmolality

    • Appearance (clear, colorless solution after reconstitution)

    • Particulate matter

    • Container closure integrity

For GMP production, each of these attributes must be tested using validated analytical methods with appropriate acceptance criteria. The manufacturing process should be designed to consistently produce rhIFN-γ meeting these specifications, with appropriate in-process controls to monitor critical parameters throughout production.

How does recombinant IFN-gamma compare to natural IFN-gamma in terms of post-translational modifications and biological functions?

Recombinant human IFN-gamma (rhIFN-γ) produced in E. coli differs from natural human IFN-gamma in several important aspects, primarily related to post-translational modifications and structural features:

  • Post-translational modifications:

    • Natural IFN-γ is glycosylated, while E. coli-derived rhIFN-γ lacks glycosylation

    • Natural IFN-γ may contain other modifications such as phosphorylation and acetylation

    • E. coli-derived rhIFN-γ typically contains an N-terminal methionine not present in the mature natural form

  • Structural differences:

    • The rhIFN-γ sequence typically encompasses amino acids Gln24-Gln166 of the natural protein, with an additional N-terminal methionine

    • Both natural and recombinant IFN-γ form homodimers, but subtle conformational differences may exist due to the absence of glycosylation in the recombinant form

  • Biological function comparison:

PropertyNatural IFN-γE. coli-derived rhIFN-γ
Receptor bindingHigh affinityComparable affinity
Specific activityVariable, typically 10^7-10^8 IU/mg2-4 × 10^7 IU/mg
Half-life in vivoLonger due to glycosylationShorter due to lack of glycosylation
ImmunogenicityLowPotentially higher due to lack of glycosylation
StabilityHigher stabilityMay have reduced stability
Cellular effectsFull spectrum of activitiesComparable spectrum of activities
  • Functional equivalence:
    Despite these differences, E. coli-derived rhIFN-γ retains the core biological activities of natural IFN-γ, including:

    • Macrophage activation

    • MHC class II upregulation (particularly HLA-DR on monocytes)

    • Antiviral protection

    • Th1 immune response promotion

    • Anti-parasitic defense stimulation

When designing experiments, researchers should consider these differences, particularly when translating in vitro findings to in vivo systems where pharmacokinetics may be affected by the absence of glycosylation in E. coli-derived rhIFN-γ. For applications requiring closer mimicry of natural IFN-γ, mammalian cell-derived recombinant forms (e.g., from CHO cells) that include appropriate glycosylation may be preferable, despite their typically higher cost and lower yield.

What are the current approaches for combining IFN-gamma with immune checkpoint inhibitors in cancer immunotherapy research?

Current approaches for combining IFN-gamma with immune checkpoint inhibitors in cancer immunotherapy research focus on exploiting synergistic effects while mitigating potential antagonistic interactions:

For researchers designing preclinical or clinical studies of this combination, careful monitoring of immunological parameters is essential, including assessment of monocyte subpopulations, PD-L1 expression dynamics, and tumor microenvironment changes. The unexpected finding of reduced irAE incidence with this combination warrants further mechanistic investigation and may represent an important advantage over other immunotherapy combinations.

How can researchers leverage IFN-gamma-induced gene signatures for biomarker development in immunotherapy research?

Researchers can leverage IFN-γ-induced gene signatures for biomarker development in immunotherapy research through several strategic approaches:

  • Characterization of IFN-γ response signatures:

    • Identify core gene sets consistently upregulated by IFN-γ across different cell types and tissues

    • Distinguish between early response genes (directly induced by STAT1 activation) and secondary response genes

    • Map the temporal dynamics of IFN-γ-induced transcriptional changes

  • Biomarker identification strategies:

    • Single-cell RNA sequencing to resolve cellular heterogeneity in IFN-γ responses

    • Proteomics to identify secreted factors that could serve as blood-based biomarkers

    • Epigenetic profiling to detect stable chromatin modifications following IFN-γ exposure

    • Integration of multi-omics data to develop robust signature panels

  • Clinical application approaches:

    • Development of tissue-based assays measuring IFN-γ-responsive gene expression in tumor biopsies

    • Blood-based assays detecting circulating proteins induced by IFN-γ signaling

    • Identification of minimal gene sets with maximal predictive power for clinical outcomes

  • Biomarker validation methodology:

    • Initial demonstration of IFN-γ-responsiveness in controlled in vitro systems

    • Confirmation in relevant animal models

    • Retrospective analysis in existing clinical specimen collections

    • Prospective validation in clinical trials

  • Specific biomarker categories:

    • Predictive biomarkers: Identifying patients likely to respond to IFN-γ-based therapies

    • Pharmacodynamic biomarkers: Confirming biological activity of administered IFN-γ

    • Response biomarkers: Early indicators of therapeutic efficacy

    • Resistance biomarkers: Signatures indicating development of resistance to IFN-γ effects

  • Technological platforms:

    • Digital PCR for precise quantification of selected gene panels

    • NanoString technology for targeted gene expression profiling

    • Mass cytometry for high-dimensional protein-level analysis

    • Spatial transcriptomics to map IFN-γ responses within tissue architecture

When designing biomarker studies, researchers should consider that IFN-γ induces significant changes in HLA-DR expression on monocytes and PD-L1 expression on intermediate and non-classical monocytes . These easily accessible circulating immune cells provide convenient pharmacodynamic biomarkers for confirming IFN-γ activity in vivo. Additionally, the interaction between IFN-γ and PD-L1 expression highlights the importance of developing integrated biomarker panels that capture both the pro-inflammatory effects of IFN-γ and the counter-regulatory mechanisms it induces.

What are the methodological considerations for investigating IFN-gamma's role in neuroinflammatory and neurodegenerative disease models?

Investigating IFN-gamma's role in neuroinflammatory and neurodegenerative disease models requires specialized methodological approaches that address the unique challenges of neuroimmunology research:

  • Model selection considerations:

    • In vitro neuronal cultures (primary or cell lines) for direct IFN-γ effects

    • Mixed glial-neuronal co-cultures to examine cell-cell interactions

    • Brain organoids for three-dimensional tissue-like responses

    • Animal models with varying degrees of blood-brain barrier integrity

    • Human samples (CSF, brain tissue) for translational validation

  • Delivery and dosing strategies:

    • Direct application to cultured cells (typically 0.15-0.75 ng/mL for ED50)

    • Intracerebroventricular injection in animal models

    • Osmotic pump delivery for sustained central exposure

    • Engineered delivery systems for crossing the blood-brain barrier

    • Gene therapy approaches for localized IFN-γ production

  • Neural-specific readouts:

    • Electrophysiological measurements of neuronal activity

    • Neurite outgrowth and synaptic density quantification

    • Neurotransmitter release and receptor expression

    • Axonal transport dynamics

    • Neuronal-glial communication assessment

  • Glial-specific assessments:

    • Microglial polarization states (M1/M2 paradigm)

    • Astrocyte reactivity and A1/A2 phenotyping

    • Oligodendrocyte maturation and myelination capacity

    • Blood-brain barrier integrity evaluation

  • Application-specific experimental designs:

    For neurodegenerative models:

    • Timing of IFN-γ intervention relative to disease onset

    • Assessment of protein aggregation (Aβ, tau, α-synuclein)

    • Neuronal survival quantification

    • Behavioral testing for functional outcomes

    For neuroinflammatory models:

    • IFN-γ effects on immune cell infiltration

    • Blood-brain barrier permeability changes

    • Cytokine/chemokine cascades

    • Demyelination and remyelination dynamics

  • Specialized techniques:

    • Two-photon imaging for in vivo neural circuit visualization

    • Single-cell RNA sequencing of neural populations

    • Spatial transcriptomics for region-specific responses

    • CLARITY or iDISCO tissue clearing for whole-brain analysis

Recent research has demonstrated that IFN-γ stimulated murine and human neurons mount anti-parasitic defenses against intracellular parasites like Toxoplasma gondii , highlighting the direct responsiveness of neurons to this cytokine. This finding emphasizes the importance of examining neuron-intrinsic responses to IFN-γ rather than focusing exclusively on glial-mediated effects. Researchers should design experiments that can distinguish between direct IFN-γ signaling in neurons versus indirect effects mediated through glial activation or peripheral immune cell recruitment.

What analytical techniques are most appropriate for assessing the quality and consistency of GMP-grade recombinant IFN-gamma across production batches?

Ensuring batch-to-batch consistency of GMP-grade recombinant IFN-gamma requires comprehensive analytical characterization using complementary techniques:

  • Physicochemical characterization:

    • Size exclusion chromatography (SEC): Quantifies monomer/dimer ratios and aggregation states

    • Reversed-phase HPLC (RP-HPLC): Assesses hydrophobic variants and chemical modifications

    • Capillary electrophoresis (CE): Evaluates charge variants and isoelectric properties

    • Mass spectrometry (MS): Precise molecular weight determination and detection of modifications

    • Peptide mapping: Confirmation of primary sequence and identification of modified residues

  • Structural analysis:

    • Circular dichroism (CD): Assessment of secondary structure elements

    • Fourier-transform infrared spectroscopy (FTIR): Complementary secondary structure analysis

    • Differential scanning calorimetry (DSC): Thermal stability evaluation

    • Hydrogen-deuterium exchange mass spectrometry (HDX-MS): Higher-order structure analysis

    • Small-angle X-ray scattering (SAXS): Solution-state structural characterization

  • Functional testing:

    • Cytopathic antiviral assays: Gold standard for specific activity determination (2-4 × 10^7 IU/mg)

    • Cell-based reporter assays: Measurement of IFN-γ-responsive promoter activation

    • Flow cytometry: Assessment of HLA-DR upregulation on monocytes

    • Surface plasmon resonance (SPR): Receptor binding kinetics and affinity determination

    • Cell proliferation/viability assays: Measurement of IFN-γ-mediated growth inhibition

  • Impurity profiling:

    • Host cell protein ELISA: Quantification of process-related protein impurities

    • qPCR: Residual DNA quantification

    • Limulus amebocyte lysate (LAL) test: Endotoxin determination

    • Colorimetric assays: Detection of process-related chemical impurities

    • Specific immunoassays: Detection of potential contaminating cytokines or growth factors

  • Stability-indicating methods:

    • Accelerated and real-time stability studies: Evaluation under various storage conditions

    • Forced degradation studies: Identification of degradation pathways and products

    • In-use stability testing: Assessment of stability after reconstitution

  • Comparative analytical strategy:

    • Reference standard comparison for each analytical method

    • Statistical process control with appropriate control charts

    • Trend analysis across multiple batches

    • Establishment of acceptable quality limits based on process capability

For GMP production, analytical methods must be validated according to ICH guidelines, demonstrating specificity, accuracy, precision, linearity, range, and robustness. Implementation of orthogonal methods addressing similar quality attributes provides greater confidence in results. Advanced analytical approaches such as multi-attribute monitoring (MAM) using LC-MS can simultaneously assess multiple quality attributes, enabling more comprehensive batch comparisons.

A systematic analytical comparison of batches should include radar plots or similarity scores integrating multiple quality attributes, with appropriate statistical methods to determine significant deviations that might impact clinical performance.

How might emerging technologies enhance the production and application of recombinant IFN-gamma for research and therapeutic purposes?

Emerging technologies are poised to transform both the production and application of recombinant IFN-gamma, offering opportunities for improved yield, quality, and therapeutic efficacy:

  • Advanced production platforms:

    • Cell-free protein synthesis: Rapid production without cell culture, enabling rapid iteration

    • Continuous manufacturing: Integrated processes replacing batch production for consistent quality

    • Automated micro-bioreactors: High-throughput optimization of expression conditions

    • Synthetic biology approaches: Engineered chassis organisms with enhanced secretion capabilities

    • Alternative expression systems: Insect cells, plant-based systems, or novel microbial hosts

  • Protein engineering innovations:

    • Computational design: In silico prediction of stabilizing mutations

    • Directed evolution: High-throughput screening for enhanced properties

    • Non-natural amino acid incorporation: Site-specific modification for enhanced half-life

    • Domain fusion strategies: Creation of bifunctional molecules with targeted activity

    • Structure-guided engineering: Rational design based on receptor-ligand interfaces

  • Delivery technologies:

    • Stimuli-responsive nanoparticles: Environmentally triggered release at target sites

    • Exosome-based delivery: Natural vesicles for enhanced cellular uptake

    • Tissue-specific targeting: Antibody-cytokine fusion proteins

    • Self-assembling peptide depots: Long-term local release systems

    • mRNA delivery systems: In situ production of IFN-γ using nucleic acid therapeutics

  • Advanced analytical methods:

    • Single-molecule characterization: Direct visualization of protein structure and dynamics

    • Multi-attribute monitoring (MAM): Simultaneous assessment of multiple quality attributes

    • Artificial intelligence: Predictive models for critical quality attributes

    • Automated high-throughput analytics: Real-time process monitoring and adjustment

    • Digital twins: Computational models of production processes for optimization

  • Therapeutic application innovations:

    • Precision dosing algorithms: Individualized dosing based on biomarker response

    • Combination therapy optimization: Synergistic regimens with checkpoint inhibitors

    • Cell-based delivery systems: Engineered cells producing IFN-γ in response to specific stimuli

    • Organ-on-chip models: Improved preclinical evaluation of efficacy and toxicity

    • Digital biomarkers: Remote monitoring of treatment responses

These emerging technologies promise to address current limitations in rhIFN-γ production and application, potentially leading to higher yields exceeding the current benchmark of 40 mg g⁻¹ cell mass , enhanced stability beyond current formulations , and more targeted therapeutic approaches with improved efficacy and reduced side effects. Integration of computational approaches with experimental technologies is particularly promising for accelerating optimization across the development pipeline.

What are the current challenges and potential solutions in scaling up GMP production of recombinant IFN-gamma for clinical applications?

Scaling up GMP production of recombinant IFN-gamma for clinical applications presents several challenges that require innovative solutions:

  • Upstream processing challenges:

    Challenges:

    • Inclusion body formation in E. coli expression systems

    • Balancing protein expression with cell growth

    • Maintaining genetic stability of production strains

    • Process variability at increased scales

    Solutions:

    • Optimized fermentation parameters with real-time monitoring

    • Genetically engineered host strains with enhanced secretion

    • Controlled induction strategies to maximize yield

    • Scale-down models for accurate process development

  • Downstream processing challenges:

    Challenges:

    • Efficient solubilization of inclusion bodies

    • Maximizing refolding efficiency at large scale

    • Chromatographic separation scalability

    • Maintaining consistent dimer percentage

    Solutions:

    • Optimized solubilization buffers with chaotropic agents

    • Controlled dilution systems for refolding at 50-100 μg/ml concentration range

    • Continuous chromatography processes

    • In-line monitoring of dimer formation

  • Analytical challenges:

    Challenges:

    • Development of high-throughput potency assays

    • Real-time monitoring of critical quality attributes

    • Establishing clinically relevant specifications

    • Method transfer and validation across manufacturing sites

    Solutions:

    • Reporter-cell based potency assays replacing viral cytopathic assays

    • Process analytical technology (PAT) implementation

    • Quality by design (QbD) approach to specification setting

    • Robust method validation with appropriate system suitability criteria

  • Regulatory challenges:

    Challenges:

    • Evolving regulatory requirements for biologics

    • Comparability assessments after process changes

    • International harmonization of standards

    • Post-approval manufacturing changes

    Solutions:

    • Early and frequent regulatory interactions

    • Comprehensive comparability protocols

    • Implementation of international standards (ICH, WHO)

    • Risk-based approaches to post-approval changes

  • Economic challenges:

    Challenges:

    • High production costs affecting affordability

    • Batch failures at commercial scale

    • Cold chain requirements

    • Competition from biosimilars

    Solutions:

    • Process intensification to increase yield to >40 mg g⁻¹ cell mass

    • Predictive maintenance and quality monitoring

    • Development of stable formulations

    • Continuous improvement programs

The current benchmark for rhIFN-γ yield using optimized reversed phase chromatography and refolding procedures is approximately 40 mg g⁻¹ cell mass with >90% dimer formation . While this represents a significant improvement over earlier methods, further optimization of each production stage through implementation of advanced technologies could potentially increase yields while maintaining the required specific activity of 2-4 × 10^7 IU/mg protein .

How can computational approaches advance our understanding of IFN-gamma signaling networks and improve therapeutic targeting?

Computational approaches offer powerful tools for advancing our understanding of IFN-gamma signaling networks and improving therapeutic targeting through multiple complementary strategies:

  • Network-level modeling approaches:

    • Ordinary differential equation (ODE) models: Quantitative description of signaling dynamics

    • Boolean network models: Qualitative representation of pathway activation states

    • Bayesian networks: Probabilistic modeling of signaling relationships

    • Agent-based models: Simulation of cell-cell interactions mediated by IFN-γ

    • Multi-scale models: Integration of molecular, cellular, and tissue-level effects

  • Data-driven computational methods:

    • Machine learning for biomarker discovery: Identification of predictive signatures

    • Network inference from omics data: Reconstruction of IFN-γ responsive networks

    • Single-cell trajectory analysis: Mapping cellular state transitions after IFN-γ exposure

    • Natural language processing: Automated extraction of IFN-γ knowledge from literature

    • Deep learning for image analysis: Quantification of IFN-γ-induced phenotypic changes

  • Structural biology computations:

    • Molecular dynamics simulations: Investigation of IFN-γ-receptor interactions

    • In silico mutagenesis: Prediction of functional consequences of IFN-γ variants

    • Protein-protein docking: Modeling of IFN-γ interactions with novel binding partners

    • Pharmacophore modeling: Design of small molecules targeting IFN-γ signaling

    • Quantum mechanics calculations: Analysis of critical binding interactions

  • Therapeutic targeting applications:

    • Systems pharmacology models: Prediction of drug combinations with IFN-γ

    • Virtual patient cohorts: In silico clinical trials for IFN-γ-based therapies

    • Network controllability analysis: Identification of optimal intervention points

    • Cell-specific response prediction: Personalized dosing strategies

    • Toxicity prediction models: Anticipation of potential adverse effects

  • Integration with experimental approaches:

    • Active learning frameworks: Computational guidance of experimental design

    • Digital twin development: Computational models calibrated to specific systems

    • Hybrid modeling approaches: Combining mechanistic and data-driven methods

    • Model validation workflows: Systematic testing of computational predictions

    • Iterative refinement cycles: Continuous improvement through experiment-computation loops

Computational approaches are particularly valuable for understanding the complex effects of IFN-γ on immune checkpoint expression, such as the observed increase in PD-L1 expression on intermediate monocytes following IFN-γ administration . By modeling these feedback mechanisms, researchers can better predict the outcomes of combination therapies involving IFN-γ and checkpoint inhibitors, potentially explaining observations such as the reduced incidence of immune-related adverse events in combination therapy .

For therapeutic applications, computational methods can help define optimal dosing regimens to balance pro-inflammatory effects with counter-regulatory mechanisms, identify patient populations most likely to benefit from IFN-γ-based therapies, and design novel protein variants or delivery systems with improved pharmacological properties.

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.