دورية أكاديمية

Analysis of interferon type I signature for differential diagnosis of diseases of the immune system ( review of literature).

التفاصيل البيبلوغرافية
العنوان: Analysis of interferon type I signature for differential diagnosis of diseases of the immune system ( review of literature).
عنوان ترانسليتريتد: Анализ профиля экспрессии интерферонзависимых генов для дифференциальной диагностики заболеваний иммунной системы (обзор литературы).
المؤلفون: Suspitsin EN; St.-Petersburg State Pediatric Medical University.; N.N. Petrov Institute of Oncology., Raupov RK; St.-Petersburg State Pediatric Medical University., Kuchinskaya EM; Almazov National Medical Research Centre., Kostik MM; St.-Petersburg State Pediatric Medical University.; Almazov National Medical Research Centre.
المصدر: Klinicheskaia laboratornaia diagnostika [Klin Lab Diagn] 2021 May 23; Vol. 66 (5), pp. 279-284.
نوع المنشور: Journal Article; Review
اللغة: English
بيانات الدورية: Publisher: Meditsina Country of Publication: Russia (Federation) NLM ID: 9432021 Publication Model: Print Cited Medium: Print ISSN: 0869-2084 (Print) Linking ISSN: 08692084 NLM ISO Abbreviation: Klin Lab Diagn Subsets: MEDLINE
أسماء مطبوعة: Original Publication: Moskva : "Meditsina", 1992-
مواضيع طبية MeSH: Interferon Type I*/genetics , Lupus Erythematosus, Systemic*/diagnosis , Lupus Erythematosus, Systemic*/genetics, Diagnosis, Differential ; Humans ; Immune System
مستخلص: Type 1 interferons (IFN1) are both key molecules of antiviral defense and potent inflammatory mediators. In 2003, increased expression of a variety of interferon 1-regulated genes was observed in a blood cells of patients with systemic lupus erythematosus (SLE). This phenomenon was called the type 1 interferon signature (IFN1-signature). Since then, expression patterns indicating the presence of an IFN1-signature were consistently detected in a range of monogenic and complex autoimmune and autoinflammatory conditions. A quantitative indicator reflecting the degree of hyperactivation of the IFN1 pathway is known as interferon score. This review discusses the possible causes of upregulated expression of interferon 1-induced genes, the laboratory approaches to the interferon score analysis, as well as the practical use of this indicator for the diagnosis of various conditions.
Competing Interests: The authors declare no conflict of interest.
References: Ivashkiv L.B. IFNγ: signalling, epigenetics and roles in immunity, metabolism, disease and cancer immunotherapy. Nat. Rev. Immunol. 2018; 18:545–58.
Rizza P., Moretti F., Belardelli F. Recent advances on the immunomodulatory effects of IFN-α: Implications for cancer immunotherapy and autoimmunity. Autoimmunity. 2010; 43(3):204–9.
Barrat F.J., Crow M.K., Ivashkiv L.B. Interferon target-gene expression and epigenomic signatures in health and disease. Nat. Immunol. 2019; 20:1574–83.
Hertzog P., Forster S., Samarajiwa S. Systems biology of interferon responses. J. Interferon Cytokine Res. 2011; 31(1):5-11.
Forster S. Interferon signatures in immune disorders and disease. Immunol. Cell Biol. 2012; 90:520–27.
Kavanagh D., McGlasson S., Jury A., Williams J., Scolding N., Bellamy C. et al. Type I interferon causes thrombotic microangiopathy by a dose-dependent toxic effect on the microvasculature. Blood. 2016; 128(24):2824–33.
Nasonov E.L., Avdeeva A.S. Immunoinflammatory rheumatic diseases associated with type I interferon:new evidence. Nauchno-prakticheskaya revmatologiya. 2019;57(4):452-61. (in Russian).
Demirkaya E., Sahin S., Romano M., Zhou Q., Aksentijevich I. New Horizons in the Genetic Etiology of Systemic Lupus Erythematosus and Lupus-Like Disease: Monogenic Lupus and Beyond. J. Clin. Med. 2020; 9(3):712.
Rönnblom L. The type i interferon system in the etiopathogenesis of autoimmune diseases. Ups. J. Med. Sci. 2011; 18:227–37.
Hakkim A., Fürnrohr B.G., Amann K., Laube B., Abed U.A., Brinkmann V. et al. Impairment of neutrophil extracellular trap degradation is associated with lupus nephritis. Proc. Natl. Acad. Sci. USA. 2010; 107(21):9813–18.
Hagberg N., Rönnblom L. Systemic Lupus Erythematosus – A Disease with A Dysregulated Type I Interferon System. Scand. J. Immunol. 2015; 82:199–7.
Rice G.I., Del Toro Duany Y., Jenkinson E.M., Forte G.M.A., Anderson B.H., Ariaudo G. et al. Gain-of-function mutations in IFIH1 cause a spectrum of human disease phenotypes associated with upregulated type i interferon signaling. Nat. Genet. 2014; 46(5):503–9.
Meuwissen M.E.C., Schot R., Buta S., Oudesluijs G., Tinschert S., Speer S.D. et al. Human USP18 deficiency underlies type 1 interferonopathy leading to severe pseudo-TORCH syndrome. J. Exp. Med. 2016; 213(7):1163–74.
Davidson S., Steiner A., Harapas C.R., Masters S.L. An Update on Autoinflammatory Diseases: Interferonopathies. Curr. Rheumatol. Rep. 2018; 20(7):38.
Rodero M.P., Decalf J., Bondet V., Hunt D., Rice G.I., Werneke S. et al. Detection of interferon alpha protein reveals differential levels and cellular sources in disease. J. Exp. Med. 2017; 214(5):1547–55.
Mathian A., Mouries-Martin S., Dorgham K., Devilliers H., Barnabei L., Ben Salah E. et al. Monitoring Disease Activity in Systemic Lupus Erythematosus With Single-Molecule Array Digital Enzyme-Linked Immunosorbent Assay Quantification of Serum Interferon-α. Arthritis Rheumatol. 2019; 71(5):756–65.
Melki I., Devilliers H., Gitiaux C., Bondet V., Belot A., Bodemer C. et al. Circulating Interferon-α Measured With a Highly Sensitive Assay as a Biomarker for Juvenile Inflammatory Myositis Activity: Comment on the Article by Mathian et al. Arthritis Rheumatol. 2020; 72(1):195-7.
Kuri T., Habjan M., Penski N., Weber F. Species-independent bioassay for sensitive quantification of antiviral type i interferons. Virol. J. 2010; 7:50.
Widman D.G. Bioassay for the measurement of type-i interferon activity. Methods Mol. Biol. 2013; 1031:91–6.
Rees P.A., Lowy R.J. Measuring type I interferon using reporter gene assays based on readily available cell lines. J. Immunol. Methods 2018; 461:63–72.
Bennett L., Palucka A.K., Arce E., Cantrell V., Borvak J., Banchereau J. et al. Interferon and granulopoiesis signatures in systemic lupus erythematosus blood. J. Exp. Med. 2003; 197(6):711–23.
Baechler E.C., Batliwalla F.M., Karypis G., Gaffney P.M., Ortmann W.A., Espe K.J. et al. Interferon-inducible gene expression signature in peripheral blood cells of patients with severe lupus. Proc. Natl. Acad. Sci. U S A. 2003; 100(5):2610–15.
Liu M., Liu J., Hao S., Wu P., Zhang X., Xiao Y. et al. Higher activation of the interferon-gamma signaling pathway in systemic lupus erythematosus patients with a high type I IFN score: relation to disease activity. Clin. Rheumatol. 2018; 37(10):2675–84.
Psarras A., Emery P., Vital E.M. Type I interferon-mediated autoimmune diseases: Pathogenesis, diagnosis and targeted therapy. Rheumatology (United Kingdom). 2017; 56: 1662–75.
Greenberg S.A., Higgs B.W., Morehouse C., Walsh R.J., Won Kong S., Brohawn P. et al. Relationship between disease activity and type 1 interferon- and other cytokine-inducible gene expression in blood in dermatomyositis and polymyositis. Genes Immun. 2012; 13(3):207–13.
Brkic Z., Van Bon L., Cossu M., Van Helden-Meeuwsen C.G., Vonk M.C., Knaapen H. et al. The interferon type i signature is present in systemic sclerosis before overt fibrosis and might contribute to its pathogenesis through high BAFF gene expression and high collagen synthesis. Ann. Rheum. Dis. 2016; 75(8):1567–73.
Lübbers J., Brink M., Van De Stadt L.A., Vosslamber S., Wesseling J.G., Van Schaardenburg D. et al. The type i IFN signature as a biomarker of preclinical rheumatoid arthritis. Ann. Rheum. Dis. 2013; 72(5):776–80.
Bodewes I.L.A., Versnel M.A. Interferon activation in primary Sjögren’s syndrome: recent insights and future perspective as novel treatment target. Expert Rev. Clin. Immunol. 2018; 14(10):817-29.
Yao Y., Richman L., Higgs B.W., Morehouse C.A., De Los Reyes M., Brohawn P. et al. Neutralization of interferon-α/β-inducible genes and downstream effect in a phase I trial of an anti-interferon-α monoclonal antibody in systemic lupus erythematosus. Arthritis Rheum. 2009; 60(6):1785–96.
Higgs B.W., Liu Z., White B., Zhu W., White W.I., Morehouse C. et al. Patients with systemic lupus erythematosus, myositis, rheumatoid arthritis and scleroderma share activation of a common type I interferon pathway. Ann. Rheum. Dis. 2011; 70(11):2029–36.
Rice G.I., Melki I., Frémond M.L., Briggs T.A., Rodero M.P., Kitabayashi N. et al. Assessment of Type I Interferon Signaling in Pediatric Inflammatory Disease. J. Clin. Immunol. 2017; 37(2):123–32.
Rice G.I., Forte G.M.A., Szynkiewicz M., Chase D.S., Aeby A., Abdel-Hamid M.S. et al. Assessment of interferon-related biomarkers in Aicardi-Goutières syndrome associated with mutations in TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, and ADAR: A case-control study. Lancet Neurol. 2013; 12(12):1159–69.
Lamot L., Niemietz I., Brown K.L. Methods for type I interferon detection and their relevance for clinical utility and improved understanding of rheumatic diseases. Clin. Exp. Rheumatol. 2019; 37(6):1077-83.
Geiss G.K., Bumgarner R.E., Birditt B., Dahl T., Dowidar N., Dunaway D.L. et al. Direct multiplexed measurement of gene expression with color-coded probe pairs. Nat. Biotechnol. 2008; 26(3):317–25.
Pescarmona R., Belot A., Villard M., Besson L., Lopez J., Mosnier I. et al. Comparison of RT-qPCR and Nanostring in the measurement of blood interferon response for the diagnosis of type I interferonopathies. Cytokine. 2019; 113:446–52.
Kim H., De Jesus A.A., Brooks S.R., Liu Y., Huang Y., Vantries R. et al. Development of a Validated Interferon Score Using NanoString Technology. J. Interf. Cytokine Res. 2018; 38(4):171–85.
Skrabl-Baumgartner A., Plecko B., Schmidt W.M., König N., Hershfield M., Gruber-Sedlmayr U. et al. Autoimmune phenotype with type i interferon signature in two brothers with ADA2 deficiency carrying a novel CECR1 mutation. Pediatr. Rheumatol. 2017; 22;15(1).
Volpi S., Tsui J., Mariani M., Pastorino C., Caorsi R., Sacco O. et al. Type I interferon pathway activation in COPA syndrome. Clin. Immunol. 2018; 187:33–6.
König N., Fiehn C., Wolf C., Schuster M., Cura Costa E., Tüngler V. et al. Familial chilblain lupus due to a gain-of-function mutation in STING. Ann. Rheum. Dis. 2017; 76(2):468–72.
de Jesus A.A., Hou Y., Brooks S., Malle L., Biancotto A., Huang Y. et al. Distinct interferon signatures and cytokine patterns define additional systemic autoinflammatory diseases. J. Clin. Invest. 2020; 130(4):1669–82.
El-Sherbiny Y.M., Psarras A., Yusof M.Y.M., Hensor E.M.A., Tooze R., Doody G. et al. A novel two-score system for interferon status segregates autoimmune diseases and correlates with clinical features. Sci Rep. 2018; 8(1).
Chiche L., Jourde-Chiche N., Whalen E., Presnell S., Gersuk V., Dang K. et al. Modular transcriptional repertoire analyses of adults with systemic lupus erythematosus reveal distinct type I and type II interferon signatures. Arthritis Rheumatol. 2014; 66(6):1583–95.
Pin A., Monasta L., Taddio A., Piscianz E., Tommasini A., Tesser A. An Easy and Reliable Strategy for Making Type I Interferon Signature Analysis Comparable among Research Centers. Diagnostics (Basel, Switzerland) 2019; 9(3).
Yip L., Fuhlbrigge R., Atkinson M.A., Fathman C.G. Impact of blood collection and processing on peripheral blood gene expression profiling in type 1 diabetes. BMC Genomics 2017; 18(1):1–16.
Menke A., Rex-Haffner M., Klengel T., Binder E.B., Mehta D. Peripheral blood gene expression: It all boils down to the RNA collection tubes. BMC Res. Notes. 2012; 5:1.
Skogholt A.H., Ryeng E., Erlandsen S.E., Skorpen F., Schønberg S.A., Sætrom P. Gene expression differences between PAXgene and Tempus blood RNA tubes are highly reproducible between independent samples and biobanks. BMC Res. Notes. 2017; 10(1):136.
Lamot L., Niemietz I., Brown K.L. Comparable type i interferon score determination from PAXgene and Tempus whole blood RNA collection and isolation systems. BMC Res. Notes. 2019; 12(1).
Brohawn P.Z., Streicher K., Higgs B.W., Morehouse C., Liu H., Illei G. et al. Type I interferon gene signature test–low and –high patients with systemic lupus erythematosus have distinct gene expression signatures. Lupus 2019; 28(13):1524–33.
Md Yusof M.Y., Psarras A., El-Sherbiny Y.M., Hensor E.M.A., Dutton K., Ul-Hassan S. et al. Prediction of autoimmune connective tissue disease in an at-risk cohort: Prognostic value of a novel two-score system for interferon status. Ann. Rheum. Dis. 2018; 77(10).
Wither J., Johnson S.R., Liu T., Noamani B., Bonilla D., Lisnevskaia L. et al. Presence of an interferon signature in individuals who are anti-nuclear antibody positive lacking a systemic autoimmune rheumatic disease diagnosis. Arthritis Res. Ther. 2017; 19(1).
Lambers W.M., De Leeuw K., Doornbos-Van Der Meer B., Diercks G.F.H., Bootsma H., Westra J. Interferon score is increased in incomplete systemic lupus erythematosus and correlates with myxovirus-resistance protein A in blood and skin. Arthritis Res. Ther. 2019; 21(1).
Rigolet M., Hou C., Baba Amer Y., Aouizerate J., Periou B., Gherardi R.K. et al. Distinct interferon signatures stratify inflammatory and dysimmune myopathies. RMD Open 2019; 5(1).
Bauer J.W., Petri M., Batliwalla F.M., Koeuth T., Wilson J., Slattery C. et al. Interferon-regulated chemokines as biomarkers of systemic lupus erythematosus disease activity: A validation study. Arthritis Rheum. 2009; 60(10):3098–107.
Nikpour M., Dempsey A.A., Urowitz M.B., Gladman D.D., Barnes D.A. Association of a gene expression profile from whole blood with disease activity in systemic lupus erythaematosus. Ann. Rheum. Dis. 2008; 67(8):1069–75.
Kirou K.A., Lee C., George S., Louca K., Peterson M.G.E., Crow M.K. Activation of the interferon-α pathway identifies a subgroup of systemic lupus erythematosus patients with distinct serologic features and active disease. Arthritis Rheum. 2005; 52(5):1491–503.
Feng X., Wu H., Grossman J.M., Hanvivadhanakul P., FitzGerald J.D., Park G.S. et al. Association of increased interferon-inducible gene expression with disease activity and lupus nephritis in patients with systemic lupus erythematosus. Arthritis Rheum. 2006; 54(9):2951–62.
Landolt-Marticorena C., Bonventi G., Lubovich A., Ferguson C., Unnithan T., Su J. et al. Lack of association between the interferon-α signature and longitudinal changes in disease activity in systemic lupus erythematosus. Ann. Rheum. Dis. 2009; 68(9):1440–46.
Brkic Z., Maria N.I., Van Helden-Meeuwsen C.G., Van De Merwe J.P., Van Daele P.L., Dalm V.A. et al. Prevalence of interferon type I signature in CD14 monocytes of patients with Sjögren’s syndrome and association with disease activity and BAFF gene expression. Ann. Rheum. Dis. 2013; 72(5):728–35.
Landolt-Marticorena C., Wither R., Reich H., Herzenberg A., Scholey J., Gladman D.D. et al. Increased expression of B cell activation factor supports the abnormal expansion of transitional B Cells in systemic lupus erythematosus. J. Rheumatol. 2011; 38(4):642–51.
Reed A.M., Peterson E., Bilgic H., Ytterberg S.R., Amin S., Hein M.S. et al. Changes in novel biomarkers of disease activity in juvenile and adult dermatomyositis are sensitive biomarkers of disease course. Arthritis Rheum. 2012 Dec; 64(12):4078–86.
Walsh R.J., Sek W.K., Yao Y., Jallal B., Kiener P.A., Pinkus J.L. et al. Type I interferon-inducible gene expression in blood is present and reflects disease activity in dermatomyositis and polymyositis. Arthritis Rheum. 2007; 56(11):3784–92.
Kim H., Gunter-Rahman F., McGrath J.A., Lee E., De Jesus A.A., Targoff I.N. et al. Expression of interferon-regulated genes in juvenile dermatomyositis versus Mendelian autoinflammatory interferonopathies. Arthritis Res Ther. 2020;22 (1).
Avdeeva A.S., Tchetina E.V., Cherkasova M.V., Markova G.A., Artyuhov A.S., Dashinimaev E.B., Nasonov E.L. The expression of interferon-stimulated genes (interferon “signature”) in patients with rheumatoid arthritis (preliminary results). Nauchno-prakticheskaya revmatologiya. 2020; 58(6):673-77. (in Russian).
Nasonov E.L., Avdeeva A.S., Lila A.M. Efficacy and safety of tofacitinib for immunemediated inflammatory rheumatic diseases (part 1). Nauchno-prakticheskaya revmatologiya. 2020; 58(2):214-24. (in Russian).
Merrill J.T., Wallace D.J., Petri M., Kirou K.A., Yao Y., White W.I. et al. Safety profile and clinical activity of sifalimumab, a fully human anti-interferon α monoclonal antibody, in systemic lupus erythematosus: A phase I, multicentre, double-blind randomised study. Ann. Rheum. Dis. 2011; 70(11):1905–13.
Khamashta M., Merrill J.T., Werth V.P., Furie R., Kalunian K., Illei G.G. et al. Sifalimumab, an anti-interferon-α monoclonal antibody, in moderate to severe systemic lupus erythematosus: A randomised, double-blind, placebo-controlled study. Ann. Rheum. Dis. 2016; 75(11):1909–16.
Higgs B.W., Zhu W., Morehouse C., White W.I., Brohawn P., Guo X. et al. A phase 1b clinical trial evaluating sifalimumab, an anti-IFN-α monoclonal antibody, shows target neutralisation of a type I IFN signature in blood of dermatomyositis and polymyositis patients. Ann. Rheum. Dis. 2014; 73(1):256–62.
Bizzini B., Drouet B., Zagury D., Abitbol M., Burny A., Boissier M.C. Kinoids: A family of immunogens for active anticytokine immunotherapy applied to autoimmune diseases and cancer. Immunotherapy. 2010; 2(3):347-65.
Pellerin A., Otero K., Czerkowicz J.M., Kerns H.M., Shapiro R.I., Ranger A.M. et al. Anti- BDCA 2 monoclonal antibody inhibits plasmacytoid dendritic cell activation through Fc-dependent and Fc-independent mechanisms. EMBO Mol. Med. 2015; 7(4):464–76.
Furie R., Werth V.P., Merola J.F., Stevenson L., Reynolds T.L., Naik H. et al. Monoclonal antibody targeting BDCA2 ameliorates skin lesions in systemic lupus erythematosus. J. Clin. Invest. 2019; 129(3):1359–71.
معلومات مُعتمدة: 20-45-01005 The Russian Science Foundation
فهرسة مساهمة: Keywords: autoimmune disease; interferon score; interferonopathy; review; type I interferons
المشرفين على المادة: 0 (Interferon Type I)
تواريخ الأحداث: Date Created: 20210528 Date Completed: 20210531 Latest Revision: 20210531
رمز التحديث: 20221213
DOI: 10.51620/0869-2084-2021-66-5-279-284
PMID: 34047513
قاعدة البيانات: MEDLINE
الوصف
تدمد:0869-2084
DOI:10.51620/0869-2084-2021-66-5-279-284