Fig. 2.1
Comparison of the different in vitro and in vivo systems currently available for HBV research with respect to application and limitation
In 2002 a bipotent progenitor cell line, the HepaRG cells, that was derived from a liver tumor associated with chronic HCV infection, was found to become permissive for HBV infection after being cultured for some weeks under specific conditions promoting the enhancement of various hepatocyte-specific functions [31]. As observed in PHH cultures, the addition of dimethyl sulfoxide (DMSO), hydrocortisone and polyethylene glycol (PEG) is necessary to increase cell differentiation and hence the efficacy of HBV infection [32, 33]. The use of these cells offered new possibilities to study the infectivity of clinical HBV isolates and variants [34], as well as to explore the capacities of peptides derived from the preS-envelope protein of HBV to block the entry of both hepatitis B and Delta virus in vitro [35]. Moreover, cell entry was shown to occur in a polarized manner in these hepatocyte-like cells [13], whereas additional host factors appeared to mediate virus endocytosis . An in vitro study showed for instance that caveolin-1-mediated endocytosis is required for HBV entry in HepaRG cells [36]. HepaRG cultures were also shown to produce infectious HBV particles for more than 100 days and were successfully used for antiviral studies [37]. Using both transfected hepatoma cell lines and HepaRG cells, non-immunosuppressive cyclophilin inhibitors , such as alisporivir, have recently been shown to affect HBV replication and HBsAg production [38].
Since differentiated HepaRG cells represent a mixture of hepatocyte-like and biliary-like epithelial cells, only a subset of these cells becomes susceptible to HBV infection [13, 39]. It is also worth noting that no intracellular cccDNA amplification or spreading of HBV infection was observed in HepaRG cultures. This may be in part due to the inefficient and very slow conversion of the input relaxed circular DNA into cccDNA determined in these cells [39]. A newly described hepatoma cell line, named HLCZ01, was shown to be permissive for infection with both HBV and HCV clinical isolates [40]. Interestingly, sustained viral replication, for up to 90 days, without evidence of overt HBV and HCV interference could be shown.
A highly variable susceptibility to HBV infection is commonly observed in primary human hepatocyte (PHH) cultures [41, 42]. This may be in part due to divergences in host genetic susceptibility to infection, as well as to the loss of hepatic phenotype shortly after plating [11, 12]. To enhance susceptibility of HBV infection and virion productivity, Shlomai and colleagues showed that micropatterning and coculturing of primary human hepatocytes with stromal cells (MPCCs) maintained prolonged infection that could be further boosted by suppression of the innate immune responses. Moreover, to limit donor variability bias, the authors also obtained HBV infection using induced pluripotent stem cells differentiated into hepatocyte-like cells (iHeps) [43]. Since HBV permissiveness occurred in these cells in a differentiation-dependent manner, this system may be used to study host factors involved in HBV infection establishment and productivity.
Only recently, the discovery of the cellular receptor has opened new possibilities to investigate the initial steps of HBV infection in vitro [9]. In this elegant study, Li and colleagues used a synthetic peptide corresponding to the myristoylated N-terminus of the preS1 protein of the HBV envelope to identify the cellular receptor responsible for viral entry. By using photo-cross-linking and tandem affinity purification procedures, the authors showed that the preS1 peptide specifically interacts with a sodium taurocholate cotransporting polypeptide (NTCP), a transmembrane transporter exclusively localized to the basolateral membrane of high differentiated primary hepatocytes. It is indeed the rapid decrease of NTCP expression observed in cultured PHHs, as well as the lack of this receptor in most of the hepatoma cell lines that account for the rapid loss or lack of HBV susceptibility in these in vitro systems . NTCP mediates the transport of conjugated bile salts and of some drugs from the portal blood into the liver.
Based on the discovery that NTCP is the functional HBV entry receptor , hepatoma cell lines constitutively expressing the human NTCP gene have been created and have demonstrated the successful establishment of HBV infection in a significant proportion of NTCP-transfected hepatoma cells [9, 44, 45].
The availability of in vitro assays permitting investigation of the early steps of infection, as well as rapid screening of new anti-HBV agents, is expected to open new opportunities in HBV research. By using HBV-susceptible cells, it has been observed that DDX3, a member of the DEAD-box RNA helicase family, can affect cccDNA transcription [46]. A sophisticated study involving the use of both human and duck in vitro systems provided evidence that HBV utilizes the cell tyrosyl-DNA-phosphodiesterases (TDP1 and TDP2) to release the terminal protein of the polymerase from the rcDNA to initiate cccDNA biogenesis [47]. NTCP-transduced hepatoma cell lines are also expected to accelerate the acquisition of data revealing the interplay between HBV and host factors, as documented by studies showing that binding of the preS1 domain of the HBV envelope to NTCP inhibits its function [45]. Nevertheless, additional hepatocyte-specific factors appear to be involved in the HBV infection process, since infection rates and virion productivity are generally low in NTCP expressing cell lines. Intriguingly, the establishment of infection with the Hepatitis Delta Virus (HDV) but not productive HBV infection could be demonstrated in murine cells engineered to express the human NTCP [48]. Since both HBV and HDV use the same envelope proteins for cell entry, further species-specific differences or the lack of essential cellular factors within the murine hepatocytes may be responsible for such discrepancies [44, 49].
Primary hepatocytes isolated from Tupaia belangeri represents a good alternative to carry out in vitro infection studies with HBV [8]. It should be noted that the use of in vitro systems based both on primary tupaia hepatocytes and hepatoma cell lines have been fundamental even to identify the cellular receptor and other factors involved in the HBV attachment process [9, 50]. In general, being faster and more convenient than in vivo experiments, cell culture studies are very useful to carry out high throughput antiviral screening approaches and single cell level analyses in well-controlled experimental settings (Fig. 2.1). However, in spite of the existence of highly sophisticated primary hepatocyte-based systems, cultured cells may respond differently to infections and to other stimuli than cells in the intact liver [51]. Having lost the capability to express various hepatocyte-specific genes, discrepancies between data obtained in vitro and in vivo have to be considered. Thus, insights gained using cell-based in vitro systems need to be verified in in vivo systems.
Animal Models
The Duck Model
A major advantage of employing the DHBV model is that primary hepatocytes from ducklings or embryos are easily accessible and domestic Pekin ducks can be used under normal laboratory conditions. Furthermore, ducks show high infectivity rates in vivo [52] and generally reach high levels of DHBV replication and antigen expression. In vitro and in vivo studies with DHBV have contributed substantially to elucidate the replication mechanism adopted by the hepadnaviruses [1, 53–56] and mechanisms involved in the biogenesis of the cccDNA [47]. Furthermore, antiviral studies with polymerase inhibitors indicated that a stronger cccDNA reduction could be achieved in animals displaying higher cell proliferation rates [57], suggesting that hepatocyte turnover may destabilize the cccDNA pool in infected cells. Although various antiviral compounds have been tested in the duck model [58–61], these animals appear to be less sensitive to potential toxic effects than woodchucks [62]. Therefore, the results of antiviral drug screening might be of limited value for human HBV infection due to specific features of DHBV and the corresponding host hepatocytes. For instance, nucleocapsid inhibitors of the HAP family which can potently inhibit HBV replication appeared inactive on DHBV [63, 64]. It should be also kept in mind that the DHBV genome is smaller than HBV and is the most distantly related virus, since it shares little primary nucleotide sequence homology (40 %) with HBV. Moreover, DHBV uses the duck carboxypeptidase D (DCPD) and not like HBV the NTCP as receptor for viral entry and the infection is mostly not associated with liver disease and development of hepatocellular carcinoma (HCC) (Fig. 2.1).
The Woodchuck Model
Because of the higher similarities of WHV to HBV in terms of genomic organization, experimentally induced infection of woodchucks, the American Marmota monax, with WHV has been fundamental in the preclinical evaluation of antiviral drugs now in use for treatment of HBV infection [65–71]. Moreover, experimental infection of newborn woodchucks almost invariably leads to chronic infection, whereas animals infected at older ages generally develop acute hepatitis. Since acute and chronic WHV infection in woodchucks show serological profiles similar to those of HBV infection in humans, the woodchuck system has provided important insight about virological factors involved in the establishment of virus infection [2] and persistence [72]. Viral integrations, which frequently lead to proto-oncogene activation of the myc gene [73], are commonly found in woodchucks even after resolution of transient infection with WHV [74], while their frequency increases dramatically in chronically infected animals [5]. Interestingly, viral integrations were used as genetic markers to estimate the fate of infected hepatocytes and the amount of cell turnover occurring in the course of infection [5, 74]. These studies revealed the existence of cccDNA-free hepatocytes containing “traces” of the infection in form of viral integrations, thus indicating that cccDNA loss can occur also without destruction of the infected cells [5]. Since nearly all neonatally infected woodchucks develop hepatocellular carcinoma, this is the most used model of viral-induced HCC and has contributed to the development of new imaging agents for enhancement of detection of hepatic neoplasms by ultrasound and magnetic resonance imaging [73].
Numerous nucleoside analogues currently used in the clinic have been first assessed in woodchucks. Lamivudine, for instance, acted as a nontoxic antiviral drug in woodchucks and was shown to operate synergistically with interferon alpha [70, 75]. Using the woodchuck model, an antiviral activity comparable to lamivudine was reported for adefovir [67, 76, 77] and tenofovir [71], while drugs displaying higher antiviral efficacy, like entecavir, induced profound viremia reduction and a stronger delay of viral rebound after drug withdrawal [78]. Moreover, long-term suppression of WHV replication showed to delay the development of HCC [79]. Drug-resistant mutants have been also found after prolonged lamivudine treatment, while supplemental treatment with adefovir could restore viral suppression, thus mimicking clinical observations [73]. Therapeutic strategies involving the inhibition of PD-L1 [80] or gene therapy-based approaches enabling prolonged expression of IL-12 have been employed to study the efficacy of these drugs to break immunological tolerance [81, 82]. The characterization of the transcriptional response of these animals to WHV persistent infection, by performing sequencing of the woodchuck transcriptome and generation of custom woodchuck microarrays, indicated that chronic WHV infection, like CHB, is associated with a limited type I interferon response and induction of markers that, like in CHB infection, are associated with T cell exhaustion [83]. Improved sequencing information is expected to facilitate studies also aiming at developing therapeutic vaccines [84]. Regarding the screening of novel immune modulatory substances, the treatment of woodchucks with the oral TLR7 agonist GS-9620 has shown to induce sustained antiviral responses and even seroconversion in a substantial proportion of treated animals [85]. Although woodchucks are difficult to breed in captivity and they do not fulfill the requirements for an easy to handle experimental animal, the use of these outbred animals in HBV research plays an important role for the assessment of new immune therapeutic approaches (Fig. 2.1).
The Chimpanzee Model of HBV Infection [63]
The chimpanzee is the only immunocompetent experimental animal model fully susceptible to HBV infection, as demonstrated by the induction of acute infection and hepatitis after injection of serum from human HBV carriers [86]. Even though chimpanzees rarely develop chronic liver disease, they develop cellular immune responses largely resembling those observed in humans acutely infected with HBV [87]. Thus, researchers have relied upon chimpanzees to study the pathogenesis of acute HBV infection [88] and these high primates have played an essential role in the development of a safe vaccine, for the preclinical evaluation of HBV-specific monoclonal antibodies [89] and production of neutralizing HBV-specific antibodies [90–95]. The half-life of circulating HBV virions was also determined first in these animals [96]. Both protective immunity and the efficacy of the hepatitis B vaccine against antiviral drug-resistant HBV mutants have been assessed by rechallenging the chimpanzees either with homologous or heterologous viruses [95]. Because sequential liver biopsies can be obtained throughout the course of infection, chimpanzees represent an extremely valuable infection system for the analysis of intrahepatic virological changes and immune responses (Fig. 2.1). Such studies revealed for instance that non-cytolytic downregulation of HBV replication may play an important role in HBV clearance, because most HBV-DNA could be cleared from the liver and the blood of experimentally infected chimpanzees before T-cell infiltration and liver injury occurred [97]. It was also shown that hepatocellular injury is predominately immune mediated [98] and that a strong and polyclonal CD8 T cell response to HBV proteins characterizes the acute self-limited HBV infection [99]. Moreover, the depletion of CD4 T cells indicated that these immune cells do not directly participate in viral clearance, but rather contribute to the induction and maintenance of B and CD8 T cell responses [100]. Interestingly, low levels of cccDNA were shown to persist in the liver of chimpanzees even after resolution of infection [101], while the presence of HBV DNA integrates could demonstrate the clonal expansion of hepatocytes in livers of chronically infected chimpanzees [102]. Infection kinetic and microarray analyses of serial liver biopsy samples obtained from experimentally infected chimpanzees revealed that HBV does not induce significant changes in the expression of intrahepatic innate response genes and production of type I IFNs in the first weeks following HBV infection and spreading [101]. The relevance of these studies could be confirmed in CHB patients [103]. HBV infection studies in chimpanzees also indicated that the size of the inoculum affects not only the kinetics of viral spread but also the outcome of infection, since injections with few virions were associated with a strong CD4 T-cell priming delay and development of persistent HBV infection [104]. A recent example of preclinical assessment of antiviral drugs in chronically infected chimpanzees regarded the evaluation of the therapeutic efficacy of TLR7 agonists, where oral administration of this immune modulatory compound reduced viral loads and induced enhancement of antiviral immune responses, such as elevation of interferon stimulated genes (ISGs) in liver and blood [105]. Establishment of HBV infection also appeared to be limited by the antiviral effects and enhancement of type I interferon responses that HCV induced in the liver of chimpanzees previously infected with HCV [106].
Although chimpanzees are a uniquely valuable species for research with human hepatotropic viruses, the strong ethical constraints and high costs encountered by working with primates have increasingly restricted their use (Fig. 2.1). Moreover, the extremely limited number of chimpanzees available for research represents a serious restriction for the evaluation of antiviral drug efficacy, while recent advances in alternate research tools, including cell-based and other animal models, are further reducing the necessity to use chimpanzees as research subjects [107–109].
The Tupaia Model
Inoculation of tree shrews with HBV-positive human serum was shown to result in a transient HBV infection, characterized by low levels of viral replication and production of antibodies to HBsAg and HBeAg [8]. Moreover, HBV virions produced in tupaias were successfully passed through several generations and infection of these animals could be specifically blocked by immunization with hepatitis B vaccine [110]. Infection of adult tupaias causes only a mild, transient infection with low viral titers. Recent data suggest that infection of neonate animals with HBV is inducing chronic infection. Although viremia reached only moderate levels (up to one million HBV DNA copies/ml), immunopathologic changes in the liver of long-term infected animals, including forms of bridging necrosis and fibrosis were observed [111]. It is however worth noting that even if experimental infection of tree shrews causes only a very mild and transient infection in these animals, primary hepatocytes isolated from T. belangeri can be efficiently infected in vitro (Fig. 2.1) and hence represent a valuable alternative source of HBV-permissive cells to study the early steps of HBV infection [7, 9]. Cultures of primary tupaia hepatocytes have been successfully used to show that polymerase inhibitors like adefovir and lamivudine can reduce but not prevent the formation of cccDNA upon hepatocyte infection [112]. Moreover, the woolly monkey hepatitis B virus (WMHV) was isolated from an endangered new world primate [113], the woolly monkey Lagothrix lagotricha. Phylogenetic analysis of the nucleotide sequences of WMHBV indicated that this virus may represent a progenitor of the human virus [113]. Interestingly, WMHBV is not infectious for chimpanzees [113], but primary tupaia hepatocytes are highly susceptible to infection with this HBV-closely related hepadnavirus [114, 115].
Mouse Models
Mouse Models of HBV Replication
Transgenic Mice
To investigate specific aspects of HBV replication, as well as the role of distinct viral proteins in HBV pathogenesis using convenient inbred animal models, embryo microinjection technologies have enabled the development of mice harboring either single HBV genes or terminally redundant over-length HBV-DNA constructs [116–120]. The first HBV-replicating transgenic mice, which were developed by Chisari and colleagues in 1995 [119], demonstrated the feasibility to produce in murine hepatocytes infectious HBV virions morphologically indistinguishable from human-derived virions [97]. As the immune system of transgenic animals recognizes during e mbryonic development the virus as “self,” these studies provided the first evidence that HBV replication does not induce hepatocellular injury [121]. To show that HBV-related pathogenesis is largely mediated by the host immune responses, induction of acute hepatitis and hepatocellular injury was demonstrated after adoptive transfer of HBV-antigen specific CTLs [122–124]. CTL-mediated release of cytokines also showed to suppress viral replication by non-cytolytic mechanisms [125, 126], while the recruitment of antigen nonspecific inflammatory cells amplified the severity of liver damage initiated by antigen-specific CTLs [127]. CD8+ T cells isolated from mice and engineered to express HBV specific chimeric antigen receptors (S-CARs) w ere also shown to engraft and expand in immunocompetent HBV-transgenic mice. After adoptive transfer, these cells were shown to control HBV replication, while causing only transient liver damage. Since these effector T cells, can be developed regardless of their HLA type, the adoptive transfer of such genetically modified HBV-specific T cells may represent a promising immunotherapeutic approach deserving further investigations [128, 129].
HBV transgenic mice have been successfully used to evaluate the impact of various polymerase inhibitors, such as lamivudine [130], adefovir dipivoxil and entecavir [131, 132] on HBV replication. Therapeutic approaches involving the use of HBV-specific small interfering RNAs (siRNAs ) were also tested in HBV-transgenic mice [133, 134]. To combine gene silencing with the induction of interferon responses, Protzer and colleagues recently employed 5′-triphosphorylated small interfering RNAs targeting highly conserved sequences on HBV RNA transcripts and showed that by triggering RIG-I-mediated innate immune responses these bifunctional antiviral molecules suppressed HBV replication more efficiently than siRNAs lacking a triphosphate group [135].
A major limitation encountered by using HBV transgenic mice is that they do not allow investigation of viral entry and spreading (Fig. 2.1). Moreover, no cccDNA is built in the liver of transgenic mice and the chromosomally integrated HBV genome cannot be eliminated from the host genome. As a consequence, viral clearance and cccDNA eradication cannot be achieved in this model. To break tolerance to HBV antigens and investigate the mechanisms of viral clearance, alternative mouse models have been developed which rely on transfecting or transducing the viral genome into mouse hepatocytes by different means, such as using recombinant adenoviral vectors , or by hydrodynamic injection of naked DNA in mice.
Vector-Mediated Transfer of HBV Genome
Adenoviral vectors containing a replication-competent HBV genome (Ad-HBV) have been shown to permit efficient transfer of the HBV genome into the liver of immunocompetent mice [136]. After intravenous injection of such adenoviral derived vectors, HBV proteins are produced under the control of endogenous HBV promoters and enhancers and viral replication could be demonstrated for up to 3 months. Viral clearance was accompanied by mild to moderate liver inflammation with elevated serum alanine transaminase activities [137]. After the induction of adaptive immune responses, anti-HBs seroconversion and development of neutralizing antibodies the infection was cleared. Inflammation and liver damage were also shown to be controlled by regulatory T-cells [138]. Because of the acute, self-limiting character of such adenoviral-mediated HBV infection, the system offers good possibilities to investigate the mechanisms of immune-mediated viral clearance also involving intrahepatic expansion of cytotoxic T cells (CTL) and NKT cells [139, 140]. By injecting relative low doses of adenoviral vectors [141] or by injecting the viruses intrahepatically into neonatal mice [142], persistent infection in immunocompetent mice could be established. This type of tolerance resembles immunological features of chronic HBV infection in humans. Moreover, even if mice do not establish cccDNA, using adenoviral vectors the HBV genome is in an extrachromosomal organization and hence its clearance can be achieved. Recombinant adeno-associated viruses (AAV) were also used to transfer replication-competent HBV genomes in a mouse strain carrying human leukocyte antigen A2/DR1 transgenes [143]. In these animals, viremia and antigenemia persisted for at least 1 year. Notably, a higher number of regulatory T-cells and no significant liver inflammation were determined in those livers, while impairment of functional T cell responses indicated the occurrence of tolerance. However, establishment of long-term viral replication with Ad-HBV vectors is limited by the immune responses against these vectors and occurrence of adenovirus-mediated cytotoxic effects may also limit their application. Since a functional cccDNA, associated with histone and non-histone proteins is not built in murine hepatocytes, these models are not suitable for the development of drugs or antiviral strategies targeting the natural template of HBV transcription and replication (Fig. 2.1).
Hydrodynamic Injection of HBV Genome
Hydrodynamic injection techniques , which involve the rapid injection of a large volume (10 % of the animal weight) of a solution containing naked DNA into the tail vein of mice, are quite stressful for the animals but allow crossing species-specific barriers and permit efficient HBV DNA transfer [144]. Moreover, the rapid injection of liquid induces significant liver damage and ALT elevation shortly after injection. Hydrodynamic injection of replication-competent HBV genomes in mice resulted in viremia titres up to 1 × 107 HBV DNA/ml [144]. Although HBV replication initiated already 1 day post-injection, replication levels decreased after 1 week and HBV was cleared from blood within 2–3 weeks, as soon as specific antiviral antibodies and CD8+ T cells appeared. However, HBV infection persisted for 3 months after hydrodynamic injection of mice lacking adaptive immune cells and natural killer cells, thus demonstrating that the outcome of hydrodynamic transfection of HBV depended on the host immune response [144]. Hydrodynamic injection studies also showed that simultaneous delivery of HBV expressing plasmids and HBV-specific siRNAs prevented HBV replication [133, 134], while by injecting modified HBV DNA plasmids into C57BL/6 mice, a significant immune clearance of HBV could be achieved [145]. A recent report showed that the use of a lentiviral backbone instead of an AAV vector led to increased and prolonged HBV replication (>56 days post injection) [146]. In an attempt to generate cccDNA-like molecules in mice, a monomeric HBV genome precursor plasmid (pr-cccDNA), that can be converted by Cre/loxP-mediated DNA recombination into a 3.3-kb cccDNA, has been recently used [147]. Although such recombinant cccDNA could be detected in the nuclei of murine hepatocytes, the induced immune response rapidly limited viral replication in vivo [147].
To investigate whether cccDNA molecules could be directly targeted for destruction, the CRISPR/Cas9 system has recently been employed both for in vitro [148, 149] and in vivo studies, using the HBV-hydrodynamic-mouse models [150]. In these studies, the levels of HBV-expressing vectors and different markers of viral replication were significantly reduced, and without evidence of toxicity, suggesting that the CRISPR/Cas9 system could be recruited to the HBV-expressing vectors. Although many issues regarding the efficiency and safety of the system remain to be addressed, these findings are the first to demonstrate nuclease-mediated disruption of a HBV expressing vector, as a model of cccDNA, thus opening new possibilities for the development of innovative antiviral strategies aimed at disrupting the cccDNA.
Despite the relatively short span of viral replication available, mice transfected by hydrodynamic injection are suitable not only for short-term antiviral studies but also for testing the consequences of specific mutations within the viral genomes. In comparison to HBV-transgenic mice, hydrodynamic-based procedures permit investigation of immune response emerging during acute infection. Since the viral genome is not integrated into the host genome as a transgene, viral clearance commonly occurs in these systems. A clear advantage of the system is that different HBV genotypes and variants can be injected into mice and analyzed in vivo in relatively short time (Fig. 2.1). Nevertheless, the rapid injection of large volume of fluids causes not only strong discomfort to the animals but also significant damage in the liver, which may alter cell function and signaling analyses. Since reinfection of the mouse hepatocytes is not possible, viral clearance is easier to achieve in these non-transgenic murine models than in humans, and therefore, the efficacy of antiviral treatments should be validated in systems permissive for HBV infection. Interestingly, all results available so far indicate that murine cells engineered to express the human NTCP do not become susceptible for HBV infection [44]. It appears that species-specific differences or the lack of cellular factors involved in post-entry steps are responsible for these discrepancies.
Mouse Models of HBV Infection
The inability to reproduce the entire infection cycle of HBV in murine hepatocytes has hindered our understanding of the mechanisms by which HBV interacts with hepatocyte-specific functions, as well as to study mechanisms of viral entry, cccDNA formation and spreading in a well-controlled in vivo laboratory system . Because of these restraints, many efforts have concentrated on the development of models based on the use of the natural target of HBV infection: the human hepatocyte. However, the susceptibility of cultured primary human hepatocytes to HBV infection is highly variable and cultured hepatocytes rapidly lose the expression of essential hepatocyte-specific factors, such as the NTCP receptor [9]. The generation of mice carrying human hepatocytes permits to overcome most of these limitations.
The Trimera Mouse
These mice represented the first human–mouse chimeric system that was developed by transplanting human liver fragments under the kidney capsule of normal Balb/c mice. To avoid rejection of the implanted tissue, these mice were preconditioned by total body irradiation and reconstituted with SCID mouse bone marrow cells [151]. After ex vivo infection of the small human liver specimens with HBV, low levels of viremia, that remained detectable for approximately 20 days, could be determined in implanted animals. Interestingly, mice ectopically carrying human liver fragments could also be engrafted with human peripheral blood mononuclear cells (BPMC), so that the effects of polyclonal anti-HBs antibodies against HBV could be assessed [89, 152]. Nevertheless, due to the extra-hepatic location of the implanted tissues, human hepatocytes remained functional only for limited time and in vivo infection with HBV or other human hepatotropic viruses could not be established.
Human Liver Chimeric Mice
To achieve long-term survival of primary human hepatocytes permissive for HBV infection in vivo, isolated cells need to be integrated in the mouse liver. The requirements to achieve this goal are (1) the damage of the endogenous murine hepatocytes to create the space and the regenerative stimulus necessary for transplanted hepatocytes to reconstitute the diseased mouse liver; and (2) the absence of murine adaptive immune cells and NK cells to permit engraftment and survival of transplanted xenogeneic hepatocytes. Different human-liver chimeric mouse models are currently available. The Alb-urokinase-type plasminogen activator (uPA) transgenic mouse was the first model describing the strong regeneration capacity of healthy transplanted hepatocytes. In this system, over-expression of the hepatotoxic uPA transgene, which is driven by the mouse albumin promoter, induces high levels of uPA in plasma, hypo-fibrinogenemia and subacute liver failure in young mice [153]. To generate mice with human liver chimerism, uPA transgenic mice have been backcrossed with immunodeficient mouse strains, such as the RAG2−/− [154–156], the Severe Combined Immune Deficient (SCID) [157], which lack functional B and T cells or SCID/beige mice, which also lack NK-cell functions (shortly USB mice ) [158, 159] and NOD/SCID/gamma(c)(null) (shortly uPA-NOG) [178]. Following intra-splenic injection of one million freshly isolated or cryopreserved-thawed human hepatocytes, the transplanted cells migrate via the splenic and portal veins to the liver, where cells integrate into the liver parenchyma. Engrafted human hepatocytes proliferate to form larger regenerative nodules that eventually merge together to replace the diseased liver parenchyma. Reconstitution of the mouse liver takes around 2 months and the levels of human chimerism can be estimated by determining the concentration of human serum albumin in mouse blood [159, 160]. Within the mouse liver, the transplanted human hepatocytes maintain normal metabolic functions [157, 161]. To delay the production of the toxic transgene, which makes transplantation procedures necessary in the first month of life, alternative mouse models, where the expression of the uPA transgene is inducible [162] or is regulated by the MUP (major urinary protein) promoter, have been also developed [163]. To generate an animal model where hepatocyte failure can be induced at will in adult mice, alternative human-liver chimeric mice, based on the use of fumaryl acetoacetate hydrolase-deficient (FAH) mice, were established [164, 165]. FAH plays a crucial role in the tyrosine breakdown pathway and its deficiency leads to accumulation of toxic tyrosine catabolites and liver failure. However, accumulation of these catabolites can be avoided by administering the drug NTBC (2-(2-nitro-4-trifluoromethylbenzyol)-cyclohexane-1,3-dione), a pathway inhibitor that protects the animals from the occurrence of liver injury until drug administration is withdrawn. More recently, by transplanting higher amounts of human hepatocytes, or by performing repeated hepatocyte transplantation , high rates of human hepatocyte chimerism could be achieved in mice also lacking the Rag2 and the gamma-chain of the receptor for IL-2 genes (shortly FRG mice ) [166]. Human hepatocytes were also successfully transplanted in mice expressing the herpes simplex type-1 thymidine kinase (TK) transgene that were backcrossed with NOG (NOD/SCID/gamma(c)(null)) mice [167]. Even in this case, human hepatocyte transplantation can be performed in adult mice, since mouse liver cells expressing the TK-transgene can be selectively destroyed upon administration of ganciclovir [168].
From the first successful transplantation of human hepatocytes into uPA/RAG2 mice and establishment of de novo infection with HBV [156], several groups performed HBV infection studies [160, 169, 170], as well as demonstrated infection with other human hepatitis viruses , like HCV and HDV, in humanized uPA/SCID or uPA/SCID/beige (USB) mice [160, 171–173]. Notably, after intra-peritoneal inoculation of HBV infectious serum or cell culture derived virions, productive HBV infection, which requires the establishment of a functional cccDNA in hepatocyte nuclei, is first achieved in a minority of human hepatocytes and several weeks are needed to accomplish viral spreading [159]. After that, nearly all human hepatocytes stain HBcAg-positive and viremia reaches a stable plateau which, to a certain extent, correlates with the levels of human chimerism. The use of patient serum samples as virus inoculum allows the functional analysis of distinct HBV genotypes, naturally occurring variants and drug-resistance mutants. Genetically engineered viruses can therefore be used to investigate the role of distinct viral proteins in human infected hepatocytes. Using this type of approach, the expression of the regulatory HBx protein provided in trans was shown to be essential for cccDNA-driven HBV replication in infected human hepatocytes [170]. Studies focusing on investigating how HBV may affect cellular pathways [161] and innate immune responses of the human hepatocytes are just starting to emerge and showed that expression of metabolic genes [161] and innate immune response genes in these cells resembles well the expression pattern determined in human livers [158]. To this regard, a recent study indicated that binding of HBV to its cellular receptor alters the hepatocellular uptake of bile salts and the expression profile of genes of the bile acid metabolism [161]. The occurrence of such alterations was also confirmed in patient biopsy samples. Using humanized mice , both HBV and HCV have been recently shown to contribute to the induction and accumulation of aberrant DNA methylation in human hepatocytes through the activation of NK-cell-dependent innate immune responses [174].
Both the antiviral activity of clinically approved polymerase inhibitors [115, 158, 169] and the in vivo efficacy of novel polymerase or capsid inhibitors [175, 176] have been assessed in human liver chimeric mice. The model also served to evaluate the in vivo efficacy of lipopeptides derived from the large envelope protein of HBV to prevent de novo HBV [177] and HDV infection [172], as well as to investigate the ability of the most clinically advanced entry inhibitor, Myrcludex-B, to block HBV spreading post-infection [178]. Moreover, the serial passage of infected hepatocytes isolated from infected mice and transplanted into new recipients has permitted to gain insight about the impact of hepatocyte proliferation on cccDNA stability and activity in vivo [179]. The drastic reduction of intrahepatic cccDNA loads induced by cell division even in the absence of antiviral therapy revealed a weak point in HBV persistence that deserves further investigations. Thus, these systems offer unique opportunities to investigate factors that can affect the stability and/or activity of the cccDNA minichromosome, as well as the direct antiviral effects of cytokines and immune modulatory substances, such as interferons (Fig. 2.1). To this regard, human liver chimeric mice have been used to assess whether HBV can circumvent the induction of antiviral defense mechanisms [158]. Upon administration of regular human IFNα, HBV was shown to hinder the nuclear accumulation of STAT-1, thus providing a potential mechanism for the reduced induction of interferon stimulated genes (ISGs) determined in HBV-infected human hepatocytes [158]. On the other hand, studies in these mice also showed that IFNα can mediate epigenetic repression of the cccDNA minichromosome [29], while the repeated administrations of the longer-active pegylated IFNα was shown to be able to breach the impairment of HBV-infected hepatocyte responsiveness and induce sustained enhancement of human interferon stimulated genes (ISG) [180]. In this study, the stronger antiviral effects of peg-IFNα exerted on the human hepatocytes were shown to trigger a substantial decline of circulating HBsAg and HBeAg levels in chimeric mice, without claiming the involvement of immune cell responses [180]. Moreover, the comparative analyses of the innate immune responses revealed that type I, II and III IFNs are differently induced in murine and human hepatocytes and that the effects of distinct IFNs may differ between animal species, thus underlying the importance of validating results obtained in murine systems also by employing primary human hepatocytes [181].
Immune Competent Human Liver Chimeric Mice
Human hepatocytes can be very abundant within the chimeric livers but non-parenchymal cells, such as sinusoidal endothelial cells and Kupffer cells, are of murine origin. As a consequence, development of fibrosis and pathophysiologic processes that are commonly associated with chronic viral hepatitis infections but involve a cross talk between the hepatocytes and other liver resident cells are not observed in the above mentioned systems. Moreover, since these chimeric animals are genetically immune deficient they are not suited for vaccine studies and for evaluation of adaptive immune responses. To circumvent these limitations, partially haplotype-identical human peripheral blood mononuclear cells (PBMC) have been recently transferred in uPA/SCID chimeric mice after the establishment of HBV infection [182]. Notably, infiltrating human immune cells caused severe hepatocyte degeneration, while treatment with anti-Fas antibodies or depletion of dendritic cells prevented the death of human hepatocytes.
A different attempt to generate a humanized mouse model harboring both human liver cells and a human immune system was the development of the AFC8 model [183]. These mice were obtained by crossing BALB/c-RAG2−/−γc−/− mice, which lack functional B, T and natural Killer cells, with mice carrying a liver-specific suicidal transgene with inducible activity based on the induction of caspase 8 in mouse hepatocytes. These animals were used to transplant simultaneously human fetal hepatocytes and hematopoietic stem cells that were obtained by the digestion of human fetal liver tissues (15–18 weeks of gestation period). Since the fetal liver provides both types of cells, reconstitution of both cell lineages is syngeneic and hepatocyte rejection by the human immune system is not expected. Although HBV infection studies were not performed, after inoculation of hepatitis C virus, low levels of intrahepatic viral replication, as well as T-cell responses and development of fibrosis could be determined. Despite partial success and the ethical restrains encountered by employing human fetal tissues, the generation of chimeric systems equipped with both a human liver and a functional human immune system, with a matched major histocompatibility complex, still remains a major challenge. The latest advances reported by Gutti et al. showed the feasibility to reconstitute an uPA-NOG mouse strain with both adult human hepatocytes and hematolymphoid cells [184]. In this study, dual reconstitution was achieved by transplanting fetal or even adult hepatocytes and mismatched hematopoietic stem cells (CD34+ HSCs) derived from either a fetal liver or umbilical cord blood. As an alternative and well-tolerated procedure to total body irradiation, mice received 3 days of a treosulfan-based chemotherapeutic conditioning before HSC injection. The presence of CD8+ and CD4+ human cells and of human hepatocyte clusters was observed in the liver of these animals. No cell-mediated rejection but also no evidence of cell interactions were determined in animals reconstituted with mismatched HSCs. Although the applicability of these approaches for the study of HBV and HDV associated immune pathological processes needs further research, these technical advances have paved the way for the development of dually reconstituted humanized systems.
Conclusions
In the recent years fundamental progresses were made concerning the development of both in vitro and in vivo systems that offer new opportunities to researchers to choose between different models on the base of the specific questions addressed. The recent discovery of the long-searched cellular receptor for HBV was a milestone that has permitted the creation of HBV permissive hepatoma cell lines which can recapitulate the entire HBV life cycle. Together with the HBV permissive cell line HepaRG and the new described cell line HLCZ01 these tools shall allow high-throughput screening of large compound collections for innovative substances interfering with the different steps of the life cycle. In the light of the success of direct antiviral substances in HCV field, it can be expected that new direct antiviral substances targeting different steps of the life cycle will arise in the near future. Nevertheless, and in contrast to HCV infection, HBV persistence is guaranteed by the stable episomal cccDNA minichromosome . Thus, the task of curing HBV infection will require development and testing of new substances targeting key steps of HBV life cycle, such as viral entry [177], cccDNA formation [47], stability [185] and activity [29], as well as enhancing immune responses. Since the maintenance of hepatocyte specific functions that are often lost in hepatoma cell lines appears fundamental to identify the factors involved in the infection process, it can be expected that the use of convenient hepatoma cell lines culture systems will keep bearing important limitations. Thus, further development of more sophisticated in vitro HBV infection systems, possibly based on the use of engineered nonhuman hepatocyte-like cells or humanized murine hepatocytes, appears mandatory.
Regarding the in vivo models of HBV infection, the classical reference was the chimpanzee. However, due to stronger ethical restrains this model is practically not available for HBV research and needs to be replaced by different older and newer systems. As a consequence, important research progresses focusing, for instance, on identifying host factors involved in cccDNA formation mechanisms still relay on the use of duck and tupaia hepatocytes, while the woodchuck system, despite its known virological and host-related differences, still offers unique advantages to accomplish preclinical vaccination studies and testing of immune modulatory substances. Human liver chimeric mice represent already a well establish HBV infection model for preclinical in vivo testing of direct antiviral agents and for studying how human hepatitis viruses interact with their natural target of infection, the human hepatocyte. Moreover, co-transplantation of human immune cells into these mice may further brighten their use by allowing the study of human immune responses to HBV. Ideally, immune competent HBV-permissive murine models may fulfill most of the unmet needs in preclinical research. Nevertheless, species-differences both in term of pharma cokinetics and immune responses may be misguiding and it can be expected that all systems will keep having specific advantages and limitations, depending on the research purpose. Thus, validation of results in multiple systems should always be encouraged.
References
1.
Mason WS, Aldrich C, Summers J, Taylor JM. Asymmetric replication of duck hepatitis B virus DNA in liver cells: free minus-strand DNA. Proc Natl Acad Sci U S A. 1982;79(13):3997–4001.PubMedCentralPubMed