You are on page 1of 13

Antiviral Research 150 (2018) 47–59

Contents lists available at ScienceDirect

Antiviral Research
journal homepage: www.elsevier.com/locate/antiviral

A natural small molecule inhibitor corilagin blocks HCV replication and T


modulates oxidative stress to reduce liver damage
B. Uma Reddya, Ranajoy Mullicka, Anuj Kumara, Geetika Sharmaa, Paromita Baga,
Chaitrali Laha Roya, Govindarajan Sudhab, Himani Tandonb, Pratik Davea, Ashutosh Shuklaa,
Priyanka Srinivasana, Madhusudhan Nandhithaa, Narayanaswamy Srinivasanb, Saumitra Dasa,∗
a
Department of Microbiology and Cell Biology, Indian Institute of Science, Bangalore, 560012, India
b
Molecular Biophysics Unit, Indian Institute of Science, Bangalore, 560012, India

A R T I C L E I N F O A B S T R A C T

Keywords: Hepatitis C virus (HCV) infection causes chronic liver disease, which often leads to hepatocellular carcinoma.
Hepatitis C virus Earlier, we have demonstrated anti-HCV property of the methanolic extract of Phyllanthus amarus, an age-old
Antiviral folk-medicine against viral hepatitis. Here, we report identification of a principal bioactive component ‘cor-
Small molecule inhibitor ilagin’, which showed significant inhibition of the HCV key enzymes, NS3 protease and NS5B RNA-dependent-
Oxidative stress
RNA-polymerase. This pure compound could effectively inhibit viral replication in the infectious cell culture
system, displayed strong antioxidant activity by blocking HCV induced generation of reactive oxygen species and
suppressed up-regulation of NOX4 and TGF-β mRNA levels. Oral administration of corilagin in BALB/c mice
demonstrated its better tolerability and systemic bioavailability. More importantly, corilagin could restrict
serum HCV RNA levels, decrease collagen deposition and hepatic cell denaturation in HCV infected chimeric
mice harbouring human hepatocytes. Taken together, results provide a basis towards developing a pure natural
drug as an alternate therapeutic strategy for restricting viral replication and prevent liver damage towards better
management of HCV induced pathogenesis.

1. Introduction DAAs based regimens could be further explored as one of the most
commendable alternate strategy at this moment [Seeff et al., 2008;
Hepatitis C virus (HCV), a member of Flaviviridae family, has in- Ganta et al., 2017].
fected over 170 million people worldwide and is the leading cause of Several classes of plant extracts and their derivatives such as puni-
liver fibrosis, cirrhosis and hepatocellular carcinoma [Averhoff et al., calagin [Reddy et al., 2014], silymarin [Wagoner et al., 2010] and
2012]. There is no vaccine available and the previous standard-of- curcumin [Chen et al., 2012] were reported to exhibit anti-HCV prop-
care HCV therapy consisting pegylated interferon alpha and ribavirin erties. Earlier, we reported that methanolic extract of Phyllanthus
provides only 50% sustained virological response (SVR). Recent ad- amarus could inhibit HCV RNA replication by inhibiting the NS3 serine
dition of direct-acting antivirals (DAAs) has revolutionized hepatitis C protease and NS5B-RdRp enzyme activities [Ravikumar et al., 2011].
virus (HCV) patient care and offers cure to many patients worldwide Different bioactive candidates of P. amarus, particularly polyphenols,
[Pawlotsky et al., 2016]. However, it is impossible to provide DAAs to showed wide spectrum of pharmacological activities including anti-
170 million chronically infected patients [Bankwitz et al., 2017]. HBV [Ott et al., 1997] and anti-HIV [Notka et al., 2004] properties by
Further, treatment-induced cure does not protect from HCV re-infec- regulating various signalling pathways.
tion [Bankwitz et al., 2017]. Moreover, HCV prevention in high-risk In the present study, we identified corilagin as the principal
groups has seen only limited improvement with DAAs [Liang, 2013]. bioactive component from P. amarus, which showed significant in-
Thus, use of natural plant products to supplement or circumvent IFN/ hibition of HCV enzymes and replication. It also prevented HCV

Abbreviations: HCV, Hepatitis C virus; DAAs, direct-acting antivirals; SVR, Sustained Virological Response; EGFP, enhanced-green-fluorescent-protein; RdRp, RNA dependent RNA
polymerase; JFH1, strain from a patient with fulminant hepatitis; TGFβ, Transforming growth factor beta; Huh 7, human hepatocellular carcinoma cells; ROS, Reactive Oxygen Species;
RIPA buffer, Radioimmunoprecipitation assay buffer; Poly(A), polyadenylation; UMP, Uridine monophosphate; OECD, Organisation for Economic Co-operation and Development; NF-κB,
Nuclear Factor Kappa B; ERK, extracellular signal–regulated kinase

Corresponding author.
E-mail address: sdas@iisc.ac.in (S. Das).

https://doi.org/10.1016/j.antiviral.2017.12.004
Received 24 March 2017; Received in revised form 1 December 2017; Accepted 6 December 2017
Available online 07 December 2017
0166-3542/ © 2017 Elsevier B.V. All rights reserved.
B.U. Reddy et al. Antiviral Research 150 (2018) 47–59

(caption on next page)

mediated generation of reactive oxygen species (ROS) and offered 2. Materials and methods
protection against liver damage by modulating oxidative stress.
Corilagin was found to be non-toxic and systemically bioavailable in- 2.1. Plasmids
vivo. An oral administration of corilagin could restrict serum HCV-RNA
levels and reduce the amount of collagen deposition in HCV infected Plasmids pYB-43 encoding single-chain NS3/4A and pYB-44 ex-
chimeric PXB-mice harboring human hepatocytes. pressing fusion protein consisting an enhanced-green-fluorescent-

48
B.U. Reddy et al. Antiviral Research 150 (2018) 47–59

Fig. 1. Effect of different fractions and corilagin from P. amarus on HCV enzymes. (A) NS3 protease assay: The crude leaf methanolic extract and its fractions-1, 2, 3 and residue-3 were
incubated in increasing concentrations (1, 2.5, 5, 7.5 and 10 μg/ml) with recombinant HCV NS3-protease and fusion substrate protein (EGFP-NS5A/B-CBD). Cleavage of this substrate by
NS3 results in emission of fluorescent light that is easily detected and quantitated by fluorimetry. [‘C’ represents vehicle control (0.01% DMSO)], ‘MeOH ext’ represents crude leaf
methanolic extract of Phyllanthus amarus. Fraction-1, 2, 3 and Residue-3 correspond to n-hexane, chloroform, ethyl acetate fractions and left over residue of crude extract, respectively.
The relative NS3 protease activity was normalized with the vehicle control. (B) NS5B polymerase assay: The purified recombinant NS5B protein, poly-A template and oligo dT12 were pre-
incubated with or without fractions (1, 2 and 3) at two concentrations (5 and 30 μg/mL) followed by the addition of α-32P-UTPs. Incorporation of radiolabelled UMPs into RNA products
was quantified using a liquid scintillation counter. Here, ‘C’ represents the relative NS5B RdRp activity normalized with the DMSO vehicle control. (C) Chemical structure of corilagin
(drawn by using ACD/chemsketch freeware). (D) Similar to panel A, NS3 protease assay was also performed with increasing concentrations (0.1, 0.5, 0.75, 1, 2, 5, 15, 30 μM) of purified
corilagin. Telaprevir (Tela, 1 μM) and hypophyllanthin (Hypo, 30 μM) were used as positive and negative controls respectively. (E) Effect of corilagin (at 25, 50 and 100 μM) on cellular
trypsin activity against FITC conjugated casein substrate was determined by fluorimetry. Here, trypsin inhibitor (at 10, 25, 50 μM) from soybean was used as a positive control (F) NS5B
assay was carried out using increasing concentrations (5, 20 and 30 μM) of corilagin. Sofuosbuvir (5 μM) was used as a positive control of inhibition. ‘C’ denotes vehicle control. Results
are expressed as mean percentage of inhibition with error bars of standard deviations from the three independent experiments.

Table 1 2.4. Extraction and bioassay guided fractionation of active constituents


Effect of Phyllanthus amarus and its fractions on HCV enzymes activities.
Dried leaves of P. amarus were refluxed with 90% ethanol. The re-
Phyllanthus amarus HCV NS3/4A protease HCV NS5BCΔ21RdRp assay
(dried leaf) assay (IC50) (IC50)
sultant crude extract was partitioned successively from non-polar to
polar solvents and concentrated by rotary evaporator (IKA, Germany).
MeOH extract (ME) < 1.0 μg/ml 5.00 μg/ml Fractions-1, 2, 3 and residue-3 were evaluated for their inhibitory po-
[Ravikumar et al., 2011] tentials against HCV enzymes (NS3 and NS5B). Major compound from
Fraction-1 of ME 5.0 μg/ml Could not achieve IC50
(n-hexane)
the active fraction-3 was purified by column chromatography and
Fraction-2 of ME 2.5 μg/ml 10.0 μg/ml confirmed for its purity and identity by analytical HPLC
(Chloroform) (Supplementary Fig. S1).
Fraction-3 of ME < 1.0 μg/ml 10.0 μg/ml
(Ethyl acetate)

2.5. HCV NS3 protease based cleavage assay


Table 2
Effect of corilagin on HCV enzyme activities, replication and cell viability. To examine the effect of corilagin on the HCV NS3/4A protease
activity, a high throughput in-vitro fluorimetric assay [Berdichevsky
Compound HCV NS3/ HCV NS5B HCV JFH1 Cell Viability Selectivity et al., 2003] was performed in black 96-well microtiter plate. The single
4A RdRp Replication assay (CC50) Index Ratio
chain NS3/4A protease (scNS3/4A from HCV genotype 1b) and fusion
Protease assay (IC50) (EC50) (SI =
assay CC50/EC50)
protein (EGFP-NS5A/B-CBD) were overexpressed in E. coli BL21 cells
(IC50) and purified. The purified enzyme scNS3/4A (0.1 μM) was pre-in-
cubated with increasing concentrations of test samples followed by the
Corilagin < 1.0 μM 20.0 μM < 50.0 μM > 1.0 mM > 20.0 addition of purified fusion substrate protein (EGFP-NS5A/B-CBD,
0.5 μM) and further incubation for another one hour at 37 °C. The re-
action was terminated by adding the cellulose slurry (40 μM) into the
protein (EGFP), cellulose binding domain and NS3 cleavage site at the
reaction mix followed by rocking the tubes. Then, tubes were cen-
middle [EGFP-NS5A/B-CBD] were received from Dr. Itai Benhar, Tel
trifuged, and the supernatants were separated. Finally, the total fluor-
Aviv University, Israel [Berdichevsky et al., 2003]. Plasmid
escence signal evolved from the cleaved EGFP substrate in the super-
pThNS5BCΔ21 [Kaushik-Basu et al., 2008] was obtained from Dr.
natant was quantified by fluorimeter (Modulus TM Microplate
Neerja Kaushik-Basu, UMDNJ-New Jersey Medical School, USA. The
Multimode Reader) using excitation filter 484 nm and emission filter
HCV JFH-1 plasmid construct [Wakita et al., 2005] and pSGR-Luc/JFHI
538 nm. Telaprevir (MedChem Express) was used as a positive control.
was a generous gift from Dr. Takaji Wakita, National Institute of In-
fectious Diseases, Tokyo [Kato et al., 2005]. Myc-tagged-HCV NS5A
clone was received from Dr. Krishnan H Harshan from Centre for Cel-
2.6. Cellular protease assay
lular and Molecular Biology, India.

A simple high throughput in-vitro cellular serine protease (trypsin)


2.2. Cell lines assay was performed as per the manufacturer's instructions using pro-
tease fluorescent detection kit (PF0100, Sigma-Aldrich). In this assay,
HCV replicon harboring hepatoma cell line (Rep2a) was obtained cellular trypsin recognizes and cleaves casein substrate labelled with
from Dr. Ralf Bartenschlager, Heidelberg University [Frese et al., 2003]. fluorescein isothiocyanate (FITC). The intensity of fluorescence signal
Huh7.5 cell line was received from Dr. C. M. Rice, Rockefeller Uni- evolving from the cleaved FITC conjugated casein substrate was
versity, USA. Cells were cultured in Dulbecco's modified Eagle's quantified by fluorometer using excitation filter 485 nm and emission
medium (DMEM; Sigma-Aldrich) supplemented with 10% fetal bovine filter 538 nm in the presence or absence of test compounds. The trypsin
serum (FBS; Gibco), 100 U/ml of penicillin (HiMedia), and 100 μg/ml inhibitor from Glycine max (Sigma Aldrich) was used as positive con-
of streptomycin sulphate (HiMedia) at 37 °C in 5% CO2 conditions. trol.
Hygromycin (25 μg/ml) was additionally used to culture replicon
containing cells.
2.7. Western blotting
2.3. Collection and identification of plant
Cells were harvested, and cell lysates were prepared using radio-
Fresh plants of Phyllanthus amarus Schum and Thonn immunoprecipitation (RIPA) lysis buffer. The proteins from these ly-
(Euphorbiaceae) were collected from GKVK campus, Bangalore, and sates were resolved in SDS PAGE and transferred onto nitrocellulose
identified. The herbarium was prepared and deposited with voucher membrane. The protein of interest was detected using respective pri-
specimen number 74099 in the Foundation for Revitalisation of Local mary antibody followed by HRP conjugated secondary antibody and
Health Traditions, Bangalore. enhanced chemiluminescence method.

49
B.U. Reddy et al. Antiviral Research 150 (2018) 47–59

(caption on next page)

50
B.U. Reddy et al. Antiviral Research 150 (2018) 47–59

Fig. 2. Effect of corilagin on cell viability and HCV replication (A) Huh7 cells were treated with increasing concentrations of corilagin (0.1–1.0 mM) followed by measurement of cell
viability by measuring the absorbance at 560 nm after treatment with MTT reagent. ‘C’ represents Huh7 cells with no treatment as control. (B) Huh7.5 cells were infected with JFH-1
followed by corilagin treatment (10–200 μM) and cell viability was determined; ‘C’ represents JFH1 infected Huh7.5 cells as control. (C) Schematic representation of pSGR-luc/JFH1
(adapted from Kato et al., 2005). Huh 7 cells were transfected with HCV pSGR-Luc/JFH1 RNA and treated with increasing concentrations (10, 25, 50, 100, 200 μM) of corilagin. No
treatment was used as a control i.e., ‘C’ (representing 100% luciferase activity). Telaprevir (Tela, 20 μM) was used as a positive control. (D) Schematic representation of p-JFH1 (adapted
from Wakita et al., 2005). Huh7.5 cells were infected with JFH-1 for 4 h and treated with increasing concentrations of corilagin (25, 50, and 100 μM). At 72 h, HCV negative strand
synthesis was quantified by qRT-PCR and data was normalized with GAPDH was used as an internal control. Sofosbuvir (10 μM) was used as a positive control. ‘JFH1’ represents control
i.e., Huh7.5 cells infected with JFH-1 in the absence of any inhibitor. Data represent mean ± SD from three independent experiments.

2.8. HCV NS5B RdRp assay primers used here were as follows:

The RdRp assay was performed as described previously [Kaushik- HCV149F:5′ TGCGGAACCGGTGAGTACA3′
Basu et al., 2008]. Briefly, the purified NS5BCΔ21(HCV genotype 1b), HCV342R:5′CTTAAGGTTTAGGATTCGTGCTCAT 3′
poly (A) template (Amersham) and oligo-dT12 were incubated on ice GAPDH F:5′ CATGAGAAGTATGACAACAGCCT 3′
with or without test samples. This was followed by addition of α-32P- GAPDH R: 5′ AGTCCTTCCACGATACCAAAGT 3′
UTPs and further incubation for another 1 h at 30 °C. Reactions were
terminated by 5% ice cold trichloroacetic acid in 0.5 mM sodium pyr-
2.14. Measurement of intracellular ROS production
ophosphate. Resulting polymeric RNAs were precipitated at −20 °C for
1 h and transferred to GF/C filters (Sigma-Aldrich). These filters were
Induction of intracellular ROS production in Huh7.5 cells by JFH-1
washed successively with 5% trichloroacetic acid and distilled water.
infection or Huh7 cells by over expression of HCV proteins (core and
Filters were dried at room temperature. The amount of radiolabelled
NS5A) in the presence or absence of corilagin was quantified by mea-
UMPs incorporated into RNA products present on the filters was
suring fluorescent signals obtained from the cleaved DCFDA (2′,7′ –
quantified using a liquid scintillation counter.
dichlorofluorescindiacetate) by fluorimeter. Here, H2O2 (0.03%) was
used as positive ROS inducer. Additionally, the intracellular ROS pro-
2.9. MTT assay
duction in Huh7.5-core stable cells (4 × 104) with or without corilagin
was analyzed by FACS (BD Biosciences).
Cytotoxic effects of corilagin on Huh7 cells and HCV infected
Huh7.5 cells were assessed 48 h post-treatment by using MTT reagent [3-
(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] (Sigma- 2.15. Estimation of NOX4/TGF-β mRNA levels
Aldrich) as per the standard protocol [Ciofani et al., 2010].
Hepatic fibrosis is a hallmark of hepatitis C virus infection. Viral
2.10. HCV replication assay in replicon 2a cells hepatitis results in increased level of NOX4 (a member of the NADPH
oxidase, Nox family), which is a direct contributor of oxidative stress in
The Huh7 cells harbouring HCV replicon 2a were treated with a TGF-β mediated pathway [Boudreau et al., 2009]. The levels of HCV
corilagin and incubated for additional 24 h. Then total RNA was ex- induced NOX4/TGF-β mRNA expressions in JFH-1 infected
tracted using TRIzol reagent (Sigma Aldrich). The HCV RNA levels were Huh7.5 cells and Huh7 cells over expressed with HCV proteins (core
determined using real time RT-PCR as described below. Sofosbuvir and NSS5A) in the presence or absence of corilagin treatment were
(MyHep™, Gilead Sciences, Ireland) was used as the standard control. quantified by real time RT-PCR using NOX4 and TGF-β specific primers.
GAPDH expression served as an internal control.
2.11. Luciferase assay Following primers were used:

Huh7 cells were transfected with pSGR-Luc/JFH1 RNA using lipo- Nox4 F: 5′ TGGCTCTCCATGAATGTCCT 3′
fectamine-2000 (Invitrogen). At 4 h post transfection, cells were treated Nox4R: 5′CTTAGACACAATCCCCCAACA3′
with increasing concentrations of corilagin. After 24 h, cells were lysed, TGF-βF: 5′TGCCCCCTCTGCCACCTGGGGCGGT 3′
and luciferase assay was performed using dual luciferase assay kit TGF-βR: 5′ TCGGCCAGGGCGGAGCGCAGCAGG 3’.
(Promega). The luciferase signals were measured using luminometer
and the TCID50 (50% effective HCV replication inhibitory concentra- 2.16. Analysis of TGF-β pathway by Cignal finder 10 pathway reporter
tion) of corilagin was calculated. array

2.12. HCV replication assay using HCV JFH-1 The transcription factors/signalling molecules involved in the TGF-
β pathway was investigated using Cignal Finder Immune Signalling 10-
Huh 7.5 cells were infected with HCV JFH-1 (genotype 2a) virus pathway Reporter Luciferase Kit (CCA-008L, SA Biosciences) following
(RNA titer 1 × 1010 copies/ml). At 4 h post infection, cells were wa- manufacturer's instructions.
shed with PBS, changed to complete medium and treated with in-
creasing concentrations of corilagin. At 72 h post-treatment, cells were
2.17. Ethics statement for animal studies
harvested, and total cellular RNA was isolated by Trizol method and
subjected to real time-PCR.
Animal experiments were approved by ‘Institutional Animal Ethics
Committee’. Animals were maintained in accordance with the guide-
2.13. Real Time-PCR
lines of the Indian National Law on animal care and use.
Total cellular RNA was reverse transcribed using HCV forward
primer (HCV149F) corresponding to IRES region and GAPDH reverse 2.18. Determination of systemic toxicity in mice
primer (GAPDH R). cDNA was diluted (1: 2) and added in SYBR Green
PCR Master Mix and subjected to amplification using real-time ABI For the safety assessment, the acute and sub-acute toxicity of cor-
PRISM 7900 (Applied Biosystems) PCR machine. GAPDH (a house- ilagin was determined according to OECD guidelines [OECD-423 and
keeping gene) was used as an internal control for normalization. The OECD-407].

51
B.U. Reddy et al. Antiviral Research 150 (2018) 47–59

(caption on next page)

52
B.U. Reddy et al. Antiviral Research 150 (2018) 47–59

Fig. 3. Effect of corilagin on HCV induced ROS production. (A) Huh7 cells were either incubated with H2O2 (0.03%) (positive control) or transiently transfected with HCV proteins (core
and NS5A) and then treated cells with increasing concentrations of purified corilagin (25–100 μM). At 48 h post transfection cells were treated with DCFDA stain. The amount of ROS
production was quantified by measuring the amount of fluorescence produced upon oxidation of DCFDA to fluorescent DCF on fluorescent plate reader. Here, ‘C’ represents cells alone,
‘vc’ is cells treated with only DMSO (0.01%) as vehicle control. (B) For the experiment performed in panel A, the level of HCV core was evaluated by Western blotting using anti-core
antibody. (C) In order to reconfirm the ROS inhibitory effects of corilagin, Huh7.5-cells (4 × 104) stably expressing core were treated with corilagin at concentration 50–100 μM and
incubated for 48 h. Cells were then stained with 10 μM of DCFDA and results were analyzed with BD FACS Diva software. The peak ‘a’ represents Huh7.5-core stable cells that are neither
treated with DCFDA stain nor with pure corilagin. The peak ‘b’ represents Huh7.5-core stable cells treated with only DCFDA (10 μM). The peaks ‘c’ and ‘d’ represents Huh7.5-core stable
cells treated with DCFDA (10 μM) and increasing concentrations of pure corilagin (50–100 μM). (D) The quantification of the data from panel ‘C’ is shown here in the form of a bar graph.
(E and F) Experiment similar to panel A, was performed with Huh7.5 cells infected with JFH1 in the presence or absence of corilagin (25–100 μM). Here, fluorescence intensity was
measured at 72 h post infection. Corresponding HCV RNA levels were also measured and plotted as fold change.

2.19. Bioavailability of corilagin in mice and rats verses treated) were done by two-tailed unpaired t-test using GraphPad
Prism 6.0 software and significant differences between the groups are
The bioavailability of corilagin in BALB/c mice and Wistar rats were represented by an asterisk (*p < 0.05, **p < 0.01 and
measured using protocol described previously [Haid et al., 2012]. ***p < 0.001).

2.20. In-vivo tolerability and efficacy of corilagin in HCV chimeric mice 3. Results
model
3.1. Fractionation and purification of the anti-HCV components of P.
In order to demonstrate the safety of corilagin, 1 mM/day of cor- amarus
ilagin was administered orally for 7 days to chimeric mice harboring
human liver (engraftment of primary human hepatocytes is around In order to identify the bioactive constituents of P. amarus, its crude
70–80% in uPA-SCID mice, carried at Phoenix Bio Inc. Japan). After leaf methanolic (MeOH) extract and fractions 1 to 3 (n-hexane,
confirming its tolerability in chimeric mice, the therapeutic efficacy of chloroform, ethyl acetate, respectively) were assessed for both HCV-
corilagin was explored. To study this, first uPA-SCID mice were en- NS3 protease and NS5B-RdRp inhibitory activities. All the fractions
grafted with human hepatocytes (> 70% replacement), then these an- were found to inhibit NS3 protease in a dose dependent manner but
imals were infected with HCV-1b genotype and kept them under ob- fraction-3 showed highest inhibitory activity (Fig. 1A and Table 1).
servation for 3 weeks to reach the HCV RNA titre at optimum levels Interestingly, fractions 2 and- 3 also showed significant inhibition of
prior to challenge study. During this course of period, the HCV positive NS5B activity (Fig. 1B) suggesting these could be potent inhibitors of
strand RNA copies in the sera of chimeric mice were determined reg- HCV replication. Since fraction 3 was active against both the viral en-
ularly every week by qRT-PCR. Those mice in which serum HCV RNA zymes, the corresponding principal bioactive constituent of fraction 3
copy number achieved to ≥107, were selected for efficacy study and was identified as corilagin (Fig. 1C) by high performance liquid chro-
randomly assigned into two groups of 3 mice each. Group-1 mice (ad- matography and confirmed by LC-ESI-MS (Supplementary Fig. S1 and
ministered with water) were used as control. Group- 2 mice were S2).
treated with corilagin for 14 days. During this treatment window, body
weight and h-albumin (a direct readout of replacement of human he-
3.2. Corilagin inhibits HCV NS3/4A protease and NS5B RdRp activities in-
patocytes into chimeric mice liver) levels of mice from both groups
vitro
were recorded. HCV serum RNA from infected mice serum was ex-
tracted on 0th, 7th and 14th day of treatment from both the group and
HCV encoded NS3 protease is essential for viral replication [Reed
quantified by qRT-PCR. Here, the level of HCV RNA copies on 0th day of
and Rice, 2000; Kolykhalov et al., 2000] and has long been considered
corilagin treatment was approximately ≥107 in chimeric mice sera.
as an attractive target for the therapeutic intervention [Lamarre et al.,
Subsequently the treatment of corilagin was continued for 14 con-
2003]. Results from both flourimetric and gel based cleavage assays
secutive days. The HCV RNA levels in the serum of chimeric mice
indicate that corilagin significantly inhibited the NS3 protease activity
treated with corilagin (group 2) were compared with the chimeric mice
in a dose dependent manner (IC50 < 1 μM) (Fig. 1D, Supplementary
treated with water (group 1). This experiment was done through con-
Fig S3A, Table 2). However, corilagin did not show inhibitory effect on
tract research with PhoenixBio, Japan.
the cellular trypsin activity in-vitro even at 100 μM concentration
(Fig. 1E). The specificity of corilagin action was also checked by testing
2.21. Histochemical analysis of chimeric mice liver its effect on cleavage of eIF4GI [Kerekatte et al., 1999] by Coxsackie
virus 2A protease in HeLa cells. No reduction in the cleavage of eIF4GI
HCV infection in chimeric mice humanized liver leads to up-reg- by coxsackievirus 2A protease (Supplementary Fig. S3B) suggested that
ulation of fibrotic genes with consequential liver fibrosis and cirrhosis this small molecule inhibitor could be specific for HCV NS3 protease.
[Keng et al., 2016]. To examine the amount of fibrillar collagen de- The effect of corilagin on the HCV polymerase activity was also
position at five weeks post HCV infection in both control and corilagin checked. The purified corilagin also showed significant inhibition of
treated groups, the histological analysis of NBF fixed chimeric mice NS5B RdRp activity in a concentration dependent manner with an IC50
liver tissues was performed. To distinguish the collagen deposition from of 20 μM (Fig. 1F and Table 2). Since, corilagin was inhibiting both the
the normal hepatocytes into the processed liver sections, collagen viral enzymes (NS3/4A protease and NS5B polymerase), further we
specific stain, sirius red (Sigma-Aldrich) was used. Here, fast green stain made an attempt to understand the structural interactions of corilagin
was used to stain the normal hepatocytes [Bility et al., 2014]. Image J with these HCV enzymes. For this we performed structure guided
software was used to quantify the percent area of collagen distribution docking studies. These studies were performed using the available 3D
in HCV infected chimeric mice liver sections. structures in PubChem and PDB databases to understand the possible
nature of interactions between corilagin and viral proteins, NS3 and
2.22. Software and statistical analyses NS5B (as explained in Supplementary text, Schrodinger). The results of
these docking studies support that corilagin can inhibit both NS3 pro-
All data are representative of a minimum of three independent ex- tease and NS5B polymerase activities. This can be attributed to its in-
periments. The data are expressed as the mean ± standard deviations teractions with crucial amino acid residues of viral enzymes, such as
(SD). Statistical comparisons between two independent groups (control catalytic triad and β-hairpin of NS3 and NS5B enzymes respectively

53
B.U. Reddy et al. Antiviral Research 150 (2018) 47–59

(caption on next page)

54
B.U. Reddy et al. Antiviral Research 150 (2018) 47–59

Fig. 4. Effect of corilagin on HCV mediated induction of NOX4 and TGF-β expressions. (A) Quantification of the cellular NOX4 mRNA levels from HCV infected Huh7.5 cells also HCV
core/NS5A over expressing Huh7 cells by real time qPCR in the presence or absence of corilagin. (B) Similarly, the quantification of the cellular TGF-β mRNA levels from HCV infected
Huh7.5 cells as well as HCV core and NS5A over expressing Huh7 cells in the presence or absence of corilagin by qRT-PCR. C represents control (either JFH1 infection, HCV core, NS5A
over expression) in respective treatment groups. (C–D) Huh7 cells were over expressed with HCV core and NS5A in the presence of corilagin. The levels of these proteins (core and NS5A)
were checked by Western blotting using anti-core and anti-myc antibodies (Here ‘+’ = 25 μM, ‘++’ = 50 μM and ‘+++’ = 100 μM of corilagin). (E) Analysis of the 10-pathway
reporter assay for TGF-β pathway from uninfected and infected Huh7.5 cells treated with or without corilagin. Results were plotted as relative fold change in Fluc/Rluc values (Y axis). X
axis denotes the concentrations of corilagin used in uninfected and infected cells. Results shown as mean ± SD from three biologically independent experiments.

(Supplementary Figure S4). (Fig. 3C and D). Further, we have also confirmed the reduction of ROS
production in HCV-JFH1 infected Huh7.5 cells (Fig. 3E). The corre-
3.3. Corilagin is non-toxic in cell culture sponding decrease in the level of HCV RNA in these cells were found to
be consistent (Fig. 3F) with the reduction in ROS production. In par-
Results based on cell viability assay showed that treatment of cor- allel, we have checked the levels of HCV core protein in JFH1 infection,
ilagin up to 1000 μM to Huh7 cells did not induce any toxicity (Fig. 2A) however, in contrast to core protein over expression experiment
(CC50 > 1 mM). Further, treatment of corilagin in increasing con- (Fig. 3B), the level of HCV core protein in the context of HCV-JFH1
centrations (10–200 μM) rescued HCV-JFH1induced cell death in in- virus (Supplementary Fig S6B) infection didn't decrease upon corilagin
fectious cell culture system (Fig. 2B). treatment.

3.4. Corilagin inhibits HCV replication in cell culture 3.6. Corilagin suppressed the HCV induced ROS production via NOX4 in a
TGF-β dependent pathway
To investigate the effect of corilagin on HCV replication, HCV
genotype 2a sub genomic reporter replicon (pSGR-Luc/JFH1) was used Our results showed that, either JFH-1 infected Huh7.5 cells or over
for transient transfection. Results showed a considerable decrease in expression of HCV proteins, particularly HCV core and NS5A proteins in
luciferase activity upon treatment with corilagin (TCID50 = 29.5 μM) Huh7 cells, significantly upregulated the expression of both NOX4 and
(Fig. 2C). Similarly, HCV RNA levels reduced upon corilagin treatment TGF-β mRNA levels. The treatment of corilagin could dramatically re-
on non-reporter replicon 2a harboring Huh7 cells (Supplementary Fig duce the expression of TGF-β and NOX4 mRNA levels under these
S5A-C). Sofosbuvir was used as a positive control. conditions (Fig. 4A and B; also see Supplementary Tables S2 and S3).
Next, we studied the effect of corilagin on HCV replication using The levels of core and NS5A in overexpressed cells were checked by
JFHI infectious virus. Huh7.5 cells were infected with HCV genotype 2a Western blotting (Fig. 4C and D).
virus JFH1 (Fig. 2D) for 72 h in absence and presence of increasing In addition, we also confirmed the effect of corilagin on the TGF-β
concentrations of corilagin (post incubation). Sofosbuvir was used as levels from uninfected and JFH1 infected Huh7.5 cells by the immune
positive control in this study. The inhibitory effect of corilagin was signalling. Result showed down regulation of the transcription factors
observed against negative strand synthesis in the infectious cell culture SMAD2/3/4 (involved in the TGF-β pathway) upon treatment with
system (EC50 = ∼50 μM). Even though the replication levels are very corilagin (Fig. 4E).
low (Fig. 2D, Table 2 and Supplementary Fig. S5D-E, Supplementary
Table S1). Here, as expected, levels of NS5B decreased in a dose de- 3.7. Corilagin is well tolerable in mice
pendent manner with increasing concentrations of corilagin
(Supplementary Fig. S6B). Next, we have checked the in-vivo tolerability of corilagin in BALB/c
mice. It was observed that an acute dose of corilagin up to 3500 mg/kg
3.5. Corilagin suppressed HCV induced ROS production b. wt. showed maximum tolerance (LD50 lies between 3500 and
5000 mg/kg b.wt.). Suggesting corilagin can be considerably non-toxic
Generation of ROS plays an important role in the development and even at higher dosage (Fig. 5A). In sub-acute toxicity studies, the re-
progression of inflammatory liver disease mediated by HCV. The in- peated oral administrations of corilagin (1000 mg/kg/day b.wt.) for 28
hibitory activity of corilagin (if any) against ROS production was also days to mice did not exhibit any undesirable changes in behavior and
investigated. Several folds increase in the ROS production was observed body weight of the mice (Fig. 5B). Importantly, corilagin treatment did
due to over expression of HCV proteins (core and NS5A) in Huh7 cells not affect the histology of mice liver in both acute and sub-acute toxi-
(Fig. 3A). This increase was significantly decreased upon treatment of city studies (Fig. 5C).
corilagin, proving its potent antioxidant and hepatoprotective proper-
ties. Interestingly, immunoblot analysis showed that corilagin (25, 50 3.8. Orally administered corilagin is systemically transported to mice and
and 100 μM) treatment leads to specific decrease of HCV core protein rat blood plasma
dose dependently in Huh7 cells over expressed with HCV core protein
(Fig. 3B). The levels of core protein were rescued by MG132 (data not As orally administered corilagin did not show any toxicity in acute
shown) suggesting that corilagin might degrade the HCV-core protein and sub-acute toxicity studies, it was important to check if this com-
via proteosomal pathway. Further, corilagin also suppressed the NS5A pound is systemically available in blood or not. For this, 1500 mg/kg
(over expressed) induced ROS (Fig. 3A). However, the levels of NS5A corilagin was orally fed in mice and rats followed by HPLC-ESI-MS
did not show any change upon treatment of corilagin (Supplementary analyses of plasma samples collected at different time points. Results
Fig. S6A). We have also ruled out the possibility that decrease in the showed systemic maximum bioavailability at 2hr (Tmax = 2 h) with half
fluorescence intensity (observed in Fig. 3A) is not due to effect of cor- life time of the compound (t1/2) = ∼6 h and maximum concentration
ilagin in quenching the fluorescence of GFP. For this, we have ad- absorbed (Cmax) = 57.0 and 52.37 μg/ml of blood in mice and rats
ditionally performed an experiment where Huh7 cells were co-trans- respectively (Fig. 5D).
fected with pCDNA vector expressions of HCV-core/NS5A followed by
pCD-GFP and treated with or without corilagin. However, no change in 3.9. Corilagin restricts HCV-RNA levels in chimeric mouse model
GFP fluorescence suggested that corilagin indeed causes ROS suppres-
sion without affecting the fluorescence (Supplementary Fig. S7A-C). To investigate the tolerability and in-vivo efficacy of the corilagin,
Similar inhibitory effect of corilagin against generation of ROS by the HCV challenge chimeric mice model (as described in materials and
Huh 7.5 stable cell line expressing core protein in FACS analyses methods) was used. Tolerability result showed that treatment of

55
B.U. Reddy et al. Antiviral Research 150 (2018) 47–59

Fig. 5. In-vivo toxicity and bioavailability of purified corilagin. (A) Acute toxicity study. Body weights of BALB/c mice (n = 3) were recorded for 14 consecutive days after oral
administration of an acute maximum tolerance dose of corilagin (3500 mg/kg b.wt. p.o.) (B) Sub-acute toxicity study. Repeated oral doses of corilagin (1000 mg/kg b.wt./day p.o.) into
BALB/c mice (n = 3) for 28 consecutive days were administered and body weight was measured. Results from corilagin treated mice in both acute (A) and sub-acute toxicity (B) studies
were compared with mice from their respective control groups treated only sterile water (vehicle control for corilagin). (C) Histopathological examination of mice liver. A representative
picture of the liver sections of mice from different treatment groups in acute toxicity studies {(i) Water control, (ii) corilagin 3500 mg/kg b.wt.}. Similarly, representative pictures of liver
sections of mice from different groups in subacute toxicity studies, {(iii) Water control, (iv) corilagin (1000 mg/kg b.wt./day)}. (D) Pharmacokinetics of corilagin in BALB/c mice and
Wistar rats. An acute dose of corilagin (1500 mg/kg b. wt.) was administered orally to animals. Concentrations of corilagin in plasma at different time points (0–24 h) post treatment was
measured by LC-ESI-MS. Values used to plot AUC are mean ± SD from the three mice or rats (pre-administered with corilagin) at each time point.

corilagin up to 1 mM/day p.o. for 7 consecutive days is well tolerated in Further, to check the therapeutic efficacy of corilagin, HCV (gt-1b)
chimeric mice with no signs of toxicity (behavioural changes and infected chimeric mice were treated with corilagin (1 mM/day p.o.) for
mortality). Body weights of mice were also found to be normal 14 days. No alterations in the body weight ratio (Fig. 6B) or h-albumin
(Fig. 6A). levels (Fig. 6C) were observed. With respect to virus titre, a steady

56
B.U. Reddy et al. Antiviral Research 150 (2018) 47–59

(caption on next page)

57
B.U. Reddy et al. Antiviral Research 150 (2018) 47–59

Fig. 6. The tolerability and efficacy of corilagin in HCV infected chimeric mice. (A) Tolerability study of corilagin in chimeric/non PXB mice, n = 3 (with < 70% replacement index of
primary human hepatocytes). Body weights of these mice were recorded from 0 to 7 days during for repeated administrations of corilagin at a dose of 1 mM/day. (B–C) Efficacy study of
corilagin in HCV infected chimeric/PXB-mice, n = 3 (> 70% replacement index of primary human hepatocytes). To study this, uPA/SCID chimeric mice were infected with HCV
genotype 1b three weeks prior to challenge with corilagin. The body weights and levels of serum h-albumin of post infected PXB mice were monitored daily for 14 days during corilagin (1
mM/day) treatment period (i.e., on 4th and 5th weeks of infected PXB mice). Similar observations were also recorded in the same time for those post infected PXB mice from control
group. (D) Levels of serum HCV RNA titre in control (no treatment) and corilagin treated chimeric/PXB-mice (n = 3). (E) Histochemistry of HCV infected chimeric mice liver: On 15th
day post treatment, post infected chimeric/PXB-mice from control and treated groups were sacrificed, dissected the liver. Then liver sections were examined for collagen deposition using
sirius red and fast green stains (collagen stained with red-coloured by Sirius red stain and normal hepatocytes stained with blueish green-colour by fast green stain). (E i, iii, v)
Representative pictures from the liver sections of three different HCV infected chimeric mice from control group. (E ii, iv, vi) Representative pictures from the liver sections of three
different HCV infected chimeric/PXB-mice from corilagin treated group, (F) Results from the panel E was represented in bar graph format. Here, approximately 10 representative fields
were selected from each HCV infected chimeric mice liver sections of both water control and corilagin treated groups. The image J software was used to quantify the positive area of
collagen distribution from the liver sections. (Fi) Bar graph of control mouse-1 verses corilagin treated mouse-1, (Fii) represents control mouse-2 verses corilagin treated mouse-2, (Fiii)
represents control mouse-3 verses corilagin treated mouse-3. Here, C represents HCV infected control mice (administered only with water). (For interpretation of the references to colour
in this figure legend, the reader is referred to the Web version of this article.)

increase in the control group (treated with water) was observed. expressed alone (Fig. 3B).
However, in the corilagin treated mice group, the virus titer was re- These contrasting but interesting observations may reveal that al-
stricted to a considerable extent (Fig. 6D; Supplementary Table S4). though corilagin decrease ROS production and NOX4/TGF-β mRNA
This study acts a proof of concept for the potential of plant-derived levels, however mechanisms for these are different in core over-
compounds, such as corilagin, to act as antivirals against HCV. expressed cells versus HCV infected cells which need to be further ex-
plored in near future. Previous studies showed that corilagin isolated
3.10. Corilagin treatment alleviates the HCV induced liver fibrosis in from Phyllanthus niruri L. has a potential antitumor effect against
chimeric mice ovarian cancer cells via the targeted action on the TGF-β/ERK/AKT/
Smad signalling pathways [Jia et al., 2013]. Corilagin was also shown
To investigate whether corilagin can offer protection against HCV to be considerably effective in reducing the in-vivo growth of xeno-
induced liver damage, we performed liver histochemical analyses of grafted Hep3B hepatocellular carcinoma cells [Hau et al., 2010]. Reg-
HCV infected PXB chimeric mice liver sections using two stains, sirius ulation of notch signalling pathway by corilagin helps suppressing the
red and fast green specifically to stain fibrillar collagen and normal development of cholangiocarcinoma [Gu et al., 2016]. Corilagin at-
hepatocytes, respectively (Fig. 6E i-vi). Results from the quantification tenuates bleomycin-induced epithelial injury and fibrosis via blocking
of percent area of collagen distribution in corilagin treated group the oxidative stress, release of proinflammatory cytokines and TGF-β1
showed approximately 4 folds reduction in fibrillar collagen deposition and NF-κB signalling [Wang et al., 2014]. Corilagin isolated from P.
in between/around portal tracts as compared to liver sections from the amarus showed a high degree of antiviral activity against HIV infection
control group (Fig. 6F i-iii). Thus, it appears corilagin could suppress [Notka et al., 2004], Human enterovirus 71 and Coxsackie-virus A16
HCV mediated liver fibrosis and hepatic cell denaturation. [Yeo et al., 2015]. Another study showed that, corilagin exhibit anti-
inflammatory effects in HSV 1 infected microglia [Guo et al., 2010].
4. Discussion Corilagin reduces the cytotoxicity induced by EV71 or Coxsackie virus
A16 on Vero cells [Yeo et al., 2015]. All these observations indicate that
There are increasing evidences that viral cure does not eliminate the corilagin might be potentiated in the clinical setting after testing in
risk for developing progressive liver disease once fibrosis is established (pre) clinical trials.
[El-Serag et al., 2016]. Infact, the DAAs might not be effective once The in-vivo HCV challenge study in PXB chimeric mice showed re-
chronic HCV infection reaches liver fibrosis, cirrhosis and HCC stages. striction of HCV RNA levels in the serum at a steady state level upon
Here, we have demonstrated possible use of corilagin (derived from a oral administration of corilagin. Interestingly, administration of cor-
plant P.amarus) in inhibiting HCV replication; oxidative stress as well as ilagin to chimeric mice, significantly reduced HCV induced liver fi-
reducing the HCV induced liver damage. brosis, suggesting that corilagin might possess a direct antifibrotic role.
Corilagin could inhibit HCV NS3 protease enzyme activity at sub- Thus, our findings open up a new strategy offering the use of the an-
micromolar concentrations; it also inhibited NS5B-RdRp activity but at tifibrotic property of corilagin in HCV induced liver fibrosis for better
slightly higher concentrations. management of disease progression.
HCV infection creates oxidative stress that impairs the basic cellular Taken together, it appears, corilagin might have multifaceted role
functions by DNA damage, modifying lipids and proteins and altering for better management of HCV infected liver. Also, the remarkable
mitochondrial membrane potential [Pal et al., 2010; Ivanov et al., tolerance and stability of this compound in blood circulation might
2013]. Our study has demonstrated that corilagin could effectively re- offer greater advantage in reducing the risk of HCV induced patho-
duce HCV induced oxidative stress by suppressing the HCV protein genesis. More importantly, the study unfolds the ultimate mysteries of
induced enhanced expressions of NOX4 mRNA levels via TGF-β medi- the ‘folk medicinal use’ of the wonder plant ‘Phyllanthus amarus’ against
ated signalling pathway implicated in profibrotic processes [Carmona- viral hepatitis in ancient time.
Cuenca et al., 2008]. In fact, our result obtained with core over-
expressed cells is consistent with earlier study which showed that Conflicts of interest
blocking TGF-β signalling with neutralizing antibodies could drastically
abrogate HCV core protein mediated induction of Nox4 and subsequent The authors declare no conflict of interest.
ROS production [Boudreau et al., 2009]. However, the increased core
level in JFH1 cell culture upon corilagin treatment (Supplementary Fig Acknowledgements
S6B) suggests that corilagin might first stabilize core in this system
which may lead to reduced HCV replication (as observed in replication We are thankful to Itai Benhar, Takaji Wakita, Neerja Kaushik-Basu,
assays) by inhibiting HCV IRES activity in subsequent HCV life cycles Ralf Bartenschlager, Charles M. Rice and Krishnan H Harshan for
[Shimoike T et al., 1999; Shimoike T et al., 2006]. Alternatively, Core sharing useful plasmid constructs and cell lines. We are also grateful to
protein is stabilized (protected) from corilagin when probed in the Central animal facility, Microtome, NMR, FACS, Analytical spectro-
context of JFH1 infection (Supplementary Fig S6B). But found to be scopy and other central facilities of our Institute (funded by DST-FIST
decreased upon Corilagin treatment when Core protein is over and DBT-partnership programme).

58
B.U. Reddy et al. Antiviral Research 150 (2018) 47–59

We thank Dr. Upasana Ray and Dr. Partho Sarothi Ray for critical K., Hasegawa, I., Fujiwara, K., Ito, K., Ozasa, A., Mizokami, M., Wakita, T., 2005.
comments on the manuscript and our other laboratory members for the Detection of anti-hepatitis C virus effects of interferon and ribavirin by a sensitive
replicon system. J. Clin. Microbiol. 43, 5679–5684.
helpful discussion. This work was supported by grant from Department Keng, C.T., Sza, C.W., Zheng, D., Zheng, Z., Young, K.S.M., Tan, S.Q., Ong, J.J.Y., Tan,
of Biotechnology (DBT) (BT/01/COE/06/02/10-IISc), Government of S.Y., Loh, E., Upadya, M.H., Kuick, C.H., Hotta, H., Lim, S.G., Tan, T.C., Chang,
India to SD. BUR is supported by the research fellowship from Science K.T.E., Hong, W., Chen, J., Tan, Y.-J., Chen, Q., 2016. Characterization of liver pa-
thogenesis, human immune responses and drug testing in a humanised mouse model
and Engineering Research Board, DST. AK, GS and PD were supported of HCV infection. Gut 65, 1744–1753.
by the research fellowship from CSIR. SD and NS also acknowledge JC Kerekatte, V., Keiper, B.D., Badorff, C., Cai, A., Knowlton, K.U., Rhoads, R.E., 1999.
Bose fellowship from DST for research support. Cleavage of Poly(A)-binding protein by coxsakievirus 2A protease in vitro and in
vivo: another mechanism for host protein synthesis shutoff? J. Virol. 73, 709–717.
Kaushik-Basu, N., Bopda-Waffo, A., Talele, T.T., Basu, A., Costa, P.R., da Silva, A.J.,
Appendix A. Supplementary data Sarafianos, S.G., Noel, F., 2008. Identification and characterization of coumestans as
novel HCV NS5B polymerase inhibitors. Nucleic Acids Res. 36, 1482–1496.
Kolykhalov, A.A., Mihalik, K., Feinstone, S.M., Rice, C.M., 2000. Hepatitis C virus-en-
Supplementary data related to this article can be found at http://dx.
coded enzymatic activities and conserved RNA elements in the 3’ nontranslated re-
doi.org/10.1016/j.antiviral.2017.12.004. gion are essential for virus replication in vivo. J. Virol. 74, 2046–2051.
Lamarre, D., Anderson, P.C., Bailey, M., Beaulieu, P., Bolger, G., Bonneau, P., Bös, M.,
References Cameron, D.R., Cartier, M., Cordingley, M.G., Faucher, A.M., Goudreau, N., Kawai,
S.H., Kukolj, G., Lagacé, L., LaPlante, S.R., Narjes, H., Poupart, M.A., Rancourt, J.,
Sentjens, R.E., St George, R., Simoneau, B., Steinmann, G., Thibeault, D., Tsantrizos,
Averhoff, F.M., Glass, N., Holtzman, D., 2012. Global burden of hepatitis C: considera- Y.S., Weldon, S.M., Yong, C.L., Llinàs-Brunet, M., 2003. An NS3 protease inhibitor
tions for healthcare providers in the United States. Clin. Infect. Dis. 55, S10–S15. with antiviral effects in humans infected with hepatitis C virus. Nature 426, 186–189.
Bankwitz, D., Doepke, M., Hueging, K., Weller, R., Bruening, J., Behrendt, P., Lee, J.Y., Liang, T.J., 2013. Current progress in development of hepatitis C virus vaccines. Nat.
Vondran, F.W.R., Manns, M.P., Bartenschlager, R., Pietschmann, T., 2017. Med. 19, 869–878.
Maturation of secreted HCV particles by incorporation of secreted ApoE protects from Notka, F., Meier, G., Wagner, R., 2004. Concerted inhibitory activities of Phyllanthus
antibodies by enhancing infectivity. J. Hepatol. 30251–30259 S0168–8278. amarus on HIV replication in vitro and ex vivo. Antivir. Res. 64, 93–102.
Berdichevsky, Y., Zemel, R., Bachmatov, L., Abramovich, A., Koren, R., Sathiyamoorthy, Organization for Economic Cooperation and Development, 2001. OECD Guidelines for the
P., Golan-Goldhirsh, A., Tur-Kaspa, R., Benhar, I., 2003. A novel high throughput Testing of Chemicals. OECD 423. Acute Oral Toxicity-acute Toxic Class Method.
screening assay for HCV NS3 serine protease inhibitors. J. Virol. Methods 107, Paris. .
245–255. Organization for Economic Cooperation and Development, 2006. OECD Report of the
Bility, M.T., Cheng, L., Zhang, Z., Luan, Y., Li, F., Chi, L., Zhang, L., Tu, Z., Gao, Y., Fu, Y., Validation of the Updated Test Guideline 407: Repeat Dose 28-day Oral Toxicity
Niu, J., Wang, F., Su, L., 2014. Hepatitis B virus infection and immunopathogenesis in Study in Laboratory Rats. Series on Testing and Assessment No 59, ENV/JM/MONO.
a humanized mouse model: induction of human-specific liver fibrosis and M2-like pp. 26.
macrophages. PLoS Pathog. 10, e1004032. Ott, M., Thyagarajan, S.P., Gupta, S., 1997. Phyllanthus amarus suppresses hepatitis B
Boudreau, H.E., Emerson, S.U., Korzeniowska, A., Jendrysik, M.A., Leto, T.L., 2009. virus by interrupting interactions between HBV enhancer I and cellular transcription
Hepatitis C virus (HCV) proteins induce NADPH oxidase 4 expression in a trans- factors. Eur. J. Clin. Invest. 27, 908–915.
forming growth factor beta-dependent manner: a new contributor to HCV-induced Pal, S., Polyak, S.J., Bano, N., Qiu, W.C., Carithers, R.L., Shuhart, M., Gretch, D.R., Das,
oxidative stress. J. Virol. 83, 12934–12946. A., 2010. Hepatitis C virus induces oxidative stress, DNA damage and modulates the
Carmona-Cuenca, I., Roncero, C., Sancho, P., Caja, L., Fausto, N., Fernandez, M., DNA repair enzyme NEIL1. J. Gastroenterol. Hepatol. 25, 627–634.
Fabregat, I., 2008. Upregulation of the NADPH oxidase NOX4 by TGF-beta in hepa- Pawlotsky, J.M., 2016. Hepatitis C virus resistance to direct-acting antiviral drugs in
tocytes is required for its pro-apoptotic activity. J. Hepatol. 49, 965–976. interferon-free regimens. Gastroenterology 151, 70–86.
Chen, M.H., Lee, M.Y., Chuang, J.J., Li, Y.Z., Ning, S.T., Chen, J.C., Liu, Y.W., 2012. Ravikumar, Y.S., Ray, U., Nandhitha, M., Perween, A., Raja Naika, H., Khanna, N., Das, S.,
Curcumin inhibits HCV replication by induction of heme oxygenase-1 and suppres- 2011. Inhibition of hepatitis C virus replication by herbal extract: phyllanhus amarus
sion of AKT. Int. J. Mol. Med. 30, 1021–1028. as potent natural source. Virus Res. 158, 89–97.
Ciofani, G., Danti, S., D'Alessandro, D., Moscato, S., Menciassi, A., 2010. Assessing cy- Reddy, B.U., Mullick, R., Kumar, A., Sudha, G., Srinivasan, N., Das, S., 2014. Small mo-
totoxicity of boron nitride nanotubes: interference with the MTT assay. Biochem. lecule inhibitors of HCV replication from pomegranate. Sci. Rep. 4, 5411.
Biophys. Res. Commun. 394, 405–411. Reed, K.E., Rice, C.M., 2000. Overview of hepatitis C virus genome structure, polyprotein
El-Serag, H.B., Kanwal, F., Richardson, P., Kramer, J., 2016. Risk of hepatocellular car- processing and protein properties. Curr. Top. Microbiol. Immunol. 242, 55–84.
cinoma after sustained virological response in veterans with hepatitis C virus infec- Schrodinger.The PyMOL molecular Graphics System Version 1.5.0.4 Schrodinger, LLC.
tion. Hepatology 64, 130–137. Seeff, L.B., Curto, T.M., Szabo, G., Everson, G.T., Bonkovsky, H.L., Dienstag, J.L.,
Frese, M., Barth, K., Kaul, A., Lohmann, V., Schwarzle, V., Bartenschlager, R., 2003. Shiffman, M.L., Lindsay, K.L., Lok, A.S., Di Bisceglie, A.M., Lee, W.M., Ghany, M.G.,
Hepatitis C virus RNA replication is resistant to tumour necrosis factor-alpha. J. Gen. HALT-C-Trial Group, 2008. Herbal product use by persons enrolled in the hepatitis C
Virol. 84, 1253–1259. antiviral long-term treatment against cirrhosis (HALT-C) Trial. Hepatology 47,
Ganta, K.K., Mandal, A., Debnath, S., Hazra, B., Chaubey, B., 2017. Anti-HCV activity 605–612.
from semi-purified methanolic root extracts of Valeriana wallichii. Phytother Res. 31, Shimoike, T., Mimori, S., Tani, H., Matsuura, Y., Miyamura, T., 1999. Interaction of he-
433–440. patitis C virus core protein with viral sense RNA and suppression of its translation. J.
Gu, Y., Xiao, L., Ming, Y., Zheng, Z., Li, W., 2016. Corilagin suppresses cholangiocarci- Virol. 73, 9718–9725.
noma progression through Notch signalling pathway in vitro and in-vivo. Int. J. Shimoike, T., Koyama, C., Murakami, K., Suzuki, R., Matsuura, Y., Miyamura, T., Suzuki,
Oncol. 48, 1868–1876. T., 2006. Downregulation of the internal ribosome entry site (IRES)-mediated
Guo, Y.J., Zhao, L., Li, X.F., Mei, Y.W., Zhang, S.L., Tao, J.Y., Zhou, Y., Dong, J.H., 2010. translation of the hepatitis C virus: critical role of binding of the stem-loop IIId do-
Effect of corilagin on anti-inflammation in HSV-1 encephalitis and HSV-1 infected main of IRES and the viral core protein. Virology 345, 434–445.
microglias. Eur. J. Pharmacol. 635, 79–86. Wagoner, J., Negash, A., Kane, O.J., Martinez, L.E., Nahmias, Y., Bourne, N., Owen, D.M.,
Haid, S., Novodomska, A., Gentzsch, J., Grethe, C., Geuenich, S., Bankwitz, D., Chhatwal, Grove, J., Brimacombe, C., McKeating, J.A., Pecheur, E.I., Graf, T.N., Oberlies, N.H.,
P., Jannack, B., Hennebelle, T., Bailleul, F., Keppler, O.T., Poenisch, M., Lohmann, V., Cao, F., Tavis, J.E., Polyak, S.J., 2010. Multiple effects of silymarin on
Bartenschlager, R., Hernandez, C., Lemasson, M., Rosenberg, A.R., Wong-Staal, F., the hepatitis C virus life cycle. Hepatology 51, 1912–1921.
Davioud-Charvet, E., Pietschmann, T., 2012. A plant-derived flavonoid inhibits entry Wang, Z., Guo, Q.Y., Zhang, X.J., Li, X., Li, W.T., Ma, X.T., Ma, L.J., 2014. Corilagin
of all HCV genotypes into human hepatocytes. Gastroenterology 143, 213–222. attenuates aerosol bleomycin-induced experimental lung injury. Int. J. Mol. Sci. 15,
Hau, D.K., Zhu, G.Y., Leung, A.K., Wong, R.S., Cheng, G.Y., Lai, P.B., Tong, S.W., Lau, 9762–9779.
F.Y., Chan, K.W., Wong, W.Y., Lam, K.H., Cheng, C.H., Cheung, F., Chui, C.H., Wakita, T., Pietschmann, T., Kato, T., Date, T., Miyamoto, M., Zhao, Z., Murthy, K.,
Gambari, R., Fong, D.W., 2010. In vivo anti-tumour activity of corilagin on Hep3B Habermann, A., Krausslich, H.G., Mizokami, M., Bartenschlager, R., Liang, T.J., 2005.
hepatocellular carcinoma. Phytomedicine 18, 11–15. Production of infectious hepatitis C virus in tissue culture from a cloned viral
Ivanov, A.V., Bartosch, B., Smirnova, O.A., Isaguliants, M.G., Kochetkov, S.N., 2013. HCV genome. Nat. Med. 11, 791–796.
and oxidative stress in the liver. Viruses 5, 439–469. Yeo, S.G., Song, J.H., Hong, E.H., Lee, B.R., Kwon, Y.S., Chang, S.Y., Kim, S.H., Lee, S.W.,
Jia, L., Jin, H., Zhou, J., Chen, L., Lu, Y., Ming, Y., Yu, Y., 2013. A potential anti-tumor Park, J.H., Ko, H.J., 2015. Antiviral effects of Phyllanthus urinaria containing cor-
herbal medicine, Corilagin, inhibits ovarian cancer cell growth through blocking the ilagin against human enterovirus 71 and Coxsackievirus A16 in vitro. Arch Pharm.
TGF-β signaling pathways. BMC Compl. Alternative Med. 13, 33. Res. (Seoul) 38, 193–202.
Kato, T., Date, T., Miyamoto, M., Sugiyama, M., Tanaka, Y., Orito, E., Ohno, T., Sugihara,

59

You might also like