Rapid reversal of innate immune dysregulation in blood of patients and livers of humanized mice with HCV following DAA therapy

Matthew A Burchill, Justin A Roby, Nanette Crochet, Megan Wind-Rotolo, Amy E Stone, Michael G Edwards, Rachael J Dran, Michael S Kriss, Michael Gale Jr, Hugo R Rosen, Matthew A Burchill, Justin A Roby, Nanette Crochet, Megan Wind-Rotolo, Amy E Stone, Michael G Edwards, Rachael J Dran, Michael S Kriss, Michael Gale Jr, Hugo R Rosen

Abstract

Results: First, in patients receiving two different combinations of DAAs, we found that DAAs induced not only rapid viral clearance, but also a re-setting of antiviral immune responses in the peripheral blood. Specifically, we see a rapid decline in the expression of genes associated with chronic IFN stimulation (IFIT3, USP18, IFIT1) as well as a rapid decline in genes associated with inflammation (IL1β, CXCL10, CXCL11) in the peripheral blood that precedes the complete removal of virus from the blood. Interestingly, this rapid reversal of innate immune activation was not seen in patients who successfully clear chronic HCV infection using IFN-based therapy. Next, using a novel humanized mouse model (Fah-/-RAG2-/-IL2rgnull-FRG), we assessed the changes that occur in the hepatic tissue following DAA treatment. DAA-mediated rapid HCV clearance resulted in blunting of the expression of proinflammatory responses while functionally restoring the RIG-I/MAVS axis in the liver of humanized mice.

Conclusions: Collectively, our data demonstrate that the rapid viral clearance following treatment with DAAs results in the rebalancing of innate antiviral response in both the peripheral blood and the liver as well as enhanced antiviral signaling within previously infected hepatocytes.

Conflict of interest statement

Competing Interests: MRW is an employee of and holds stock in Bristol-Myers Squibb and MAB received funding from Gilead Sciences. This does not alter our adherence to PLOS ONE policies on sharing data and materials.

Figures

Fig 1. DAA therapy results in a…
Fig 1. DAA therapy results in a reduction in innate immune activation in the peripheral blood.
(A) Graphical representation of the measured or predicted activation state (IPA) of innate immune signaling molecules at the end of DAA therapy compared to pretreatment. Green is inhibited whereas red represents increased expression. (B) Semi-quantitative RT-PCR of select genes associated with HCV-induced inflammation from PBMCs treated with two different regimens of DAA at end of treatment compared to pretreatment in DAA cohort 1 (n = 18, top) or DAA cohort 2 (n = 11, bottom). (C) Western blot analysis of the levels of pS-NFκB, and NFκB. Top: representative images of 6 patient samples prior to (Pre) and twelve weeks following completion of DAA therapy (Post) in DAA cohort 2. (D) Quantification of the ratio of pS-NFκB to NFκB (right) prior to treatment (circles) and at twelve weeks following the end of treatment (squares). Lines represent individual patient samples. *P = 0.03, Wilcoxon matched-pairs signed rank test. N = 11 patients.
Fig 2. DAA therapy induces rapid suppression…
Fig 2. DAA therapy induces rapid suppression of some but not all antiviral signaling molecules.
Kinetic analysis of transcriptional levels of CXCL10 (black), CXCL11 (purple), RIG-I (yellow), IRF3 (blue), IFIT1(ISG56) (red), IFITM1 (grey), USP18 (green), IL1β (black circles, dotted line) and plasma viral load (black crosses with black dashed line) from PBMCs in DAA patient cohort 2 (n = 11).
Fig 3. Differential transcriptional changes are associated…
Fig 3. Differential transcriptional changes are associated with IFN-free DAA therapy and IFN/Ribavirin therapy.
(A) Representation of the shared and different transcriptional changes in DAA therapy (EOT) compared to IFNα/Ribavirin therapy (wk 10) (19). The top 1000 genes statistically significantly changed from pretreatment in both data sets were compared to the expression of the given gene in the other treatment cohort. (B) List of the 32 genes that were statistically changed from post treatment to pretreatment in both data sets. (C) Quantitative RT-PCR comparing fold change from pretreatment at twelve weeks following DAA therapy (Black Bars) and twenty-four weeks following IFNα/Ribavirin therapy (White bars). P value represents comparison between both treatments. P<0.05, ***P<0.001, NS = Not significant. N = 8-11patients per group.
Fig 4. Restoration of HCV-suppressed antiviral signaling…
Fig 4. Restoration of HCV-suppressed antiviral signaling in the liver of humanized mice treated with DAAs.
(A) Serum levels of HCV in mice injected with 1.4 x 106 IU HCV intravenously. (B) HCV viral loads after treatment with Sofosbuvir (2.0mg daily), Daclatasvir (0.3mg daily), Asunaprevir (0.5mg twice daily) or DMSO vehicle control via oral gavage. (C) Protein expression of NS3, MAVS, IFITM1, and IRF3 following 14 days of DMSO treatment or DAA treatment in whole liver tissue via immunofluorescence staining. N = 3 mice per group. ****P<0.0001, NS = Not significant.

References

    1. Everson G, Cooper C, Hezode C, Shiffman ML, Yoshida E, Beltran-Jaramillo T, et al. DAUPHINE: a randomized phase II study of danoprevir/ritonavir plus peginterferon alpha-2a/ribavirin in HCV genotypes 1 or 4. Liver international: official journal of the International Association for the Study of the Liver. 2015;35(1):108–19. doi: .
    1. Burchill MA, Golden-Mason L, Wind-Rotolo M, Rosen HR. Memory re-differentiation and reduced lymphocyte activation in chronic HCV-infected patients receiving direct-acting antivirals. Journal of viral hepatitis. 2015;22(12):983–91. doi: .
    1. Conjeevaram HS, Fried MW, Jeffers LJ, Terrault NA, Wiley-Lucas TE, Afdhal N, et al. Peginterferon and ribavirin treatment in African American and Caucasian American patients with hepatitis C genotype 1. Gastroenterology. 2006;131(2):470–7. doi: .
    1. Burton JR Jr., Klarquist J, Im K, Smyk-Pearson S, Golden-Mason L, Castelblanco N, et al. Prospective analysis of effector and regulatory CD4+ T cells in chronic HCV patients undergoing combination antiviral therapy. J Hepatol. 2008;49(3):329–38. doi: .
    1. McMahan RH, Porsche CE, Edwards MG, Rosen HR. Free Fatty Acids Differentially Downregulate Chemokines in Liver Sinusoidal Endothelial Cells: Insights into Non-Alcoholic Fatty Liver Disease. PLoS One. 2016;11(7):e0159217 doi: .
    1. Ellis EC, Naugler WE, Parini P, Mork LM, Jorns C, Zemack H, et al. Mice with chimeric livers are an improved model for human lipoprotein metabolism. PLoS One. 2013;8(11):e78550 doi: .
    1. Azuma H, Paulk N, Ranade A, Dorrell C, Al-Dhalimy M, Ellis E, et al. Robust expansion of human hepatocytes in Fah-/-/Rag2-/-/Il2rg-/- mice. Nat Biotechnol. 2007;25(8):903–10. doi: .
    1. Horner SM, Gale M Jr. Regulation of hepatic innate immunity by hepatitis C virus. Nat Med. 2013;19(7):879–88. doi: .
    1. Tsuchida T, Friedman SL. Mechanisms of hepatic stellate cell activation. Nat Rev Gastroenterol Hepatol. 2017. doi: .
    1. Rosen HR. Emerging concepts in immunity to hepatitis C virus infection. J Clin Invest. 2013;123(10):4121–30. doi: .
    1. Negash AA, Ramos HJ, Crochet N, Lau DT, Doehle B, Papic N, et al. IL-1beta production through the NLRP3 inflammasome by hepatic macrophages links hepatitis C virus infection with liver inflammation and disease. PLoS Pathog. 2013;9(4):e1003330 doi: .
    1. Miura K, Kodama Y, Inokuchi S, Schnabl B, Aoyama T, Ohnishi H, et al. Toll-like receptor 9 promotes steatohepatitis by induction of interleukin-1beta in mice. Gastroenterology. 2010;139(1):323–34 e7. doi: .
    1. Gieling RG, Wallace K, Han YP. Interleukin-1 participates in the progression from liver injury to fibrosis. Am J Physiol Gastrointest Liver Physiol. 2009;296(6):G1324–31. doi: .
    1. Dienstag JL, McHutchison JG. American Gastroenterological Association technical review on the management of hepatitis C. Gastroenterology. 2006;130(1):231–64; quiz 14–7. doi: .
    1. Schaefer M, Capuron L, Friebe A, Diez-Quevedo C, Robaeys G, Neri S, et al. Hepatitis C infection, antiviral treatment and mental health: a European expert consensus statement. J Hepatol. 2012;57(6):1379–90. doi: .
    1. Schaefer M, Sarkar R, Knop V, Effenberger S, Friebe A, Heinze L, et al. Escitalopram for the prevention of peginterferon-alpha2a-associated depression in hepatitis C virus-infected patients without previous psychiatric disease: a randomized trial. Ann Intern Med. 2012;157(2):94–103. doi: .
    1. Fried MW. Side effects of therapy of hepatitis C and their management. Hepatology. 2002;36(5 Suppl 1):S237–44. doi: .
    1. Meissner EG, Wu D, Osinusi A, Bon D, Virtaneva K, Sturdevant D, et al. Endogenous intrahepatic IFNs and association with IFN-free HCV treatment outcome. J Clin Invest. 2014;124(8):3352–63. doi: .
    1. Taylor MW, Tsukahara T, McClintick JN, Edenberg HJ, Kwo P. Cyclic changes in gene expression induced by Peg-interferon alfa-2b plus ribavirin in peripheral blood monocytes (PBMC) of hepatitis C patients during the first 10 weeks of treatment. J Transl Med. 2008;6:66 doi: .
    1. Bissig KD, Wieland SF, Tran P, Isogawa M, Le TT, Chisari FV, et al. Human liver chimeric mice provide a model for hepatitis B and C virus infection and treatment. J Clin Invest. 2010;120(3):924–30. Epub 2010/02/25. doi: .
    1. Grompe M, al-Dhalimy M, Finegold M, Ou CN, Burlingame T, Kennaway NG, et al. Loss of fumarylacetoacetate hydrolase is responsible for the neonatal hepatic dysfunction phenotype of lethal albino mice. Genes Dev. 1993;7(12A):2298–307. .
    1. Arnaud N, Dabo S, Akazawa D, Fukasawa M, Shinkai-Ouchi F, Hugon J, et al. Hepatitis C virus reveals a novel early control in acute immune response. PLoS Pathog. 2011;7(10):e1002289 doi: .
    1. 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. Proc Natl Acad Sci U S A. 2011;108(35):14590–5. doi: .
    1. Li XD, Sun L, Seth RB, Pineda G, Chen ZJ. Hepatitis C virus protease NS3/4A cleaves mitochondrial antiviral signaling protein off the mitochondria to evade innate immunity. Proc Natl Acad Sci U S A. 2005;102(49):17717–22. doi: .
    1. Wilkins C, Woodward J, Lau DT, Barnes A, Joyce M, McFarlane N, et al. IFITM1 is a tight junction protein that inhibits hepatitis C virus entry. Hepatology. 2013;57(2):461–9. doi: .
    1. Bachofner JA, Valli PV, Kroger A, Bergamin I, Kunzler P, Baserga A, et al. Direct antiviral agent treatment of chronic hepatitis C results in rapid regression of transient elastography and fibrosis markers fibrosis-4 score and aspartate aminotransferase-platelet ratio index. Liver international: official journal of the International Association for the Study of the Liver. 2017;37(3):369–76. doi: .
    1. Horner SM. Activation and evasion of antiviral innate immunity by hepatitis C virus. J Mol Biol. 2014;426(6):1198–209. doi: .
    1. Kurelac I, Lepej SZ, Grlgic I, Gorenec L, Papic N, Dusek D, et al. Chemokine CXCL10 at week 4 of treatment predicts sustained virological response in patients with chronic hepatitis C. J Interferon Cytokine Res. 2012;32(8):386–91. doi: .
    1. Spaan M, van Oord G, Kreefft K, Hou J, Hansen BE, Janssen HL, et al. Immunological Analysis During Interferon-Free Therapy for Chronic Hepatitis C Virus Infection Reveals Modulation of the Natural Killer Cell Compartment. The Journal of infectious diseases. 2016;213(2):216–23. doi: .
    1. Serti E, Chepa-Lotrea X, Kim YJ, Keane M, Fryzek N, Liang TJ, et al. Successful Interferon-Free Therapy of Chronic Hepatitis C Virus Infection Normalizes Natural Killer Cell Function. Gastroenterology. 2015;149(1):190–200 e2. doi: .

Source: PubMed

3
Abonnieren