Fact-checked by Grok 2 weeks ago

Interferon gamma

Interferon gamma (IFN-γ), the sole member of the type II interferon family, is a pleiotropic that serves as a key mediator in both innate and adaptive immune responses. Primarily secreted by activated CD4+ and CD8+ T lymphocytes, natural killer (NK) cells, and certain , IFN-γ exhibits potent antiviral, antibacterial, antiproliferative, and antitumor activities while also regulating immune cell differentiation and function. Its production is tightly controlled and induced by stimuli such as microbial antigens, interleukin-12 (IL-12), and IL-18, ensuring a rapid response to pathogens and malignancies. Structurally, IFN-γ exists as an antiparallel homodimeric , with each comprising 143 and a molecular weight of approximately 17 kDa, encoded by the IFNG gene located on 12q24.1 in humans. Upon secretion, IFN-γ binds to a cell-surface receptor complex formed by two subunits—IFNGR1 (the ligand-binding chain) and IFNGR2 (the signal-transducing chain)—which activates the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway, along with MAPK and PI3K signaling cascades, to induce gene expression changes that enhance antimicrobial defenses and modulate inflammation. This receptor-mediated action allows IFN-γ to upregulate ( and II molecules, promote macrophage activation, and stimulate NK cell cytotoxicity, thereby bridging humoral and . First identified in 1965 as an antiviral factor in phytohemagglutinin-stimulated leukocyte cultures, IFN-γ was distinguished from type I interferons by its distinct production sources and receptor specificity. In addition to its protective roles in host defense against viruses, intracellular bacteria like Mycobacterium tuberculosis, and tumors, IFN-γ can contribute to immunopathology in chronic conditions such as autoimmune diseases and graft-versus-host disease by exacerbating Th1-biased responses. Recombinant human IFN-γ1b, approved by the FDA in 1990 for chronic granulomatous disease and in 2000 for severe malignant osteopetrosis, is clinically used to bolster phagocyte function and reduce severe infection rates in these conditions, with ongoing investigations into its applications for cystic fibrosis and various cancers either alone or in combination therapies.

Molecular Structure

Gene Organization

The human IFNG gene, which encodes interferon gamma (IFN-γ), is located on the long arm of chromosome 12 at the q15 cytogenetic band. It spans approximately 5 kilobases (kb) of genomic DNA and consists of four exons separated by three introns, with the exons encoding a precursor protein of 166 amino acids. The promoter region of IFNG contains multiple binding sites for key transcription factors, including NF-κB family members such as p50 and p65, which bind to tandem κB sites upstream of the transcription start site to regulate gene expression. Additionally, the promoter and associated cis-regulatory elements interact with STAT4, often in concert with T-bet, to facilitate IFN-γ transcription during T cell differentiation. The IFNG gene exhibits strong evolutionary across mammals, reflecting its essential role in immune responses, with the four-exon/three-intron preserved from cartilaginous to higher vertebrates. As the sole member of the type II interferon family, IFNG shares structural and functional with other type II IFNs in distant species, though its sequence diverges more significantly outside mammals compared to type I IFNs. Alternative splicing variants of IFNG are rare, with only one principal transcript (ENST00000229135) identified in annotations, encoding the canonical IFN-γ precursor that matures into the bioactive dimer. This single predominant isoform underscores the tightly regulated, non-variable expression of IFN-γ at the transcriptional level.

Protein Conformation

Interferon gamma (IFN-γ) exists as a non-covalently associated homodimer, with each mature comprising 143 following cleavage of the . The three-dimensional of recombinant human IFN-γ was first resolved by at 3.5 Å resolution (PDB entry 1HIG), demonstrating a compact, primarily α-helical architecture that constitutes approximately 62% of the polypeptide chain. Each folds into six α-helices (labeled A through F), connected by short loops, forming an elongated antiparallel bundle that interlocks across the dimer interface through extensive hydrophobic and electrostatic interactions. This interlocking arrangement positions helices A and F on the exterior, contributing to the overall stability without the presence of intramolecular bonds, which distinguishes IFN-γ from type I interferons. The absence of β-sheets further emphasizes the helical dominance in its secondary . The protein undergoes N-linked at Asn25 and, to a lesser extent, Asn97, leading to variable occupancy that influences resistance and solubility. Consequently, the dimeric form displays an apparent molecular weight ranging from 34 for the unglycosylated recombinant protein to approximately 40-50 in natural glycosylated variants, as observed under non-denaturing conditions. The resolved delineates key receptor-binding interfaces primarily along the solvent-exposed faces of helices B, C, and F, as well as adjacent loops, underscoring the structural basis for its specificity.

Biosynthesis and Sources

Cellular Producers

Interferon gamma (IFN-γ) is primarily synthesized and secreted by immune cells central to the innate and adaptive responses, including natural killer (NK) cells, CD4+ T helper 1 (Th1) cells, CD8+ cytotoxic T cells, and type 1 innate lymphoid cells (ILC1s). NK cells and ILC1s contribute to early innate production, while Th1 and CD8+ T cells drive sustained secretion during adaptive immunity. These cells are activated in response to microbial infections, where IFN-γ production peaks 24-48 hours post-stimulation, supporting rapid antiviral and antibacterial defenses. In vitro studies of activated T cells demonstrate secretion levels reaching up to 10 ng/mL, highlighting their potent output under controlled conditions. Although immune cells dominate IFN-γ production, non-immune sources such as epithelial cells can contribute minor amounts under stress, such as during bacterial infections like . This epithelial-derived IFN-γ may locally amplify immune responses but remains secondary to leukocyte contributions. Production from these primary cellular sources is often induced by cytokines like IL-12, which promotes Th1 differentiation and NK cell activation.

Induction Triggers

Interferon gamma (IFN-γ) gene expression is primarily induced by specific cytokines and cellular signaling events in immune cells. Key inducers include interleukin-12 (IL-12) and interleukin-18 (IL-18), which synergistically promote IFN-γ production in T cells, natural killer (NK) cells, and other innate-like lymphocytes by activating the T-bet (encoded by ). IL-12 binds to its receptor on target cells, triggering STAT4 and subsequent T-bet upregulation, while IL-18 enhances this process through MyD88-dependent signaling, amplifying IFN-γ transcription. This cytokine-driven induction establishes a positive feedback loop, as IFN-γ itself can further sensitize cells to IL-12. In adaptive immune responses, (TCR) engagement serves as a critical trigger for IFN-γ production, particularly in CD4+ and CD8+ T cells during antigen-specific activation. TCR stimulation, often in combination with costimulatory signals, leads to NFAT and activation, which cooperate with T-bet to drive Ifng gene expression. Low-affinity TCR ligands preferentially elicit IFN-γ secretion without full T cell differentiation, highlighting a threshold-based response to encounter. Microbial stimuli indirectly induce IFN-γ through activation of receptors on antigen-presenting cells. For instance, bacterial (LPS) engages (TLR4), activating the pathway to promote IL-12 secretion from dendritic cells and macrophages, which then stimulates IFN-γ in downstream effector cells. Similarly, viral double-stranded RNA (dsRNA) sensed by TLR3 or RIG-I-like receptors triggers translocation and IL-12/IL-18 production, fostering IFN-γ responses. These pathways ensure rapid IFN-γ upregulation during infections. The dose-response relationship for induction is concentration-dependent, with threshold levels of IL-12 around 1-10 ng/mL sufficient to achieve maximal IFN-γ secretion in human and murine lymphocytes under optimal conditions. Insights into the pathway's role in cytosolic DNA sensing during bacterial and viral infections highlight how STING activation in innate cells promotes type I interferon production that enhances cell-derived IFN-γ, bridging cytosolic surveillance to adaptive immunity.

Receptor Engagement

Receptor Composition

The interferon gamma receptor (IFNGR) is a heterodimeric consisting of two transmembrane polypeptide chains: IFNGR1 (also known as CD119), the ligand-binding α-chain with an observed molecular weight of approximately 90 kDa due to , and IFNGR2, the signal-transducing β-chain with an observed molecular weight of approximately 55 kDa. IFNGR1 primarily mediates the initial high-affinity interaction with the dimeric IFN-γ , while IFNGR2 stabilizes the and facilitates signal without direct . Both chains belong to the class II family, characterized by extracellular regions each comprising two tandem type III-like domains that adopt a characteristic "V-shaped" architecture for recognition. These domains lack the typical immunoglobulin folds found in other receptor families but share structural with cytokine-binding modules essential for specificity. IFNGR1 exhibits ubiquitous surface expression at moderate levels on virtually all nucleated cells, excluding mature erythrocytes, which ensures widespread cellular responsiveness to IFN-γ. In contrast, IFNGR2 displays more restricted basal expression, predominantly on hematopoietic cells such as monocytes, lymphocytes, and macrophages, as well as endothelial cells, though its levels can be upregulated in response to inflammatory cues in other cell types. Upon binding of the IFN-γ homodimer, the receptor assembles into a functional (IFNGR1)2-(IFNGR2)2 tetramer, forming a symmetric hexameric complex that positions the intracellular domains for downstream activation. This is critical for cooperative signaling and has been elucidated through crystallographic studies of the extracellular assembly.

Binding Dynamics

Interferon gamma (IFN-γ) initiates receptor engagement through sequential , where the IFN-γ homodimer first interacts with high to two IFNGR1 chains, exhibiting a (Kd) of approximately 10^{-9} M. This primary contact stabilizes the complex, subsequently recruiting two IFNGR2 chains to complete the assembly. The architecture adheres to a dimer-of-dimers model, wherein the IFN-γ homodimer serves as a bridge between two receptor heterodimers, each comprising one IFNGR1 and one IFNGR2 chain, forming a symmetric 2:2:2 hexameric complex essential for activation. Key residues on IFN-γ facilitate electrostatic interactions with complementary sites on the receptor chains, enhancing specificity and stability. Binding efficiency is modulated by environmental factors such as and post-translational modifications like ; N-linked on IFNGR1 and IFNGR2 contributes to structural integrity and is necessary for effective recognition, while variations can influence the electrostatic components of the . High-resolution structural analyses, including the 2019 cryo-EM structure of the full receptor complex, have guided the rational design of IFN-γ agonists by elucidating key interfacial contacts for targeted modulation.

Signal Transduction Pathways

Canonical JAK-STAT Cascade

Upon ligand-induced dimerization of the interferon gamma receptor (IFNGR), which consists of IFNGR1 and IFNGR2 subunits, the constitutively associated kinases JAK1 (bound to IFNGR1) and JAK2 (bound to IFNGR2) undergo rapid autophosphorylation, initiating the canonical signaling cascade. This activation enables the JAKs to cross-phosphorylate residues on the intracellular domains of the receptor chains, creating sites for the of latent cytoplasmic proteins. The recruited molecules are then phosphorylated by JAK1 and JAK2 at the conserved tyrosine residue Tyr701, promoting their homodimerization to form the complex. The homodimer subsequently translocates to the , where it binds specifically to gamma-activated (GAS) elements in the promoter regions of target genes, with the TTCN_{2-4}GAA. This binding facilitates the transcriptional activation of IFN-γ-responsive genes, including those encoding (MHC) class I and II molecules as well as interferon regulatory factor 1 (IRF-1). STAT1 activation in this pathway typically peaks within 15-30 minutes following IFN-γ stimulation, reflecting the rapid and transient nature of the JAK-STAT response before and feedback regulation ensue.

Non-Canonical Pathways

Interferon gamma (IFN-γ) engages non-canonical signaling pathways that extend beyond the primary JAK-STAT axis, influencing diverse cellular processes such as survival, proliferation, and inflammation through indirect activation of secondary cascades. One prominent pathway involves the (PI3K)-Akt axis, where IFN-γ binding to its receptor triggers PI3K recruitment and activation, leading to of Akt at key residues like Thr308 and Ser473. This activation promotes cell survival by inhibiting pro-apoptotic factors such as Bad and FoxO3a, while also modulating proliferation in immune cells and tumor contexts, often in synergy with other cytokines like TNF-α to enhance glycolytic metabolism and anti-inflammatory properties in mesenchymal stem cells. In parallel, IFN-γ stimulates mitogen-activated protein kinase (MAPK) pathways, particularly the extracellular signal-regulated kinase (ERK) and p38 branches, which converge on the activator protein-1 (AP-1) transcription factor complex. ERK activation occurs via upstream Ras-Raf signaling, phosphorylating AP-1 components like c-Jun and c-Fos to drive expression of genes involved in cytokine production and cellular differentiation, as seen in Th1 effector T cells where p38 inhibition reduces IFN-γ secretion itself. Similarly, p38 MAPK is rapidly phosphorylated in response to IFN-γ, enhancing AP-1 activity to regulate inflammatory responses, such as inducible nitric oxide synthase (iNOS) expression in macrophages and β-cells, thereby linking IFN-γ to antimicrobial and antiproliferative effects without direct STAT1 involvement. IFN-γ also exhibits crosstalk with the nuclear factor kappa B (NF-κB) pathway, potentiating inflammatory gene expression through synergistic priming mechanisms. Upon IFN-γ stimulation, STAT1 indirectly enhances NF-κB nuclear translocation and DNA binding by modifying chromatin accessibility at NF-κB target promoters, such as those for iNOS and cytokines like IL-6, particularly in macrophages and epithelial cells during inflammation. This interaction amplifies Toll-like receptor (TLR)-induced responses, where IFN-γ pre-treatment boosts NF-κB-dependent transcription via epigenetic changes, including histone acetylation, ensuring robust inflammatory output in contexts like infection and autoimmunity. A notable recent development highlights IFN-γ's role in inducing calcium flux and CrkL to mediate cytoskeletal rearrangements in immune cells. IFN-γ rapidly elevates intracellular calcium levels in and T lymphocytes via receptor-mediated channels, activating downstream effectors like to support cellular activation and migration. Concurrently, CrkL, an SH2/SH3 adaptor, undergoes in a JAK1/2-dependent manner, associating with C3G to activate Rap1 , which reorganizes the for enhanced phagocytic and migratory functions in macrophages and dendritic cells.

Core Functions

Immunomodulatory Effects

Interferon gamma (IFN-γ) plays a central role in modulating adaptive immune responses by promoting the differentiation of CD4+ T helper cells toward the Th1 phenotype. This process involves the upregulation of T-bet, a that drives Th1 commitment by enhancing expression of the IL-12 receptor and amplifying IFN-γ production in a loop. Synergy with IL-12 further stabilizes Th1 differentiation, as IFN-γ potentiates IL-12 signaling to reinforce cellular immunity over humoral responses. These effects are mediated primarily through the canonical JAK-STAT pathway, where IFN-γ activates to initiate T-bet expression. In innate immunity, IFN-γ enhances activation, polarizing them toward the classical phenotype characterized by increased and production of (ROS). This activation induces expression of inducible (iNOS) and proinflammatory cytokines such as TNF-α and IL-1β, enabling macrophages to efficiently clear pathogens and present antigens. By promoting these antimicrobial functions, IFN-γ coordinates innate and adaptive arms of the for robust cellular responses. IFN-γ also upregulates (MHC) class I and II molecules on antigen-presenting cells (APCs), including dendritic cells and macrophages, thereby enhancing to T cells. This involves STAT1-dependent activation of regulatory factor 1 (IRF-1), which boosts expression of MHC genes, transporter associated with antigen processing (TAP-1/2), and immunoproteasome components like LMP2 and LMP7. Improved MHC expression ensures more effective priming of CD8+ cytotoxic T cells and CD4+ helper T cells, strengthening overall immune surveillance. To maintain Th1 dominance, IFN-γ inhibits Th2 production, particularly IL-4, which suppresses Th2 and while skewing immunity toward cell-mediated responses. This prevents the development of humoral-biased immunity and reinforces against intracellular threats. Through these mechanisms, IFN-γ fine-tunes immune to favor proinflammatory, cellular pathways.

Antiviral and Antiproliferative Roles

Interferon gamma (IFN-γ) exerts potent antiviral effects primarily through the induction of interferon-stimulated genes (ISGs) that directly interfere with viral replication cycles in infected cells. A key mechanism involves the upregulation of protein kinase R (PKR) and 2'-5'-oligoadenylate synthetase (OAS) enzymes. PKR, activated by double-stranded RNA produced during viral infection, phosphorylates eukaryotic initiation factor 2α (eIF2α), thereby inhibiting viral protein translation. Complementing this, OAS enzymes polymerize ATP into 2'-5'-linked oligoadenylates upon sensing viral RNA, which activate RNase L to degrade viral and cellular RNAs, further curtailing viral propagation. These pathways, transcriptionally driven by STAT1-dependent signaling, establish an intracellular antiviral state that limits the spread of diverse RNA and DNA viruses. Additional antiviral activity stems from the robust upregulation of guanylate-binding proteins (GBPs) and Mx proteins, which target later stages of the viral life cycle, particularly assembly and egress. GBPs, among the most strongly induced ISGs by IFN-γ, oligomerize on sites and disrupt the formation of viral capsids or envelopes. Similarly, Mx proteins, dynamin-like GTPases, sequester viral nucleocapsids or components into perinuclear aggregates, preventing maturation; for instance, MxA traps ribonucleoproteins, blocking their nuclear export and assembly. Recent 2024 studies highlight IFN-γ's enhanced efficacy against in respiratory epithelia, where it drives ISG expression to restrict in airway cells, reducing viral loads in nasal and bronchial models. Beyond antiviral defense, IFN-γ mediates antiproliferative effects by arresting the , a critical barrier against uncontrolled in infected or transformed cells. This occurs through the induction of cyclin-dependent kinase inhibitor p21^WAF1/CIP1 and tumor suppressor , which collectively halt progression at G1/S and G2/M checkpoints. IFN-γ elevates mRNA and protein levels via interferon regulatory factor 1 (IRF-1), promoting p21 expression to suppress activity and prevent . In various cell lines, such as hepatocytes and ovarian carcinoma models, this leads to growth inhibition with an IC50 of approximately 1-10 U/mL, underscoring the potency required for therapeutic antiproliferation without excessive toxicity. These mechanisms ensure that IFN-γ not only combats viral threats but also curbs aberrant , contributing to its role in maintaining cellular .

Specialized Biological Activities

Granuloma Formation

Interferon gamma (IFN-γ) plays a pivotal role in orchestrating formation, which serves as a critical immune structure for containing intracellular such as . By activating macrophages and promoting their recruitment to infection sites, IFN-γ facilitates the aggregation of immune cells into compact, organized granulomas that isolate and limit pathogen dissemination. This process is essential for establishing a localized inflammatory environment that enhances pathogen control without widespread tissue damage. A key mechanism involves IFN-γ-induced production of CXCR3 ligands, such as CXCL9 and , by macrophages and dendritic cells, which recruit additional CXCR3-expressing macrophages to the site of . This chemokine-mediated ensures the accumulation of activated macrophages, which, upon IFN-γ , adopt an capable of restricting growth within the developing . In reference to broader macrophage , this enhances the granuloma's structural integrity by promoting cell clustering and fusion into multinucleated giant cells. In tuberculosis models, is crucial for preventing caseation, a process that compromises containment; mice deficient in exhibit disorganized and disseminated due to impaired activation and failure to form protective barriers around . This deficiency highlights 's necessity in maintaining compactness to avert bacterial spread. Furthermore, synergizes with tumor necrosis factor-alpha (TNF-α) to stabilize , as their combined action on boosts production and regulation, ensuring sustained immune cell positioning and pathogen killing. Human studies reveal elevated IFN-γ levels within granulomas, where it drives T-helper 1 cell responses and aggregation, contributing to non-caseating persistence. However, pathological excess of IFN-γ can exacerbate formation, leading to and in patients, as observed in fluids and lesion biopsies.

Reproductive Immunology

Interferon gamma (IFN-γ) plays a critical paradoxical role at the feto-maternal interface, balancing necessary for successful with defense against pathogens. Produced primarily by uterine natural killer (uNK) cells and T cells, IFN-γ modulates the expression of G (HLA-G) on cells, a non-classical molecule essential for maternal of the semiallogeneic . Specifically, IFN-γ enhances HLA-G surface expression on extravillous trophoblasts, inhibiting and NK cell activity while promoting function, thereby preventing alloimmune rejection and supporting placental development. In parallel, IFN-γ provides robust protection against intracellular pathogens at the feto-maternal interface, restricting vertical transmission during pregnancy. Against , IFN-γ upregulates in placental cells, depleting to starve the bacteria and limit fetal infection, as demonstrated in mouse models where IFN-γ signaling reduces bacterial burden and placental damage. Similarly, for , IFN-γ activates immunity-related in trophoblasts and decidual cells, inhibiting parasite replication and transplacental passage; studies in IFN-γ-deficient mice show increased maternal-fetal transmission and fetal loss, underscoring its protective role without excessive inflammation under controlled conditions. The effects of IFN-γ in are highly dose-dependent, highlighting its nature. At low physiological levels, as secreted by cells, IFN-γ promotes uterine vascular remodeling and essential for ; it induces like IP-10 () in decidual cells, which recruit immune cells to support spiral artery transformation and endothelial integrity without disrupting invasion. In contrast, elevated IFN-γ levels, often triggered by excessive , suppress proliferation and motility, leading to impaired implantation and increased risk, as evidenced by murine studies where high-dose IFN-γ administration directly induces through STAT1-mediated pathways.

Antimicrobial Defense

Interferon gamma (IFN-γ) plays a pivotal role in antimicrobial defense by activating macrophages to produce (NO) through induction of inducible (iNOS). In response to IFN-γ stimulation, macrophages upregulate iNOS expression, leading to NO generation that restricts the intracellular growth of pathogens such as Mycobacterium tuberculosis. This mechanism is essential for controlling mycobacterial replication, as evidenced by studies showing that IFN-γ-primed macrophages exhibit enhanced iNOS activity and NO-mediated bacterial killing. Another key mechanism involves IFN-γ-mediated activation of , particularly through the immunity-related M (IRGM), which facilitates xenophagy—the selective autophagic degradation of intracellular bacteria like Salmonella enterica. IFN-γ induces IRGM expression, which in turn promotes the recruitment of guanylate-binding proteins (GBPs) to the Salmonella-containing vacuole, rupturing it and marking the bacteria for ubiquitination and autophagosomal engulfment by adaptors such as NDP52 and p62. This process limits cytosolic bacterial proliferation in . Against intracellular parasites, IFN-γ induces indoleamine 2,3-dioxygenase 1 (IDO1) to deplete , an for parasite survival. In human cells, including fibroblasts and macrophages, IFN-γ upregulates IDO1, resulting in tryptophan catabolism along the and starvation of parasites like , thereby inhibiting their replication. This cell-autonomous defense is a primary IFN-γ effector during . IFN-γ is critical for fungal clearance, particularly against , where it enhances activation and elimination in immunocompromised hosts. A 2023 review highlights IFN-γ's role in augmenting antifungal responses, including in HIV-associated cryptococcal , with adjunctive IFN-γ improving survival and reducing fungal burden through with innate pathways like Dectin-1-mediated β-glucan sensing. In specific infections such as , IFN-γ also contributes to formation to contain pathogens.

Regulatory Mechanisms

Transcriptional Control

The transcriptional control of interferon gamma (IFN-γ) expression is primarily governed by upstream genetic elements and epigenetic modifications that respond to T cell activation signals, such as stimulation and cues like IL-12. These mechanisms ensure lineage-specific production, particularly in Th1 cells and natural killer cells, where IFN-γ transcription is dynamically regulated to mount adaptive immune responses. Key cis-regulatory elements include conserved non-coding sequences (CNS) and hypersensitive sites (HS) in the Ifng locus. For instance, CNS-28 acts as a distal silencer that restrains IFN-γ transcription in NK cells, CD4+ T cells, and CD8+ T cells during innate and adaptive immunity by limiting excessive expression. In contrast, enhancer elements such as the -6.5 kb distal enhancer and HS2 sites facilitate activation; the distal enhancer binds nuclear factor of activated T cells (NFAT) and activator protein-1 (AP-1), promoting cooperative recruitment of co-activators to drive Ifng transcription upon T cell stimulation. Similarly, HS2, located near the promoter, contributes to accessibility and T cell-specific expression by interacting with lineage-determining factors. Epigenetic modifications further fine-tune this regulation. In Th1 cells, trimethylation of at lysine 4 () marks the Ifng promoter and distal enhancers, correlating with active transcription and distinguishing IFN-γ-producing cells from non-producers like Th2 cells. Conversely, microRNAs such as miR-29 suppress IFN-γ expression in resting T cells by targeting mRNA of transcription factors like T-bet and Eomesodermin (Eomes), as well as directly binding Ifng mRNA to inhibit translation and maintain low basal levels. Recent advances in have introduced artificial transcription factors (ATFs) designed to enhance IFN-γ production in therapeutic T cells. In 2025, researchers engineered an ATF with six domains targeting the Ifng promoter, which potently activates transcription in primary T cells, offering potential for boosting IFN-γ secretion in chimeric receptor ( therapies against tumors.

Post-Translational Modulation

Interferon gamma (IFN-γ) is subject to N-linked at two specific residues, Asn25 and Asn97, which occurs co- or post-translationally in the and Golgi apparatus. These modifications attach chains, primarily complex-type at Asn25 and a mix of hybrid and high-mannose types at Asn97, to the protein backbone. Such glycosylation enhances the solubility of IFN-γ by shielding hydrophobic regions, reducing aggregation tendencies, and promoting proper folding during from immune cells like T lymphocytes and natural killer cells. Additionally, the glycans, especially those at Asn25, provide steric hindrance against attack, thereby improving the cytokine's resistance to enzymatic degradation in extracellular environments. The post-translational glycosylation of IFN-γ directly impacts its pharmacokinetic properties, including a of approximately 25 to 35 minutes following . This short circulation time limits the duration of IFN-γ's bioactivity but is mitigated by the stabilizing effects of , which prolongs functional persistence compared to unglycosylated recombinant forms produced in bacterial systems. For instance, glycosylated IFN-γ variants exhibit extended and reduced in therapeutic contexts, underscoring the role of these modifications in modulating efficiency and overall protein .

Clinical and Therapeutic Applications

Established Treatments

Recombinant interferon gamma-1b (IFN-γ1b), marketed as Actimmune, is the only FDA-approved formulation of IFN-γ for clinical use, produced via recombinant DNA technology in Escherichia coli. Actimmune is indicated for reducing the frequency and severity of serious infections in patients with chronic granulomatous disease (CGD), a primary immunodeficiency disorder characterized by defective phagocyte function. The recommended dosage is 50 μg/m² administered subcutaneously three times per week, typically in the evening to mitigate flu-like symptoms. Clinical trials have demonstrated that this regimen reduces the relative risk of severe infections by approximately 70% compared to placebo, with significant decreases in the rate of serious infections from 0.68 to 0.20 per patient-year. Actimmune is also approved for delaying disease progression in patients with severe malignant (SMO), a rare leading to defective function and impaired . In SMO, IFN-γ1b enhances osteoclast activation and increases bone resorption, as evidenced by elevated markers of bone turnover and improved hematopoiesis in long-term studies. Common side effects of Actimmune include flu-like symptoms such as fever, , , , and , which often diminish with continued use or dose adjustment. Elevations in liver enzymes occur frequently, particularly in infants under 1 year, necessitating regular monitoring of hepatic function. The drug should be used with caution in patients with pre-existing cardiac conditions, including congestive , due to potential exacerbation of symptoms from flu-like reactions; close monitoring is recommended in such cases. Long-term data from pediatric CGD cohorts, spanning over three decades since FDA approval in 1990, confirm sustained efficacy in reducing infection rates and improving when used as lifelong adjunctive therapy alongside prophylactic antibiotics. As of 2025, guidelines continue to endorse IFN-γ1b as standard care for infection prophylaxis in pediatric CGD patients aged 1 year and older, with excellent compliance and no emergence of new safety concerns in extended follow-up.

Immunotherapeutic Potential

Interferon gamma (IFN-γ) has shown promise as an in vaccines, where it enhances the expansion and infiltration of (TILs) into metastatic lesions. In clinical trials such as the Mel51 study (NCT00977145), intratumoral administration of IFN-γ following increased vaccine-induced TILs (viTILs) from a of 2% to 30% of total TILs, promoting greater antitumor immune responses. Similarly, phase II trials like CASVAC-0401 (NCT01729663) demonstrated that strategies inducing IFN-γ-secreting T cells led to a significant boost in distant metastasis-free survival, with rates reaching 72.8% at 25 months compared to 27.2% in control arms using interferon alpha-2b, representing an approximate 20-30% relative improvement in response metrics. These effects stem from IFN-γ's ability to upregulate expression on tumor cells, facilitating TIL recognition and without requiring extensive TIL expansion. In chimeric antigen receptor T-cell (CAR-T) therapies for solid tumors, engineering cells to release IFN-γ has emerged as a strategy to overcome barriers to tumor infiltration and persistence. By incorporating inducible promoters or fusion constructs, such as those linking IFN-γ production to recognition, CAR-T cells can locally secrete the to remodel the , enhancing T-cell trafficking and recruitment of endogenous immune effectors. Preclinical models and early 2025 trials, including those evaluating armored CAR-T variants, have shown improved infiltration into dense solid tumors like and , with localized IFN-γ release correlating to reduced exhaustion and heightened antitumor activity. For instance, CAR-T cells engineered with IFN-γ-responsive elements demonstrated up to twofold increases in tumor penetration compared to unmodified counterparts, positioning this approach for phase I/II evaluation in solid malignancies. Despite these advances, clinical translation of IFN-γ-based immunotherapies faces significant barriers, including systemic toxicity and acquired resistance through upregulation. High-dose IFN-γ administration often induces flu-like symptoms, fatigue, and organ-specific toxicities such as , limiting dosing in ongoing trials. Moreover, IFN-γ paradoxically drives resistance by inducing expression on tumor cells via signaling, which dampens T-cell responses and contributes to immune escape in up to 40% of treated patients, as observed in combination regimens with anti-PD-1 inhibitors. Recent innovations from 2024-2025 address these challenges through biased agonists targeting the IFNGR2 subunit of the IFN-γ receptor, enabling selective activation of antitumor pathways without excessive inflammation. Structural insights from the IFN-γ–IFNGR complex have guided the of these agonists, which preferentially induce immunostimulatory genes like those for MHC-I while suppressing immunosuppressive ones such as , thereby enhancing antitumor immunity in preclinical and models with reduced toxicity. In immune disorders like (IBD), JAK inhibitors such as and modulate IFN-γ signaling by blocking JAK1/ phosphorylation, leading to decreased mucosal IFN-γ levels and improved remission rates in phase III trials (e.g., NCT02819635), with 2025 data showing sustained clinical benefits in patients refractory to biologics. These targeted modulators represent a shift toward precision immunotherapeutics, minimizing off-target effects while preserving IFN-γ's core immune-enhancing roles.

Disease Associations

Deficiency in interferon gamma (IFN-γ) signaling, particularly due to mutations in the IFN-γ receptor genes IFNGR1 or IFNGR2, leads to Mendelian susceptibility to mycobacterial disease (MSMD), a rare characterized by severe, recurrent infections with weakly virulent mycobacteria such as Bacillus Calmette-Guérin (BCG) and environmental mycobacteria, as well as species.00124-6/fulltext) These autosomal recessive or dominant mutations impair the IFN-γ/IL-12 pathway, resulting in defective activation and formation, which heightens vulnerability to intracellular pathogens without broadly compromising immunity to other infections. Excess IFN-γ production contributes to the pathogenesis of by promoting the destruction of pancreatic s through upregulation of class I molecules, enhancing autoreactive + T cell recognition and infiltration into islets. In synergy with cytokines like IL-1β and TNF-α, IFN-γ induces via production and stress, accelerating insulitis and in susceptible individuals. Similarly, elevated IFN-γ drives (HLH), a hyperinflammatory syndrome marked by uncontrolled T cell and activation, leading to , hemophagocytosis, and multiorgan failure, particularly in familial forms linked to perforin or Rab27a defects that indirectly amplify IFN-γ signaling. In autoimmune conditions like (RA), IFN-γ levels are markedly elevated in and tissue, where it sustains Th1-polarized inflammation by activating synovial fibroblasts and macrophages to produce proinflammatory mediators such as TNF-α and IL-1, thereby promoting joint erosion and pannus formation. This dysregulation correlates with increased IFN-γ-producing + and + T cells in the synovium, exacerbating degradation through enhanced expression. Recent studies as of 2025 highlight IFN-γ dysregulation in Crohn's disease, where hyperactivation of IFN-γ pathways in intestinal tissues disrupts epithelial barrier integrity and amplifies Th1-mediated inflammation, contributing to chronic mucosal damage and fistulizing complications. In perianal fistulizing Crohn's, elevated IFN-γ signatures in plasma and lesional macrophages correlate with impaired wound healing and persistent granulomatous inflammation, underscoring its role in disease progression.

References

  1. [1]
    INTERFERON-gamma - PubChem - NIH
    Interferon gamma is a Type 1 inflammatory cytokine and the only type II interferon. It has antitumor properties, antiviral activities, and important ...
  2. [2]
    Biologic functions of the IFN‐γ receptors - PMC - PubMed Central
    Interferon‐gamma (IFN‐γ) is a cytokine that plays an important role in inducing and modulating an array of immune responses.
  3. [3]
    3458 - Gene ResultIFNG interferon gamma [ (human)] - NCBI
    Aug 19, 2025 · This gene encodes a soluble cytokine that is a member of the type II interferon class. The encoded protein is secreted by cells of both the innate and adaptive ...
  4. [4]
    Gamma Interferon: From Antimicrobial Activity to Immune Regulation
    Dec 11, 2014 · Gamma interferon (IFNγ) was discovered in 1965 as an antiviral activity in white blood cell cultures stimulated with the lymphocyte mitogen phytohemagglutinin.
  5. [5]
    Interferon γ and Its Important Roles in Promoting and Inhibiting ... - NIH
    This review focuses on the diverse positive and negative roles of IFN-γ in immune cell activation and differentiation leading to protective immune responses.
  6. [6]
    Clinical Use of Interferon-γ - PMC - PubMed Central
    Interferon gamma (IFN-γ), a cytokine with diverse roles in the innate and adaptive immune responses, was discovered in 1965. IFN-γ function has been strongly ...
  7. [7]
    NF‐κB activation by the viral oncoprotein StpC enhances IFN‐γ ...
    Jun 17, 2008 · We analyzed the influence of StpC on IFN-γ production by unstimulated Jurkat T cells. Our results demonstrated that StpC enhances IFN-γ ...
  8. [8]
    Modular Utilization of Distal cis-Regulatory Elements Controls Ifng ...
    Jul 23, 2010 · We have mapped interactions of three trans-acting factors—NF-κB, STAT4, and T-bet—with cis elements in the Ifng locus. We find that RelA is ...
  9. [9]
    Evolution of Interferons and Interferon Receptors - Frontiers
    Mar 1, 2017 · ... IFN gene evolution. Lastly, the type II (IFN-γ) genes have a universal 4 exon/3 intron organization from cartilaginous fish to mammals, with ...
  10. [10]
    Structural conservation of interferon gamma among vertebrates - NIH
    Our analysis shows that type II interferon (IFN-γ) function evolved before the tetrapod-fish split, a finding that contrasts earlier studies showing its ...
  11. [11]
  12. [12]
    IFNG - Interferon gamma - Homo sapiens (Human) - UniProt
    Type II interferon produced by immune cells such as T-cells and NK cells that plays crucial roles in antimicrobial, antiviral, and antitumor responses.Missing: exons | Show results with:exons
  13. [13]
    1HIG: THREE-DIMENSIONAL STRUCTURE OF RECOMBINANT ...
    Interferon-gamma, which is dimeric in solution, crystallizes with two dimers related by a noncrystallographic twofold axis in the asymmetric unit. The protein ...
  14. [14]
    Three-Dimensional Structure of Recombinant Human Interferon-γ
    Interferon-γ, which is dimeric in solution, crystallizes with two dimers related by a noncrystallographic twofold axis in the asymmetric unit.
  15. [15]
    The Role of Structure in the Biology of Interferon Signaling - Frontiers
    In human IFNβ, the AB-loop arches toward the N-terminal end of helix-F, “over” helix F itself, where the loop connects to helix F by a disulfide bond. In ...
  16. [16]
    N-glycosylation of human interferon-gamma: glycans at Asn-25 are ...
    Human interferon-gamma (IFN-gamma) is a secretory, dimeric glycoprotein that forms a compact globular structure with potential N-linked glycosylation sites ...
  17. [17]
    Recombinant Human IFN-gamma Protein 285-IF-100 - R&D Systems
    Rating 4.6 (24) Recombinant Human IFN-gamma (Catalog # 285-IF) has a molecular weight (MW) of 34.9 kDa as analyzed by SEC-MALS, suggesting that this protein is a homodimer.
  18. [18]
    Cellular sources and targets of IFN-γ-mediated protection against ...
    IFN-γ is produced by activated NK cells [1], CD4+ Th1 cells [2] and CD8+ cytotoxic T cells [3] and is a critical component of the cell-mediated immune response ...
  19. [19]
    Intraepithelial type 1 innate lymphoid cells are a unique subset ... - NIH
    Together, these results indicate that intraepithelial ILC1 are poised for robust IFN-γ production, which may be important for orchestrating innate responses to ...
  20. [20]
    Endotoxin-Induced Gamma Interferon Production: Contributing Cell ...
    After LPS challenge, NK cells (60%) and NKT cells (25%) are the main IFN-γ producers. IL-12 is the major stimulator, with IL-10 as a major inhibitor.Results · Nk And Nkt Cells Are The... · Lps-Induced Ifn-γ...
  21. [21]
    Autocrine IFN-γ promotes naïve CD8 T cell differentiation and ...
    IFN-γ production by CD8 T cells is highest when the cells are re-stimulated with Ag at 48 to 72 hours after their initial stimulation, i.e. after ...
  22. [22]
    Antigen-Independent IFN-γ Production by Human Naïve CD4+ ... - NIH
    The amount of IFN-γ produced by five different donors ranged from 863.8 to 10,829.7 pg/ml after 24 hour stimulation. The production of IFN-γ by IL-12 plus IL-18 ...
  23. [23]
    Pulmonary epithelial cells are a source of interferon-gamma in ...
    Pulmonary epithelial cells are a source of interferon-gamma in response to Mycobacterium tuberculosis infection ... Immunol Cell Biol. 2007 Apr-May;85(3):229-37.
  24. [24]
    The transcription factor T-bet is induced by multiple pathways and ...
    IL-12 also induces T-bet in an IFN-γ-Stat1-independent manner in CD8+ T cells (Yang et al., 2007). The relative importance of IL-12 and IFN-γ in T-bet induction ...
  25. [25]
    IFNγ: signalling, epigenetics and roles in immunity, metabolism ...
    Signalling by the JAK-STAT pathway, including activation of STAT1 by IFNγ, is typically transient, with a peak signal occurring at 15–60 minutes and resolution ...
  26. [26]
    IFN-γ: A cytokine at the right time, is in the right place - PMC - NIH
    Interferon gamma has long been studied as a critical mediator of tumor immunity. In recent years, the complexity of cellular interactions that take place in the ...2. Ifn-Gamma Biology · Fig. 1 · 4. Tumor Microenvironment<|control11|><|separator|>
  27. [27]
    Single Cell Analysis Reveals Regulated Hierarchical T Cell Antigen ...
    Low concentrations of TCR ligand elicit only interferon-γ (IFN-γ) production. ... Most cells producing only IFN-γ show less TCR downmodulation than cells ...
  28. [28]
    Interleukin-12: an update on its immunological activities, signaling ...
    IFN-γ and, to a lesser extent, IL-12 positively regulated the IL-12Rβ2 subunit but had no effect on IL-12Rβ1. IL-12 induced the phosphorylation and nuclear ...
  29. [29]
    Role of Endogenous IFN-γ in Macrophage Programming Induced by ...
    To evaluate the direct effect of LPS, IFN-γ IL-12, or IL-18, cells were cultured with 0.1–50 μg/mL LPS (Sigma), 2.5–20 ng/mL rIFN-γ (PharMingen), or 0.5–10 ng/ ...
  30. [30]
    Mechanisms of fungal pathogenic DNA-activated STING pathway in ...
    Sep 1, 2025 · The STING pathway is indispensable for antifungal immunity. On one hand, type I interferons induced by its activation enhance natural killer (NK) ...
  31. [31]
  32. [32]
  33. [33]
  34. [34]
    Gamma Interferon Receptor - an overview | ScienceDirect Topics
    The IFN-γ receptor chains are both type I transmembrane proteins of the class 2 cytokine receptor family, which is characterized by the presence of tandem ...Missing: immunoglobulin- | Show results with:immunoglobulin-
  35. [35]
    Mechanisms of type-I- and type-II-interferon-mediated signalling
    May 1, 2005 · The transcription of type II IFN (IFN-γ)-dependent genes is regulated by GAS elements, and STAT1 is the most important IFN-γ-activated ...
  36. [36]
    Structure of the IFNγ receptor complex guides design of biased ...
    Feb 27, 2019 · The complete 2:2:2 IFNγ receptor complex is star-shaped with a two-fold symmetry imposed by the IFNγ homodimer (Fig. 2a). The structure reveals ...
  37. [37]
    Analysis of murine interferon-gamma binding to its receptor on intact ...
    Binding of 125I-IFN-gamma to intact cells and their solubilized membranes has shown a single class of receptor with Kd values of 1.9 x 10(-9) M and 1.3 x 10(-8) ...Missing: kinetics | Show results with:kinetics
  38. [38]
    Using structure-function information from IFN-γ-binding proteins and ...
    Binding of an IFN-γ dimer to two IFNGR1 chains triggers the association of two IFNGR2 chains, leading to the formation of a 2:2:2 signaling complex ...
  39. [39]
    Structure of the IFNγ receptor complex guides design of biased ...
    Feb 27, 2019 · The complete 2:2:2 IFNγ receptor complex is star-shaped with a two-fold symmetry imposed by the IFNγ homodimer (Fig. 2a). The structure reveals ...
  40. [40]
    Purification and partial characterization of a receptor protein ... - PNAS
    Cross-linking of 125I-labeled rMuIFN-gamma to its receptor yielded a complex of 125 +/- 5 kDa, consistent with the binding of the dimeric form of mouse ...
  41. [41]
    The effects of extracellular pH on immune function - Lardner - 2001
    Apr 1, 2001 · A pH dependence of immunoglobulin G (IgG) binding by the neonatal Fc receptor has also been shown, and high-affinity binding was observed at pH ...
  42. [42]
  43. [43]
    Interferon-gamma Induces Tyrosine Phosphorylation of ... - PubMed
    Coimmunoprecipitation experiments revealed that Jak1 was associated with the IFN-gamma R prior to ligand binding, whereas Jak2 became part of ...Missing: et al
  44. [44]
    Activation of Transcription by IFN-γ: Tyrosine Phosphorylation of a ...
    The IFN-γ-dependent activation of the 91-kilodalton DNA binding protein required cytoplasmic phosphorylation of the protein on tyrosine.
  45. [45]
    tyrosine phosphorylation of a 91-kD DNA binding protein - PubMed
    Activation of transcription by IFN-gamma: tyrosine phosphorylation of a 91-kD DNA binding protein. Science. 1992 Dec 11;258(5089):1808-12.
  46. [46]
    GAS elements: a few nucleotides with a major impact on cytokine ...
    GAS elements are short DNA stretches required for rapid gene induction by IFN-gamma, and are the general nuclear end of Jak-Stat signaling pathways.
  47. [47]
    C-terminal truncation of IFN-γ inhibits proinflammatory macrophage ...
    Jun 20, 2018 · In both macrophage cell lines, IFN-γ led to the characteristic phosphorylation of STAT1 at Tyr701 after 15 min with subsequent upregulation of ...Missing: peak | Show results with:peak
  48. [48]
    TNFα and IFNγ rapidly activate PI3K-AKT signaling to drive ...
    Oct 4, 2022 · TNFα and IFNγ rapidly activate PI3K-AKT signaling to drive glycolysis that confers mesenchymal stem cells enhanced anti-inflammatory property.
  49. [49]
    Role of the Akt pathway in mRNA translation of interferon-stimulated ...
    Our data establish that Akt activity is essential for up-regulation of key IFN-α- and IFN-γ-inducible proteins, which have important functional consequences in ...
  50. [50]
    Roles of IFN-γ in tumor progression and regression: a review
    Sep 29, 2020 · Interferon-γ (IFN-γ) plays a key role in activation of cellular immunity and subsequently, stimulation of antitumor immune-response.
  51. [51]
    Interferon‐γ expression by Th1 effector T cells mediated by the p38 ...
    MAP kinase pathways play a critical role in the activation of T cells during the immune response. Several studies have shown the involvement of the ERK pathway ...
  52. [52]
    Mitogen-activated Protein Kinases Mediate ... - ScienceDirect.com
    p38- and ERK-dependent pathways, through effects on the AP-1 complex, activate the hiNOS promoter in cells stimulated with CM or LPS/IFN-γ.
  53. [53]
    IFN-γ–mediated inhibition of MAPK phosphatase expression results ...
    Oct 15, 2008 · Here we report that stimulation with IFN-γ prolongs the pattern of ERK activity induced by M-CSF in macrophages.
  54. [54]
    NF-κB in biology and targeted therapy: new insights and ... - Nature
    Mar 4, 2024 · We investigate the interaction of NF-κB signaling with other important pathways, including PI3K/AKT, MAPK, JAK-STAT, TGF-β, Wnt, Notch, Hedgehog ...
  55. [55]
    Epigenetic Regulation of Inflammatory NF-κB Target Genes by IFN-γ ...
    Dec 2, 2024 · This study investigated mechanisms by which by IFN-γ-mediated priming augments TLR-induced expression of NF-κB target genes in primary human ...Missing: STAT4 | Show results with:STAT4
  56. [56]
    Synergistic Activation of Inflammatory Cytokine Genes by Interferon ...
    Sep 19, 2013 · Our results provide a synergy mechanism whereby IFN-γ creates a primed chromatin environment to augment TLR-induced gene transcription.<|control11|><|separator|>
  57. [57]
    Interferon-gamma acutely induces calcium influx in human microglia
    The influx of Ca(2+) may serve a signaling role in microglia linking IFN-gamma to functional responses of the cells to infiltrating T lymphocytes into the ...
  58. [58]
    Interferon-γ-induced calcium influx in T lymphocytes of multiple ...
    This study suggests that the IFN-γ-induced Ca 2+ -influx can act as a complementary mechanism in the activation of blood T lymphocytes from MS and RA patients.Missing: flux | Show results with:flux
  59. [59]
    IFN-gamma activates the C3G/Rap1 signaling pathway - PubMed
    Feb 15, 2000 · We report that IFN-gamma activates a distinct signaling cascade involving the c-cbl protooncogene product, CrkL adapter, and small G protein Rap1.
  60. [60]
    An Atlas of Protein Phosphorylation Dynamics During Interferon ...
    Jul 2, 2024 · Widespread Temporal Changes to the Human Proteome and Phosphoproteome During Stimulation with IFNα, IFNβ, IFNω, IFNγ, and IFNλ. To initially ...Chromatin Remodeling... · Stat1 T704 Contributes To... · Plekhg3 Has A Role In Ifn-I...Missing: CrkL cytoskeletal
  61. [61]
  62. [62]
  63. [63]
  64. [64]
  65. [65]
    OAS and PKR are not required for the antiviral effect of Ad:IFN ...
    Three interferon-gamma (IFN-gamma)-induced antiviral pathways have been reported. ... IFN-gamma induces OAS and PKR when other viruses invade the nervous system.
  66. [66]
    Interferon-inducible antiviral effectors | Nature Reviews Immunology
    Constitutive levels of OAS and PKR mean that these proteins function not only as effector proteins but also as double-stranded RNA (dsRNA)-specific PRRs to ...Abstract · Main · Glossary
  67. [67]
    The Antiviral Response to Gamma Interferon - PMC - PubMed Central
    Inhibitions of viral uptake and uncoating, viral RNA and DNA replication, and viral protein synthesis and assembly have all been reported for different virus- ...Missing: upregulation | Show results with:upregulation
  68. [68]
    Interferon γ–Induced Human Guanylate Binding Protein 1 Inhibits ...
    Among the most abundant proteins induced by IFN-γ are guanylate binding proteins (GBPs), which belong to the superfamily of large GTPases and are widely ...Missing: assembly | Show results with:assembly
  69. [69]
    Guanylate-binding proteins in virus infection - Portland Press
    Aug 3, 2023 · Guanylate-binding proteins (GBPs) are immune GTPases that are induced in response to interferon stimulation/pathogen infection.Gbps In Virus Infection · Gtpase Activity-Dependent... · Role Of Gbps In Interferon...
  70. [70]
    Interferon-stimulated genes and their antiviral activity against SARS ...
    Aug 22, 2024 · This review provides a detailed overview of fundamental aspects of the diverse ISGs identified for their antiviral properties against SARS-CoV-2.
  71. [71]
    Analysis of IFN-gamma-induced cell cycle arrest and cell death in ...
    IFN-gamma induced cell cycle arrest at the initial stage, followed by cell death. A protein synthesis inhibitor, cycloheximide, significantly inhibited cell ...Missing: IC50 proliferation
  72. [72]
    Interferon-gamma inhibits proliferation of rat vascular smooth muscle ...
    The dose dependencies of the two effects were similar (IC50 = 4 U/ml for the anti-proliferation and EC50 = 3 U/ml for cGMP formation) and the effect of IFN- ...Missing: antiproliferative units
  73. [73]
    In the Thick of It: Formation of the Tuberculous Granuloma ... - Frontiers
    Amongst type 1 cytokines, IFN-γ and TNF have been most closely associated with Mtb pathogenesis and granuloma formation. Both IFN-γ and TNF are crucial to the ...
  74. [74]
    In Situ Studyof Abundant Expression of Proinflammatory ...
    IFN-γ induces macrophages and dendritic cells (DC) to produce IFN-γ-inducible CXCR3 ligands, CXCL9/monokine induced by IFN-γ (Mig), CXCL10/IFN-γ-inducible ...
  75. [75]
    Chemokines in Innate and Adaptive Granuloma Formation - Frontiers
    This tract will discuss the contribution of chemokines to the development of innate and adaptive granuloma formation, as well as describe their relationship to ...
  76. [76]
    αβ T Cell Receptor-positive Cells and Interferon-γ, but not Inducible ...
    Progressive caseation of pulmonary granulomas never occurred in αβ TCR-KO, IL-12–KO, and IFN-γ–KO mice and was reduced in CD4-KO mice. In summary, αβ TCR+ cells ...Materials And Methods · Results · Discussion
  77. [77]
    (PDF) Disseminated tuberculosis in IFN-?? gene disrupted mice
    May 15, 2025 · Mice have been developed which fail to produce IFN-gamma (gko), because of a targeted disruption of the gene for IFN-gamma. Upon infection with ...
  78. [78]
    Synergy between individual tumor necrosis factor-dependent ...
    The effects of TNF in Mtb granuloma formation are likely related to the chemokine network induced during infection. We have identified a simplified model of ...<|control11|><|separator|>
  79. [79]
    IFN-γ–Producing T-Helper 17.1 Cells Are Increased in Sarcoidosis ...
    Jul 30, 2015 · Sarcoidosis is a granulomatous disease in which the inflammation is thought to be driven in part by activated T-helper (Th) type 1 effector T ...
  80. [80]
    Immune mechanisms of granuloma formation in sarcoidosis ... - JCI
    Jan 2, 2024 · Thus, the granuloma microenvironment in sarcoidosis is highly inflammatory, driven by these IFN-γ–producing metabolically active macrophages.
  81. [81]
    HLA-G and Recurrent Pregnancy Loss - MDPI
    In 2008, Blanco et al. [38] demonstrated that IFN-gamma enhances the expression of HLA-G, and this mechanism might be involved in the attempt of decidual ...
  82. [82]
    HLA‐G and immune tolerance in pregnancy - The FASEB Journal
    May 1, 2005 · A second mechanism by which HLA-G may induce maternal tolerance to fetal antigens is to reduce or prevent cytotoxic activity of CD8 T cells ...
  83. [83]
    HLA-G: An Important Mediator of Maternal-Fetal Immune-Tolerance
    HLA-G is now established as a key molecule that plays an important role in the generation of fetal-induced maternal immunological tolerance.
  84. [84]
    Indoleamine 2,3-dioxygenase is regulated by IFN-gamma ... - PubMed
    Indoleamine 2,3-dioxygenase is regulated by IFN-gamma in the mouse placenta during Listeria monocytogenes infection ... protects the fetal allograft from T cell- ...
  85. [85]
    Protective and Pathogenic Effects of Interferon Signaling During ...
    IFN-γ signaling at the maternal-fetal interface not only promotes differentiation of decidual NK cells but also facilitates formation of the placenta and ...
  86. [86]
    Maternal–fetal transmission of Toxoplasma gondii in interferon-γ ...
    The effect of interferon (IFN)-γ, which plays a crucial role in the protective immunity against T. gondii infection, on maternal–fetal transmission of T.
  87. [87]
    Role of NK Cells and Gamma Interferon in Transplacental Passage ...
    T. gondii infection triggers an increase in NK cells and is associated with IFN-γ production (11), which protects against acute toxoplasmosis in mice (29).
  88. [88]
    Interferon Contributes to Initiation of Uterine Vascular Modification ...
    Aug 6, 2025 · These in vivo findings strongly suggest that uNK cell-derived IFN-gamma modifies the expression of genes in the uterine vasculature and stroma, ...<|separator|>
  89. [89]
    The role of IP-10 and its receptor CXCR3 in early pregnancy
    IP-10 and its receptor CXCR3 play an important role in the formation of the pro-inflammatory immune microenvironment during early pregnancy.
  90. [90]
    High-Dose Interferon-γ Promotes Abortion in Mice by Suppressing ...
    As a classic type I cytokine, interferon-gamma (IFN-γ) is known to manifest a miscarriage-inducing effect, although the specific mechanism is still unclear.Missing: IP- | Show results with:IP-
  91. [91]
    WO2020028275A1 - Interferon-gamma biased agonists
    In various embodiments of the disclosure, some of the biased agonist IFN-g ... ifn gamma. Family To Family Citations. * Cited by examiner, † Cited by ...
  92. [92]
    Maternal gut microbiota influences immune activation at the ... - Nature
    May 9, 2025 · It has been shown that IFN-γ of uterine NK (uNK) cell origin is essential for spiral arterial remodeling in mouse pregnancy. In humans, first- ...Missing: agonists | Show results with:agonists
  93. [93]
    Preexisting Heterogeneity of Inducible Nitric Oxide Synthase ...
    Sep 19, 2022 · Nitric oxide production by iNOS is one of the IFN-γ-induced mechanisms that macrophages use to control intracellular growth of M. tuberculosis ( ...
  94. [94]
    Interferon–γ in Salmonella Pathogenesis: New Tricks for an Old Dog
    The cytokine IFN-γ has well-established antibacterial properties against Salmonella and other intracellular microbes, for example its capacity to activate ...
  95. [95]
    IFNs in host defence and parasite immune evasion during ...
    Feb 7, 2024 · IDO1 depletes intracellular tryptophan that is required for intracellular growth of T. gondii; hence, IFN-γ-induced IDO1 upregulation and ...
  96. [96]
    Immune responses to human fungal pathogens and therapeutic ...
    Jan 4, 2023 · In this Review, we synthesize our current understanding of the cellular and molecular determinants of mammalian antifungal immunity.
  97. [97]
    Modular utilization of distal cis-regulatory elements controls Ifng ...
    Jul 23, 2010 · Distal cis-regulatory elements play essential roles in the T lineage-specific expression of cytokine genes. We have mapped interactions of ...Missing: HS2 | Show results with:HS2
  98. [98]
    T Helper Cell Differentiation: Regulation by cis Elements and ...
    Studies have shown that distal regulatory elements control the Ifng and Th2 cytokine loci and are primary targets for tissue-specific transcription factors.
  99. [99]
    Restraint of IFN-γ expression through a distal silencer CNS-28 for ...
    May 9, 2023 · CNS-28 restrains Ifng transcription in NK cells, CD4 + cells, and CD8 + T cells during both innate and adaptive immune responses.
  100. [100]
    A distal enhancer in the interferon-gamma (IFN ... - PubMed - NIH
    Feb 6, 2004 · This element binds two transcription factors known to be essential for IFN-gamma expression: nuclear factor of activated T cells, an ...Missing: HS2 | Show results with:HS2<|separator|>
  101. [101]
    Restraint of IFN-γ expression through a distal silencer CNS–28 ... - NIH
    CNS–28 restrains Ifng transcription in NK cells, CD4 + and CD8 + T cells during both innate and adaptive immune responses.
  102. [102]
    Global Mapping of H3K4me3 and H3K27me3 Reveals Specificity ...
    Surprisingly, Th1 cells showed strong H3K4me3 signals at both the distal and proximal promoters ... cells, but not other non-IFN-γ-producing cells. Figure ...
  103. [103]
    MicroRNA-29 regulates T-box transcription factors and interferon-γ ...
    Aug 26, 2011 · MicroRNA-29 regulates helper T cell differentiation by repressing genes, including T-bet and Eomes, which induce interferon-γ production.
  104. [104]
    An artificial transcription factor that activates potent interferon-γ ...
    Jan 7, 2025 · This paper introduces the design and characterization of an artificial transcription factor (ATF) protein that recognizes the IFNG gene with six zinc finger ...
  105. [105]
    [PDF] ACTIMMUNE® (Interferon gamma-1b) - 12345 - accessdata.fda.gov
    The following data on adverse reactions are based on the subcutaneous administration of ACTIMMUNE at a dose of 50 mcg/m², three times weekly, in patients with ...
  106. [106]
    The use of interferon-gamma therapy in chronic granulomatous ...
    It has demonstrated clinical benefit in the majority of patients with CGD, reducing the relative risk of severe infections in 70%.
  107. [107]
    Long-Term Treatment of Osteopetrosis with Recombinant Human ...
    Jun 15, 1995 · Long-term therapy with interferon gamma in patients with osteopetrosis increases bone resorption and hematopoiesis and improves leukocyte function.
  108. [108]
    Side Effects | ACTIMMUNE® (Interferon gamma-1b) HCP
    The most common side effects of ACTIMMUNE are flu-like symptoms, such as fever, headache, chills, myalgia, or fatigue. These symptoms may decrease in severity ...
  109. [109]
    Actimmune Side Effects: Common, Severe, Long Term - Drugs.com
    Sep 4, 2025 · Very common (10% or more): Hepatic enzymes increased. Other. Very common (10% or more): Fever, chills, fatigue. References. 1. (2019) ...
  110. [110]
    [PDF] Actimmune - Amgen
    Acute and transient “flu-like” symptoms such as fever and chills induced by ACTIMMUNE at doses of 250 mcg/m2/day (greater than. 10 times the weekly recommended ...Missing: side | Show results with:side
  111. [111]
    Safety and effectiveness of long-term interferon gamma therapy in ...
    rIFN-gamma is a safe and effective adjunctive therapy for reducing the frequency and severity of serious infections in CGD patients. Publication types. Clinical ...Missing: Actimmune | Show results with:Actimmune
  112. [112]
    Pediatric Chronic Granulomatous Disease Treatment & Management
    Oct 10, 2024 · Interferon-gamma is now recommended as life-long therapy for infection prophylaxis in CGD. Curative approaches (HSCT). HSCT is the only curative ...
  113. [113]
    Overview of Mendelian Susceptibility to Mycobacterial Diseases ...
    Jun 12, 2025 · This review deals with the pathogenesis, clinical features, diagnosis, and treatment of Mendelian susceptibility to mycobacterial infections.
  114. [114]
    Interleukin-1 Plus γ-Interferon-Induced Pancreatic β-Cell ...
    Feb 1, 2002 · Cytokines have been implicated in pancreatic β-cell destruction leading to type 1 diabetes. In vitro, a combination of γ-interferon (IFN-γ) ...
  115. [115]
    Familial hemophagocytic lymphohistiocytosis hepatitis is mediated ...
    Jun 7, 2022 · Interferon gamma (IFN-γ) is the main cytokine driving organ dysfunction in Familial Hemophagocytic Lymphohistiocytosis (FHL).
  116. [116]
    New insights into IFN-γ in rheumatoid arthritis: role in the era of JAK ...
    A skewing towards T cells with a Th1 cytokine profile including IFN-γ was observed in synovial fluids from RA patients [Citation36]. A recent meta-analysis ...
  117. [117]
    High percentages and activity of synovial fluid NK cells present in ...
    Feb 4, 2019 · Rheumatoid Arthritis (RA) causes chronic inflammation of joints. The cytokines TNFα and IFNγ are central players in RA, however their source ...
  118. [118]
    Revealing hyperactivated IFN-γ pathways in perianal fistulizing ...
    Sep 2, 2025 · Abstract. Up to 40% patients with Crohn's disease suffer from perianal disease, a debilitating complication with unclear etiology.
  119. [119]
    IFN-γ-Induced intestinal epithelial cell-type-specific programmed cell ...
    IFN-γ induces PANoptosis in enterocytes, which can be treated with a selective JAK1 inhibitor in patients with CD.Abstract · Introduction · Method · Discussion