#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Activation of Type I and III Interferon Response by Mitochondrial and Peroxisomal MAVS and Inhibition by Hepatitis C Virus


Mammalian cells developed several defense mechanisms against viral infection. One major strategy involves pattern recognition receptors (PRRs) recognizing non-self motifs in viral RNA and triggering the production of type I and III interferon (IFN) that induce an antiviral state. One central signaling molecule in this cascade is MAVS (Mitochondrial Antiviral Signaling protein), residing on mitochondria, mitochondria-associated membranes of the endoplasmic reticulum, and peroxisomes. Here we characterized the role of mitochondrial and peroxisomal MAVS for the activation of the IFN response and their counteraction by the hepatitis C virus (HCV), a major causative agent of chronic liver disease with a high propensity to establish persistence. By using various functional and genetic knock-out cell systems reconstituted to express exclusively mitochondrial or peroxisomal MAVS, we observed comparable activation of type I and III IFN response by either MAVS species. In addition, we found that the HCV protease residing in nonstructural protein 3 (NS3) efficiently cleaves MAVS independent from its subcellular localization. This cleavage is required for suppression of the IFN response and might contribute to HCV persistence. Our results indicate a largely localization-independent activation of the IFN response by MAVS in hepatocytes and its efficient counteraction by the HCV NS3 protease.


Vyšlo v časopise: Activation of Type I and III Interferon Response by Mitochondrial and Peroxisomal MAVS and Inhibition by Hepatitis C Virus. PLoS Pathog 11(11): e32767. doi:10.1371/journal.ppat.1005264
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1005264

Souhrn

Mammalian cells developed several defense mechanisms against viral infection. One major strategy involves pattern recognition receptors (PRRs) recognizing non-self motifs in viral RNA and triggering the production of type I and III interferon (IFN) that induce an antiviral state. One central signaling molecule in this cascade is MAVS (Mitochondrial Antiviral Signaling protein), residing on mitochondria, mitochondria-associated membranes of the endoplasmic reticulum, and peroxisomes. Here we characterized the role of mitochondrial and peroxisomal MAVS for the activation of the IFN response and their counteraction by the hepatitis C virus (HCV), a major causative agent of chronic liver disease with a high propensity to establish persistence. By using various functional and genetic knock-out cell systems reconstituted to express exclusively mitochondrial or peroxisomal MAVS, we observed comparable activation of type I and III IFN response by either MAVS species. In addition, we found that the HCV protease residing in nonstructural protein 3 (NS3) efficiently cleaves MAVS independent from its subcellular localization. This cleavage is required for suppression of the IFN response and might contribute to HCV persistence. Our results indicate a largely localization-independent activation of the IFN response by MAVS in hepatocytes and its efficient counteraction by the HCV NS3 protease.


Zdroje

1. Pichlmair A, Schulz O, Tan CP, Naslund TI, Liljestrom P, Weber F, et al. RIG-I-mediated antiviral responses to single-stranded RNA bearing 5'-phosphates. Science. 2006;314(5801):997–1001. doi: 10.1126/science.1132998 17038589.

2. Yoneyama M, Kikuchi M, Natsukawa T, Shinobu N, Imaizumi T, Miyagishi M, et al. The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nature immunology. 2004;5(7):730–7. doi: 10.1038/ni1087 15208624.

3. Kato H, Takeuchi O, Sato S, Yoneyama M, Yamamoto M, Matsui K, et al. Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nature. 2006;441(7089):101–5. doi: 10.1038/nature04734 16625202.

4. Sun L, Wu J, Du F, Chen X, Chen ZJ. Cyclic GMP-AMP synthase is a cytosolic DNA sensor that activates the type I interferon pathway. Science. 2013;339(6121):786–91. doi: 10.1126/science.1232458 23258413; PubMed Central PMCID: PMC3863629.

5. Wu J, Sun L, Chen X, Du F, Shi H, Chen C, et al. Cyclic GMP-AMP is an endogenous second messenger in innate immune signaling by cytosolic DNA. Science. 2013;339(6121):826–30. doi: 10.1126/science.1229963 23258412; PubMed Central PMCID: PMC3855410.

6. Pichlmair A, Schulz O, Tan CP, Rehwinkel J, Kato H, Takeuchi O, et al. Activation of MDA5 requires higher-order RNA structures generated during virus infection. Journal of virology. 2009;83(20):10761–9. doi: 10.1128/JVI.00770-09 19656871; PubMed Central PMCID: PMC2753146.

7. Baum A, Sachidanandam R, Garcia-Sastre A. Preference of RIG-I for short viral RNA molecules in infected cells revealed by next-generation sequencing. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(37):16303–8. doi: 10.1073/pnas.1005077107 20805493; PubMed Central PMCID: PMC2941304.

8. Binder M, Eberle F, Seitz S, Mucke N, Huber CM, Kiani N, et al. Molecular mechanism of signal perception and integration by the innate immune sensor retinoic acid-inducible gene-I (RIG-I). The Journal of biological chemistry. 2011;286(31):27278–87. doi: 10.1074/jbc.M111.256974 21659521; PubMed Central PMCID: PMC3149321.

9. Seth RB, Sun L, Ea CK, Chen ZJ. Identification and characterization of MAVS, a mitochondrial antiviral signaling protein that activates NF-kappaB and IRF 3. Cell. 2005;122(5):669–82. doi: 10.1016/j.cell.2005.08.012 16125763.

10. Meylan E, Curran J, Hofmann K, Moradpour D, Binder M, Bartenschlager R, et al. Cardif is an adaptor protein in the RIG-I antiviral pathway and is targeted by hepatitis C virus. Nature. 2005;437(7062):1167–72. doi: 10.1038/nature04193 16177806.

11. Xu LG, Wang YY, Han KJ, Li LY, Zhai Z, Shu HB. VISA is an adapter protein required for virus-triggered IFN-beta signaling. Molecular cell. 2005;19(6):727–40. doi: 10.1016/j.molcel.2005.08.014 16153868.

12. Kawai T, Takahashi K, Sato S, Coban C, Kumar H, Kato H, et al. IPS-1, an adaptor triggering RIG-I- and Mda5-mediated type I interferon induction. Nature immunology. 2005;6(10):981–8. doi: 10.1038/ni1243 16127453.

13. Gack MU, Shin YC, Joo CH, Urano T, Liang C, Sun L, et al. TRIM25 RING-finger E3 ubiquitin ligase is essential for RIG-I-mediated antiviral activity. Nature. 2007;446(7138):916–20. doi: 10.1038/nature05732 17392790.

14. Wu B, Hur S. How RIG-I like receptors activate MAVS. Current opinion in virology. 2015;12:91–8. doi: 10.1016/j.coviro.2015.04.004 25942693.

15. Hou F, Sun L, Zheng H, Skaug B, Jiang QX, Chen ZJ. MAVS forms functional prion-like aggregates to activate and propagate antiviral innate immune response. Cell. 2011;146(3):448–61. doi: 10.1016/j.cell.2011.06.041 21782231; PubMed Central PMCID: PMC3179916.

16. Loo YM, Fornek J, Crochet N, Bajwa G, Perwitasari O, Martinez-Sobrido L, et al. Distinct RIG-I and MDA5 signaling by RNA viruses in innate immunity. Journal of virology. 2008;82(1):335–45. doi: 10.1128/JVI.01080-07 17942531; PubMed Central PMCID: PMC2224404.

17. Yoneyama M, Kikuchi M, Matsumoto K, Imaizumi T, Miyagishi M, Taira K, et al. Shared and unique functions of the DExD/H-box helicases RIG-I, MDA5, and LGP2 in antiviral innate immunity. Journal of immunology. 2005;175(5):2851–8. 16116171.

18. Kotenko SV, Gallagher G, Baurin VV, Lewis-Antes A, Shen M, Shah NK, et al. IFN-lambdas mediate antiviral protection through a distinct class II cytokine receptor complex. Nature immunology. 2003;4(1):69–77. doi: 10.1038/ni875 12483210.

19. Sheppard P, Kindsvogel W, Xu W, Henderson K, Schlutsmeyer S, Whitmore TE, et al. IL-28, IL-29 and their class II cytokine receptor IL-28R. Nature immunology. 2003;4(1):63–8. doi: 10.1038/ni873 12469119.

20. Prokunina-Olsson L, Muchmore B, Tang W, Pfeiffer RM, Park H, Dickensheets H, et al. A variant upstream of IFNL3 (IL28B) creating a new interferon gene IFNL4 is associated with impaired clearance of hepatitis C virus. Nature genetics. 2013;45(2):164–71. doi: 10.1038/ng.2521 23291588; PubMed Central PMCID: PMC3793390.

21. Ank N, West H, Bartholdy C, Eriksson K, Thomsen AR, Paludan SR. Lambda interferon (IFN-lambda), a type III IFN, is induced by viruses and IFNs and displays potent antiviral activity against select virus infections in vivo. Journal of virology. 2006;80(9):4501–9. doi: 10.1128/JVI.80.9.4501-4509.2006 16611910; PubMed Central PMCID: PMC1472004.

22. Park H, Serti E, Eke O, Muchmore B, Prokunina-Olsson L, Capone S, et al. IL-29 is the dominant type III interferon produced by hepatocytes during acute hepatitis C virus infection. Hepatology. 2012;56(6):2060–70. doi: 10.1002/hep.25897 22706965; PubMed Central PMCID: PMC3581145.

23. Marcello T, Grakoui A, Barba-Spaeth G, Machlin ES, Kotenko SV, MacDonald MR, et al. Interferons alpha and lambda inhibit hepatitis C virus replication with distinct signal transduction and gene regulation kinetics. Gastroenterology. 2006;131(6):1887–98. doi: 10.1053/j.gastro.2006.09.052 17087946.

24. Laidlaw SM, Dustin LB. Interferon lambda: opportunities, risks, and uncertainties in the fight against HCV. Frontiers in immunology. 2014;5:545. doi: 10.3389/fimmu.2014.00545 25400636; PubMed Central PMCID: PMC4215632.

25. Sommereyns C, Paul S, Staeheli P, Michiels T. IFN-lambda (IFN-lambda) is expressed in a tissue-dependent fashion and primarily acts on epithelial cells in vivo. PLoS pathogens. 2008;4(3):e1000017. doi: 10.1371/journal.ppat.1000017 18369468; PubMed Central PMCID: PMC2265414.

26. Dickensheets H, Sheikh F, Park O, Gao B, Donnelly RP. Interferon-lambda (IFN-lambda) induces signal transduction and gene expression in human hepatocytes, but not in lymphocytes or monocytes. Journal of leukocyte biology. 2013;93(3):377–85. doi: 10.1189/jlb.0812395 23258595; PubMed Central PMCID: PMC3579021.

27. Zhou Z, Hamming OJ, Ank N, Paludan SR, Nielsen AL, Hartmann R. Type III interferon (IFN) induces a type I IFN-like response in a restricted subset of cells through signaling pathways involving both the Jak-STAT pathway and the mitogen-activated protein kinases. Journal of virology. 2007;81(14):7749–58. doi: 10.1128/JVI.02438-06 17507495; PubMed Central PMCID: PMC1933366.

28. Jacobson IM, Davis GL, El-Serag H, Negro F, Trepo C. Prevalence and challenges of liver diseases in patients with chronic hepatitis C virus infection. Clinical gastroenterology and hepatology: the official clinical practice journal of the American Gastroenterological Association. 2010;8(11):924–33; quiz e117. doi: 10.1016/j.cgh.2010.06.032 20713178.

29. Perz JF, Armstrong GL, Farrington LA, Hutin YJ, Bell BP. The contributions of hepatitis B virus and hepatitis C virus infections to cirrhosis and primary liver cancer worldwide. Journal of hepatology. 2006;45(4):529–38. doi: 10.1016/j.jhep.2006.05.013 16879891.

30. Bartenschlager R, Lohmann V, Penin F. The molecular and structural basis of advanced antiviral therapy for hepatitis C virus infection. Nature reviews Microbiology. 2013;11(7):482–96. doi: 10.1038/nrmicro3046 23748342.

31. Foy E, Li K, Sumpter R Jr., Loo YM, Johnson CL, Wang C, et al. Control of antiviral defenses through hepatitis C virus disruption of retinoic acid-inducible gene-I signaling. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(8):2986–91. doi: 10.1073/pnas.0408707102 15710892; PubMed Central PMCID: PMC549461.

32. Horner SM, Liu HM, Park HS, Briley J, Gale M Jr. Mitochondrial-associated endoplasmic reticulum membranes (MAM) form innate immune synapses and are targeted by hepatitis C virus. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(35):14590–5. doi: 10.1073/pnas.1110133108 21844353; PubMed Central PMCID: PMC3167523.

33. Bellecave P, Sarasin-Filipowicz M, Donze O, Kennel A, Gouttenoire J, Meylan E, et al. Cleavage of mitochondrial antiviral signaling protein in the liver of patients with chronic hepatitis C correlates with a reduced activation of the endogenous interferon system. Hepatology. 2010;51(4):1127–36. doi: 10.1002/hep.23426 20044805.

34. Dixit E, Boulant S, Zhang Y, Lee AS, Odendall C, Shum B, et al. Peroxisomes are signaling platforms for antiviral innate immunity. Cell. 2010;141(4):668–81. doi: 10.1016/j.cell.2010.04.018 20451243; PubMed Central PMCID: PMC3670185.

35. Odendall C, Dixit E, Stavru F, Bierne H, Franz KM, Durbin AF, et al. Diverse intracellular pathogens activate type III interferon expression from peroxisomes. Nature immunology. 2014;15(8):717–26. doi: 10.1038/ni.2915 24952503; PubMed Central PMCID: PMC4106986.

36. Lin R, Lacoste J, Nakhaei P, Sun Q, Yang L, Paz S, et al. Dissociation of a MAVS/IPS-1/VISA/Cardif-IKKepsilon molecular complex from the mitochondrial outer membrane by hepatitis C virus NS3-4A proteolytic cleavage. Journal of virology. 2006;80(12):6072–83. doi: 10.1128/JVI.02495-05 16731946; PubMed Central PMCID: PMC1472616.

37. Loo YM, Owen DM, Li K, Erickson AK, Johnson CL, Fish PM, et al. Viral and therapeutic control of IFN-beta promoter stimulator 1 during hepatitis C virus infection. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(15):6001–6. doi: 10.1073/pnas.0601523103 16585524; PubMed Central PMCID: PMC1458687.

38. Jones CT, Catanese MT, Law LM, Khetani SR, Syder AJ, Ploss A, et al. Real-time imaging of hepatitis C virus infection using a fluorescent cell-based reporter system. Nature biotechnology. 2010;28(2):167–71. doi: 10.1038/nbt.1604 20118917; PubMed Central PMCID: PMC2828266.

39. Bartenschlager R, Ahlborn-Laake L, Mous J, Jacobsen H. Nonstructural protein 3 of the hepatitis C virus encodes a serine-type proteinase required for cleavage at the NS3/4 and NS4/5 junctions. Journal of virology. 1993;67(7):3835–44. 8389908; PubMed Central PMCID: PMC237748.

40. Cheng G, Zhong J, Chisari FV. Inhibition of dsRNA-induced signaling in hepatitis C virus-infected cells by NS3 protease-dependent and -independent mechanisms. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(22):8499–504. doi: 10.1073/pnas.0602957103 16707574; PubMed Central PMCID: PMC1482521.

41. Oshiumi H, Miyashita M, Matsumoto M, Seya T. A distinct role of Riplet-mediated K63-Linked polyubiquitination of the RIG-I repressor domain in human antiviral innate immune responses. PLoS pathogens. 2013;9(8):e1003533. doi: 10.1371/journal.ppat.1003533 23950712; PubMed Central PMCID: PMC3738492.

42. Baril M, Racine ME, Penin F, Lamarre D. MAVS dimer is a crucial signaling component of innate immunity and the target of hepatitis C virus NS3/4A protease. Journal of virology. 2009;83(3):1299–311. doi: 10.1128/JVI.01659-08 19036819; PubMed Central PMCID: PMC2620913.

43. Mi Z, Ma Y, Tong Y. Avian influenza virus H5N1 induces rapid interferon-beta production but shows more potent inhibition to retinoic acid-inducible gene I expression than H1N1 in vitro. Virology journal. 2012;9:145. doi: 10.1186/1743-422X-9-145 22862800; PubMed Central PMCID: PMC3464129.

44. Abe K, Ishigami T, Shyu AB, Ohno S, Umemura S, Yamashita A. Analysis of interferon-beta mRNA stability control after poly(I:C) stimulation using RNA metabolic labeling by ethynyluridine. Biochemical and biophysical research communications. 2012;428(1):44–9. doi: 10.1016/j.bbrc.2012.09.144 23063848; PubMed Central PMCID: PMC3672056.

45. Jiang Z, Georgel P, Du X, Shamel L, Sovath S, Mudd S, et al. CD14 is required for MyD88-independent LPS signaling. Nature immunology. 2005;6(6):565–70. doi: 10.1038/ni1207 15895089.

46. Dodt G, Braverman N, Wong C, Moser A, Moser HW, Watkins P, et al. Mutations in the PTS1 receptor gene, PXR1, define complementation group 2 of the peroxisome biogenesis disorders. Nature genetics. 1995;9(2):115–25. doi: 10.1038/ng0295-115 7719337.

47. Muntau AC, Roscher AA, Kunau WH, Dodt G. The interaction between human PEX3 and PEX19 characterized by fluorescence resonance energy transfer (FRET) analysis. European journal of cell biology. 2003;82(7):333–42. doi: 10.1078/0171-9335-00325 12924628.

48. Agrawal G, Subramani S. Emerging role of the endoplasmic reticulum in peroxisome biogenesis. Frontiers in physiology. 2013;4:286. doi: 10.3389/fphys.2013.00286 24115935; PubMed Central PMCID: PMC3792350.

49. Shalem O, Sanjana NE, Hartenian E, Shi X, Scott DA, Mikkelsen TS, et al. Genome-scale CRISPR-Cas9 knockout screening in human cells. Science. 2014;343(6166):84–7. doi: 10.1126/science.1247005 24336571; PubMed Central PMCID: PMC4089965.

50. Binder M, Kochs G, Bartenschlager R, Lohmann V. Hepatitis C virus escape from the interferon regulatory factor 3 pathway by a passive and active evasion strategy. Hepatology. 2007;46(5):1365–74. doi: 10.1002/hep.21829 17668876.

51. Fujiki Y, Rachubinski RA, Lazarow PB. Synthesis of a major integral membrane polypeptide of rat liver peroxisomes on free polysomes. Proceedings of the National Academy of Sciences of the United States of America. 1984;81(22):7127–31. 6594687; PubMed Central PMCID: PMC392090.

52. Suzuki Y, Orii T, Takiguchi M, Mori M, Hijikata M, Hashimoto T. Biosynthesis of membrane polypeptides of rat liver peroxisomes. Journal of biochemistry. 1987;101(2):491–6. 3584097.

53. Imanaka T, Shiina Y, Takano T, Hashimoto T, Osumi T. Insertion of the 70-kDa peroxisomal membrane protein into peroxisomal membranes in vivo and in vitro. The Journal of biological chemistry. 1996;271(7):3706–13. 8631984.

54. Hoepfner D, Schildknegt D, Braakman I, Philippsen P, Tabak HF. Contribution of the endoplasmic reticulum to peroxisome formation. Cell. 2005;122(1):85–95. doi: 10.1016/j.cell.2005.04.025 16009135.

55. Neuspiel M, Schauss AC, Braschi E, Zunino R, Rippstein P, Rachubinski RA, et al. Cargo-selected transport from the mitochondria to peroxisomes is mediated by vesicular carriers. Current biology: CB. 2008;18(2):102–8. doi: 10.1016/j.cub.2007.12.038 18207745.

56. Jones JM, Morrell JC, Gould SJ. PEX19 is a predominantly cytosolic chaperone and import receptor for class 1 peroxisomal membrane proteins. The Journal of cell biology. 2004;164(1):57–67. doi: 10.1083/jcb.200304111 14709540; PubMed Central PMCID: PMC2171958.

57. Kinoshita N, Ghaedi K, Shimozawa N, Wanders RJ, Matsuzono Y, Imanaka T, et al. Newly identified Chinese hamster ovary cell mutants are defective in biogenesis of peroxisomal membrane vesicles (Peroxisomal ghosts), representing a novel complementation group in mammals. The Journal of biological chemistry. 1998;273(37):24122–30. 9727033.

58. Muntau AC, Mayerhofer PU, Paton BC, Kammerer S, Roscher AA. Defective peroxisome membrane synthesis due to mutations in human PEX3 causes Zellweger syndrome, complementation group G. American journal of human genetics. 2000;67(4):967–75. doi: 10.1086/303071 10958759; PubMed Central PMCID: PMC1287898.

59. Ghaedi K, Honsho M, Shimozawa N, Suzuki Y, Kondo N, Fujiki Y. PEX3 is the causal gene responsible for peroxisome membrane assembly-defective Zellweger syndrome of complementation group G. American journal of human genetics. 2000;67(4):976–81. doi: 10.1086/303086 10968777; PubMed Central PMCID: PMC1287899.

60. Eno CO, Eckenrode EF, Olberding KE, Zhao G, White C, Li C. Distinct roles of mitochondria- and ER-localized Bcl-xL in apoptosis resistance and Ca2+ homeostasis. Molecular biology of the cell. 2012;23(13):2605–18. doi: 10.1091/mbc.E12-02-0090 22573883; PubMed Central PMCID: PMC3386223.

61. Tagami S, Eguchi Y, Kinoshita M, Takeda M, Tsujimoto Y. A novel protein, RTN-XS, interacts with both Bcl-XL and Bcl-2 on endoplasmic reticulum and reduces their anti-apoptotic activity. Oncogene. 2000;19(50):5736–46. doi: 10.1038/sj.onc.1203948 11126360.

62. Hsu YT, Wolter KG, Youle RJ. Cytosol-to-membrane redistribution of Bax and Bcl-X(L) during apoptosis. Proceedings of the National Academy of Sciences of the United States of America. 1997;94(8):3668–72. 9108035; PubMed Central PMCID: PMC20498.

63. Bauhofer O, Ruggieri A, Schmid B, Schirmacher P, Bartenschlager R. Persistence of HCV in quiescent hepatic cells under conditions of an interferon-induced antiviral response. Gastroenterology. 2012;143(2):429–38 e8. Epub 2012/04/24. doi: 10.1053/j.gastro.2012.04.018 22522091.

64. Ding S, Robek MD. Peroxisomal MAVS activates IRF1-mediated IFN-lambda production. Nature immunology. 2014;15(8):700–1. doi: 10.1038/ni.2924 25045870.

65. Marukian S, Andrus L, Sheahan TP, Jones CT, Charles ED, Ploss A, et al. Hepatitis C virus induces interferon-lambda and interferon-stimulated genes in primary liver cultures. Hepatology. 2011;54(6):1913–23. doi: 10.1002/hep.24580 21800339; PubMed Central PMCID: PMC3219820.

66. Thomas E, Gonzalez VD, Li Q, Modi AA, Chen W, Noureddin M, et al. HCV infection induces a unique hepatic innate immune response associated with robust production of type III interferons. Gastroenterology. 2012;142(4):978–88. doi: 10.1053/j.gastro.2011.12.055 22248663; PubMed Central PMCID: PMC3435150.

67. Hiet MS, Bauhofer O, Zayas M, Roth H, Tanaka Y, Schirmacher P, et al. Control of temporal activation of hepatitis C virus-induced interferon response by domain 2 of nonstructural protein 5A. Journal of hepatology. 2015. doi: 10.1016/j.jhep.2015.04.015 25908268.

68. Sheahan T, Imanaka N, Marukian S, Dorner M, Liu P, Ploss A, et al. Interferon lambda alleles predict innate antiviral immune responses and hepatitis C virus permissiveness. Cell host & microbe. 2014;15(2):190–202. Epub 2014/02/18. doi: 10.1016/j.chom.2014.01.007 24528865; PubMed Central PMCID: PMC4104123.

69. Bolen CR, Ding S, Robek MD, Kleinstein SH. Dynamic expression profiling of type I and type III interferon-stimulated hepatocytes reveals a stable hierarchy of gene expression. Hepatology. 2014;59(4):1262–72. doi: 10.1002/hep.26657 23929627; PubMed Central PMCID: PMC3938553.

70. Onoguchi K, Yoneyama M, Takemura A, Akira S, Taniguchi T, Namiki H, et al. Viral infections activate types I and III interferon genes through a common mechanism. The Journal of biological chemistry. 2007;282(10):7576–81. doi: 10.1074/jbc.M608618200 17204473.

71. Jewell NA, Cline T, Mertz SE, Smirnov SV, Flano E, Schindler C, et al. Lambda interferon is the predominant interferon induced by influenza A virus infection in vivo. Journal of virology. 2010;84(21):11515–22. doi: 10.1128/JVI.01703-09 20739515; PubMed Central PMCID: PMC2953143.

72. Mordstein M, Neugebauer E, Ditt V, Jessen B, Rieger T, Falcone V, et al. Lambda interferon renders epithelial cells of the respiratory and gastrointestinal tracts resistant to viral infections. Journal of virology. 2010;84(11):5670–7. doi: 10.1128/JVI.00272-10 20335250; PubMed Central PMCID: PMC2876583.

73. Pietschmann T, Kaul A, Koutsoudakis G, Shavinskaya A, Kallis S, Steinmann E, et al. Construction and characterization of infectious intragenotypic and intergenotypic hepatitis C virus chimeras. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(19):7408–13. doi: 10.1073/pnas.0504877103 16651538; PubMed Central PMCID: PMC1455439.

74. Reiss S, Rebhan I, Backes P, Romero-Brey I, Erfle H, Matula P, et al. Recruitment and activation of a lipid kinase by hepatitis C virus NS5A is essential for integrity of the membranous replication compartment. Cell host & microbe. 2011;9(1):32–45. doi: 10.1016/j.chom.2010.12.002 21238945; PubMed Central PMCID: PMC3433060.

75. Kaufmann T, Schlipf S, Sanz J, Neubert K, Stein R, Borner C. Characterization of the signal that directs Bcl-x(L), but not Bcl-2, to the mitochondrial outer membrane. The Journal of cell biology. 2003;160(1):53–64. doi: 10.1083/jcb.200210084 12515824; PubMed Central PMCID: PMC2172731.

76. Fransen M, Wylin T, Brees C, Mannaerts GP, Van Veldhoven PP. Human pex19p binds peroxisomal integral membrane proteins at regions distinct from their sorting sequences. Molecular and cellular biology. 2001;21(13):4413–24. doi: 10.1128/MCB.21.13.4413-4424.2001 11390669; PubMed Central PMCID: PMC87101.

77. Siegel R, Eskdale J, Gallagher G. Regulation of IFN-lambda1 promoter activity (IFN-lambda1/IL-29) in human airway epithelial cells. Journal of immunology. 2011;187(11):5636–44. doi: 10.4049/jimmunol.1003988 22058416.

78. Kaul A, Stauffer S, Berger C, Pertel T, Schmitt J, Kallis S, et al. Essential role of cyclophilin A for hepatitis C virus replication and virus production and possible link to polyprotein cleavage kinetics. PLoS pathogens. 2009;5(8):e1000546. doi: 10.1371/journal.ppat.1000546 19680534; PubMed Central PMCID: PMC2718831.

79. Grandvaux N, Servant MJ, tenOever B, Sen GC, Balachandran S, Barber GN, et al. Transcriptional profiling of interferon regulatory factor 3 target genes: direct involvement in the regulation of interferon-stimulated genes. Journal of virology. 2002;76(11):5532–9. 11991981; PubMed Central PMCID: PMC137057.

80. Yoneyama M, Suhara W, Fukuhara Y, Fukuda M, Nishida E, Fujita T. Direct triggering of the type I interferon system by virus infection: activation of a transcription factor complex containing IRF-3 and CBP/p300. The EMBO journal. 1998;17(4):1087–95. doi: 10.1093/emboj/17.4.1087 9463386; PubMed Central PMCID: PMC1170457.

81. Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) Method. Methods. 2001;25(4):402–8. doi: 10.1006/meth.2001.1262 11846609.

82. Schindelin J, Arganda-Carreras I, Frise E, Kaynig V, Longair M, Pietzsch T, et al. Fiji: an open-source platform for biological-image analysis. Nature methods. 2012;9(7):676–82. doi: 10.1038/nmeth.2019 22743772; PubMed Central PMCID: PMC3855844.

83. Heigwer F, Kerr G, Boutros M. E-CRISP: fast CRISPR target site identification. Nature methods. 2014;11(2):122–3. doi: 10.1038/nmeth.2812 24481216.

84. Friebe P, Boudet J, Simorre JP, Bartenschlager R. Kissing-loop interaction in the 3' end of the hepatitis C virus genome essential for RNA replication. Journal of virology. 2005;79(1):380–92. Epub 2004/12/15. doi: 10.1128/JVI.79.1.380-392.2005 15596831; PubMed Central PMCID: PMC538730.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2015 Číslo 11
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#