1887

Abstract

The mechanistic target of rapamycin (mTOR), an important kinase that assimilates several upstream signals, associates into two functional complexes, mTORC1 and mTORC2. In this study, we demonstrate that HCV infection activates mTORC1 that functions in important antiviral response. Pharmacological inhibition of mTOR complexes augmented cellular HCV RNA levels, the observation confirmed further by Raptor depletion, indicating antiviral roles of mTORC1. ULK1 depletion phenocopied mTOR inhibition and thus suggested that mTORC1 restricts HCV replication through ULK1. We reveal that ULK1 depletion augmented the levels of miR-122, a critical host factor for HCV replication, thus possibly regulating HCV replication. The increase in HCV RNA levels, however, failed to augment intracellular infectious virion production, reflecting a lower rate of virion assembly. Higher intracellular HCV RNA levels, however, did not result in a corresponding increase in HCV RNA and infectious titres in mTOR inhibited supernatants, but in contrast showed a consistent drop, confirming defective viral assembly caused by the inhibition. Consistent with this, the mTOR activator caused a significant drop in HCV RNA levels both in infected cells and in the supernatant. Our results demonstrate that ULK1 depletion did not affect autophagy, suggesting that ULK1-mediated HCV regulation is autophagy independent. Together, our data demonstrate that mTORC1 functions to suppress HCV RNA replication, but facilitates the virion packaging and release. Our studies reveal that the activation of mTOR by HCV infection is an antiviral measure by the cells.

Loading

Article metrics loading...

/content/journal/jgv/10.1099/jgv.0.001356
2020-01-01
2024-04-25
Loading full text...

Full text loading...

/deliver/fulltext/jgv/101/1/86.html?itemId=/content/journal/jgv/10.1099/jgv.0.001356&mimeType=html&fmt=ahah

References

  1. Choo Q, Kuo G, Weiner A, Overby L, Bradley D et al. Isolation of a cDNA clone derived from a blood-borne non-A, non-B viral hepatitis genome. Science 1989; 244:359–362 [View Article]
    [Google Scholar]
  2. Hadigan C, Kottilil S. Hepatitis C virus infection and coinfection with human immunodeficiency virus: challenges and advancements in management. Jama 2011; 306:294–301
    [Google Scholar]
  3. Manns MP, Wedemeyer H, Cornberg M. Treating viral hepatitis C: efficacy, side effects, and complications. Gut 2006; 55:1350–1359 [View Article]
    [Google Scholar]
  4. Fried MW. Side effects of therapy of hepatitis C and their management. Hepatology 2002; 36:S237–244
    [Google Scholar]
  5. Takeda H, Ueda Y, Inuzuka T, Yamashita Y, Osaki Y et al. Evolution of multi-drug resistant HCV clones from pre-existing resistant-associated variants during direct-acting antiviral therapy determined by third-generation sequencing. Sci Rep 2017; 7:45605 [View Article]
    [Google Scholar]
  6. Maio CD V, Cento V, Aragri M, Paolucci S, Pollicino T et al. Frequent NS5A and multiclass resistance in almost all HCV genotypes at DAA failures: what are the chances for second-line regimens?. Journal of hepatology 2017
    [Google Scholar]
  7. Jopling CL, Yi M, Lancaster AM, Lemon SM, Sarnow P. Modulation of hepatitis C virus RNA abundance by a liver-specific microRNA. Science 2005; 309:1577–1581 [View Article]
    [Google Scholar]
  8. Shimakami T, Yamane D, Jangra RK, Kempf BJ, Spaniel C et al. Stabilization of hepatitis C virus RNA by an Ago2-miR-122 complex. Proc Natl Acad Sci U S A 2012; 109:941–946 [View Article]
    [Google Scholar]
  9. Li Y, Masaki T, Lemon SM. miR-122 and the hepatitis C RNA genome: more than just stability. RNA biology 2013; 10:919–923
    [Google Scholar]
  10. Masaki T, Arend KC, Li Y, Yamane D, McGivern DR 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 2015; 17:217–228 [View Article]
    [Google Scholar]
  11. Kambara H, Fukuhara T, Shiokawa M, Ono C, Ohara Y et al. Establishment of a novel permissive cell line for the propagation of hepatitis C virus by expression of microRNA miR122. J Virol 2012; 86:1382–1393 [View Article]
    [Google Scholar]
  12. Sabatini DM, Erdjument-Bromage H, Lui M, Tempst P, Snyder SH. RAFT1: a mammalian protein that binds to FKBP12 in a rapamycin-dependent fashion and is homologous to yeast TORS. Cell 1994; 78:35–43 [View Article]
    [Google Scholar]
  13. Brown EJ, Albers MW, Bum Shin T, ichikawa K, Keith CT et al. A mammalian protein targeted by G1-arresting rapamycin–receptor complex. Nature 1994; 369:756–758 [View Article]
    [Google Scholar]
  14. Saxton RA, Sabatini DM. mTOR signaling in growth, metabolism, and disease. Cell 2017; 168:960–976 [View Article]
    [Google Scholar]
  15. Kim D-H, Sarbassov DD, Ali SM, King JE, Latek RR et al. mTOR interacts with raptor to form a nutrient-sensitive complex that signals to the cell growth machinery. Cell 2002; 110:163–175 [View Article]
    [Google Scholar]
  16. Wullschleger S, Loewith R, Hall MN. Tor signaling in growth and metabolism. Cell 2006; 124:471–484 [View Article]
    [Google Scholar]
  17. Laplante M, Sabatini DM. mTOR signaling in growth control and disease. Cell 2012; 149:274–293 [View Article]
    [Google Scholar]
  18. Hsieh AC, Costa M, Zollo O, Davis C, Feldman ME et al. Genetic dissection of the oncogenic mTOR pathway reveals druggable addiction to translational control via 4EBP-eIF4E. Cancer Cell 2010; 17:249–261 [View Article]
    [Google Scholar]
  19. Haghighat A, Mader S, Pause A, Sonenberg N. Repression of cap-dependent translation by 4E-binding protein 1: competition with p220 for binding to eukaryotic initiation factor-4E. Embo J 1995; 14:5701–5709 [View Article]
    [Google Scholar]
  20. Richter JD, Sonenberg N. Regulation of cap-dependent translation by eIF4E inhibitory proteins. Nature 2005; 433:477–480 [View Article]
    [Google Scholar]
  21. Egan D, Kim J, Shaw RJ, Guan K-L. The autophagy initiating kinase ULK1 is regulated via opposing phosphorylation by AMPK and mTOR. Autophagy 2011; 7:643–644 [View Article]
    [Google Scholar]
  22. Tanida I, Fukasawa M, Ueno T, Kominami E, Wakita T et al. Knockdown of autophagy-related gene decreases the production of infectious hepatitis C virus particles. Autophagy 2009; 5:937–945 [View Article]
    [Google Scholar]
  23. Dreux M, Gastaminza P, Wieland SF, Chisari FV. The autophagy machinery is required to initiate hepatitis C virus replication. Proc Natl Acad Sci U S A 2009; 106:14046–14051 [View Article]
    [Google Scholar]
  24. Sarbassov DD, Guertin DA, Ali SM, Sabatini DM. Phosphorylation and regulation of Akt/PKB by the rictor-mTOR complex. Science 2005; 307:1098–1101 [View Article]
    [Google Scholar]
  25. Sarbassov DD, Ali SM, Sengupta S, Sheen J-H, Hsu PP et al. Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB. Mol Cell 2006; 22:159–168 [View Article]
    [Google Scholar]
  26. Choo AY, Yoon S-O, Kim SG, Roux PP, Blenis J. Rapamycin differentially inhibits S6Ks and 4E-BP1 to mediate cell-type-specific repression of mRNA translation. Proc Natl Acad Sci U S A 2008; 105:17414–17419 [View Article]
    [Google Scholar]
  27. Thoreen CC, Kang SA, Chang JW, Liu Q, Zhang J et al. An ATP-competitive mammalian target of rapamycin inhibitor reveals rapamycin-resistant functions of mTORC1. J Biol Chem 2009; 284:8023–8032 [View Article]
    [Google Scholar]
  28. Feldman ME, Apsel B, Uotila A, Loewith R, Knight ZA et al. Active-Site inhibitors of mTOR target rapamycin-resistant outputs of mTORC1 and mTORC2. PLoS Biol 2009; 7:e1000038 [View Article]
    [Google Scholar]
  29. Tsukiyama-Kohara K, Iizuka N, Kohara M, Nomoto A. Internal ribosome entry site within hepatitis C virus RNA. J Virol 1992; 66:1476–1483
    [Google Scholar]
  30. Wang C, Sarnow P, Siddiqui A. A conserved helical element is essential for internal initiation of translation of hepatitis C virus RNA. J Virol 1994; 68:7301–7307
    [Google Scholar]
  31. Stöhr S, Costa R, Sandmann L, Westhaus S, Pfaender S et al. Host cell mTORC1 is required for HCV RNA replication. Gut 2016; 65:2017–2028 [View Article]
    [Google Scholar]
  32. Shao RX, Zhang L, Peng LF, Sun E, Chung WJ et al. Suppressor of cytokine signaling 3 suppresses hepatitis C virus replication in an mTOR-dependent manner. J Virol 2010; 84:6060–6069 [View Article]
    [Google Scholar]
  33. Shrivastava S, Bhanja Chowdhury J, Steele R, Ray R, Ray RB. Hepatitis C virus upregulates Beclin1 for induction of autophagy and activates mTOR signaling. J Virol 2012; 86:8705–8712 [View Article]
    [Google Scholar]
  34. Mannova P, Beretta L. Activation of the N-Ras-PI3K-Akt-mTOR pathway by hepatitis C virus: control of cell survival and viral replication. J Virol 2005; 79:8742–8749 [View Article]
    [Google Scholar]
  35. George A, Panda S, Kudmulwar D, Chhatbar SP, Nayak SC et al. Hepatitis C virus NS5A binds to the mRNA cap-binding eukaryotic translation initiation 4F (eIF4F) complex and up-regulates host translation initiation machinery through eIF4E-binding protein 1 inactivation. J Biol Chem 2012; 287:5042–5058 [View Article]
    [Google Scholar]
  36. Zhao J, Zhai B, Gygi SP, Goldberg AL. mTOR inhibition activates overall protein degradation by the ubiquitin proteasome system as well as by autophagy. Proc Natl Acad Sci U S A 2015; 112:15790–15797 [View Article]
    [Google Scholar]
  37. Rance E, Tanner JE, Alfieri C. Inhibition of IκB kinase by thalidomide increases hepatitis C virus RNA replication. J Viral Hepat 2012; 19:e73–e80 [View Article]
    [Google Scholar]
  38. Komurian-Pradel F, Perret M, Deiman B, Sodoyer M, Lotteau V et al. Strand specific quantitative real-time PCR to study replication of hepatitis C virus genome. J Virol Methods 2004; 116:103–106 [View Article]
    [Google Scholar]
  39. Plaskon NE, Adelman ZN, Myles KM. Accurate strand-specific quantification of viral RNA. PLoS One 2009; 4:e7468 [View Article]
    [Google Scholar]
  40. Huang H, Kang R, Wang J, Luo G, Yang W et al. Hepatitis C virus inhibits AKT-tuberous sclerosis complex (TSC), the mechanistic target of rapamycin (mTOR) pathway, through endoplasmic reticulum stress to induce autophagy. Autophagy 2013; 9:175–195 [View Article]
    [Google Scholar]
  41. Ray U, Das S. Interplay between NS3 protease and human La protein regulates translation-replication switch of hepatitis C virus. Sci Rep 2011; 1: [View Article]
    [Google Scholar]
  42. Joshi A, Iyengar R, Joo JH, Li-Harms XJ, Wright C et al. Nuclear ULK1 promotes cell death in response to oxidative stress through PARP1. Cell Death Differ 2016; 23:216–230 [View Article]
    [Google Scholar]
  43. Wendel H-G, Silva RLA, Malina A, Mills JR, Zhu H et al. Dissecting eIF4E action in tumorigenesis. Genes Dev 2007; 21:3232–3237 [View Article]
    [Google Scholar]
  44. Herker E, Harris C, Hernandez C, Carpentier A, Kaehlcke K et al. Efficient hepatitis C virus particle formation requires diacylglycerol acyltransferase-1. Nat Med 2010; 16:1295–1298 [View Article]
    [Google Scholar]
  45. Gastaminza P, Cheng G, Wieland S, Zhong J, Liao W et al. Cellular determinants of hepatitis C virus assembly, maturation, degradation, and secretion. J Virol 2008; 82:2120–2129 [View Article]
    [Google Scholar]
  46. Mankouri J, Walter C, Stewart H, Bentham M, Park WS et al. Release of Infectious Hepatitis C Virus from Huh7 Cells Occurs via a trans -Golgi Network-to-Endosome Pathway Independent of Very-Low-Density Lipoprotein Secretion. J Virol 2016; 90:7159–7170 [View Article]
    [Google Scholar]
  47. Lai C-K, Jeng K-S, Machida K, Lai MMC. Hepatitis C virus egress and release depend on endosomal trafficking of core protein. J Virol 2010; 84:11590–11598 [View Article]
    [Google Scholar]
  48. Laplante M, Sabatini DM. An emerging role of mTOR in lipid biosynthesis. Current Biology 2009; 19:R1046–R1052 [View Article]
    [Google Scholar]
  49. Jiang X, Yeung RS. Regulation of microtubule-dependent protein transport by the TSC2/mammalian target of rapamycin pathway. Cancer Res 2006; 66:5258–5269 [View Article]
    [Google Scholar]
  50. SH R, Jung CH, Hahn WS, Xu X, Kim YM et al. Distinct functions of ULK1 and ULK2 in the regulation of lipid metabolism in adipocytes. Autophagy 2013; 9:2103–2114
    [Google Scholar]
  51. Caino MC, Chae YC, Vaira V, Ferrero S, Nosotti M et al. Metabolic stress regulates cytoskeletal dynamics and metastasis of cancer cells. J Clin Invest 2013; 123:2907–2920 [View Article]
    [Google Scholar]
  52. Chan EYW, Kir S, Tooze SA. siRNA screening of the kinome identifies ULK1 as a multidomain modulator of autophagy. J Biol Chem 2007; 282:25464–25474 [View Article]
    [Google Scholar]
  53. Loo Y-M, Loo Y-M, Foy E, Li K, Yoneyama M et al. Regulating intracellular antiviral defense and permissiveness to hepatitis C virus RNA replication through a cellular RNA helicase, RIG-I. J Virol 2005; 79:2689–2699 [View Article]
    [Google Scholar]
  54. Lindenbach BD et al. Complete replication of hepatitis C virus in cell culture. Science 2005; 309:623–626 [View Article]
    [Google Scholar]
http://instance.metastore.ingenta.com/content/journal/jgv/10.1099/jgv.0.001356
Loading
/content/journal/jgv/10.1099/jgv.0.001356
Loading

Data & Media loading...

Supplements

Supplementary material 1

PDF
This is a required field
Please enter a valid email address
Approval was a Success
Invalid data
An Error Occurred
Approval was partially successful, following selected items could not be processed due to error