viral persistence, liver disease and host response in

28
Viral persistence, liver disease and host response in Hepatitis C-like virus rat model. Authors: Sheetal Trivedi 1 , Satyapramod Murthy 1 , Himanshu Sharma 1 , Alex S. Hartlage 1,2 , Arvind Kumar 1 , Sashi Gadi 3 , Peter Simmonds 4 , Lokendra V. Chauhan 5 , Troels K. H. Scheel 6,7 , Eva Billerbeck 7 , Peter D. Burbelo 8 , Charles M. Rice 7 , W. Ian Lipkin 5 , Kurt Vandergrift 9 , John M. Cullen 3 and Amit Kapoor 1,10 * Affiliations: 1 Center for Vaccines and Immunity, The Research Institute at Nationwide Children’s Hospital, Columbus, OH 43205, USA. 2 Medical Scientist Training Program, College of Medicine and Public Health, Ohio State University, Columbus, OH 43210 3 College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA. 4 Nuffield Department of Medicine, University of Oxford, Oxford, OX1 3SY, UK. 5 Center for Infection and Immunity, Columbia University, New York, NY 10032, USA. 6 Copenhagen Hepatitis C Program, Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark 7 Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York 10065 8 Dental Clinical Research Core, NIDCR, NIH, Bethesda, MD 9 Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, PA 16802 10 Department of Pediatrics, College of Medicine and Public Health, Ohio State University, Columbus, OH 43210 *Corresponding author. E-mail: [email protected] Amit Kapoor, Ph.D. Associate Professor Department of Pediatrics, College of Medicine The Ohio State University Center for Vaccines and Immunity The Research Institute at Nationwide Children's Hospital 700 Children's Drive Columbus, Ohio 43205 Short title: Rat model to study hepacivirus persistence and liver disease. This article has been accepted for publication and undergone full peer review but has not been through the copyediting, typesetting, pagination and proofreading process which may lead to differences between this version and the Version of Record. Please cite this article as doi: 10.1002/hep.29494 This article is protected by copyright. All rights reserved.

Upload: others

Post on 11-Nov-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Viral persistence, liver disease and host response in

Viral persistence, liver disease and host response in

Hepatitis C-like virus rat model.

Authors: Sheetal Trivedi1, Satyapramod Murthy

1, Himanshu Sharma

1, Alex S. Hartlage

1,2, Arvind

Kumar1, Sashi Gadi

3, Peter Simmonds

4, Lokendra V. Chauhan

5, Troels K. H. Scheel

6,7, Eva

Billerbeck7, Peter D. Burbelo

8, Charles M. Rice

7, W. Ian Lipkin

5, Kurt Vandergrift

9, John M. Cullen

3

and Amit Kapoor1,10

*

Affiliations: 1Center for Vaccines and Immunity, The Research Institute at Nationwide Children’s Hospital, Columbus, OH

43205, USA. 2Medical Scientist Training Program, College of Medicine and Public Health, Ohio State University,

Columbus, OH 43210 3College of Veterinary Medicine, North Carolina State University, Raleigh, NC 27607, USA.

4 Nuffield Department of Medicine, University of Oxford, Oxford, OX1 3SY, UK.

5Center for Infection and Immunity, Columbia University, New York, NY 10032, USA.

6Copenhagen Hepatitis C Program, Department of Infectious Diseases, Copenhagen University Hospital,

Hvidovre, and Department of Immunology and Microbiology, Faculty of Health and Medical Sciences,

University of Copenhagen, Copenhagen, Denmark 7Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, New York 10065

8Dental Clinical Research Core, NIDCR, NIH, Bethesda, MD

9Center for Infectious Disease Dynamics, The Pennsylvania State University, University Park, PA 16802

10Department of Pediatrics, College of Medicine and Public Health, Ohio State University, Columbus, OH

43210

*Corresponding author. E-mail: [email protected]

Amit Kapoor, Ph.D. Associate Professor

Department of Pediatrics, College of Medicine

The Ohio State University

Center for Vaccines and Immunity

The Research Institute at Nationwide Children's Hospital

700 Children's Drive

Columbus, Ohio 43205

Short title: Rat model to study hepacivirus persistence and liver disease.

This article has been accepted for publication and undergone full peer review but has not beenthrough the copyediting, typesetting, pagination and proofreading process which may lead todifferences between this version and the Version of Record. Please cite this article asdoi: 10.1002/hep.29494

This article is protected by copyright. All rights reserved.

Page 2: Viral persistence, liver disease and host response in

Page | 2

ABSTRACT

The lack of a relevant, tractable, and immunocompetent animal model for hepatitis C virus (HCV)

has severely impeded investigations of viral persistence, immunity and pathogenesis. In the absence

of immunocompetent models with robust HCV infection, homolog hepaciviruses in their natural host

could potentially provide useful surrogate models. We isolated a rodent hepacivirus (RHV) from wild

rats (Rattus norvegicus), RHV-rn1, acquired the complete viral genome sequence and developed an

infectious reverse genetics system. RHV-rn1 resembles HCV in genomic features including the

pattern of polyprotein cleavage sites and secondary structures in the viral 5’ and 3’ UTRs. We used

site-directed and random mutagenesis to determine that only the first of the two miR-122 seed sites in

viral 5’UTR is required for viral replication and persistence in rats. Next, we used the clone derived

virus progeny to infect several inbred and outbred rat strains. Our results determined that RHV-rn1

possesses several HCV-defining hallmarks: hepatotropism, propensity to persist, and the ability of

induce gradual liver damage. Histological examination of liver samples revealed the presence of

lymphoid aggregates, parenchymal inflammation and macro/micro vesicular steatosis in chronically

infected rats. Gene expression analysis demonstrated that the intrahepatic response during RHV-rn1

infection in rats mirrors that of HCV infection, including persistent activation of interferon signaling

pathways. Finally, we determined that the backbone drug of HCV direct acting antiviral (DAA)

therapy, Sofosbuvir, effectively suppresses chronic RHV-rn1 infection in rats. Taken together, we

developed RHV-rn1 infected rats as a fully immunocompetent and informative surrogate model to

delineate the mechanisms of HCV-related viral persistence, immunity and pathogenesis.

Page 2 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 3: Viral persistence, liver disease and host response in

Page | 3

INTRODUCTION

Hepatitis C virus (HCV) is a hepatotropic RNA virus classified within the genus Hepacivirus

of the family Flaviviridae and a leading cause of chronic liver disease globally. Following acute

infection, HCV establishes lifelong persistence in 60-80% of individuals, significantly elevating the

risk for developing liver cirrhosis and hepatocellular carcinoma(1). The host and viral determinants

governing this acute to chronic transition of HCV infection are mostly unknown, in large part due to

the absence of immunocompetent animal models for its in vivo characterization(2). Beside humans,

HCV only infects chimpanzees, and therefore, unlike the in-depth in vitro characterization, most of

our knowledge of HCV pathogenesis and immunity is gained through studies of infected humans or

experimentally infected chimpanzees, limited by either the lack of appropriate human samples or an

insufficient number of animals to enable an unambiguous analysis. Furthermore, the use of

chimpanzees in the elucidation of host response patterns against HCV and for the testing of

preclinical therapeutic and vaccine modalities has been stopped or restricted in most countries

because of ethical considerations. Thus, a relevant, tractable, and immunocompetent animal model is

urgently needed if the mechanisms behind HCV unique pathogenesis in humans are to be deciphered

(3, 4).

GBV-B virus, a hepacivirus of mysterious origin, is a well-recognized HCV surrogate but

unlike HCV, it usually causes only acute resolving infections in tamarins (3, 5). Nonetheless, GBV-B

infection in tamarins helped in elucidating the functional importance of several viral genome features

including the relevance of miR-122 binding sites in the viral 5’ UTR and the importance of different

sequence elements of the 3’ UTR (6, 7). Over the years several genetically engineered mouse models

of HCV have been described, but in addition of being technically cumbersome, these platforms suffer

from an underlying requirement to blunt innate and/or adaptive immune responses to support even

mediocre levels of viral replication, precluding detailed analysis of HCV associated immunity(8, 9).

Recently, we and others have identified multiple hepacivirus species in wild rodents (10).

These rodent hepaciviruses (RHV) share common genetic and evolutionary features with HCV and

therefore, with their natural hosts, could provide relevant surrogate models for the study of HCV

(11). The animal host of an ideal surrogate, however, should be an accessible and well-characterized

laboratory model with numerous molecular and genetic tools available for examining the mechanisms

Page 3 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 4: Viral persistence, liver disease and host response in

Page | 4

of viral pathogenesis. For this reason, we chose to establish a model system using a variant of a

hepacivirus species identified during a metagenomics survey of pathogens in Norway rats (Rattus

norvegicus) (12). We determined that this rat hepacivirus isolate, RHV-rn1 fails to establish

persistent infection in immunocompetent laboratory mice (13). Here, we describe that RHV-rn1

infection in laboratory rats mirror HCV infection in humans and therefore provide an informative,

tractable and fully immunocompetent surrogate model for studies of HCV persistence, immunity and

liver pathogenesis.

RESULTS

Isolation, genome sequencing and reverse genetic system for RHV-rn1

Since only a partial genome of Rat hepacivirus was previously sequenced (12), we first

infected four Sprague Dawley (SD) rats with a serum sample from an RHV-rn1 infected wild rat. All

infected rats developed high-titer viremia at one week post-infection (p.i.) and remained viremic for

the entire duration of the study (80 days p.i.) (Fig.1A). A terminal serum sample of a chronically

infected female was then used to acquire the complete viral genome using next generation sequencing

(Illumina HiSeq). After de novo assembly of the viral genome, the polyprotein coding sequence was

re-confirmed by Sanger dideoxy sequencing. Thereafter, both genomic termini were acquired using a

rapid amplification of cDNA ends and a phosphorylated oligonucleotide adapter ligation approach for

5’ and 3’ ends, respectively (14, 15). The resulting viral genome was henceforth termed RHV-rn1 as

it shared 8450/8975 (94%) nucleotide and 2936/2991 (98%) amino acid identities with the published

partial genome sequence of the Norway rat hepacivirus (GenBank accession no. KJ950938).

The complete genome of RHV-rn1 contains 9656 nucleotides and encodes a single 2991

amino acid long polyprotein (GenBank accession no. KX905133, Fig.1B). Cleavage sites

documented for HCV are conserved in RHV-rn1 and allowed a robust prediction of the sizes of the

replication-associated proteins; a separate analysis using signalaseP (16) predicted cleavage sites that

would produce core, E1, E2 and p7 proteins comparable in size to those of HCV and other

hepaciviruses (Fig.1B). The 5’ UTR is 485 bases long and terminates with a methionine codon in the

optimal Kozak context. The 3’ end of the 5’ UTR is predicted to form a type IV internal ribosomal

entry site despite a high degree of sequence divergence with HCV and other hepaciviruses in this

Page 4 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 5: Viral persistence, liver disease and host response in

Page | 5

region (Fig.1C). Furthermore, the 5’UTR of RHV-rn1 contains two miR-122 seed sites (CACUCC),

which are important for HCV replication and hepatotropism (17, 18). The 3’UTR is 297 bases long,

contains a short poly-C region followed by three stem loops (SLI to SLIII) and a U as terminal base,

similar to the HCV 3’ X region (Fig.S1) (19).

Next, we constructed a complete genomic clone of RHV-rn1 using the consensus sequence

described above (Fig.2A). To test the infectivity of clone-derived transcripts, we injected 5µg of

RHV-rn1 RNA directly into the livers of immunocompetent Brown Norway (BN) rats. All inoculated

rats developed high-titer viremia at three days p.i. and remained viremic throughout the study period

(> 17 wk p.i.) (Fig. 2B). Injected rats developed anti-NS3 IgG within 3-4 weeks p.i. No significant

fluctuations in serum ALT levels were observed. In situ hybridization using a RHV-rn1 helicase

specific probe showed presence of viral genomes in the cytoplasm of hepatocytes (Fig. 2C).

Comparison of viral titers in different organs and a negative strand RNA assay further confirmed the

hepatotropic nature of the virus (Fig. 2D and E). Sequencing of serum virus from infected rats

showed 100% nucleotide identity to the plasmid clone. A serum sample from one rat with high-titer

viremia was then used to infect eight naïve rats, all of which developed persistent infection (data not

shown). Together, these results confirmed that the RHV-rn1 clone contains all necessary genomic

elements to produce infectious virus.

Importance of the two miR-122 seed sites in the viral 5’UTR

The RHV-rn1 reverse genetics system provides an unlimited resource to generate natural and

mutant viruses for the functional and biological characterization of different genomic features. As a

proof of principle, we next focused our studies on the functional role of the two miR-122 binding

sites within the viral 5’UTR. First, we created two pools of RHV-rn1 mutant plasmids, RM-1 and

RM-2, using random mutagenesis of either of the two miR-122 seed sites (Fig.3A). RNA transcripts

of these pools were sequenced to confirm an approximately equimolar presence of all four

ribonucleotides at the six base positions theoretically generating 4,095 unique mutants in each pool.

Then, 5 µg of RM-1 and RM-2 transcripts were separately injected into the liver of individual BN

rats. Both rats developed a high-titer viremia three days p.i. and remained persistently infected for the

duration of the study period (82 days p.i.). All serial serum samples of the RM-1 injected rat,

including the earliest sample (3 days p.i.), showed RHV-rn1 variants with the natural miR-122 seed

Page 5 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 6: Viral persistence, liver disease and host response in

Page | 6

site sequence (CACUCC), indicating the strong selection for this seed site (Fig.3B). However, all the

serial serum samples of RM-2 injected rat showed selection of an RHV-rn1 variant containing a

unique sequence (ACAGUG-U) in place of the canonical miR-122 seed site sequence (Fig.3B). Even

the earliest appearing serum virus, 3 days p.i., had this unique sequence, indicating rapid selection of

this novel variant. Interestingly, this new sequence remained unchanged till the end of the study (90

days p.i.), indicating the dispensability of the second miR-122 seed site for viral replication and

persistence. To further confirm, we infected two more rats with RM-2. Both these rats developed

viremia, but, interestingly, the sequencing of serum virus showed a swarm of RM-2 variants from 3-

13 days p.i. in one rat and up to 23 days p.i. in the other, after that only the wild type RHV-rn1

(CACUCC) was present in both rats. Although these results indicated the dispensability of the second

miR-122 seed site for RHV-rn1 infection, this seed site apparently also conferred a replication

advantage to the virus.

Next, we used site-directed mutagenesis to confirm the requirement of the first miR-122 seed

site sequence (Fig.3A). Rats infected with SM-3 (auxiliary pairing mutant) and SM-12 (seed site

mutant) viruses showed an early appearance of viremia that persisted for the duration of the study

period (90 days p.i., Fig.3C). While the SM-3 virus did not revert back to the wild type sequence, the

SM-12 virus reverted back in all three infected animals after 10 days. The SM-3 viruses maintained

an intact binding site 1 for miR-122. To confirm that the SM-12 reversion is not due to the disruption

of a small predicted stem loop structure, we created a double mutant virus where position 21 was

changed to pair with the mutated SM-12 site. Interestingly, this double mutant failed to induce

infection or seroconversion in the two injected rats. Taken together, these results indicate that only

the first canonical miR-122 seed site is crucial for the virus in vivo; however, the cytidine residue at

the third position in the RHV-rn1 genome, predicted to bind the 3’ region of rat miR-122, has no

effect on viral replication and persistence. Interestingly, mutations at the SM-3 site in HCV can

confer resistance against the miR-122 antagomir in humans (20).

Infection outcomes and liver diseases in laboratory rats.

While BN rats provide an exceptional in vivo platform for evaluating the role of viral genomic

features, we failed to observe even partial control of RHV-rn1 infection in this inbred strain. To

evaluate whether host heterogeneity influences RHV-rn1 infection outcome, we infected four

Page 6 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 7: Viral persistence, liver disease and host response in

Page | 7

different outbred lines of rats: Sprague-Dawley (SD), Holtzman (HTZ), Long Evans (LE), and Wistar

Han (WH). While LE, WH, and SD rats showed limited suppression of viral replication, HTZ rats

exhibited clear differences in viral titers between individuals within the first 80 days of infection

(Fig. 4). Since HTZ rats displayed the largest suppression of viremia, four rats of this strain were

kept for a longer period to study the viral persistence and development of liver diseases. All six

infected rats also showed elevation of ALT that fluctuated overtime similar to HCV infection in

humans (Fig.5A) (21). One of the four rats started suppressing the viremia around day 150 and

achieved complete virus clearance by day 275. This rat no. 52 remained clear of virus for the duration

of the study (325 days), indicating complete and spontaneous resolution of viral infection (Fig.5A).

To study the histopathological changes in the livers of infected rats, two serial liver biopsies

were obtained from four infected rats (no. 49, 50, 52 and 53) at days 85 and 235. A third liver biopsy

was collected from one rat (no. 49) on day 287 to confirm continued progression of liver diseases. At

day 85 p.i., all four infected rats exhibited moderate hepatic inflammation characterized by a

consistent pattern of dense lymphocytic aggregates focused on the portal tracts. At this stage there

was focal evidence of extension of the inflammation beyond the margins of the limiting plate,

indicative of mild piecemeal necrosis (Fig.5B). At day 235 p.i. the histologic features of the livers

had changed with evident, but diminished portal tract lymphoid infiltrates. At this point there was

more parenchymal damage with hepatic plate disarray and isolated lymphocytic foci within the

hepatic parenchyma and often associated with apoptotic hepatocytes. Another feature evident in the

later biopsies and characteristic of human HCV infection was the presence of both macrovesicular

and micro-vesicular steatosis in hepatocytes. Bile ducts were indistinct in scattered portal tracts,

suggesting injury, but clearly evident injury to the ducts was rare. Next, we used in situ hybridization

to co-localize virus and liver pathological changes. Interestingly, we found steatosis in the

hepatocytes infected with the virus suggesting that the virus replication can directly triggers steatosis

in the hepatocytes (Fig.5C).

Intrahepatic responses during RHV-rn1 infection

HCV infection is known to induce potent liver responses during acute as well as chronic

infection (22-25). We used RNA-seq analysis of liver samples collected on days 20, 85 and 235 p.i,

from HTZ rats of two different class-I haplotypes, RT-1l and RT-1

u. Comparison of differentially

expressed genes (DEG) between infected and uninfected rats indicated an increased number of DEG

Page 7 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 8: Viral persistence, liver disease and host response in

Page | 8

in the later phase of infection (Fig.6A and B). Uninfected rats with a different MHC class I molecule

shared 200-285 of the DEG; most of these were involved in metabolism and physiological processes.

Interestingly, we identified 416 and 457 DEG unique to infected rats with RT-1l and RT-1

u genotypes,

respectively. Analysis of the predicted biological functions of these DEG revealed their primary role

in cellular and host responses against viral infection, including higher expression of several DEG

involved in the interferon signaling pathway (Fig.6C). We further identified 123 DEG that were

exclusive to the infected rats of both genotypes. These DEG include genes primarily involved in

leukocyte and interleukin signaling. Upstream regulators included IFN-γ, TNF, CSF2, IL6, MYD88,

STAT1 and IL2. Noticeably, while MHC class II molecules were consistently overexpressed after

RHV-rn1 infection, the class I molecules were significantly repressed, albeit with an inconsistent

pattern (Fig.6C).

Notably, the magnitude of RHV-rn1 induced DEG of IFN pathways increased during the late

phase of infection indicating the persistent nature of the antiviral host responses (Fig.7 A and B).

DEG shared among class I, II and III IFNs were most significantly overexpressed including OASL,

OAS3, CXCL10, TNFRSF14, STAT1, STAT4, IRF7 and IFITM3. Increased IL-8 signaling was

noticed in the liver at day 85 and 235 p.i. indicating an increase in angiogenesis and inflammation.

There was a significant increase in the expression of genes that are known to play a role in pattern

recognition receptor signaling like TLR7, TLR8, MDA-5, NALP3, PKC, IRF7, NFKB and

RANTES. Suppressed biological pathways included RhoGDI signaling and Liver X-receptor

activation. HCV infection in vitro leads to sequestration of cellular miR-122 (26). Our RNA-seq data

of rats with the RT-1l

haplotype indicated de-repression of a number of miR-122 regulated genes

(ACTR1A, CCNG1, CREB1, CYP2C9, GIT1, GNPDA1, 1QGAP, KHDRBS1, LAMC1, MAP4,

MTHFD2, PKM, PTPN1, RBM3, SEMA4D, SEPT2, SH3GRL3, VAV3 and VIM). However, de-

repression of DEGs was also observed for genes regulated by miR-21 and let-7 cluster in the livers of

infected rats. While there were no significant differences in the expression levels of known HCV

entry factors (SCRAB1, CD81. CLD1 and OCLN), the expression of a recently identified entry factor

Cadherin was consistently increased in the livers of all infected rats (27).

RHV-rn1 infected rats as model for HCV Direct Acting Antiviral (DAA) therapy.

Page 8 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 9: Viral persistence, liver disease and host response in

Page | 9

To assess the feasibility of our RHV infected rat model to study changes in the viral

population and virus-specific T cell immunity after DAA cure, we treated three rats with established

chronic RHV-rn1 infection with Sofosbuvir and ribavirin (Fig. 8). Daily doses of Sofosbuvir (40

mg/kg/day) and ribavirin (70 mg/kg/day in two doses each day) were comparable to the

recommended human doses adjusted to rat pharmacokinetics. As controls, three chronically infected

rats were treated with only ribavirin and one rat was put on a low dose Sofosbuvir (5 mg/kg/day) and

ribavirin. Our results unambiguously showed that the high-dose sofosbuvir and ribavirin combination

rapidly suppressed RHV-rn1 replication to below the detection threshold in all three treated rats. Low

dose sofosbuvir with ribavirin and ribavirin alone, failed to suppress the viremia in treated rats.

Interestingly, the viremia rapidly rebounded in all three rats a week after the drugs were discontinued

(Fig. 8), suggesting that longer period of high-dose sofosbuvir and ribavirin treatment is required to

achieve a sustained viral response in the rats.

DISCUSSIONS

We here isolated RHV-rn1 from samples of a wild rat, developed an infectious clone and

produced homogenous virus stocks to infect several strains of laboratory rats, expecting to develop an

informative model to study HCV-like viral persistence and liver diseases. Our results show that

infected rats spontaneously develop chronic hepacivirus infections making this model of hepacivirus

infection in its natural host of great relevance for understanding human HCV infection and immunity.

We further determined that RHV-rn1 is hepatotropic and the chronic viral infection leads to slow

progressive liver diseases (Fig.2).

Development of an infectious clone for RHV-rn1 will allow studies of virus mutagenesis and

facilitate the development of in vitro systems. Similar developments for HCV took several years,

mainly due to failure to identify the 3’ end of the viral genome and lack of a tractable model (28). We

recently established a similar system for the equine non-primate hepacivirus in horses (15). We used

our reverse genetic system to study the biological importance of miR-122 sites in the 5’UTR of

RHV-rn1. Our results indicate that only the first of the two miR-122 sites is indispensable for RHV-

rn1 replication and persistence. We also used a novel random mutagenesis approach that allowed

identification of a RHV-rn1 variant with a novel sequence in place of the second miR-122 seed site.

Page 9 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 10: Viral persistence, liver disease and host response in

Page | 10

Further studies using these different viruses, with one or both the wild type miR-122 sites may shed

new light on their role in virus infection outcomes and pathogenesis. Recently, miR-122 “sponging”

by HCV genomes has been suggested to play a role in regulating the gene expression of miR-122

target genes in hepatocytes, including oncogenes, thereby providing an environment fertile for end

stage liver diseases such as hepatocellular carcinoma(26). Here, we showed de-repression of selected

miR-122 targets in infected rats. RHV-rn1 miR-122 mutants will be useful to study the de-repression

of miR-122 regulated genes in vivo, and any effect this may have on virus induced pathogenesis.

Host factors play an important role in determining the infection and the pathogenic outcomes

of HCV infections in humans (2, 29). While 20-30% of infected individuals clear the virus within the

first year of infection, the remaining 70-80% of individuals develop a life-long chronic virus infection

(1, 30-32). The acute phase of HCV infection in humans is generally asymptomatic with the

exception of mild elevations in serum ALT levels indicating liver injury (33). After several years of

chronic infection, some of these patients develop liver diseases including chronic hepatitis, steatosis,

fibrosis, cirrhosis and hepatocellular carcinoma (34). The variables that can dictate or contribute to

these differences remain unknown, largely due to lack of a fully immunocompetent tractable model.

Here, we used homogenous virus stock derived from our reverse genetics system, yet the infected

HTZ rats differed in the course of viremia, infection outcomes and liver pathology (Fig.5),

resembling HCV infection in humans. Thus, we believe that RHV-rn1 infection in outbred or

genetically heterogeneous rats provides a unique model to identify host determinants of hepacivirus

infection outcomes and pathogenesis.

HCV liver disease is characterized by the histologic triad of lymphoid aggregates in portal

tracts, epithelial damage of small bile ducts and micro- and macro- vesicular steatosis of hepatocytes

(35). We determined that RHV-rn1 infected rats develop 2 of these 3 features with a suggestion of

biliary epithelial injury as well within 32 weeks post infection. The presence of macro- and micro-

vesicular steatosis in livers of RHV-rn1 infected rats is a finding of major significance. We used in

situ hybridization to determine that several virus infected hepatocytes had steatosis, suggesting direct

induction by virus infection (Fig.5C). Uninfected rats and one of the infected rats did not develop

steatosis. More interestingly, in rat no. 52, virus clearance lead to reversion of inflammation,

confirming the direct link with the virus. Many human patients with chronic HCV also develop a

Page 10 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 11: Viral persistence, liver disease and host response in

Page | 11

degree of hepatic steatosis. Moreover, several clinical studies have now confirmed that hepatic

steatosis is an independent risk factor of HCC in patients with chronic HCV (36, 37). Development of

parenchymal inflammation, hepatitis, necrosis and steatosis within eight months of RHV-rn1

infection indicates that some chronically infected rats could consequently progress to cirrhosis and

HCC.

In vitro studies of HCV suggest that the viral NS3/4A protease can block IRF3-mediated

transcriptional activation of IFNβ by cleavage and inactivation of the mitochondrial antiviral

signaling (MAVS) and TIR-domain-containing adapter (TRIF) proteins (38, 39). However, strong

innate immune responses are observed in humans and chimpanzees infected with HCV (22, 25, 40,

41). Notably, HCV is known to induce stronger innate immune responses than the other MAVS

cleaving hepatotropic RNA virus, hepatitis A virus (22-25). Moreover, chronic liver inflammation

during HCV infection contributes towards a procarcinogenic microenvironment and paves the way

for development of fibrosis/cirrhosis and HCC (42). Our liver transcriptomics of RHV-rn1 infected

rats during acute and chronic phase shows significant over-expression of genes associated with

inflammatory responses. Interestingly, the magnitude of interferon stimulated gene (ISG) expression

increased during the chronic phase compared to the early infection phase indicating persistent

activation of interferon and innate immune responses during RHV-rn1 infection. Additionally,

several signaling pathways of the adaptive immunity were also persistently activated. Differentiation

of naïve T-cells requires antigenic stimulation mediated by the T-cell receptor and co-receptor CD4

with the antigen-MHC-II complex. IL-2 and IFN-γ are the two most important cytokines for Th1

differentiation (43). Our results indicate persistent activation of the Th1 pathway during RHV-rn1

infection in rats despite their failure to clear the viral persistence. Virus specific functional T cell

responses against Core, NS3 and NS4 proteins were also detected using IFN-γ-ELISpot assays in

RHV-nr1 infected HTZ rats (data not shown). MHC class-II molecules were found significantly

overexpressed. Interestingly, our data showed repression of class I molecules, the expression of

which is necessary for effective targeting of infected hepatocytes by cytotoxic CD8+ T cells. Overall,

these results confirm that RHV-rn1 infected rats offer a suitable in vivo platform to elucidate the role

of various ISGs and immune responses in hepacivirus pathogenesis. Notably, RNA-seq analysis of

outbred animals is challenging due to inherent differences in host genetics and therefore our data

needs to be confirmed using a lager cohort of infected rats and MHC matched controls.

Page 11 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 12: Viral persistence, liver disease and host response in

Page | 12

An interferon-free regimen of DAA can achieve sustained virologic response (SVR) in most

HCV infected subjects(44). Considering that persistent antigenic stimulation drives T cell exhaustion,

DAA cure has the potential to reverse defects and restore protective antiviral functions (43, 45).

However, recovery of T cell immunity after antiviral therapy and protection from reinfection are the

critical unknowns for translational research in the post-DAA era. The rat model is suitable to

characterize the function and fate of T cells during hepacivirus infection and after DAA mediated

cure. Most importantly, availability of RHV-rn1 and related viruses will allow us to precisely

determine if the cured rats develop protection against homologous or heterologous challenge virus

infection.

Considering the strict species tropism of HCV, identification of animal hepaciviruses

promised the development of a fully immunocompetent and informative surrogate model useful to

delineate the mechanisms of viral persistence and immunity (10). The non-primate hepaciviruses

(NPHV) or equine hepaciviruses are interesting given that they remain the closest genetic relatives of

HCV, and share many features of its natural history (46). However, they mostly cause acute resolving

infection with no clinical disease in horses (15, 47) and studies are limited by the number of animals,

expenses and lack of species specific research reagents. We therefore focused our efforts on

developing a tractable lab rodent surrogate model for HCV, and selected the rat hepacivirus RHV-

rn1. We recently determined that RHV-rn1 infection is rapidly cleared by immunocompetent mice

(13) and that the duration of viremia was shorter than that observed for NPHV and GBV-B infections

in horses and marmosets, receptively (5, 48). However, the availability of numerous genetic variants

and immunological reagents for lab mice makes this model extremely useful for characterizing the

nature of host responses associated with acute virus clearance and protective immunity (13).

Additionally, it is plausible that the rat virus can be further adapted to establish a fully

immunocompetent mouse model for persistent hepacivirus infections. Importantly, RHV-rn1

infection in inbred and outbred rats closely resembles HCV infection outcomes since most infected

animals develop life-long persistent hepatotropic infection associated with chronic inflammation and

liver diseases. As with human HCV infection, full susceptibility to viral persistence probably reflects

hundreds of years of co-evolution between RHV-rn1 and its natural rat host. Thus, rats being an

immunocompetent and fully susceptible host of RHV-rn1 could allow optimization of effective

vaccine designs to prevent hepaciviral persistence in the natural context. Overall, we conclude that

Page 12 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 13: Viral persistence, liver disease and host response in

Page | 13

laboratory rats infected with RHV-rn1 can serve as an informative, fully immunocompetent surrogate

to study the mechanisms of HCV persistence, immunity, and pathogenesis.

Materials and Methods

Animals. Brown Norway, Sprague Dawley, Wistar Han, Holtzman, and Long Evans rats were

obtained from commercial suppliers (Charles River or Envigo). All animal studies were performed at

North Carolina State University and The Research Institute at Nationwide Children’s Hospital using

protocols approved by respective Institutional Animal Care and Use Committees.

Viral genome, infectious clone, mutagenesis and RNA transcription. The complete genome of

RHV-rn1 was sequenced as reported by us previously for other rodent hepaciviruses(14, 15). The

RHV-rn1 molecular clone was synthesized and modified to introduce desired restriction sites using

standard molecular techniques (Fig.2). Site-directed and random mutagenesis was performed using

commercial PCR mutagenesis kits (New England Biolabs). After linearization of the plasmid, viral

transcripts were synthesized using Hi-Script Transcription kit (Promega). After a DNAase-I digestion

step, purified RNA was used for intrahepatic injection into rats. Complete genome of RHV-rn1 is

deposited in GenBank (accession no. KX905133).

RHV-rn1 infection, viral titers, and antibody detection. Rats were infected intravenously with

105-10

7 VGE of RHV-rn1 by tail vein injection. For experiments involving the RHV-rn1 molecular

clone, animals were injected intrahepatically with 5 µg of purified RNA.

Viral RNA from animal sera was extracted using the QIAamp Viral RNA Mini Kit (Qiagen). After

reverse transcription, quantitative PCR was done using AmpliTaq Gold 360 Master Mix (Applied

Biosystems) and following cycling conditions: 50º C for 2 min, 95º C for 2 min and 40 cycles of 95º

C for 15 sec, 53º C for 45 sec, and 60º C for 45 sec. Primers and probe sequences used for this

protocol were TACATGGCTAAGCAATACGG (sense), AAGCGCAGCACCAATTCC (antisense),

and [6-FAM]CTCACGTACATGACGTACGGCATG[BHQ1a-6FAM] (probe). To detect negative-

strand RNA replicative intermediates, we used a novel RNA polyuridylation approach. Briefly, total

RNA was polyuridylated via poly-U polymerase (NEB) followed by cDNA generation using

Superscript II Reverse Transcriptase and a unique primer containing a polyadenylated 3’ end

Page 13 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 14: Viral persistence, liver disease and host response in

Page | 14

(GAATCGAGCACCAGTTACGCATGCCGAGGTCGACTTCCTAGAAAAAAAAAAAAAAAA -

AACCA). PCR was then performed using a truncated form of the unique RT primer

(GAATCGAGCACCAGTTACG) and an RHV-rn1 specific negative-strand primer

(CCATGTAGAGGAGATAAGTCC).

To detect antibody responses in infected animals, we used a sensitive luciferase immunoprecipation

system (LIPS) assay to screen serum samples for the presence of antibodies against the RHV-rn1

NS3 helicase protein. A 240 amino acid fragment (positions 1139 to 1379) of the RHV-rn1 helicase

protein (NS3) was subcloned downstream of Renilla luciferase (Rluc) using the pREN2 vector to

express Rluc-fused viral antigen. LIPS assays were performed as described in our previous

publications (46).

Tissue histology. Liver sections (6µm) were fixed with 10% neutral buffered formaldehyde,

processed routinely and stained using hematoxylin and eosin. In situ hybridization was performed

using RNAscope and probe targeting RHV-rn1 helicase sequence.

Serum ALT quantification. Serum alanine aminotransferase (ALT) levels in rats were determined

using an ALT assay kit (BiooScientific) as per manufacturer’s instructions.

RNA-seq analysis of infected liver samples. Liver sections and biopsies were immediately

immersed in RNAlater (Ambion) and kept at -80ºC until RNA extraction by RNeasy kit (Qiagen).

DNA and rRNA from total RNA was removed using DNAse-I and Ribo-zero (Illumina) and then

samples with RNA integrity value of >7 were used for RNA-seq. We used Illumina Scriptseq

(stranded RNAseq) for library prep and paired end 150 bp SBS Chemistry to generate sequences

using the Illumina HiSeq4000 platform. Sixty million reads were generated for each liver sample.

Quality filtering of reads was done using FastQC (v0.11.3) and then sequences were mapped to

reference genome of Rattus norvegicus, NCBI Rnor_6.0 (ftp://igenome:G3nom3s4u@ussd-

ftp.illumina.com/Rattus_norvegicus/NCBI/Rnor_6.0/Rattus_norvegicus_NCBI_Rnor_6.0.tar.gz)

using TopHat2 (v2.1.1). The Cufflinks v2.2.1 package was used for transcript assembly and

calculation of differential gene expression. Cuffmerge (v1.0.0) tool was used to merge and generate a

combined transcript. Merged transcript and reference genome fasta was used by Cuffdiff (v2.2.1) tool

Page 14 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 15: Viral persistence, liver disease and host response in

Page | 15

to generate differential gene expression for any two different sample conditions. RNA-Seq analysis

was performed using a previously described protocol (49). Gene function and pathway analysis was

performed by Ingenuity Pathway Analysis software (Qiagen Inc.). Cutoff parameters used to identify

differentially expressed genes were p-value <0.05, false discovery rate q<0.05 and fold change

>0.5849, (14). We used custom in house developed R-scripts and R-packages, Limma for

vennDiagram and gplots for making heatmaps.

Acknowledgements: Research in the Kapoor lab is supported by National Institutes of Health grants

AI10763, NSF grant 1619072 Ecology and Evolution of Infectious Diseases program and The

Research Institute at Nationwide Children’s Hospital. C.M.R and E.B are supported by R01

AI131688. P.D.B.’s work in this study was supported by the Intramural Research Program of the

National Institute of Dental and Craniofacial Research.

Authors Contributions: S.T., J.C. and A.K. conceived and led the study. All authors participated in

the analysis and interpretation of data. S.T., S.M., A.S.H., P.S., T.K.H.S., J.C. and A.K. wrote the

paper.

Page 15 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 16: Viral persistence, liver disease and host response in

Page | 16

REFERENCES

1. Hoofnagle JH. Course and outcome of hepatitis C. Hepatology 2002;36:S21-29.

2. Thimme R, Bukh J, Spangenberg HC, Wieland S, Pemberton J, Steiger C, Govindarajan S, et al.

Viral and immunological determinants of hepatitis C virus clearance, persistence, and disease. Proc Natl

Acad Sci U S A 2002;99:15661-15668.

3. Bukh J. Animal models for the study of hepatitis C virus infection and related liver disease.

Gastroenterology 2012;142:1279-1287 e1273.

4. Dorner M, Rice CM, Ploss A. Study of hepatitis C virus entry in genetically humanized mice.

Methods 2013;59:249-257.

5. Manickam C, Rajakumar P, Wachtman L, Kramer JA, Martinot AJ, Varner V, Giavedoni LD, et al.

Acute Liver Damage Associated with Innate Immune Activation in a Small Nonhuman Primate Model of

Hepacivirus Infection. J Virol 2016;90:9153-9162.

6. Nam JH, Faulk K, Engle RE, Govindarajan S, St Claire M, Bukh J. In vivo analysis of the 3'

untranslated region of GB virus B after in vitro mutagenesis of an infectious cDNA clone: persistent

infection in a transfected tamarin. J Virol 2004;78:9389-9399.

7. Sagan SM, Sarnow P, Wilson JA. Modulation of GB virus B RNA abundance by microRNA-122:

dependence on and escape from microRNA-122 restriction. J Virol 2013;87:7338-7347.

8. von Schaewen M, Ploss A. Murine models of hepatitis C: what can we look forward to? Antiviral

Res 2014;104:15-22.

9. Winer BY, Ding Q, Gaska JM, Ploss A. In vivo models of hepatitis B and C virus infection. FEBS

Lett 2016;590:1987-1999.

10. Hartlage AS, Cullen JM, Kapoor A. The Strange, Expanding World of Animal Hepaciviruses. Annu

Rev Virol 2016;3:53-75.

11. Drexler JF, Corman VM, Muller MA, Lukashev AN, Gmyl A, Coutard B, Adam A, et al. Evidence for

novel hepaciviruses in rodents. PLoS Pathog 2013;9:e1003438.

12. Firth C, Bhat M, Firth MA, Williams SH, Frye MJ, Simmonds P, Conte JM, et al. Detection of

zoonotic pathogens and characterization of novel viruses carried by commensal Rattus norvegicus in

New York City. MBio 2014;5:e01933-01914.

13. Billerbeck E, Wolfisberg R, Fahnoe U, Xiao JW, Quirk C, Luna JM, Cullen JM, et al. Mouse models

of acute and chronic hepacivirus infection. Science 2017;357:204-208.

14. Kapoor A, Simmonds P, Scheel TK, Hjelle B, Cullen JM, Burbelo PD, Chauhan LV, et al.

Identification of rodent homologs of hepatitis C virus and pegiviruses. MBio 2013;4:e00216-00213.

15. Scheel TK, Kapoor A, Nishiuchi E, Brock KV, Yu Y, Andrus L, Gu M, et al. Characterization of

nonprimate hepacivirus and construction of a functional molecular clone. Proc Natl Acad Sci U S A

2015;112:2192-2197.

16. Petersen TN, Brunak S, von Heijne G, Nielsen H. SignalP 4.0: discriminating signal peptides from

transmembrane regions. Nat Methods 2011;8:785-786.

17. Jopling CL, Yi M, Lancaster AM, Lemon SM, Sarnow P. Modulation of hepatitis C virus RNA

abundance by a liver-specific MicroRNA. Science 2005;309:1577-1581.

18. Li Y, Masaki T, Yamane D, McGivern DR, Lemon SM. Competing and noncompeting activities of

miR-122 and the 5' exonuclease Xrn1 in regulation of hepatitis C virus replication. Proc Natl Acad Sci U S

A 2013;110:1881-1886.

19. Yi M, Lemon SM. Structure-function analysis of the 3' stem-loop of hepatitis C virus genomic

RNA and its role in viral RNA replication. RNA 2003;9:331-345.

Page 16 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 17: Viral persistence, liver disease and host response in

Page | 17

20. van der Ree MH, de Vree JM, Stelma F, Willemse S, van der Valk M, Rietdijk S, Molenkamp R, et

al. Safety, tolerability, and antiviral effect of RG-101 in patients with chronic hepatitis C: a phase 1B,

double-blind, randomised controlled trial. Lancet 2017;389:709-717.

21. Puoti C, Magrini A, Stati T, Rigato P, Montagnese F, Rossi P, Aldegheri L, et al. Clinical,

histological, and virological features of hepatitis C virus carriers with persistently normal or abnormal

alanine transaminase levels. Hepatology 1997;26:1393-1398.

22. Bigger CB, Guerra B, Brasky KM, Hubbard G, Beard MR, Luxon BA, Lemon SM, et al. Intrahepatic

gene expression during chronic hepatitis C virus infection in chimpanzees. J Virol 2004;78:13779-13792.

23. Lanford RE, Feng Z, Chavez D, Guerra B, Brasky KM, Zhou Y, Yamane D, et al. Acute hepatitis A

virus infection is associated with a limited type I interferon response and persistence of intrahepatic

viral RNA. Proc Natl Acad Sci U S A 2011;108:11223-11228.

24. Li K, Lemon SM. Innate immune responses in hepatitis C virus infection. Semin Immunopathol

2013;35:53-72.

25. Bigger CB, Brasky KM, Lanford RE. DNA microarray analysis of chimpanzee liver during acute

resolving hepatitis C virus infection. J Virol 2001;75:7059-7066.

26. Luna JM, Scheel TK, Danino T, Shaw KS, Mele A, Fak JJ, Nishiuchi E, et al. Hepatitis C virus RNA

functionally sequesters miR-122. Cell 2015;160:1099-1110.

27. Li Q, Sodroski C, Lowey B, Schweitzer CJ, Cha H, Zhang F, Liang TJ. Hepatitis C virus depends on

E-cadherin as an entry factor and regulates its expression in epithelial-to-mesenchymal transition. Proc

Natl Acad Sci U S A 2016;113:7620-7625.

28. Lindenbach BD, Meuleman P, Ploss A, Vanwolleghem T, Syder AJ, McKeating JA, Lanford RE, et

al. Cell culture-grown hepatitis C virus is infectious in vivo and can be recultured in vitro. Proc Natl Acad

Sci U S A 2006;103:3805-3809.

29. Su AI, Pezacki JP, Wodicka L, Brideau AD, Supekova L, Thimme R, Wieland S, et al. Genomic

analysis of the host response to hepatitis C virus infection. Proc Natl Acad Sci U S A 2002;99:15669-

15674.

30. Ge D, Fellay J, Thompson AJ, Simon JS, Shianna KV, Urban TJ, Heinzen EL, et al. Genetic variation

in IL28B predicts hepatitis C treatment-induced viral clearance. Nature 2009;461:399-401.

31. Osburn WO, Fisher BE, Dowd KA, Urban G, Liu L, Ray SC, Thomas DL, et al. Spontaneous control

of primary hepatitis C virus infection and immunity against persistent reinfection. Gastroenterology

2010;138:315-324.

32. Thomas DL, Thio CL, Martin MP, Qi Y, Ge D, O'Huigin C, Kidd J, et al. Genetic variation in IL28B

and spontaneous clearance of hepatitis C virus. Nature 2009;461:798-801.

33. Park SH, Rehermann B. Immune responses to HCV and other hepatitis viruses. Immunity

2014;40:13-24.

34. Bandiera S, Billie Bian C, Hoshida Y, Baumert TF, Zeisel MB. Chronic hepatitis C virus infection

and pathogenesis of hepatocellular carcinoma. Curr Opin Virol 2016;20:99-105.

35. Goodman ZD, Ishak KG. Histopathology of hepatitis C virus infection. Semin Liver Dis 1995;15:70-

81.

36. Ohata K, Hamasaki K, Toriyama K, Matsumoto K, Saeki A, Yanagi K, Abiru S, et al. Hepatic

steatosis is a risk factor for hepatocellular carcinoma in patients with chronic hepatitis C virus infection.

Cancer 2003;97:3036-3043.

37. Tanaka A, Uegaki S, Kurihara H, Aida K, Mikami M, Nagashima I, Shiga J, et al. Hepatic steatosis

as a possible risk factor for the development of hepatocellular carcinoma after eradication of hepatitis C

virus with antiviral therapy in patients with chronic hepatitis C. World J Gastroenterol 2007;13:5180-

5187.

Page 17 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 18: Viral persistence, liver disease and host response in

Page | 18

38. Meylan E, Curran J, Hofmann K, Moradpour D, Binder M, Bartenschlager R, Tschopp J. Cardif is

an adaptor protein in the RIG-I antiviral pathway and is targeted by hepatitis C virus. Nature

2005;437:1167-1172.

39. Li K, Foy E, Ferreon JC, Nakamura M, Ferreon AC, Ikeda M, Ray SC, et al. Immune evasion by

hepatitis C virus NS3/4A protease-mediated cleavage of the Toll-like receptor 3 adaptor protein TRIF.

Proc Natl Acad Sci U S A 2005;102:2992-2997.

40. Schoggins JW, Rice CM. Innate immune responses to hepatitis C virus. Curr Top Microbiol

Immunol 2013;369:219-242.

41. Horner SM, Gale M, Jr. Regulation of hepatic innate immunity by hepatitis C virus. Nat Med

2013;19:879-888.

42. Mitchell JK, Lemon SM, McGivern DR. How do persistent infections with hepatitis C virus cause

liver cancer? Curr Opin Virol 2015;14:101-108.

43. Wherry EJ, Kurachi M. Molecular and cellular insights into T cell exhaustion. Nat Rev Immunol

2015;15:486-499.

44. Chan J, Chung RT. Perspectives on HCV: Current Therapeutic Regimens and Drug-Drug

Interactions. Clin Pharmacol Drug Dev 2017;6:147-163.

45. Walker CM. Designing an HCV vaccine: a unique convergence of prevention and therapy? Curr

Opin Virol 2017;23:113-119.

46. Burbelo PD, Dubovi EJ, Simmonds P, Medina JL, Henriquez JA, Mishra N, Wagner J, et al.

Serology-enabled discovery of genetically diverse hepaciviruses in a new host. J Virol 2012;86:6171-

6178.

47. Pfaender S, Walter S, Grabski E, Todt D, Bruening J, Romero-Brey I, Gather T, et al. Immune

protection against reinfection with nonprimate hepacivirus. Proc Natl Acad Sci U S A 2017;114:E2430-

E2439.

48. Pfaender S, Cavalleri JM, Walter S, Doerrbecker J, Campana B, Brown RJ, Burbelo PD, et al.

Clinical course of infection and viral tissue tropism of hepatitis C virus-like non-primate hepaciviruses.

Hepatology 2014.

49. Trapnell C, Roberts A, Goff L, Pertea G, Kim D, Kelley DR, Pimentel H, et al. Differential gene and

transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat Protoc

2012;7:562-578.

Page 18 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 19: Viral persistence, liver disease and host response in

Page | 19

FIGURE LEGENDS

Fig.1. Genetic characterization of RHV-rn1. (A). Isolation and infection outcome of RHV-rn1

in Sprague Dawley rats. The serum sample pointed by red arrow was used to acquire complete

RHV-rn1 genome and for construction of the genome clone (B). Genome diagram of RHV-rn1

depicting cleavage sites predicted by alignment and homology to sites in the non-structural genes

previously characterized in other hepaciviruses (HCV, NPHV and GBV-B). Cleavage sites in

structural gene regions were independently predicted using the SignalP 4.1 server (16). Predicted

N-linked glycosylation sites (Nx[S/T]) in envelope proteins are depicted by vertical arrows. (C).

The predicted secondary structure of RHV-rn1 5’ UTR. The location and sequence of the two

miR-122 sites are shown as filled circles.

Fig.2. The reverse genetic system and tissue tropism of RHV-rn1. (A). Schematics and

restriction map of the RHV-rn1 infectious clone. (B). Kinetics of viremia in the four rats injected

with RHV-rn1 transcripts. Anti-NS3 IgG titers for two rats are shown in shaded areas (C). In situ

hybridization of uninfected (left image) and infected (right image) rat liver sections. Images were

captured at 400X resolution and antiviral probe was labeled with Fast-Red dye. (D). Mean viral

titers in different organs of four infected HTZ rats. Two different liver sections were used for

each of the four rats. (E). Results of negative strand RNA assay to detect RHV-rn1 replicative

forms in different organs of the infected rats.

Fig. 3. Mutagenesis of miR-122 bindings sites, infection outcomes and evolution of RHV-

rn1 mutants. (A). Schematics of mutagenesis targeting the two miR-122 sites and the names of

different mutants. The blue circle represent a mixture of all four nucleotides confirmed by

sequencing of pooled RNA shown below as the electropherograms of dideoxy sequencing. (B).

Kinetics of viremia and evolution of mutated sites in vivo. The RM-1 infected rat rapidly selected

wild type virus (CACUCC). One of the three rats infected with RM-2 selected a unique variant

(ACAGUGU, green color line) that established persistent infection. The other two rats, shown

with black lines, selected the wild type virus. (C). In the three SM-3 infected rats, the mutant

virus remain mutant type while in the three SM12 infected rats, the mutant virus reverted back to

wild type after 10 days p.i.. The double mutant with complementary SM12 and SM21 mutations

failed to initiate infection in the two injected rats (pink line).

Page 19 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 20: Viral persistence, liver disease and host response in

Page | 20

Fig.4. RHV-rn1 infection and outcomes in outbred rat strains. Six rats of each strain were

infected with 106 genomic copies the clone derived RHV-rn1.

Fig.5. RHV-rn1 infection and disease outcomes outbred HTZ rats. (A). Long-term follow-up

study of four HTZ rats. Viral titers and ALT are shown. All rats seroconverted 20-24 days post

infection (anti-NS3 IgG). (B) Histopathological examination of liver biopsies from these four

rats collected at day 85 and 235 p.i. Portal lymphocytic aggregates are shown by the black arrow

in top-left panel (rat no. 50), hepatocellular steatosis is shown by the back arrow in the middle-

left panel (rat no. 50). Top and middle right side panels are liver sections from rat no. 53. Bottom

panels (rat no. 50) show an apoptotic hepatocyte (left) and higher magnification of

microvesicular steatosis (right), indicated by black arrows. (C) In situ hybridization of rat no. 49

liver sections serially obtained at day 85, 235 and 287 days p.i.. A large lymphocytic aggregate

in shown by a black arrow in the middle panel. Microvesicular and macrovesicular steatotis is

shown by red and pink arrows, respectively in the bottom panel.

Fig.6. RNA-seq analysis of rat livers after RHV-rn1 infection. (A). Venn diagrams of liver

DEG (p value <0.05 and q value <0.05) between control and infected rats of different class I

haplotypes. RT-1l and RT-1

u are shown as letters L and U, respectively. Days post infection are

shown as numbers as suffix. (B). Dynamics of 8,201 DEG significantly changed at different

times p.i. in the livers of RHV-rn1 infected rats. (C). Changes in 62 DEG known to be associated

with anti-viral responses at different times p.i.

Fig.7. Dynamics of DEG associated with IFN-γ and IFN-λ1 responses during RHV-rn1

infection. Log2 fold difference in expression of DEGs compared to the uninfected control rats is

shown. All gene expression data was normalized using fragment per kilo base per million reads

(FPKM) values.

Fig.8. DAA therapy of chronically infected rats. HTZ rats infected with RHV-rn1 were treated

with different doses of sofosbuvir and ribavirin and also ribavirin alone. The treatment periods is

indicated by broken black arrows.

Page 20 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 21: Viral persistence, liver disease and host response in

Fig.1. Genetic characterization of RHV-rn1. (A). Isolation and infection outcome of RHV-rn1 in Sprague Dawley rats. The serum sample pointed by red arrow was used to acquire complete RHV-rn1 genome and

for construction of the genome clone (B). Genome diagram of RHV-rn1 depicting cleavage sites predicted by

alignment and homology to sites in the non-structural genes previously characterized in other hepaciviruses (HCV, NPHV and GBV-B). Cleavage sites in structural gene regions were independently predicted using the SignalP 4.1 server (16). Predicted N-linked glycosylation sites (Nx[S/T]) in envelope proteins are depicted by vertical arrows. (C). The predicted secondary structure of RHV-rn1 5’ UTR. The location and sequence of

the two miR-122 sites are shown as filled circles.

254x190mm (300 x 300 DPI)

Page 21 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 22: Viral persistence, liver disease and host response in

Fig.2. The reverse genetic system and tissue tropism of RHV-rn1. (A). Schematics and restriction map of the RHV-rn1 infectious clone. (B). Kinetics of viremia in the four rats injected with RHV-rn1 transcripts. Anti-NS3 IgG titers for two rats are shown in shaded areas (C). In situ hybridization of uninfected (left image) and

infected (right image) rat liver sections. Images were captured at 400X resolution and antiviral probe was labeled with Fast-Red dye. (D). Mean viral titers in different organs of four infected HTZ rats. Two different liver sections were used for each of the four rats. (E). Results of negative strand RNA assay to detect RHV-

rn1 replicative forms in different organs of the infected rats.

254x190mm (300 x 300 DPI)

Page 22 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 23: Viral persistence, liver disease and host response in

Fig. 3. Mutagenesis of miR-122 bindings sites, infection outcomes and evolution of RHV-rn1 mutants. (A). Schematics of mutagenesis targeting the two miR-122 sites and the names of different mutants. The blue

circle represent a mixture of all four nucleotides confirmed by sequencing of pooled RNA shown below as the

electropherograms of dideoxy sequencing. (B). Kinetics of viremia and evolution of mutated sites in vivo. The RM-1 infected rat rapidly selected wild type virus (CACUCC). One of the three rats infected with RM-2 selected a unique variant (ACAGUGU, green color line) that established persistent infection. The other two rats, shown with black lines, selected the wild type virus. (C). In the three SM-3 infected rats, the mutant

virus remain mutant type while in the three SM12 infected rats, the mutant virus reverted back to wild type after 10 days p.i.. The double mutant with complementary SM12 and SM21 mutations failed to initiate

infection in the two injected rats (pink line).

254x190mm (300 x 300 DPI)

Page 23 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 24: Viral persistence, liver disease and host response in

Fig.4. RHV-rn1 infection and outcomes in outbred rat strains. Six rats of each strain were infected with 106 genomic copies the clone derived RHV-rn1.

254x190mm (300 x 300 DPI)

Page 24 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 25: Viral persistence, liver disease and host response in

Fig.5. RHV-rn1 infection and disease outcomes outbred HTZ rats. (A). Long-term follow-up study of four HTZ rats. Viral titers and ALT are shown. All rats seroconverted 20-24 days post infection (anti-NS3 IgG). (B) Histopathological examination of liver biopsies from these four rats collected at day 85 and 235 p.i. Portal

lymphocytic aggregates are shown by the black arrow in top-left panel (rat no. 50), hepatocellular steatosis is shown by the back arrow in the middle-left panel (rat no. 50). Top and middle right side panels are liver

sections from rat no. 53. Bottom panels (rat no. 50) show an apoptotic hepatocyte (left) and higher magnification of microvesicular steatosis (right), indicated by black arrows. (C) In situ hybridization of rat no. 49 liver sections serially obtained at day 85, 235 and 287 days p.i.. A large lymphocytic aggregate in

shown by a black arrow in the middle panel. Microvesicular and macrovesicular steatotis is shown by red and pink arrows, respectively in the bottom panel.

254x190mm (300 x 300 DPI)

Page 25 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 26: Viral persistence, liver disease and host response in

Fig.6. RNA-seq analysis of rat livers after RHV-rn1 infection. (A). Venn diagrams of liver DEG (p value <0.05 and q value <0.05) between control and infected rats of different class I haplotypes. RT-1l and RT-1u are shown as letters L and U, respectively. Days post infection are shown as numbers as suffix. (B). Dynamics

of 8,201 DEG significantly changed at different times p.i. in the livers of RHV-rn1 infected rats. (C). Changes in 62 DEG known to be associated with anti-viral responses at different times p.i.

254x190mm (300 x 300 DPI)

Page 26 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 27: Viral persistence, liver disease and host response in

Fig.7. Dynamics of DEG associated with IFN-γ and IFN-λ1 responses during RHV-rn1 infection. Log2 fold difference in expression of DEGs compared to the uninfected control rats is shown. All gene expression data

was normalized using fragment per kilo base per million reads (FPKM) values.

254x190mm (300 x 300 DPI)

Page 27 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.

Page 28: Viral persistence, liver disease and host response in

Fig.8. DAA therapy of chronically infected rats. HTZ rats infected with RHV-rn1 were treated with different doses of sofosbuvir and ribavirin and also ribavirin alone. The treatment periods is indicated by broken black

arrows.

254x190mm (300 x 300 DPI)

Page 28 of 29

Hepatology

Hepatology

This article is protected by copyright. All rights reserved.