IRDS Genes: Interfaces and Pathways: Comparison
Please note this is a comparison between Version 2 by Vivi Li and Version 1 by Monikaben Padariya.

Interferon (IFN)-related DNA damage resistant signature (IRDS) genes are a subgroup of interferon-stimulated genes (ISGs) found upregulated in different cancer types, which promotes resistance to DNA damaging chemotherapy and radiotherapy.

  • DNA damage
  • IRDS genes
  • DNA
  • RNA
  • ATP
  • functional site
  • viruses
  • receptors
  • resistance
  • interferon
  • chemotherapy and radiotherapy
  • protein interfaces
  • upstream regulator
Please wait, diff process is still running!

References

  1. Deng, L.; Liang, H.; Fu, S.; Weichselbaum, R.R.; Fu, Y.X. From DNA damage to nucleic acid sensing: A strategy to enhanceradiation therapy. Clin. Cancer Res. 2016, 22, 20–25.
  2. Burnette, B.C.; Liang, H.; Lee, Y.; Chlewicki, L.; Khodarev, N.N.; Weichselbaum, R.R.; Fu, Y.X.; Auh, S.L. The efficacy of radiotherapy relies upon induction of type i interferon-dependent innate and adaptive immunity. Cancer Res. 2011, 71, 2488–2496.
  3. Lim, J.Y.; Gerber, S.A.; Murphy, S.P.; Lord, E.M. Type I interferons induced by radiation therapy mediate recruitment and effector function of CD8(+) T cells. Cancer Immunol. Immunother. 2014, 63, 259–271.
  4. Sarhan, J.; Liu, B.C.; Muendlein, H.I.; Weindel, C.G.; Smirnova, I.; Tang, A.Y.; Ilyukha, V.; Sorokin, M.; Buzdin, A.; Fitzgerald, K.A.; et al. Constitutive interferon signaling maintains critical threshold of MLKL expression to license necroptosis. Cell Death Differ. 2019, 26, 332–347.
  5. Snyder, A.G.; Hubbard, N.W.; Messmer, M.N.; Kofman, S.B.; Hagan, C.E.; Orozco, S.L.; Chiang, K.; Daniels, B.P.; Baker, D.; Oberst, A. Intra tumoral activation of the necroptotic pathway components RIPK1 and RIPK3 potentiates antitumor immunity. Sci. Immunol. 2019, 4, eaaw2004.
  6. Erdal, E.; Haider, S.; Rehwinkel, J.; Harris, A.L.; McHugh, P.J. A prosurvival DNA damage-induced cytoplasmic interferon responseis mediated by end resection factors and is limited by Trex1. Genes Dev. 2017, 31, 353–369.
  7. Berman, H.M.; Westbrook, J.; Feng, Z.; Gilliland, G.; Bhat, T.N.; Weissig, H.; Shindyalov, I.N.; Bourne, P.E. The Protein DataBank. Nucleic Acids Res. 2000, 28, 235–242.
  8. Isaacs, A.; Lindenmann, J. Virus interference. I. The interferon. Proc. R. Soc. Lond. B Biol. Sci. 1957, 147, 258–267.
  9. Platanias, L.C. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat. Rev. Immunol. 2005, 5, 375–386.
  10. Kotredes, K.P.; Gamero, A.M. Interferons as inducers of apoptosis in malignant cells. J. Interferon Cytokine Res. 2013, 33, 162–170.
  11. Gresser, I.; Maury, C.; Brouty-Boyé, D. Mechanism of the antitumou reffect of interferon in mice. Nature 1972, 239, 167–168.
  12. Kaplan, D.H.; Shankaran, V.; Dighe, A.S.; Stockert, E.; Aguet, M.; Old, L.J.; Schreiber, R.D. Demonstration of an interferon dependent tumor surveillance system in immunocompetent mice. Proc. Natl. Acad. Sci. USA 1998, 95, 7556–7561.
  13. Borden, E.C.; Sen, G.C.; Uze, G.; Silverman, R.H.; Ransohoff, R.M.; Foster, G.R.; Stark, G.R. Interferons at age 50: Past, current and future impact on biomedicine. Nat. Rev. Drug Discov. 2007, 6, 975–990.
  14. Walter, M.R. The role of structure in the biology of interferon signaling. Front. Immunol. 2020, 11, 606489.
  15. Ivashkiv, L.B.; Donlin, L.T. Regulation of type I interferon responses. Nat. Rev. Immunol. 2014, 14, 36–49.
  16. Staeheli, P. Interferon induced proteins and the antiviral state. Adv. Virus Res. 1990, 38, 147–200.
  17. Thomas, C.; Moraga, I.; Levin, D.; Krutzik, P.O.; Podoplelova, Y.; Trejo, A.; Lee, C.; Yarden, G.; Vleck, S.E.; Glenn, J.S.; et al. Structural linkage between ligand discrimination and receptor activation by type I interferons. Cell 2011, 146, 621–632.
  18. Mendoza, J.L.; Escalante, N.K.; Jude, K.M.; SotolongoBellon, J.; Su, L.; Horton, T.M.; Tsutsumi, N.; Berardinelli, S.J.; Haltiwanger, R.S.; Piehler, J.; et al. Structure of the IFNγ receptor complex guides design of biased agonists. Nature 2019, 567, 56–60.
  19. Mendoza, J.L.; Schneider, W.M.; Hoffmann, H.H.; Vercauteren, K.; Jude, K.M.; Xiong, A.; Moraga, I.; Horton, T.M.; Glenn, J.S.; De Jong, Y.P.; et al. The IFN-λ-IFN-λR1-IL-10Rβ Complex reveals structural features underlying type III IFN functional plasticity. Immunity 2017, 46, 379–392.
  20. Mesev, E.V.; LeDesma, R.A.; Ploss, A. Decoding type I and III interferon signalling during viral infection. Nat. Microbiol. 2019, 4, 914–924.
  21. Oon, S.; Wilson, N.J.; Wicks, I. Targeted therapeutics in SLE: Emerging strategies to modulate the interferon pathway. Clin. Transl. Immunol. 2016, 5, e79.
  22. Krause, C.D.; Pestka, S. Evolution of the Class 2 cytokines and receptors, and discovery of new friends and relatives. Pharmacol. Ther. 2005, 106, 299–346.
  23. Novick, D.; Cohen, B.; Rubinstein, M. The human interferon alpha/beta receptor: Characterization and molecular cloning. Cell 1994, 77, 391–400.
  24. Roisman, L.C.; Jaitin, D.A.; Baker, D.P.; Schreiber, G. Mutational analysis of the IFNAR1 binding site on IFN alpha 2 reveals the architecture of a weak ligand-receptor binding site. J. Mol. Biol. 2005, 353, 271–281.
  25. Jaitin, D.A.; Roisman, L.C.; Jaks, E.; Gavutis, M.; Piehler, J.; Vander Heyden, J.; Uze, G.; Schreiber, G. Inquiring into the differential action of interferons (IFNs): An IFN-alpha2 mutant with enhanced affinity to IFNAR1 is functionally similar to IFN-beta. Mol. Cell Biol. 2006, 26, 1888–1897.
  26. Piehler, J.; Schreiber, G. Mutational and structural analysis of the binding interface between type I interferons and their receptor Ifnar2. J. Mol. Biol. 1999, 294, 223–237.
  27. Stark, G.R.; Kerr, I.M.; Williams, B.R.; Silverman, R.H.; Schreiber, R.D. How cells respond to interferons. Annu. Rev. Biochem. 1998, 67, 227–264.
  28. Hamilton, J.A.; Whitty, G.A.; Kola, I.; Hertzog, P.J. Endogenous IFN-alpha beta suppresses colony-stimulating factor(CSF) 1 stimulated macrophage DNA synthesis and mediates inhibitory effects of lipopolysaccharide and TNF-alpha. J. Immunol. 1996, 156, 2553–2557.
  29. Marié, I.; Durbin, J.E.; Levy, D.E. Differential viral induction of distinct interferon-alpha genes by positive feedback through interferon regulatory factor-7. EMBO J. 1998, 17, 6660–6669.
  30. Vogel, S.N.; Fertsch, D. Endogenous interferon production by endotoxin-responsive macrophages provides an autostimulatory differentiation signal. Infect. Immun. 1984, 45, 417–423.
  31. Takaoka, A.; Mitani, Y.; Suemori, H.; Sato, M.; Yokochi, T.; Noguchi, S.; Tanaka, N.; Taniguchi, T. Crosstalk between interferon-gamma and -alpha/beta signaling components in caveolar membrane domains. Science 2000, 288, 2357–2360.
  32. Foster, G.R.; Rodrigues, O.; Ghouze, F.; Schulte-Frohlinde, E.; Testa, D.; Liao, M.J.; Stark, G.R.; Leadbeater, L.; Thomas, H.C. Different relative activities of human cell derived interferon-alpha subtypes: IFN-alpha 8 has very high antiviral potency. J. Interferon Cytokine Res. 1996, 16, 1027–1033.
  33. Hibbert, L.; Foster, G.R. Human type I interferons differ greatly in their effects on the proliferation of primary B cells. J. Interferon Cytokine Res. 1999, 19, 309–318.
  34. Sadler, A.J.; Williams, B.R. Interferon inducible antiviral effectors. Nat. Rev. Immunol. 2008, 8, 559–568.
  35. Sistigu, A.; Yamazaki, T.; Vacchelli, E.; Chaba, K.; Enot, D.P.; Adam, J.; Vitale, I.; Goubar, A.; Baracco, E.E.; Remédios, C.; et al. Cancer cell autonomous contribution of type I interferon signaling to the efficacy of chemotherapy. Nat. Med. 2014, 20, 1301–1309.
  36. Deng, L.; Liang, H.; Xu, M.; Yang, X.; Burnette, B.; Arina, A.; Li, X.D.; Mauceri, H.; Beckett, M.; Darga, T.; et al. STING-dependent cytosolic DNA sensing promotes radiation induced Type I interferon dependent antitumor immunity in immunogenic tumors. Immunity 2014, 41, 843–852.
  37. Yang, H.; Wang, H.; Ren, J.; Chen, Q.; Chen, Z.J. cGAS is essential for cellular senescence. Proc. Natl. Acad. Sci. USA 2017, 114, E4612–E4620.
  38. Gasser, S.; Zhang, W.Y.L.; Tan, N.Y.J.; Tripathi, S.; Suter, M.A.; Chew, Z.H.; Khatoo, M.; Ngeow, J.; Cheung, F.S.G. Sensing of dangerous DNA. Mech. Ageing Dev. 2017, 165, 33–46.
  39. Hong, C.; Tijhuis, A.E.; Foijer, F. The cGAS Paradox: Contrasting roles for cGAS-STING pathway in chromosomal instability. Cells 2019, 8, 1228.
  40. Nastasi, C.; Mannarino, L.; D’Incalci, M. DNA damage response and immune defense. Int. J. Mol. Sci. 2020, 21, 7504.
  41. Jiang, M.; Chen, P.; Wang, L.; Li, W.; Chen, B.; Liu, Y.; Wang, H.; Zhao, S.; Ye, L.; He, Y.; et al. cGAS-STING, an important pathway in cancer immunotherapy. J. Hematol. Oncol. 2020, 13, 81.
  42. Luecke, S.; Holleufer, A.; Christensen, M.H.; Jønsson, K.L.; Boni, G.A.; Sørensen, L.K.; Johannsen, M.; Jakobsen, M.R.; Hartmann, R.; Paludan, S.R. cGAS is activated by DNA in a length-dependent manner. EMBO Rep. 2017, 18, 1707–1715.
  43. Marcus, A.; Mao, A.J.; Lensink-Vasan, M.; Wang, L.; Vance, R.E.; Raulet, D.H. Tumor derived cGAMP triggers a STING mediated interferon response in non-tumor cells to activate the NK cell response. Immunity 2018, 49, 754–763.e4.
  44. Kanda, N.; Shimizu, T.; Tada, Y.; Watanabe, S. IL-18 enhances IFN-γ induced production of CXCL9, CXCL10, and CXCL11 in humank eratinocytes. Eur. J. Immunol. 2007, 37, 338–350.
  45. Yu, H.; Pardoll, D.; Jove, R. STATs in cancer inflammation and immunity: A leading role for STAT3. Nat. Rev. Cancer 2009, 9, 798–809.
  46. Jones, L.M.; Broz, M.L.; Ranger, J.J.; Ozcelik, J.; Ahn, R.; Zuo, D.; Ursini-Siegel, J.; Hallett, M.T.; Krummel, M.; Muller, W.J. STAT3 Establishes an immuno suppressive micro environment during the early stages of breast carcinogenes is to promote tumor growth and metastasis. Cancer Res. 2016, 76, 1416–1428.
  47. Kortylewski, M.; Kujawski, M.; Wang, T.; Wei, S.; Zhang, S.; Pilon-Thomas, S.; Niu, G.; Kay, H.; Mulé, J.; Kerr, W.G.; et al. Inhibiting Stat3 signaling in the hematopoietic system elicits multi component antitumor immunity. Nat. Med. 2005, 11, 1314–1321.
  48. Grégoire, C.; Chasson, L.; Luci, C.; Tomasello, E.; Geissmann, F.; Vivier, E.; Walzer, T. The trafficking of natural killer cells. Immunol. Rev. 2007, 220, 169–182.
  49. Elsea, C.R.; Roberts, D.A.; Druker, B.J.; Wood, L.J. Inhibition of p38MAPK suppresses inflammatory cytokine induction by Etoposide, 5-Fluorouracil, and Doxorubicin without affecting tumoricidal activity. PLoS ONE 2008, 3, e02355.
  50. Li, A.; Yi, M.; Qin, S.; Song, Y.; Chu, Q.; Wu, K. Activating cGAS-STING pathway for the optimal effect of cancer immunotherapy. J. Hematol. Oncol. 2019, 12, 35.
  51. Parkes, E.E.; Walker, S.M.; Taggart, L.E.; McCabe, N.; Knight, L.A.; Wilkinson, R.; McCloskey, K.D.; Buckley, N.E.; Savage, K.I.; Salto-Tellez, M.; et al. Activation of STING-dependent innate immune signaling by S-Phase specific DNA damage in breast cancer. J. Natl. Cancer Inst. 2017, 109, djw199.
  52. Harding, S.M.; Benci, J.L.; Irianto, J.; Discher, D.E.; Minn, A.J.; Greenberg, R.A. Mitotic progression following DNA damage enables pattern recognition within micro nuclei. Nature 2017, 548, 466–470.
  53. Gerber, S.A.; Sedlacek, A.L.; Cron, K.R.; Murphy, S.P.; Frelinger, J.G.; Lord, E.M. IFN-γ mediates the antitumor effects of radiation therapy in a murine colon tumor. Am. J. Pathol. 2013, 182, 2345–2354.
  54. Tanimoto, T.; Yamamoto, S.; Taniai, M.; Taniguchi, M.; Ariyasu, H.; Ushio, C.; Aga, M.; Mukai, Y.; Tsutsumi, Y.; Ariyasu, T.; et al. The combination of IFN-alpha 2 and IFN-alpha 8 exhibits synergistic antiproliferative activity on renal cell carcinoma (RCC) cell lines through increased binding affinity for IFNAR-2. J. Interferon Cytokine Res. 2007, 27, 517–523.
  55. DiFranco, S.; Turdo, A.; Todaro, M.; Stassi, G. Role of Type I and II Interferons in colorectal cancer and melanoma. Front. Immunol. 2017, 8, 878.
  56. Minn, A.J. Interferons and the immunogenic effects of cancer therapy. Trends Immunol. 2015, 36, 725–737.
  57. Khodarev, N.N.; Beckett, M.; Labay, E.; Darga, T.; Roizman, B.; Weichselbaum, R.R. STAT1 is overexpressed in tumors selected for radioresistance and confers protection from radiation in transduced sensitive cells. Proc. Natl. Acad. Sci. USA 2004, 101, 1714–1719.
  58. Harris, A.L. Hypoxia- a key regulatory factor in tumour growth. Nat. Rev. Cancer 2002, 2, 38–47.
  59. Hallahan, D.E.; Haimovitz-Friedman, A.; Kufe, D.W.; Fuks, Z.; Weichselbaum, R.R. The role of cytokines in radiation oncology. Important Adv. Oncol. 1993, 71–80.
  60. Marples, B.; Scott, S.D.; Hendry, J.H.; Embleton, M.J.; Lashford, L.S.; Margison, G.P. Development of synthetic promoters for radiation-mediated gene therapy. Gene Ther. 2000, 7, 511–517.
  61. Kita, K.; Sugaya, S.; Zhai, L.; Wu, Y.P.; Wano, C.; Chigira, S.; Nomura, J.; Takahashi, S.; Ichinose, M.; Suzuki, N. Involvement of LEU13 in interferon-induced refractoriness of human RSa cells to cell killing by Xrays. Radiat. Res. 2003, 160, 302–308.
  62. Benci, J.L.; Johnson, L.R.; Choa, R.; Xu, Y.; Qiu, J.; Zhou, Z.; Xu, B.; Ye, D.; Nathanson, K.L.; June, C.H.; et al. Opposing functions of interferon coordinate adaptive and innate immune responses to cancer immune check point blockade. Cell 2019, 178, 933–948.e14.
  63. Duarte, C.W.; Willey, C.D.; Zhi, D.; Cui, X.; Harris, J.J.; Vaughan, L.K.; Mehta, T.; McCubrey, R.O.; Khodarev, N.N.; Weichselbaum, R.R.; et al. Expression signature of IFN/STAT1 signaling genes predicts poor survival outcome in glioblastoma multiforme in a subtype-specific manner. PLoS ONE 2012, 7, e029653.
  64. Tsai, M.H.; Cook, J.A.; Chandramouli, G.V.; DeGraff, W.; Yan, H.; Zhao, S.; Coleman, C.N.; Mitchell, J.B.; Chuang, E.Y. Gene expression profiling of breast, prostate, and glioma cells following single versus fractionated doses of radiation. Cancer Res. 2007, 67, 3845–3852.
  65. Liauw, S.L.; Connell, P.P.; Weichselbaum, R.R. New paradigms and future challenges in radiation oncology: An update of biological targets and technology. Sci. Transl. Med. 2013, 5, 173sr2.
  66. Khodarev, N.N.; Minn, A.J.; Efimova, E.V.; Darga, T.E.; Labay, E.; Beckett, M.; Mauceri, H.J.; Roizman, B.; Weichselbaum, R.R. Signal transducer and activator of transcription 1 regulates both cytotoxic and prosurvival functions in tumor cells. Cancer Res. 2007, 67, 9214–9220.
  67. Cheon, H.; Stark, G.R. Unphosphorylated STAT1 prolongs the expression of interferon-induced immune regulatory genes. Proc. Natl. Acad. Sci. USA 2009, 106, 9373–9378.
  68. Cheon, H.; Yang, J.; Stark, G.R. The functions of signal transducers and activators of transcriptions 1 and 3 as cytokine-inducible proteins. J. Interferon Cytokine Res. 2011, 31, 33–40.
  69. Cheon, H.; Holvey-Bates, E.G.; Schoggins, J.W.; Forster, S.; Hertzog, P.; Imanaka, N.; Rice, C.M.; Jackson, M.W.; Junk, D.J.; Stark, G.R. IFNβ dependent increases in STAT1, STAT2, and IRF9 mediate resistance to viruses and DNA damage. EMBO J. 2013, 32, 2751–2763.
  70. Khodarev, N.N.; Roach, P.; Pitroda, S.P.; Golden, D.W.; Bhayani, M.; Shao, M.Y.; Darga, T.E.; Beveridge, M.G.; Sood, R.F.; Sutton, H.G.; et al. STAT1 pathway mediates amplification of metastatic potential and resistance to therapy. PLoS ONE 2009, 4, e05821.
  71. Khodarev, N.N.; Roizman, B.; Weichselbaum, R.R. Molecular pathways: Interferon/stat1 pathway: Role in the tumor resistance to genotoxic stress and aggressive growth. Clin. Cancer Res. 2012, 18, 3015–3021.
  72. Takeuchi, O.; Akira, S. Pattern recognition receptors and inflammation. Cell 2010, 140, 805–820.
  73. Mackenzie, K.J.; Carroll, P.; Martin, C.A.; Murina, O.; Fluteau, A.; Simpson, D.J.; Olova, N.; Sutcliffe, H.; Rainger, J.K.; Leitch, A.; et al. cGAS surveillance of micronuclei links genome instability to innate immunity. Nature 2017, 548, 461–465.
  74. Cheon, H.; Borden, E.C.; Stark, G.R. Interferons and their stimulated genes in the tumor microenvironment. Semin. Oncol. 2014, 41, 156–173.
  75. Weichselbaum, R.R.; Ishwaran, H.; Yoon, T.; Nuyten, D.S.; Baker, S.W.; Khodarev, N.; Su, A.W.; Shaikh, A.Y.; Roach, P.; Kreike, B.; et al. An interferon-related gene signature for DNA damage resistance is a predictive marker for chemotherapy and radiation for breast cancer. Proc. Natl. Acad. Sci. USA 2008, 105, 18490–18495.
  76. Wallace, T.A.; Martin, D.N.; Ambs, S. Interactions among genes, tumor biology and the environment in cancer health disparities: Examining the evidence on a national and global scale. Carcinogenesis 2011, 32, 1107–1121.
  77. UniProt Consortium. UniProt: A worldwide hub of protein knowledge. Nucleic Acids Res. 2019, 47, D506–D515.
  78. Raudvere, U.; Kolberg, L.; Kuzmin, I.; Arak, T.; Adler, P.; Peterson, H.; Vilo, J. g:Profiler: A web server for functional enrichment analysis and conversions of gene lists (2019 update). Nucleic Acids Res. 2019, 47, W191–W198.
  79. Reimand, J.; Isserlin, R.; Voisin, V.; Kucera, M.; Tannus-Lopes, C.; Rostamianfar, A.; Wadi, L.; Meyer, M.; Wong, J.; Xu, C.; et al. Pathway enrichment analysis and visualization of omics data using g:Profiler, GSEA, Cytoscape and Enrichment Map. Nat. Protoc. 2019, 14, 482–517.
  80. Shannon, P.; Markiel, A.; Ozier, O.; Baliga, N.S.; Wang, J.T.; Ramage, D.; Amin, N.; Schwikowski, B.; Ideker, T. Cytoscape: A software environment for integrated models of biomolecular interaction networks. Genome Res. 2003, 13, 2498–2504.
  81. Rusinova, I.; Forster, S.; Yu, S.; Kannan, A.; Masse, M.; Cumming, H.; Chapman, R.; Hertzog, P.J. Interferome v2.0: An updated database of annotated interferon-regulated genes. Nucleic Acids Res. 2013, 41, D1040–D1046.
  82. Stark, G.R.; Darnell, J.E. The JAK-STAT pathway at twenty. Immunity 2012, 36, 503–514.
  83. Savoldi-Barbosa, M.; Sakamoto-Hojo, E.T. Influence of interferon-gamma on radiation-induced apoptosis in normal and a taxia-telangiectasia fibroblast cell lines. Teratog. Carcinog. Mutagen. 2001, 21, 417–429.
  84. Sirota, N.P.; Bezlepkin, V.G.; Kuznetsova, E.A.; Lomayeva, M.G.; Milonova, I.N.; Ravin, V.K.; Gaziev, A.I.; Bradbury, R.J. Modifying effect in vivo of interferon alpha on induction and repair of lesions of DNA of lymphoid cells of gamma-irradiated mice. Radiat. Res. 1996, 146, 100–105.
  85. Wu, B.; Hur, S. How RIG-I like receptors activate MAVS. Curr. Opin. Virol. 2015, 12, 91–98.
  86. Boelens, M.C.; Wu, T.J.; Nabet, B.Y.; Xu, B.; Qiu, Y.; Yoon, T.; Azzam, D.J.; Twyman-SaintVictor, C.; Wiemann, B.Z.; Ishwaran, H.; et al. Exosome transfer from stromal to breast cancer cells regulates therapy resistance pathways. Cell 2014, 159, 499–513.
  87. Gonzalez, K.J.; Moncada-Giraldo, D.M.; Gutierrez, J.B. In silico identification of potential inhibitors against human 2′-5′-oligoadenylate synthetase (OAS) proteins. Comput. Biol. Chem. 2020, 85, 107211.
  88. Souissi, I.; Ladam, P.; Cognet, J.A.; LeCoquil, S.; Varin-Blank, N.; Baran-Marszak, F.; Metelev, V.; Fagard, R. A STAT3-inhibitory hairpin decoy oligodeoxy nucleotide discriminates between STAT1 and STAT3 and induces death in a human colon carcinoma cell line. Mol. Cancer 2012, 11, 12.
  89. Joo, C.H.; Shin, Y.C.; Gack, M.; Wu, L.; Levy, D.; Jung, J.U. Inhibition of interferon regulatory factor7 (IRF7) mediated interferon signal transduction by the Kaposi’s sarcoma-associated herpes virus viral IRF homolog vIRF3. J. Virol. 2007, 81, 8282–8292.
  90. Kalathiya, U.; Padariya, M.; Faktor, J.; Coyaud, E.; Alfaro, J.A.; Fahraeus, R.; Hupp, T.R.; Goodlett, D.R. Interfaces with structure dynamics of the workhorses from cells revealed through cross-linking mass spectrometry (CLMS). Biomolecules 2021, 11, 382.
  91. Chen, X.; Vinkemeier, U.; Zhao, Y.; Jeruzalmi, D.; Darnell, J.E., Jr.; Kuriyan, J. Crystal structure of atyrosine phosphorylated STAT-1 dimer bound to DNA. Cell 1998, 93, 827–839.
  92. Ibsen, M.S.; Gad, H.H.; Andersen, L.L.; Hornung, V.; Julkunen, I.; Sarkar, S.N.; Hartmann, R. Structural and functional analysis reveals that human OASL binds dsRNA to enhance RIG-I signaling. Nucleic Acids Res. 2015, 43, 5236–5248.
  93. Panne, D.; Maniatis, T.; Harrison, S.C. An atomic model of the interferon-beta enhanceosome. Cell 2007, 129, 1111–1123.
  94. Dar, A.C.; Dever, T.E.; Sicheri, F. Higher-order substrate recognition of eIF2 alpha by the RNA-dependent protein kinase PKR. Cell 2005, 122, 887–900.
  95. Chen, M.H.; Ben-Efraim, I.; Mitrousis, G.; Walker-Kopp, N.; Sims, P.J.; Cingolani, G. Phospholipid scramblase 1 contains a nonclassical nuclear localization signal with unique binding site in importin alpha. J. Biol. Chem. 2005, 280, 10599–10606.
  96. Wu, B.; Peisley, A.; Richards, C.; Yao, H.; Zeng, X.; Lin, C.; Chu, F.; Walz, T.; Hur, S. Structural basis for dsRNA recognition, filament formation, and antiviral signal activation by MDA5. Cell 2013, 152, 276–289.
  97. Yang, H.; Wang, J.; Jia, X.; McNatt, M.W.; Zang, T.; Pan, B.; Meng, W.; Wang, H.W.; Bieniasz, P.D.; Xiong, Y. Structural insight into the mechanisms of enveloped virus tethering by tetherin. Proc. Natl. Acad. Sci. USA 2010, 107, 18428–18432.
  98. Abbas, Y.M.; Laudenbach, B.T.; Martínez-Montero, S.; Cencic, R.; Habjan, M.; Pichlmair, A.; Damha, M.J.; Pelletier, J.; Nagar, B. Structure of human IFIT1 with capped RNA reveals adaptable mRNA binding and mechanisms for sensing N1 and N2 ribose 2′-Omethylations. Proc. Natl. Acad. Sci. USA 2017, 114, E2106–E2115.
  99. Donovan, J.; Whitney, G.; Rath, S.; Korennykh, A. Structural mechanism of sensing long dsRNA via an oncatalytic domain in human oligoadenylate synthetase 3. Proc. Natl. Acad. Sci. USA 2015, 112, 3949–3954.
  100. Donovan, J.; Dufner, M.; Korennykh, A. Structural basis for cytosolic double-stranded RNA surveillance by human oligoadenylate synthetase 1. Proc. Natl. Acad. Sci. USA 2013, 110, 1652–1657.
  101. Rennie, M.L.; McKelvie, S.A.; Bulloch, E.M.; Kingston, R.L. Transient dimerization of human MxA promotes GTP hydrolysis, resulting in a mechanical power stroke. Structure 2014, 22, 1433–1445.
  102. Fribourgh, J.L.; Nguyen, H.C.; Matreyek, K.A.; Alvarez, F.J.D.; Summers, B.J.; Dewdney, T.G.; Aiken, C.; Zhang, P.; Engelman, A.; Xiong, Y. Structural insight into HIV-1 restriction by MxB. Cell Host Microbe 2014, 16, 627–638.
  103. Mazza, C. Human Type I 17 Beta-Hydroxysteroid Dehydrogenase: Site Directed Mutagenesis and X-ray Crystallography Structure Function Analysis. Ph.D. Thesis, Universite Joseph Fourier, Grenoble, France, 1997.
  104. Wathelet, M.G.; Lin, C.H.; Parekh, B.S.; Ronco, L.V.; Howley, P.M.; Maniatis, T. Virus infection induces the assembly of coordinately activated transcription factors on the IFN-beta enhancer in vivo. Mol. Cell 1998, 1, 507–518.
  105. Yang, H.; Lin, C.H.; Ma, G.; Baffi, M.O.; Wathelet, M.G. Interferon regulatory factor-7 synergizes with other transcription factors through multiple interactions with p300/CBP coactivators. J. Biol. Chem. 2003, 278, 15495–15504.
  106. Ning, S.; Pagano, J.S.; Barber, G.N. IRF7: Activation, regulation, modification and function. Genes Immun. 2011, 12, 399–414.
  107. Lin, R.; Génin, P.; Mamane, Y.; Hiscott, J. Selective DNA binding and association with the CREB binding protein coactivator contribute to differential activation of alpha/beta interferon genes by interferon regulatory factors 3 and 7. Mol. Cell Biol. 2000, 20, 6342–6353.
  108. Kileng, O.; Bergan, V.; Workenhe, S.T.; Robertsen, B. Structural and functional studies of an IRF-7-like gene from Atlantics almon. Dev. Comp. Immunol. 2009, 33, 18–27.
  109. Kristiansen, H.; Gad, H.H.; Eskildsen-Larsen, S.; Despres, P.; Hartmann, R. The oligoadenylate synthetase family: An ancient protein family with multiple antiviral activities. J. Interferon Cytokine Res. 2011, 31, 41–47.
  110. Kristiansen, H.; Scherer, C.A.; McVean, M.; Iadonato, S.P.; Vends, S.; Thavachelvam, K.; Steffensen, T.B.; Horan, K.A.; Kuri, T.; Weber, F.; et al. Extracellular 2′-5′ oligoadenylate synthetase stimulates RNaseL-independent antiviral activity: A novel mechanism of virus induced innate immunity. J. Virol. 2010, 84, 11898–11904.
  111. Latham, K.E.; Cosenza, S.; Reichenbach, N.L.; Mordechai, E.; Adelson, M.E.; Kon, N.; Horvath, S.E.; Charubala, R.; Mikhailov, S.N.; Pfeiderer, W.; et al. Inhibition of growth of estrogen receptor positive and estrogen receptor negative breast cancer cells in culture by AA-etherA, a stable 2-5A derivative. Oncogene 1996, 12, 827–837.
  112. Feng, Z.; Zheng, W.; Tang, Q.; Cheng, L.; Li, H.; Ni, W.; Pan, X. Fludarabine inhibits STAT1-mediated up-regulation of caspase-3 expression indexamethasone-induced osteoblasts apoptosis and slows the progression of steroid induced a vascular necrosis of the femoral head in rats. Apoptosis 2017, 22, 1001–1012.
  113. Gunning, P.T.; Katt, W.P.; Glenn, M.; Siddiquee, K.; Kim, J.S.; Jove, R.; Sebti, S.M.; Turkson, J.; Hamilton, A.D. Isoform selective inhibition of STAT1 or STAT3 homo-dimerization via peptidomimetic probes: Structural recognition of STAT SH2 domains. Bioorg. Med. Chem. Lett. 2007, 17, 1875–1878.
  114. Zhou, X.X.; Gao, P.J.; Sun, B.G. Pravastatin attenuates interferon-gamma action via modulation of STAT1 to prevent aortic atherosclerosis in apolipoprotein E-knockout mice. Clin. Exp. Pharmacol. Physiol. 2009, 36, 373–379.
  115. Böhmer, F.D.; Friedrich, K. Protein tyrosine phosphatases as wardens of STAT signaling. JAKSTAT 2014, 3, e28087.
  116. Porritt, R.A.; Hertzog, P.J. Dynamic control of type I IFN signalling by an integrated network of negative regulators. Trends Immunol. 2015, 36, 150–160.
  117. Rytinki, M.M.; Kaikkonen, S.; Pehkonen, P.; Jääskeläinen, T.; Palvimo, J.J. PIA Sproteins: Pleiotropic interactors associated with SUMO. Cell. Mol. Lif eSci. 2009, 66, 3029–3041.
  118. Usmani, S.Z.; Sexton, R.; Ailawadhi, S.; Shah, J.J.; Valent, J.; Rosenzweig, M.; Lipe, B.; Zonder, J.A.; Fredette, S.; Durie, B.; et al. Phase I safety data of lenalidomide, bortezomib, dexamethasone, and elotuzumab as induction therapy for newly diagnosed symptomatic multiple myeloma: SWOG S1211. Blood Cancer J. 2015, 5, e334.
  119. Lan, Q.; Peyvandi, S.; Duffey, N.; Huang, Y.T.; Barras, D.; Held, W.; Richard, F.; Delorenzi, M.; Sotiriou, C.; Desmedt, C.; et al. Type I interferon/IRF7 axis instigates chemotherapy-induced immunological dormancy in breastc ancer. Oncogene 2019, 38, 2814–2829.
  120. Liang, Q.; Fu, B.; Wu, F.; Li, X.; Yuan, Y.; Zhu, F. ORF45 of Kaposi’s sarcoma-associated herpes virus inhibits phosphorylation of interferon regulatory factor 7 by IKKε and TBK1 as an alternatives ubstrate. J. Virol. 2012, 86, 10162–10172.
  121. Jammi, N.V.; Whitby, L.R.; Beal, P.A. Small molecule inhibitors of the RNA-dependent protein kinase. Biochem. Biophys. Res. Commun. 2003, 308, 50–57.
  122. Sanfilippo, C.; Pinzone, M.R.; Cambria, D.; Longo, A.; Palumbo, M.; DiMarco, R.; Condorelli, F.; Nunnari, G.; Malaguarnera, L.; DiRosa, M. OAS gene family expression is associated with HIV-Related neurocognitive disorders. Mol. Neurobiol. 2018, 55, 1905–1914.
  123. Field, L.L.; Bonnevie-Nielsen, V.; Pociot, F.; Lu, S.; Nielsen, T.B.; Beck-Nielsen, H. OAS1 splice site polymorphism controlling antiviral enzyme activity influences susceptibility to type 1 diabetes. Diabetes 2005, 54, 1588–1591.
  124. Banerjee, S.; Gusho, E.; Gaughan, C.; Dong, B.; Gu, X.; Holvey-Bates, E.; Talukdar, M.; Li, Y.; Weiss, S.R.; Sicheri, F.; et al. OAS-RNase L innate immune pathway mediates the cytotoxicity of a DNA-demethylating drug. Proc. Natl. Acad. Sci. USA 2019, 116, 5071–5076.
  125. Kodigepalli, K.M.; Bowers, K.; Sharp, A.; Nanjundan, M. Roles and regulation of phospholipid scramblases. FEBS Lett. 2015, 589, 3–14.
  126. Tufail, Y.; Cook, D.; Fourgeaudm, L.; Powers, C.J.; Merten, K.; Clark, C.L.; Hoffman, E.; Ngo, A.; Sekiguchi, K.J.; O’Shea, C.C.; et al. Phosphatidyl serine exposure controls viral innate immune responses by microglia. Neuron 2017, 93, 574–586.e8.
  127. Chow, K.T.; Gale, M., Jr.; Loo, Y.M. RIG-I and other RNA sensors in antiviral immunity. Annu. Rev. Immunol. 2018, 36, 667–694.
  128. Kasumba, D.M.; Hajake, T.; Oh, S.W.; Kotenko, S.V.; Kato, H.; Fujita, T. A Plant-derived nucleic acid reconciles type I IFN and a Pyroptotic-like event in immunity against respiratory viruses. J. Immunol. 2017, 199, 2460–2474.
  129. Oberson, A.; Spagnuolo, L.; Puddinu, V.; Barchet, W.; Rittner, K.; Bourquin, C. NAB2 is a novel immune stimulator of MDA-5 that promotes a strong type I interferon response. Oncotarget 2017, 9, 5641–5651.
  130. Johnson, B.; VanBlargan, L.A.; Xu, W.; White, J.P.; Shan, C.; Shi, P.Y.; Zhang, R.; Adhikari, J.; Gross, M.L.; Leung, D.W.; et al. Human IFIT3 modulates IFIT1 RNA binding specificity and protein stability. Immunity 2018, 48, 487–499.e5.
  131. Pidugu, V.K.; Wu, M.M.; Yen, A.H.; Pidugu, H.B.; Chang, K.W.; Liu, C.J.; Lee, T.C. IFIT1 and IFIT3 promote oral squamous cell carcinoma metastasis and contribute to the anti-tumor effect of gefitinib via enhancing p-EGFR recycling. Oncogene 2019, 38, 3232–3247.
  132. Mahauad-Fernandez, W.D.; Okeoma, C.M. B49, a BST-2-based peptide, inhibits adhesion and growth of breast cancer cells. Sci. Rep. 2018, 8, 4305.
  133. Cheng, Y.; Ma, X.L.; Wei, Y.Q.; Wei, X.W. Potential roles and targeted therapy of the CXCLs/CXCR2 axis in cancer and inflammatory diseases. Biochim. Biophys. Acta Rev. Cancer 2019, 1871, 289–312.
  134. Chrétien, I.; Marcuz, A.; Courtet, M.; Katevuo, K.; Vainio, O.; Heath, J.K.; White, S.J.; DuPasquier, L. CTX, a Xenopus thymocyte receptor, defines a molecular family conserved throughout vertebrates. Eur. J. Immunol. 1998, 28, 4094–4104.
  135. Padariya, M.; Kalathiya, U.; Mikac, S.; Dziubek, K.; Tovar Fernandez, M.C.; Sroka, E.; Fahraeus, R.; Sznarkowska, A. Viruses, cancer and non-self recognition. Open Biol. 2021, 11, 200348.
  136. Villarreal, L.P. The source of self: Genetic parasites and the origin of adaptive immunity. Ann. N. Y. Acad. Sci. 2009, 1178, 194–232.
  137. Chuong, E.B.; Elde, N.C.; Feschotte, C. Regulatory evolution of innate immunity through co-option of endogenous retroviruses. Science 2016, 351, 1083–1087.
  138. Didcock, L.; Young, D.F.; Goodbourn, S.; Randall, R.E. The V protein of simianvirus 5 inhibits interferon signalling by targeting STAT1 for proteasome-mediated degradation. J. Virol. 1999, 73, 9928–9933.
  139. Andrejeva, J.; Young, D.F.; Goodbourn, S.; Randall, R.E. Degradation of STAT1 and STAT2 by the V proteins of simianvirus 5 and human parainfluenza virus type 2, respectively: Consequences for virus replication in the presence of alpha/beta and gamma interferons. J. Virol. 2002, 76, 2159–2167.
  140. Look, D.C.; Roswit, W.T.; Frick, A.G.; Gris-Alevy, Y.; Dickhaus, D.M.; Walter, M.J.; Holtzman, M.J. Direct suppression of Stat1 function during adenoviral infection. Immunity 1998, 9, 871–880.
  141. Najarro, P.; Traktman, P.; Lewis, J.A. Vaccinia virus blocks gamma interferon signal transduction: Viral VH1 phosphatase reverses Stat1 activation. J. Virol. 2001, 75, 3185–3196.
  142. Arbiza, J.; Mirazo, S.; Fort, H. Viral quasispecies profiles as the result of the interplay of competition and cooperation. BMC Evol. Biol. 2010, 10, 137.
  143. Ojosnegros, S.; Perales, C.; Mas, A.; Domingo, E. Quasispecies as a matter of fact: Viruses and beyond. Virus Res. 2011, 162, 203–215.
  144. Lauring, A.S.; Andino, R. Quasispecies theory and the behavior of RNA viruses. PLoS Pathog. 2010, 6, e1001005.
  145. Villarreal, L.P.; Witzany, G. Rethinking quasispecies theory: From fittest type to cooperative consortia. World J. Biol. Chem. 2013, 4, 79–90.
  146. Domingo, E.; Perales, C. Viral quasispecies. PLoS Genet. 2019, 15, e1008271.
  147. Huang, W.T.; Lin, C.W. EBV-encoded miR-BART20-5p and miR-BART8 inhibitthe IFN-γ-STAT1 pathway associated with disease progression in nasal NK-cell lymphoma. Am. J. Pathol. 2014, 184, 1185–1197.
  148. Drappier, M.; Michiels, T. Inhibition of the OAS/RNaseL pathway by viruses. Curr. Opin. Virol. 2015, 15, 19–26.
  149. Han, J.Q.; Townsend, H.L.; Jha, B.K.; Paranjape, J.M.; Silverman, R.H.; Barton, D.J. A phylogenetically conserved RNA structure in the poliovirus open reading frame inhibits the antiviral endoribonuclease RNaseL. J. Virol. 2007, 81, 5561–5572.
  150. Han, J.Q.; Barton, D.J. Activation and evasion of the antiviral 2′-5′ oligoadenylate synthetase/ribonucleaseL pathway by hepatitis C virus mRNA. RNA 2002, 8, 512–525.
  151. Min, J.Y.; Krug, R.M. The primary function of RNA binding by the influenza a virus NS1 protein in infected cells: Inhibiting the 2′-5′ oligo(A)synthetase/RNaseL pathway. Proc. Natl. Acad. Sci. USA 2006, 103, 7100–7105.
  152. Chang, H.W.; Watson, J.C.; Jacobs, B.L. The E3L gene of vaccinia virus encodes an inhibitor of the interferon-induced, double-stranded RNA-dependent protein kinase. Proc. Natl. Acad. Sci. USA 1992, 89, 4825–4829.
  153. Huismans, H.; Joklik, W.K. Reovirus-coded polypeptides in infected cells: Isolation of two native monomeric polypeptides with affinity for single-stranded and double-stranded RNA, respectively. Virology 1976, 70, 411–424.
  154. Schröder, H.C.; Ugarković, D.; Wenger, R.; Reuter, P.; Okamoto, T.; Müller, W.E. Binding of Tat protein to TAR region of human immunodeficiency virus type 1 blocks TAR-mediated activation of (2′-5′)oligoadenylate synthetase. AIDS Res. Hum. Retrovir. 1990, 6, 659–672.
  155. Burgess, H.M.; Mohr, I. Cellular 5′-3′ mRNA exonuclease Xrn1 controls double-stranded RNA accumulation and anti-viral responses. Cell Host Microbe 2015, 17, 332–344.
  156. Liu, S.W.; Katsafanas, G.C.; Liu, R.; Wyatt, L.S.; Moss, B. Poxvirus decapping enzymes enhance virulence by preventing the accumulation of dsRNA and the induction of innate antiviral responses. Cell Host Microbe 2015, 17, 320–331.
  157. Zhao, L.; Jha, B.K.; Wu, A.; Elliott, R.; Ziebuhr, J.; Gorbalenya, A.E.; Silverman, R.H.; Weiss, S.R. Antagonism of the interferon-induced OAS-RNaseL pathway by murine coronavirus ns2 protein is required for virus replication and liver pathology. Cell Host Microbe 2012, 11, 607–616.
  158. Zhang, R.; Jha, B.K.; Ogden, K.M.; Dong, B.; Zhao, L.; Elliott, R.; Patton, J.T.; Silverman, R.H.; Weiss, S.R. Homologous 2′,5′-phosphodiesterases from disparate RNA viruses antagonize antiviral innate immunity. Proc. Natl. Acad. Sci. USA 2013, 110, 13114–13119.
  159. Ogden, K.M.; Hu, L.; Jha, B.K.; Sankaran, B.; Weiss, S.R.; Silverman, R.H.; Patton, J.T.; Prasad, B.V. Structural basis for 2′-5′-oligoadenylate binding and enzyme activity of aviral RNaseL antagonist. J. Virol. 2015, 89, 6633–6645.
  160. Cayley, P.J.; Davies, J.A.; McCullagh, K.G.; Kerr, I.M. Activation of the ppp(A2′p)nA system in interferon-treated, herpes simplex virus-infected cells and evidence for novel inhibitors of the ppp(A2′p)nA-dependent RNase. Eur. J. Biochem. 1984, 143, 165–174.
  161. Kato, H.; Takeuchi, O.; Sato, S.; Yoneyama, M.; Yamamoto, M.; Matsui, K.; Uematsu, S.; Jung, A.; Kawai, T.; Ishii, K.J.; et al. Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature 2006, 441, 101–105.
  162. Hornung, V.; Ellegast, J.; Kim, S.; Brzózka, K.; Jung, A.; Kato, H.; Poeck, H.; Akira, S.; Conzelmann, K.K.; Schlee, M.; et al. 5′-Triphosphate RNA is the ligand for RIG-I. Science 2006, 314, 994–997.
  163. Peisley, A.; Lin, C.; Wu, B.; Orme-Johnson, M.; Liu, M.; Walz, T.; Hur, S. Cooperative assembly and dynamic disassembly of MDA5 filaments for viral dsRNA recognition. Proc. Natl. Acad. Sci. USA 2011, 108, 21010–21015.
  164. Gorman, J.A.; Hundhausen, C.; Errett, J.S.; Stone, A.E.; Allenspach, E.J.; Ge, Y.; Arkatkar, T.; Clough, C.; Dai, X.; Khim, S.; et al. The A946T variant of the RNA sensor IFIH1 mediates an interferon program that limits viral infection but increases the risk for autoimmunity. Nat. Immunol. 2017, 18, 744–752.
  165. Leung, D.W.; Shabman, R.S.; Farahbakhsh, M.; Prins, K.C.; Borek, D.M.; Wang, T.; Mühlberger, E.; Basler, C.F.; Amarasinghe, G.K. Structural and functional characterization of Reston Ebola virus VP35 interferon inhibitory domain. J. Mol. Biol. 2010, 399, 347–357.
  166. Ramanan, P.; Edwards, M.R.; Shabman, R.S.; Leung, D.W.; Endlich-Frazier, A.C.; Borek, D.M.; Otwinowski, Z.; Liu, G.; Huh, J.; Basler, C.F.; et al. Structural basis for Marburg virus VP35-mediated immune evasion mechanisms. Proc. Natl. Acad. Sci. USA 2012, 109, 20661–20666.
  167. Motz, C.; Schuhmann, K.M.; Kirchhofer, A.; Moldt, M.; Witte, G.; Conzelmann, K.K.; Hopfner, K.P. Paramyxovirus V proteins disrupt the fold of the RNA sensor MDA5 to inhibit antiviral signaling. Science 2013, 339, 690–693.
  168. Davis, M.E.; Wang, M.K.; Rennick, L.J.; Full, F.; Gableske, S.; Mesman, A.W.; Gringhuis, S.I.; Geijtenbeek, T.B.; Duprex, W.P.; Gack, M.U. Antagonism of the phosphatase PP1 by the measles virus V protein is required for innate immune escape of MDA5. Cell Host Microbe 2014, 16, 19–30.
  169. Chatterjee, S.; Basler, C.F.; Amarasinghe, G.K.; Leung, D.W. Molecular Mechanisms of innate immune inhibition bynon-segmented negative-sense RNA viruses. J. Mol. Biol. 2016, 428, 3467–3482.
  170. Zhuang, T.; Yi, X.; Chen, J.; Kang, P.; Chen, X.; Chen, J.; Cui, T.; Chang, Y.; Ye, Z.; Ni, Q.; et al. Intracellular virus sensor MDA5 exacerbates vitiligo by inducing the secretion of chemokines in keratinocytes under virus invasion. Cell Death Dis. 2020, 11, 453.
  171. Bailey, C.C.; Zhong, G.; Huang, I.C.; Farzan, M. IFITM-family Proteins: The cell’s first line of antiviral defense. Annu. Rev. Virol. 2014, 1, 261–283.
  172. Brass, A.L.; Huang, I.C.; Benita, Y.; John, S.P.; Krishnan, M.N.; Feeley, E.M.; Ryan, B.J.; Weyer, J.L.; Vander Weyden, L.; Fikrig, E.; et al. The IFITM proteins mediate cellular resistance to influenza AH1N1 virus, West Nilevirus, and denguevirus. Cell 2009, 139, 1243–1254.
  173. Huang, I.C.; Bailey, C.C.; Weyer, J.L.; Radoshitzky, S.R.; Becker, M.M.; Chiang, J.J.; Brass, A.L.; Ahmed, A.A.; Chi, X.; Dong, L.; et al. Distinct patterns of IFITM-mediated restriction of filoviruses, SARS coronavirus, and influenza A virus. PLoS Pathog. 2011, 7, e1001258.
  174. Wilkins, C.; Woodward, J.; Lau, D.T.; Barnes, A.; Joyce, M.; McFarlane, N.; McKeating, J.A.; Tyrrell, D.L.; Gale, M., Jr. IFITM1 is a tight junction protein that inhibit shepatitis C virus entry. Hepatology 2013, 57, 461–469.
  175. Habjan, M.; Hubel, P.; Lacerda, L.; Benda, C.; Holze, C.; Eberl, C.H.; Mann, A.; Kindler, E.; Gil-Cruz, C.; Ziebuhr, J.; et al. Sequestration by IFIT1 impairs translation of 2′O-unmethylated capped RNA. PLoS Pathog. 2013, 9, e1003663.
  176. Hyde, J.L.; Gardner, C.L.; Kimura, T.; White, J.P.; Liu, G.; Trobaugh, D.W.; Huang, C.; Tonelli, M.; Paessler, S.; Takeda, K.; et al. A viral RNA structural element alters host recognition of nonself RNA. Science 2014, 343, 783–787.
  177. Kumar, P.; Sweeney, T.R.; Skabkin, M.A.; Skabkina, O.V.; Hellen, C.U.; Pestova, T.V. Inhibition of translation by IFIT family members is determined by their ability to interact selectively with the 5′-terminal regions of cap0-, cap1- and 5′ppp-mRNAs. Nucleic Acids Res. 2014, 42, 3228–3245.
  178. Diamond, M.S. IFIT1: A dual sens or and effect or molecule that detects non-2′-Omethylated viral RNA and inhibits its translation. Cytokine Growth Factor Rev. 2014, 25, 543–550.
  179. Wang, C.; Pflugheber, J.; Sumpter, R., Jr.; Sodora, D.L.; Hui, D.; Sen, G.C.; Gale, M., Jr. Alpha interferon induces distinct translational control programs to suppress hepatitis C virus RNA replication. J. Virol. 2003, 77, 3898–3912.
  180. Pichlmair, A.; Lassnig, C.; Eberle, C.A.; Górna, M.W.; Baumann, C.L.; Burkard, T.R.; Bürckstümmer, T.; Stefanovic, A.; Krieger, S.; Bennett, K.L.; et al. IFIT1 is an antiviral protein that recognizes 5′-triphosphate RNA. Nat. Immunol. 2011, 12, 624–630.
  181. Bouloy, M.; Plotch, S.J.; Krug, R.M. Globin mRNAs are primers for the transcription of influenza viral RNA in vitro. Proc. Natl. Acad. Sci. USA 1978, 75, 4886–4890.
  182. Plotch, S.J.; Bouloy, M.; Ulmanen, I.; Krug, R.M. A unique cap(m7GpppXm)-dependent influenza virion endonuclease cleaves capped RNAs to generate the primers that initiate viral RNA transcription. Cell 1981, 23, 847–858.
  183. Pyper, J.M.; Clements, J.E.; Zink, M.C. The nucleolusis the site of Borna disease virus RNA transcription and replication. J. Virol. 1998, 72, 7697–7702.
  184. Reynaud, J.M.; Kim, D.Y.; Atasheva, S.; Rasalouskaya, A.; White, J.P.; Diamond, M.S.; Weaver, S.C.; Frolova, E.I.; Frolov, I. IFIT1 Differentially Interferes with translation and replication of alpha virus genomes and promotes induction of Type I interferon. PLoS Pathog. 2015, 11, e1004863.
  185. Li, S.; Labrecque, S.; Gauzzi, M.C.; Cuddihy, A.R.; Wong, A.H.; Pellegrini, S.; Matlashewski, G.J.; Koromilas, A.E. The human papillomavirus(HPV)-18E6 oncoprotein physically associates with Tyk2 and impairs Jak-STAT activation by interferon-alpha. Oncogene 1999, 18, 5727–5737.
  186. Zhou, C.; Tuong, Z.K.; Frazer, I.H. Papillomavirus immune evasion strategies target the infected cell and the local immune system. Front. Oncol. 2019, 9, 682.
  187. Chang, Y.E.; Laimins, L.A. Microarray analysis identifies interferon-inducible genes and Stat-1 as major transcriptional targets of human papillomavirus type 31. J. Virol. 2000, 74, 4174–4182.
  188. Hong, S.; Mehta, K.P.; Laimins, L.A. Suppression of STAT-1 expression by human papillomaviruses is necessary for differentiation-dependent genome amplification and plasmid maintenance. J. Virol. 2011, 85, 9486–9494.
  189. Nees, M.; Geoghegan, J.M.; Hyman, T.; Frank, S.; Miller, L.; Woodworth, C.D. Papillomavirus type 16 oncogenes downregulate expression of interferon-responsive genes and upregulate proliferation-associated and NF-kappaB-responsive genes in cervical keratinocytes. J. Virol. 2001, 75, 4283–4296.
  190. Barnard, P.; McMillan, N.A. The human papillomavirus E7 oncoprotein abrogates signaling mediated by interferon-alpha. Virology 1999, 259, 305–313.
  191. Barnard, P.; Payne, E.; McMillan, N.A. The human papillomavirus E7 protein is able to inhibit the antiviral and anti-growth functions of interferon-alpha. Virology 2000, 277, 411–419.
  192. Boccardo, E.; Lepique, A.P.; Villa, L.L. The role of inflammation in HPV carcinogenesis. Carcinogenesis 2010, 31, 1905–1912.
  193. Schneider, A.; Papendick, U.; Gissmann, L.; DeVilliers, E.M. Interferon treatment of human genital papillomavirus infection: Importance of viral type. Int. J. Cancer 1987, 40, 610–614.
  194. Rincon-Orozco, B.; Halec, G.; Rosenberger, S.; Muschik, D.; Nindl, I.; Bachmann, A.; Ritter, T.M.; Dondog, B.; Ly, R.; Bosch, F.X.; et al. Epigenetic silencing of interferon-kappa in human papillomavirus type 16-positive cells. Cancer Res. 2009, 69, 8718–8725.
More
Video Production Service