Skip to main content
Erschienen in: Archives of Virology 10/2017

Open Access 16.06.2017 | Original Article

Nucleoside analogue 2’-C-methylcytidine inhibits hepatitis E virus replication but antagonizes ribavirin

verfasst von: Changbo Qu, Lei Xu, Yuebang Yin, Maikel P. Peppelenbosch, Qiuwei Pan, Wenshi Wang

Erschienen in: Archives of Virology | Ausgabe 10/2017

Abstract

Hepatitis E virus (HEV) infection has emerged as a global health issue, but no approved medication is available. The nucleoside analogue 2’-C-methylcytidine (2CMC), a viral polymerase inhibitor, has been shown to inhibit infection with a variety of viruses, including hepatitis C virus (HCV). Here, we report that 2CMC significantly inhibits the replication of HEV in a subgenomic replication model and in a system using a full-length infectious virus. Importantly, long-term treatment with 2CMC did not result in a loss of antiviral potency, indicating a high barrier to drug resistance development. However, the combination of 2CMC with ribavirin, an off-label treatment for HEV, exerts antagonistic effects. Our results indicate that 2CMC serves as a potential antiviral drug against HEV infection.

Introduction

Hepatitis E virus (HEV) is a single-stranded, positive-sense RNA virus, and its genome contains three open reading frames (ORFs). ORF1 encodes a polyprotein that serves as a precursor of all of the nonstructural proteins needed for HEV replication. ORF2 encodes the capsid protein of the HEV virion. ORF3 encodes a small multifunctional protein with a molecular mass of 13 kDa [11]. HEV was initially thought to cause acute infection only in developing countries. However, over the last decade, hepatitis E cases have frequently been reported in developed countries and have been recognized mainly as autochthonous cases rather than an imported disease [11, 12]. Generally, HEV infection is self-limiting and asymptomatic, and as a consequence, its mortality rate is low. However, it can cause high mortality in pregnant women. In immunocompromised patients receiving organ transplantation, more than 60% of HEV-infected patients develop chronic disease and quickly progress towards severe liver complications such as fibrosis and cirrhosis [20]. In addition to hepatitis, this virus has been associated with a broad range of extrahepatic manifestations, in particular, renal and neurological injuries [14, 21]. Therefore, the development of specific antiviral drugs for HEV infection is urgently required.
Nucleoside analogues have been used clinically for almost 50 years and represent the cornerstones for treatment of patients with cancer or viral infection. Ribavirin (RBV) has been used as an off-label antiviral drug, showing high efficacy in many chronic HEV patients, but HEV mutations associated with ribavirin treatment failure have been reported [4, 7]. Sofosbuvir (SOF), a potent direct-acting agent (DAA) against hepatitis C virus (HCV) [2], has been recently suggested to inhibit HEV replication in cell culture and exert an additive effect when combined with ribavirin [4]. However, other in vitro and clinical studies have demonstrated that sofosbuvir is not very effective against HEV infection [8, 18, 19], suggesting that this drug might not be a promising candidate for the treatment of chronic HEV patients.
2’-C-methylcytidine (2CMC) was initially identified as a competitive inhibitor of the HCV RNA-dependent RNA polymerase (RdRp). In addition to HCV, it has been shown to inhibit the replication of a variety of other viruses (e.g., dengue virus and norovirus) [13, 15]. It also has been reported to inhibit cutthroat trout virus, a non-pathogenic fish virus that is remarkable similar to HEV [6]. In this study, we have demonstrated that 2CMC efficiently inhibits HEV replication and thus is a potential candidate for anti-HEV drug development.

Materials and methods

Reagents and antibodies

2CMC, RBV, guanosine triphosphate (GTP) and cytidine 5’-triphosphate (CTP) were purchased from Sigma-Aldrich and were dissolved in dimethyl sulfoxide (DMSO) (Sigma-Aldrich, St Louis, MO). The HEV-specific antibody was purchased from EMD Millipore (MAB8002).

HEV cell culture models

Multiple cell lines were employed in this study, including human hepatoma cell lines (Huh7 and PLC/PRF/5), a human embryonic kidney cell line (HEK293), a human primary glioblastoma cell line (U87), and a human fetal lung fibroblast cell line (MRC5). Huh7 and U87 cells were kindly provided by Professor Bart Haagmans from the Department of Viroscience, Erasmus Medical Center. The human embryonic kidney 293 cell line, PLC/PRF/5 and MRC5 were originally obtained from ATCC (http://​www.​atcc.​org). These cells were cultured in Dulbecco’s modified Eagle medium (Lonza Biowhittaker, Verviers, Belgium) supplemented with 10% fetal bovine serum, 100 IU of penicillin per ml, and 100 μg of streptomycin per ml. For the full-length HEV model, a plasmid construct containing the full-length HEV genome (Kernow-C1 p6 clone; GenBank accession number JQ679013) was employed to generate HEV genomic RNA using an Ambion mMessage mMachine in vitro RNA transcription Kit (Thermo Fisher Scientific Life Sciences) [16]. Huh7, PLC/PRF/5, HEK293, U87 and MRC5 cells were electroporated with full-length HEV genomic RNA to generate consecutive HEV-infected cell models (Huh7-p6, PLC/PRF/5-p6, HEK293-p6, U87-p6 and MRC5-p6). To generate the subgenomic (p6-Luc) HEV model, a plasmid construct containing subgenomic HEV was used. This plasmid has an HEV sequence in which the 5’ portion of HEV ORF2 was replaced with the in-frame Gaussia princeps luciferase reporter gene [16]. Huh7, U87 and HEK293 cells were electroporated with HEV subgenomic RNA to generate HEV subgenomic models (Huh7-p6-Luc, U87-p6-Luc, and HEK293-p6-Luc). To normalize nonspecific effects of 2CMC on the luciferase signal, Huh7 cells stably expressing a non-secreted firefly luciferase under the control of the human phosphoglycerate kinase (PGK) promotor (PGK-Luc) were used [18]. In addition, Huh7 cells harboring a subgenomic HCV bicistronic replicon (I389/NS3-3 V/LucUbiNeo-ET) (Huh7-HCV-Luc) were used as positive control of antiviral activity.

Quantification of HEV replication

For Gaussia luciferase, the secreted luciferase activity in the cell culture medium was measured using a BioLux® Gaussia Luciferase Flex Assay Kit (New England Biolabs). Gaussia luciferase activity was quantified using a LumiStar Optima luminescence counter (BMG LabTech, Offenburg, Germany). For the full-length infectious models (HEV-p6), intracellular viral RNA was quantified. RNA was isolated using a Machery-Nucleo Spin RNA II kit (Bioke, Leiden, The Netherlands) and quantified using a NanoDrop ND-1000 spectrophotometer (Wilmington, DE, USA). cDNA was prepared from total RNA using a cDNA Synthesis Kit (Takara Bio Inc, USA). The HEV RNA level was quantified using a SYBR Green–based real-time PCR assay (Applied Biosystems® SYBR® Green PCR Master Mix, Life Technologies, CA, USA) according to the manufacturer’s instructions. The PCR steps consisted of a 10 min holding stage (95 °C) followed by 40 cycles of 15 s at 95 °C, 30 s at 58 °C, and 30 s at 72 °C. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used as a reference gene to normalize gene expression. Relative gene expression was normalized to GAPDH using the formula 2−ΔΔCT (ΔΔCT = ΔCTsample − ΔCTcontrol). The HEV primer sequences were as follows: HEV-F, 5’-ATTGGCCAGAAGTTGGTTTTCAC-3’; HEV-R, 5’-CCGTGGCTATAATTGTGGTCT-3’; GAPDH-F, 5’-TGTCCCCACCCCCAATGTATC-3’; GAPDH-R, 5’CTCCGATGCCTGCTTCACTACCTT-3’.

MTT assay

The cells were seeded in a 96-well plate, and 10 mM 3-(4,5-dimethylthiazol-2-yl)-2,5 diphenyltetrazolium bromide (MTT) (Sigma) was added to the cells. Subsequently, the cells were incubated at 37 °C with 5% CO2 for 3 h. The culture medium was then removed, and 100 μl of DMSO was added to each well. The absorbance of each well was read in a microplate absorbance reader (Bio-Rad, Japan) at wavelength of 490 nm.

Long-term treatment assay

For the long-term treatment assay of the subgenomic model (Huh7-p6-luc), the cells were seeded into a 96-well plate with 5000 cells per well. The cells of the CTR and 2CMC treatment groups were passaged and seeded with the same number of cells every 3 days (d), and cells incubated with vehicle (non-treatment) or 2CMC (10 µM) were maintained throughout the entire incubation period. For the long-term treatment assay of infectious model (Huh7-p6), the cells were seeded into a 48-well plate with 2 × 104 cells per well. The cells of the CTR or 2CMC treatment groups were passaged and seeded with the same number of cells every 3 days, and cells incubated with vehicle (non-treatment) or 2CMC (10 µM) were maintained throughout the entire incubation period.

Western blot assay

Cultured cells were lysed in Laemmli sample buffer containing 0.1 M DTT and heated for 5 min at 95 °C, followed by loading onto a 10% sodium dodecyl sulfate polyacrylamide gel and separation by electrophoresis for 90 min at 120 V, after which the proteins were electrophoretically transferred onto a polyvinylidene difluoride membrane (Invitrogen) for 1.5 h with an electric current of 250 mA. Subsequently, the membrane was blocked with a mixture of 2.5 ml of blocking buffer (Odyssey, USA) and 2.5 ml of phosphate-buffered saline containing 0.05% Tween 20. This was followed by overnight incubation with anti-HEV capsid protein primary antibodies (1:1000) at 4 °C. The membrane was then washed three times, followed by incubation for 1 h with goat anti-mouse IRDye-conjugated secondary antibody (Li-COR Biosciences, Lincoln, USA) (1:5000). After washing three times, protein bands were detected using an Odyssey 3.0 Infrared Imaging System.

IC50 and CC50 calculation

The 50% inhibitory concentration (IC50) value and 50% cytotoxic concentration (CC50) were calculated based on the model Y¼Bottom þ (Top-Bottom)/ (1 þ 10^((LogIC50-X)*HillSlope)) using GraphPad Prism 5 software (GraphPad Prism 5; GraphPad Software Inc., La Jolla, CA, USA).

Statistical analysis

Statistical analysis was performed using the nonpaired, nonparametric test with the Mann-Whitney test and one-way ANOVA with Tukey’s multiple comparison post-test (GraphPad Prism version 5.01; GraphPad Software). P-values less than 0.05 were considered statistically significant.

Results

In this study, the potential anti-HEV effect of 2CMC was investigated in HEV replication models with concentrations ranging from 0.1 μΜ to 10 μM. We demonstrated that 2CMC significantly reduced HEV-driven luciferase activity, and the anti-HEV activity was comparable with its anti-HCV effect at the concentration of 10 µM (Fig. 1A). The IC50 value of 2CMC against HEV replication was 1.64 µM, the CC50 of 2CMC in Huh7 cells was 111.2 µM, and the selectivity index (SI, CC50/IC50) was 67.8 (Fig. 1B). The anti-HEV effect of 2CMC was further confirmed in the full-length (Kernow-C1, p6) infectious model of HEV genotype 3 by both RT-PCR assay (Fig. 1C) and western blot assay (Fig. 1D).
Since HEV-related extrahepatic manifestations have been reported [12], we extended our study to some other hepatic and nonhepatic cell lines. HEV infectious or replication models were established in HEK293, PLC/PRF/5, MRC5 and U87 cells. The anti-HEV potential of 2CMC in these cell lines was tested. In line with the results observed in Huh7-based HEV replication and infectious models, we observed a similar anti-HEV effect of 2CMC in all these cell models without affecting the cell viability (Fig. 1E to G).
Drug resistance is one of the main factors that limit the effectiveness of antiviral treatment. To characterize 2CMC in this respect, we performed experiments in which both HEV replication and infectious models were constantly exposed to 2CMC (10 μM). Interestingly, 2CMC retained its anti-HEV activity in both models even after long-term exposure (Fig. 2A and B). Furthermore, the negative control retained high levels of luciferase activity after long-term incubation with 2CMC, excluding the loss of cell viability during the experimental period (Fig. 2C). Taken together, 2CMC displays a high barrier for drug resistance development.
Theoretically, nucleoside/nucleotide analogs can serve as potential direct-acting antivirals because they bind to the viral RNA polymerase active site to block viral replication. To evaluate the inhibitory specificity of 2CMC against HEV replication, we performed a competition assay employing the substrate cytidine triphosphate (CTP) as an analogous competitor of 2CMC. Our results indicated that CTP reversed the inhibitory effects of 2CMC on HEV replication activity in a dose-dependent manner. In contrast, guanosine triphosphate (GTP) exerted no effect, implying the inhibitory specificity of 2CMC against HEV replication (Fig. 3A and B). Another nucleoside analogue, ribavirin, has been used clinically as an off-label treatment for HEV infection. Thus, its anti-HEV effect in combination with 2CMC was tested. Interestingly, a moderate antagonistic effect (-36.93 µM2 %) was observed, implying that they employ a similar antiviral mechanism (Fig. 4A and B).

Discussion

A variety of nucleoside analogues have been widely used to treat viral infections due to their potent antiviral effects and high barrier to drug resistance development. Ribavirin, an guanosine analogue, is the drug of choice for treating most chronic HEV patients. However, treatment failure has been observed in some cases. Sofosbuvir, a prodrug of a uridine nucleoside analogue that is very effective against HCV, has been investigated recently for its anti-HEV potency. However, there has been debate regarding its potency against HEV [9, 17]. 2CMC, a cytidine nucleoside analogue, has been shown to inhibit infection with a variety of viruses, including HCV and HIV [5]. In this study, we have demonstrated that 2CMC potently inhibits HEV replication in different cell models, albeit with slight differences (Fig. 1A and C). A possible explanation is that these models recapitulate the different steps of the HEV life cycle. The full-length infectious clone (Huh7-p6) models the entire cycle of HEV infection, whereas the subgenomic model (Huh7-p6-luc) only mimics viral replication due to the lack of ORF2 and ORF3.
Encouragingly, the anti-HEV activity was comparable with its anti-HCV effect at particular concentrations. More importantly, in the long-term treatment experiment, 2CMC displayed a high barrier to resistance development, and we demonstrated that this was a specific anti-HEV effect and not due to cytotoxicity. It has been suggested that after it is absorbed by the cells, 2CMC is converted to its 5′-triphosphate form (2CMC-CTP), which serves as an active molecule that competes with the natural substrate CTP. Consistent with this, our results demonstrated that CTP but not GTP reverses the anti-HEV effect of 2CMC, revealing a potential mechanism of action of 2CMC against HEV.
Since ribavirin has been widely used to treat chronic HEV patients, a combined therapy of ribavirin with 2CMC might be envisaged. To test this, the combinatory effects of both drugs were investigated. Unexpectedly, an antagonistic effect was observed. These findings are in agreement with the earlier observation of the combinatory effects of ribavirin and 2CMC on HCV and HIV [3].
Of note, the potential adverse effects of 2CMC should be carefully evaluated in future studies. The clinical applications of nucleoside analogues have been limited in some cases due to off- target effects. Mitochondrial DNA polymerase is an important unintended target for many nucleoside analogues. It has been reported that a nucleoside analogue containing a 2-C-methyl (2-CM) group can reduce mitochondrial transcription and oxidative phosphorylation, resulting in dysfunction of cell metabolism [1, 10]. Therefore, it is recommended that future anti-HEV drug development efforts should focus on the design of less-toxic agents based on the main chemical structure of 2CMC.
In conclusion, 2CMC exerts potent anti-HEV effects in well-established cell culture models, and serves as a potential backbone for anti-HEV drug design. To achieve better efficacy and fewer side effects, future research is still required for drug optimization based on the chemical structure of 2CMC.

Acknowledgements

The authors would like to thank Dr. Suzanne U. Emerson (National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD) for kindly providing the plasmids to generate subgenomic and full-length HEV genotype 3 cell culture systems; Prof. Ralf Bartenschlager and Dr. Volker Lohmann (University of Heidelberg, Germany) for generously providing the Huh7 subgenomic HCV replication cells. The authors also thank the Dutch Digestive Foundation (MLDS) for a career development grant (No. CDG 1304), the Daniel den Hoed Foundation for a Centennial Award grant (to Q. Pan), and the China Scholarship Council for funding PhD fellowships for C. Qu (No.201509110121), L. Xu (No.201306300027), Y. Yin (No.201307720045) and W. Wang (No. 201303250056).

Compliance with ethical standards

This article does not contain any studies with human participants or animals performed by any of the authors and is in compliance with ethical standards for research.

Conflict of interest

The authors declare no conflict of interest.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

© Springer Medizin

Bis 11. April 2024 bestellen und im ersten Jahr 50 % sparen!

e.Med Innere Medizin

Kombi-Abonnement

Mit e.Med Innere Medizin erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Innere Medizin, den Premium-Inhalten der internistischen Fachzeitschriften, inklusive einer gedruckten internistischen Zeitschrift Ihrer Wahl.

© Springer Medizin

Bis 11. April 2024 bestellen und im ersten Jahr 50 % sparen!

e.Med Allgemeinmedizin

Kombi-Abonnement

Mit e.Med Allgemeinmedizin erhalten Sie Zugang zu allen CME-Fortbildungen und Premium-Inhalten der allgemeinmedizinischen Zeitschriften, inklusive einer gedruckten Allgemeinmedizin-Zeitschrift Ihrer Wahl.

© Springer Medizin

Bis 11. April 2024 bestellen und im ersten Jahr 50 % sparen!

Literatur
1.
Zurück zum Zitat Arnold JJ, Sharma SD, Feng JY, Ray AS, Smidansky ED, Kireeva ML, Cho A, Perry J, Vela JE, Park Y, Xu Y, Tian Y, Babusis D, Barauskus O, Peterson BR, Gnatt A, Kashlev M, Zhong W, Cameron CE (2012) Sensitivity of mitochondrial transcription and resistance of RNA polymerase II dependent nuclear transcription to antiviral ribonucleosides. PLoS Pathog 8:e1003030CrossRefPubMedPubMedCentral Arnold JJ, Sharma SD, Feng JY, Ray AS, Smidansky ED, Kireeva ML, Cho A, Perry J, Vela JE, Park Y, Xu Y, Tian Y, Babusis D, Barauskus O, Peterson BR, Gnatt A, Kashlev M, Zhong W, Cameron CE (2012) Sensitivity of mitochondrial transcription and resistance of RNA polymerase II dependent nuclear transcription to antiviral ribonucleosides. PLoS Pathog 8:e1003030CrossRefPubMedPubMedCentral
2.
Zurück zum Zitat Cholongitas E, Papatheodoridis GV (2014) Sofosbuvir: a novel oral agent for chronic hepatitis C. Ann Gastroenterol 27:331–337PubMedPubMedCentral Cholongitas E, Papatheodoridis GV (2014) Sofosbuvir: a novel oral agent for chronic hepatitis C. Ann Gastroenterol 27:331–337PubMedPubMedCentral
3.
Zurück zum Zitat Coelmont L, Paeshuyse J, Windisch MP, De Clercq E, Bartenschlager R, Neyts J (2006) Ribavirin antagonizes the in vitro anti-hepatitis C virus activity of 2’-C-methylcytidine, the active component of valopicitabine. Antimicrob Agents Chemother 50:3444–3446CrossRefPubMedPubMedCentral Coelmont L, Paeshuyse J, Windisch MP, De Clercq E, Bartenschlager R, Neyts J (2006) Ribavirin antagonizes the in vitro anti-hepatitis C virus activity of 2’-C-methylcytidine, the active component of valopicitabine. Antimicrob Agents Chemother 50:3444–3446CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Dao Thi VL, Debing Y, Wu X, Rice CM, Neyts J, Moradpour D, Gouttenoire J (2016) Sofosbuvir inhibits hepatitis E virus replication in vitro and results in an additive effect when combined with ribavirin. Gastroenterology 150(82–85):e84 Dao Thi VL, Debing Y, Wu X, Rice CM, Neyts J, Moradpour D, Gouttenoire J (2016) Sofosbuvir inhibits hepatitis E virus replication in vitro and results in an additive effect when combined with ribavirin. Gastroenterology 150(82–85):e84
5.
6.
Zurück zum Zitat Debing Y, Winton J, Neyts J, Dallmeier K (2013) Cutthroat trout virus as a surrogate in vitro infection model for testing inhibitors of hepatitis E virus replication. Antiviral Res 100:98–101CrossRefPubMed Debing Y, Winton J, Neyts J, Dallmeier K (2013) Cutthroat trout virus as a surrogate in vitro infection model for testing inhibitors of hepatitis E virus replication. Antiviral Res 100:98–101CrossRefPubMed
7.
Zurück zum Zitat Debing Y, Gisa A, Dallmeier K, Pischke S, Bremer B, Manns M, Wedemeyer H, Suneetha PV, Neyts J (2014) A mutation in the hepatitis E virus RNA polymerase promotes its replication and associates with ribavirin treatment failure in organ transplant recipients. Gastroenterology 147:1008–1011 e1007 (quiz e1015–1006) CrossRefPubMed Debing Y, Gisa A, Dallmeier K, Pischke S, Bremer B, Manns M, Wedemeyer H, Suneetha PV, Neyts J (2014) A mutation in the hepatitis E virus RNA polymerase promotes its replication and associates with ribavirin treatment failure in organ transplant recipients. Gastroenterology 147:1008–1011 e1007 (quiz e1015–1006) CrossRefPubMed
8.
Zurück zum Zitat Donnelly MC, Imlach SN, Abravanel F, Ramalingam S, Johannessen I, Petrik J, Fraser AR, Campbell JD, Bramley P, Dalton HR, Hayes PC, Kamar N, Simpson KJ (2017) Sofosbuvir and daclatasvir anti-viral therapy fails to clear HEV viremia and restore reactive T cells in a HEV/HCV co-infected liver transplant recipient. Gastroenterology 152:300–301CrossRefPubMed Donnelly MC, Imlach SN, Abravanel F, Ramalingam S, Johannessen I, Petrik J, Fraser AR, Campbell JD, Bramley P, Dalton HR, Hayes PC, Kamar N, Simpson KJ (2017) Sofosbuvir and daclatasvir anti-viral therapy fails to clear HEV viremia and restore reactive T cells in a HEV/HCV co-infected liver transplant recipient. Gastroenterology 152:300–301CrossRefPubMed
9.
Zurück zum Zitat Eyer L, Zouharova D, Sirmarova J, Fojtikova M, Stefanik M, Haviernik J, Nencka R, de Clercq E, Ruzek D (2017) Antiviral activity of the adenosine analogue BCX4430 against West Nile virus and tick-borne flaviviruses. Antiviral Res 142:63–67CrossRefPubMed Eyer L, Zouharova D, Sirmarova J, Fojtikova M, Stefanik M, Haviernik J, Nencka R, de Clercq E, Ruzek D (2017) Antiviral activity of the adenosine analogue BCX4430 against West Nile virus and tick-borne flaviviruses. Antiviral Res 142:63–67CrossRefPubMed
10.
Zurück zum Zitat Feng JY, Xu Y, Barauskas O, Perry JK, Ahmadyar S, Stepan G, Yu H, Babusis D, Park Y, McCutcheon K, Perron M, Schultz BE, Sakowicz R, Ray AS (2016) Role of mitochondrial RNA polymerase in the toxicity of nucleotide inhibitors of hepatitis C Virus. Antimicrob Agents Chemother 60:806–817CrossRefPubMedPubMedCentral Feng JY, Xu Y, Barauskas O, Perry JK, Ahmadyar S, Stepan G, Yu H, Babusis D, Park Y, McCutcheon K, Perron M, Schultz BE, Sakowicz R, Ray AS (2016) Role of mitochondrial RNA polymerase in the toxicity of nucleotide inhibitors of hepatitis C Virus. Antimicrob Agents Chemother 60:806–817CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Fujiwara S, Yokokawa Y, Morino K, Hayasaka K, Kawabata M, Shimizu T (2014) Chronic hepatitis E: a review of the literature. J Viral Hepat 21:78–89CrossRefPubMed Fujiwara S, Yokokawa Y, Morino K, Hayasaka K, Kawabata M, Shimizu T (2014) Chronic hepatitis E: a review of the literature. J Viral Hepat 21:78–89CrossRefPubMed
12.
Zurück zum Zitat Kamar N, Bendall R, Legrand-Abravanel F, Xia NS, Ijaz S, Izopet J, Dalton HR (2012) Hepatitis E. Lancet 379:2477–2488CrossRefPubMed Kamar N, Bendall R, Legrand-Abravanel F, Xia NS, Ijaz S, Izopet J, Dalton HR (2012) Hepatitis E. Lancet 379:2477–2488CrossRefPubMed
13.
Zurück zum Zitat Lee JC, Tseng CK, Wu YH, Kaushik-Basu N, Lin CK, Chen WC, Wu HN (2015) Characterization of the activity of 2’-C-methylcytidine against dengue virus replication. Antiviral Res 116:1–9CrossRefPubMed Lee JC, Tseng CK, Wu YH, Kaushik-Basu N, Lin CK, Chen WC, Wu HN (2015) Characterization of the activity of 2’-C-methylcytidine against dengue virus replication. Antiviral Res 116:1–9CrossRefPubMed
14.
Zurück zum Zitat Pischke S, Hartl J, Pas SD, Lohse AW, Jacobs BC, Van der Eijk AA (2017) Hepatitis E virus: Infection beyond the liver? J Hepatol 66:1082–1095CrossRefPubMed Pischke S, Hartl J, Pas SD, Lohse AW, Jacobs BC, Van der Eijk AA (2017) Hepatitis E virus: Infection beyond the liver? J Hepatol 66:1082–1095CrossRefPubMed
15.
Zurück zum Zitat Rocha-Pereira J, Jochmans D, Dallmeier K, Leyssen P, Cunha R, Costa I, Nascimento MS, Neyts J (2012) Inhibition of norovirus replication by the nucleoside analogue 2’-C-methylcytidine. Biochem Biophys Res Commun 427:796–800CrossRefPubMed Rocha-Pereira J, Jochmans D, Dallmeier K, Leyssen P, Cunha R, Costa I, Nascimento MS, Neyts J (2012) Inhibition of norovirus replication by the nucleoside analogue 2’-C-methylcytidine. Biochem Biophys Res Commun 427:796–800CrossRefPubMed
16.
Zurück zum Zitat Shukla P, Nguyen HT, Faulk K, Mather K, Torian U, Engle RE, Emerson SU (2012) Adaptation of a genotype 3 hepatitis E virus to efficient growth in cell culture depends on an inserted human gene segment acquired by recombination. J Virol 86:5697–5707CrossRefPubMedPubMedCentral Shukla P, Nguyen HT, Faulk K, Mather K, Torian U, Engle RE, Emerson SU (2012) Adaptation of a genotype 3 hepatitis E virus to efficient growth in cell culture depends on an inserted human gene segment acquired by recombination. J Virol 86:5697–5707CrossRefPubMedPubMedCentral
17.
Zurück zum Zitat Todt D, Gisa A, Radonic A, Nitsche A, Behrendt P, Suneetha PV, Pischke S, Bremer B, Brown RJ, Manns MP, Cornberg M, Bock CT, Steinmann E, Wedemeyer H (2016) In vivo evidence for ribavirin-induced mutagenesis of the hepatitis E virus genome. Gut 65:1733–1743CrossRefPubMedPubMedCentral Todt D, Gisa A, Radonic A, Nitsche A, Behrendt P, Suneetha PV, Pischke S, Bremer B, Brown RJ, Manns MP, Cornberg M, Bock CT, Steinmann E, Wedemeyer H (2016) In vivo evidence for ribavirin-induced mutagenesis of the hepatitis E virus genome. Gut 65:1733–1743CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat van der Valk M, Zaaijer HL, Kater AP, Schinkel J (2017) Sofosbuvir shows antiviral activity in a patient with chronic hepatitis E virus infection. J Hepatol 66:242–243CrossRefPubMed van der Valk M, Zaaijer HL, Kater AP, Schinkel J (2017) Sofosbuvir shows antiviral activity in a patient with chronic hepatitis E virus infection. J Hepatol 66:242–243CrossRefPubMed
19.
Zurück zum Zitat Wang W, Hakim MS, Nair VP, de Ruiter PE, Huang F, Sprengers D, Van Der Laan LJ, Peppelenbosch MP, Surjit M, Pan Q (2016) Distinct antiviral potency of sofosbuvir against hepatitis C and E viruses. Gastroenterology 151:1251–1253CrossRefPubMed Wang W, Hakim MS, Nair VP, de Ruiter PE, Huang F, Sprengers D, Van Der Laan LJ, Peppelenbosch MP, Surjit M, Pan Q (2016) Distinct antiviral potency of sofosbuvir against hepatitis C and E viruses. Gastroenterology 151:1251–1253CrossRefPubMed
20.
Zurück zum Zitat Zhou X, de Man RA, de Knegt RJ, Metselaar HJ, Peppelenbosch MP, Pan Q (2013) Epidemiology and management of chronic hepatitis E infection in solid organ transplantation: a comprehensive literature review. Rev Med Virol 23:295–304CrossRefPubMed Zhou X, de Man RA, de Knegt RJ, Metselaar HJ, Peppelenbosch MP, Pan Q (2013) Epidemiology and management of chronic hepatitis E infection in solid organ transplantation: a comprehensive literature review. Rev Med Virol 23:295–304CrossRefPubMed
21.
Zurück zum Zitat Zhou X, Huang F, Xu L, Lin Z, de Vrij FM, Ayo-Martin AC, van der Kroeg M, Zhao M, Yin Y, Wang W, Cao W, Wang Y, Kushner SA, Peron JM, Alric L, de Man RA, Jacobs BC, van Eijk JJ, Aronica EM, Sprengers D, Metselaar HJ, de Zeeuw CI, Dalton HR, Kamar N, Peppelenbosch MP, Pan Q (2017) Hepatitis E virus infects neurons and brains. J Infect Dis 215:1197–1206CrossRefPubMed Zhou X, Huang F, Xu L, Lin Z, de Vrij FM, Ayo-Martin AC, van der Kroeg M, Zhao M, Yin Y, Wang W, Cao W, Wang Y, Kushner SA, Peron JM, Alric L, de Man RA, Jacobs BC, van Eijk JJ, Aronica EM, Sprengers D, Metselaar HJ, de Zeeuw CI, Dalton HR, Kamar N, Peppelenbosch MP, Pan Q (2017) Hepatitis E virus infects neurons and brains. J Infect Dis 215:1197–1206CrossRefPubMed
Metadaten
Titel
Nucleoside analogue 2’-C-methylcytidine inhibits hepatitis E virus replication but antagonizes ribavirin
verfasst von
Changbo Qu
Lei Xu
Yuebang Yin
Maikel P. Peppelenbosch
Qiuwei Pan
Wenshi Wang
Publikationsdatum
16.06.2017
Verlag
Springer Vienna
Erschienen in
Archives of Virology / Ausgabe 10/2017
Print ISSN: 0304-8608
Elektronische ISSN: 1432-8798
DOI
https://doi.org/10.1007/s00705-017-3444-8

Weitere Artikel der Ausgabe 10/2017

Archives of Virology 10/2017 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.