1 Summary
The development and evaluation of effective therapies and vaccines for the hepatitis C virus (HCV) and the study of its interactions with the mammalian host have been hindered for a long time by the absence of suitable small animal models. Immune compromised mouse models that recapitulate the complete HCV life cycle have been useful to investigate many aspects of the HCV life cycle including antiviral interventions. However, HCV has a high propensity to establish persistence and associated histopathological manifestations such as steatosis, fibrosis, cirrhosis and hepatocellular carcinoma (HCC). Better understanding of these processes requires the development of a permissive and fully immunocompetent small animal model. In this review we summarize the in vivo models that are available for the study of HCV.
Keywords: viral hepatitis, antiviral therapy, animal models, pathology
2 Introduction to experimental models for HCV research
After its discovery 26 years ago [1], it quickly became apparent that chimpanzees were the only other species, besides humans, to be susceptible to HCV infection. Over the years several landmark advances have been achieved including the replicon system in 1999 [2,3] and the identification of a full-length isolate of HCV (JFH1) in 2005 [4-6] that could recapitulate the full viral life cycle in the human hepatoma cell line Huh7, so called cell culture HCV (HCVcc). Together with the (sub)genomic replicon, this HCVcc system has been the working horse for screening and identification of new antiviral drugs [7]. HCV infects and replicates in hepatocytes, which in vivo are characterized by a highly-polarized morphology and reside in a complex liver architecture [8]. Advancing research tools beyond HCVcc and human hepatoma cells has remained challenging. Primary HCV isolates show a very limited ability to replicate in tissue culture and the use of primary hepatocytes in culture is complicated by rapid loss of differentiation and poor reproducibility [9,10]. Some advances have been made in the use of primary hepatocyte culture systems [11-14] and more recently with induced pluripotent stem cell (iPSC) derived hepatocyte-like cells [15-17], but only HCVcc reproducibly infect such models and no reliable in vitro system is able to robustly support primary HCV isolates of different genotypes.
3 Animal models to study HCV infection
Besides humans, the experimental infection of chimpanzees has played a pivotal role in the discovery of HCV and has proven very valuable for deciphering host-virus interactions, particularly of cellular immunity, and preclinical analysis of antiviral strategies [18]. Genomes that acquired cell culture adaptive mutations were found to be highly attenuated in chimpanzees or reverted to the wild type sequence once injected in mice with humanized liver, again underscoring the discrepancies between these models and restrictions to the biological relevance of in vitro systems [19,20]. However, even between chimpanzees and humans subtle differences in HCV became apparent. Whereas a minority of humans spontaneously clears the infection, few chimpanzees evolve to chronicity and to date no fibrosis and only one hepatocellular carcinoma (HCC) has been observed.
Because of growing ethical constraints, limited availability and the high costs associated with chimpanzee studies other animals have been tested in their ability to support HCV infection. Although HCV can infect iPSC derived hepatocyte-like cells from pigtail macaques [21], to date no viremia has been shown in macaques or other primates besides chimpanzees [22]. In addition to primates, several other species have been evaluated for HCV susceptibility but most showed to be resistant to infection. One exception is the tree shrew (Tupaia belangeri), a non-rodent squirrel-like mammal that was found to be permissive for persistent low-level HCV viremia including HCV-related liver disorders in some animals [23].
Due to this narrow host tropism of HCV, the development of practical small animal models for HCV, e.g. laboratory mice and rats, has been challenging [24]. Whilst rodents are naturally resistant to HCV infection different approaches have been undertaken to enable the study of the virus in mice. Four different approaches will be highlighted in this review: the study of novel HCV homologs that naturally infect rodents, the adaptation of HCV to the murine environment, humanization of the mouse through genetic approaches, and by xenotransplantation methods.
3.1 HCV HOMOLOGS
Alternatives to human and chimpanzee samples for the study of HCV biology in vivo could be found in HCV-related viruses that infect other animal species (Figure 1, first panel). Until 2011, the only known homolog of HCV was GBV-B, named after a surgeon (initials GB) suffering from acute hepatitis whose serum was used to infect tamarins that subsequently developed acute hepatitis [25,26]. While GBV-B infection of New World monkeys could be used as an HCV surrogate model, persistent infection is rare [27]. Indeed, an ideal surrogate model should not only resemble HCV’s hepatotropism, but essentially also its ability to establish persistent infection, associated immune responses and ultimately pathogenesis. Recent deep sequencing virome analyses have led to the identification of a number of previously unknown HCV-related hepaci- and pegiviruses in dogs, horses, rodents, bats and non-human primates, thereby greatly broadening the hepacivirus genus [28]. The equine non-primate hepaciviruses (NPHV) is the best studied of the novel hepaciviridae. While NPHV infection can indeed be persistent, mildly elevated liver enzymes, infiltration of lymphocytes into the liver and even mild hepatitis have been observed in horses that clear infection [29,30]. In addition, a novel equine pegivirus (EPgV) has been identified that is linked to liver disease in horses, designated Theiler’s disease associated virus (TDAV) [31].
Figure 1. Different approaches to study HCV in animal models.
First panel: HCV-related viruses that infect animal species such as wild mice, rats, tamarins, bats and horses. These infections can be studied in their natural host, or eventually immune competent laboratory inbred mouse strains. Second panel: in vitro adaptation of HCV to mouse hepatoma cells may allow the isolation of viral variants that can establish an infection in wild type mice. Third panel: transient or stable expression of human factors that are essential to support infection of wild type HCV. Fourth panel: in xenotransplantation models, the genetic background of the host permits repopulation of the liver upon transplantation of human hepatocytes. Additional transplantation of HLA-compatible hematopoietic stem cells results in dually reconstituted mice.
Obviously, the large size and associated animal care costs of horses, compared to conventional laboratory animal models, is a huge drawback. Therefore, novel rodent hepaciviruses are of particular interest. Infections of these viruses in their natural host, or eventually immune competent laboratory inbred mouse strains, warrant further investigation. Methodical searches for hepaciviruses in several species of wild rodents have led to the identification of potential small animal models for HCV, such as deer mice (Peromyscus sps.), the hispid pocket mouse (Chaetodipus hispidus), the desert wood rat (Neotoma lepida), bank voles (Myodes glareolus), South African four-striped mice (Rhabdomys pumilio) and rats (Rattus norvegicus) from New York City [32-34]. Importantly, hepatotropism and histopathological signs of liver inflammation have been observed in some of these rodents, e.g. in bank voles and rats. However, to confirm the utility of rodent hepacivirus infections as surrogate HCV model systems, it remains to be examined whether the pathogenesis of these viruses parallels that seen for HCV in humans.
3.2 VIRAL ADAPTATION
Using the error-prone replication of most RNA viruses, one approach to infect murine cells is to adapt HCV to the mouse environment. Long-term in vitro cultivation of HCV in the presence of mouse cells or cells expressing murine entry proteins could allow the virus to adapt to these murine factors (Figure 1, second panel). Indeed, using this approach Bitzegeio and colleagues discovered adaptive mutations in the viral envelope proteins E1 and E2 enabling the virus to utilize murine CD81 and OCLN for cell entry [35]. This is important because residues in both mouse CD81 and OCLN were previously shown to form the species barrier of HCV infection of mouse cells [36]. Recently, the same group reported that this mouse adapted virus was able to infect, replicate and produce new infectious viral particles in immortalized mouse liver cell lines with defective innate immunity in the absence of any human cofactors [37]. Whether this variant needs additional adaptations to productively infect primary mouse hepatocytes in vivo and persist in the animals is subject to further investigation.
3.3 THE GENETICALLY HUMANIZED MOUSE MODEL
Complimenting the strategy of adapting HCV to the mouse environment, genetic manipulation of the host can be used to knock down murine factors that hamper productive HCV infection; or to complement the mouse with human factors that are essential for this process. Therefore, knowledge of the barriers that determine the human tropism of HCV is essential. This approach has most efficiently been performed with the HCV entry factors, where transient expression of the human restriction factors CD81 and OCLN by adenoviral delivery supported HCVcc entry into the mouse hepatocytes and allowed evaluation of entry inhibitors and vaccine candidates (Figure 1, third panel) [38-40]. Building on this work, mice that were transgenic for four human entry factors (4hEF) and deficient in several innate immune signaling pathways not only allowed HCVcc entry but also low level replication, with infectious virus being recovered from the mouse serum [41]. Using a similar CD81/OCLN transgenic approach this was recently expanded to HCV isolates beyond HCVcc and in mice that have an intact innate immune system, albeit with very low level of viremia [42]. Differences between the ICR mouse background in this latter study and C57BL/6 mice used in the 4hEF model suggest murine genetic differences may be important for replication of patient-derived HCV isolates. Apolipoprotein E, known to enhance HCV production in mouse hepatoma cells [43], was higher expressed and the liver-specific facilitator for HCV replication, miR-122 [44], was much better induced during HCV infection in the ICR vs. C57BL/6 background upon human CD81/OCLN expression [42]. These and potentially other unique hepatic components critical for HCV replication and assembly partially explain the observed HCV persistent infections. Further work is needed to confirm the utility of these models to study adaptive immune responses, immune evasion and vaccination strategies. It will be interesting to see if long-term infection in these models leads to fibrosis and/or HCC development.
3.4 THE HUMAN-LIVER XENOGRAFT MOUSE MODEL
The introduction of mice of which the liver was xenografted with human hepatocytes was an important innovation for the study of HCV, and hepatotropic pathogens in general. Liver chimeric mice are generated by intraspenically infusing primary human hepatocytes into mice with a genetic immune deficiency combined with a constitutive or inducible liver injury (Figure 1, fourth panel) [45-50]. While the liver injury provides a niche for hepatocellular engraftment, the immunodeficiency is needed to avoid rejection of xenogeneic grafts. Hepatocytes have a remarkable regenerative capacity that, after migration to and integration into the liver, allows them to extensively and sometimes nearly completely repopulate the diseased liver parenchyma [51-55]. Although two different recipient lines have predominantly been used for generating human-liver chimeric mice: the uPA-SCID mouse and the FRG mouse [56], susceptibility to HCV infection has also been shown in alternative immunodeficient liver injury models such as MUP-uPA [57] and TK-NOG mice [58]. While subtle differences exist, they are essentially very similar and allow robust and reproducible infection of both natural virus isolates and cell-culture produced HCV [50,59-61]. These humanized mice have been very useful for the study of the basic aspects of the HCV life cycle [20,62-70], the evaluation of passive immunization strategies [60,71-74] and the assessment of novel antiviral therapies [50,75-86].
Since the primary human hepatocytes that reside is the mouse liver have retained many of their functions, human-liver chimeric mice have also been extensively used for the study of other hepatotropic microorganisms such as HBV [46,87-92], HDV [93,94] and Plasmodium falciparum, the causative agent of malaria [95-101]. Their applications extend beyond infectious diseases to the evaluation of the human-type metabolism and potential toxicity of medicinal compounds [102-107]; and the basic cell biology of the human hepatocyte [108,109].
3.5 IMMUNOCOMPETENT XENOGRAFT MOUSE MODELS
In order to prevent rejection of engrafted human hepatocytes, human-liver chimeric mouse models require an immune deficiency. However, studies on HCV immunopathogenesis, primary (human) adaptive immune response and vaccine efficacy in mice would require both a human liver graft and a functional (human) immune system in one and the same recipient animal. This has recently been achieved by combining adult human hepatocytes and human CD34+ hematopoietic stem cells (HSCs) from different human donors [110,111]. Although the authors did not show HCV infection, the high human engraftment levels suggest that these mice could become viremic upon HCV challenge. In addition to this allogeneic system, a syngeneic double reconstituted mouse model was generated by intrahepatic injection of human hepatoblasts (hepatocyte progenitor cells) and CD34+ HSCs from a single fetal donor (Figure 1, fourth panel) [112]. Importantly, HCV challenged mice developed a human anti-HCV T cell response and liver fibrosis. Although HCV RNA was present in liver extracts, the low level of human liver reconstitution in this model did not support detectable viremia. More recently the same group published another double syngeneic reconstituted model, which they used to study HBV immunopathogenesis [113]. Yet again, the rather low level of human hepatocyte engraftment will probably hamper similar studies on HCV infections. In parallel, fine-tuning of human immune cell repopulation and stimulation of specific immune subset development including myeloid and natural killer cells [114], could eventually be used to investigate involvement of these immune cells in HCV infections.
4 Conclusions and future perspectives
Recently approved direct-acting antivirals have dramatically changed the clinical HCV landscape. From a chronic disease that, even after prolonged harsh therapies, could be cured in only a subset of patients, HCV now can be eradicated in almost all patients with minimal side effects. Nevertheless, HCV animal models remain relevant, particularly for two reasons. To this date we have a limited understanding why only a subset of patients develop fibrosis and HCC, nor how to regress fibrosis and prevent HCC after curing HCV. Second, given its widespread prevalence a prophylactic vaccine remains desirable to prevent ongoing HCV spread in certain high-risk populations and geographical areas. Over the past decade several independent approaches have led to small HCV animal models that can complement or replace studies in chimpanzees. These model systems, together with the recent identification of several HCV homologs, including rodent hepaciviridae, open the possibility to further these two goals in mice. Given their inherent limitations all lines of HCV mouse models will probably serve their own usefulness for specific scientific questions. For example, vaccine studies using HCV homologs in wild type mice may become more relevant than HCV adapted to entry factor transgenic animals that lack essential innate immune pathways. Human-liver chimeric mice in their current state completely lack or contain only very limited cellular immunity, and therefore are not yet useful for vaccine studies. However, if human HCC could be induced or engrafted in human liver chimeric mice, such models may become the preferred model to study various aspects of HCV-HCC interactions that may be more translationally relevant to patient tumors than murine HCC. Therefore further development and characterization of HCV animal models will remain useful until HCV has been eradicated worldwide.
Highlights.
Ethical restraints on the use of chimpanzees have opened a quest for alternative animal models to study HCV
Mice require humanization of the liver to become susceptible to HCV infection
Alternatively the virus can be adapted to the mouse environment
Immunocompetent mice with (genetically) humanized liver are available but entail further optimization
Recently identified non-primate hepaciviruses may provide novel surrogate animal models
Table 1. Characteristics of different animal models for the study of HCV.
Animal model | Detectable virus production |
Assessment of antiviral efficacy |
Liver disease |
Adaptive immune response |
Availability/ throughput |
---|---|---|---|---|---|
CHIMPANZEE | Persistent viremia or acute self- limited |
Vaccination, passive immunization[ 115,116], antiviral therapies [117,118] |
+/− | + | Very low |
TUPAIA | Intermittent viremia [23] |
− | + | − | low |
VIRAL ADAPTATION | − | − | − | − | high |
GENETICALLY HUMANIZED MOUSE MODEL |
No viremia [38] Persistent viremia [41,42] |
DAA [41,42] Entry inhibitors and vaccine candidates [38] (indirect assesment) |
− | + | high |
HUMAN-LIVER CHIMERIC MOUSE MODEL |
Persistent viremia |
Passive immunization [39,60,71,72], DAAs [77,79,80], Entry inhibitors [76,78,81,82] Other antiviral therapies [50,84] |
− | − | Low |
HCV HOMOLOGS IN NATURAL HOST |
Persistent viremia [29,30] |
− | +/− | + | Low (NPHV)/ high (rodent hepacivirus) |
HUMAN IMMUNOCOMPETE NT LIVER MOUSE MODEL |
In liver extracts [112] |
− | + | + | low |
Acknowledgements
The research described herein, performed in our laboratory, has been funded by the Ghent University (Concerted Action Grants 01G00507 and 01G01712), The Research Foundation – Flanders (FWO-Vlaanderen; projects 1500910N, G0212.10N and G052112N), The Agency for Innovation by Science and Technology (IWT project #140045, HILIM-3D), the Belgian Federal Government (IUAP P6/36-HEPRO and P7/47-HEPRO-2), and the European Union (FP6 HEPACIVAC; FP7, HepaMab). YPJ is supported by the National Institute of Diabetes, Digestive and Kidney Disease (K08DK090576). KV is supported by a Fellowship of the Belgian American Educational Foundation. Finally, we want to thank Ms. Julie Vercauteren for graphical assistance.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of interest: Nothing to disclose.
References
- 1.Choo QL, Kuo G, Weiner AJ, Overby LR, Bradley DW, Houghton M. Isolation of a cDNA clone derived from a blood-borne non-A, non-B viral hepatitis genome. Science. 1989;244:359–362. doi: 10.1126/science.2523562. [DOI] [PubMed] [Google Scholar]
- 2.Lohmann V, Korner F, Koch J, Herian U, Theilmann L, Bartenschlager R. Replication of subgenomic hepatitis C virus RNAs in a hepatoma cell line. Science. 1999;285:110–113. doi: 10.1126/science.285.5424.110. [DOI] [PubMed] [Google Scholar]
- 3.Blight KJ, Kolykhalov AA, Rice CM. Efficient initiation of HCV RNA replication in cell culture. Science. 2000;290:1972–1974. doi: 10.1126/science.290.5498.1972. [DOI] [PubMed] [Google Scholar]
- 4.Wakita T, Pietschmann T, Kato T, Date T, Miyamoto M, Zhao Z, Murthy K, Habermann A, Krausslich HG, Mizokami M, et al. Production of infectious hepatitis C virus in tissue culture from a cloned viral genome. Nat Med. 2005;11:791–796. doi: 10.1038/nm1268. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Zhong J, Gastaminza P, Cheng G, Kapadia S, Kato T, Burton DR, Wieland SF, Uprichard SL, Wakita T, Chisari FV. Robust hepatitis C virus infection in vitro. Proc Natl Acad Sci U S A. 2005;102:9294–9299. doi: 10.1073/pnas.0503596102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Lindenbach BD, Evans MJ, Syder AJ, Wolk B, Tellinghuisen TL, Liu CC, Maruyama T, Hynes RO, Burton DR, McKeating JA, et al. Complete replication of hepatitis C virus in cell culture. Science. 2005;309:623–626. doi: 10.1126/science.1114016. [DOI] [PubMed] [Google Scholar]
- 7.Bartenschlager R, Lohmann V, Penin F. The molecular and structural basis of advanced antiviral therapy for hepatitis C virus infection. Nat Rev Microbiol. 2013;11:482–496. doi: 10.1038/nrmicro3046. [DOI] [PubMed] [Google Scholar]
- 8.Wang L, Boyer JL. The maintenance and generation of membrane polarity in hepatocytes. Hepatology. 2004;39:892–899. doi: 10.1002/hep.20039. [DOI] [PubMed] [Google Scholar]
- 9.Sheahan T, Jones CT, Ploss A. Advances and challenges in studying hepatitis C virus in its native environment. Expert Rev Gastroenterol Hepatol. 2010;4:541–550. doi: 10.1586/egh.10.53. [DOI] [PubMed] [Google Scholar]
- 10.Podevin P, Carpentier A, Pene V, Aoudjehane L, Carriere M, Zaidi S, Hernandez C, Calle V, Meritet JF, Scatton O, et al. Production of infectious hepatitis C virus in primary cultures of human adult hepatocytes. Gastroenterology. 2010;139:1355–1364. doi: 10.1053/j.gastro.2010.06.058. [DOI] [PubMed] [Google Scholar]
- 11.Ploss A, Khetani SR, Jones CT, Syder AJ, Trehan K, Gaysinskaya VA, Mu K, Ritola K, Rice CM, Bhatia SN. Persistent hepatitis C virus infection in microscale primary human hepatocyte cultures. Proc Natl Acad Sci U S A. 2010;107:3141–3145. doi: 10.1073/pnas.0915130107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Andrus L, Marukian S, Jones CT, Catanese MT, Sheahan TP, Schoggins JW, Barry WT, Dustin LB, Trehan K, Ploss A, et al. Expression of paramyxovirus V proteins promotes replication and spread of hepatitis C virus in cultures of primary human fetal liver cells. Hepatology. 2011;54:1901–1912. doi: 10.1002/hep.24557. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Marukian S, Andrus L, Sheahan TP, Jones CT, Charles ED, Ploss A, Rice CM, Dustin LB. Hepatitis C virus induces interferon-lambda and interferon-stimulated genes in primary liver cultures. Hepatology. 2011;54:1913–1923. doi: 10.1002/hep.24580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Lagaye S, Shen H, Saunier B, Nascimbeni M, Gaston J, Bourdoncle P, Hannoun L, Massault PP, Vallet-Pichard A, Mallet V, et al. Efficient replication of primary or culture hepatitis C virus isolates in human liver slices: a relevant ex vivo model of liver infection. Hepatology. 2012;56:861–872. doi: 10.1002/hep.25738. [DOI] [PubMed] [Google Scholar]
- 15.Wu X, Robotham JM, Lee E, Dalton S, Kneteman NM, Gilbert DM, Tang H. Productive hepatitis C virus infection of stem cell-derived hepatocytes reveals a critical transition to viral permissiveness during differentiation. PLoS Pathog. 2012;8:e1002617. doi: 10.1371/journal.ppat.1002617. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Roelandt P, Obeid S, Paeshuyse J, Vanhove J, Van Lommel A, Nahmias Y, Nevens F, Neyts J, Verfaillie CM. Human pluripotent stem cell-derived hepatocytes support complete replication of hepatitis C virus. J Hepatol. 2012;57:246–251. doi: 10.1016/j.jhep.2012.03.030. [DOI] [PubMed] [Google Scholar]
- 17.Schwartz RE, Fleming HE, Khetani SR, Bhatia SN. Pluripotent stem cell-derived hepatocyte-like cells. Biotechnol Adv. 2014;32:504–513. doi: 10.1016/j.biotechadv.2014.01.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Bukh J. A critical role for the chimpanzee model in the study of hepatitis C. Hepatology. 2004;39:1469–1475. doi: 10.1002/hep.20268. [DOI] [PubMed] [Google Scholar]
- 19.Bukh J, Pietschmann T, Lohmann V, Krieger N, Faulk K, Engle RE, Govindarajan S, Shapiro M, St Claire M, Bartenschlager R. Mutations that permit efficient replication of hepatitis C virus RNA in Huh-7 cells prevent productive replication in chimpanzees. Proc Natl Acad Sci U S A. 2002;99:14416–14421. doi: 10.1073/pnas.212532699. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Kaul A, Woerz I, Meuleman P, Roels G, Bartenschlager R. Cell culture adaptation of hepatitis C virus and in vivo viability of an adapted variant. J Virol. 2007;81:13168–13179. doi: 10.1128/JVI.01362-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Sourisseau M, Goldman O, He W, Gori JL, Kiem HP, Gouon-Evans V, Evans MJ. Hepatic cells derived from induced pluripotent stem cells of pigtail macaques support hepatitis C virus infection. Gastroenterology. 2013;145:966–969. e967. doi: 10.1053/j.gastro.2013.07.026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Abe K, Kurata T, Teramoto Y, Shiga J, Shikata T. Lack of susceptibility of various primates and woodchucks to hepatitis C virus. J Med Primatol. 1993;22:433–434. [PubMed] [Google Scholar]
- 23.Amako Y, Tsukiyama-Kohara K, Katsume A, Hirata Y, Sekiguchi S, Tobita Y, Hayashi Y, Hishima T, Funata N, Yonekawa H, et al. Pathogenesis of hepatitis C virus infection in Tupaia belangeri. J Virol. 2010;84:303–311. doi: 10.1128/JVI.01448-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Mailly L, Robinet E, Meuleman P, Baumert TF, Zeisel MB. Hepatitis C virus infection and related liver disease: the quest for the best animal model. Front Microbiol. 2013;4:213. doi: 10.3389/fmicb.2013.00212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Deinhardt F, Holmes AW, Capps RB, Popper H. Studies on the transmission of human viral hepatitis to marmoset monkeys. I. Transmission of disease, serial passages, and description of liver lesions. J Exp Med. 1967;125:673–688. doi: 10.1084/jem.125.4.673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Stapleton JT, Foung S, Muerhoff AS, Bukh J, Simmonds P. The GB viruses: a review and proposed classification of GBV-A, GBV-C (HGV), and GBV-D in genus Pegivirus within the family Flaviviridae. J Gen Virol. 2011;92:233–246. doi: 10.1099/vir.0.027490-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Takikawa S, Engle RE, Faulk KN, Emerson SU, Purcell RH, Bukh J. Molecular evolution of GB virus B hepatitis virus during acute resolving and persistent infections in experimentally infected tamarins. J Gen Virol. 2010;91:727–733. doi: 10.1099/vir.0.015750-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28*.Scheel TK, Simmonds P, Kapoor A. Surveying the global virome: Identification and characterization of HCV-related animal hepaciviruses. Antiviral Res. 2014;115C:83–93. doi: 10.1016/j.antiviral.2014.12.014. This review summarizes the recent identification of a number of previously unknown HCV-related hepaci- and pegiviruses in dogs, horses, bats, non-human primates and rodents. The latter potentially paving the way to surrogate small animal model systems for HCV.
- 29.Pfaender S, Cavalleri JM, Walter S, Doerrbecker J, Campana B, Brown RJ, Burbelo PD, Postel A, Hahn K, Anggakusuma, et al. Clinical course of infection and viral tissue tropism of hepatitis C virus-like nonprimate hepaciviruses in horses. Hepatology. 2015;61:448–459. doi: 10.1002/hep.27440. [DOI] [PubMed] [Google Scholar]
- 30.Scheel TK, Kapoor A, Nishiuchi E, Brock KV, Yu Y, Andrus L, Gu M, Renshaw RW, Dubovi EJ, McDonough SP, et al. Characterization of nonprimate hepacivirus and construction of a functional molecular clone. Proc Natl Acad Sci U S A. 2015 doi: 10.1073/pnas.1500265112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Chandriani S, Skewes-Cox P, Zhong W, Ganem DE, Divers TJ, Van Blaricum AJ, Tennant BC, Kistler AL. Identification of a previously undescribed divergent virus from the Flaviviridae family in an outbreak of equine serum hepatitis. Proc Natl Acad Sci U S A. 2013;110:E1407–1415. doi: 10.1073/pnas.1219217110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Kapoor A, Simmonds P, Scheel TK, Hjelle B, Cullen JM, Burbelo PD, Chauhan LV, Duraisamy R, Sanchez Leon M, Jain K, et al. Identification of rodent homologs of hepatitis C virus and pegiviruses. MBio. 2013;4:e00216–00213. doi: 10.1128/mBio.00216-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Drexler JF, Corman VM, Muller MA, Lukashev AN, Gmyl A, Coutard B, Adam A, Ritz D, Leijten LM, van Riel D, et al. Evidence for novel hepaciviruses in rodents. PLoS Pathog. 2013;9:e1003438. doi: 10.1371/journal.ppat.1003438. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Firth C, Bhat M, Firth MA, Williams SH, Frye MJ, Simmonds P, Conte JM, Ng J, Garcia J, Bhuva NP, 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. doi: 10.1128/mBio.01933-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Bitzegeio J, Bankwitz D, Hueging K, Haid S, Brohm C, Zeisel MB, Herrmann E, Iken M, Ott M, Baumert TF, et al. Adaptation of hepatitis C virus to mouse CD81 permits infection of mouse cells in the absence of human entry factors. PLoS Pathog. 2010;6:e1000978. doi: 10.1371/journal.ppat.1000978. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Ploss A, Evans MJ, Gaysinskaya VA, Panis M, You HN, de Jong YP, Rice CM. Human occludin is a hepatitis C virus entry factor required for infection of mouse cells. Nature. 2009;457:882–886. doi: 10.1038/nature07684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37*.Frentzen A, Anggakusuma, Gurlevik E, Hueging K, Knocke S, Ginkel C, Brown RJ, Heim M, Dill MT, Kroger A, et al. Cell entry, efficient RNA replication, and production of infectious hepatitis C virus progeny in mouse liver-derived cells. Hepatology. 2014;59:78–88. doi: 10.1002/hep.26626. The life cycle of a mouse adapted HCV variant that uses murine CD81 and OCLN for cell entry is completely recapitulated in mouse liver-derived cell lines upon innate immunity blunting and overexpression of miR-122, ApoE and HCV entry factors in the absence of any human cofactors.
- 38.Dorner M, Horwitz JA, Robbins JB, Barry WT, Feng Q, Mu K, Jones CT, Schoggins JW, Catanese MT, Burton DR, et al. A genetically humanized mouse model for hepatitis C virus infection. Nature. 2011;474:208–211. doi: 10.1038/nature10168. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Giang E, Dorner M, Prentoe JC, Dreux M, Evans MJ, Bukh J, Rice CM, Ploss A, Burton DR, Law M. Human broadly neutralizing antibodies to the envelope glycoprotein complex of hepatitis C virus. Proc Natl Acad Sci U S A. 2012;109:6205–6210. doi: 10.1073/pnas.1114927109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Anggakusuma, Colpitts CC, Schang LM, Rachmawati H, Frentzen A, Pfaender S, Behrendt P, Brown RJ, Bankwitz D, Steinmann J, et al. Turmeric curcumin inhibits entry of all hepatitis C virus genotypes into human liver cells. Gut. 2013 doi: 10.1136/gutjnl-2012-304299. [DOI] [PubMed] [Google Scholar]
- 41*.Dorner M, Horwitz JA, Donovan BM, Labitt RN, Budell WC, Friling T, Vogt A, Catanese MT, Satoh T, Kawai T, et al. Completion of the entire hepatitis C virus life cycle in genetically humanized mice. Nature. 2013;501:237–241. doi: 10.1038/nature12427. Innate immune defective C57BL/6 mice transgenic for human HCV entry factors allow productive infection of cell culture-derived HCV.
- 42*.Chen J, Zhao Y, Zhang C, Chen H, Feng J, Chi X, Pan Y, Du J, Guo M, Cao H, et al. Persistent hepatitis C virus infections and hepatopathological manifestations in immune-competent humanized mice. Cell Res. 2014;24:1050–1066. doi: 10.1038/cr.2014.116. Immune competent ICR mice transgenic for human HCV entry factors allow productive infection of primary HCV isolates.
- 43.Long G, Hiet MS, Windisch MP, Lee JY, Lohmann V, Bartenschlager R. Mouse hepatic cells support assembly of infectious hepatitis C virus particles. Gastroenterology. 2011;141:1057–1066. doi: 10.1053/j.gastro.2011.06.010. [DOI] [PubMed] [Google Scholar]
- 44.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. doi: 10.1126/science.1113329. [DOI] [PubMed] [Google Scholar]
- 45**.Mercer DF, Schiller DE, Elliott JF, Douglas DN, Hao C, Rinfret A, Addison WR, Fischer KP, Churchill TA, Lakey JR, et al. Hepatitis C virus replication in mice with chimeric human livers. Nat Med. 2001;7:927–933. doi: 10.1038/90968. The first use of the human-liver chimeric uPA-SCID mouse model for HCV infections.
- 46.Dandri M, Burda MR, Torok E, Pollok JM, Iwanska A, Sommer G, Rogiers X, Rogler CE, Gupta S, Will H, et al. Repopulation of mouse liver with human hepatocytes and in vivo infection with hepatitis B virus. Hepatology. 2001;33:981–988. doi: 10.1053/jhep.2001.23314. [DOI] [PubMed] [Google Scholar]
- 47.Meuleman P, Libbrecht L, De Vos R, de Hemptinne B, Gevaert K, Vandekerckhove J, Roskams T, Leroux-Roels G. Morphological and biochemical characterization of a human liver in a uPA-SCID mouse chimera. Hepatology. 2005;41:847–856. doi: 10.1002/hep.20657. [DOI] [PubMed] [Google Scholar]
- 48.Azuma H, Paulk N, Ranade A, Dorrell C, Al-Dhalimy M, Ellis E, Strom S, Kay MA, Finegold M, Grompe M. Robust expansion of human hepatocytes in Fah−/−/Rag2−/−/Il2rg−/− mice. Nat Biotechnol. 2007;25:903–910. doi: 10.1038/nbt1326. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Bissig KD, Le TT, Woods NB, Verma IM. Repopulation of adult and neonatal mice with human hepatocytes: a chimeric animal model. Proc Natl Acad Sci U S A. 2007;104:20507–20511. doi: 10.1073/pnas.0710528105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50**.Bissig KD, Wieland SF, Tran P, Isogawa M, Le TT, Chisari FV, Verma IM. Human liver chimeric mice provide a model for hepatitis B and C virus infection and treatment. J Clin Invest. 2010;120:924–930. doi: 10.1172/JCI40094. The first description of using the FRG mouse model with inducible liver injury for HCV infections.
- 51.Sandgren EP, Palmiter RD, Heckel JL, Daugherty CC, Brinster RL, Degen JL. Complete hepatic regeneration after somatic deletion of an albumin-plasminogen activator transgene. Cell. 1991;66:245–256. doi: 10.1016/0092-8674(91)90615-6. [DOI] [PubMed] [Google Scholar]
- 52.Overturf K, Al-Dhalimy M, Tanguay R, Brantly M, Ou CN, Finegold M, Grompe M. Hepatocytes corrected by gene therapy are selected in vivo in a murine model of hereditary tyrosinaemia type I. Nat Genet. 1996;12:266–273. doi: 10.1038/ng0396-266. [DOI] [PubMed] [Google Scholar]
- 53.Fausto N. Hepatocytes break the rules of senescence in serial transplantation studies. Is there a limit to their replicative capacity? Am J Pathol. 1997;151:1187–1189. [PMC free article] [PubMed] [Google Scholar]
- 54.Fausto N. Liver regeneration. J Hepatol. 2000;32:19–31. doi: 10.1016/s0168-8278(00)80412-2. [DOI] [PubMed] [Google Scholar]
- 55.Fausto N, Campbell JS, Riehle KJ. Liver regeneration. Hepatology. 2006;43:S45–53. doi: 10.1002/hep.20969. [DOI] [PubMed] [Google Scholar]
- 56.Vercauteren K, de Jong YP, Meuleman P. HCV animal models and liver disease. J Hepatol. 2014;61:S26–33. doi: 10.1016/j.jhep.2014.07.013. [DOI] [PubMed] [Google Scholar]
- 57.Tesfaye A, Stift J, Maric D, Cui Q, Dienes HP, Feinstone SM. Chimeric mouse model for the infection of hepatitis B and C viruses. PLoS One. 2013;8:e77298. doi: 10.1371/journal.pone.0077298. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.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. doi: 10.1016/j.bbrc.2013.10.040. [DOI] [PubMed] [Google Scholar]
- 59.Bukh J, Meuleman P, Tellier R, Engle RE, Feinstone SM, Eder G, Satterfield WC, Govindarajan S, Krawczynski K, Miller RH, et al. Challenge pools of hepatitis C virus genotypes 1-6 prototype strains: replication fitness and pathogenicity in chimpanzees and human liver-chimeric mouse models. J Infect Dis. 2010;201:1381–1389. doi: 10.1086/651579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Meuleman P, Bukh J, Verhoye L, Farhoudi A, Vanwolleghem T, Wang RY, Desombere I, Alter H, Purcell RH, Leroux-Roels G. In vivo evaluation of the cross-genotype neutralizing activity of polyclonal antibodies against hepatitis C virus. Hepatology. 2011;53:755–762. doi: 10.1002/hep.24171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Lindenbach BD, Meuleman P, Ploss A, Vanwolleghem T, Syder AJ, McKeating JA, Lanford RE, Feinstone SM, Major ME, Leroux-Roels G, et al. Cell culture-grown hepatitis C virus is infectious in vivo and can be recultured in vitro. P Natl Acad Sci USA. 2006;103:3805–3809. doi: 10.1073/pnas.0511218103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Vassilaki N, Friebe P, Meuleman P, Kallis S, Kaul A, Paranhos-Baccala G, Leroux-Roels G, Mavromara P, Bartenschlager R. Role of the hepatitis C virus core+1 open reading frame and core cis-acting RNA elements in viral RNA translation and replication. J Virol. 2008;82:11503–11515. doi: 10.1128/JVI.01640-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Joyce MA, Walters KA, Lamb SE, Yeh MM, Zhu LF, Kneteman N, Doyle JS, Katze MG, Tyrrell DL. HCV induces oxidative and ER stress, and sensitizes infected cells to apoptosis in SCID/Alb-uPA mice. PLoS pathogens. 2009;5:e1000291. doi: 10.1371/journal.ppat.1000291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Walters KA, Syder AJ, Lederer SL, Diamond DL, Paeper B, Rice CM, Katze MG. Genomic analysis reveals a potential role for cell cycle perturbation in HCV-mediated apoptosis of cultured hepatocytes. PLoS Pathog. 2009;5:e1000269. doi: 10.1371/journal.ppat.1000269. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Pietschmann T, Zayas M, Meuleman P, Long G, Appel N, Koutsoudakis G, Kallis S, Leroux-Roels G, Lohmann V, Bartenschlager R. Production of infectious genotype 1b virus particles in cell culture and impairment by replication enhancing mutations. PLoS Pathog. 2009;5:e1000475. doi: 10.1371/journal.ppat.1000475. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Fafi-Kremer S, Fofana I, Soulier E, Carolla P, Meuleman P, Leroux-Roels G, Patel AH, Cosset FL, Pessaux P, Doffoel M, et al. Viral entry and escape from antibody-mediated neutralization influence hepatitis C virus reinfection in liver transplantation. J Exp Med. 2010;207:2019–2031. doi: 10.1084/jem.20090766. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Gottwein JM, Jensen TB, Mathiesen CK, Meuleman P, Serre SB, Lademann JB, Ghanem L, Scheel TK, Leroux-Roels G, Bukh J. Development and application of hepatitis C reporter viruses with genotype 1 to 7 core-nonstructural protein 2 (NS2) expressing fluorescent proteins or luciferase in modified JFH1 NS5A. J Virol. 2011;85:8913–8928. doi: 10.1128/JVI.00049-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Brown RJ, Hudson N, Wilson G, Rehman SU, Jabbari S, Hu K, Tarr AW, 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. doi: 10.1128/JVI.01079-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Doerrbecker J, Meuleman P, Kang J, Riebesehl N, Wilhelm C, Friesland M, Pfaender S, Steinmann J, Pietschmann T, Steinmann E. Thermostability of seven hepatitis C virus genotypes in vitro and in vivo. Journal of viral hepatitis. 2013;20:478–485. doi: 10.1111/jvh.12055. [DOI] [PubMed] [Google Scholar]
- 70.Singaravelu R, Chen R, Lyn RK, Jones DM, O’Hara S, Rouleau Y, Cheng J, Srinivasan P, Nasheri N, Russell RS, et al. Hepatitis C virus induced up-regulation of microRNA-27: a novel mechanism for hepatic steatosis. Hepatology. 2014;59:98–108. doi: 10.1002/hep.26634. [DOI] [PubMed] [Google Scholar]
- 71.Vanwolleghem T, Bukh J, Meuleman P, Desombere I, Meunier JC, Alter H, Purcell RH, 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. doi: 10.1002/hep.22244. [DOI] [PubMed] [Google Scholar]
- 72.Law M, Maruyama T, Lewis J, Giang E, Tarr AW, Stamataki Z, Gastaminza P, Chisari FV, Jones IM, Fox RI, et al. Broadly neutralizing antibodies protect against hepatitis C virus quasispecies challenge. Nat Med. 2008;14:25–27. doi: 10.1038/nm1698. [DOI] [PubMed] [Google Scholar]
- 73.Akazawa D, Moriyama M, Yokokawa H, Omi N, Watanabe N, Date T, Morikawa K, Aizaki H, Ishii K, Kato T, et al. Neutralizing antibodies induced by cell culture-derived hepatitis C virus protect against infection in mice. Gastroenterology. 2013;145:447–455. e441–444. doi: 10.1053/j.gastro.2013.05.007. [DOI] [PubMed] [Google Scholar]
- 74.de Jong YP, Dorner M, Mommersteeg MC, Xiao JW, Balazs AB, Robbins JB, Winer BY, Gerges S, Vega K, Labitt RN, et al. Broadly neutralizing antibodies abrogate established hepatitis C virus infection. Sci Transl Med. 2014;6:254ra129. doi: 10.1126/scitranslmed.3009512. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.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. doi: 10.1053/j.gastro.2007.07.007. [DOI] [PubMed] [Google Scholar]
- 76.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. doi: 10.1002/hep.22547. [DOI] [PubMed] [Google Scholar]
- 77.Kneteman NM, Howe AY, Gao T, Lewis J, Pevear D, Lund G, Douglas D, Mercer DF, Tyrrell DL, Immermann F, et al. HCV796: A selective nonstructural protein 5B polymerase inhibitor with potent anti-hepatitis C virus activity in vitro, in mice with chimeric human livers, and in humans infected with hepatitis C virus. Hepatology. 2009;49:745–752. doi: 10.1002/hep.22717. [DOI] [PubMed] [Google Scholar]
- 78.Meuleman P, Albecka A, Belouzard S, Vercauteren K, Verhoye L, Wychowski C, Leroux-Roels G, Palmer KE, Dubuisson J. Griffithsin Has Antiviral Activity against Hepatitis C Virus. Antimicrob Agents Chemother. 2011;55:5159–5167. doi: 10.1128/AAC.00633-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Narjes F, Crescenzi B, Ferrara M, Habermann J, Colarusso S, Ferreira Mdel R, Stansfield I, Mackay AC, Conte I, Ercolani C, et al. Discovery of (7R)-14-cyclohexyl-7-{[2-(dimethylamino)ethyl](methyl) amino}-7,8-dihydro-6H-indolo[1,2-e][1,5]benzoxazocine-11-carboxylic acid (MK-3281), a potent and orally bioavailable finger-loop inhibitor of the hepatitis C virus NS5B polymerase. J Med Chem. 2011;54:289–301. doi: 10.1021/jm1013105. [DOI] [PubMed] [Google Scholar]
- 80.Ohara E, Hiraga N, Imamura M, Iwao E, Kamiya N, Yamada I, Kono T, Onishi M, Hirata D, Mitsui F, et al. Elimination of hepatitis C virus by short term NS3-4A and NS5B inhibitor combination therapy in human hepatocyte chimeric mice. J Hepatol. 2011;54:872–878. doi: 10.1016/j.jhep.2010.08.033. [DOI] [PubMed] [Google Scholar]
- 81.Meuleman P, Catanese MT, Verhoye L, Desombere I, Farhoudi A, Jones CT, Sheahan T, Grzyb K, Cortese R, Rice CM, 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. doi: 10.1002/hep.24692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Lacek K, Vercauteren K, Grzyb K, Naddeo M, Verhoye L, Slowikowski MP, Fafi-Kremer S, Patel AH, Baumert TF, Folgori A, et al. Novel human SR-BI antibodies prevent infection and dissemination of HCV in vitro and in humanized mice. J Hepatol. 2012;57:17–23. doi: 10.1016/j.jhep.2012.02.018. [DOI] [PubMed] [Google Scholar]
- 83.Vercauteren K, Van Den Eede N, Mesalam AA, Belouzard S, Catanese MT, Bankwitz D, Wong-Staal F, Cortese R, Dubuisson J, Rice CM, et al. Successful anti-scavenger receptor class B type I (SR-BI) monoclonal antibody therapy in humanized mice after challenge with HCV variants with in vitro resistance to SR-BI-targeting agents. Hepatology. 2014;60:1508–1518. doi: 10.1002/hep.27196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Hsu EC, Hsi B, Hirota-Tsuchihara M, Ruland J, Iorio C, Sarangi F, Diao J, Migliaccio G, Tyrrell DL, Kneteman N, et al. Modified apoptotic molecule (BID) reduces hepatitis C virus infection in mice with chimeric human livers. Nat Biotechnol. 2003;21:519–525. doi: 10.1038/nbt817. [DOI] [PubMed] [Google Scholar]
- 85.Mailly L, Xiao F, Lupberger J, Wilson G, Aubert P, Duong G, Calabrese D, Leboeuf C, Fofana I, Thumann C, et al. Clearance of persistent hepatitis C virus infection using a monoclonal antibody specific for tight junction protein claudin-1. Nature Biotechnology. 2015 doi: 10.1038/nbt.3179. in press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Fukasawa M, Nagase S, Shirasago Y, Iida M, Yamashita M, Endo K, Yagi K, Suzuki T, Wakita T, Hanada K, et al. Monoclonal Antibodies against Extracellular Domains of Claudin-1 Block Hepatitis C Virus Infection in A Mouse Model. J Virol. 2015 doi: 10.1128/JVI.03676-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Meuleman P, Libbrecht L, Wieland S, De Vos R, Habib N, Kramvis A, Roskams T, Leroux-Roels G. Immune suppression uncovers endogenous cytopathic effects of the hepatitis B virus. J Virol. 2006;80:2797–2807. doi: 10.1128/JVI.80.6.2797-2807.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Petersen J, Dandri M, Mier W, Lutgehetmann M, Volz T, von Weizsacker F, Haberkorn U, Fischer L, Pollok JM, Erbes B, et al. Prevention of hepatitis B virus infection in vivo by entry inhibitors derived from the large envelope protein. Nat Biotechnol. 2008;26:335–341. doi: 10.1038/nbt1389. [DOI] [PubMed] [Google Scholar]
- 89.Meuleman P, Leroux-Roels G. The human liver-uPA-SCID mouse: a model for the evaluation of antiviral compounds against HBV and HCV. Antiviral Res. 2008;80:231–238. doi: 10.1016/j.antiviral.2008.07.006. [DOI] [PubMed] [Google Scholar]
- 90.Lutgehetmann M, Volz T, Kopke A, Broja T, Tigges E, Lohse AW, Fuchs E, Murray JM, Petersen J, Dandri M. In vivo proliferation of hepadnavirus-infected hepatocytes induces loss of covalently closed circular DNA in mice. Hepatology. 2010;52:16–24. doi: 10.1002/hep.23611. [DOI] [PubMed] [Google Scholar]
- 91.Belloni L, Allweiss L, Guerrieri F, Pediconi N, Volz T, Pollicino T, Petersen J, Raimondo G, Dandri M, Levrero M. IFN-alpha inhibits HBV transcription and replication in cell culture and in humanized mice by targeting the epigenetic regulation of the nuclear cccDNA minichromosome. J Clin Invest. 2012;122:529–537. doi: 10.1172/JCI58847. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Volz T, Lutgehetmann M, Allweiss L, Warlich M, Bierwolf J, Pollok JM, Petersen J, Matthes E, Dandri M. Strong antiviral activity of the new l-hydroxycytidine derivative, l-Hyd4FC, in HBV-infected human chimeric uPA/SCID mice. Antivir Ther. 2012;17:623–631. doi: 10.3851/IMP2075. [DOI] [PubMed] [Google Scholar]
- 93.Lutgehetmann M, Mancke LV, Volz T, Helbig M, Allweiss L, Bornscheuer T, Pollok JM, Lohse AW, Petersen J, Urban S, et al. Humanized chimeric uPA mouse model for the study of hepatitis B and D virus interactions and preclinical drug evaluation. Hepatology. 2012;55:685–694. doi: 10.1002/hep.24758. [DOI] [PubMed] [Google Scholar]
- 94.Giersch K, Helbig M, Volz T, Allweiss L, Mancke LV, Lohse AW, Polywka S, Pollok JM, Petersen J, Taylor J, et al. Persistent hepatitis D virus mono-infection in humanized mice is efficiently converted by hepatitis B virus to a productive co-infection. J Hepatol. 2014;60:538–544. doi: 10.1016/j.jhep.2013.11.010. [DOI] [PubMed] [Google Scholar]
- 95.Sacci JB, Jr., Alam U, Douglas D, Lewis J, Tyrrell DL, Azad AF, Kneteman NM. Plasmodium falciparum infection and exoerythrocytic development in mice with chimeric human livers. Int J Parasitol. 2006;36:353–360. doi: 10.1016/j.ijpara.2005.10.014. [DOI] [PubMed] [Google Scholar]
- 96.Vaughan AM, Mikolajczak SA, Wilson EM, Grompe M, Kaushansky A, Camargo N, Bial J, Ploss A, Kappe SH. Complete Plasmodium falciparum liver-stage development in liver-chimeric mice. J Clin Invest. 2012;122:3618–3628. doi: 10.1172/JCI62684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.VanBuskirk KM, O’Neill MT, De La Vega P, Maier AG, Krzych U, Williams J, Dowler MG, Sacci JB, Jr., Kangwanrangsan N, Tsuboi T, et al. Preerythrocytic, live-attenuated Plasmodium falciparum vaccine candidates by design. Proc Natl Acad Sci U S A. 2009;106:13004–13009. doi: 10.1073/pnas.0906387106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Behet MC, Foquet L, van Gemert GJ, Bijker EM, Meuleman P, Leroux-Roels G, Hermsen CC, Scholzen A, Sauerwein RW. Sporozoite immunization of human volunteers under chemoprophylaxis induces functional antibodies against pre-erythrocytic stages of Plasmodium falciparum. Malar J. 2014;13:136. doi: 10.1186/1475-2875-13-136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Foquet L, Hermsen CC, van Gemert GJ, Van Braeckel E, Weening KE, Sauerwein R, Meuleman P, Leroux-Roels G. Vaccine-induced monoclonal antibodies targeting circumsporozoite protein prevent Plasmodium falciparum infection. The Journal of clinical investigation. 2014;124:140–144. doi: 10.1172/JCI70349. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.van Schaijk BC, Ploemen IH, Annoura T, Vos MW, Lander F, van Gemert GJ, Chevalley-Maurel S, van de Vegte-Bolmer M, Sajid M, Franetich JF, et al. A genetically attenuated malaria vaccine candidate based on gene-deficient sporozoites. Elife. 2014:3. doi: 10.7554/eLife.03582. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Foquet L, Hermsen CC, Verhoye L, van Gemert GJ, Cortese R, Nicosia A, Sauerwein RW, Leroux-Roels G, Meuleman P. Anti-CD81 but not anti-SR-BI blocks Plasmodium falciparum liver infection in a humanized mouse model. J Antimicrob Chemother. 2015 doi: 10.1093/jac/dkv019. [DOI] [PubMed] [Google Scholar]
- 102.Lootens L, Meuleman P, Pozo OJ, Van Eenoo P, Leroux-Roels G, Delbeke FT. uPA+/+−SCID mouse with humanized liver as a model for in vivo metabolism of exogenous steroids: methandienone as a case study. Clin Chem. 2009;55:1783–1793. doi: 10.1373/clinchem.2008.119396. [DOI] [PubMed] [Google Scholar]
- 103.Pozo OJ, Van Eenoo P, Deventer K, Lootens L, Van Thuyne W, Parr MK, Schanzer W, Sancho JV, Hernandez F, Meuleman P, et al. Detection and characterization of a new metabolite of 17alpha-methyltestosterone. Drug Metab Dispos. 2009;37:2153–2162. doi: 10.1124/dmd.109.028373. [DOI] [PubMed] [Google Scholar]
- 104.Strom SC, 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. doi: 10.1007/978-1-60761-688-7_27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Cohen J. Toxicology. ‘Humanized’ mouse detects deadly drug side effects. Science. 2014;344:244–245. doi: 10.1126/science.344.6181.244. [DOI] [PubMed] [Google Scholar]
- 106.Bateman TJ, Reddy VG, Kakuni M, Morikawa Y, Kumar S. Application of chimeric mice with humanized liver for study of human-specific drug metabolism. Drug Metab Dispos. 2014;42:1055–1065. doi: 10.1124/dmd.114.056978. [DOI] [PubMed] [Google Scholar]
- 107.Xu D, Nishimura T, Nishimura S, Zhang H, Zheng M, Guo YY, Masek M, Michie SA, Glenn J, Peltz G. Fialuridine induces acute liver failure in chimeric TK-NOG mice: a model for detecting hepatic drug toxicity prior to human testing. PLoS Med. 2014;11:e1001628. doi: 10.1371/journal.pmed.1001628. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Hermant P, Demarez C, Mahlakoiv T, Staeheli P, Meuleman P, Michiels T. Human but not mouse hepatocytes respond to interferon-lambda in vivo. PLoS One. 2014;9:e87906. doi: 10.1371/journal.pone.0087906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Steeland S, Puimege L, Vandenbroucke RE, Van Hauwermeiren F, Haustraete J, Devoogdt N, Hulpiau P, Leroux-Roels G, Laukens D, Meuleman P, et al. Generation and characterization of small single domain antibodies inhibiting human TNF receptor 1. J Biol Chem. 2014 doi: 10.1074/jbc.M114.617787. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Gutti TL, Knibbe JS, Makarov E, Zhang J, Yannam GR, Gorantla S, Sun Y, Mercer DF, Suemizu H, Wisecarver JL, et al. Human hepatocytes and hematolymphoid dual reconstitution in treosulfan-conditioned uPA-NOG mice. Am J Pathol. 2014;184:101–109. doi: 10.1016/j.ajpath.2013.09.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Wilson EM, Bial J, Tarlow B, Bial G, Jensen B, Greiner DL, Brehm MA, Grompe M. Extensive double humanization of both liver and hematopoiesis in FRGN mice. Stem Cell Res. 2014;13:404–412. doi: 10.1016/j.scr.2014.08.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112*.Washburn ML, Bility MT, Zhang L, Kovalev GI, Buntzman A, Frelinger JA, Barry W, Ploss A, Rice CM, Su L. A humanized mouse model to study hepatitis C virus infection, immune response, and liver disease. Gastroenterology. 2011;140:1334–1344. doi: 10.1053/j.gastro.2011.01.001. Syngeneic human-immune and human-liver double reconstituted mice developed a human anti-HCV T cell response and liver fibrosis, however without detectable viremia.
- 113.Bility MT, Cheng L, Zhang Z, Luan Y, Li F, Chi L, Zhang L, Tu Z, Gao Y, Fu Y, et al. Hepatitis B virus infection and immunopathogenesis in a humanized mouse model: induction of human-specific liver fibrosis and M2-like macrophages. PLoS Pathog. 2014;10:e1004032. doi: 10.1371/journal.ppat.1004032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Rongvaux A, Willinger T, Martinek J, Strowig T, Gearty SV, Teichmann LL, Saito Y, Marches F, Halene S, Palucka AK, et al. Development and function of human innate immune cells in a humanized mouse model. Nat Biotechnol. 2014;32:364–372. doi: 10.1038/nbt.2858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Bukh J, Forns X, Emerson SU, Purcell RH. Studies of hepatitis C virus in chimpanzees and their importance for vaccine development. Intervirology. 2001;44:132–142. doi: 10.1159/000050040. [DOI] [PubMed] [Google Scholar]
- 116.Folgori A, Capone S, Ruggeri L, Meola A, Sporeno E, Ercole BB, Pezzanera M, Tafi R, Arcuri M, Fattori E, et al. A T-cell HCV vaccine eliciting effective immunity against heterologous virus challenge in chimpanzees. Nat Med. 2006;12:190–197. doi: 10.1038/nm1353. [DOI] [PubMed] [Google Scholar]
- 117.Lanford RE, Hildebrandt-Eriksen ES, Petri A, Persson R, Lindow M, Munk ME, Kauppinen S, Orum H. Therapeutic silencing of microRNA-122 in primates with chronic hepatitis C virus infection. Science. 2010;327:198–201. doi: 10.1126/science.1178178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Olsen DB, Davies ME, Handt L, Koeplinger K, Zhang NR, Ludmerer SW, Graham D, Liverton N, MacCoss M, Hazuda D, et al. Sustained viral response in a hepatitis C virus-infected chimpanzee via a combination of direct-acting antiviral agents. Antimicrob Agents Chemother. 2011;55:937–939. doi: 10.1128/AAC.00990-10. [DOI] [PMC free article] [PubMed] [Google Scholar]