Interferons are a family of cytokines produced by host cells in response to viral infections and other pathogens, acting as key mediators of the innate immune response by inducing an antiviral state in neighboring cells and coordinating broader immune defenses.[1] Discovered in 1957 by Alick Isaacs and Jean Lindenmann during experiments on viral interference in influenza-infected chick embryo cells, interferons were initially characterized as soluble proteins that confer resistance to viral replication without directly neutralizing the virus.[2][3]Interferons are categorized into three types based on their structure, receptor usage, and cellular sources: type I interferons (including over a dozen IFN-α subtypes, IFN-β, IFN-ε, IFN-κ, and IFN-ω), which are produced by most nucleated cells and bind the ubiquitously expressed IFNAR receptor; type II interferon (IFN-γ), secreted primarily by activated T cells and natural killer (NK) cells and binding the IFNGR receptor; and type III interferons (IFN-λ1 through IFN-λ4), mainly expressed by epithelial cells at mucosal surfaces and utilizing the IFNLR1/IL-10R2 receptor complex.[1][4][5]These proteins exert pleiotropic effects, including direct antiviral activity through the upregulation of hundreds of interferon-stimulated genes (ISGs) such as MxA, OAS, and PKR that inhibit viral replication and spread; antiproliferative effects on tumor cells by arresting cell cycle progression and promoting apoptosis; and immunomodulatory roles by enhancing NK cell cytotoxicity, macrophage activation, antigen presentation via MHC class I molecules, and differentiation of T cells and dendritic cells.[1][6][4]Type I and III interferons share significant functional overlap in establishing early antiviral barriers, particularly at epithelial interfaces, while type II interferon uniquely drives Th1 immune responses and macrophage polarization critical for combating intracellular bacteria and tumors.[7][6] Their signaling converges on the JAK-STAT pathway, leading to transcriptional activation of ISGs, but dysregulation can contribute to autoimmune diseases like systemic lupus erythematosus or exacerbate chronic inflammation.[6][8]Clinically, recombinant interferons have been pivotal in treating conditions such as chronic hepatitis B and C infections, multiple sclerosis, and certain malignancies including hairy cell leukemia and melanoma, though their use has evolved with the advent of more targeted therapies due to side effects like flu-like symptoms and cytopenias.[2][9] Ongoing research highlights their roles in emerging viral threats, cancer immunotherapy, and mucosal immunity, underscoring their enduring impact on biomedicine six decades after discovery.[9][5]
Classification and Types
Type I Interferons
Type I interferons constitute a large family of cytokines that play a central role in the innate immune response to viral infections. In humans, this family includes multiple subtypes of IFN-α, specifically 13 functional genes encoding the IFN-α subtypes (IFN-α1, IFN-α2, IFN-α4, IFN-α5, IFN-α6, IFN-α7, IFN-α8, IFN-α10, IFN-α13, IFN-α14, IFN-α16, IFN-α17, and IFN-α21), along with single genes for IFN-β, IFN-ε, IFN-κ, and IFN-ω, totaling 17 distinct subtypes. These proteins share a conserved structure consisting of five α-helices (A–E) arranged in a characteristic long-chain α-helical bundle, which facilitates their binding to cellular receptors and enables diverse biological activities.[8][10][11]The genomic organization of type I interferon genes is compact and clustered on the short arm of chromosome 9p21.3, spanning approximately 400 kb, with most genes lacking introns and consisting of a single exon. This arrangement, including 17 functional genes and several pseudogenes, reflects evolutionary duplication events that expanded the family. Notably, the IFN-κ gene is an exception, located separately on chromosome 9 but still part of the broader cluster. Among these, IFN-α genes exhibit the greatest multiplicity, while IFN-β is encoded by a single inducible gene and exists as a monomeric protein.[12][13][14]Key properties of type I interferons include their production by virtually all nucleated cells in response to viral pathogens, leading to rapid induction—often within hours of infection—via pattern recognition receptors. They bind exclusively to the heterodimeric IFNAR receptor complex (IFNAR1 and IFNAR2) on the cell surface, triggering shared signaling pathways that amplify antiviral states. These interferons primarily establish an antiviral milieu by inducing hundreds of interferon-stimulated genes, though their pleiotropic effects extend to immune modulation.[15][16][11]
Type II Interferon
Type II interferon refers to a single cytokine, interferon gamma (IFN-γ), which is a dimeric protein lacking structural similarity to the alpha-helical bundles found in type I and type III interferons.[17] Unlike the monomeric forms of other interferon types, IFN-γ forms a homodimer through non-covalent bonds in a unique head-to-tail orientation, featuring an intercalated arrangement of its six alpha helices per subunit.[11] This compact structure, with a molecular weight of approximately 34-40 kDa for the dimer, enables high receptor specificity and distinct signaling capabilities.[18]In humans, IFN-γ is encoded by the IFNG gene located on the long arm of chromosome 12 at position 12q15.[19] The gene spans about 5 kb and consists of four exons separated by three introns, with the mature protein derived from a 166-amino-acid precursor after cleavage of a 20-residue signal peptide.[20] This genomic organization supports the production of a secreted glycoprotein that is heavily glycosylated at specific asparagine residues, contributing to its stability and bioactivity.[21]IFN-γ is primarily secreted by activated natural killer (NK) cells and T lymphocytes, particularly CD4+ Th1 cells and CD8+ cytotoxic T cells, in response to immune challenges.[21] It binds to a heterodimeric receptor complex composed of IFNGR1 and IFNGR2 subunits, initiating signaling that prominently activates macrophages to enhance antimicrobial activity and antigen presentation.[22] This cytokine plays a pivotal role in promoting Th1-biased immune responses, driving cell-mediated immunity through upregulation of major histocompatibility complex class I and II molecules on target cells.[19]
Type III Interferons
Type III interferons, also known as lambda interferons (IFN-λ), constitute a distinct subclass of interferons that exhibit structural homology to the interleukin-10 (IL-10) family of cytokines, with approximately 11–13% amino acid sequence identity to IL-10 and its relatives.[23] These cytokines were identified as antiviral agents in the early 2000s and are characterized by their role in mucosal immunity, bridging innate antiviral responses with barrier-specific defense mechanisms.[24]In humans, the type III interferon family comprises four members: IFN-λ1 (also designated IL-29), IFN-λ2 (IL-28A), IFN-λ3 (IL-28B), and IFN-λ4.[7] These proteins share 15–19% amino acid sequence homology with type I interferons, despite their classification in a separate family due to distinct receptor usage and expression patterns.[23] The genes encoding IFN-λ1 through IFN-λ3 are tightly clustered on the long arm of human chromosome 19 (19q13.13), spanning a region of about 40 kb, while the IFN-λ4 gene lies upstream of the IFNL3 gene; each gene is organized into five exons, reflecting a conserved genomic architecture typical of the IL-10 family.[23] This clustering facilitates coordinated regulation during viral infections.A hallmark of type III interferons is their preferential production by epithelial cells lining mucosal barriers, such as those in the respiratory, gastrointestinal, and reproductive tracts, in response to viral pathogens like RNA viruses.[25] They exert their effects by binding to a unique heterodimeric receptor complex consisting of the IFN-λ-specific receptor subunit IFNLR1 (also known as IL-28Rα) and the shared IL-10 receptor β chain (IL10RB), which is distinct from the type I IFN receptor but activates similar Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathways.[7] This receptor binding induces an antiviral state in target cells, comparable to that of type I interferons, by upregulating interferon-stimulated genes that inhibit viral replication, though the effects are more localized due to restricted IFNLR1 expression primarily on epithelial and select immune cells like dendritic cells and neutrophils.[26] The epithelial-biased expression and limited receptor distribution of type III interferons minimize systemic inflammation, providing targeted protection at entry points of infection without the widespread immune activation associated with type I interferons.[25]Notably, genetic variations in the type III interferon locus influence disease outcomes; for instance, a dinucleotide frameshift variant (ss469415590) in the IFN-λ4 gene creates a functional protein but is associated with impaired spontaneous clearance of hepatitis C virus (HCV) infection, as individuals carrying the deleterious allele exhibit higher viral persistence rates in genome-wide association studies. This polymorphism highlights the clinical relevance of type III interferons in viral hepatitis, where IFN-λ4 expression can modulate the balance between antiviral efficacy and immune pathology. Overall, the barrier-specific properties of type III interferons position them as key sentinels for mucosal antiviral defense, complementing the broader actions of other interferon classes.
Production and Induction
Cellular Sources
Interferons are produced by a variety of cells across different tissues, with production often being inducible in response to stimuli, though many cell types also exhibit basal low-level expression to maintain homeostatic antiviral readiness.[27] This context-dependent expression allows interferons to contribute to both innate and adaptive immune responses, with specific cell types serving as primary producers for each interferon class.[28]For type I interferons, plasmacytoid dendritic cells (pDCs) represent the major source of IFN-α, capable of producing vast quantities upon viral encounter due to their specialized Toll-like receptor expression.[8] In contrast, IFN-β is predominantly secreted by fibroblasts, epithelial cells, and macrophages, which respond to a broader array of pathogens and provide localized antiviral protection.[29] These cells, including endothelial cells, demonstrate widespread potential for type I interferon production, underscoring the ubiquity of this response in non-immune tissues.[8]Type II interferon, IFN-γ, is primarily produced by adaptive immune cells such as CD4+ T helper type 1 (Th1) cells, CD8+ cytotoxic T cells, and natural killer (NK) cells, which release it during adaptive and innate immune activation.[30] Additionally, monocytes and macrophages can generate IFN-γ upon appropriate stimulation, contributing to inflammatory contexts where myeloid cells amplify immune signaling.[31]Group 1 innate lymphoid cells (ILC1s) also serve as innate sources, linking early defense to subsequent T cell responses.[32]Type III interferons, including IFN-λ, are chiefly produced by epithelial cells, particularly those lining mucosal barriers, where they enforce compartmentalized defense against enteric and respiratory pathogens.[25] In the liver, hepatocytes emerge as key producers of IFN-λ during viral hepatitis infections, such as hepatitis B and C, supporting organ-specific antiviral activity.[33] Myeloid cells, like dendritic cells and macrophages, can also contribute to IFN-λ production in response to viral stimuli, though epithelial sources predominate at barrier sites.[25]
Induction Pathways
Interferons are primarily induced in response to pathogen-associated molecular patterns (PAMPs) detected by pattern recognition receptors (PRRs) in innate immune cells. Viral infections trigger type I interferon production through endosomal and cytosolic sensors. Toll-like receptor 3 (TLR3), located in endosomes, recognizes double-stranded RNA (dsRNA) from viruses, recruiting the adaptor protein TRIF to activate both NF-κB and interferon regulatory factor 3 (IRF3), which translocate to the nucleus to drive interferon gene expression.[34] Cytoplasmic viral RNAs are sensed by RIG-I-like receptors (RLRs), including retinoic acid-inducible gene I (RIG-I) for short dsRNA with 5'-triphosphate ends and melanoma differentiation-associated protein 5 (MDA5) for longer dsRNAs; both interact with mitochondrial antiviral-signaling protein (MAVS) to phosphorylate and activate IRF3 and IRF7 via kinases such as TBK1 and IKKε.[35] For DNA viruses or bacteria releasing cytosolic DNA, cyclic GMP-AMP synthase (cGAS) detects it and synthesizes the second messenger cGAMP, which binds and activates stimulator of interferon genes (STING) on the endoplasmic reticulum, leading to TBK1-mediated phosphorylation of IRF3 and subsequent type I interferon transcription.[36]Bacterial and parasitic infections engage additional pathways beyond viral sensors. Cytosolic bacterial DNA, such as from Listeria monocytogenes or Mycobacterium tuberculosis, activates the STING pathway similarly to viral DNA, promoting IRF3-dependent type I interferon induction to enhance macrophage responses.[36] Nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs), including NOD1 and NOD2, recognize bacterial peptidoglycans in the cytosol; NOD2, for instance, cooperates with RLRs to amplify IFN-β production during bacterial invasion by potentiating MAVS signaling.[37] Parasitic triggers, such as protozoan DNA, also engage STING to induce type I interferons, contributing to control of infections like those caused by Toxoplasma gondii.[35]Type I interferons (IFN-α and IFN-β) are induced via promoters containing interferon-stimulated response elements (ISREs) and NF-κB binding sites, where phosphorylated IRF3 and IRF7 form homodimers or heterodimers with NF-κB to initiate transcription; IRF7 is particularly critical for IFN-α subtypes in plasmacytoid dendritic cells.[38] Type III interferons are induced through pathways similar to type I, via PRR recognition of viral PAMPs leading to IRF3/IRF7 activation and transcription from promoters with ISRE and NF-κB sites, predominantly in epithelial cells.[39] In contrast, type II interferon (IFN-γ) production is mainly driven by adaptive immune signals, with T-cell receptor (TCR) engagement in CD4+ and CD8+ T cells, combined with interleukin-12 (IL-12) stimulation from antigen-presenting cells, activating STAT4 and T-bet to promote IFN-γ gene expression in Th1 cells and natural killer cells.[40]Transcriptional regulation of interferon genes relies on the cooperative action of NF-κB, which drives proinflammatory genes alongside interferons, and IRFs, which specifically target interferon promoters; for example, the IFN-β enhanceosome assembles IRF3/7, NF-κB, and AP-1 on the promoter to ensure robust induction.[41]Negative feedback mechanisms, such as suppressors of cytokine signaling (SOCS) proteins—particularly SOCS1 induced by interferons themselves—inhibit Janus kinase (JAK)-STAT signaling upstream, limiting excessive interferon production to prevent immunopathology.[42]
Signal Transduction
Receptors and Binding
Type I interferons bind to a cell-surface receptor composed of the IFNAR1 and IFNAR2 subunits, forming a heterodimeric complex that facilitates high-affinity ligand engagement. IFNAR2 primarily mediates the initial high-affinity interaction with type I IFNs, exhibiting nanomolar binding affinity (Kd ≈ 0.1–100 nM depending on the IFN subtype), while IFNAR1 contributes lower-affinitybinding (Kd ≈ 1–5 μM for IFNα subtypes). The resulting ternary complex achieves an overall affinity in the picomolar range (Kd ≈ 10^{-10} M), enabling efficient signal initiation across diverse cell types.[43][44][45][46]Type II interferon (IFNγ) engages a receptor system comprising IFNGR1 and IFNGR2 subunits, which exist as preformed homodimers on the cell surface. Ligand binding by the dimeric IFNγ induces clustering of two IFNGR1 chains followed by recruitment of two IFNGR2 chains, assembling a 2:2:2 hexameric complex essential for signaling. This ligand-induced oligomerization enhances binding stability, with the IFNGR1-IFNγ interaction characterized by high affinity (Kd ≈ 10^{-9} to 10^{-10} M).[47][48]Type III interferons (IFNλs) signal through a heterodimeric receptor of IFNLR1 and IL10RB subunits, with expression predominantly restricted to epithelial cells and certain immune cells at barrier sites. IFNLR1 provides the ligand-specific binding site (Kd ≈ 10^{-7} to 10^{-8} M for IFNλ subtypes), while IL10RB serves as a shared low-affinity subunit common to IL-10 family cytokines. This epithelial-restricted distribution limits type III IFN responsiveness compared to the ubiquitous type I receptor.[49][50]Structural studies reveal conserved cytokine-receptor interfaces across interferon types, featuring helical bundles in the ligands that dock into fibronectin type III domains on receptor chains. For type I IFNs, key interactions involve helix A and F of the IFN with IFNAR2's D1 domain and IFNAR1's membrane-proximal regions, stabilized by hydrogen bonds and hydrophobic contacts at the binding interface. Similar architecture in type III complexes highlights IFNLR1's role in anchoring IFNλ via its N-terminal helices. Receptor ubiquitination, particularly of IFNAR1 via SCF^βTrCP E3 ligase, promotes lysosomal degradation and desensitization, preventing prolonged signaling after ligand binding.[51][52][53]Binding kinetics for interferons involve rapid association rates, such as 10^6–10^7 M^{-1} s^{-1} for type I IFNs to IFNAR, followed by quick dissociation modulated by subtype-specific affinities. Upon engagement, receptor-ligand complexes undergo swift clathrin-mediated endocytosis, internalizing the complex within minutes to regulate signal duration and localization. This endocytic trafficking is crucial for balancing acute responses and avoiding overstimulation.[43][54]
Downstream Signaling Cascades
Upon binding to their respective receptors, type I and type III interferons activate the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway through associated JAK1 and TYK2 kinases.[6] This leads to tyrosine phosphorylation of STAT1 and STAT2, which heterodimerize and associate with interferon regulatory factor 9 (IRF9) to form the ISGF3 complex.[55] The ISGF3 complex translocates to the nucleus and binds to interferon-stimulated response elements (ISRE) in the promoters of target genes, inducing their transcription.[56] In contrast, type II interferon (IFN-γ) engages JAK1 and JAK2 kinases, resulting in phosphorylation and homodimerization of STAT1, which then binds to gamma-activated sites (GAS) to drive gene expression.[6] These distinct STAT complexes ensure type-specific transcriptional programs while sharing core pathway components.[57]Beyond the canonical JAK-STAT axis, interferons activate parallel pathways that modulate cellular responses. The phosphoinositide 3-kinase (PI3K)/AKT pathway is engaged to promote cell survival and enhance translation of interferon-stimulated genes (ISGs) via mTOR/p70 S6 kinase activation.[58] Additionally, mitogen-activated protein kinase (MAPK) cascades, including p38 and ERK, are induced to regulate proliferation and contribute to antiproliferative effects.[6] These non-canonical signals integrate with JAK-STAT outputs to fine-tune interferon responses without altering the primary transcriptional machinery.[59]Interferon signaling culminates in the induction of approximately 300 ISGs, including exemplars like MxA (which inhibits viral nucleocapsid assembly), OAS (which activates RNase L for RNA degradation), and PKR (which phosphorylates eIF2α to halt protein synthesis).[60] These genes are primarily regulated through ISRE elements bound by ISGF3 for type I/III responses or GAS for type II.[56] Signal amplification occurs via autocrine and paracrine loops, where induced interferons upregulate IFNAR expression to sustain pathway activation and propagate signals to neighboring cells.[61] This feedback enhances the robustness of the antiviral and immunomodulatory state.[62]
Biological Functions
Antiviral Mechanisms
Interferons establish an antiviral state in infected and neighboring cells primarily through the induction of interferon-stimulated genes (ISGs), which encode effector proteins that target multiple stages of the viral replication cycle.[63] This state inhibits viral propagation by directly interfering with viral processes such as entry, replication, and protein synthesis, without relying on adaptive immune responses.[63]One key mechanism involves the inhibition of viral protein synthesis via the double-stranded RNA (dsRNA)-activated protein kinase R (PKR). Upon viralinfection, dsRNA produced during replication activates PKR, leading to its autophosphorylation and subsequent phosphorylation of the eukaryotic initiation factor 2 alpha (eIF2α) at serine 51. This phosphorylation inhibits the guanine nucleotide exchange factor eIF2B, preventing the recycling of eIF2 and thereby blocking the initiation of cap-dependent translation, which selectively impairs viral protein production while allowing some host translation to continue.[64] PKR's role was first elucidated in studies showing its interferon-inducible activation as a core component of the antiviral response.Another major pathway is the 2'-5'-oligoadenylate synthetase (OAS)-RNase L system, which targets viral RNA for degradation. Interferon-induced OAS enzymes detect viral dsRNA and synthesize 2'-5'-linked oligoadenylates (2-5A), which bind and activate the latent endoribonuclease RNase L. Activated RNase L then cleaves single-stranded viral and cellular RNAs at UU/UA dinucleotides, resulting in the degradation of viral genomes and the production of small RNA fragments that can further amplify interferon signaling.[65] This pathway's antiviral efficacy was demonstrated in early biochemical assays identifying OAS activation as a direct interferon effector.Specific ISGs also provide targeted antiviral actions. Mx GTPases, dynamin-like proteins induced by type I interferons, trap viral nucleocapsids and prevent their trafficking or assembly; for instance, human MxA inhibits influenza A virus by binding and sequestering viral ribonucleoproteins in the cytoplasm, blocking nuclear import essential for replication.[66] APOBEC3G, an interferon-upregulated cytidine deaminase, incorporates into retroviral particles and hypermutates the viral genome by deaminating cytosines to uracils in the single-stranded DNA intermediate during reverse transcription, introducing deleterious G-to-A mutations that inactivate the provirus.[67] Similarly, IFITM proteins, particularly IFITM3, restrict viral entry by altering endosomal membrane fluidity and cholesterol levels, thereby inhibiting fusion between viral and host membranes for enveloped viruses like influenza and dengue.[68]The establishment of this antiviral state is most effective when interferons are administered prior to or shortly after viral exposure, as pre-treatment allows full ISG expression and confers robust protection against subsequent infection, whereas concurrent treatment during active replication is less efficient due to partial pathway engagement.[69]
Immunomodulatory Roles
Interferons play pivotal roles in modulating both innate and adaptive immune responses by facilitating communication between immune cells and promoting their differentiation and activation. Type I interferons (IFN-α and IFN-β) and type II interferon (IFN-γ) enhance the maturation of dendritic cells (DCs), which are crucial antigen-presenting cells. Specifically, type I IFNs upregulate the expression of major histocompatibility complex (MHC) class I and II molecules on DCs, alongside costimulatory molecules such as CD80 and CD86, thereby improving antigen presentation to T cells without halting antigen processing.[70] This maturation process is essential for bridging innate and adaptive immunity, as demonstrated in studies showing that systemic type I IFN responses are required for DC maturation and subsequent CD4+ T cell immunity during infections.[71] Similarly, IFN-γ contributes to DC activation by enhancing MHC expression and promoting a pro-inflammatory phenotype that supports effective immune priming.[72]In T cell responses, interferons direct differentiation toward protective effector subsets. IFN-γ, the canonical type II interferon, is a key driver of Th1 cell differentiation from naive CD4+ T cells, promoting the production of pro-inflammatory cytokines like IL-2 and further IFN-γ to amplify cellular immunity against intracellular pathogens.[73] Type I IFNs complement this by enhancing the expansion and function of cytotoxic CD8+ T cells, increasing their survival and effector capabilities through STAT1-dependent signaling that boosts perforin and granzyme expression.[74] Additionally, type I IFNs suppress Th2 responses by reversing Th2 commitment in human CD4+ T cells and inhibiting IL-4-mediated differentiation, thereby preventing allergic or humoral-biased immunity in favor of cell-mediated responses.[75]Natural killer (NK) cells are activated by IFN-γ, which enhances their cytotoxicity and cytokine production in a feedback loop that sustains innate antiviral and antitumor defenses. IFN-γ promotes NK cell proliferation and IFN-γ secretion itself, creating an autocrine amplification that recruits and activates other immune effectors.[72] For B cells, type I IFNs influence class-switch recombination, particularly toward IgG subclasses like IgG2a and IgG3, by directly stimulating B cellactivation and indirectly via DC-mediated help, thereby enhancing humoral responses against viruses.[76] Type III interferons (IFN-λ) exhibit similar but more localized effects on B cell maturation in mucosal tissues.[77]Interferons engage in complex crosstalk with other cytokines to fine-tune immune networks. IFN-γ synergizes with IL-12, produced by DCs and macrophages, to robustly induce Th1 differentiation and NK cell IFN-γ production, amplifying protective responses against infections and tumors.[78] Conversely, interferons antagonize IL-10, an anti-inflammatorycytokine; for instance, IFN-γ suppresses IL-10 production in macrophages, preventing dampening of pro-inflammatory signals and sustaining effector functions.[79] Type I IFNs similarly counteract IL-10 by inhibiting its suppressive effects on DC maturation, ensuring sustained immune activation.[80] This balanced interplay underscores interferons' role in orchestrating adaptive immune polarization.
Antiproliferative Effects
Interferons of all types exert antiproliferative effects through shared and distinct mechanisms, primarily involving the JAK-STAT signaling pathway to induce cell cycle arrest, apoptosis, and suppression of tumor growth. These effects are mediated by the transcriptional activation of interferon-stimulated genes (ISGs) that regulate cell proliferation and survival.[81]Type I interferons (IFN-α and IFN-β), upon binding to the IFNAR receptor, activate JAK1 and TYK2, leading to phosphorylation of STAT1 and STAT2, which form the ISGF3 complex and drive ISG expression. A central mechanism is the STAT1-dependent upregulation of the cyclin-dependent kinase inhibitor p21 (CDKN1A), which induces G1/S phase arrest by inhibiting CDK2 and preventing retinoblastoma protein (Rb) phosphorylation, thereby halting cell cycle progression in tumor and proliferating cells.[6] Type I IFNs also promote apoptosis through pathways involving PKR activation, which phosphorylates eIF2α to inhibit protein synthesis, and upregulation of pro-apoptotic factors like TRAIL and Bax, contributing to their efficacy against malignancies such as leukemia and melanoma.[81]Type II interferon (IFN-γ) signals via the IFNGR receptor, primarily activating JAK1 and JAK2 to phosphorylate STAT1 homodimers that directly induce antiproliferative genes. IFN-γ similarly upregulates p21 and downregulates cyclins (e.g., cyclin D1), enforcing G1 arrest, and enhances apoptosis in synergy with TNF-α by amplifying Fas and TRAIL pathways. These effects are prominent in immune and epithelial cells, supporting antitumor immunity and restricting pathogen-induced proliferation.[81][6]Type III interferons (IFN-λ) exert antiproliferative effects primarily through activation of the JAK-STAT signaling pathway, leading to the transcriptional regulation of genes that control cell growth and survival. Upon binding to the IFN-λ receptor complex (IFNLR1/IL10RB), IFN-λ induces phosphorylation of STAT1 and STAT2, forming the ISGF3 complex that drives expression of interferon-stimulated genes (ISGs) involved in growth inhibition.[82] This mechanism shares similarities with type I IFNs (IFN-α/β), though IFN-λ often exhibits more sustained STAT activation and differential potency in epithelial and tumor cells.[82]A key antiproliferative pathway involves STAT1-mediated upregulation of the cyclin-dependent kinase inhibitor p21 (CDKN1A), which promotes G1 phase cell cycle arrest by inhibiting CDK2 activity and preventing retinoblastoma protein (Rb) phosphorylation.[83] IFN-λ also downregulates cyclins, such as cyclin D1 and E, further stalling the cell cycle progression in responsive cells like esophageal carcinoma lines and melanoma models.[84] These effects mirror the G1 arrest induced by IFN-α/β but are more pronounced in IFN-λ-treated keratinocytes, where prolonged signaling enhances growth suppression.[82]In addition to cell cycle control, IFN-λ induces apoptosis in tumor cells through upregulation of pro-apoptotic factors, including TRAIL and FAS ligands, activating caspase cascades such as caspase-3/7.[85] This pathway contributes to direct cytotoxicity, with increased Bax expression and p21 accumulation amplifying death signals in IFN-λ-sensitive malignancies.[86] Unlike IFN-γ, which synergizes with TNF-α to potentiate FAS/TRAIL-mediated apoptosis, IFN-λ's effects are more autonomous but can enhance overall antitumor responses in epithelial contexts.[6]IFN-λ plays a role in cellular senescence by reinforcing p21-mediated growth arrest, contributing to long-term suppression of proliferation in aging or stressed cells, though this is less dominant than its acute antiproliferative actions.[87] In normal cell homeostasis, these mechanisms maintain epithelial integrity by limiting unchecked division, particularly at barrier sites.[88]In tumor suppression, IFN-λ inhibits angiogenesis by inducing chemokines like IP-10 (CXCL10), which recruit effector T cells and block vascular endothelial growth factor (VEGF)-driven vessel formation in models of melanoma and lung cancer.[89] This indirect antiproliferative effect complements direct ISG-mediated growth control, enhancing tumor microenvironment remodeling without broad immune activation.[77]
Interactions with Pathogens
Host Resistance to Viruses
Interferons play a pivotal role in the systemic host response to viral infections, orchestrating both innate and adaptive immunity through distinct temporal phases. Type I interferons, including IFN-α and IFN-β, initiate an early innate wave that rapidly establishes an antiviral state in infected and neighboring cells, limiting viral replication and spread shortly after pathogen detection.00746-9) This phase is crucial for containing initial infection before adaptive responses mature. Subsequently, Type II interferon (IFN-γ), produced primarily by activated natural killer cells and T lymphocytes, sustains the response by enhancing adaptive immunity, promoting cytotoxic T cell differentiation and antibody production to facilitate viral clearance.[90] Together, these interferon types integrate innate control with long-term adaptive defenses, preventing dissemination and supporting resolution of acute infections.[15]Specific examples illustrate interferons' contributions to organ-specific viral resistance. In influenza A virus infections, IFN-λ restricts viral propagation in the upper airways by inducing localized antiviral effects in epithelial cells, thereby preventing progression to the lower respiratory tract and reducing transmission risk.[91] Similarly, in hepatitis B virus (HBV) infection, a robust interferon response correlates with viral control, whereas weak or impaired interferon signaling—characterized by diminished induction of interferon-stimulated genes (ISGs)—is associated with progression to chronicity, allowing persistent viral replication in hepatocytes.[92]Experimental models underscore interferons' essentiality in host survival against viruses. In interferon receptor knockout mice, infection with lymphocytic choriomeningitis virus (LCMV) leads to lethal immune-mediated pathology due to unchecked viral dissemination and dysregulated inflammation, highlighting the protective balance interferons impose on both antiviral defense and immune homeostasis.[93]Age and sex also modulate interferon efficacy; females generally exhibit stronger Type I interferon responses, conferring greater resistance to viral infections compared to males, while aging diminishes this response in both sexes, increasing susceptibility.[94] These differences arise from sex-specific immune regulation and age-related declines in interferon signaling efficiency.[95]Long-term immunity benefits from interferon-induced epigenetic modifications that imprint a "memory" on immune cells. In chronic viral infections, Type I interferons drive epigenetic changes in ISGs within memory B cells, enabling faster and more robust reactivation upon re-exposure, thus contributing to sustained protection without ongoing inflammation.00137-7) This mechanism ensures durable antiviral readiness, bridging acute responses to lifelong immunity.
Viral Countermeasures
Viruses have evolved diverse strategies to counteract the interferon (IFN) response, primarily by targeting its induction and signaling pathways to facilitate replication and dissemination. One prominent mechanism is the inhibition of IFN induction, where viral proteins interfere with pattern recognition receptors (PRRs) or downstream transcription factors. For instance, the nonstructural protein 1 (NSP1) of SARS-CoV prevents IFN production by blocking the phosphorylation and activation of interferon regulatory factor 3 (IRF3), a key transcription factor in the RIG-I/MDA5 pathway.00027-6/fulltext) Similarly, the NS1 protein of influenza A virus sequesters double-stranded RNA (dsRNA), a viral replication byproduct that would otherwise activate RIG-I and PKR, thereby suppressing IFN-β transcription and downstream antiviral gene expression.[96]Another critical evasion tactic involves blockade of IFN receptors and signaling cascades, preventing the transduction of antiviral signals into the host cell. Poxviruses such as vaccinia employ soluble decoy receptors like B18R, which bind type I IFNs with high affinity in the extracellular space, competing with cellular receptors and inhibiting JAK-STAT activation.[97] In HIV-1, the trans-activator of transcription (Tat) protein disrupts IFN-γ signaling by suppressing STAT1phosphorylation, thereby impairing the expression of IFN-stimulated genes (ISGs) essential for restricting viral spread.[98]Certain virus families exhibit specialized countermeasures that modulate the IFN environment more broadly. Herpesviruses, including Epstein-Barr virus and human cytomegalovirus, encode homologs of interleukin-10 (vIL-10), which suppress pro-inflammatory cytokine production and indirectly dampen IFN responses by promoting an anti-inflammatory milieu that favors viral persistence.[99] Picornaviruses, such as enterovirus 71, utilize their 2A protease to cleave mitochondrial antiviral-signaling protein (MAVS), disrupting the RIG-I signaling adaptor and abolishing type I IFN induction at the mitochondrial membrane.[100] RNA viruses often generate abundant immunostimulatory nucleic acids during replication, necessitating robust antagonism to evade rapid PRR detection.[101]This ongoing evolutionary arms race between viruses and host PRRs has driven the diversification of IFN evasion mechanisms, with viral proteins adapting to counter emerging host defenses while hosts evolve enhanced sensing capabilities.30295-0)
Response in Coronaviruses
In SARS-CoV-2 infections, the type I interferon (IFN) response is characteristically delayed, permitting extensive viral replication in the early phase and subsequently driving hyperinflammation through exaggerated proinflammatory cytokine production.[102][103] This temporal dysregulation contrasts with more robust early IFN responses seen in milder cases or other respiratory viruses, where prompt activation limits viral spread and mitigates tissue damage.[104] In parallel, type III IFN (IFN-λ) exerts a protective effect confined to the upper airways, inducing antiviral interferon-stimulated genes (ISGs) in epithelial cells to restrict viral dissemination without eliciting the systemic inflammation associated with type I IFN.[105][106] Elevated IFN-λ1 and IFN-λ3 levels in the nasopharynx correlate with asymptomatic or mild COVID-19 outcomes, underscoring their role in frontline mucosal defense.[107]Variant-specific differences in IFN induction further illustrate the nuanced host response to coronaviruses. The Omicron variant elicits a stronger type I and III IFN response than Delta, resulting in elevated ISG expression and reduced viral antagonism of innate signaling in primary nasal epithelial cells.[108][109] This heightened sensitivity contributes to Omicron's lower pathogenicity in the lower respiratory tract compared to earlier strains like Delta.[110] Genetic factors also modulate severity; inborn errors in the type I IFN pathway, including autosomal recessive or dominant deficiencies in TLR3-, IRF7-, or IRF9-mediated signaling, impair antiviral immunity and predispose individuals to life-threatening COVID-19pneumonia, even without prior severe infections.[111][112] Such monogenic defects account for up to 5% of critical cases, emphasizing the pathway's essentiality.[113]Clinical investigations into IFN therapeutics have yielded promising results for coronavirus management. Phase II trials conducted in 2023 evaluated inhaled IFN-β1a (SNG001) in adults with mild-to-moderate COVID-19, demonstrating safety and a non-statistically significant reduction in hospitalization risk, alongside trends toward accelerated viral clearance and symptom resolution.[114][115] These findings build on earlier evidence of IFN-β's antiviral potency against SARS-CoV-2, supporting its targeted delivery to the airways for enhanced local efficacy without broad immunosuppressive effects.[116]SARS-CoV-2 counters the IFN response through accessory proteins that disrupt signaling cascades. Notably, ORF6 binds to nucleoporins and karyopherin complexes to block STAT1 nuclear translocation, thereby inhibiting type I IFN gene expression and ISG induction.[117][118] Similarly, ORF9b localizes to the nucleolus and endoplasmic reticulum, where it interferes with STAT1 phosphorylation and translocation, enhancing viral evasion in infected cells.[119][120] These mechanisms enable persistent replication and are amplified in certain variants, linking them to observed delays in host immunity.[121]Persistent IFN dysregulation may underlie long COVID pathogenesis. Patients with ongoing post-acute sequelae exhibit chronically low serum levels of IFN-β and IFN-α, correlating with prolonged pulmonary complications and immune exhaustion.[122][123] This subdued response contrasts with acute infection dynamics and suggests a failure to fully resolve the antiviral state, potentially sustaining low-grade inflammation.[124]Emerging SARS-CoV-2 variants continue to evolve in IFN sensitivity, with progressive resistance observed across lineages of concern. Successive variants, including sublineages of Omicron, display diminished responsiveness to exogenous type I IFN in vitro, attributed to enhanced expression of antagonists like ORF6 and ORF9b, which may facilitate immune escape and prolonged shedding.[125][119] Despite this, some post-Omicron strains retain partial susceptibility, informing ongoing surveillance and therapeutic strategies up to 2025.[126]
Therapeutic Applications
Clinical Uses in Diseases
Interferons have established roles in treating various viral infections, particularly through type I interferons. Pegylated interferon-alpha (IFN-α), often combined with ribavirin, was a standard therapy for chronic hepatitis C virus (HCV) infection prior to the introduction of direct-acting antivirals (DAAs), which have largely replaced it since achieving sustained virologic response (SVR) rates exceeding 95% with improved tolerability; historically, it achieved SVR rates of approximately 45-50% in patients with genotype 1 HCV.[127][128] For chronic hepatitis B virus (HBV) infection, pegylated IFN-α-2a induces hepatitis B e-antigen (HBeAg) seroconversion in up to 36% of HBeAg-positive patients six months post-treatment.[129] Type I IFN-β is approved for relapsing-remitting multiple sclerosis (MS), where it reduces the annualized relapse rate by about 30% compared to placebo in large randomized trials.[130]In oncology, IFN-α serves as an adjuvant therapy for high-risk cutaneous melanoma, particularly in stage III disease, where high-dose regimens improve relapse-free survival without consistent overall survival benefits.[131] For hairy cell leukemia, IFN-α treatment yields overall response rates exceeding 80%, though complete remissions are achieved in fewer than 20% of cases, with durable responses in many patients.[132]Type II IFN-γ is indicated for chronic granulomatous disease (CGD), a primary immunodeficiency, where subcutaneous administration reduces the frequency and severity of serious infections by enhancing phagocyte function, as demonstrated in controlled trials showing significant prophylactic efficacy.[133] Emerging investigational applications for interferons include type III IFN-λ in autoimmune and infectious conditions. Recent 2024 clinical data support IFN-λ's role in COVID-19 management, with trials demonstrating its antiviral activity against SARS-CoV-2, including reduced viral loads and potential prophylactic benefits in high-risk populations when administered as pegylated formulations; as of 2025, pegylated IFN-λ remains investigational for COVID-19.[134] However, interferon therapies face limitations in HIV treatment due to poor long-term tolerance, including adverse effects like flu-like symptoms, mood disorders, and potential immune exhaustion, leading to limited efficacy and near-abandonment as a primary antiviral strategy.[135]
Formulations and Administration
Interferon therapies are primarily available as recombinant formulations, with key examples including IFN-α2a (Roferon-A) and IFN-α2b (Intron A), which are produced via bacterial expression systems for treating viral infections and malignancies.[136] Pegylated versions, such as IFN-α2a (Pegasys) and IFN-α2b (PegIntron), incorporate polyethylene glycol to extend half-life and enable less frequent dosing compared to non-pegylated forms.[136] For multiple sclerosis, IFN-β formulations include IFN-β1a (Avonex, administered intramuscularly) and IFN-β1b (Betaseron, administered subcutaneously).[137]Administration typically occurs via subcutaneous or intramuscular injection, with pegylated interferons given once weekly to maintain therapeutic levels.[138] For multiple sclerosis, dosing schedules vary: Avonex at 30 mcg intramuscularly once weekly, often titrated from 7.5 mcg to mitigate initial symptoms; Rebif (another IFN-β1a) at 44 mcg subcutaneously three times weekly after gradual escalation.[139] Inhalation via nebulization is used for respiratory viral infections, such as with IFN-β1a (SNG001) or IFN-α2b aerosol, delivering the agent directly to the lungs to enhance local antiviral effects while minimizing systemic exposure.[140]Common side effects include flu-like symptoms (fever, chills, myalgia) occurring shortly after injection, affecting up to 60% of patients initially, as well as depression and thyroid dysfunction, particularly with IFN-α therapies.[141][142] Management involves dose titration—starting at 20-25% of the full dose and increasing over weeks—to reduce symptom severity, alongside symptomatic treatments like analgesics or antidepressants.[143][144]By 2025, biosimilars for IFN-α2a and other interferons have entered markets, offering comparable efficacy at 20-30% lower costs through competitive pricing and streamlined approvals.[145] Emerging nanoparticle delivery systems, such as poly(lactic-glycolic acid) or chitosan-based carriers, are in preclinical and early clinical trials to enable sustained release, targeted lung or oral administration, and reduced toxicity by limiting systemic distribution.[146][147]
Historical Development
Discovery and Early Characterization
The discovery of interferon took place in 1957 at the National Institute for Medical Research in London, where Alick Isaacs and Jean Lindenmann investigated the phenomenon of viral interference using fragments of chick embryo chorioallantoic membrane.[148] They observed that cells exposed to heat-inactivated influenza virus became resistant to infection by live virus, a protection mediated by a diffusible, soluble factor released from the initially infected cells.[148] This factor induced an antiviral state in uninfected neighboring cells without directly neutralizing the virus, distinguishing it from antibodies or other known immune components.[148]Isaacs and Lindenmann named the substance "interferon" to reflect its role in interfering with viral replication.[148]In the early 1960s, research expanded to mammalian systems, with initial animal models demonstrating interferon's protective effects in vivo; for instance, administration of interferon preparations reduced mortality from encephalomyocarditis virus in mice and protected rabbits from vaccinia virus lesions. By 1965, E. Frederick Wheelock reported the production of an interferon-like antiviral protein in human leukocytes stimulated by the mitogen phytohemagglutinin, marking the first identification of what would later be classified as a distinct type. During this decade, interferons began to be classified based on their cellular sources and properties: leukocyte-derived interferon (later designated alpha) and fibroblast-derived interferon (later beta) were distinguished by differences in antigenicity and production stimuli, though formal typing awaited further characterization. Alick Isaacs died prematurely in 1967 at age 46.[9]The 1970s brought significant challenges in purifying interferon due to its low yields from natural sources—typically only 10^6 to 10^8 units per liter of cell culture supernatant or buffy coat from blood—necessitating processing of vast quantities of material for even small amounts of pure protein.[9] Efforts to isolate homogeneous interferon involved complex multi-step procedures like chromatography and electrophoresis, but instability and heterogeneity often resulted in substantial losses of activity. Despite these hurdles, partial purifications enabled early insights into its glycoprotein nature and species specificity, with human leukocyte interferon first purified to homogeneity in 1978. These advances set the stage for molecular studies, culminating in the cloning of interferon genes in the early 1980s.[9]
Recombinant Production and Advances
The advent of recombinant DNA technology in the late 1970s enabled the large-scale production of interferons, overcoming the limitations of extracting them from biological sources such as virus-infected cells. In 1980, scientists at Genentech successfully cloned and expressed the human leukocyte interferon alpha (IFN-α) gene in Escherichia coli, marking the first production of recombinant IFN-α in a bacterial system and yielding biologically active protein at levels sufficient for initial therapeutic evaluation.[149] This breakthrough, published in Nature, demonstrated that prokaryotic hosts could synthesize functional human cytokines, paving the way for industrial-scale manufacturing. Similarly, in the early 1980s, recombinant IFN-β was developed using Chinese hamster ovary (CHO) cells to produce a glycosylated form more akin to the native protein, addressing the need for proper post-translational modifications absent in bacterial expression systems.[150]These early recombinant efforts culminated in regulatory milestones, with the U.S. Food and Drug Administration (FDA) approving IFN-α2a (Roferon-A) and IFN-α2b (Intron A) in 1986 for the treatment of hairy cell leukemia, the first approvals for any recombinant interferon and highlighting their antiproliferative potential in oncology.[151] However, production challenges emerged, particularly with bacterial hosts like E. coli, where the lack of glycosylation led to reduced stability, shorter serum half-life, and increased immunogenicity due to the formation of neutralizing antibodies in patients.[152] Scale-up to biopharmaceutical levels also posed difficulties, requiring optimized fermentation processes to achieve high yields while minimizing inclusion body formation and endotoxin contamination, often necessitating refolding steps that complicated purification.[153]Advances in the 1990s focused on improving pharmacokinetics through chemical modification. Pegylation, the covalent attachment of polyethylene glycol (PEG) chains to IFN-α, was developed to extend circulating half-life from hours to days, reducing dosing frequency from daily to weekly injections and mitigating immunogenicity.[154] This innovation, first explored in preclinical models around 1990 and leading to FDA approvals for pegylated IFN-α2a (Pegasys) and IFN-α2b (Peg-Intron) in 2002 for hepatitis C, represented a key refinement in recombinant interferon formulations.[155]The 2000s saw the expansion of the interferon family with the discovery and recombinant development of type III interferons, known as IFN-λ (or lambda interferons). Identified in 2003 through genomic screening, IFN-λ genes were cloned and expressed in mammalian systems like CHO cells, revealing their unique receptor specificity on epithelial cells and potential for targeted antiviral therapy with reduced systemic side effects compared to type I IFNs.[156] Initial recombinant production emphasized glycosylation to ensure bioactivity, with early studies demonstrating efficacy against hepatitis C and influenza in preclinical models.[157]Recent progress from 2023 to 2025 has incorporated gene therapy approaches to achieve sustained interferon expression, minimizing repeated dosing and immunogenicity risks associated with exogenous proteins. Adenoviral vectors, such as nadofaragene firadenovec (Adstiladrin), which delivers the IFN-α2b gene for intravesical expression in bladder cancer, entered expanded clinical trials evaluating long-term safety and efficacy, with FDA approval of additional manufacturing facilities in 2025 supporting broader access.[158] In September 2025, Japan's Pharmaceuticals and Medical Devices Agency (PMDA) accepted a New Drug Application (NDA) filing for nadofaragene firadenovec for high-risk BCG-unresponsive non-muscle invasive bladder cancer.[159] In October 2025, Ferring Pharmaceuticals presented new real-world research on its clinical use, including three studies at a medical conference.[160] Ongoing phase II/III trials for similar vectors in viral infections and cancers aim to optimize transgene integration for durable, localized IFN production, building on vector engineering to enhance transduction efficiency and reduce off-target effects.[161] These developments underscore a shift toward integrated biomanufacturing strategies, combining recombinant expression with advanced delivery systems for more precise therapeutic modulation.
Interferons in Specific Organisms
Human Interferons
Human interferons are encoded by a multigene family clustered primarily on specific chromosomes. The type I interferon genes, including 13 IFN-α genes (12 of which are functional and encode distinct subtypes), one IFN-β gene (IFNB1), one IFN-ε gene (IFNE1), one IFN-κ gene (IFNK), and one IFN-ω gene (IFNW1), are located in a cluster spanning approximately 400 kb on chromosome 9p21.3.[162] The type II interferon, IFN-γ (IFNG), is encoded by a single gene on chromosome 12q15.[163] Type III interferons, comprising IFN-λ1 (IFNL1), IFN-λ2 (IFNL2), IFN-λ3 (IFNL3), and IFN-λ4 (IFNL4, which functions as a pseudogene in many individuals but produces a protein in others), are clustered on chromosome 19q13.2 within a 55 kb region.[164] This genomic organization reflects evolutionary duplication events, with the type I cluster containing at least nine pseudogenes, including several IFN-α pseudogenes such as IFNA1P, IFNA11P, and IFNA12P, which contribute to the complexity of interferon regulation without producing functional proteins.[13]Genetic variations in human interferon genes influence antiviral responses and disease susceptibility. A prominent polymorphism, rs12979860 in the IFN-λ3 gene (IFNL3), significantly affects spontaneous clearance and treatment response in hepatitis C virus (HCV) infection, with the CC genotype conferring a favorable outcome by enhancing interferon signaling efficiency.[165]Sex differences are evident in IFN-α expression signatures, where females exhibit stronger type I interferon responses upon stimulation, driven by higher TLR7 expression on plasmacytoid dendritic cells and X-chromosome-linked factors, contributing to observed sex biases in autoimmune conditions.[166]Recent single-cell RNA sequencing (scRNA-seq) analyses have mapped interferon expression across human tissues, revealing cell-type-specific patterns that highlight their role in immune homeostasis. For instance, IFN-α and IFN-β are predominantly expressed in immune cells like plasmacytoid dendritic cells and monocytes in lymphoid tissues, while IFN-ε shows constitutive expression in epithelial cells of reproductive tissues; these insights from multi-organ atlases underscore dynamic regulation during homeostasis and infection.[167] Non-coding RNAs further modulate interferon gene expression, with long non-coding RNAs such as LUCAT1 acting as negative feedback regulators to dampen type I interferon responses and prevent excessive inflammation.[168]Human interferon genes bear signatures of evolutionary adaptation, including archaic introgression from Neanderthals at loci like OAS1-OAS3, which encode interferon-stimulated genes enhancing antiviral activity against RNA viruses.[169] Genome-wide association studies (GWAS) updated in 2025 have linked interferon pathway variants to autoimmune diseases, such as systemic lupus erythematosus (SLE), where trans-effects on interferon signaling genes like USP18 account for up to 9% of genetic risk, emphasizing their dual role in immunity and pathology.[170]
Teleost Fish Interferons
Teleost fish, comprising the majority of extant fish species, possess a diverse interferon (IFN) system that is integral to their innate antiviral immunity, differing significantly from that in higher vertebrates due to evolutionary adaptations such as whole-genome duplications.[171] Type I IFNs are the primary antiviral cytokines in teleosts, classified into three main groups based on structural features and phylogenetic analysis: group I (subgroups α, δ, ε, η), group II (subgroups β, γ, ζ, θ), and a more recently identified group III.[172] Group I IFNs, characterized by four conserved cysteine residues forming two disulfide bonds, are present across all teleostspecies examined, while group II IFNs, with two cysteines forming a single disulfide bond, are restricted to specific lineages such as cyprinids (e.g., zebrafish, carp), salmonids (e.g., Atlantic salmon, rainbow trout), and some perciforms.[173] This structural diversity enables fish type I IFNs to bind distinct receptor complexes, enhancing their responsiveness to a broad spectrum of aquatic viruses.[174]Structurally, teleost type I IFNs belong to the class II α-helical cytokine family, featuring a characteristic four-helix bundle, but they exhibit greater sequence variability and genomic multiplicity compared to mammalian counterparts.[175] Phylogenetic studies indicate that fish type I IFNs form a distinct clade separate from avian and mammalian type I IFNs, reflecting an ancient divergence before the teleost-specific genome duplication events that amplified IFN gene numbers—up to 17 in salmonids like rainbow trout.[175] For instance, in zebrafish (Danio rerio), multiple IFN genes (e.g., ifnphi1 to ifnphi4) encode proteins with varying abilities to induce antiviral states, underscoring the system's adaptability to diverse viral threats in aquatic environments.[176] Type II IFNs, represented by IFN-γ, are single-chain molecules that promote Th1-like responses and macrophage activation, while type III IFNs (IFN-λ) provide mucosal immunity, though less characterized in fish; a novel type IV IFN, identified in 2022, functions similarly to type I but signals through unique cytokine receptor family B (CRFB) members.[177][178]Receptors for teleost IFNs are encoded by the CRFB gene family, which expanded in fish to include at least 17 members, allowing for specialized signaling.[177] Type I group I IFNs primarily bind CRFB1/5/99 complexes, activating the JAK-STAT pathway to induce interferon-stimulated genes (ISGs) like mx, vig1, and isg15, which establish an antiviral state.[172] In contrast, group II IFNs interact with CRFB2/5/99 or CRFB6/17/99 pairs, eliciting potent antiviral activity against RNA viruses such as viral hemorrhagic septicemia virus (VHSV) in rainbow trout.[173] Experimental overexpression of these IFNs in cell lines, such as epithelioma papulosum cyprini (EPC) cells, demonstrates dose-dependent protection against fish rhabdoviruses, with group II IFNs often showing higher potency due to their streamlined structure.[171]The functional role of teleost IFNs extends beyond antiviral defense to immunomodulation, with type I IFNs upregulating MHC class I expression and enhancing NK cell-like activity in species like ginbuna crucian carp.[179] Seminal studies, including the initial cloning of fish IFN genes in the early 2000s, revealed that poly I:C induction triggers IFN production via RIG-I-like receptors (RLRs), mirroring mammalian pathways but with greater reliance on LGP2 in fish.[175]In vivo, recombinant IFN treatments in salmonids have reduced mortality from infectious salmon anemia virus by over 80%, highlighting therapeutic potential in aquaculture.[180] However, the extraordinary diversity of fish type I IFNs—likely an evolutionary counter to sophisticated viral evasion tactics in water—also poses challenges, as some viruses like infectious hematopoietic necrosis virus (IHNV) can suppress IFN signaling via non-structural proteins.[176]Recent advances, such as crystal structure analyses of type I IFNα from large yellow croaker (Larimichthys crocea), confirm the conserved helical fold but reveal unique loop regions that modulate receptor affinity, providing insights into ligand specificity absent in mammals.[181] These findings, building on earlier work like the 2005 phylogenetic classification, emphasize the teleost IFN system's role as a model for understanding cytokineevolution and antiviral strategies in poikilothermic vertebrates.[175]