Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

The yin and yang of hepatitis C: synthesis and decay of hepatitis C virus RNA

Key Points

  • The number of hepatitis C virus (HCV) RNA copies present within infected hepatocytes is determined by the rate of viral RNA synthesis in relation to the rates of intracellular HCV RNA decay and the packaging and export of newly replicated genomes in virions. Both viral RNA and viral proteins are typically present at low abundance, and this may contribute to cell survival, viral escape from the immune response and long-term viral persistence.

  • HCV RNA is synthesized by replicase complexes associated with the membranous web, which is a collection of cytoplasmic single- and double-membrane vesicles that are formed as protrusions from the endoplasmic reticulum. Formation of the membranous web is dependent on phosphatidylinositol 4-kinase IIIα and multiple nonstructural HCV proteins, including NS4B and NS5A.

  • Newly synthesized HCV positive-strand RNAs ((+)RNAs) are subject to four possible fates: cycling to ribosomes to direct translation of more viral protein; translocating to lipid droplets for packaging into new virions for export from the cell; remaining within the membranous web to act as a template for synthesis of negative-strand RNA; or being degraded by 5′-to-3′ exoribonuclease 1 (XRN1).

  • A network of variably conserved RNA structures extends across the entire HCV (+)RNA genome, but structure is especially prominent within 3′ and 5′ untranslated regions and an internal cis-acting replication element. RNA–RNA interactions within and between these structures, as well as RNA–protein interactions involving poly(rC)-binding protein 2 and possibly other host proteins, modulate the structure of the genome, including its circularization, and thereby probably regulate genome engagement in different stages of the viral life cycle.

  • HCV infection induces oxidative stress. This leads to cellular lipid peroxidation, which acts in an inhibitory manner on the replicase complex to reduce viral replication, thereby providing a negative feedback loop that helps to maintain viral RNA and protein abundance at low levels within the liver.

  • The microRNA miR-122 is an abundant, evolutionarily conserved liver-specific microRNA. It binds two sites near the 5′ end of the HCV genome in association with Argonaute 2, and it promotes replication of the virus by stabilizing and protecting the genome against XRN1-mediated decay and directly stimulating viral RNA synthesis by reducing the proportion of RNA genomes engaged in protein translation, thereby increasing those available to act as templates in RNA synthesis.

Abstract

Hepatitis C virus (HCV) is an unusual RNA virus that has a striking capacity to persist for the remaining life of the host in the majority of infected individuals. In order to persist, HCV must balance viral RNA synthesis and decay in infected cells. In this Review, we focus on interactions between the positive-sense RNA genome of HCV and the host RNA-binding proteins and microRNAs, and describe how these interactions influence the competing processes of viral RNA synthesis and decay to achieve stable, long-term persistence of the viral genome. Furthermore, we discuss how these processes affect hepatitis C pathogenesis and therapeutic strategies against HCV.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: The hepatitis C virus RNA genome and polyprotein.
Figure 2: The yin of hepatitis C virus: RNA synthesis.
Figure 3: The yang of hepatitis C virus: RNA decay.

Similar content being viewed by others

References

  1. Horner, S. M. & Gale, M. Jr. Regulation of hepatic innate immunity by hepatitis C virus. Nat. Med. 19, 879–888 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Walker, C. M. Adaptive immunity to the hepatitis C virus. Adv. Virus Res. 78, 43–86 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Bartenschlager, R., Lohmann, V. & Penin, F. The molecular and structural basis of advanced antiviral therapy for hepatitis C virus infection. Nat. Rev. Microbiol. 11, 482–496 (2013).

    CAS  PubMed  Google Scholar 

  4. Holmberg, S. D., Spradling, P. R., Moorman, A. C. & Denniston, M. M. Hepatitis C in the United States. N. Engl. J. Med. 368, 1859–1861 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Thomas, D. L. et al. The natural history of hepatitis C virus infection: host, viral, and environmental factors. JAMA 284, 450–456 (2000).

    CAS  PubMed  Google Scholar 

  6. Prokunina-Olsson, L. et al. A variant upstream of IFNL3 (IL28B) creating a new interferon gene IFNL4 is associated with impaired clearance of hepatitis C virus. Nat. Genet. 45, 164–171 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Liang, Y. et al. Visualizing hepatitis C virus infections in human liver by two-photon microscopy. Gastroenterology 137, 1448–1458 (2009).

    CAS  PubMed  Google Scholar 

  8. Kandathil, A. J. et al. Use of laser capture microdissection to map hepatitis C virus-positive hepatocytes in human liver. Gastroenterology 145, 1404–1413 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Wieland, S. et al. Simultaneous detection of hepatitis C virus and interferon stimulated gene expression in infected human liver. Hepatology 59, 2121–2130 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Teixeira, R., Marcos, L. A. & Friedman, S. L. Immunopathogenesis of hepatitis C virus infection and hepatic fibrosis: new insights into antifibrotic therapy in chronic hepatitis C. Hepatol. Res. 37, 579–595 (2007).

    CAS  PubMed  Google Scholar 

  11. Westbrook, R. H. & Dusheiko, G. Natural history of hepatitis C. J. Hepatol. 61, S58–S68 (2014).

    PubMed  Google Scholar 

  12. Walters, K. A. et al. Genomic analysis reveals a potential role for cell cycle perturbation in HCV-mediated apoptosis of cultured hepatocytes. PLoS Pathog. 5, e1000269 (2009).

    PubMed  PubMed Central  Google Scholar 

  13. Kannan, R. P., Hensley, L. L., Evers, L., Lemon, S. M. & McGivern, D. R. Hepatitis C virus infection causes cell cycle arrest at the level of entry to mitosis. J. Virol. 85, 7989–8001 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Yamane, D. et al. Regulation of the hepatitis C virus RNA replicase by endogenous lipid peroxidation. Nat. Med. 20, 927–935 (2014). This paper describes the unique manner in which the HCV replicase is negatively regulated by cellular lipid peroxidation.

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Shimakami, T. et al. Stabilization of hepatitis C virus RNA by an Ago2–miR-122 complex. Proc. Natl Acad. Sci. USA 109, 941–946 (2012).

    CAS  PubMed  Google Scholar 

  16. Zeisel, M. B., Felmlee, D. J. & Baumert, T. F. Hepatitis C virus entry. Curr. Top. Microbiol. Immunol. 369, 87–112 (2013).

    CAS  PubMed  Google Scholar 

  17. Shulla, A. & Randall, G. Hepatitis C virus — host interactions, replication, and viral assembly. Curr. Opin. Virol. 2, 725–732 (2012).

    CAS  PubMed  Google Scholar 

  18. Honda, M., Beard, M. R., Ping, L. H. & Lemon, S. M. A phylogenetically conserved stem-loop structure at the 5′ border of the internal ribosome entry site of hepatitis C virus is required for cap-independent viral translation. J. Virol. 73, 1165–1174 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Friebe, P., Lohmann, V., Krieger, N. & Bartenschlager, R. Sequences in the 5′ nontranslated region of hepatitis C virus required for RNA replication. J. Virol. 75, 12047–12057 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Yi, M. & Lemon, S. M. 3′ nontranslated RNA signals required for replication of hepatitis C virus RNA. J. Virol. 77, 3557–3568 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Yi, M. & Lemon, S. M. Structure–function analysis of the 3′ stem-loop of hepatitis C virus genomic RNA and its role in viral RNA replication. RNA. 9, 331–345 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Friebe, P. & Bartenschlager, R. Role of RNA structures in genome terminal sequences of the hepatitis C virus for replication and assembly. J. Virol. 83, 11989–11995 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Li, Y., Masaki, T., Yamane, D., McGivern, D. R. & Lemon, S. M. Competing and noncompeting activities of miR-122 and the 5′ exonuclease Xrn1 in regulation of hepatitis C virus replication. Proc. Natl Acad. Sci. USA 110, 1881–1886 (2013). References 15 and 23 provide the first direct evidence that miR-122, acting in association with AGO2, stabilizes and protects the HCV RNA genome from XRN1-mediated decay.

    CAS  PubMed  Google Scholar 

  24. Niepmann, M. Hepatitis C virus RNA translation. Curr. Top. Microbiol. Immunol. 369, 143–166 (2013).

    CAS  PubMed  Google Scholar 

  25. Moradpour, D. & Penin, F. Hepatitis C virus proteins: from structure to function. Curr. Top. Microbiol. Immunol. 369, 113–142 (2013).

    CAS  PubMed  Google Scholar 

  26. Lohmann, V. et al. Replication of subgenomic hepatitis C virus RNAs in a hepatoma cell line. Science 285, 110–113 (1999).

    CAS  PubMed  Google Scholar 

  27. Gosert, R. et al. Identification of the hepatitis C virus RNA replication complex in Huh-7 cells harboring subgenomic replicons. J. Virol. 77, 5487–5492 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Chatel-Chaix, L. & Bartenschlager, R. Dengue virus- and hepatitis C virus-induced replication and assembly compartments: the enemy inside — caught in the web. J. Virol. 88, 5907–5911 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Appleby, T. C. et al. Viral replication. Structural basis for RNA replication by the hepatitis C virus polymerase. Science 347, 771–775 (2015).

    CAS  PubMed  Google Scholar 

  30. Romero-Brey, I. et al. Three-dimensional architecture and biogenesis of membrane structures associated with hepatitis C virus replication. PLoS Pathog. 8, e1003056 (2012). This study uses a combination of light microscopy- and electron microscopy-based methods to examine the morphology of the membranous web induced by HCV.

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Ferraris, P., Blanchard, E. & Roingeard, P. Ultrastructural and biochemical analyses of hepatitis C virus-associated host cell membranes. J. Gen. Virol. 91, 2230–2237 (2010).

    CAS  PubMed  Google Scholar 

  32. Paul, D., Hoppe, S., Saher, G., Krijnse-Locker, J. & Bartenschlager, R. Morphological and biochemical characterization of the membranous hepatitis C virus replication compartment. J. Virol. 87, 10612–10627 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Miyanari, Y. et al. Hepatitis C virus non-structural proteins in the probable membranous compartment function in viral genome replication. J. Biol. Chem. 278, 50301–50308 (2003).

    CAS  PubMed  Google Scholar 

  34. Sakamoto, H. et al. Host sphingolipid biosynthesis as a target for hepatitis C virus therapy. Nat. Chem. Biol. 1, 333–337 (2005).

    CAS  PubMed  Google Scholar 

  35. Egger, D. et al. Expression of hepatitis C virus proteins induces distinct membrane alterations including a candidate viral replication complex. J. Virol. 76, 5974–5984 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Berger, K. L., Kelly, S. M., Jordan, T. X., Tartell, M. A. & Randall, G. Hepatitis C virus stimulates the phosphatidylinositol 4-kinase III α-dependent phosphatidylinositol 4-phosphate production that is essential for its replication. J. Virol. 85, 8870–8883 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Reiss, S. et al. The lipid kinase phosphatidylinositol-4 kinase III α regulates the phosphorylation status of hepatitis C virus NS5A. PLoS Pathog. 9, e1003359 (2013). This article describes the role of PI4KIIIα in regulating the phosphorylation of NS5A and the morphogenesis of the membranous web.

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Berger, C. et al. Daclatasvir-like inhibitors of NS5A block early biogenesis of hepatitis C virus-induced membranous replication factories, independent of RNA replication. Gastroenterology 147, 1094–1105 (2014).

    CAS  PubMed  Google Scholar 

  39. McGivern, D. R. et al. Kinetic analyses reveal potent and early blockade of hepatitis C virus assembly by NS5A inhibitors. Gastroenterology 147, 453–462 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Targett-Adams, P. et al. Small molecules targeting hepatitis C virus-encoded NS5A cause subcellular redistribution of their target: insights into compound modes of action. J. Virol. 85, 6353–6368 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Yao, N., Reichert, P., Taremi, S. S., Prosise, W. W. & Weber, P. C. Molecular views of viral polyprotein processing revealed by the crystal structure of the hepatitis C virus bifunctional protease–helicase. Structure 7, 1353–1363 (1999).

    CAS  PubMed  Google Scholar 

  42. Bressanelli, S., Tomei, L., Rey, F. A. & De Francesco, R. Structural analysis of the hepatitis C virus RNA polymerase in complex with ribonucleotides. J. Virol. 76, 3482–3492 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Love, R. A., Brodsky, O., Hickey, M. J., Wells, P. A. & Cronin, C. N. Crystal structure of a novel dimeric form of NS5A domain I protein from hepatitis C virus. J. Virol. 83, 4395–4403 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Tellinghuisen, T. L., Marcotrigiano, J. & Rice, C. M. Structure of the zinc-binding domain of an essential component of the hepatitis C virus replicase. Nature 435, 374–379 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Quinkert, D., Bartenschlager, R. & Lohmann, V. Quantitative analysis of the hepatitis C virus replication complex. J. Virol. 79, 13594–13605 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Yang, F. et al. A major determinant of cyclophilin dependence and cyclosporine susceptibility of hepatitis C virus identified by a genetic approach. PLoS Pathog. 6, e1001118 (2010).

    PubMed  PubMed Central  Google Scholar 

  47. Evans, M. J., Rice, C. M. & Goff, S. P. Phosphorylation of hepatitis C virus nonstructural protein 5A modulates its protein interactions and viral RNA replication. Proc. Natl Acad. Sci. USA 101, 13038–13043 (2004).

    CAS  PubMed  Google Scholar 

  48. Liu, Z., Yang, F., Robotham, J. M. & Tang, H. Critical role of cyclophilin A and its prolyl-peptidyl isomerase activity in the structure and function of the hepatitis C virus replication complex. J. Virol. 83, 6554–6565 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Rosnoblet, C. et al. Hepatitis C virus NS5B and host cyclophilin A share a common binding site on NS5A. J. Biol. Chem. 287, 44249–44260 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Gao, L., Aizaki, H., He, J. W. & Lai, M. M. Interactions between viral nonstructural proteins and host protein hVAP-33 mediate the formation of hepatitis C virus RNA replication complex on lipid raft. J. Virol. 78, 3480–3488 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Hamamoto, I. et al. Human VAP-B is involved in hepatitis C virus replication through interaction with NS5A and NS5B. J. Virol. 79, 13473–13482 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Gallay, P. A. & Lin, K. Profile of alisporivir and its potential in the treatment of hepatitis C. Drug Des. Devel. Ther. 7, 105–115 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Mauger, D. M. et al. The functionally conserved architecture of hepatitis C virus RNA genomes. Proc. Natl Acad. Sci. USA 112, 3692–3697 (2015).

    CAS  PubMed  Google Scholar 

  54. Lukavsky, P. J. Structure and function of HCV IRES domains. Virus Res. 139, 166–171 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Kolykhalov, A. A., Feinstone, S. M. & Rice, C. M. Identification of a highly conserved sequence element at the 3′ terminus of hepatitis C virus genome RNA. J. Virol. 70, 3363–3371 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Kolykhalov, A. A., Mihalik, K., Feinstone, S. M. & Rice, C. M. Hepatitis C virus-encoded enzymatic activities and conserved RNA elements in the 3′ nontranslated region are essential for virus replication in vivo. J. Virol. 74, 2046–2051 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Tuplin, A., Struthers, M., Simmonds, P. & Evans, D. J. A twist in the tail: SHAPE mapping of long-range interactions and structural rearrangements of RNA elements involved in HCV replication. Nucleic Acids Res. 40, 6908–6921 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Romero-Lopez, C., Barroso-Deljesus, A., Garcia-Sacristan, A., Briones, C. & Berzal-Herranz, A. End-to-end crosstalk within the hepatitis C virus genome mediates the conformational switch of the 3′X-tail region. Nucleic Acids Res. 42, 567–582 (2014). This work uses SHAPE studies to document complex, long-range RNA–RNA interactions involving the 5′ and 3′ UTRs and CRE of the HCV genome.

    CAS  PubMed  Google Scholar 

  59. McMullan, L. K. et al. Evidence for a functional RNA element in the hepatitis C virus core gene. Proc. Natl Acad. Sci. USA 20, 2879–2884 (2007).

    Google Scholar 

  60. You, S., Stump, D. D., Branch, A. D. & Rice, C. M. A cis-acting replication element in the sequence encoding the NS5B RNA-dependent RNA polymerase is required for hepatitis C virus RNA replication. J. Virol. 78, 1352–1366 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Diviney, S. et al. A hepatitis C virus cis-acting replication element forms a long-range RNA–RNA interaction with upstream RNA sequences in NS5B. J. Virol. 82, 9008–9022 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Friebe, P., Boudet, J., Simorre, J. P. & Bartenschlager, R. Kissing-loop interaction in the 3′ end of the hepatitis C virus genome essential for RNA replication. J. Virol. 79, 380–392 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. You, S. & Rice, C. M. 3′ RNA elements in hepatitis C virus replication: kissing partners and long poly(U). J. Virol. 82, 184–195 (2008).

    CAS  PubMed  Google Scholar 

  64. Chu, D. et al. Systematic analysis of enhancer and critical cis-acting RNA elements in the protein-encoding region of the hepatitis C virus genome. J. Virol. 87, 5678–5696 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. Romero-Lopez, C., Barroso-Deljesus, A., Garcia-Sacristan, A., Briones, C. & Berzal-Herranz, A. The folding of the hepatitis C virus internal ribosome entry site depends on the 3′-end of the viral genome. Nucleic Acids Res. 40, 11697–11713 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Kim, J. H. et al. A cellular RNA-binding protein enhances internal ribosomal entry site-dependent translation through an interaction downstream of the hepatitis C virus polyprotein initiation codon. Mol. Cell. Biol. 24, 7878–7890 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Ray, U. & Das, S. Interplay between NS3 protease and human La protein regulates translation–replication switch of hepatitis C virus. Sci. Rep. 1, 1 (2011).

    PubMed  PubMed Central  Google Scholar 

  68. Scheller, N. et al. Translation and replication of hepatitis C virus genomic RNA depends on ancient cellular proteins that control mRNA fates. Proc. Natl Acad. Sci. USA 106, 13517–13522 (2009).

    CAS  PubMed  Google Scholar 

  69. Weinlich, S. et al. IGF2BP1 enhances HCV IRES-mediated translation initiation via the 3'UTR. RNA 15, 1528–1542 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Isken, O. et al. Nuclear factors are involved in hepatitis C virus RNA replication. RNA 13, 1675–1692 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Rosenfeld, A. B. & Racaniello, V. R. Hepatitis C virus internal ribosome entry site-dependent translation in Saccharomyces cerevisiae is independent of polypyrimidine tract-binding protein, poly(rC)-binding protein 2, and La protein. J. Virol. 79, 10126–10137 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Wang, L., Jeng, K. S. & Lai, M. M. Poly(C)-binding protein 2 interacts with sequences required for viral replication in the hepatitis C virus (HCV) 5′ untranslated region and directs HCV RNA replication through circularizing the viral genome. J. Virol. 85, 7954–7964 (2011). This paper provides evidence that PCBP2 mediates circularization of the HCV genome through a protein–protein bridge.

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Hashem, Y. et al. Hepatitis-C-virus-like internal ribosome entry sites displace eIF3 to gain access to the 40S subunit. Nature 503, 539–543 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Li, Y., Masaki, T., Shimakami, T. & Lemon, S. M. hnRNP L and NF90 interact with hepatitis C virus 5′ terminal untranslated RNA and promote efficient replication. J. Virol. 88, 7199–7209 (2014).

    PubMed  PubMed Central  Google Scholar 

  75. Gontarek, R. R. et al. hnRNP C and polypyrimidine tract-binding protein specifically interact with the pyrimidine-rich region within the 3′NTR of the HCV RNA genome. Nucleic Acids Res. 27, 1457–1463 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Spangberg, K., Wiklund, L. & Schwartz, S. HuR, a protein implicated in oncogene and growth factor mRNA decay, binds to the 3′ ends of hepatitis C virus RNA of both polarities. Virology 274, 378–390 (2000).

    CAS  PubMed  Google Scholar 

  77. Ito, T. & Lai, M. M. An internal polypyrimidine-tract-binding protein-binding site in the hepatitis C virus RNA attenuates translation, which is relieved by the 3′-untranslated sequence. Virology 254, 288–296 (1999).

    CAS  PubMed  Google Scholar 

  78. Luo, G. Cellular proteins bind to the poly(U) tract of the 3′ untranslated region of hepatitis C virus RNA genome. Virology 256, 105–118 (1999).

    CAS  PubMed  Google Scholar 

  79. Petrik, J., Parker, H. & Alexander, G. J. Human hepatic glyceraldehyde-3-phosphate dehydrogenase binds to the poly(U) tract of the 3′ non-coding region of hepatitis C virus genomic RNA. J. Gen. Virol. 80, 3109–3113 (1999).

    CAS  PubMed  Google Scholar 

  80. Randall, G. et al. Cellular cofactors affecting hepatitis C virus infection and replication. Proc. Natl Acad. Sci. USA 104, 12884–12889 (2007). This investigation consists of a comprehensive RNAi screen of protein factors suspected of interacting with or otherwise regulating HCV RNA replication.

    CAS  PubMed  Google Scholar 

  81. Oakland, T. E., Haselton, K. J. & Randall, G. EWSR1 binds the hepatitis C virus cis-acting replication element and is required for efficient viral replication. J. Virol. 87, 6625–6634 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Li, Q., Pene, V., Krishnamurthy, S., Cha, H. & Liang, T. J. Hepatitis C virus infection activates an innate pathway involving IKK-α in lipogenesis and viral assembly. Nat. Med. 19, 722–729 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  83. Oshiumi, H., Sakai, K., Matsumoto, M. & Seya, T. DEAD/H box 3 (DDX3) helicase binds the RIG-I adaptor IPS-1 to up-regulate IFN-β-inducing potential. Eur. J. Immunol. 40, 940–948 (2010).

    CAS  PubMed  Google Scholar 

  84. Chang, J. et al. miR-122, a mammalian liver-specific microRNA, is processed from hcr mRNA and may downregulate the high affinity cationic amino acid transporter CAT-1. RNA Biol. 1, 106–113 (2004).

    CAS  PubMed  Google Scholar 

  85. Lanford, R. E. et al. Therapeutic silencing of microRNA-122 in primates with chronic hepatitis C virus infection. Science 327, 198–201 (2010). This report details the impact of therapeutic sequestration of miR-122 on HCV replication and the intrahepatic transcriptome in chimpanzees.

    CAS  PubMed  Google Scholar 

  86. Tsai, W. C. et al. MicroRNA-122 plays a critical role in liver homeostasis and hepatocarcinogenesis. J. Clin. Invest. 122, 2884–2897 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  87. Jopling, C. L., Yi, M., Lancaster, A. M., Lemon, S. M. & Sarnow, P. Modulation of hepatitis C virus RNA abundance by a liver-specific microRNA. Science 309, 1577–1581 (2005). This paper demonstrates that miR-122 interacts directly with the HCV 5′ UTR and promotes amplification of the genome.

    CAS  PubMed  Google Scholar 

  88. Jopling, C. L., 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 4, 77–85 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Machlin, E. S., Sarnow, P. & Sagan, S. M. Masking the 5′ terminal nucleotides of the hepatitis C virus genome by an unconventional microRNA–target RNA complex. Proc. Natl Acad. Sci. USA 108, 3193–3198 (2011).

    CAS  PubMed  Google Scholar 

  90. Pang, P. S. et al. Structural map of a microRNA-122:hepatitis C virus complex. J. Virol. 86, 1250–1254 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Shimakami, T. et al. Base pairing between hepatitis C virus RNA and microRNA 122 3′ of its seed sequence is essential for genome stabilization and production of infectious virus. J. Virol. 86, 7372–7383 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Roberts, A. P., Lewis, A. P. & Jopling, C. L. miR-122 activates hepatitis C virus translation by a specialized mechanism requiring particular RNA components. Nucleic Acids Res. 39, 7716–7729 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Luna, J. M. et al. Hepatitis C virus RNA functionally sequesters miR-122. Cell 160, 1099–1110 (2015). This AGO-based CLIP analysis of the HCV genome demonstrates that HCV RNA exerts an miR-122 sponge effect in infected cell cultures.

    CAS  PubMed  PubMed Central  Google Scholar 

  94. Jangra, R. K., Yi, M. & Lemon, S. M. Regulation of hepatitis C virus translation and infectious virus production by the microRNA miR-122. J. Virol. 84, 6615–6625 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  95. Mortimer, S. A. & Doudna, J. A. Unconventional miR-122 binding stabilizes the HCV genome by forming a trimolecular RNA structure. Nucleic Acids Res. 41, 4230–4240 (2013). This study paper describes the trimolecular complex formed by two copies of miR-122 at the 5′ end of the HCV RNA genome.

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Janssen, H. L. et al. Treatment of HCV infection by targeting microRNA. N. Engl. J. Med. 368, 1685–1694 (2013).

    CAS  PubMed  Google Scholar 

  97. Henke, J. I. et al. microRNA-122 stimulates translation of hepatitis C virus RNA. EMBO J. 27, 3300–3310 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  98. Masaki, T. et al. miR-122 stimulates hepatitis C virus RNA synthesis by altering the balance of viral RNAs engaged in replication versus translation. Cell Host Microbe 17, 217–228 (2015). This paper shows that miR-122 directly promotes HCV RNA synthesis in an AGO2- and PCBP2-dependent manner.

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Wilson, J. A., Zhang, C., Huys, A. & Richardson, C. D. Human Ago2 is required for efficient miR-122 regulation of HCV RNA accumulation and translation. J. Virol. 85, 2342–2350 (2011).

    CAS  PubMed  Google Scholar 

  100. Conrad, K. D. et al. microRNA-122 dependent binding of Ago2 protein to hepatitis C virus RNA Is associated with enhanced RNA stability and translation stimulation. PLoS ONE 8, e56272 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Pichlmair, A. et al. RIG-I-mediated antiviral responses to single-stranded RNA bearing 5′ phosphates. Science. 314, 999–1001 (2006).

    Google Scholar 

  102. Sumpter, R. Jr. et al. Regulating intracellular antiviral defense and permissiveness to hepatitis C virus RNA replication through a cellular RNA helicase, RIG-I. J. Virol. 79, 2689–2699 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  103. Li, Y. P., Gottwein, J. M., Scheel, T. K., Jensen, T. B. & Bukh, J. MicroRNA-122 antagonism against hepatitis C virus genotypes 1–6 and reduced efficacy by host RNA insertion or mutations in the HCV 5′ UTR. Proc. Natl Acad. Sci. USA 108, 4991–4996 (2011).

    CAS  PubMed  Google Scholar 

  104. Israelow, B. et al. Hepatitis C virus genetics affects miR-122 requirements and response to miR-122 inhibitors. Nat. Commun. 5, 5408 (2014).

    PubMed  PubMed Central  Google Scholar 

  105. Ottosen, S. et al. In vitro antiviral activity and preclinical and clinical resistance profile of miravirsen, a novel anti-hepatitis C virus therapeutic targeting the human factor miR-122. Antimicrob. Agents Chemother. 59, 599–608 (2015).

    PubMed  Google Scholar 

  106. Thibault, P. A., Huys, A., Dhillon, P. & Wilson, J. A. MicroRNA-122-dependent and -independent replication of Hepatitis C virus in Hep3B human hepatoma cells. Virology 436, 179–190 (2013).

    CAS  PubMed  Google Scholar 

  107. Bai, Y., Zhou, K. & Doudna, J. A. Hepatitis C virus 3′UTR regulates viral translation through direct interactions with the host translation machinery. Nucleic Acids Res. 41, 7861–7874 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Garcia-Sacristan, A. et al. A magnesium-induced RNA conformational switch at the internal ribosome entry site of hepatitis C virus genome visualized by atomic force microscopy. Nucleic Acids Res. 43, 565–580 (2014).

    PubMed  PubMed Central  Google Scholar 

  109. Spangberg, K., Wiklund, L. & Schwartz, S. Binding of the La autoantigen to the hepatitis C virus 3′ untranslated region protects the RNA from rapid degradation in vitro. J. Gen. Virol. 82, 113–120 (2001).

    CAS  PubMed  Google Scholar 

  110. Romero-Lopez, C. & Berzal-Herranz, A. A long-range RNA–RNA interaction between the 5′ and 3′ ends of the HCV genome. RNA 15, 1740–1752 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  111. Quintavalle, M. et al. Hepatitis C virus NS5A is a direct substrate of casein kinase I-α, a cellular kinase identified by inhibitor affinity chromatography using specific NS5A hyperphosphorylation inhibitors. J. Biol. Chem. 282, 5536–5544 (2007).

    CAS  PubMed  Google Scholar 

  112. Ross-Thriepland, D., Mankouri, J. & Harris, M. Serine phosphorylation of the hepatitis C virus NS5A protein controls the establishment of replication complexes. J. Virol. 89, 3123–3135 (2015).

    CAS  PubMed  Google Scholar 

  113. Masaki, T. et al. Involvement of hepatitis C virus NS5A hyperphosphorylation mediated by casein kinase I-α in infectious virus production. J. Virol. 88, 7541–7555 (2014).

    PubMed  PubMed Central  Google Scholar 

  114. Huang, H., Chen, Y. & Ye, J. Inhibition of hepatitis C virus replication by peroxidation of arachidonate and restoration by vitamin E. Proc. Natl Acad. Sci. USA 104, 18666–18670 (2007).

    CAS  PubMed  Google Scholar 

  115. Wang, T. & Weinman, S. A. Interactions between hepatitis C virus and mitochondria: impact on pathogenesis and innate immunity. Curr. Pathobiol. Rep. 1, 179–187 (2013).

    PubMed  PubMed Central  Google Scholar 

  116. Garneau, N. L., Wilusz, J. & Wilusz, C. J. The highways and byways of mRNA decay. Nat. Rev. Mol. Cell Biol. 8, 113–126 (2007).

    CAS  PubMed  Google Scholar 

  117. Song, M. G., Li, Y. & Kiledjian, M. Multiple mRNA decapping enzymes in mammalian cells. Mol. Cell 40, 423–432 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Jones, C. I., Zabolotskaya, M. V. & Newbury, S. F. The 5′→3′ exoribonuclease XRN1/Pacman and its functions in cellular processes and development. Wiley Interdiscip. Rev. RNA 3, 455–468 (2012).

    CAS  PubMed  Google Scholar 

  119. Liu, Q., Greimann, J. C. & Lima, C. D. Reconstitution, activities, and structure of the eukaryotic RNA exosome. Cell 127, 1223–1237 (2006).

    CAS  PubMed  Google Scholar 

  120. Lykke-Andersen, S., Tomecki, R., Jensen, T. H. & Dziembowski, A. The eukaryotic RNA exosome: same scaffold but variable catalytic subunits. RNA Biol. 8, 61–66 (2011).

    CAS  PubMed  Google Scholar 

  121. Wang, Z. & Kiledjian, M. Functional link between the mammalian exosome and mRNA decapping. Cell 107, 751–762 (2001).

    CAS  PubMed  Google Scholar 

  122. van Hoof, A. & Wagner, E. J. A brief survey of mRNA surveillance. Trends Biochem. Sci. 36, 585–592 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  123. Balistreri, G. et al. The host nonsense-mediated mRNA decay pathway restricts mammalian RNA virus replication. Cell Host Microbe 16, 403–411 (2014).

    CAS  PubMed  Google Scholar 

  124. Li, Y., Yamane, D. & Lemon, S. M. Dissecting the roles of the 5′ exoribonucleases Xrn1 and Xrn2 in restricting hepatitis C virus replication. J. Virol. 89, 4857–4865 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  125. Jinek, M., Coyle, S. M. & Doudna, J. A. Coupled 5′ nucleotide recognition and processivity in Xrn1-mediated mRNA decay. Mol. Cell 41, 600–608 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Moon, S. L. et al. XRN1 stalling in the 5′ UTR of hepatitis C virus and bovine viral diarrhea virus is associated with dysregulated host mRNA stability. PLoS Pathog. 11, e1004708 (2015).

    PubMed  PubMed Central  Google Scholar 

  127. Moon, S. L. et al. A noncoding RNA produced by arthropod-borne flaviviruses inhibits the cellular exoribonuclease XRN1 and alters host mRNA stability. RNA 18, 2029–2040 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Sedano, C. D. & Sarnow, P. Hepatitis C virus subverts liver-specific miR-122 to protect the viral genome from exoribonuclease Xrn2. Cell Host Microbe 16, 257–264 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Miki, T. S. & Grosshans, H. The multifunctional RNase XRN2. Biochem. Soc. Trans. 41, 825–830 (2013).

    CAS  PubMed  Google Scholar 

  130. Nagarajan, V. K., Jones, C. I., Newbury, S. F. & Green, P. J. XRN 5′→3′ exoribonucleases: structure, mechanisms and functions. Biochim. Biophys. Acta 1829, 590–603 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Jones, D. M., Domingues, P., Targett-Adams, P. & McLauchlan, J. Comparison of U2OS and Huh-7 cells for identifying host factors that affect hepatitis C virus RNA replication. J. Gen. Virol. 91, 2238–2248 (2010).

    CAS  PubMed  Google Scholar 

  132. Silverman, R. H. Viral encounters with 2′,5′-oligoadenylate synthetase and RNase L during the interferon antiviral response. J. Virol. 81, 12720–12729 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  133. Degols, G., Eldin, P. & Mechti, N. ISG20, an actor of the innate immune response. Biochimie 89, 831–835 (2007).

    CAS  PubMed  Google Scholar 

  134. Chakrabarti, A., Jha, B. K. & Silverman, R. H. New insights into the role of RNase L in innate immunity. J. Interferon Cytokine Res. 31, 49–57 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Kwon, Y. C., Kang, J. I., Hwang, S. B. & Ahn, B. Y. The ribonuclease L-dependent antiviral roles of human 2′,5′-oligoadenylate synthetase family members against hepatitis C virus. FEBS Lett. 587, 156–164 (2013).

    CAS  PubMed  Google Scholar 

  136. Malathi, K. et al. RNase L releases a small RNA from HCV RNA that refolds into a potent PAMP. RNA 16, 2108–2119 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Washenberger, C. L. et al. Hepatitis C virus RNA: dinucleotide frequencies and cleavage by RNase L. Virus Res. 130, 85–95 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Zhou, Z. et al. Antiviral activities of ISG20 in positive-strand RNA virus infections. Virology 409, 175–188 (2011).

    CAS  PubMed  Google Scholar 

  139. Jiang, D. et al. Identification of three interferon-inducible cellular enzymes that inhibit the replication of hepatitis C virus. J. Virol. 82, 1665–1678 (2008).

    CAS  PubMed  Google Scholar 

  140. Lin, R. J. et al. MCPIP1 ribonuclease exhibits broad-spectrum antiviral effects through viral RNA binding and degradation. Nucleic Acids Res. 41, 3314–3326 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  141. Lin, R. J. et al. MCPIP1 suppresses hepatitis C virus replication and negatively regulates virus-induced proinflammatory cytokine responses. J. Immunol. 193, 4159–4168 (2014).

    CAS  PubMed  Google Scholar 

  142. Zhu, Y., Wang, X., Goff, S. P. & Gao, G. Translational repression precedes and is required for ZAP-mediated mRNA decay. EMBO J. 31, 4236–4246 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  143. Shapiro, J. S. et al. Drosha as an interferon-independent antiviral factor. Proc. Natl Acad. Sci. USA 111, 7108–7113 (2014).

    CAS  PubMed  Google Scholar 

  144. Dougherty, J. D., White, J. P. & Lloyd, R. E. Poliovirus-mediated disruption of cytoplasmic processing bodies. J. Virol. 85, 64–75 (2011).

    CAS  PubMed  Google Scholar 

  145. Bigger, C. B. et al. Intrahepatic gene expression during chronic hepatitis C virus infection in chimpanzees. J. Virol. 78, 13779–13792 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  146. Kiledjian, M., Wang, X. & Liebhaber, S. A. Identification of two KH domain proteins in the α-globin mRNP stability complex. EMBO J. 14, 4357–4364 (1995).

    CAS  PubMed  PubMed Central  Google Scholar 

  147. Korf, M., Jarczak, D., Beger, C., Manns, M. P. & Krüger, M. Inhibition of hepatitis C virus translation and subgenomic replication by siRNAs directed against highly conserved HCV sequence and cellular HCV cofactors. J. Hepatol 43, 225–234 (2005).

    CAS  PubMed  Google Scholar 

  148. Guedj, J. et al. Modeling shows that the NS5A inhibitor daclatasvir has two modes of action and yields a shorter estimate of the hepatitis C virus half-life. Proc. Natl Acad. Sci. USA 110, 3991–3996 (2013).

    CAS  PubMed  Google Scholar 

  149. Sohlenius-Sternbeck, A. K. Determination of the hepatocellularity number for human, dog, rabbit, rat and mouse livers from protein concentration measurements. Toxicol. In Vitro 20, 1582–1586 (2006).

    CAS  PubMed  Google Scholar 

  150. Graw, F. et al. Inferring viral dynamics in chronically HCV infected patients from the spatial distribution of infected hepatocytes. PLoS Comput. Biol. 10, e1003934 (2014). This paper describes mathematical modelling of infection events that may explain the spatial distribution of HCV-infected hepatocytes within the liver.

    PubMed  PubMed Central  Google Scholar 

  151. Villordo, S. M. & Gamarnik, A. V. Genome cyclization as strategy for flavivirus RNA replication. Virus Res. 139, 230–239 (2009).

    CAS  PubMed  Google Scholar 

  152. Filomatori, C. V. et al. A 5′ RNA element promotes dengue virus RNA synthesis on a circular genome. Genes Dev. 20, 2238–2249 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  153. Herold, J. & Andino, R. Poliovirus RNA replication requires genome circularization through a protein–protein bridge. Mol. Cell 7, 581–591 (2001).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors apologize to the many colleagues whose work they have been unable to cite and thank K. McKnight and A. Perelson for helpful discussions. This work was supported in part by grants from the US National Institutes of Health (R01-AI095690 and R01-CA164029) and the University of North Carolina Cancer Research Fund.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Stanley M. Lemon.

Ethics declarations

Competing interests

S.M.L.'s laboratory has received research funding from industry (Merck, Scynexis and Johnson & Johnson) as well as from the US National Institutes of Health, and he has served as a consultant to Merck, AbbVie, Hoffman-LaRoche, Gilead, Santaris, Achillion and Janssen with respect to the clinical and preclinical development of direct-acting antivirals against hepatitis C virus. Y.L., D.Y. and T.M. declare no competing interests.

PowerPoint slides

Glossary

Stem–loop

Also called a hairpin element. An element of RNA secondary structure formed when two segments of an RNA molecule have complementary nucleotide sequences and base pair to form a two-stranded helix with an intervening single-stranded loop segment.

Internal ribosome entry site

(IRES). A highly structured RNA sequence that recruits ribosomes and initiates translation of the RNA independently of ribosome scanning from a 5′-cap. The 5′ untranslated region of the hepatitis C virus RNA genome contains an IRES. A variety of IRES elements with different RNA structures are found in other viruses, as well as in a subset of host mRNAs, where they permit translation when cap-dependent translation is inhibited during mitosis, apoptosis or hypoxia.

Lipid droplets

Ubiquitous, dynamic cellular organelles that are rich in lipids and mainly composed of cholesteryl esters and triglycerides. They function in energy storage by sequestering fatty acids in triglycerides and as a platform for the metabolism and transport of lipids, as well as playing a part in cellular signalling through the generation of bioactive lipids.

Retinoic acid-inducible gene I

(RIG-I). A cytoplasmic RNA helicase that senses viral RNAs and initiates signalling that leads to host interferon production. RIG-I preferentially binds short double-stranded, AU-rich RNAs with 5′ di- or triphosphates and subsequently activates its downstream adaptor, mitochondrial antiviral signalling protein (MAVS), through interactions between shared caspase activation and recruitment domains (CARDs). RIG-I and a second RIG-I-like receptor, MDA5 (also known as IFIH1), contribute to innate immune sensing of distinct types of viruses.

Protein kinase R

(PKR). An interferon-induced protein kinase that is activated in response to double-stranded RNA and is central to cellular responses to a variety of stresses, such as viral infection, cytokine exposure, nutrient depletion, irradiation and endoplasmic reticulum stress. PKR-mediated phosphorylation of eukaryotic translation initiation factor 2A (EIF2A) results in global translational repression.

Pseudoknot

A higher-order RNA structure that contains at least two stem–loops and in which the bases in one half of one stem pair with those in the loop of a second stem–loop.

Ribosome scanning

The process by which a 40S ribosome subunit with associated initiation factors moves in a 3′ direction along an mRNA molecule in search of a start codon (typically AUG) following its initial interactions with and binding to the 5′ terminal 7-methylguanylate cap structure. This is an essential component of eukaryotic cap-dependent translation initiation. Assembly of the mature 80S ribosome and the initiation of polypeptide synthesis commence at the start codon.

Selective 2′-hydroxyl acylation and primer extension

(SHAPE). An experimental method that assesses the backbone flexibility of an RNA molecule at a single-nucleotide resolution based on the reactivity of 2′-hydroxyl groups towards an electrophile. Nucleotides that are flexible (unpaired) are preferentially chemically modified and are recognized as stops in a subsequent primer extension reaction, allowing unpaired bases to be distinguished from those for which flexibility is constrained by base-pairing.

RNAi

A mechanism that was initially discovered in plants and involves small non-coding RNAs suppressing gene expression by targeting specific mRNA transcripts in association with an RNA-induced silencing complex. Synthetic short interfering RNAs (siRNAs) may be transfected into cells to deplete a specific protein.

RNA-induced silencing complex

(RISC). A ribonucleoprotein complex with a core comprising a small non-coding RNA (typically a microRNA or short interfering RNA) guide strand loaded into an Argonaute protein (AGO1–AGO4). The RISC acts to suppress translation and, in some cases, directly cleave an mRNA target.

Antagomir

A synthetic, typically nuclease-resistant, chemically modified oligonucleotide that is complementary to the sequence of a microRNA and antagonizes its activity by binding to and sequestering the microRNA from its mRNA target, as well as possibly blocking microRNA biogenesis.

Small nucleolar RNA

(snoRNA). A class of small non-coding RNAs that act to guide specific post-transcriptional modifications, such as 2′-O-methylation or uridine isomerization, and occasionally cleavage, of other RNAs (primarily rRNAs and tRNAs).

Polysome

A large complex formed by multiple mature 80S ribosomes bound to an mRNA that is actively undergoing translation.

Flaviviridae

A family of enveloped positive-strand RNA viruses comprising four genera: Flavivirus (dengue virus, yellow fever virus and others), Hepacivirus (hepatitis C virus), Pestivirus and Pegivirus.

P-bodies

(Processing bodies). Cytoplasmic aggregates of proteins engaged in the transport, storage and decay of host mRNA. These bodies represent foci of accumulation of 5′-to-3′ exonuclease 1 (XRN1).

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, Y., Yamane, D., Masaki, T. et al. The yin and yang of hepatitis C: synthesis and decay of hepatitis C virus RNA. Nat Rev Microbiol 13, 544–558 (2015). https://doi.org/10.1038/nrmicro3506

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrmicro3506

This article is cited by

Search

Quick links

Nature Briefing Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing: Microbiology