#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Supporting Role for GTPase Rab27a in Hepatitis C Virus RNA Replication through a Novel miR-122-Mediated Effect


Eukaryotic cells constantly expel a variety of small vesicles that are loaded with proteins, nucleic acids and other small compounds that were produced inside the cell. One particular kind of vesicle is called exosome. Exosomes are initially located in multivesicular compartments inside cells and are docked at the cell surface membrane by the small GTPase Rab27a. In the liver, high expression of Rab27a correlates with the development of hepatocellular carcinoma, suggesting a high trafficking capacity for exosomes. Also, it has been shown that hepatitis C virus (HCV) can spread from cell to cell via exosomes. We discovered that Rab27a abundance affects HCV virion abundance that independent from its role in exosome secretion. The presence of Rab27a in membrane-enriched replication complexes and nearby lipid droplets points to functions of Rab27a in the viral life cycle. Depletion of Rab27a resulted in a lower abundance of the liver-specific microRNA miR-122. It is known that two molecules of miR-122 form an oligomeric complex with the 5’ end of the viral RNA leading to protection of the viral RNA against cellular nucleases. However, we show that the Rab27a-mediated loss of miR-122 was independent of its role in protecting the viral RNA, very likely by the downregulation of a cellular inhibitor of HCV gene expression. These findings argue for novel, hitherto undetected roles for miR-122 in the viral life cycle.


Vyšlo v časopise: Supporting Role for GTPase Rab27a in Hepatitis C Virus RNA Replication through a Novel miR-122-Mediated Effect. PLoS Pathog 11(8): e32767. doi:10.1371/journal.ppat.1005116
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1005116

Souhrn

Eukaryotic cells constantly expel a variety of small vesicles that are loaded with proteins, nucleic acids and other small compounds that were produced inside the cell. One particular kind of vesicle is called exosome. Exosomes are initially located in multivesicular compartments inside cells and are docked at the cell surface membrane by the small GTPase Rab27a. In the liver, high expression of Rab27a correlates with the development of hepatocellular carcinoma, suggesting a high trafficking capacity for exosomes. Also, it has been shown that hepatitis C virus (HCV) can spread from cell to cell via exosomes. We discovered that Rab27a abundance affects HCV virion abundance that independent from its role in exosome secretion. The presence of Rab27a in membrane-enriched replication complexes and nearby lipid droplets points to functions of Rab27a in the viral life cycle. Depletion of Rab27a resulted in a lower abundance of the liver-specific microRNA miR-122. It is known that two molecules of miR-122 form an oligomeric complex with the 5’ end of the viral RNA leading to protection of the viral RNA against cellular nucleases. However, we show that the Rab27a-mediated loss of miR-122 was independent of its role in protecting the viral RNA, very likely by the downregulation of a cellular inhibitor of HCV gene expression. These findings argue for novel, hitherto undetected roles for miR-122 in the viral life cycle.


Zdroje

1. Bartenschlager R, Penin F, Lohmann V, Andre P. Assembly of infectious hepatitis C virus particles. Trends in microbiology. 2011;19(2):95–103. Epub 2010/12/15. doi: 10.1016/j.tim.2010.11.005 21146993.

2. Murray CL, Rice CM. Turning hepatitis C into a real virus. Annual review of microbiology. 2011;65:307–27. doi: 10.1146/annurev-micro-090110-102954 21682640.

3. Jopling CL, Schutz S, Sarnow P. Position-dependent function for a tandem microRNA miR-122-binding site located in the hepatitis C virus RNA genome. Cell host & microbe. 2008;4(1):77–85. Epub 2008/07/16. doi: 10.1016/j.chom.2008.05.013 18621012; PubMed Central PMCID: PMC3519368.

4. Machlin ES, Sarnow P, Sagan SM. Masking the 5' terminal nucleotides of the hepatitis C virus genome by an unconventional microRNA-target RNA complex. Proceedings of the National Academy of Sciences of the United States of America. 2011;108(8):3193–8. Epub 2011/01/12. doi: 10.1073/pnas.1012464108 21220300; PubMed Central PMCID: PMC3044371.

5. Li Y, Masaki T, Yamane D, McGivern DR, Lemon SM. Competing and noncompeting activities of miR-122 and the 5' exonuclease Xrn1 in regulation of hepatitis C virus replication. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(5):1881–6. doi: 10.1073/pnas.1213515110 23248316; PubMed Central PMCID: PMC3562843.

6. Sedano CD, Sarnow P. Hepatitis C Virus Subverts Liver-Specific miR-122 to Protect the Viral Genome from Exoribonuclease Xrn2. Cell host & microbe. 2014;16(2):257–64. doi: 10.1016/j.chom.2014.07.006 25121753.

7. Hoofnagle JH. Course and outcome of hepatitis C. Hepatology. 2002;36(5 Suppl 1):S21–9. Epub 2002/10/31. doi: 10.1053/jhep.2002.36227 12407573.

8. Lavanchy D. Evolving epidemiology of hepatitis C virus. Clinical microbiology and infection: the official publication of the European Society of Clinical Microbiology and Infectious Diseases. 2011;17(2):107–15. Epub 2010/11/26. doi: 10.1111/j.1469-0691.2010.03432.x 21091831.

9. Janssen HL, Reesink HW, Lawitz EJ, Zeuzem S, Rodriguez-Torres M, Patel K, et al. Treatment of HCV infection by targeting microRNA. The New England journal of medicine. 2013;368(18):1685–94. Epub 2013/03/29. doi: 10.1056/NEJMoa1209026 23534542.

10. Paul D, Bartenschlager R. Architecture and biogenesis of plus-strand RNA virus replication factories. World journal of virology. 2013;2(2):32–48. doi: 10.5501/wjv.v2.i2.32 24175228; PubMed Central PMCID: PMC3785047.

11. Romero-Brey I, Bartenschlager R. Membranous replication factories induced by plus-strand RNA viruses. Viruses. 2014;6(7):2826–57. doi: 10.3390/v6072826 25054883; PubMed Central PMCID: PMC4113795.

12. Dreux M, Garaigorta U, Boyd B, Decembre E, Chung J, Whitten-Bauer C, et al. Short-range exosomal transfer of viral RNA from infected cells to plasmacytoid dendritic cells triggers innate immunity. Cell host & microbe. 2012;12(4):558–70. Epub 2012/10/23. doi: 10.1016/j.chom.2012.08.010 23084922; PubMed Central PMCID: PMC3479672.

13. Masciopinto F, Giovani C, Campagnoli S, Galli-Stampino L, Colombatto P, Brunetto M, et al. Association of hepatitis C virus envelope proteins with exosomes. European journal of immunology. 2004;34(10):2834–42. doi: 10.1002/eji.200424887 15368299.

14. Ramakrishnaiah V, Thumann C, Fofana I, Habersetzer F, Pan Q, de Ruiter PE, et al. Exosome-mediated transmission of hepatitis C virus between human hepatoma Huh7.5 cells. Proceedings of the National Academy of Sciences of the United States of America. 2013;110(32):13109–13. doi: 10.1073/pnas.1221899110 23878230; PubMed Central PMCID: PMC3740869.

15. Tamai K, Shiina M, Tanaka N, Nakano T, Yamamoto A, Kondo Y, et al. Regulation of hepatitis C virus secretion by the Hrs-dependent exosomal pathway. Virology. 2012;422(2):377–85. Epub 2011/12/06. doi: 10.1016/j.virol.2011.11.009 22138215.

16. Ostrowski M, Carmo NB, Krumeich S, Fanget I, Raposo G, Savina A, et al. Rab27a and Rab27b control different steps of the exosome secretion pathway. Nature cell biology. 2010;12(1):19–30; sup pp 1–13. Epub 2009/12/08. doi: 10.1038/ncb2000 19966785.

17. Gerber PP, Cabrini M, Jancic C, Paoletti L, Banchio C, von Bilderling C, et al. Rab27a controls HIV-1 assembly by regulating plasma membrane levels of phosphatidylinositol 4,5-bisphosphate. The Journal of cell biology. 2015;209(3):435–52. Epub 2015/05/06. doi: 10.1083/jcb.201409082 25940347; PubMed Central PMCID: PMC4427790.

18. Nagashima S, Jirintai S, Takahashi M, Kobayashi T, Tanggis, Nishizawa T, et al. Hepatitis E virus egress depends on the exosomal pathway, with secretory exosomes derived from multivesicular bodies. The Journal of general virology. 2014. Epub 2014/06/28. doi: 10.1099/vir.0.066910-0 24970738.

19. Bello-Morales R, Crespillo AJ, Fraile-Ramos A, Tabares E, Alcina A, Lopez-Guerrero JA. Role of the small GTPase Rab27a during herpes simplex virus infection of oligodendrocytic cells. BMC microbiology. 2012;12:265. Epub 2012/11/21. doi: 10.1186/1471-2180-12-265 23164453; PubMed Central PMCID: PMC3554593.

20. Tolmachova T, Ramalho JS, Anant JS, Schultz RA, Huxley CM, Seabra MC. Cloning, mapping and characterization of the human RAB27A gene. Gene. 1999;239(1):109–16. Epub 1999/11/26. 10571040.

21. Pan Q, Ramakrishnaiah V, Henry S, Fouraschen S, de Ruiter PE, Kwekkeboom J, et al. Hepatic cell-to-cell transmission of small silencing RNA can extend the therapeutic reach of RNA interference (RNAi). Gut. 2012;61(9):1330–9. Epub 2011/12/27. doi: 10.1136/gutjnl-2011-300449 22198713.

22. Bukong TN, Momen-Heravi F, Kodys K, Bala S, Szabo G. Exosomes from hepatitis C infected patients transmit HCV infection and contain replication competent viral RNA in complex with Ago2-miR122-HSP90. PLoS pathogens. 2014;10(10):e1004424. doi: 10.1371/journal.ppat.1004424 25275643; PubMed Central PMCID: PMC4183590.

23. Liu Z, Zhang X, Yu Q, He JJ. Exosome-associated hepatitis C virus in cell cultures and patient plasma. Biochemical and biophysical research communications. 2014;455(3–4):218–22. doi: 10.1016/j.bbrc.2014.10.146 25449270.

24. Berger KL, Cooper JD, Heaton NS, Yoon R, Oakland TE, Jordan TX, et al. Roles for endocytic trafficking and phosphatidylinositol 4-kinase III alpha in hepatitis C virus replication. Proceedings of the National Academy of Sciences of the United States of America. 2009;106(18):7577–82. Epub 2009/04/21. doi: 10.1073/pnas.0902693106 19376974; PubMed Central PMCID: PMC2678598.

25. Kato T, Date T, Miyamoto M, Furusaka A, Tokushige K, Mizokami M, et al. Efficient replication of the genotype 2a hepatitis C virus subgenomic replicon. Gastroenterology. 2003;125(6):1808–17. Epub 2004/01/16. 14724833.

26. Egger D, Wolk B, Gosert R, Bianchi L, Blum HE, Moradpour D, et al. Expression of hepatitis C virus proteins induces distinct membrane alterations including a candidate viral replication complex. Journal of virology. 2002;76(12):5974–84. Epub 2002/05/22. 12021330; PubMed Central PMCID: PMC136238.

27. Ferraris P, Beaumont E, Uzbekov R, Brand D, Gaillard J, Blanchard E, et al. Sequential biogenesis of host cell membrane rearrangements induced by hepatitis C virus infection. Cellular and molecular life sciences: CMLS. 2013;70(7):1297–306. Epub 2012/11/28. doi: 10.1007/s00018-012-1213-0 23184194.

28. Gosert R, Egger D, Lohmann V, Bartenschlager R, Blum HE, Bienz K, et al. Identification of the hepatitis C virus RNA replication complex in Huh-7 cells harboring subgenomic replicons. Journal of virology. 2003;77(9):5487–92. Epub 2003/04/15. 12692249; PubMed Central PMCID: PMC153965.

29. Romero-Brey I, Merz A, Chiramel A, Lee JY, Chlanda P, Haselman U, et al. Three-dimensional architecture and biogenesis of membrane structures associated with hepatitis C virus replication. PLoS pathogens. 2012;8(12):e1003056. Epub 2012/12/14. doi: 10.1371/journal.ppat.1003056 23236278; PubMed Central PMCID: PMC3516559.

30. Jopling CL, Yi M, Lancaster AM, Lemon SM, Sarnow P. Modulation of hepatitis C virus RNA abundance by a liver-specific MicroRNA. Science. 2005;309(5740):1577–81. doi: 10.1126/science.1113329 16141076.

31. Gatfield D, Le Martelot G, Vejnar CE, Gerlach D, Schaad O, Fleury-Olela F, et al. Integration of microRNA miR-122 in hepatic circadian gene expression. Genes Dev. 2009;23(11):1313–26. Epub 2009/06/03. doi: 10.1101/gad.1781009 19487572; PubMed Central PMCID: PMC2701584.

32. Cox EM, Sagan SM, Mortimer SA, Doudna JA, Sarnow P. Enhancement of hepatitis C viral RNA abundance by precursor miR-122 molecules. RNA. 2013;19(12):1825–32. Epub 2013/10/10. doi: 10.1261/rna.040865.113 24106328; PubMed Central PMCID: PMC3884667.

33. Pegtel DM, Cosmopoulos K, Thorley-Lawson DA, van Eijndhoven MA, Hopmans ES, Lindenberg JL, et al. Functional delivery of viral miRNAs via exosomes. Proceedings of the National Academy of Sciences of the United States of America. 2010;107(14):6328–33. doi: 10.1073/pnas.0914843107 20304794; PubMed Central PMCID: PMC2851954.

34. Miyanari Y, Atsuzawa K, Usuda N, Watashi K, Hishiki T, Zayas M, et al. The lipid droplet is an important organelle for hepatitis C virus production. Nature cell biology. 2007;9(9):1089–97. Epub 2007/08/28. doi: 10.1038/ncb1631 17721513.

35. Vogt DA, Camus G, Herker E, Webster BR, Tsou CL, Greene WC, et al. Lipid droplet-binding protein TIP47 regulates hepatitis C Virus RNA replication through interaction with the viral NS5A protein. PLoS pathogens. 2013;9(4):e1003302. doi: 10.1371/journal.ppat.1003302 23593007; PubMed Central PMCID: PMC3623766.

36. Chen TS, Lai RC, Lee MM, Choo AB, Lee CN, Lim SK. Mesenchymal stem cell secretes microparticles enriched in pre-microRNAs. Nucleic acids research. 2010;38(1):215–24. Epub 2009/10/24. doi: 10.1093/nar/gkp857 19850715; PubMed Central PMCID: PMC2800221.

37. Gibbings DJ, Ciaudo C, Erhardt M, Voinnet O. Multivesicular bodies associate with components of miRNA effector complexes and modulate miRNA activity. Nature cell biology. 2009;11(9):1143–9. Epub 2009/08/18. doi: 10.1038/ncb1929 19684575.

38. Garaigorta U, Heim MH, Boyd B, Wieland S, Chisari FV. Hepatitis C virus (HCV) induces formation of stress granules whose proteins regulate HCV RNA replication and virus assembly and egress. Journal of virology. 2012;86(20):11043–56. Epub 2012/08/03. doi: 10.1128/JVI.07101-11 22855484; PubMed Central PMCID: PMC3457181.

39. Pager CT, Schutz S, Abraham TM, Luo G, Sarnow P. Modulation of hepatitis C virus RNA abundance and virus release by dispersion of processing bodies and enrichment of stress granules. Virology. 2013;435(2):472–84. Epub 2012/11/13. doi: 10.1016/j.virol.2012.10.027 23141719; PubMed Central PMCID: PMC3534916.

40. Luna JM, Scheel TK, Danino T, Shaw KS, Mele A, Fak JJ, et al. Hepatitis C virus RNA functionally sequesters miR-122. Cell. 2015;160(6):1099–110. doi: 10.1016/j.cell.2015.02.025 25768906; PubMed Central PMCID: PMC4386883.

41. Shimakami T, Yamane D, Jangra RK, Kempf BJ, Spaniel C, Barton DJ, et al. Stabilization of hepatitis C virus RNA by an Ago2-miR-122 complex. Proceedings of the National Academy of Sciences of the United States of America. 2012;109(3):941–6. Epub 2012/01/05. doi: 10.1073/pnas.1112263109 22215596; PubMed Central PMCID: PMC3271899.

42. Wilson JA, Zhang C, Huys A, Richardson CD. Human Ago2 is required for efficient microRNA 122 regulation of hepatitis C virus RNA accumulation and translation. Journal of virology. 2011;85(5):2342–50. Epub 2010/12/24. doi: 10.1128/JVI.02046-10 21177824; PubMed Central PMCID: PMC3067765.

43. Zhong J, Gastaminza P, Cheng G, Kapadia S, Kato T, Burton DR, et al. Robust hepatitis C virus infection in vitro. Proceedings of the National Academy of Sciences of the United States of America. 2005;102(26):9294–9. Epub 2005/06/09. doi: 10.1073/pnas.0503596102 15939869; PubMed Central PMCID: PMC1166622.

44. Yi M, Villanueva RA, Thomas DL, Wakita T, Lemon SM. Production of infectious genotype 1a hepatitis C virus (Hutchinson strain) in cultured human hepatoma cells. Proceedings of the National Academy of Sciences of the United States of America. 2006;103(7):2310–5. Epub 2006/02/08. doi: 10.1073/pnas.0510727103 16461899; PubMed Central PMCID: PMC1413728.

45. Schlegel A, Giddings TH Jr., Ladinsky MS, Kirkegaard K. Cellular origin and ultrastructure of membranes induced during poliovirus infection. Journal of virology. 1996;70(10):6576–88. 8794292; PubMed Central PMCID: PMC190698.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2015 Číslo 8
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#