Next Article in Journal
Development of Personalized Therapeutic Strategies by Targeting Actionable Vulnerabilities in Metastatic and Chemotherapy-Resistant Breast Cancer PDXs
Next Article in Special Issue
Identification of Keratin 23 as a Hepatitis C Virus-Induced Host Factor in the Human Liver
Previous Article in Journal / Special Issue
The Role of Micronutrients in the Infection and Subsequent Response to Hepatitis C Virus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Humanized Mouse Models for the Study of Hepatitis C and Host Interactions

1
Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A*STAR), Proteos, Singapore 138673, Singapore
2
Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore
3
Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
*
Author to whom correspondence should be addressed.
Cells 2019, 8(6), 604; https://doi.org/10.3390/cells8060604
Submission received: 15 May 2019 / Revised: 9 June 2019 / Accepted: 13 June 2019 / Published: 17 June 2019
(This article belongs to the Special Issue Hepatitis C Virus and Host Interactions)

Abstract

:
Hepatitis C virus (HCV) infection is commonly attributed as a major cause of chronic hepatotropic diseases, such as, steatosis, cirrhosis and hepatocellular carcinoma. As HCV infects only humans and primates, its narrow host tropism hampers in vivo studies of HCV-mammalian host interactions and the development of effective therapeutics and vaccines. In this context, we will focus our discussion on humanized mice in HCV research. Here, these humanized mice are defined as animal models that encompass either only human hepatocytes or both human liver and immune cells. Aspects related to immunopathogenesis, anti-viral interventions, drug testing and perspectives of these models for future HCV research will be discussed.

1. Introduction

First identified in 1989, hepatitis C virus (HCV) is an enveloped, positive-sense, single-stranded ribonucleic acid (RNA) virus belonging to the genus Hepacivirus of family Flaviviridae [1,2]. It is hepatotropic and a leading cause of liver disease including acute hepatitis, chronic hepatitis, cirrhosis and hepatocellular carcinoma. The global prevalence of HCV infection is estimated at 3%, with 170 million chronically infected individuals [3,4,5]. Infection is transmitted through blood and most frequently occurs by perinatal transmission, unsafe invasive healthcare procedures and improper injection practices [6,7].
To date, there are 7 major HCV genotypes (1–7) [8]. Despite their different geographical distributions, there is about 30% heterogeneity in the viral genomic sequence between each genotype [9,10]. For example, genotype 4 is found in Egypt, genotype 5 in South Africa, genotype 7 in Democratic Republic of Congo (DRC) and genotypes 1a, 1b, 2 and 3 within United States of America [9,11]. On the other hand, having one of the highest global incidences of HCV, the predominant genotypes in Asia are genotypes 1 (Singapore, Indonesia and Philippines), 3 (Malaysia and Thailand) and 6 (Vietnam, Myanmar and Laos) [12].
A spectrum of in vitro and in vivo models including; cell culture, tree shrew, zebrafish, chimpanzee and viral protein transgenic mouse models have been used to study HCV [4]. Even though these models have advanced parts of HCV research, the inability to fully recapitulate relevant clinical features as observed in human patients, create a need for improved animal models to enable the effective study of HCV immunopathogenesis and evaluation of new therapeutics and prophylactic vaccines [13].
Hepatitis C virus exhibits a narrow host tropism, infecting the hepatocytes of a limited range of species, particularly human and chimpanzee [14,15]. Being genetically similar to humans, the chimpanzee has been instrumental in the study of HCV infection. Its contributions to research include the identification of HCV, understanding HCV immunopathogenesis, validation of molecular tools for drug discovery and evaluation of drug safety/efficacy [16]. The accumulation of scientific discoveries, has translated into several Food and Drug Administration (FDA)-approved antivirals [17]. While the chimpanzee models have provided valuable insights to HCV research, the use of these models is limited due to the lack of human cells/immune system, ethics and economic reasons [16].
Existing limitations and the need to further our understanding of HCV viral immunopathogenesis and treatments; have led scientists to establish HCV infected humanized mouse models [18,19,20,21]. In this context, humanized mice are defined as mice engrafted with only human hepatocytes or with human immune system and hepatocytes. These small animal models allow in vivo HCV infection as per clinical settings, therefore enabling the analysis of human-specific host immune responses [18,19,20,21]. In this review, we provide an overview of the currently available humanized mouse models that have proven valuable for the study of HCV and discuss their main benefits and weaknesses.

2. Life Cycle of HCV

The HCV life cycle is only partially understood as there is a complex network of cell surface molecules involved in mediating viral entry, hence making it challenging to establish a reliable in vitro model of replication [22]. It has been shown that there are seven steps to the lifecycle of HCV, namely; attachment, entry, uncoating, translation, replication, assembly and maturation. Viral particles of HCV circulate the blood either as free particles or are surrounded by low-density lipoproteins from the host. These HCV virions then enter the cells via clathrin-mediated endocytosis by sequentially binding various receptor molecules of the target cell membrane [22,23,24].
Low pH of the endosome triggers fusion of cellular and viral membranes, in turn causing the capsid to be disorganized. Upon disruption of the viral capsid in the endocytic compartment, single-stranded RNA genome is uncoated and released into the cytoplasm. Following this, the RNA genome is translated at the rough endoplasmic reticulum (ER) to form a single polyprotein precursor which is cleaved by cellular and viral proteases into single proteins. Products from this process include, structural core and envelope glycoproteins E1 and E2; non-structural proteins important for viral assembly and release, p7 viroporin and non-structural protein 2 (NS2); protease complex, NS3 and NS4A; membrane-associated protein which mediates interactions between virus and host, NS4B; Zinc-binding and proline-rich hydrophilic phosphoprotein essential for replication of HCV RNA, NS5A and a RNA polymerase, NS5B [22,23,24].
Through a minus-strand replicative intermediate, an array of host factors and non-structural proteins form a replication complex which makes multiple copies of HCV RNA genome. Virions undergo maturation and are enveloped by endogenous lipoproteins as they are being assembled in an ER-derived compartment and finally released through exocytosis via a Golgi-dependent secretory pathway or transmitted to other cells by a cell-free mechanism [22,23,24].
Among the most frequently utilized methods to study in vitro replication of HCV, cell culture HCV (HCVcc) and HCV trans-complemented particles (HCVTCP) are most widely applied. Application of these cell lines has not only allowed the identification of HCV entry factors but also established the virion structure of HCV, determined its biochemical properties and evaluated relevant therapeutics. However, all in vitro methods use Huh-7 cells. Even though these cells are permissive to HCV replication, it has different mechanisms, locations of HCV receptors and absent polarity as compared to primary hepatocytes [22,23,24]. As a result, the life cycle of HCV is not accurately reproduced in this model, hence making humanized mice an attractive platform to further understand the life cycle of HCV.

3. Immunopathogenesis of HCV

Once hepatocytes are infected with HCV, the immune system is activated to clear the virus [25,26]. To do this, a cascade of immunological events is triggered, dendritic cells (DC), hepatic stellate cells and Kupffer cells secrete cytokines (MIP-1α, IL-12, IL-15 and IL-18) to recruit NK cells, which produce IFNγ in the liver [27,28]. Type I and III interferons produced, then activates sinusoidal endothelial cells to secrete chemokines (CXCL10, MIG), which attracts T cells for viral clearance [29].
However, the highly mutable HCV is capable of evading host immune system to develop chronic infection. It achieves this through a number of ways, first via the disruption of DCs. It has been shown that HCV can increase levels of indoleamine-2, 3, -deoxygenase to dysregulate DC maturation and antigen-presenting functions [30]. Induce plasmacytoid dendritic cell (pDC) apoptosis and disrupt proteasomal subunits of DC to affect their phagocytic functions [31]. Unresponsiveness to chemokine CCL21 also impairs DC migration to lymphoid tissues and downregulates HCV-sensing toll-like receptors (TLRs) as well as critical adaptor molecules (TRIF, TRAF6) [32]. Impaired DC function, early in HCV infection results in low levels of NK cell maturation and an immunosuppressive regulatory T-cell phenotype due to defective priming of CD4+ and CD8+ T cells [33,34,35].
Second, even though it has been shown that there is no difference in the cytotoxicity of natural killer (NK) cells between groups of individuals including HCV infected, recovered patients and healthy donors [36,37,38,39]. Researchers have demonstrated that NK cells may be inhibited by ways including but not limited to, reduction in NK cell expression of microRNA-155, which upregulates inhibitory receptor Tim-3; disrupted NK cell activation receptor expression and core-induced stabilization of human leukocyte antigen E (HLA-E) [36,37,38,39,40,41].
Third, assisting HCV in evading the immune system also include CD56+ natural killer T cells (NKT), which are found to be at low levels in acute HCV infection [42]. Additionally, in chronic infection, naïve antigen-specific CD8+ T cells have been shown to be primed by NKT cells to produce an immunosuppressive environment, further enabling HCV to evade the immune system [43].
Lastly, early adaptive immune responses are crucial in determining the acute or chronic outcome of HCV infection. About 30% of infected individuals are able to eliminate virus-infected cells via strong and sustained responses by CD8+ T cells and CD4+ T cells [44,45,46,47,48]. A defined role for B cells in HCV infection has not been dissected and current results are controversial [49]. In chronic infection, the inability of T cells to control HCV infection could be due to a range of reasons, such as, defective priming of T-cells by DC, abnormal T cell priming by intrahepatic antigen-presenting cells, T cell anergy and high expression levels of viral antigens in hepatocytes that causes elevated levels of regulatory T cell (Treg) subsets, which in turn creates an immunosuppressive immune environment [28,50,51,52].

4. The Role of HCV in the Progression of Liver-Associated Diseases

In acute hepatitis infection, components within the extracellular matrix, including glycoproteins and proteoglycans are synthesized in a tridimensional network to limit inflammatory reactions during the early phases of infection [53]. However, when infected individuals are unable to clear the virus, chronic HCV infection occurs. During this state, activated hepatic stellate cells acquire a myofibroblastic phenotype which allows it to attract leukocytes, proliferate and produce extracellular matrix proteins and collagen, eventually resulting in hepatic fibrogenesis where these components are deposited in the liver and eventually damage the organ [54].
The transition from liver fibrogenesis and cirrhosis involves inflammatory factors, vascular occlusion and angiogenesis. The activity of hepatic stellate cells is mainly regulated by important elements such as, transforming growth factor β (TGF-β), chemokines and adipokines [54]. Similar to fibrogenesis, both the formation of angiogenesis and cirrhosis are mediated by extracellular matrix remodeling and the activation of growth factors, including but not exclusive of TGF-β, vascular endothelial growth factor (VEGF), and platelet-derived growth factor (PDGF), and increased gene expression of adhesion molecules [55,56].
Hepatocellular carcinoma (HCC) is the most common type of primary liver malignancy and the fourth leading cause of cancer-related deaths globally [57]. In most cases, HCV-associated hepatocarcinogenesis is preceded and driven by hepatic inflammation, oxidative stress and cellular deoxyribonucleic acid (DNA) damage [54,58]. All of which, is progressively caused by hepatic fibrogenesis and cirrhosis [53,59]. Once infected, hepatitis C virus does not integrate into the host’s genome. Instead, it remains as an episome in the endoplasmic reticulum (ER), encoding for survival and growth proteins, which induces the expansion of infected hepatocytes [60,61]. Proliferation of infected hepatocyte activates signaling pathways, such as Wnt/β-catenin and MAPK/ERK, which results in mutations implicated during the transition of chronic viral hepatitis to HCC [62,63,64].

5. Types of Humanized Mice for HCV Studies

5.1. Humanized Mouse Models with Only Human Hepatocytes

As HCV has a narrow host tropism, there is a lack of in vivo models that can recapitulate clinical settings, therefore limiting the development of effective vaccines and treatments [65]. Even though non-human primate animal models have contributed significantly to HCV research, ethical, financial and a lack of human immune system have restricted their use in recent years [16].
This limitation raised enormous demands for a small, easy to manipulate and cost-effective model that is able to recapitulate clinical scenarios [14]. These include different types of humanized mice, of which immunodeficient mice engrafted with either only human hepatocytes or both human immune system and hepatocytes [18,19,21,66,67,68,69,70,71,72,73,74,75,76]. These models allow the study of HCV infection and discovery of potential treatments. Here, we review humanized mouse models currently available for HCV research and have included important information, such as details of their study and advantages as well as limitations for each model.

5.1.1. Alb-uPA/SCID

The Alb-uPA/SCID model was designed to investigate neonatal bleeding disorders [77]. The over-expression of murine urokinase-type plasminogen activator (uPA) with an albumin promoter resulted in an increase in plasma uPA levels, severe hemorrhagic events, accelerated hepatocytes and neonatal death [77,78]. However, in newborn mice, scientists observed that the elevated uPA levels gradually decreased to normal levels through random somatic deletions of the uPA transgene within hepatocytes and therefore abolished transgene expression. These transgenic uPA deficient cells exhibited significant replicative advantage, capable of selective proliferation and complete liver regeneration [78] (Table 1).
In a study to investigate the replicative capacity of adult mouse hepatocytes into newborn Alb-uPA/SCID mice, these mature hepatocytes replaced up to 80% of the hepatocytes in recipient liver and restored liver deficiency [78,79]. By transplanting normal human hepatocytes into Alb-uPA/SCID mice, in vivo human HCV infection was demonstrated for the first time by Mercer et al. [80]. These chimeric Alb-uPA/SCID mice were inoculated with HCV infected human serum and developed prolonged HCV infections of up to 35 weeks with high viral titers. The human liver Alb-uPA/SCID mouse models have proven valuable for its contributions to understanding the basic biology of HCV, evaluation of antiviral therapies and neutralizing antibodies [81,82,83,84,85,86,87,88,89].
Yet, limitations of this model include, high neonatal mortality rate, susceptibility to kidney disorders, small body size and weight, limited time window for transplantation, breeding difficulties and the inability to expand engrafted human hepatocytes due to the spontaneous genotype reversion in recipient mouse hepatocytes, that effectively outcompete the transplanted human cells during liver repopulation [4,90].

5.1.2. cDNA-uPA/SCID

To address existing issues, variants of uPA mice, such as hemizygous cDNA-uPA/SCID was established and successfully infected with HCV in vivo [91]. This strain was generated using embryonic stem cell techniques and attained appropriate levels of uPA expression. As compared to the original strain, the liver of this model was not detrimentally damaged, experienced fewer kidney disorders, and had higher body weight and longer survival rates. However, even though HCV viremia was significantly higher in this model, it was unable to maintain viremia for longer than 8 weeks [70].

5.1.3. MUP-uPA SCID

The other variant, MUP-uPA SCID/Bg model was constructed by backcrossing transgenic mice carrying the uPA gene driven by MUP promoter onto a SCID/Beige background [92]. As compared to the original strain, the MUP-uPA SCID/Bg mouse is healthier, has a longer time window of up to 1 year of age for hepatocyte transplantation and is susceptible to infection with HCV genotype 1-6 [71].

5.1.4. uPA/NOG

Constructed on severely immunodeficient non-obese diabetic (NOD)/Shi-scid/IL-2Rγnull (NOG) mice, the third variant is the uPA/NOG mouse model [93]. This model offers several advantages over classical Alb-uPA/SCID mice, enabling, minimal neonatal lethality, increased breeding efficiency, improved recipient survival, simplified surgical manipulation and higher xenogeneic cell engraftment. At present, HCV infection has not been reported in uPA/NOG mice.

5.1.5. Fah−/−/Rag2−/−/Il2rg−/− (FRG)

The FRG strain of mice is a triple knockout mouse model [73]. Deletion of fumarylacetoacetate (Fah), a tyrosine catabolic enzyme results in the accumulation of hepatotoxic metabolites such as Fah and maleylacetoacetate which induces liver damage and is lethal [94] (Table 2). Oral administration of 2-(2-nitro-4-trifluoro-methylbenzoyl)1,3-cyclohexedione (NTBC) blocks the enzyme hydroxyphenylpyruvate dioxygenase which prevents the accumulation of hepatotoxic metabolites, liver damage and maintains FRG mice in a healthy state [94]. Upon NTBC withdrawal, mouse liver can be repopulated with human hepatocytes [73,94].
The FRG model is advantageous to classical Alb-uPA/SCID mice in a range of ways including, that it has a higher rate of chimerism, is easy to breed and there is absolutely no spontaneous transgene revision, renal disorders and limitations in the time frame for transplantation as liver repopulation is controlled by NTBC withdrawal [74].
An absence of spontaneous transgene reversion in FRG enables serial transplantation of human hepatocytes as the Fah deficient mouse hepatocytes are unable to compete with transplanted human cells during liver repopulation. Because of this, an infinite number of hepatocytes from a single donor can be produced via serial transplantation over a few generations of mice, hence making it a cost-effective model, attractive for large scale studies requiring human hepatocytes or mice.
It has been reported that a high rate of liver chimerism of up to 95% human hepatocytes in FRG mice is generated by high transplantation dose of human hepatocytes. These mice were successfully infected with 4 HCV genotypes and were responsive to antiviral and neutralizing antibodies [74,95,96]. Improvements to FRG models are constantly being developed by adding human oncostatin-M to enhance human hepatoblastoma repopulation in recipient mouse liver by 5-100-fold [96]. These syngeneic liver and immune system mice are reconstituted with functional human T and B lymphocytes, monocytes and NK cells, which are able to support HCV infection, hence making it an ideal model for the study of HCV infection in the liver.

5.1.6. TK-NOG

In 2011, a new mouse model expressing a transgene, herpes simplex virus type 1 thymidine kinase (HSVtk), within the liver of immunodeficient NOG mice (TK-NOG) [97] was created. Mouse liver cells diminished after the exposure of ganciclovir (GCV) and human hepatocytes were stably maintained without exogenous drugs. It has been shown that serum alanine aminotransferase (ALT) levels are increased in TK-NOG mice after the HCV infection induced by GCV treatment, rendering these mice useful for the study of HCV virology [76,97]. However, drawback of this model includes a lack of human immune system, absence of liver disease post-infection, low body weight and high rates of renal disorders and mortality [76,97].

5.2. Humanized Mouse Models with Human Immune System and Hepatocytes

Human liver chimeric mice have provided valuable insights into HCV infection, as well as the evaluation of antiviral treatments. However, a major limitation includes the lack of a functional human immune system, which impedes the understanding of human-specific immune responses during HCV immunopathogenesis and novel therapeutics/vaccines [67,98].
Histopathological features associated with chronically infected HCV patients, such as hepatic inflammation, fibrosis, cirrhosis and HCC has not been reported in human liver chimeric mice. This indicates that the human immune system plays a key role in disease progression. Therefore, humanized mouse models with both human hepatocytes and immune system should be used for HCV research.

5.2.1. AFC8-hu HSC/Hep

Driven on an albumin promoter (AFC8), the AFC8-hu HSC/Hep strain of transgenic mouse expresses FK506 binding protein (FKBP) and caspase 8 [66,67]. To induce liver cell death, recipient mouse is administered AP20187, a synthetic drug that induces dimerization of active caspase 8, expressed specifically in mouse liver cells [66,67].
To study HCV pathogenesis, human CD34+ hematopoietic stem cells (HSC) and human hepatocyte progenitors were co-transplanted into Balb/C-Rag2−/−-γC−/− mice and infected with HCV isolates [66]. Infected mice demonstrated elevated levels of ALT, pDC, NK cells, macrophages, T cells, liver inflammation and fibrosis [66,67].
Limitations of this model include, low human liver chimerism of 15%, undetectable HCV viremia in the blood, absence of HCV-specific antibodies and hepatitis C virus genomic RNA in the liver, is detectable in only half of infected mice, after 1-4-month post-infection [66,67] (Table 3).

5.2.2. NSG-DRB*0101

Even though current humanized mouse models with both human hepatocytes and immune system are crucial for modelling diseases and testing drugs, it has been reported to have some functional deficiencies. The NSG-DRB*0101 mouse model was developed to determine if the inclusion of human leukocyte antigen (HLA) would improve the development of functional human T and B cells [99,100,101,102,103].
Expression of HLA in humanized mice allowed the development of a partially functional adaptive human immune system during viral infection and the generation of HLA-A2 restricted virus-specific T cells. However, drawbacks in this model include difficulty in sourcing HSC with the same HLA type, minute levels of human NK cells, defective recruitment and homing of human immune cells, and restricted inter-species cross-reactivity of cytokines and chemokines produced during infection [68].

5.2.3. HIL Mice

A humanized mouse model with both human hepatocytes and immune system (HIL mice) was established by intrahepatically injecting mice with human fetal liver cells [18,104,105,106]. Unlike other chimeric liver mouse models, these mice do not require additional drug treatments or transgene modifications [18,104].
Similar to clinical scenarios, these mice developed liver inflammation, upregulated human cytokines and developed fibrosis [18,19,20,21]. Long-term effects of HCV in HIL mice were monitored for up to 28 weeks, where infected mice demonstrated higher incidences of fibrosis. Immune profile analysis of HCV infected mice showed elevated numbers of T cells and monocytes/macrophages in granulomatous inflammation [18,19,20,21] (Table 4).
In addition, at 28 weeks’ post-infection, human proinflammatory cytokines such as interferon gamma (IFN-γ), monocyte chemoattractant protein 1 (MCP-1) and interleukin 18 (IL-18) were significantly increased in the plasma of HIL mice [19]. When treated with pegylated-interferon-α-2A (PEG-IFNα-2A), progression of HCV liver pathogenesis was blocked by PEG-IFNα-2A, demonstrating that HIL mice are able to reproduce HCV infection and immunopathogenesis [107,108,109,110,111,112]. The main drawback of this model is the low levels of B cells detected as compared to clinical settings [19].

6. Hepatitis C Treatment

Humanized mouse models with only human hepatocytes have been crucial in initial therapeutic tests. However, due to the lack of a functional human immune system, the study of in vivo drug interactions and vaccine development has been precluded [113,114]. On the other hand, humanized mouse models with both human immune system and hepatocytes are able to efficiently recapitulate immune-mediated events in HCV and are essential in developing novel vaccines and treatments.
Depending on the genotype of HCV, different combinations of therapeutics including interferon (IFN), direct acting antiviral (DAA) and ribavirin (RBV) are usually prescribed [115,116,117,118]. Some obstacles faced in current therapies include factors such as, patients being resistant to existing treatments, poor tolerance to side effects which prevent individuals from completing therapy, limited efficacy, high cost and emergence of drug-resistant viral variants [119,120]. For these reasons, constant research and development is needed to develop effective treatment and vaccine options. In this review, some antivirals such as Claudin-1 (CLDN1), Interferon-λ, NA808, direct acting antivirals (DAA) and PEG-IFNα-2A will be discussed.

6.1. Claudin-1 Antibody

As a tight junction protein, functions of CLDN1 include regulation of HCV entry and transmission from cell to cell [121]. Claudin-1 antibody functions by eliminating HCV viral activity through inhibition of HCV cell entry. When Alb-uPA/SCID mice were administered CLDN1, virus-mediated signaling pathways were induced and persistent HCV infection was cleared, demonstrating effectiveness of the treatment [95,111] (Table 5).

6.2. Interferon-λ

Interferon is a cytokine that is essential in mediating the innate immune system and is the first response against HCV viral infection. The innate immunity is crucial for initial defense against viral pathogens; therefore, antiviral drugs that can activate innate immune response are ideal candidates for HCV treatment [122,123,124].

6.3. NA808

An alternative method of activating the innate immune system is through a complex, cationic liposome (LIC) and synthetic double-stranded RNA analog, polyinosinic-polycytidylic acid (Poly I:C) (LIC-pIC). Activation of this complex induces IFN-λ mediated antiviral response of HCV-infected human hepatocytes and is a potential for overcoming resistance to therapies across different HCV genotypes [124,125].
Katsume et al. [126] identified a novel class of serine palmitoyltransferase (SPT) inhibitor, known as NA808. Derived from fungal metabolites, this SPT inhibitor prevents the synthesis of sphingolipid, in turn disrupting HCV replication complex, hence inhibiting HCV replication.

6.4. DAA

Studies have shown that even in the absence of immunomodulatory IFN, direct acting antivirals (DAA) are able to activate the innate immune system. Administration of DAA suppresses HCV expression and significantly reduces levels of chemokines [96]. Post-DAA treatment, there is an increase in protein expression of Cardif and IFITM1, suggesting that DAA are able to block HCV expansion in hepatic cells [96,127].
Currently, an array of DAA is being developed as novel strategies to target HCV [11]. These drugs have been shown to provide shorter treatment times, higher cure rates and reduced side effects. The main classes of DAA are NS3/4 protease inhibitors (PIs), NS5A inhibitor, Nucleoside and nucleotide NS5B polymerase inhibitors. These drugs directly target viral proteins halt HCV replication in host cells. In clinics, treatments for HCV infected patients include combinations of DAA. Two significant groups of DAA are widely used, Boceprevir and Telaprevir blocks NS3/4A serine protease and inhibits crucial proteins for viral cycle, while NS5A inhibits protease to halt HCV replication [128,129,130,131].

6.5. PEG-IFNα-2A

Human PEG-IFNα-2A is often used in clinical treatment of HCV and has a specific role in inhibiting HCV replication and regressing HCV-associated disease progression. It has been shown that HIL mice are able to reproduce HCV infection, immunopathogenesis and drug response as per clinical settings. When tested with PEG-IFNα-2A, progression of HCV liver pathogenesis was halted. In addition, it was observed that serum ALT levels decreased and mice were protected from liver damage and fibrosis [18,132].

7. HCV Vaccines

Although current treatments have improved the cure rates of HCV, existing limitations that prevent complete disease eradication include; high cost of therapy, limited worldwide accessibility, potential development of drug resistance and an inability to ameliorate long-term effects of chronic infection [4]. Therefore, to relive a worldwide burden of the disease, it is crucial to develop an effective vaccine to prevent the transmission of virus and liver damage [133].
Over the years, a range of adjuvants, vectors and vaccination regimens have been tested [4]. At present, two ways are being used to design HCV vaccines. First, through inducing an antibody response that targets the exterior surface of viruses [134]. However, as HCV is highly variable among strains and mutates quickly, this method is extremely challenging. Second, generate broadly neutralizing antibodies to induce viral inactivity and third, stimulate broad T cell responses to clear infected hepatocytes [134,135,136,137].
The task of developing a vaccine against HCV is an extremely challenging process. Humanized mouse models can be readily used to better understand the immunopathogenesis of HCV and also as a preclinical platform to determine the effectiveness of novel vaccines and therapeutics.
Table 5. Antiviral treatments for HCV.
Table 5. Antiviral treatments for HCV.
Drug nameDAAPEG-IFNα-2ANA808Interferon-λClaudin-1
Trade nameMany different DAA in the market with individual namesPegasys---
Manufacturing companyA range of companies manufacture DAARoche Pharmaceuticals---
Mechanism of actionDisrupts HCV viral life cycle by shortening the length of therapy, minimizing size effects, targeting the virus, improving sustained virological response ratesActs as interferon within the immune systemHalts HCV replication via non-competitive inhibition of Serine Palmitoyltransferase (SPT), hence reducing viral load in miceIFN-λ binds to heterodimeric IFN-λ receptor, activates STAT phosphorylation-dependent signal cascade which induces hundreds of IFN-stimulated genes, which in turn modulates a range of immune functionsBlocks entry of HCV
Commonly used in combinationPEG-IFNα-2A and RibavirinDAA and RibavirinPEG-IFNα-2A, HCV polymerase/ protease inhibitorsNot fully characterizedRibavirin
Rate of SVR~95%~79% in genotype
1~89% in genotype 2 or 3
Not fully characterizedNot fully characterizedNot fully characterized
Stage of clinical trial-Completed and in market---
Side effectsFatigue, gastrointestinal symptoms, anemia, headache and dyspneaHeadache, fatigue, depression, insomnia, nausea, pain at site of injection, fever, psychosis, autoimmune disorders, blood clots and infectionNot fully characterized
Inhibition of host enzyme might result in mechanism-related toxicities/side effects
Not fully characterizedNot fully characterized
Advantages
  • Effective
  • Wide range of DAA
  • Safe
  • Effective particularly in patients with IL28B genotype
  • No development of resistant mutants
  • Able to prevent replication of HCV genotypes 1a, 1b, 2a, 3a, and 4a
Not fully characterized Not fully characterized
Limitations
  • Expensive
  • Unavailable in some regions of the world
  • Extensive and systemic side effects
  • Limited efficacy
  • Viral and host factors can result in non-responders
Not fully characterizedNot fully characterizedNot fully characterized
ReferencesWilliford et al. (2016) [3]Huang et al. (2017) [138]Katsume et al. (2013) [126]Bruening et al. (2018) [139]
  • Colpitts et al. (2018) [140]
  • Evans et al. (2017) [121]
  • Meertens et al. (2008) [136]
Abbreviations - DAA: Direct-acting antiviral, HCV: Hepatitis C virus, PEG-IFNα-2A: Pegylated-interferon-α-2A, SVR: Sustained virological response, SPT: Serine Palmitoyltransferase
Table 6. Immunotherapy for HCC.
Table 6. Immunotherapy for HCC.
Drug nameNivolumabPembrolizumabTremelimumabDurvalumabIpilimumab
Commercial nameOpdivoKeytruda-ImfinziYervoy
CompanyBristol-Myers Squibb (BMS)Merck Sharp & Dohme (MSD)MedImmuneMedImmuneBMS
Target moleculePD-1PD-1CTLA-4PDL-1CTLA-4
Target cellT lymphocyteT lymphocyteT lymphocyteTumor cellT lymphocyte
Stage of clinical trialApproved by FDA and commercially availableApproved by FDA and commercially availablePhase IIIApproved by FDA and commercially availableApproved by FDA and commercially available
ReferencesEl-Khoueiry AB et al. (2017) [141]Zhu et al. (2018) [142]Sangro et al. (2013) [143]Wainberg et al. (2017) [144]-
Abbreviation: BMS: Bristol-Myers Squibb, MSD: Merck Sharp & Dohme.

8. Immunotherapy for Hepatocellular Carcinoma

Globally, Hepatocellular carcinoma (HCC) is one of the most debilitating and fatal cancers [145]. Despite promising data from preclinical and clinical trials, current strategies for cancer treatments are limited [146]. The establishment of humanized mice has advanced knowledge of important immunopathogenesis and oncogenic signaling pathways within the diseased microenvironment of HCC [21]. In particular, HIL mice has allowed the dissection of cancer initiation and progression, as well as the opportunity to test and evaluate a diverse range of immune-oncological interventions including but not limited to, Nivolumab, Pembrolizumab, Tremelimumab, Durvalumab and Ipilimuma (Table 6).

9. Future direction and Conclusion

Humanized mouse models with chimeric human liver or both human immune system and hepatocytes are imperative for the characterization of HCV infection and development of therapeutics and vaccines [18,19,20,21,66,68]. As these models are of utmost importance, constant improvements are necessary to push boundaries and create models with superior clinical accuracy.
Even though current models of humanized mice are able to support HCV infection, some limitations that need to be improved on include; first, humanization levels of hepatocytes and immune cells can be further enhanced in humanized mice. This can be done through the supplementation of cytokines (IL-1β, IL-2, IL-7, and GM-CSF), to enable differentiation and maturation of HSC which can give rise to a range of immune cell subsets [147,148,149]. A more human-specific microenvironment will enable in-depth characterization of HCV immunopathogenesis and therapeutic development. Second, elimination of selective host-specific factors may improve HCV infection efficiency. For example, studies have demonstrated that very-low-density lipoprotein (VLDL) blocks HCV cell attachment; therefore, removing VLDL may increase the infection efficiency of HCV [150].
Third, the liver sinusoidal endothelium (LSEC) has a paramount role in shaping intrahepatic immune responses by mediating antigen-presentation and immune cell homing into the liver but is of mouse origin [151,152]. Finding methods to humanize this anatomy will provide valuable insights into human immune cell migration via the liver endothelium. Fourth, to fully mimic clinical settings, induced pluripotent stem cells (iPSC) may be used to create patient matched mice for HCV and HCC studies [21,153].
The lack of effective therapies and vaccine for HCV highlights an unmet clinical need. Advancements in developing humanized mouse models will provide insights into the complexity, redundancy, interdependence and regulatory mechanisms of acute and chronic HCV infection, therefore providing exciting opportunities for in vivo characterization of HCV virus-host interaction and the identification of novel vaccine and treatment strategies.

Author Contributions

K.S.M.Y. took the lead in writing the manuscript. Z.S. and Q.C. contributed to writing. Q.C. supervised the preparation of manuscript.

Funding

This work was supported by the following grants: National Research Foundation Fellowship Singapore NRF-NRFF2017-03 (Q. Chen.), National Research Foundation Singapore Competitive Research Programme NRF2016-CRP001-103 (Q. Chen), Eradication of HBV TCR Program: NMRC/TCR/014-NUHS/2015 and NMRC/OFLCG/003/2018 from National Medical Research Council, Singapore (Q. Chen) and EMULSION programme H18/01/a0/017 Industry Alignment Fund Pre-positioning (IAF-PP) from A*STAR Singapore (Q. Chen).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Major, M.E.; Feinstone, S.M. The molecular virology of hepatitis C. Hepatology 1997, 25, 1527–1538. [Google Scholar] [CrossRef] [PubMed]
  2. Frei, B. To C or not to C, that is the question! J. Am. Coll. Cardiol. 2003, 42, 253–255. [Google Scholar] [CrossRef]
  3. Williford, S.E.; McGivern, D.R. Mechanism of action of direct-acting antivirals: new insights into the HCV life cycle. In Hepatitis C Virus II.; Miyamura, T.L.S., Walker, C., Wakita, T., Eds.; Springer: Tokyo, Japan, 2016. [Google Scholar]
  4. Burm, R.; Collignon, L.; Mesalam, A.A.; Meuleman, P. Animal models to study hepatitis C virus infection. Front Immunol. 2018, 9, 1032. [Google Scholar] [CrossRef] [PubMed]
  5. Lavanchy, D. The global burden of Hepatitis C. Liver Int. 2009, 29, 74–81. [Google Scholar] [CrossRef] [PubMed]
  6. Gish, R.G.; Afdhal, N.H.; Dieterich, D.T.; Reddy, K.R. Management of hepatitis C virus in special populations: Patient and treatment considerations. Clin. Gastroenterol. Hepatol. 2005, 3, 311–318. [Google Scholar] [CrossRef]
  7. Liang, T.J.; Rehermann, B.; Seeff, L.B.; Hoofnagle, J.H. Pathogenesis, Natural History, Treatment, and Prevention of Hepatitis C. Ann. Intern. Med. 2000, 132, 296–305. [Google Scholar] [CrossRef] [Green Version]
  8. Smith, D.B.; Bukh, J.; Kuiken, C.; Muerhoff, A.S.; Rice, C.M.; Stapleton, J.T.; Simmonds, P. Expanded classification of hepatitis C virus into 7 genotypes and 67 subtypes: Updated criteria and genotype assignment web resource. Hepatology 2014, 59, 318–327. [Google Scholar] [CrossRef]
  9. Murphy, D.G.; Sablon, E.; Chamberland, J.; Fournier, E.; Dandavino, R.; Tremblay, C.L. Hepatitis C virus genotype 7, a new genotype originating from central Africa. J. Clin. Microbiol. 2015, 53, 967–972. [Google Scholar] [CrossRef]
  10. Lindenbach, B.D.; Rice, C.M. Unravelling hepatitis C virus replication from genome to function. Nature 2005, 436, 933–938. [Google Scholar] [CrossRef]
  11. Nguyen, L.H.; Nguyen, M.H. Systematic review: Asian patients with chronic hepatitis C infection. Aliment. Pharmacol. Ther. 2013, 37, 921–936. [Google Scholar] [CrossRef] [Green Version]
  12. Wasitthankasem, R.; Vongpunsawad, S.; Siripon, N.; Suya, C.; Chulothok, P.; Chaiear, K.; Rujirojindakul, P.; Kanjana, S.; Theamboonlers, A.; Tangkijvanich, P.; et al. Genotypic distribution of hepatitis C virus in Thailand and Southeast Asia. PLoS ONE 2015, 10, e0126764. [Google Scholar] [CrossRef] [PubMed]
  13. Vercauteren, K.; de Jong, Y.P.; Meuleman, P. HCV animal models and liver disease. J. Hepatol. 2014, 61, S26–S33. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Von Schaewen, M.; Ploss, A. Murine models of hepatitis C: What can we look forward to? Antivir. Res. 2014, 104, 15–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Park, S.-H.; Rehermann, B. Immune responses to HCV and other hepatitis viruses. Immunity 2014, 40, 13–24. [Google Scholar] [CrossRef] [PubMed]
  16. Lanford, R.E.; Walker, C.M.; Lemon, S.M. The chimpanzee model of viral hepatitis: Advances in understanding the immune response and treatment of viral hepatitis. ILAR J. 2017, 58, 172–189. [Google Scholar] [CrossRef] [PubMed]
  17. Liang, T.J.; Ghany, M.G. Current and future therapies for hepatitis C virus infection. N. Engl. J. Med. 2013, 368, 1907–1917. [Google Scholar] [CrossRef] [PubMed]
  18. Keng, C.T.; Sze, C.W.; Zheng, D.; Zheng, Z.; Yong, K.S.M.; Tan, S.Q.; Ong, J.J.Y.; Tan, S.Y.; Loh, E.; Upadya, M.H.; et al. Characterisation of liver pathogenesis, human immune responses and drug testing in a humanised mouse model of HCV infection. Gut 2016, 65, 1744–1753. [Google Scholar] [CrossRef] [PubMed]
  19. Zheng, Z.; Sze, C.W.; Keng, C.T.; Al-Haddawi, M.; Liu, M.; Tan, S.Y.; Kwek, H.L.; Her, Z.; Chan, X.Y.; Barnwal, B.; et al. Hepatitis C virus mediated chronic inflammation and tumorigenesis in the humanised immune system and liver mouse model. PLoS ONE 2017, 12, e0184127. [Google Scholar] [CrossRef]
  20. Tan-Garcia, A.; Wai, L.-E.; Zheng, D.; Ceccarello, E.; Jo, J.; Banu, N.; Khakpoor, A.; Chia, A.; Tham, C.Y.L.; Tan, A.T.; et al. Intrahepatic CD206+ macrophages contribute to inflammation in advanced viral-related liver disease. J. Hepatol. 2017, 67, 490–500. [Google Scholar] [CrossRef]
  21. Zhao, Y.; Shuen, T.W.H.; Toh, T.B.; Chan, X.Y.; Liu, M.; Tan, S.Y.; Fan, Y.; Yang, H.; Lyer, S.G.; Bonney, G.K.; et al. Development of a new patient-derived xenograft humanised mouse model to study human-specific tumour microenvironment and immunotherapy. Gut 2018, 67, 1845–1854. [Google Scholar] [CrossRef]
  22. Dustin, L.B.; Bartolini, B.; Capobianchi, M.R.; Pistello, M. Hepatitis C virus: Life cycle in cells, infection and host response, and analysis of molecular markers influencing the outcome of infection and response to therapy. Clin. Microbiol. Infect. 2016, 22, 826–832. [Google Scholar] [CrossRef] [PubMed]
  23. Kim, C.W.; Chang, K.-M. Hepatitis C virus: Virology and life cycle. Clin. Mol. Hepatol. 2013, 19, 17–25. [Google Scholar] [CrossRef] [PubMed]
  24. Dubuisson, J.; Cosset, F.-L. Virology and cell biology of the hepatitis C virus life cycle—An update. J. Hepatol. 2014, 61, S3–S13. [Google Scholar] [CrossRef] [PubMed]
  25. Gremion, C.; Cerny, A. Hepatitis C virus and the immune system: A concise review. Rev. Med. Virol. 2005, 15, 235–268. [Google Scholar] [CrossRef] [PubMed]
  26. Holz, L.; Rehermann, B. T cell responses in hepatitis C virus infection: Historical overview and goals for future research. Antivir. Res. 2015, 114, 96–105. [Google Scholar] [CrossRef] [PubMed]
  27. Ahmad, A.; Alvarez, F. Role of NK and NKT cells in the immunopathogenesis of HCV-induced hepatitis. J. Leukoc. Biol. 2004, 76, 743–759. [Google Scholar] [CrossRef] [PubMed]
  28. Ryan, E.J.; O’Farrelly, C. The affect of chronic hepatitis C infection on dendritic cell function: A summary of the experimental evidence. J. Viral Hepat. 2011, 18, 601–607. [Google Scholar] [CrossRef]
  29. Giugliano, S.; Kriss, M.; Golden-Mason, L.; Dobrinskikh, E.; Stone, A.E.L.; Soto-Gutierrez, A.; Mitchell, A.; Khetani, S.R.; Yamane, D.; Stoddard, M.; et al. Hepatitis C virus infection induces autocrine interferon signaling by human liver endothelial cells and release of exosomes, which inhibits viral replication. Gastroenterology 2015, 148, 392–402. [Google Scholar] [CrossRef]
  30. Schulz, S.; Landi, A.; Garg, R.; Wilson, J.A.; van Drunen Littel-van den Hurk, S. Indolamine 2,3-dioxygenase expression by monocytes and dendritic cell populations in hepatitis C patients. Clin. Exp. Immunol. 2015, 180, 484–498. [Google Scholar] [CrossRef] [Green Version]
  31. Leone, P.; Di Tacchio, M.; Berardi, S.; Santantonio, T.; Fasano, M.; Ferrone, S.; Vacca, A.; Dammacco, F.; Racanelli, V. Dendritic cell maturation in HCV infection: Altered regulation of MHC class I antigen processing-presenting machinery. J. Hepatol. 2014, 61, 242–251. [Google Scholar] [CrossRef]
  32. Nattermann, J.; Zimmermann, H.; Iwan, A.; von Lilienfeld-Toal, M.; Leifeld, L.; Nischalke, H.D.; Langhans, B.; Sauerbruch, T.; Spengler, U. Hepatitis C virus E2 and CD81 interaction may be associated with altered trafficking of dendritic cells in chronic hepatitis C. Hepatology 2006, 44, 945–954. [Google Scholar] [CrossRef]
  33. Gondois-Rey, F.; Dental, C.; Halfon, P.; Baumert, T.F.; Olive, D.; Hirsch, I. Hepatitis C virus is a weak inducer of interferon alpha in plasmacytoid dendritic cells in comparison with influenza and human herpesvirus type-1. PLoS ONE 2009, 4, e4319. [Google Scholar] [CrossRef] [PubMed]
  34. Miyazaki, M.; Kanto, T.; Inoue, M.; Itose, I.; Miyatake, H.; Sakakibara, M.; Yakushijin, T.; Kakita, N.; Hiramatsu, N.; Takehara, T.; et al. Impaired cytokine response in myeloid dendritic cells in chronic hepatitis C virus infection regardless of enhanced expression of Toll-like receptors and retinoic acid inducible gene-I. J. Med. Virol. 2008, 80, 980–988. [Google Scholar] [CrossRef] [PubMed]
  35. Gerosa, F.; Gobbi, A.; Zorzi, P.; Burg, S.; Briere, F.; Carra, G.; Trinchieri, G. The reciprocal interaction of NK cells with plasmacytoid or myeloid dendritic cells profoundly affects innate resistance functions. J. Immunol. 2005, 174, 727–734. [Google Scholar] [CrossRef]
  36. Morishima, C.; Paschal, D.M.; Wang, C.C.; Yoshihara, C.S.; Wood, B.L.; Yeo, A.E.T.; Emerson, S.S.; Shuhart, M.C.; Gretch, D.R. Decreased NK cell frequency in chronic hepatitis C does not affect ex vivo cytolytic killing. Hepatology 2006, 43, 573–580. [Google Scholar] [CrossRef] [PubMed]
  37. Yoon, J.C.; Shiina, M.; Ahlenstiel, G.; Rehermann, B. Natural killer cell function is intact after direct exposure to infectious hepatitis C virions. Hepatology 2009, 49, 12–21. [Google Scholar] [CrossRef] [PubMed]
  38. Golden-Mason, L.; Madrigal-Estebas, L.; McGrath, E.; Conroy, M.J.; Ryan, E.J.; Hegarty, J.E.; O’Farrelly, C.; Doherty, D.G. Altered natural killer cell subset distributions in resolved and persistent hepatitis C virus infection following single source exposure. Gut 2008, 57, 1121–1128. [Google Scholar] [CrossRef] [PubMed]
  39. De Maria, A.; Fogli, M.; Mazza, S.; Basso, M.; Picciotto, A.; Costa, P.; Congia, S.; Mingari, M.C.; Moretta, L. Increased natural cytotoxicity receptor expression and relevant IL-10 production in NK cells from chronically infected viremic HCV patients. Eur. J. Immunol. 2007, 37, 445–455. [Google Scholar] [CrossRef]
  40. Cheng, Y.Q.; Ren, J.P.; Zhao, J.; Wang, J.M.; Zhou, Y.; Li, G.Y.; Moorman, J.P.; Yao, Z.Q. MicroRNA-155 regulates interferon-γ production in natural killer cells via Tim-3 signalling in chronic hepatitis C virus infection. Immunology 2015, 145, 485–497. [Google Scholar] [CrossRef]
  41. Jinushi, M.; Takehara, T.; Tatsumi, T.; Kanto, T.; Miyagi, T.; Suzuki, T.; Kanazawa, Y.; Hiramatsu, N.; Hayashi, N. Negative regulation of NK cell activities by inhibitory receptor CD94/NKG2A leads to altered NK cell-induced modulation of dendritic cell functions in chronic hepatitis C virus infection. J. Immunol. 2004, 173, 6072–6081. [Google Scholar] [CrossRef]
  42. Golden-Mason, L.; Castelblanco, N.; Farrelly, C.; Rosen, H.R. Phenotypic and functional changes of cytotoxic CD56pos natural T cells determine outcome of acute hepatitis C virus infection. J. Virol. 2007, 81, 9292–9298. [Google Scholar] [CrossRef] [PubMed]
  43. Wahl, C.; Bochtler, P.; Schirmbeck, R.; Reimann, J. Type I IFN-producing CD4 Vα14i NKT cells facilitate priming of IL-10-producing CD8 T cells by hepatocytes. J. Immunol. 2007, 178, 2083–2093. [Google Scholar] [CrossRef] [PubMed]
  44. Guidotti, L.G.; Chisari, F.V. To kill or to cure: Options in host defense against viral infection. Curr. Opin. Immunol. 1996, 8, 478–483. [Google Scholar] [CrossRef]
  45. Thimme, R.; Oldach, D.; Chang, K.-M.; Steiger, C.; Ray, S.C.; Chisari, F.V. Determinants of viral clearance and persistence during acute hepatitis C virus infection. J. Exp. Med. 2001, 194, 1395–1406. [Google Scholar] [CrossRef] [PubMed]
  46. Diepolder, H.M.; Zachoval, R.; Hoffmann, R.M.; Jung, M.C.; Pape, G.R.; Wierenga, E.A.; Santantonio, T.; Eichenlaub, D. Possible mechanism involving T-lymphocyte response to non-structural protein 3 in viral clearance in acute hepatitis C virus infection. Lancet 1995, 346, 1006–1007. [Google Scholar] [CrossRef]
  47. Missale, G.; Bertoni, R.; Lamonaca, V.; Valli, A.; Massari, M.; Mori, C.; Rumi, M.G.; Houghton, M.; Fiaccadori, F.; Ferrari, C. Different clinical behaviors of acute hepatitis C virus infection are associated with different vigor of the anti-viral cell-mediated immune response. J. Clin. Investig. 1996, 98, 706–714. [Google Scholar] [CrossRef] [PubMed]
  48. Chang, K.-M. Immunopathogenesis of hepatitis C virus infection. Clin. Liver Dis. 2003, 7, 89–105. [Google Scholar] [CrossRef]
  49. Mizuochi, T.; Ito, M.; Takai, K.; Yamaguchi, K. Peripheral blood memory B cells are resistant to apoptosis in chronic hepatitis C patients. Virus Res. 2011, 155, 349–351. [Google Scholar] [CrossRef]
  50. Niesen, E.; Schmidt, J.; Flecken, T.; Thimme, R. Suppressive effect of interleukin 10 on priming of naive hepatitis C virus—Specific CD8+ T cells. J. Infect. Dis. 2014, 211, 821–826. [Google Scholar] [CrossRef] [PubMed]
  51. Zimmermann, M.; Flechsig, C.; Monica, N.L.; Tripodi, M.; Adler, G.; Dikopoulos, N. Hepatitis C virus core protein impairs in vitro priming of specific T cell responses by dendritic cells and hepatocytes. J. Hepatol. 2008, 48, 51–60. [Google Scholar] [CrossRef] [PubMed]
  52. O’Beirne, J.; Mitchell, J.; Farzaneh, F.; Harrison, P.M. Inhibition of major histocompatibility complex Class I antigen presentation by hepatitis C virus core protein in myeloid dendritic cells. Virology 2009, 389, 1–7. [Google Scholar] [CrossRef] [Green Version]
  53. Marcellin, P.; Asselah, T.; Boyer, N. Fibrosis and disease progression in hepatitis C. Hepatology 2002, 36, S47–S56. [Google Scholar] [CrossRef] [PubMed]
  54. Lee, U.E.; Friedman, S.L. Mechanisms of hepatic fibrogenesis. Best Pract. Res. Clin. Gastroenterol. 2011, 25, 195–206. [Google Scholar] [CrossRef]
  55. Eng, F.J.; Friedman, S.L.; Fibrogenesis, I. New insights into hepatic stellate cell activation: The simple becomes complex. Am. J. Physiol. Gastrointest. Liver Physiol. 2000, 279, G7–G11. [Google Scholar] [CrossRef] [PubMed]
  56. Fernández, M.; Semela, D.; Bruix, J.; Colle, I.; Pinzani, M.; Bosch, J. Angiogenesis in liver disease. J. Hepatol. 2009, 50, 604–620. [Google Scholar] [CrossRef]
  57. Pellicoro, A.; Ramachandran, P.; Iredale, J.P.; Fallowfield, J.A. Liver fibrosis and repair: Immune regulation of wound healing in a solid organ. Nat. Rev. Immunol. 2014, 14, 181–194. [Google Scholar] [CrossRef] [PubMed]
  58. Okuda, M.; Li, K.; Beard, M.R.; Showalter, L.A.; Scholle, F.; Lemon, S.M.; Weinman, S.A. Mitochondrial injury, oxidative stress, and antioxidant gene expression are induced by hepatitis C virus core protein. Gastroenterology 2002, 122, 366–375. [Google Scholar] [CrossRef] [Green Version]
  59. Hayes, C.N.; Zhang, P.; Zhang, Y.; Chayama, K. Molecular mechanisms of hepatocarcinogenesis following sustained virological response in patients with chronic hepatitis C virus infection. Viruses 2018, 10, 531. [Google Scholar] [CrossRef]
  60. Arzumanyan, A.; Reis, H.M.; Feitelson, M.A. Pathogenic mechanisms in HBV- and HCV-associated hepatocellular carcinoma. Nat. Rev. Cancer 2013, 13, 123–135. [Google Scholar] [CrossRef]
  61. Jin, Y.M.; Yun, C.; Park, C.; Wang, H.-J.; Cho, H. Expression of hepatitis B virus X protein is closely correlated with the high periportal inflammatory activity of liver diseases. J. Viral Hepat. 2001, 8, 322–330. [Google Scholar] [CrossRef]
  62. Terris, B.; Pineau, P.; Bregeaud, L.; Valla, D.; Belghiti, J.; Tiollais, P.; Degott, C.; Dejean, A. Close correlation between β-catenin gene alterations and nuclear accumulation of the protein in human hepatocellular carcinomas. Oncogene 1999, 18, 6583–6588. [Google Scholar] [CrossRef] [PubMed]
  63. Huang, H.; Fujii, H.; Sankila, A.; Mahler-Araujo, B.M.; Matsuda, M.; Cathomas, G.; Ohgaki, H. Beta-catenin mutations are frequent in human hepatocellular carcinomas associated with hepatitis C virus infection. Am. J. Pathol. 1999, 155, 1795–1801. [Google Scholar] [CrossRef]
  64. Zhao, L.-J.; Wang, L.; Ren, H.; Cao, J.; Li, L.; Ke, J.-S.; Qi, Z.-T. Hepatitis C virus E2 protein promotes human hepatoma cell proliferation through the MAPK/ERK signaling pathway via cellular receptors. Exp. Cell Res. 2005, 305, 23–32. [Google Scholar] [CrossRef] [PubMed]
  65. Pfaender, S.; Brown, R.J.; Pietschmann, T.; Steinmann, E. Natural reservoirs for homologs of hepatitis C virus. Emerg. Microbes Infect. 2014, 3, e21. [Google Scholar] [CrossRef] [PubMed]
  66. Washburn, M.L.; Bility, M.T.; Zhang, L.; Kovalev, G.I.; Buntzman, A.; Frelinger, J.A.; Barry, W.; Ploss, A.; Rice, C.M.; Su, L. A humanized mouse model to study hepatitis C virus infection, immune response, and liver disease. Gastroenterology 2011, 140, 1334–1344. [Google Scholar] [CrossRef] [PubMed]
  67. Bility, M.T.; Zhang, L.; Washburn, M.L.; Curtis, T.A.; Kovalev, G.I.; Su, L. Generation of a humanized mouse model with both human immune system and liver cells to model hepatitis C virus infection and liver immunopathogenesis. Nat. Protoc. 2012, 7, 1608–1617. [Google Scholar] [CrossRef] [Green Version]
  68. Billerbeck, E.; Mommersteeg, M.C.; Shlomai, A.; Xiao, J.W.; Andrus, L.; Bhatta, A.; Vercauteren, K.; Michailidis, E.; Dorner, M.; Krishnan, A.; et al. Humanized mice efficiently engrafted with fetal hepatoblasts and syngeneic immune cells develop human monocytes and NK cells. J. Hepatol. 2016, 65, 334–343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Steenbergen, R.H.G.; Joyce, M.A.; Lund, G.; Lewis, J.; Chen, R.; Barsby, N.; Zhu, L.F.; Tyrrell, D.L.J.; Kneteman, N.M. Lipoprotein profiles in SCID/uPA mice transplanted with human hepatocytes become human-like and correlate with HCV infection success. Am. J. Physiol. Gastrointest. Liver Physiol. 2010, 299, G844–G854. [Google Scholar] [CrossRef] [Green Version]
  70. Uchida, T.; Imamura, M.; Kan, H.; Hiraga, N.; Hayes, C.N.; Tsuge, M.; Abe-Chayama, H.; Aikata, H.; Makokha, G.N.; Miki, D.; et al. Usefulness of humanized cDNA-uPA/SCID mice for the study of hepatitis B virus and hepatitis C virus virology. J. Gen. Virol. 2017, 98, 1040–1047. [Google Scholar] [CrossRef]
  71. Tesfaye, A.; Stift, J.; Maric, D.; Cui, Q.; Dienes, H.-P.; Feinstone, S.M. Chimeric mouse model for the infection of hepatitis B and C viruses. PLoS ONE 2013, 8, e77298. [Google Scholar] [CrossRef]
  72. Carpentier, A.; Tesfaye, A.; Chu, V.; Nimgaonkar, I.; Zhang, F.; Lee, S.B.; Thorgeirsson, S.S.; Feinstone, S.M.; Liang, T.J. Engrafted human stem cell-derived hepatocytes establish an infectious HCV murine model. J. Clin. Investig. 2014, 124, 4953–4964. [Google Scholar] [CrossRef] [PubMed]
  73. Azuma, H.; Paulk, N.; Ranade, A.; Dorrell, C.; Al-Dhalimy, M.; Ellis, E.; Strom, S.; Kay, M.A.; Finegold, M.; Grompe, M. Robust expansion of human hepatocytes in Fah-/-/Rag2-/-/Il2rg-/- mice. Nat. Biotechnol. 2007, 25, 903–910. [Google Scholar] [CrossRef] [PubMed]
  74. Bissig, K.-D.; Wieland, S.F.; Tran, P.; Isogawa, M.; Le, T.T.; Chisari, F.V.; Verma, I.M. Human liver chimeric mice provide a model for hepatitis B and C virus infection and treatment. J. Clin. Investig. 2010, 120, 924–930. [Google Scholar] [CrossRef] [PubMed]
  75. Dagur, R.S.; Wang, W.; Cheng, Y.; Makarov, E.; Ganesan, M.; Suemizu, H.; Gebhart, C.L.; Gorantla, S.; Osna, N.; Poluektova, L.Y. Human hepatocyte depletion in the presence of HIV-1 infection in dual reconstituted humanized mice. Biol. Open 2018, 7, bio029785. [Google Scholar] [CrossRef] [PubMed]
  76. Kosaka, K.; Hiraga, N.; Imamura, M.; Yoshimi, S.; Murakami, E.; Nakahara, T.; Honda, Y.; Ono, A.; Kawaoka, T.; Tsuge, M.; et al. A novel TK-NOG based humanized mouse model for the study of HBV and HCV infections. Biochem. Biophys. Res. Commun. 2013, 441, 230–235. [Google Scholar] [CrossRef] [Green Version]
  77. Heckel, J.L.; Sandgren, E.P.; Degen, J.L.; Palmiter, R.D.; Brinster, R.L. Neonatal bleeding in transgenic mice expressing urokinase-type plasminogen activator. Cell 1990, 62, 447–456. [Google Scholar] [CrossRef]
  78. Sandgren, E.P.; Palmiter, R.D.; Heckel, J.L.; Daugherty, C.C.; Brinster, R.L.; Degen, J.L. Complete hepatic regeneration after somatic deletion of an albumin-plasminogen activator transgene. Cell 1991, 66, 245–256. [Google Scholar] [CrossRef]
  79. Rhim, J.A.; Sandgren, E.P.; Degen, J.L.; Palmiter, R.D.; Brinster, R.L. Replacement of diseased mouse liver by hepatic cell transplantation. Science 1994, 263, 1149–1152. [Google Scholar] [CrossRef]
  80. Mercer, D.F.; Schiller, D.E.; Elliott, J.F.; Douglas, D.N.; Hao, C.; Rinfret, A.; Addison, W.R.; Fischer, K.P.; Churchill, T.A.; Lakey, J.R.T.; et al. Hepatitis C virus replication in mice with chimeric human livers. Nat. Med. 2001, 7, 927–933. [Google Scholar] [CrossRef]
  81. Meuleman, P.; Hesselgesser, J.; Paulson, M.; Vanwolleghem, T.; Desombere, I.; Reiser, H.; Leroux-Roels, G. Anti-CD81 antibodies can prevent a hepatitis C virus infection in vivo. Hepatology 2008, 48, 1761–1768. [Google Scholar] [CrossRef]
  82. Uchida, T.; Hiraga, N.; Imamura, M.; Yoshimi, S.; Kan, H.; Miyaki, E.; Tsuge, M.; Abe, H.; Hayes, C.N.; Aikata, H.; et al. Elimination of HCV via a non-ISG-mediated mechanism by vaniprevir and BMS-788329 combination therapy in human hepatocyte chimeric mice. Virus Res. 2016, 213, 62–68. [Google Scholar] [CrossRef] [PubMed]
  83. Meuleman, P.; Leroux-Roels, G. The human liver-uPA-SCID mouse: A model for the evaluation of antiviral compounds against HBV and HCV. Antivir. Res. 2008, 80, 231–238. [Google Scholar] [CrossRef] [PubMed]
  84. Vanwolleghem, T.; Bukh, J.; Meuleman, P.; Desombere, I.; Meunier, J.-C.; Alter, H.; Purcell, R.H.; Leroux-Roels, G. Polyclonal immunoglobulins from a chronic hepatitis C virus patient protect human liver–chimeric mice from infection with a homologous hepatitis C virus strain. Hepatology 2008, 47, 1846–1855. [Google Scholar] [CrossRef] [PubMed]
  85. Kneteman, N.M.; Weiner, A.J.; O’Connell, J.; Collett, M.; Gao, T.; Aukerman, L.; Kovelsky, R.; Ni, Z.-J.; Hashash, A.; Kline, J.; et al. Anti-HCV therapies in chimeric scid-Alb/uPA mice parallel outcomes in human clinical application. Hepatology 2006, 43, 1346–1353. [Google Scholar] [CrossRef] [PubMed]
  86. Joyce, M.A.; Walters, K.-A.; Lamb, S.-E.; Yeh, M.M.; Zhu, L.-F.; Kneteman, N.; Doyle, J.S.; Katze, M.G.; Tyrrell, D.L. HCV induces oxidative and ER stress, and sensitizes infected cells to apoptosis in SCID/Alb-uPA mice. PLoS Pathog. 2009, 5, e1000291. [Google Scholar] [CrossRef] [PubMed]
  87. Brown, R.J.P.; Hudson, N.; Wilson, G.; Rehman, S.U.; Jabbari, S.; Hu, K.; Tarr, A.W.; Borrow, P.; Joyce, M.; Lewis, J.; et al. Hepatitis C virus envelope glycoprotein fitness defines virus population composition following transmission to a new host. J. Virol. 2012, 86, 11956–11966. [Google Scholar] [CrossRef] [PubMed]
  88. Vanwolleghem, T.; Meuleman, P.; Libbrecht, L.; Roskams, T.; de Vos, R.; Leroux–Roels, G. Ultra-rapid cardiotoxicity of the hepatitis C virus protease inhibitor BILN 2061 in the urokinase-type plasminogen activator mouse. Gastroenterology 2007, 133, 1144–1155. [Google Scholar] [CrossRef]
  89. Vercauteren, K.; Brown, R.J.P.; Mesalam, A.A.; Doerrbecker, J.; Bhuju, S.; Geffers, R.; van Den Eede, N.; McClure, C.P.; Troise, F.; Verhoye, L.; et al. Targeting a host-cell entry factor barricades antiviral-resistant HCV variants from on-therapy breakthrough in human-liver mice. Gut 2016, 65, 2029–2034. [Google Scholar] [CrossRef]
  90. Strom, S.C.; Davila, J.; Grompe, M. Chimeric mice with humanized liver: Tools for the study of drug metabolism, excretion, and toxicity. Methods Mol. Biol. 2010, 640, 491–509. [Google Scholar] [CrossRef]
  91. Tateno, C.; Kawase, Y.; Tobita, Y.; Hamamura, S.; Ohshita, H.; Yokomichi, H.; Sanada, H.; Kakuni, M.; Shiota, A.; Kojima, Y.; et al. Generation of novel chimeric mice with humanized livers by using hemizygous cDNA-uPA/SCID mice. PLoS ONE 2015, 10, e0142145. [Google Scholar] [CrossRef]
  92. Heo, J.; Factor, V.M.; Uren, T.; Takahama, Y.; Lee, J.-S.; Major, M.; Feinstone, S.M.; Thorgeirsson, S.S. Hepatic precursors derived from murine embryonic stem cells contribute to regeneration of injured liver. Hepatology 2006, 44, 1478–1486. [Google Scholar] [CrossRef] [PubMed]
  93. Suemizu, H.; Hasegawa, M.; Kawai, K.; Taniguchi, K.; Monnai, M.; Wakui, M.; Suematsu, M.; Ito, M.; Peltz, G.; Nakamura, M. Establishment of a humanized model of liver using NOD/Shi-scid IL2Rgnull mice. Biochem. Biophys. Res. Commun. 2008, 377, 248–252. [Google Scholar] [CrossRef] [PubMed]
  94. Bissig, K.-D.; Le, T.T.; Woods, N.-B.; Verma, I.M. Repopulation of adult and neonatal mice with human hepatocytes: A chimeric animal model. Proc. Natl. Acad. Sci. USA 2007, 104, 20507–20511. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Mailly, L.; Xiao, F.; Lupberger, J.; Wilson, G.K.; Aubert, P.; Duong, F.H.T.; Calabrese, D.; Leboeuf, C.; Fofana, I.; Thumann, C.; et al. Clearance of persistent hepatitis C virus infection in humanized mice using a claudin-1-targeting monoclonal antibody. Nat. Biotechnol. 2015, 33, 549–554. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Burchill, M.A.; Roby, J.A.; Crochet, N.; Wind-Rotolo, M.; Stone, A.E.; Edwards, M.G.; Dran, R.J.; Kriss, M.S.; Gale, M., Jr.; Rosen, H.R. Rapid reversal of innate immune dysregulation in blood of patients and livers of humanized mice with HCV following DAA therapy. PLoS ONE 2017, 12, e0186213. [Google Scholar] [CrossRef]
  97. Hasegawa, M.; Kawai, K.; Mitsui, T.; Taniguchi, K.; Monnai, M.; Wakui, M.; Ito, M.; Suematsu, M.; Peltz, G.; Nakamura, M.; et al. The reconstituted ‘humanized liver’ in TK-NOG mice is mature and functional. Biochem. Biophys. Res. Commun. 2011, 405, 405–410. [Google Scholar] [CrossRef] [PubMed]
  98. Vuyyuru, R.; Patton, J.; Manser, T. Human immune system mice: Current potential and limitations for translational research on human antibody responses. Immunol. Res. 2011, 51, 257–266. [Google Scholar] [CrossRef]
  99. Strowig, T.; Gurer, C.; Ploss, A.; Liu, Y.-F.; Arrey, F.; Sashihara, J.; Koo, G.; Rice, C.M.; Young, J.W.; Chadburn, A.; et al. Priming of protective T cell responses against virus-induced tumors in mice with human immune system components. J. Exp. Med. 2009, 206, 1423–1434. [Google Scholar] [CrossRef] [Green Version]
  100. Jaiswal, S.; Pearson, T.; Friberg, H.; Shultz, L.D.; Greiner, D.L.; Rothman, A.L.; Mathew, A. Dengue virus infection and virus-specific HLA-A2 restricted immune responses in humanized NOD-scid IL2rγnull mice. PLoS ONE 2009, 4, e7251. [Google Scholar] [CrossRef]
  101. Woods, A.; Chen, H.Y.; Trumbauer, M.E.; Sirotina, A.; Cummings, R.; Zaller, D.M. Human major histocompatibility complex class II-restricted T cell responses in transgenic mice. J. Exp. Med. 1994, 180, 173–181. [Google Scholar] [CrossRef]
  102. Camacho, R.E.; Wnek, R.; Shah, K.; Zaller, D.M.; O’Reilly, R.J.; Collins, N.; Fitzgerald-Bocarsly, P.; Koo, G.C. Intra-thymic/splenic engraftment of human T cells in HLA-DR1 transgenic NOD/scid mice. Cell. Immunol. 2004, 232, 86–95. [Google Scholar] [CrossRef] [PubMed]
  103. Camacho, R.E.; Wnek, R.; Fischer, P.; Shah, K.; Zaller, D.M.; Woods, A.; La Monica, N.; Aurisicchio, L.; Fitzgerald-Bocarsly, P.; Koo, G.C. Characterization of the NOD/scid-[Tg]DR1 mouse expressing HLA-DRB1*01 transgene: A model of SCID-hu mouse for vaccine development. Exp. Hematol. 2007, 35, 1219–1230. [Google Scholar] [CrossRef] [PubMed]
  104. Chen, Q.; Khoury, M.; Limmon, G.; Choolani, M.; Chan, J.K.Y.; Chen, J. Human fetal hepatic progenitor cells are distinct from, but closely related to, hematopoietic stem/progenitor cells. Stem Cells 2013, 31, 1160–1169. [Google Scholar] [CrossRef] [PubMed]
  105. Yong, K.S.M.; Her, Z.; Chen, Q. Humanized mice as unique tools for human-specific studies. Arch. Immunol. Ther. Exp. 2018, 66, 245–266. [Google Scholar] [CrossRef]
  106. Yong, K.S.M.; Keng, C.T.; Tan, S.Q.; Loh, E.; Chang, K.T.; Tan, T.C.; Hong, W.; Chen, Q. Human CD34loCD133lo fetal liver cells support the expansion of human CD34hiCD133hi hematopoietic stem cells. Cell. Mol. Immunol. 2016, 13, 605–614. [Google Scholar] [CrossRef] [PubMed]
  107. Accapezzato, D.; Francavilla, V.; Paroli, M.; Casciaro, M.; Chircu, L.V.; Cividini, A.; Abrignani, S.; Mondelli, M.U.; Barnaba, V. Hepatic expansion of a virus-specific regulatory CD8+ T cell population in chronic hepatitis C virus infection. J. Clin. Investig. 2004, 113, 963–972. [Google Scholar] [CrossRef] [PubMed]
  108. Bertoletti, A.; D’Elios, M.; Boni, C.; de Carli, M.; Zignego, A.; Durazzo, M.; Missale, G.; Penna, A.; Fiaccadori, F.; Prete, G.; et al. Different Cytokine Profiles of Intrahepatic T Cells in Chronic Hepatitis B and Hepatitis C Virus Infections. Gastroenterology 1997, 112, 193–199. [Google Scholar] [CrossRef]
  109. Dolganiuc, A.; Chang, S.; Kodys, K.; Mandrekar, P.; Bakis, G.; Cormier, M.; Szabo, G. Hepatitis C virus (HCV) core protein-induced, monocyte-mediated mechanisms of reduced IFN-α and plasmacytoid dendritic cell loss in chronic HCV infection. J. Immunol. 2006, 177, 6758–6768. [Google Scholar] [CrossRef]
  110. Revie, D.; Salahuddin, S.Z. Role of macrophages and monocytes in hepatitis C virus infections. World J. Gastroenterol. 2014, 20, 2777–2784. [Google Scholar] [CrossRef]
  111. Zeremski, M.; Petrovic, L.M.; Talal, A.H. The role of chemokines as inflammatory mediators in chronic hepatitis C virus infection. J. Viral Hepat. 2007, 14, 675–687. [Google Scholar] [CrossRef]
  112. McGuinness, P.H.; Painter, D.; Davies, S.; McCaughan, G.W. Increases in intrahepatic CD68 positive cells, MAC387 positive cells, and proinflammatory cytokines (particularly interleukin 18) in chronic hepatitis C infection. Gut 2000, 46, 260–269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Meuleman, P.; Catanese, M.T.; Verhoye, L.; Desombere, I.; Farhoudi, A.; Jones, C.T.; Sheahan, T.; Grzyb, K.; Cortese, R.; Rice, C.M.; et al. A human monoclonal antibody targeting scavenger receptor class B type I precludes hepatitis C virus infection and viral spread in vitro and in vivo. Hepatology 2012, 55, 364–372. [Google Scholar] [CrossRef] [PubMed]
  114. Law, M.; Maruyama, T.; Lewis, J.; Giang, E.; Tarr, A.W.; Stamataki, Z.; Gastaminza, P.; Chisari, F.V.; Jones, I.M.; Fox, R.I.; et al. Broadly neutralizing antibodies protect against hepatitis C virus quasispecies challenge. Nat. Med. 2007, 14, 25–27. [Google Scholar] [CrossRef] [PubMed]
  115. Shindo, M.; di Bisceglie, A.M.; Cheung, L.; Shih, J.W.-K.; Cristiano, K.; Feinstone, S.M.; Hoofnagle, J.H. Decrease in serum hepatitis C vral RNA during apha-interferon therapy for chronic hepatitis C. Ann. Intern. Med. 1991, 115, 700–704. [Google Scholar] [CrossRef] [PubMed]
  116. Di Bisceglie, A.M.; Martin, P.; Kassianides, C.; Lisker-Melman, M.; Murray, L.; Waggoner, J.; Goodman, Z.; Banks, S.M.; Hoofnagle, J.H. Recombinant interferon alfa therapy for chronic hepatitis C. N. Engl. J. Med. 1989, 321, 1506–1510. [Google Scholar] [CrossRef] [PubMed]
  117. Davis, G.L.; Balart, L.A.; Schiff, E.R.; Lindsay, K.; Bodenheimer, H.C.; Perrillo, R.P.; Carey, W.; Jacobson, I.M.; Payne, J.; Dienstag, J.L.; et al. Treatment of chronic hepatitis C with recombinant interferon alfa. N. Engl. J. Med. 1989, 321, 1501–1506. [Google Scholar] [CrossRef] [PubMed]
  118. Marcellin, P.; Boyer, N.; Giostra, E.; Degott, C.; Courouce, A.M.; Degos, F.; Coppere, H.; Cales, P.; Couzigou, P.; Benhamou, J.-P. Recombinant human α-interferon in patients with chronic non-A, non-B hepatitis: A multicenter randomized controlled trial from France. Hepatology 1991, 13, 393–397. [Google Scholar] [CrossRef]
  119. Kohli, A.; Shaffer, A.; Sherman, A.; Kottilil, S. Treatment of hepatitis C: A systematic review treatment of hepatitis C treatment. JAMA 2014, 312, 631–640. [Google Scholar] [CrossRef]
  120. Schalm, S.W.; Hansen, B.E.; Chemello, L.; Bellobuono, A.; Brouwer, J.T.; Weiland, O.; Cavalletto, L.; Schvarcz, R.; Ideo, G.; Alberti, A. Ribavirin enhances the efficacy but not the adverse effects of interferon in chronic hepatitis C: Meta-analysis of individual patient data from European centers. J. Hepatol 1997, 26, 961–966. [Google Scholar] [CrossRef]
  121. Evans, M.J.; von Hahn, T.; Tscherne, D.M.; Syder, A.J.; Panis, M.; Wölk, B.; Hatziioannou, T.; McKeating, J.A.; Bieniasz, P.D.; Rice, C.M. Claudin-1 is a hepatitis C virus co-receptor required for a late step in entry. Nature 2007, 446, 801–805. [Google Scholar] [CrossRef]
  122. Kumar, H.; Kawai, T.; Akira, S. Pathogen recognition by the innate immune system. Int. Rev. Immunol. 2011, 30, 16–34. [Google Scholar] [CrossRef] [PubMed]
  123. Akira, S. Innate immunity and adjuvants. Philos. Trans. R Soc. Lond. B Biol. Sci. 2011, 366, 2748–2755. [Google Scholar] [CrossRef] [PubMed]
  124. Nakagawa, S.-I.; Hirata, Y.; Kameyama, T.; Tokunaga, Y.; Nishito, Y.; Hirabayashi, K.; Yano, J.; Ochiya, T.; Tateno, C.; Tanaka, Y.; et al. Targeted induction of interferon-λ in humanized chimeric mouse liver abrogates hepatotropic virus infection. PLoS ONE 2013, 8, e59611. [Google Scholar] [CrossRef] [PubMed]
  125. Sarrazin, C.; Zeuzem, S. Resistance to direct antiviral agents in patients with hepatitis C virus infection. Gastroenterology 2010, 138, 447–462. [Google Scholar] [CrossRef] [PubMed]
  126. Katsume, A.; Tokunaga, Y.; Hirata, Y.; Munakata, T.; Saito, M.; Hayashi, H.; Okamoto, K.; Ohmori, Y.; Kusanagi, I.; Fujiwara, S.; et al. A serine palmitoyltransferase inhibitor blocks hepatitis C virus replication in human hepatocytes. Gastroenterology 2013, 145, 865–873. [Google Scholar] [CrossRef] [PubMed]
  127. Wong, M.-T.; Chen, S.S.L. Emerging roles of interferon-stimulated genes in the innate immune response to hepatitis C virus infection. Cell. Mol. Immunol. 2016, 13, 11–35. [Google Scholar] [CrossRef]
  128. Pawlotsky, J.M.; Chevaliez, S.; McHutchison, J.G. The hepatitis C virus life cycle as a target for new antiviral therapies. Gastroenterology 2007, 132, 1979–1998. [Google Scholar] [CrossRef]
  129. De Andrade, V.G.; Yamashiroy, F.d.S.; Oliveira, C.V.; Moreira, A.; Winckler, F.C.; Silva, G.F. Insulin resistance reduction after sustained virological response with direct acting antiviral: Not every population improves. Arq. Gastroenterol 2018, 55, 274–278. [Google Scholar] [CrossRef]
  130. Hézode, C.; Forestier, N.; Dusheiko, G.; Ferenci, P.; Pol, S.; Goeser, T.; Bronowicki, J.-P.; Bourlière, M.; Gharakhanian, S.; Bengtsson, L.; et al. Telaprevir and Peginterferon with or without Ribavirin for chronic HCV infection. N. Engl. J. Med. 2009, 360, 1839–1850. [Google Scholar] [CrossRef]
  131. Li, X.-D.; Sun, L.; Seth, R.B.; Pineda, G.; Chen, Z.J. Hepatitis C virus protease NS3/4A cleaves mitochondrial antiviral signaling protein off the mitochondria to evade innate immunity. Proc. Natl. Acad. Sci. USA 2005, 102, 17717–17722. [Google Scholar] [CrossRef] [Green Version]
  132. Fried, M.W.; Shiffman, M.L.; Reddy, K.R.; Smith, C.; Marinos, G.; Gonçales, F.L.; Häussinger, D.; Diago, M.; Carosi, G.; Dhumeaux, D.; et al. Peginterferon alfa-2a plus Ribavirin for chronic hpatitis C virus infection. N. Engl. J. Med. 2002, 347, 975–982. [Google Scholar] [CrossRef] [PubMed]
  133. Kumthip, K.; Maneekarn, N. The role of HCV proteins on treatment outcomes. Virol. J. 2015, 12, 217. [Google Scholar] [CrossRef] [PubMed]
  134. Naderi, M.; Gholipour, N.; Zolfaghari, M.R.; Moradi Binabaj, M.; Yegane Moghadam, A.; Motalleb, G. Hepatitis C virus and vaccine development. Int. J. Mol. Cell. Med. 2014, 3, 207–215. [Google Scholar] [PubMed]
  135. Lin, Y.; Kwon, T.; Polo, J.; Zhu, Y.-F.; Coates, S.; Crawford, K.; Dong, C.; Wininger, M.; Hall, J.; Selby, M.; et al. Induction of broad CD4+ and CD8+ T-cell responses and cross-neutralizing antibodies against hepatitis C virus by vaccination with Th1-adjuvanted polypeptides followed by defective alphaviral particles expressing envelope glycoproteins gpE1 and gpE2 and nonstructural proteins 3, 4, and 5. J. Virol. 2008, 82, 7492–7503. [Google Scholar] [CrossRef]
  136. Wang, Y.; Keck, Z.-Y.; Foung, S.K.H. Neutralizing antibody response to hepatitis C virus. Viruses 2011, 3, 2127–2145. [Google Scholar] [CrossRef] [PubMed]
  137. Pierce, B.G.; Boucher, E.N.; Piepenbrink, K.H.; Ejemel, M.; Rapp, C.A.; Thomas, W.D.; Sundberg, E.J.; Weng, Z.; Wang, Y. Structure-based design of hepatitis C virus vaccines that elicit neutralizing antibody responses to a conserved epitope. J. Virol. 2017, 91, e01032-17. [Google Scholar] [CrossRef] [PubMed]
  138. Huang, Y.; Li, M.-H.; Hou, M.; Xie, Y. Peginterferon alfa-2a for the treatment of chronic hepatitis C in the era of direct-acting antivirals. Hepatobiliary Pancreat. Dis. Int. 2017, 16, 470–479. [Google Scholar] [CrossRef]
  139. Bruening, J.; Weigel, B.; Gerold, G. The role of type III interferons in hepatitis C virus infection and therapy. J. Immunol. Res. 2017, 2017, 7232361. [Google Scholar] [CrossRef]
  140. Colpitts, C.C.; Tawar, R.G.; Mailly, L.; Thumann, C.; Heydmann, L.; Durand, S.C.; Xiao, F.; Robinet, E.; Pessaux, P.; Zeisel, M.B.; et al. Humanisation of a claudin-1-specific monoclonal antibody for clinical prevention and cure of HCV infection without escape. Gut 2018, 67, 736–745. [Google Scholar] [CrossRef]
  141. Gandhi, S.; Khubchandani, S.; Iyer, R. Quality of life and hepatocellular carcinoma. J. Gastrointest. Oncol. 2014, 5, 296–317. [Google Scholar] [CrossRef]
  142. Righi, E.; Londero, A.; Carnelutti, A.; Baccarani, U.; Bassetti, M. Impact of new treatment options for hepatitis C virus infection in liver transplantation. World J. Gastroenterol. 2015, 21, 10760–10775. [Google Scholar] [CrossRef] [PubMed]
  143. El-Khoueiry, A.B.; Sangro, B.; Yau, T.; Crocenzi, T.S.; Kudo, M.; Hsu, C.; Kim, T.-Y.; Choo, S.-P.; Trojan, J.; Welling, T.H., 3rd.; et al. Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): An open-label, non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 2017, 389, 2492–2502. [Google Scholar] [CrossRef]
  144. Zhu, A.X.; Finn, R.S.; Edeline, J.; Cattan, S.; Ogasawara, S.; Palmer, D.; Verslype, C.; Zagonel, V.; Fartoux, L.; Vogel, A.; et al. Pembrolizumab in patients with advanced hepatocellular carcinoma previously treated with sorafenib (KEYNOTE-224): A non-randomised, open-label phase 2 trial. Lancet Oncol. 2018, 19, 940–952. [Google Scholar] [CrossRef]
  145. Sangro, B.; Gomez-Martin, C.; de la Mata, M.; Iñarrairaegui, M.; Garralda, E.; Barrera, P.; Riezu-Boj, J.I.; Larrea, E.; Alfaro, C.; Sarobe, P.; et al. A clinical trial of CTLA-4 blockade with tremelimumab in patients with hepatocellular carcinoma and chronic hepatitis C. J. Hepatol. 2013, 59, 81–88. [Google Scholar] [CrossRef] [PubMed]
  146. Wainberg, Z.A.; Segal, N.H.; Jaeger, D.; Lee, K.-H.; Marshall, J.; Antonia, S.J.; Butler, M.; Sanborn, R.E.; Nemunaitis, J.J.; Carlson, C.A.; et al. Safety and clinical activity of durvalumab monotherapy in patients with hepatocellular carcinoma (HCC). J. Clin. Oncol. 2017, 35, 4071. [Google Scholar] [CrossRef]
  147. Chen, Q.; He, F.; Kwang, J.; Chan, J.K.Y.; Chen, J. GM-CSF and IL-4 stimulate antibody responses in humanized mice by promoting T, B, and dendritic cell maturation. J. Immunol. 2012, 189, 5223–5229. [Google Scholar] [CrossRef] [PubMed]
  148. Chen, Q.; Khoury, M.; Chen, J. Expression of human cytokines dramatically improves reconstitution of specific human-blood lineage cells in humanized mice. Proc. Natl. Acad. Sci. USA 2009, 106, 21783–21788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Chen, Q.; Ye, W.; Jian Tan, W.; Mei Yong, K.S.; Liu, M.; Qi Tan, S.; Loh, E.; Te Chang, K.; Chye Tan, T.; Preiser, P.R.; et al. Delineation of natural killer cell differentiation from myeloid progenitors in human. Sci. Rep. 2015, 5, 15118. [Google Scholar] [CrossRef]
  150. Aizawa, Y.; Seki, N.; Nagano, T.; Abe, H. Chronic hepatitis C virus infection and lipoprotein metabolism. World J. Gastroenterol. 2015, 21, 10299–10313. [Google Scholar] [CrossRef]
  151. Kaplan, D.E.; Sugimoto, K.; Newton, K.; Valiga, M.E.; Ikeda, F.; Aytaman, A.; Nunes, F.A.; Lucey, M.R.; Vance, B.A.; Vonderheide, R.H.; et al. Discordant role of CD4 T-cell response relative to neutralizing antibody and CD8 T-cell responses in acute hepatitis C. Gastroenterology 2007, 132, 654–666. [Google Scholar] [CrossRef]
  152. Knolle, P.A.; Wohlleber, D. Immunological functions of liver sinusoidal endothelial cells. Cell. Mol. Immunol. 2016, 13, 347–353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  153. Taoka, K.; Arai, S.; Kataoka, K.; Hosoi, M.; Miyauchi, M.; Yamazaki, S.; Honda, A.; Aixinjueluo, W.; Kobayashi, T.; Kumano, K.; et al. Using patient-derived iPSCs to develop humanized mouse models for chronic myelomonocytic leukemia and therapeutic drug identification, including liposomal clodronate. Sci. Rep. 2018, 8, 15855. [Google Scholar] [CrossRef] [PubMed]
Table 1. Chimeric human liver mouse models (Part I).
Table 1. Chimeric human liver mouse models (Part I).
NameAlb-uPA/SCIDcDNA-uPA/SCIDMUP-uPA/SCID/Bg
Nomenclature---
Engraftment method for humanization of immune system---
Engraftment method for humanization of liverIntrasplenic injectionIntrasplenic injectionIntrasplenic injection
Source of cellsHuman hepatocyteHuman hepatocyteHuman hepatocyte
Presence of human hepatocytesYesYesYes
Presence of human immune systemNoNoNo
Method of HCV infectionIntraperitoneal injectionIntravenous injectionIntravenous injection
Strain of HCV usedPatient serum containing HCV genotype 1aSerum/culture medium of HCV (105 copies)Diluted plasma from HCV-infected chimpanzee
Duration monitored post-HCV infectionUp to 10 weeksUp to 8 weeksUp to 8 weeks
Advantages
  • Presence of mature human hepatocytes
  • This model is able to recapitulate the human immune system more efficiently than mouse models without humanization
  • Higher human hepatocytes and HCV viraemia levels as compared to TK-NOG
  • Useful for evaluation antiviral agents
  • Capable of supporting long-term HCV infection
  • The cDNA with albumin promoter/enhancer and uPA demonstrate no loss of uPA due to the deletion of transgene
  • Few renal disorders
  • High body weight
  • High survival rate
  • Presence of mature human hepatocytes
  • Higher hepatocyte reconstitution as compared to Alb-uPA/SCID mice
  • Higher concentration of serum albumin as compared to Alb-uPA/SCID mice
  • High and persistent titers of viremia
  • Capable of supporting long-term HCV infection
  • Easy to maintain colony of transgenic mice
  • High survival rate
  • Less technically challenging to engraft hepatocytes into mice, as there is a long window to engraft mice (4-12 months)
  • Each major HCV genotype was infectious in MUP-uPA/SCID mice
  • Capable of supporting long-term HCV infection
Drawbacks
  • Poor breeding efficiency
  • Short window for engraftment
  • Absence of human immune system
  • Liver environment is unsuitable for the engraftment of fetal-liver derived cells
  • Unable to reproduce pathological outcomes of HCV
  • Impossible to study HCV immunopathogenesis
  • No liver disease
  • Low level of hepatocyte reconstitution
  • Low serum levels of human albumin and HCV viremia
  • Repopulation of the liver with human cells may be cause by cell fusion
  • No liver disease
  • High mortality rate
  • Low body weight
  • High renal disorders
  • Absence of human immune system
  • Unable to reproduce pathological outcomes of HCV
  • Liver environment is unsuitable for the engraftment of fetal-liver derived cells
  • Impossible to study HCV immunopathogenesis
  • No liver disease
  • Absence of human immune system
  • Variable viral replication observed in mice
  • Unable to reproduce pathological outcomes of HCV
  • Liver environment is unsuitable for the engraftment of fetal-liver derived cells
  • Impossible to study HCV immunopathogenesis
  • No liver disease
References
  • Washburn et al. (2011) [66]
  • Steenbergen et al. (2010) [69]
  • Uchida et al. (2017) [70]
  • Tesfaye et al. (2017) [71]
  • Carpentier et al. (2014) [72]
Abbreviations: cDNA: Complementary DNA, Fah: Fumarylacetoacetate hydrolase, HCV: Hepatitis C virus, NTBC: 2-(2-nitro-4-trifluoro-methylbenzoyl)1,3-cyclohexedione.
Table 2. Chimeric human liver mouse models (Part II).
Table 2. Chimeric human liver mouse models (Part II).
NameFRGTK-NOG
NomenclatureFah−/−Rag2−/−γC−/−NOD.Cg-Prkdcscid Il2rgtm1Sug Tg(Alb-TK)7-2/ShiJic
Engraftment method for humanization of immune system--
Engraftment method for humanization of liverIntrahepatic injectionIntrasplenic injection of human hepatocytes
Source of cellsHuman hepatocytesHuman hepatocytes
Presence of human hepatocytesYesYes
Presence of human immune systemNoNo
Method of HCV infectionIntravenous injectionIntravenous injection
Strain of HCV used
  • Supernatant of Huh-7 cell culture containing 2 × 104 ffu JFH-1, 3 × 103 ffu HCV Con1/C3, 3 × 103 ffu HCV H77/C3
  • Patient serum containing 2 × 105 GE/ml HCV genotype 1a
  • Patient serum containing 2 × 105 IU/ml HCV genotype 3a
  • Patient serum containing HCV genotype 1b (2.2 x 106 copies/mL)
Duration monitored post-HCV infectionUp to 5 weeksUp to 8 weeks
Advantages
  • Simplified animal husbandry and surgery as liver disease can be controlled by NTBC
  • Mice are genetically stable
  • Pharmacological interference not needed in reconstituting FRG mice with hepatocytes
  • Can be serially engrafted with human hepatocytes
  • No renal disorders
  • High survival rate
  • Capable of supporting long-term HCV infection
  • Able to achieve endogenous liver injury and human hepatocyte engraftment
  • Cost effective as compared to uPA/SCID and FRG mice
  • Capable of supporting long-term HCV infection
Drawbacks
  • Requires maintenance under constant and costly NTBC treatment
  • Absence of human immune system
  • Primary engraftment does not occur in all recipient mice
  • Able to achieve high human hepatocyte reconstitution with only human adult liver cells
  • Unable to reproduce pathological outcomes of HCV
  • Absence of human immune system
  • Liver environment is unsuitable for the engraftment of fetal-liver derived cells
  • Unable to reproduce pathological outcomes of HCV
  • High mortality rate
  • Low body weight
  • High renal disorders
References
  • Washburn et al. (2011) [66]
  • Azuma et al. (2007) [73]
  • Bissig et al. (2010) [74]
  • Dagur et al. (2018) [75]
  • Kosaka et al. (2013) [76]
Table 3. Humanized mouse models with both human immune system and hepatocytes.
Table 3. Humanized mouse models with both human immune system and hepatocytes.
NameAFC8-hu HSC/HepNSG-DRB*0101
NomenclatureAFC8-HSC/Hep Balb/C Rag2−/−γC−/−NOD/scid-DRB*0101
Engraftment method for humanization of immune systemIntrahepatic injectionIntrahepatic injection
Engraftment method for humanization of liverIntrahepatic injectionIntrahepatic injection
Source of cellsHuman adult cellsHuman fetal liver
Presence of human hepatocytesYesYes
Presence of human immune systemYesYes
Method of HCV infectionIntravenous injectionIntravenous injection
Strain of HCV usedClinical isolate of HCV genotype 1a (1-5 x 107 genome copies/mL)Recombinant adenovirus serotype 5 (AdV5) (5 x 109 or 1010 particles)
Duration monitored post-HCV infectionUp to 20 weeksUp to 4 weeks
Advantages
  • The use of caspase 8-dependent induction of mouse hepatocyte apoptosis to promote human hepatocyte repopulation is less toxic as compared with uPA/SCID and FAH mice
  • Presence of both human immune system and hepatocytes
  • AFC8-hu HSC/Hep mice infected with HCV generates human immune responses, elevated levels of alanine aminotransferase (ALT), liver inflammation, hepatitis and fibrosis
  • Suitable for the study of hepatitis virus-induced liver immunopathogenesis
  • HCV genomic RNA is detectable in the livers of mice
  • Only small animal model capable to support the co-infection of HCV and HIV
  • Useful platform for the evaluation of antiviral drugs and immunotherapies
  • Presence of both human immune system and hepatocytes
  • Transgenic HLA expression improves human antiviral HLA-restricted T cell responses during human viral infections
  • Suitable for the study of hepatitis virus-induced liver immunopathogenesis
Drawbacks
  • Liver sinusoidal endothelium is of mouse origin
  • Low level of repopulation and immature phenotype of human hepatocytes
  • Unable to detect significant HCV viremia in the blood
  • Low serum levels of human albumin and HCV viremia
  • Cannot be used for long-term studies
  • Antiviral immune responses may not be as robust as in human patients
  • Does not fully recapitulate clinical settings
  • To analyze HCV immunopathogenesis, mice must be engrafted with both donor matched human hepatocytes and immune cells, hence making this a challenging model to establish
  • Liver sinusoidal endothelium is of mouse origin
  • Lack of complete viral clearance from the liver
  • Does not fully recapitulate clinical settings
References
  • Washburn et al. (2011) [66]
  • Bility et al. (2012) [67]
  • Billerbeck et al. (2013) [68]
Table 4. Humanized mouse models with both human immune system and hepatocytes (HIL mice).
Table 4. Humanized mouse models with both human immune system and hepatocytes (HIL mice).
NameNSGNSGNSG
NomenclatureNOD-scid Il2rg−/−NOD-scid Il2rg−/−NOD-scid Il2rg−/−
Engraftment method for humanization of immune systemIntrahepatic injectionIntrahepatic injectionIntrahepatic injection
Engraftment method for humanization of liverIntrahepatic injectionIntrahepatic injectionIntrahepatic injection
Source of cellsHLA type I matched fetal liverFetal liverFetal liver
Presence of human hepatocytesYesYesYes
Presence of human immune systemYesYesYes
Method of HCV infectionIntravenousIntravenousIntravenous
Strain of HCV usedHCV induced HCC patient derived xenograft106 FFU/mL of J6/JFH-1 HCV (genotype 2a) viruses106 or 107 ffu of HCVJ6/JFH1-P47
Duration monitored post-HCV infectionUp to 8 weeksUp to 9 weeksUp to 28 weeks
Advantages
  • Presence of both human immune system and hepatocytes
  • Liver inflammation and fibrosis are observed
  • Able to study HCV immunopathogenesis
  • Useful platform for therapeutic testing
  • Side effects of immunotherapies tested on this mouse model were similar to clinical settings
  • Presence of both human immune system and hepatocytes
  • Liver inflammation and fibrosis are observed
  • Mice are able to support HCV infection and demonstrate some clinical symptoms found in HCV-infected patients including hepatitis, robust virus-specific human immune cell and cytokine response as well as liver fibrosis and cirrhosis
  • Useful platform for therapeutic testing such as antiviral treatment, PEG-IFNα-2A
  • Presence of both human immune system and hepatocytes
  • HCV infected mice developed increase incidences of liver fibrosis, granulomatous inflammation and tumour formation in the form of hepatocellular adenomas/carcinomas by 28-weeks post-infection as compared to uninfected mice
  • Mice can recapitulate some clinical symptoms, such as, chronic inflammation, immune cell exhaustion and tumorigenesis as observed in patients
  • HCV infection is able to progress beyond 27-weeks in this model
  • Liver inflammation and fibrosis are observed
  • Able to study HCV immunopathogenesis
  • Useful platform for therapeutic testing
Drawbacks
  • Does not fully recapitulate HCV responses as observed in patients
  • Antiviral immune responses not as robust as observed in clinical settings
  • Does not fully recapitulate HCV responses as observed in patients
  • Antiviral immune responses not as robust as observed in clinical settings
  • Does not fully recapitulate HCV responses as observed in patients
  • Antiviral immune responses not as robust as observed in clinical settings
  • Effects of HCV in mice needs to be monitored for a longer duration of time to confirm if liver tumorigenesis can occur
  • Despite observation of chronic hepatitis, viremia was not detectable in plasma/liver of infected mice
  • HCV RNA was challenging to detect with non-sacrificial sampling methods
  • HCV RNA could only be detected after extracting RNA from purified human hepatocytes in infected HIL mice, although this renders the liver tissue unusable for histopathological analyses
References Zhao et al. (2018) [21] Keng et al. (2015) [18] Zheng et al. (2017) [19]

Share and Cite

MDPI and ACS Style

Yong, K.S.M.; Her, Z.; Chen, Q. Humanized Mouse Models for the Study of Hepatitis C and Host Interactions. Cells 2019, 8, 604. https://doi.org/10.3390/cells8060604

AMA Style

Yong KSM, Her Z, Chen Q. Humanized Mouse Models for the Study of Hepatitis C and Host Interactions. Cells. 2019; 8(6):604. https://doi.org/10.3390/cells8060604

Chicago/Turabian Style

Yong, Kylie Su Mei, Zhisheng Her, and Qingfeng Chen. 2019. "Humanized Mouse Models for the Study of Hepatitis C and Host Interactions" Cells 8, no. 6: 604. https://doi.org/10.3390/cells8060604

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop