null
The role of IFN gamma in inflammation, cancer and autoimmune disease

The role of IFN gamma in inflammation, cancer and autoimmune disease

Interferons are key cytokines in the immune system, crucial for combating viral and bacterial infections, with significant roles in both innate and adaptive immunity responses.

Key Takeaways

  1. Interferons (IFNs) are crucial cytokines in immune responses against viral and bacterial challenges.
  2. There are two main types: Type I (IFN-alpha and IFN-beta) and Type II (IFN-gamma), each with unique roles and receptor interactions.
  3. IFN-gamma, a Type II interferon, is pivotal in innate and adaptive immunity, produced mainly by T cells and NK cells.

What are Interferons?

Interferons (IFN) are a family of cytokines which, upon secretion, play a central role in mediating the innate and adaptive immune response to viral and bacterial challenges. Isaacs and Lindenmann first discovered a molecule they called interferon in the 1950s, which is now referred to as IFN gamma (Isaacs and Lindenmann, 1957). Following several more studies there are now two IFN family members identified; Type I IFN, which include IFNa and IFNb, and Type II IFN which is comprised of IFN gamma (van de Broek et al, 1995), both of which have distinct physiological roles, bind to different receptors, and are structural diversity; however, both Type I and Type II IFNs activate the anti-viral response. Moreover, Type I IFNs are secreted following the activation of intracellular and extracellular anti-viral sensors, mainly by macrophages and dendritic cells (Sadler and Williams, 2008); whereas Type II IFN is primarily secreted by T lymphocytes and natural killer (NK) cells in response to cytokine activation, specifically IL-12 or IL-18 (Schroder et al, 2004), with stimulation from both IFN subtypes leading to downstream cytosolic signalling and subsequent upregulation of gene expression.

Interferon Gamma

Interferon gamma, a type II interferon, is distinct in its role and effects compared to other interferons. It is mainly produced by certain immune cells, such as T cells and natural killer cells, and is a key regulator of both innate and adaptive immunity.

(a)

(b)

Fig 1 (a) IFN - gamma production by Effector T cell to induce cancer cell apoptosis. Fig 1(b) IFN-gamma production by cytotoxic T cells to induce apoptosis of infectedcells.  

IFN Gamma Function

IFN-γ exerts its influence by stimulating macrophages to enhance their antimicrobial activity and promote the destruction of infected cells. Additionally, IFN-γ contributes to the differentiation of T cells into specific subtypes (Th1 effector T Cells) that are crucial for orchestrating immune responses tailored to different types of pathogens. Importantly, both Type I and Type II IFNs were demonstrated to be central mediators of vaccine-induced responses, specifically CD4+ T cell (Th1) responses (Weir et al, 2008; Tudor et al, 2001).

One of the remarkable features of interferons, including IFN-γ, is their ability to induce an "antiviral state" in neighboring cells. This means that cells exposed to interferons become more resistant to viral replication, effectively limiting the spread of the infection. This antiviral state is achieved through the upregulation of various genes involved in inhibiting viral entry, replication, and assembly. .

Fig 2 IFN - gamma production by NK cells helps drive differentiations of naive T cells to Th1 effector cells.

IFN Gamma Pathway

IFN gamma binds to the extracellular domain of the IFN gamma R1, leading to the engagement of the IFN gamma R2 which induces signalling intracellularly. The tyrosine kinases Janus Kinase 1 (JAK1) and JAK2 are phosphorylated at the membrane upon association with activated IFN gamma R2, facilitating the binding of signal transducer and activator of transcription 1 (STAT1). Phosphorylation of STAT1 precedes its nuclear translocation and binding to the gamma-activated sequence (GAS), which are short DNA elements in the nucleus which bridge STAT binding and the activation of the transcription factor interferon response factor I (IFN1) (Varinou et al, 2003; Decker et al, 1997; Coccia et al, 1995). A large range of genes have been described to be transcribed in an IFN gamma-dependent manner, including many involved in haematopoiesis, inflammation, cell proliferation, cell differentiation and programmed cell death (Boehm et al, 1997). The IFN gamma pathway is negatively regulated by suppressor of cytokine singalling molecule 1 (SOCS1) (Alexander et al, 1999), which inhibit JAK2 and STAT1 interactions, and also by the dephosphorylation of STAT1 by the phosphatase TCP45 (Kramer et al, 2009). Notable, an alternative, or non-canonical, IFN gamma signaling pathway has been described, implicating n interaction between JAK and MyD88 adaptor-like molecule (Mal), commonly associated with TLR signaling (Ní Cheallaigh et al, 2016).

Figure 1: IFN gamma signalling through the JAK/STAT pathway. Extracellular secreted IFN gamma binds directly to the extracellular domain of the membrane-bound IFN gammaR1, which in turn transduces signalling to the IFN gammaR2 subunit. The carboxy termini of both IFN gammaR1 and IFN gammaR2 associate with JAK1 and JAK2 which are in turn phosphorylated. Janus kinase 1 (JAK1) and JAK2 subsequently phosphorylate STAT1 which induces its nuclear translocation, association with gamma-activated sequence (GAS) which mediate IRF1 activation and transcription of multiple genes governing cell proliferation, differentiation, heamatopoiesis and inflammation. Suppressor of Cytokines 1 (SOCS1) negatively regulates this pathway by inhibiting JAK/STAT1 interactions.

IFN Gamma - Clinical Significance

Genetic deficiency in the IL-12/IL-23/IFN gamma pathway leads to a robust susceptibility to mycobacterial infections (Filipe-Santos et al, 2006). The primary sources of IFN gamma are NK, NKT cells, macrophages and dendritic cells, which mediate the innate immune response, and both CD4+ and CD8+ T cells, which facilitate adaptive immunity.

IFN Gamma and Autoimmune Disease

Although IFN gamma can mediate clearance of pathogenic insults, chronic exposure to IFN gamma has been identified to be involved in several non-infectious pathologies, such as autoimmune diseases, for example rheumatoid arthritis (Nielan et al, 2004), and systemic lupus erythematous (Lu et al, 2016). In the case of autoimmune disease, IFN gamma effectively ‘primes’ and sensitizes cells to secondary ligands, such as TLR agonists and TNFa (Borges da Silva et al, 2015). Interestingly, crosstalk between NF-KB and IRF pathways has been reported (Iwanazsko and Kimmel, 2015), with the discovery of IFN gamma mediated IRF1 transcription enhancing the activation of NF-kappaB in response to human immunodeficiency virus (HIV) (Sgarbanti et al, 2008).

IFN Gamma in Immunosurveillance and Cancer

The term immunosurveillance describes the translocation of several immune cells, including T cells, NK cells, NKT cells, and macrophages into the tumour in an effort to attack the foreign cancer cells, eliciting a robust secretion of cytotoxic factors, such as IFN gamma, TNFα, FasL and TNF-related apoptosis inducing ligand (TRAIL) (Dunn et al, 2004). Although tumour immunology is a complex and constantly expanding field, current research demonstrates that cells have developed a way to evade immunosurveillance by secreting immunosuppressive molecules and reducing the cytotoxic T lymphocyte response. One mechanism cancer cells employ in order to evade the immune response is to downregulate IFN gamma-expressing cells and to limit IFN gamma secretion. IFN gamma negatively regulates tumourigenesis by suppressing tumour development by negatively regulating cell proliferation (Kominsky et al, 1998), or promoting apoptosis (Kim et al, 2002). Moreover, IFN gamma-induced IRF1 activation has been shown to synergise with NF-kappaB to upregulate the MHC-I expression on cytotoxic T cells in neuroblastoma, eliciting a protective response (Lorenzi et al, 2012). However, conversely IFN gamma has also been shown to induce tumour survival in several studies. IFN gamma is demonstrated to promote proliferation in a human melanoma cell line in vitro (Garbe et al 1990), and IFN gamma-producing macrophages have been identified in melanoma tumours (Zaidi et al, 2011).In addition to the cell-specific polarized effects of IFN gamma as a therapeutic, the potentially fatal side effect of immunochterapy termed Cytokine Release Syndrome (CRS) has been described, with IFN gamma and IL-6 rescuing mice from anti-CD3 induced CRS (Matthys et al, 1993). However, in clinical trials using anti-CD19 Chimeric Antigen Receptor (CAR) T cells to treat Acute Lymphoblastic Laukaemia, elevated levels of IFN gamma are indicative of later development of fatal CRS (Teachey et al, 2016).

IFN Gamma as a therapeutic

Overall, although there has been some success for IFNa in cancer treatment (Parker et al, 2016; Eto et al, 2015), IFN gamma has been shown to facilitate immunomodulation, and promote both pro-tumourigenic and anti-cancer properties, mainly though cell proliferation and apoptotic pathways. These opposing responses may be dose-dependent based on the intensity of the signal, and cancer type specific, dependent on a range of interacting factors in the tumour microenvironment. However, the therapeutic potential of IFN gamma may prove a worthy success in a subset of cancers. By bolstering the immune response against cancer cells, IFN-γ might help in slowing down tumor growth and increasing the effectiveness of other treatment modalities, such as checkpoint inhibitors.

Research is ongoing to develop novel therapeutic strategies centered around IFN-γ. This includes exploring targeted delivery methods to minimize potential side effects associated with systemic administration. Nanoparticles and liposomes are being investigated as carriers to deliver IFN-γ directly to the affected tissues, maximizing its therapeutic impact while minimizing off-target effects.

While IFN-γ's therapeutic potential is promising, there are challenges to address. Side effects, such as flu-like symptoms, fatigue, and potential immune system imbalances, need to be carefully managed. Additionally, finding the right dosage and timing of IFN-γ administration is crucial to achieve the desired therapeutic effects without triggering excessive immune reactions.

References

AlexanderWS,Starr R,Fenner JE,Scott CL,Handman E,Sprigg NS,Corbin JE,Cornish AL,Darwiche R,Owczarek CM,Kay TW,Nicola NA,Hertzog PJ,Metcalf D,Hilton DJ. SOCS1is a critical inhibitor of interferon gamma signaling and prevents the potentially fatal neonatal actions of this cytokine.Cell.1999. 98(5):597-608.

BoehmU,Klamp T,Groot M,Howard JC. Cellular responses to interferon-gamma.Annu RevImmunol.1997;15:749-95.

Borges da Silva H, Fonseca R, Alvarez JM, D'Império Lima MR. IFN-γ Priming Effects on the Maintenance of Effector Memory CD4(+) T Cells and on Phagocyte Function: Evidences from Infectious Diseases. J Immunol Res.2015. 202816

Coccia EM, Marziali G,Stellacci E,Perrotti E,Ilari R,Orsatti R,Battistini A. Cells resistant to interferon-beta respond to interferon-gamma via the Stat1-IRF-1 pathway.Virology.1995. 211(1):113-22.

Decker T,Kovarik P,Meinke A. GAS elements: a few nucleotides with a major impact on cytokine-induced gene expression.J Interferon Cytokine Res.1997. 17(3):121-34.

Dunn GP,Old LJ,Schreiber RD. Theimmunobiologyofcancerimmunosurveillanceand immunoediting.Immunity.2004. 21(2):137-48.

Eto M,Kawano Y,Hirao Y,Mita K,Arai Y,Tsukamoto T,Hashine K,Matsubara A,Fujioka T,Kimura G,Shinohara N,Tatsugami K,Hinotsu S,Naito S. PhaseIIclinicaltrialofsorafenibplusinterferonalphatreatmentforpatientswithmetastaticrenal cellcarcinomainJapan. BMC Cancer.2015. 15:667.

Filipe-Santos O,Bustamante J,Chapgier A,Vogt G,de Beaucoudrey L,Feinberg J,Jouanguy E,Boisson-Dupuis S,Fieschi C,Picard C,Casanova JL. Inborn errors of IL-12/23- and IFN-gamma-mediated immunity: molecular, cellular, and clinical features. Semin Immunol.2006. 18(6):347-61.

GarbeC,Krasagakis K,Zouboulis CC,Schröder K,Krüger S,Stadler R,Orfanos CE. Antitumor activities of interferon alpha, beta, and gamma and their combinations on human melanoma cells in vitro: changes of proliferation, melanin synthesis, and immunophenotype.

Isaacs A,Lindenmann J. Virus interference. I. The interferon.Proc R Soc Lond B Biol Sci.1957. 147(927):258-67.

Iwanaszko M,Kimmel M. NF-κB andIRFpathways:cross-regulationontargetgenespromoterlevel.BMC Genomics.2015. 16:307.

J Invest Dermatol.1990. 95(6 Suppl):231S-237S.

KimKB,Choi YH,KimIK,Chung CW,KimBJ,Park YM,Jung YK. Potentiation of Fas- and TRAIL-mediated apoptosis by IFN-gamma in A549 lung epithelial cells: enhancement of caspase-8 expression through IFN-response element.Cytokine.2002. 20(6):283-8.

Kominsky S, Johnson HM, Bryan G, Tanabe T, Hobeika AC, Subramaniam PS, Torres B. IFNgamma inhibition of cell growth in glioblastomas correlates with increased levels of the cyclin dependent kinase inhibitor p21WAF1/CIP1. Oncogene.1998. 17(23):2973-9.

Krämer OH,Knauer SK,Greiner G,Jandt E,Reichardt S,Gührs KH,Stauber RH,Böhmer FD,Heinzel T. A phosphorylation-acetylation switch regulates STAT1 signaling.Genes Dev.2009. 23(2):223-35.

Lorenzi S, Forloni M, Cifaldi L, Antonucci C, Citti A, Boldrini R, Pezzullo M, Castellano A, Russo V, van der Bruggen P, Giacomini P, Locatelli F, Fruci D. IRF1 and NF-kB restore MHC class I-restricted tumor antigen processing and presentation to cytotoxic T cells in aggressive neuroblastoma. PLoS One.2012. 7(10):e46928.

LuR,Munroe ME,Guthridge JM,Bean KM,Fife DA,Chen H,Slight-Webb SR,Keith MP,Harley JB,James JA. Dysregulation of innate and adaptive serum mediators precedes systemic lupus erythematosus classification and improves prognostic accuracy of autoantibodies. J Autoimmun.2016. 74:182-193.

Matthys P, Dillen C, Proost P, Heremans H, Van Damme J, Billiau A. Modification of the anti-CD3-induced cytokine release syndrome by anti-interferon-gamma or anti-interleukin-6 antibody treatment: protective effects and biphasic changes in blood cytokine levels. Eur J Immunol.1993. 23(9):2209-16.

Ní Cheallaigh C,Sheedy FJ,Harris J,Muñoz-Wolf N,Lee J,West K,McDermott EP,Smyth A,Gleeson LE,Coleman M,Martinez N,Hearnden CH,Tynan GA,Carroll EC,Jones SA,Corr SC,Bernard NJ,Hughes MM,Corcoran SE,O'Sullivan M,Fallon CM,Kornfeld H,Golenbock D,Gordon SV,O'Neill LA,Lavelle EC,Keane J. A Common Variant in the Adaptor Mal Regulates Interferon Gamma Signaling.Immunity.2016. 44(2):368-79.

Nielen MM, van Schaardenburg D, Reesink HW, van de Stadt RJ, van der Horst-Bruinsma IE, de Koning MH, Habibuw MR, Vandenbroucke JP, Dijkmans BA. Specific autoantibodies precede the symptoms of rheumatoid arthritis: a study of serial measurements in blood donors. Arthritis Rheum.2004. 50(2):380-6.

Parker BS, Rautela J, Hertzog PJ. Antitumouractionsofinterferons:implicationsforcancertherapy. Nat RevCancer.2016. 16(3):131-44.

Sadler AJ,Williams BR. Interferon-inducible antiviral effectors.Nat Rev Immunol.2008. 8(7):559-68.

Schoenborn JR, Wilson CB. Regulation of interferon-gamma during innate and adaptive immune responses. Adv Immunol. 2007. 96:41-101.

Schroder K,Hertzog PJ,Ravasi T,Hume DA. Interferon-gamma: an overview of signals, mechanisms and functions.J Leukoc Biol.2004. 75(2):163-89.

Sgarbanti M, Remoli AL, Marsili G, Ridolfi B, Borsetti A, Perrotti E, Orsatti R, Ilari R, Sernicola L, Stellacci E, Ensoli B, Battistini A. IRF-1 is required for full NF-kappaB transcriptional activity at the human immunodeficiency virustype 1 long terminal repeat enhancer. J Virol.2008. 82(7):3632-41.

Tudor D, Riffault S, Carrat C, Lefèvre F, Bernoin M, Charley B. Type I IFN modulates the immune response induced by DNA vaccination to pseudorabies virus glycoprotein C.

van den Broek MF, Müller U, Huang S, Zinkernagel RM, Aguet M. Immune defence in mice lacking type I and/or type II interferon receptors. Immunol Rev. 1995. 148:5-18.

Varinou L,Ramsauer K,Karaghiosoff M,Kolbe T,Pfeffer K,Müller M,Decker T. Phosphorylation of the Stat1 transactivation domain is required for full-fledged IFN-gamma-dependent innate immunity.Immunity.2003 Dec;19(6):793-802.

Virology.2001 Jul 20;286(1):197-205.

Weir RE, Fine PE, Floyd S, Stenson S, Stanley C, Branson K, Britton WJ, Huygen K, Singh M, Black G, Dockrell HM. Comparison of IFN-gamma responses to mycobacterial antigens as markers of response to BCG vaccination. Tuberculosis (Edinb).2008. 88(1):31-8.

ZaidiMR, Davis S,Noonan FP,Graff-Cherry C,Hawley TS,Walker RL,Feigenbaum L,Fuchs E,Lyakh L,Young HA,Hornyak TJ,Arnheiter H,Trinchieri G,Meltzer PS,De Fabo EC,Merlino G. Interferon-γ links ultraviolet radiation to melanomagenesis in mice.Nature.2011. 469(7331):548-53.



Additional Resources


22nd Aug 2023 Sinéad Kinsella PhD

Recent Posts