Anda di halaman 1dari 11

Journal of Immunological Methods 410 (2014) 3949

Contents lists available at ScienceDirect

Journal of Immunological Methods


journal homepage: www.elsevier.com/locate/jim

Review

Mouse models of hepatitis B and delta virus infection


Maura Dandri a,b,, Marc Ltgehetmann a,c
a
I. Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
b
German Center for Infection Research, HamburgLbeckBorstel Partner Site, Germany
c
Institute of Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany

a r t i c l e i n f o a b s t r a c t

Article history: Liver disease associated to persistent infection with the hepatitis B virus (HBV) continues to be a
Received 29 January 2014 major health problem of global impact. Therapeutic regimens currently available can efficiently
Received in revised form 3 March 2014 suppress HBV replication; however, the unique replication strategies employed by HBV permit
Accepted 4 March 2014 the virus to persist within the infected hepatocytes. As a consequence, relapse of viral activity is
Available online 12 March 2014
commonly observed after cessation of treatment with polymerase inhibitors. Among the HBV
chronically infected patients, more than 15 million patients are estimated to be co-infected with
Keywords: the hepatitis delta virus (HDV), a defective satellite virus that needs the HBV envelope for
Transgenic mice propagation. No specific drugs are currently available against HDV, while nucleos(t)ide analogs
Human liver chimeric mice
are not effective against HDV replication. Since chronic HBV/HDV co-infection leads to the most
Preclinical HBV/HDV infection studies
severe form of chronic viral hepatitis in men, a better understanding of the molecular mechanisms
of HDV-mediated pathogenesis and the development of improved therapeutic approaches is
urgently needed. The obvious limitations imposed by the use of great apes and the paucity of
robust experimental models of HBV infection have hindered progresses in understanding the
complex network of virushost interactions that are established in the course of HBV and HDV
infections. This review focuses on summarizing recent advances obtained with well-established
and more innovative experimental mouse models, giving emphasis on the strength of infection
systems based on the reconstitution of the murine liver with human hepatocytes, as tools for
elucidating the whole life cycle of HBV and HDV, as well as for studies on interactions with the
infected human hepatocytes and for preclinical drug evaluation.
2014 Elsevier B.V. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
1.1. HBV infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
1.2. HDV infection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
2. Mouse models of HBV replication . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
2.1. Transgenic mice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
2.2. Vector-mediated transduction of HBV in mice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
2.3. Hydrodynamic injection of replication competent HBV or HDV genomes . . . . . . . . . . . . . . . . . . . . . . . 42
3. Chimeric mouse models of HBV and HDV infections . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 42
3.1. Preclinical studies . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 45
3.2. Immune competent human liver chimeric mice . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46

Abbreviations: HBV, hepatitis B virus; HDV, hepatitis delta virus; cccDNA, covalently closed circular DNA; HBsAg, HBV surface antigen; HBcAg, HBV core
antigen; HDAg, Hepatitis delta antigen.
Corresponding author at: I. Dept. Internal Medicine, University Medical Center Hamburg-Eppendorf, Martinistr. 52, 20246 Hamburg, Germany. Tel.: +49 40
7410 52949; fax: +49 40 7410 57232.
E-mail address: m.dandri@uke.de (M. Dandri).

http://dx.doi.org/10.1016/j.jim.2014.03.002
0022-1759/ 2014 Elsevier B.V. All rights reserved.
40 M. Dandri, M. Ltgehetmann / Journal of Immunological Methods 410 (2014) 3949

Acknowledgment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 46

1. Introduction studies reported the successful establishment of HBV infection


in a significant proportion of NTCP-transfected HepG2 cells
1.1. HBV infection (Yan et al., 2012; Nkongolo et al., 2013; Yan et al., 2013).
Although large amounts of input viruses (MOI N 1000) are still
Characteristic of the hepatitis B virus (HBV), the prototype necessary to achieve HBV infection in these culture systems,
member of the Hepadnaviridae family, is its high tissue- and the availability of in vitro assays permitting investigation of the
species-specificity, as well as a unique genomic organization early steps of infection, as well as rapid screening of new
with asymmetric mechanism of replication (Nassal, 2008). anti-HBV agents, is expected to open new opportunities in
Because of the narrow host range, HBV infects exclusively HBV research. Nevertheless, despite the importance of such
humans, but can be used to experimentally infect chimpan- discovery, additional as-yet-unknown factors may be in-
zees. Since mice and rats are not permissive for any of the volved in the HBV infection process, since the production
known Hepadnaviridae, most of the advances in HBV of limited amounts of HBV particles has been observed.
research have been based on infection studies performed in Intriguingly, murine cells (both hepatoma and primary
chimpanzees and by using infection experiments with hepatocytes) engineered to express the human NTCP have
HBV-related hepadnaviruses (such as woodchuck hepatitis so far failed to permit the establishment of productive HBV
virus and duck hepatitis virus) and their respective hosts infection. Whether species-specific differences or the lack of
(Mason et al., 1980; Roggendorf and Tolle, 1995). These essential cellular factors within the murine hepatocytes are
studies have been essential for understanding various steps responsible for these discrepancies needs further investigations.
of the viral life-cycle (Mason et al., 1982) and experiments
performed with the American woodchuck have provided 1.2. HDV infection
important insights on the processes involved in infection
persistence and hepatocarcinogenesis (Zoulim et al., 1994; Among the HBV chronically infected patients, more than
Dandri et al., 1996; Summers and Mason, 2004; Mason et al., 15 million patients worldwide are co-infected with the
2005). Based on the close phylogenetic relationship between hepatitis delta virus (HDV) (Hughes et al., 2011), a defective
primates and tree shrews (von Weizsacker et al., 2004; virus, known to lead to the most aggressive form of chronic
Baumert et al., 2005), the tree shrew species Tupaia belangeri viral hepatitis, since the infection is generally associated with
has been frequently used for infection studies both with HBV an accelerated course of fibrosis progression, cirrhosis, and the
and, more often, with the woolly monkey hepatitis B virus highest risk for liver decompensation and death (Rizzetto,
(WMHV) (Walter et al., 1996). However, because of the 2009; Hughes et al., 2011). HDV is a small single-stranded RNA
restraints encountered using chimpanzees and animal virus which hijacks the host DNA-directed RNA polymerase to
models based on HBV-related viruses, it is not surprising propagate its viral genome in a RNA-directed RNA replication
that researchers focused on the development of more process. Within the hepatocyte, replication leads to accumula-
sophisticated systems based both on the generation of tion of three HDV RNAs: the circular genomic RNA, a
mouse models of HBV replication by using transduced and complementary antigenomic RNA, and a smaller linear
transgenic mice harboring over-length HBV-DNA genomes, mRNA, which is the template for the translation of the only
and on the development of mice carrying human chimeric known viral protein, the hepatitis delta antigen (HDAg). There
livers. The latter permits to perform in vivo infection studies are two HDAg isoforms: a small 24 kDa isoform, which is
and to investigate the complete HBV replication cycle in the essential for virus replication, and a large 27 kDa variant,
natural target of infection: the human hepatocyte. Never- necessary for virus morphogenesis, which is generated via a
theless, important steps of the infection and pathobiology of post-transcriptional RNA-editing event. Moreover, HDV needs
HBV have not been clarified. Only recently the discovery of the surface proteins of HBV to generate infectious viral particles
the cellular receptor has opened up new possibilities to and hence to complete its life-cycle (Sureau, 2006). As a
investigate specific steps of the life-cycle in vitro (Yan et al., consequence, infectious HDV virions can only be released from
2012). In this study, Li and colleagues used a synthetic hepatocytes that are co-infected with HBV. No drugs are
peptide corresponding to the myristoylated N-terminus of the currently available specifically targeting HDV and various
HBV preS1 protein to identify the candidate virus receptor from clinical studies have shown that nucleos(t)ide analogs are not
hepatocyte cultures. By using methods called zero distance effective against HDV replication (Hughes et al., 2011). This is
photo-cross-linking and tandem affinity purification, it was not surprising, since polymerase inhibitors efficiently suppress
shown that the preS1 peptide of the HBV envelope specifically HBV replication without affecting the transcriptional activ-
interacts with a sodium taurocholate cotransporting polypep- ity of the HBV covalently closed circular DNA (cccDNA)
tide (NTCP), a multiple transmembrane transporter localized minichromosome, from which all RNA transcripts and HBV
to the basolateral membrane of highly differentiated primary surface proteins (HBsAg) that are needed for HDV assem-
hepatocytes. NTCP mediates the transport of conjugated bile bly, are produced. Furthermore, the lack of robust HBV/HDV
acids and some drugs from the portal blood to the liver. Based infection models permitting investigation of the full
on the discovery that NTCP serves as HBV entry receptor, life-cycle of HDV has hampered our understanding of the
cell lines susceptible to HBV infection are emerging and first HDV biology, of the consequences of HDV co-infection for HBV
M. Dandri, M. Ltgehetmann / Journal of Immunological Methods 410 (2014) 3949 41

replication, as well as the development of more efficient Although HBV transgenic mice provided useful information
therapeutic strategies. related to the mechanisms of HBV pathogenesis, there are
important limitations by using these models that need to be
2. Mouse models of HBV replication taken into account. First, the infection step cannot be studied in
transgenic mice, since murine hepatocytes do not allow
2.1. Transgenic mice investigation of viral entry and spreading steps. Whether the
generation of mice expressing a human-specific cellular
The requirement of convenient inbred animal models to receptor (h-NTCP) will enable the establishment of HBV
investigate specific aspects of HBV replication and its oncogenic infection in mice is currently not clear. Most importantly, in
potential in vivo led to the development of transgenic mice transgenic mice all viral RNAs are synthesized from a
harboring either single viral genes or a terminally redundant chromosomally integrated copy of the viral genome and, for
over-length 1.3 HBV-DNA construct (Chisari et al., 1985; Araki unknown reasons, no cccDNA, the natural template of viral
et al., 1989; Kim et al., 1991; Guidotti et al., 1995; Slagle et al., transcription in human hepatocytes, is normally built in the
1996). The first HBV-replicating transgenic mice were devel- liver of transgenic mice. Only HBV transgenic mice crossed to
oped by Chisari and colleagues in 1995 (Guidotti et al., 1995). HNF1 null mice were reported to display low levels of
Since the transgene construct used contained exclusively viral cccDNA, although these mice showed impaired HBsAg expres-
promoters and regulatory elements, these studies demonstrat- sion and their use was limited by severe effects due to HNF1
ed the feasibility to produce in murine cells, a transgene-driven deficiency, such as hepatic and renal dysfunctions, phenylke-
production of infectious HBV virions morphologically indistin- tonuria and infertility (Raney et al., 2001).
guishable from human-derived virions (Guidotti et al., 1999). Interestingly, first results indicate that murine cells made
As the immune system of transgenic animals recognizes during transgenic for the human NTCP become fully susceptible for
embryonic development the virus as self, these studies HDV infection. However, recent studies indicated that restric-
provided the first evidence that HBV replication does not tions likely occurring after viral entry but prior to viral
induce hepatocellular injury and therefore, that HBV-related transcription (i.e. membrane fusion, nuclear entry or cccDNA
pathogenesis must be largely mediated by the host immune formation) hinder the establishment of HBV infection in
responses (Guidotti and Chisari, 2006). To support this, murine hepatocytes (Li et al., 2014). Since both HBV and HDV
induction of acute hepatitis and hepatocellular injury utilize the same envelope for binding to the cellular NTCP
was demonstrated after adoptive transfer of HBV-antigen receptor (Yan et al., 2013), this may be due to species-specific
specific CTLs (Moriyama et al., 1990; Guidotti et al., 1994, differences or the lack of cellular factors present only in human
1996b). These studies showed that the CTL-mediated release hepatocytes.
of cytokines (Guidotti et al., 1996a) can inhibit viral The fact that in transgenic mice the HBV genome cannot be
replication by lowering steady-state levels of HBV-RNAs eliminated from the host genome implies that a therapeutically-
also by non-cytolytic mechanisms (Heise et al., 1999) and mediated complete abrogation of virion productivity, which
that the recruitment of antigen non-specific inflammatory requires elimination of the cccDNA and knowledge of its
cells can amplify the severity of liver damage initiated by half-life, cannot be properly addressed in these systems. To
antigen-specific CTLs (Sitia et al., 2004). Studies performed break tolerance to HBV antigens and investigate mechanisms of
with HBV transgenic mice demonstrated that this small viral clearance, alternative mouse models have been developed
animal model is also suitable to evaluate the impact of which rely on transfecting or transducing the viral genome into
antiviral treatment strategies on HBV replication. For mouse hepatocytes by different means, such as using recombi-
instance, the impact of various nucleoside analogs, such as nant adenoviral vectors, or by hydrodynamic injection of naked
lamivudine (Weber et al., 2002), adefovir dipivoxil and DNA in mice.
entecavir (Julander et al., 2002; Julander et al., 2003) on HBV
replication was first evaluated in HBV-replicating transgenic
mice. Studies involving the analysis of gene expression 2.2. Vector-mediated transduction of HBV in mice
profiles also reported the occurrence of IFN-mediated
induction of components of the immune proteasome and Adenoviral vectors containing an over-length HBV genome
ubiquitin-like proteins in the liver of HBV-replicating (Ad-HBV) have been shown to permit efficient transfer of the
transgenic mice (Wieland et al., 2003). Small interfering HBV genome into the mouse liver (Sprinzl et al., 2001). After
RNAs (siRNAs) specifically recognizing HBV transcripts have intravenous injection of adenoviral vectors, HBV replication and
shown to suppress virion productivity in transgenic mice viremia production could be demonstrated for up to 3 months
(Klein et al., 2003; McCaffrey et al., 2003). As an alternative and mild to moderate liver inflammation with elevated serum
therapeutic approach aiming at combining both gene alanine transaminase activities was observed (Isogawa et al.,
silencing and the induction of interferon responses in the 2005). After the induction of a B-cell response, anti-HBs
liver, Protzer and colleagues designed 5-triphosphorylated seroconversion and development of neutralizing antibodies
small interfering RNAs targeting highly conserved sequences could be determined; this finally led to the clearance of the
on HBV RNA transcripts (Ebert et al., 2011). These bi-functional infection. Because of the acute, self-limiting character of such
antiviral molecules suppressed HBV replication in vivo more adenoviral-mediated HBV infection, the system offers good
efficiently than HBV-specific siRNAs lacking a triphosphate possibilities to investigate the mechanisms of immune-
group, which could trigger antiviral responses upon induction mediated viral clearance in mice. However, establishment of
of the helicase retinoid acid-inducible protein I (RIG-I) (Ebert long-term viral replication with Ad-HBV vectors is limited
et al., 2011). by the immune responses against these vectors, while the
42 M. Dandri, M. Ltgehetmann / Journal of Immunological Methods 410 (2014) 3949

occurrence of some adenovirus-mediated cytotoxic effects may Since re-infection of the mouse hepatocytes is not possible,
also limit their application. viral clearance may be easier to achieve in HBV-transfected
mouse systems than in the course of natural infection, and
2.3. Hydrodynamic injection of replication competent HBV or therefore, the efficacy of antiviral treatments should be
HDV genomes validated in systems permissive for HBV infection.

Hydrodynamic injection techniques, which involve the fast 3. Chimeric mouse models of HBV and HDV infections
injection of high volume of fluids containing naked DNA into
the tail vein of mice, allow crossing species-specific barriers Mouse models of HBV replication do not permit to study
and hence permit efficient HBV DNA transfer (Yang et al., specific steps of the life cycle of human hepatitis viruses,
2002). Hydrodynamic injection of replication-competent HBV such as the mechanisms of HBV (and HDV) entry, cccDNA
genomes in mice resulted in high levels of HBV replication with formation and spreading. Moreover, species-specific differ-
titres up to 1 107 HBV DNA/ml (Yang et al., 2002). Although ences and the inability to reproduce the entire infection cycle
HBV replication initiated already 1 day post-injection, replica- of these viruses in murine hepatocytes have hindered our
tion levels decreased after one week and HBV was cleared from understanding of the molecular mechanisms by which HBV
blood within 23 weeks, as soon as specific antiviral antibodies and HDV can affect hepatocyte functions and are directly
and CD8+ T cells appeared. However, HBV infection persisted involved in the carcinogenesis process. Because of these
for at least 81 days after hydrodynamic injection of mice restraints, in the last 15 years many efforts have concentrat-
lacking adaptive immune cells and natural killer cells, thus ed on the development of models based on the use of the
demonstrating that the outcome of hydrodynamic transfection natural target of HBV and HDV infections: the human
of HBV depended on the host immune response (Yang et al., hepatocyte. However, availability of highly viable primary
2002). Hydrodynamic injection studies also showed that human hepatocytes is limited, the cannot be propagated in
simultaneous delivery of HBV expressing plasmids and vitro, and their susceptibility to HBV infection is highly
HBV-specific siRNAs prevented HBV replication (Klein et al., variable and generally low, since these cells rapidly lose their
2003; McCaffrey et al., 2003), while by injecting modified HBV high-differentiation status and hence the expression of
DNA plasmids into C57BL/6 mice, a significant immune hepatocyte-specific factors, such as the NTCP receptor, that
clearance of HBV could be achieved (Lin et al., 2010). By are essential to establish the infection (Yan et al., 2012). The
avoiding to raise sufficient cellular immunity against HBcAg, generation of mice carrying human hepatocytes allows
the authors proposed a model of HBV persistence, which better overcoming most of these limitations. The Trimera mice
resembled the immune tolerant stage of chronic HBV carriers. represented the first humanmouse chimeric system that
Hydrodynamics-based transfection procedures have been was developed by transplanting human liver fragments
also used for in vivo studies of HDV replication and pathogen- under the kidney capsule of normal BALB/c mice. To
esis (Chang et al., 2001). A transitory accumulation of HDV RNA avoid rejection of the implanted tissue, these mice were
transcripts and appearance of the delta antigens (HDAg) could preconditioned by total body irradiation and reconstituted
be demonstrated in up to 3% of the mouse hepatocytes after with SCID mouse bone marrow cells (Ilan et al., 1999). Using
transfection of HDV RNA sequences. Since in the absence of this approach and by infecting ex vivo small human liver
HBV envelope proteins the newly synthesized HDV genomes specimens with HBV, low levels of viremia could be
cannot be released, viremia could not be established. To determined in most of the implanted animals, where viral
overcome this problem, a following study used hydrodynamic titres remained detectable for at least 20 days. Interestingly,
injection of HDV-containing plasmids in HBV transgenic mice. mice ectopically carrying human liver tissue fragments were
In this case, not only transient development of viremia but also shown to support the engraftment of human peripheral
the in vivo antiviral efficacy of prenylation inhibitors was blood mononuclear cells (PBMC), so that the effects of
reported (Bordier et al., 2003). Despite the relatively short span polyclonal anti-HBs antibodies against HBV could be assessed
of viral replication available, mice transfected by hydrodynam- (Bocher et al., 2000; Eren et al., 2000). Nevertheless, due to
ic injection are suitable not only for short-term antiviral studies the extra-hepatic location of the implanted tissues, human
but also for testing the consequences of specific mutations hepatocytes remained functional only for limited time and in
within the viral genomes. In comparison to HBV-transgenic vivo infection with HBV or other human hepatotropic viruses
mice, hydrodynamic-based procedures permit investigation of could not be established.
immune response emerging during acute infection. Since the To achieve long-term survival of highly differentiated
viral genome is not integrated into the host genome as a human hepatocytes permissive for HBV infection in vivo,
transgene, viral clearance, mediated by the immune system or major attempts have been made to create mice harboring
after antiviral therapy, can be achieved in these systems. A human hepatocytes stably integrated in the liver parenchy-
further advantage of the system is that many different mutant ma. To date and as summarized in Fig. 1, a handful of
viruses either cloned from patients, or specifically designed in different human-liver chimeric mouse models have been
vitro, can be injected into mice and analyzed in vivo for their established, even though the urokinase-type plasminogen
replication capacity in relatively short time. Nevertheless, it activator (uPA) transgenic mouse (Sandgren et al., 1991;
must be kept in mind that the rapid injection of large volume of Rhim et al., 1994; Dandri et al., 2001b) and the knockout
fluids causes strong distress to the animals and significant liver fumarylacetoacetate hydrolase (FAH) mouse (Grompe et al.,
damage, which may alter cell function and signaling analyses. 1993; Azuma et al., 2007) are the most used and best
Furthermore, viral clearance implies not only virus elimination characterized. The basic concept of these systems is that the
of the HBV-transfected cells, but also control of viral spread. presence of a transgene-driven hepatocyte damage creates
M. Dandri, M. Ltgehetmann / Journal of Immunological Methods 410 (2014) 3949 43

A uPA/SCID mice:
Transplantation in young mice
intrasplenic Reconstitution with
injection human hepatocytes liver injury in newborn mice
reconstitution of mouse liver
with human hepatocytes
2-3 week-old (2 months)
Hepatocyte isolation Alb-uPA/SCID/beige
from adult human livers MUP-uPA/SCID

B intrasplenic Drug withdrawal (NCTB) Reconstitution with FRG and TK-NOG mice:
injection or addition (Gancyclovir) human hepatocytes Transplantation in adult mice
liver injury conditionally activated
reconstitution of mouse liver
with human hepatocytes
Hepatocyte isolation adult Fah-/- (Rag2-IL2rgnull) (2-3 months)
from adult human livers adult TK-NOG

repeated intrasplenic injection

C intrasplenic Reconstitution with human


AFC8hu mice:
Transplantation in adult mice
injection immune cells & hepatocytes
liver injury conditionally activated
partial reconstitution of immune system
(2-3 months)
Hepatic stem cells (HSC) isolation with human cells; limited expansion of
adult AFC8
from human fetal livers the hepatocyte lineage

D intrasplenic
Reconstitution with
human hepatocytes
Reconstitution with human
immune cells after
uPA-NOG mice:
injection treosulfan-conditioning Adult hepatocytes transplantation in
(2-3 months) (2-3 months) young mice
liver injury in newborn mice
reconstitution of mouse liver
2-3 week-old
Hepatocyte isolation with human hepatocytes
Alb-uPA-NOG
from adult human livers partial reconstitution of immune system
with human HSCs

Hepatic stem cells (HSC) isolation


from human fetal livers

Fig. 1. Schematic outlines of the experimental approaches used to generate mice with human chimeric livers. A) One million fresh or cryopreserved hepatocytes
isolated from adult human livers are intrasplenic injected into young immune deficient uPA mice. Reconstitution of the mouse liver with human hepatocytes is
accomplished in 2 months. B) Isolated adult human hepatocytes are transplanted into upon induction of liver injury, which is initiated either by withdrawing the
inhibitor of the tyrosine catabolic pathway NCTB in FRG mice, or by administering gancyclovir to the TK-NOG mice. Transplantation of higher hepatocyte amounts
or a second round of hepatocyte injection was shown to increase levels of human chimerism in FRG mice. C) Hepatic stem cells (HSCs) isolated from a human fetal
liver are injected in adult AFC8 mice to reconstitute mice with cells of both hepatocyte and immune cell lineages. D) Cryopreserved adult hepatocytes were first
transplanted in young uPA-NOG mice to generate chimeric mouse livers. After confirming the presence of human albumin in peripheral blood, mismatched HSCs
derived from a human fetal liver are administered i.v. after 3 days of chemotherapeutic conditioning with treosulfan to dually reconstitute animals with human
hematolymphoid cells and hepatocytes.

the space and the regenerative stimulus necessary for the animals are not transplanted with normal mouse hepatocytes in
transplanted hepatocytes to expand and reconstitute the the first month of life (Brezillon et al., 2008). To reconstitute the
diseased mouse liver, while the absence of murine adaptive liver of these animals with xenogenic hepatocytes, the uPA mice
immune cells and NK cells permits the engraftment and survival need to be backcrossed with immunodeficient mouse strains,
of transplanted xenogenic hepatocytes for the life-span of the such as the RAG2/, the Severe Combined Immune Deficient
mice. (SCID), or SCID/beige mice, which lack functional B, T and
The Alb-urokinase-type plasminogen activator (uPA) trans- NK-cells (Rhim et al., 1995; Petersen et al., 1998; Dandri et al.,
genic mouse was the first model where the strong regeneration 2001b; Mercer et al., 2001). Following a single injection of one
capacity of healthy endogenous and transplanted hepatocytes million human hepatocytes into the mouse spleen, a proportion
was reported. In this system, over-expression of the hepatotoxic of the transplanted cells migrates via the splenic and portal
transgene, which is driven by the mouse albumin promoter, veins to the liver where cells can integrate into the liver
induces high levels of uPA in plasma, hypo-fibrinogenemia, and parenchyma. A few days post-transplantation, isolated single
subacute liver failure in young mice (Sandgren et al., 1991). clusters of engrafted human hepatocytes start to proliferate, as
Soon after birth, somatic deletion of the uPA transgene takes shown by the presence of cell-proliferation markers (PCNA and
place in a small fraction of mouse hepatocytes, so that these Ki67), to form larger regenerative nodules which eventually
cured cells suddenly have a strong selective growth advantage merge together to replace most of the diseased liver parenchy-
over the transgene-carrying diseased hepatocytes. Since ma (Fig. 2). Reconstitution of the mouse liver takes eight to ten
inactivation of both copies of the uPA transgene within the weeks, meanwhile the levels of human chimerism can be
same cell is a rare event, reconstitution of the mouse liver with estimated by determining the concentration of human serum
healthy recipient-derived hepatocytes is very inefficient and albumin in mouse blood (Meuleman and Leroux-Roels, 2008;
mortality of homozygous young adult uPA mice is high if the Dandri and Petersen, 2012). Since the transplanted human
44 M. Dandri, M. Ltgehetmann / Journal of Immunological Methods 410 (2014) 3949

hepatocytes reside in their natural environment, the liver, they However, accumulation of toxic metabolites can be avoided
appear to maintain normal metabolic functions (Tateno et al., by treating the mice with a pathway inhibitor, the drug NTBC
2004). The use of highly viable cryopreserved-thawed hepato- (2-(2-nitro-4-trifluoromethylbenzyol)-cyclohexane-1,3-dio-
cytes (Dandri et al., 2001a) gives the possibility to generate on ne), so that the animals are protected from the occurrence of
demand human chimeric mice, thus overcoming previous liver injury until drug administration is withdrawn. More
limitations due to the narrow time frame available for recently, mice lacking the Fah-gene, the recombination
transplantation. Nevertheless, high levels of human chime- activating gene 2 [Rag2] and the gamma-chain of the receptor
rism are only achieved in homozygous uPA immunodeficient for IL-2, were successfully reconstituted with human hepato-
recipients. Since these mice suffer from infertility, also the cytes, so that by transplanting higher amounts of human
breeder pairs need to be rescued by transplantation with hepatocytes, or by performing hepatocyte transplantation
normal mouse hepatocytes (Brezillon et al., 2008). To twice, high rates of human hepatocyte chimerism could be
attenuate liver injury in new-born mice and hence delay achieved in these so-called FRG mice (Bissig et al., 2010) (Fig. 1,
the production of the toxic transgene which makes trans- B). In an additional humanized mouse model that was lately
plantation procedures necessary in the first month of life, developed (Kosaka et al., 2013), human hepatocytes were also
alternative mouse models, such as mice where the expres- successfully transplanted in mice expressing the herpes
sion of the uPA transgene is inducible (Song et al., 2009) or is simplex type-1 thymidine kinase (TK) transgene that were
regulated by the MUP (major urinary protein) promoter backcrossed with NOG (NOD/SCID/gamma(c)(null)) mice.
have been developed (Tesfaye et al., 2013). Even in this case, human hepatocyte transplantation can be
In 2007, two groups reported the establishment of an performed in adult mice and at will, since mouse liver cells
alternative human-liver chimeric model, based on the use of expressing the TK-transgene can be selectively destroyed upon
fumaryl acetoacetate hydrolase-deficient (FAH) mice, where administration of ganciclovir (Hasegawa et al., 2011).
hepatocyte injury can be induced, at will, in adult mice Regardless of the mouse systems employed, the human
(Azuma et al., 2007; Bissig et al., 2007; Bissig et al., 2010). cells residing in the mouse liver have been shown to
FAH plays a crucial role in the tyrosine breakdown pathway maintain susceptibility of infection with human hepatotropic
and its deficiency leads to hypertyrosinemia and liver failure. viruses (Dandri et al., 2001b; Mercer et al., 2001; Bissig et al.,
2010; Lutgehetmann et al., 2011; Kosaka et al., 2013). From
the first successful transplantation of human hepatocytes
into uPA/RAG2 mice and establishment of de novo infection
with HBV (Dandri et al., 2001b), several groups demonstrat-
A B ed the establishment of HBV infection (Tsuge et al., 2005;
Meuleman and Leroux-Roels, 2008; Tsuge et al., 2010), as
well as of infection with other human hepatitis viruses in
these humanized mice (Fig. 2) (Mercer et al., 2001;
Meuleman and Leroux-Roels, 2008; Lutgehetmann et al.,
2012). Notably, after intra-peritoneal inoculation of HBV
infectious serum or cell culture derived virions, infection
C D establishment is first achieved in a small minority of human
hepatocytes, so that several weeks are needed to accomplish
viral spreading (Dandri and Petersen, 2012). 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 as virus inoculum allows the
functional analysis of distinct HBV genotypes, naturally
E F occurring variants and drug-resistance mutants. Moreover,
genetically engineered viruses produced in cell cultures can
be used to investigate the role of distinct viral proteins in
human infected hepatocytes and in the contest of physiolog-
ical levels of viral replication. Using this type of approach,
experiments with HBV strains lacking the precore protein
proved that the HBeAg is dispensable for viral infection and
Fig. 2. Human chimeric livers generated after human hepatocyte transplan- replication in vivo (Tsuge et al., 2005), while the expression
tation of immunodeficient (SCID/beige) uPA transgenic mice. Immunostain- of the regulatory HBx protein provided in trans was shown to
ing with human specific CK-18 antibody (brown) shows a close-up of a
be essential for HBV replication in infected human hepato-
human hepatocyte cluster (A) as well as an example of a cross-section from
a highly repopulated mouse liver. CF) Immune fluorescence staining of cytes, where viral transcription is exclusively driven by the
different viral hepatitis antigens demonstrating susceptibility of repopulated cccDNA minichromosome (Tsuge et al., 2010). Although
chimeric mice to a broad spectrum of hepatotropic viruses (HBV, HDV and studies focusing on investigating how HBV may affect cellular
HCV). C) Establishment of HBV infection (HBcAg = green) and (D) HBV/ pathways and innate immune responses of the human
HDV co-infection (HBcAg = green; HDAg = red, hCK18 = blue) in human
hepatocytes. E) Close-up of HDV positive human hepatocytes in HDV
hepatocytes are still limited (Lutgehetmann et al., 2011),
monoinfected mice (HDAg = red, hCK18 = blue) and F) HCV infected cells highly repopulated mice have proved to be useful for the
(HCV core antigen = green, human calnexin = red) within the mouse liver. study of drug metabolism (Katoh et al., 2007). In an attempt
M. Dandri, M. Ltgehetmann / Journal of Immunological Methods 410 (2014) 3949 45

to study the effects of viral infection on the host genome, genes (ISGs) determined in HBV-infected human hepato-
recent studies in human liver chimeric mice indicated that cytes (Lutgehetmann et al., 2011). On the other hand,
HBV and HCV can contribute to the induction and accumu- studies in these mice also provided evidence that IFN can
lation of aberrant DNA methylation in human hepatocytes inhibit HBV transcription by mediating epigenetic repres-
through the activation of NK-cell-dependent innate immune sion of the cccDNA minichromosome (Belloni et al., 2012),
responses (Okamoto et al., 2014). while studies in uPA/SCID/beige mice demonstrated that
repeated administrations of the longer-active pegylated IFN
3.1. Preclinical studies (peg-IFN) could breach the impairment of HBV-infected
hepatocyte responsiveness and induce sustained enhancement
Since mice are well-characterized small laboratory animals, of human interferon stimulated genes (ISGs) (Allweiss et al.,
mice with humanized livers offer unique opportunities to 2013; Allweiss et al., 2014). Because of the capacity of IFN to
evaluate the in vivo efficacy of antiviral compounds targeting induce epigenetic suppression of cccDNA-driven transcrip-
different steps of the HBV life-cycle. Besides evaluating the tion of both pregenomic RNA (pgRNA) and subgenomic HBV
effect of clinically approved HBV polymerase inhibitors, such as RNAs, the stronger antiviral effects of peg-IFN exerted on the
lamivudine (Tsuge et al., 2005), adefovir (Dandri et al., 2005) human hepatocytes were shown to trigger a substantial decline
and entecavir (Lutgehetmann et al., 2011), chimeric models of circulating HBsAg and HBeAg levels in chimeric mice,
have been also employed to assess the in vivo efficacy of novel without claiming the involvement of immune cell responses
polymerase or capsid inhibitors (Brezillon et al., 2011; Volz et (Allweiss et al., 2014). Altogether, these studies demonstrate
al., 2012). Humanized uPA mice served to assess the in vivo that mice with humanized livers permit not only the
efficacy of viral entry inhibitors, derived from the large exploration of the direct antiviral effects of distinct sub-
envelope protein of HBV, to prevent de novo HBV (Petersen stances but also the simultaneous and comparative analysis
et al., 2008) and HDV infection (Lutgehetmann et al., 2012), of the distinct innate immune responses that can be induced
as well as to investigate the ability of the most clinically in human and mouse hepatocytes within the same liver. To
advanced entry inhibitor, Myrcludex-B, to block HBV spreading this regard, it was recently reported that type I, II and III IFNs
post-infection (Volz et al., 2013). In this study, Myrcludex-B are differently induced in murine and human hepatocytes
could block HBV dissemination from initially infected human by treating chimeric mice with complexes comprising
hepatocytes also when treatment was started 3 weeks hepatotropic cationic liposomes and synthetic double-
post-infection, at a time when mice already displayed detect- stranded RNA analogs (Nakagawa et al., 2013). In contrast
able levels of circulating virions. Notably, after 6-weeks of daily to mouse hepatocytes, human hepatocytes appeared to
treatment not only the amount of HBcAg-positive hepatocytes express higher levels of IFN- receptor-encoding genes,
but also of intrahepatic cccDNA loads remained comparable to while IFN- and IFN- were produced at lower levels in
values found in mice sacrificed 3-weeks post-infection, thus these cells. Thus, these studies showed that expression and
indicating that not only intrahepatic spreading but also function of the distinct IFNs may differ between animal
intracellular cccDNA amplification mostly relayed on the species, thus underlying the importance of validating results
secretion of progeny viruses and their binding to target obtained in murine systems also by employing primary
hepatocytes (Volz et al., 2013). human hepatocytes.
The passage of infected tupaia (Lutgehetmann et al., 2010) Human liver chimeric mice can be super-infected or
or human hepatocytes (Allweiss et al., 2013) isolated from simultaneously infected with different human hepatotropic
chimeric mice and transplanted into new uPA/SCID recipient viruses (Fig. 2). Whereas such studies indicated an absence of
mice has also shown to permit investigation of the impact of interference between HBV and HCV (Hiraga et al., 2009)
hepatocyte proliferation on cccDNA stability and activity in co-infection with the delta hepatitis virus (HDV) appeared
vivo. The drastic reduction of intrahepatic cccDNA loads to hinder the kinetics of HBV infection and replication
induced by cell division even in the absence of antiviral therapy (Lutgehetmann et al., 2012). This latter study showed that
underscores a potentially interesting weak point in HBV establishment of HDV infection is highly efficient in naive
persistence that deserves further investigations. Because of chimeric mice, since the majority of human hepatocytes
the absence of adaptive immune cells in these chimeric mice, stained HDAg-positive long before HBV spreading was com-
immune-mediated elimination of the infected hepatocytes pleted, thus confirming that HDV can replicate intrahepatically
cannot occur. Nevertheless, these systems offer unique oppor- also in the absence of HBV infection. Furthermore, in the setting
tunities to investigate factors that can affect the stability and/or of HBV/HDV simultaneous infection the increase of HBV
activity of the cccDNA minichromosome, as well as the direct viremia and intrahepatic cccDNA loads appeared significantly
antiviral effects of cytokines and immune modulatory sub- slower than in HBV mono-infected mice, while HDV inocula-
stances, such as interferons. Human hepatocytes residing in the tion in HBV stably infected mice led to a light reduction
mouse liver were shown to maintain a functional innate (median 0.6log) of HBV viremia.
immune system, since they properly respond to stimuli The intrahepatic latency of large delta antigen (HDAg) has
induced by exogenously applied interferon. To this regard, been detected following liver transplantation (Samuel et al.,
human liver chimeric mice have been used to investigate 1995; Mederacke et al., 2012). To investigate whether HDV
whether HBV can circumvent the induction of antiviral defence can persist intrahepatically in the absence of HBV and be
mechanisms (Lutgehetmann et al., 2011). Upon administration rescued at later time points from HBV infection, humanized
of regular human IFN, HBV was shown to inhibit the nuclear mice have been recently infected with cell culture derived
accumulation of STAT-1, thus providing a potential mecha- HDV particles and the presence of HDAg-positive human
nism for the reduced induction of interferon stimulated hepatocytes was determined after several weeks of HDV
46 M. Dandri, M. Ltgehetmann / Journal of Immunological Methods 410 (2014) 3949

inoculation to demonstrate establishment and maintenance transplant simultaneously human fetal hepatocytes and
of intrahepatic HDV mono-infection (Giersch et al., 2014). hematopoietic stem cells that were obtained by the digestion
Although intrahepatic amounts of large HDAg and edited of human fetal liver tissues (1518 weeks of gestation period)
HDV-RNA forms increased over time in HDV mono-infected (Fig. 1). Since the fetal liver provides both types of cells,
livers, HBV super-infection led to prompt viremia develop- reconstitution of both cell lineages is syngeneic, hepatocyte
ment even after 6 weeks of latent mono-infection and the rejection by the human immune system is not expected.
infectivity of the rescued HDV virions was verified by serial Although HBV infection studies were not performed, after
passage in naive chimeric mice (Giersch et al., 2014). inoculation of hepatitis C virus, low levels of intrahepatic viral
Altogether, these in vivo studies indicated that conversion replication, as well as T-cell responses and development of
of a latent HDV infection to a productive HBV/HDV fibrosis could be determined. Despite partial success and the
co-infection may contribute to HDV persistence even in ethical restrains encountered by employing human fetal
patients with low HBV replication and in the setting of liver tissues, the generation of chimeric systems equipped with
transplantation. both a human liver and a functional human immune system,
Although the transplantation of mice is technically chal- with a matched major histocompatibility complex, still
lenging and good quality primary human hepatocytes are remains a major challenge. The latest advances reported by
needed, most of the human-liver chimeric models available Gutti et al. showed the feasibility to reconstitute a uPA-NOG
have shown great potential for the study of human hepatitis mouse strain with both adult human hepatocytes and
virus infection and preclinical drug investigation. Moreover, by hematolymphoid cells (Gutti et al., 2014). In this study, dual
using hepatocytes from different donors, possible host varia- reconstitution was achieved by transplanting fetal or even
tions and the impact of genetic polymorphisms of the donor on adult hepatocytes and mismatched hematopoietic stem cells
the efficiency of antiviral therapy can be investigated. By (CD34+ HSCs) derived from either a fetal liver or umbilical cord
employing chimeric mice carrying human hepatocytes with blood. As an alternative and well-tolerated procedure to total
different IL28B gene polymorphisms, it could be shown that body irradiation, mice received 3 days of a treosulfan-based
viral replication is suppressed with comparable efficiency upon chemotherapeutic conditioning before HSC injection. The
peg-IFN administration, thus indicating that such genetic presence of CD8+ and CD4+ human cells and of human
variations mostly affect the responsiveness of the immune cells hepatocyte clusters was observed in the liver of these animals.
rather than those of the infected cells (Watanabe et al., 2014). No cell-mediated rejection but also no evidence of cell
interactions were determined in animals reconstituted with
3.2. Immune competent human liver chimeric mice mismatched HSCs. Although the applicability of these ap-
proaches for the study of HBV and HDV associated immune
Human hepatocytes can be very abundant within the pathological processes needs further research, these technical
chimeric livers but non-parenchymal cells, such as sinusoidal advances have paved the way for the development of dually
endothelial cells and Kupffer cells, are of murine origin. As a reconstituted humanized systems.
consequence, development of fibrosis and pathophysiologic
processes that are commonly associated with chronic viral Acknowledgment
hepatitis infections but involve a crosstalk between the
hepatocytes and other liver resident cells is not observed in We thank our laboratory team, particularly Dr. Tassilo
the above-mentioned systems. Moreover, since these chime- Volz, Dr. Lena Allweiss & Katja Giersch, for their excellent
ric animals are genetically immune deficient they are not work and dedication to the model, as well as the German
suited for vaccine studies and for evaluation of adaptive Research Foundation (SFB841 (project A5 and A8) and
immune responses. To circumvent, at least in part, this DZIF-BMBF (TTU05.804)) for continuous financial support.
problem, adoptive transfer of antibodies or lymphocytes can
be applied. In the HCV field, for instance, these methods were References
used to analyze neutralizing activity of antibodies (Law et al.,
2008; Meuleman et al., 2012) or of activated lymphocytes Allweiss, L., et al., April 2013. Proliferation of hepatitis B virus infected
(Ohira et al., 2009). Partially haploidentical human periph- human hepatocytes induces suppression of viral replication and rapid
cccDNA decrease in humanized mice. April 2013. J. Hepatol. 58 (Suppl.
eral blood mononuclear cells (PBMC) were also recently 1), S56S57.
transferred in uPA/SCID chimeric mice after the establish- Allweiss, L., Volz, T., Lutgehetmann, M., Giersch, K., Bornscheuer, T., Lohse, A.
ment of HBV infection (Okazaki et al., 2012). Notably, W., Petersen, J., Ma, H., Klumpp, K., Fletcher, S.P., Dandri, M., 2014.
Immune cell responses are not required to induce substantial hepatitis B
infiltrating human immune cells caused severe hepatocyte virus antigen decline during pegylated interferon-alpha administration.
degeneration, while treatment with anti-Fas antibodies or J. Hepatol. 60, 500507.
depletion of dendritic cells prevented the death of human Araki, K., Miyazaki, J., Hino, O., Tomita, N., Chisaka, O., Matsubara, K.,
Yamamura, K., 1989. Expression and replication of hepatitis B virus
hepatocytes. genome in transgenic mice. Proc. Natl. Acad. Sci. U. S. A. 86, 207211.
A different attempt to generate a humanized mouse Azuma, H., Paulk, N., Ranade, A., Dorrell, C., Al-Dhalimy, M., Ellis, E., Strom, S.,
model harboring both human liver cells and a human immune Kay, M.A., Finegold, M., Grompe, M., 2007. Robust expansion of human
hepatocytes in Fah//Rag2//Il2rg/ mice. Nat. Biotechnol. 25,
system was the development of the AFC8 model (Washburn
903910.
et al., 2011). These mice were obtained by crossing Baumert, T.F., Yang, C., Schurmann, P., Kock, J., Ziegler, C., Grullich, C., Nassal,
BALB/c-RAG2/c/mice, which lack functional B, T and M., Liang, T.J., Blum, H.E., von Weizsacker, F., 2005. Hepatitis B virus
natural killer cells, with mice carrying a liver-specific suicidal mutations associated with fulminant hepatitis induce apoptosis in
primary tupaia hepatocytes. Hepatology 41, 247256.
transgene with inducible activity based on the induction of Belloni, L., Allweiss, L., Guerrieri, F., Pediconi, N., Volz, T., Pollicino, T.,
caspase 8 in mouse hepatocytes. These animals were used to Petersen, J., Raimondo, G., Dandri, M., Levrero, M., 2012. IFN-alpha
M. Dandri, M. Ltgehetmann / Journal of Immunological Methods 410 (2014) 3949 47

inhibits HBV transcription and replication in cell culture and in Guidotti, L.G., Matzke, B., Schaller, H., Chisari, F.V., 1995. High-level hepatitis
humanized mice by targeting the epigenetic regulation of the nuclear B virus replication in transgenic mice. J. Virol. 69, 61586169.
cccDNA minichromosome. J. Clin. Invest. 122, 529537. Guidotti, L.G., Ishikawa, T., Hobbs, M.V., Matzke, B., Schreiber, R., Chisari, F.V.,
Bissig, K.D., Le, T.T., Woods, N.B., Verma, I.M., 2007. Repopulation of adult 1996a. Intracellular inactivation of the hepatitis B virus by cytotoxic T
and neonatal mice with human hepatocytes: a chimeric animal model. lymphocytes. Immunity 4, 2536.
Proc. Natl. Acad. Sci. U. S. A. 104, 2050720511. Guidotti, L.G., Matzke, B., Pasquinelli, C., Schoenberger, J.M., Rogler, C.E.,
Bissig, K.D., Wieland, S.F., Tran, P., Isogawa, M., Le, T.T., Chisari, F.V., Chisari, F.V., 1996b. The hepatitis B virus (HBV) precore protein inhibits
Verma, I.M., 2010. Human liver chimeric mice provide a model for HBV replication in transgenic mice. J. Virol. 70, 70567061.
hepatitis B and C virus infection and treatment. J. Clin. Invest. 120, Guidotti, L.G., Rochford, R., Chung, J., Shapiro, M., Purcell, R., Chisari, F.V.,
924930. 1999. Viral clearance without destruction of infected cells during acute
Bocher, W.O., Galun, E., Marcus, H., Daudi, N., Terkieltaub, D., Shouval, D., HBV infection. Science 284, 825829.
Lohr, H.F., Reisner, Y., 2000. Reduced hepatitis B virus surface antigen- Gutti, T.L., Knibbe, J.S., Makarov, E., Zhang, J., Yannam, G.R., Gorantla, S., Sun,
specific Th1 helper cell frequency of chronic HBV carriers is associated Y., Mercer, D.F., Suemizu, H., Wisecarver, J.L., Osna, N.A., Bronich, T.K.,
with a failure to produce antigen-specific antibodies in the trimera Poluektova, L.Y., 2014. Human hepatocytes and hematolymphoid dual
mouse [in process citation]. Hepatology 31, 480487. reconstitution in treosulfan-conditioned uPA-NOG mice. Am. J. Pathol.
Bordier, B.B., Ohkanda, J., Liu, P., Lee, S.Y., Salazar, F.H., Marion, P.L., Ohashi, K. 184, 101109.
, Meuse, L., Kay, M.A., Casey, J.L., Sebti, S.M., Hamilton, A.D., Glenn, J.S., Hasegawa, M., Kawai, K., Mitsui, T., Taniguchi, K., Monnai, M., Wakui, M., Ito,
2003. In vivo antiviral efficacy of prenylation inhibitors against hepatitis M., Suematsu, M., Peltz, G., Nakamura, M., Suemizu, H., 2011. The
delta virus. J. Clin. Invest. 112, 407414. reconstituted humanized liver in TK-NOG mice is mature and
Brezillon, N.M., DaSilva, L., L'Hote, D., Bernex, F., Piquet, J., Binart, N., functional. Biochem. Biophys. Res. Commun. 405, 405410.
Morosan, S., Kremsdorf, D., 2008. Rescue of fertility in homozygous mice Heise, T., Guidotti, L.G., Cavanaugh, V.J., Chisari, F.V., 1999. Hepatitis B
for the urokinase plasminogen activator transgene by the transplanta- virus RNA-binding proteins associated with cytokine-induced
tion of mouse hepatocytes. Cell Transplant. 17, 803812. clearance of viral RNA from the liver of transgenic mice. J. Virol. 73,
Brezillon, N., Brunelle, M.N., Massinet, H., Giang, E., Lamant, C., DaSilva, L., 474481.
Berissi, S., Belghiti, J., Hannoun, L., Puerstinger, G., Wimmer, E., Neyts, J., Hiraga, N., Imamura, M., Hatakeyama, T., Kitamura, S., Mitsui, F., Tanaka, S.,
Hantz, O., Soussan, P., Morosan, S., Kremsdorf, D., 2011. Antiviral activity Tsuge, M., Takahashi, S., Abe, H., Maekawa, T., Ochi, H., Tateno, C.,
of Bay 41-4109 on hepatitis B virus in humanized Alb-uPA/SCID mice. Yoshizato, K., Wakita, T., Chayama, K., 2009. Absence of viral interference
PLoS One 6, e25096. and different susceptibility to interferon between hepatitis B virus and
Chang, J., Sigal, L.J., Lerro, A., Taylor, J., 2001. Replication of the human hepatitis C virus in human hepatocyte chimeric mice. J. Hepatol. 51,
hepatitis delta virus genome Is initiated in mouse hepatocytes following 10461054.
intravenous injection of naked DNA or RNA sequences. J. Virol. 75, Hughes, S.A., Wedemeyer, H., Harrison, P.M., 2011 Jul 2. Hepatitis delta virus.
34693473. Lancet 378 (9785), 7385.
Chisari, F.V., Pinkert, C.A., Milich, D.R., Filippi, P., McLachlan, A., Palmiter, R.D., Ilan, E., Burakova, T., Dagan, S., Nussbaum, O., Lubin, I., Eren, R., Ben-Moshe,
Brinster, R.L., 1985. A transgenic mouse model of the chronic hepatitis B O., Arazi, J., Berr, S., Neville, L., Yuen, L., Mansour, T.S., Gillard, J., Eid, A.,
surface antigen carrier state. Science 230, 11571160. Jurim, O., Shouval, D., Reisner, Y., Galun, E., 1999. The hepatitis B virus-
Dandri, M., Petersen, J., 2012. Chimeric mouse model of hepatitis B virus trimera mouse: a model for human HBV infection and evaluation of anti-
infection. J. Hepatol. 56, 493495. HBV therapeutic agents. Hepatology 29, 553562.
Dandri, M., Schirmacher, P., Rogler, C.E., 1996. Woodchuck hepatitis virus X Isogawa, M., Kakimi, K., Kamamoto, H., Protzer, U., Chisari, F.V., 2005.
protein is present in chronically infected woodchuck liver and Differential dynamics of the peripheral and intrahepatic cytotoxic T
woodchuck hepatocellular carcinomas which are permissive for viral lymphocyte response to hepatitis B surface antigen. Virology 333,
replication. J. Virol. 70, 52465254. 293300.
Dandri, M., Burda, M.R., Gocht, A., Torok, E., Pollok, J.M., Rogler, C.E., Will, H., Julander, J.G., Sidwell, R.W., Morrey, J.D., 2002. Characterizing antiviral
Petersen, J., 2001a. Woodchuck hepatocytes remain permissive for activity of adefovir dipivoxil in transgenic mice expressing hepatitis B
hepadnavirus infection and mouse liver repopulation after cryopreser- virus. Antivir. Res. 55, 2740.
vation. Hepatology 34, 824833. Julander, J.G., Colonno, R.J., Sidwell, R.W., Morrey, J.D., 2003. Characterization
Dandri, M., Burda, M.R., Torok, E., Pollok, J.M., Iwanska, A., Sommer, G., of antiviral activity of entecavir in transgenic mice expressing hepatitis B
Rogiers, X., Rogler, C.E., Gupta, S., Will, H., Greten, H., Petersen, J., 2001b. virus. Antivir. Res. 59, 155161.
Repopulation of mouse liver with human hepatocytes and in vivo Katoh, M., Sawada, T., Soeno, Y., Nakajima, M., Tateno, C., Yoshizato, K.,
infection with hepatitis B virus. Hepatology 33, 981988. Yokoi, T., 2007. In vivo drug metabolism model for human cytochrome
Dandri, M., Burda, M.R., Zuckerman, D.M., Wursthorn, K., Matschl, U., Pollok, P450 enzyme using chimeric mice with humanized liver. J. Pharm. Sci.
J.M., Rogiers, X., Gocht, A., Kock, J., Blum, H.E., von Weizsacker, F., 96, 428437.
Petersen, J., 2005. Chronic infection with hepatitis B viruses and antiviral Kim, C.M., Koike, K., Saito, I., Miyamura, T., Jay, G., 1991. HBx gene of hepatitis
drug evaluation in uPA mice after liver repopulation with tupaia B virus induces liver cancer in transgenic mice. Nature 351, 317320.
hepatocytes. J. Hepatol. 42, 5460. Klein, C., Bock, C.T., Wedemeyer, H., Wustefeld, T., Locarnini, S., Dienes, H.P.,
Ebert, G., Poeck, H., Lucifora, J., Baschuk, N., Esser, K., Esposito, I., Hartmann, Kubicka, S., Manns, M.P., Trautwein, C., 2003. Inhibition of hepatitis B
G., Protzer, U., 2011. 5 Triphosphorylated small interfering RNAs virus replication in vivo by nucleoside analogues and siRNA. Gastroen-
control replication of hepatitis B virus and induce an interferon re- terology 125, 918.
sponse in human liver cells and mice. Gastroenterology 141, 696706 Kosaka, K., Hiraga, N., Imamura, M., Yoshimi, S., Murakami, E., Nakahara, T.,
(706 e1-3.). Honda, Y., Ono, A., Kawaoka, T., Tsuge, M., Abe, H., Hayes, C.N., Miki, D.,
Eren, R., Ilan, E., Nussbaum, O., Lubin, I., Terkieltaub, D., Arazi, Y., Ben-Moshe, Aikata, H., Ochi, H., Ishida, Y., Tateno, C., Yoshizato, K., Sasaki, T.,
O., Kitchinzky, A., Berr, S., Gopher, J., Zauberman, A., Galun, E., Shouval, Chayama, K., 2013. A novel TK-NOG based humanized mouse model for
D., Daudi, N., Eid, A., Jurim, O., Magnius, L.O., Hammas, B., Reisner, Y., the study of HBV and HCV infections. Biochem. Biophys. Res. Commun.
Dagan, S., 2000. Preclinical evaluation of two human anti-hepatitis B 441, 230235.
virus (HBV) monoclonal antibodies in the HBV-trimera mouse model Law, M., Maruyama, T., Lewis, J., Giang, E., Tarr, A.W., Stamataki, Z.,
and in HBV chronic carrier chimpanzees. Hepatology 32, 588596. Gastaminza, P., Chisari, F.V., Jones, I.M., Fox, R.I., Ball, J.K., McKeating, J.
Giersch, K., Helbig, M., Volz, T., Allweiss, L., Mancke, L.V., Lohse, A.W., A., Kneteman, N.M., Burton, D.R., 2008. Broadly neutralizing antibodies
Polywka, S., Pollok, J.M., Petersen, J., Taylor, J., Dandri, M., Lutgehetmann, protect against hepatitis C virus quasispecies challenge. Nat. Med. 14,
M., 2014 Mar. Persistent hepatitis D virus mono-infection in humanized 2527.
mice is efficiently converted by hepatitis B virus to a productive co- Li, H., Zhuang, Q., Wang, Y., Zhang, T., Zhao, J., Zhang, Y., Zhang, J., Lin, Y.,
infection. J Hepatol. 2014 Mar 60 (3), 538544. Yuan, Q., Xia, N., Han, J., 2014 Mar. HBV life cycle is restricted in mouse
Grompe, M., al-Dhalimy, M., Finegold, M., Ou, C.N., Burlingame, T., hepatocytes expressing human NTCP. Cell Mol. Immunol. 11 (2),
Kennaway, N.G., Soriano, P., 1993. Loss of fumarylacetoacetate hydrolase 175183.
is responsible for the neonatal hepatic dysfunction phenotype of lethal Lin, Y.J., Huang, L.R., Yang, H.C., Tzeng, H.T., Hsu, P.N., Wu, H.L., Chen, P.J.,
albino mice. Genes Dev. 7, 22982307. Chen, D.S., 2010. Hepatitis B virus core antigen determines viral
Guidotti, L.G., Chisari, F.V., 2006. Immunobiology and pathogenesis of viral persistence in a C57BL/6 mouse model. Proc. Natl. Acad. Sci. U. S. A.
hepatitis. Annu. Rev. Pathol. 1, 2361. 107, 93409345.
Guidotti, L.G., Guilhot, S., Chisari, F.V., 1994. Interleukin-2 and alpha/beta Lutgehetmann, M., Volz, T., Kopke, A., Broja, T., Tigges, E., Lohse, A.W., Fuchs,
interferon down-regulate hepatitis B virus gene expression in vivo by E., Murray, J.M., Petersen, J., Dandri, M., 2010. In vivo proliferation of
tumor necrosis factor-dependent and -independent pathways. J. Virol. hepadnavirus-infected hepatocytes induces loss of covalently closed
68, 12651270. circular DNA in mice. Hepatology 52, 1624.
48 M. Dandri, M. Ltgehetmann / Journal of Immunological Methods 410 (2014) 3949

Lutgehetmann, M., Bornscheuer, T., Volz, T., Allweiss, L., Bockmann, J.H., Raney, A.K., Eggers, C.M., Kline, E.F., Guidotti, L.G., Pontoglio, M., Yaniv, M.,
Pollok, J.M., Lohse, A.W., Petersen, J., Dandri, M., 2011. Hepatitis B virus McLachlan, A., 2001. Nuclear covalently closed circular viral genomic
limits response of human hepatocytes to interferon-alpha in chimeric DNA in the liver of hepatocyte nuclear factor 1 alpha-null hepatitis B
mice. Gastroenterology 140, 20742083 (2083 e1-2). virus transgenic mice. J. Virol. 75, 29002911.
Lutgehetmann, M., Mancke, L.V., Volz, T., Helbig, M., Allweiss, L., Rhim, J.A., Sandgren, E.P., Degen, J.L., Palmiter, R.D., Brinster, R.L., 1994.
Bornscheuer, T., Pollok, J.M., Lohse, A.W., Petersen, J., Urban, S., Dandri, Replacement of diseased mouse liver by hepatic cell transplantation.
M., 2012. Humanized chimeric uPA mouse model for the study of Science 263, 11491152.
hepatitis B and D virus interactions and preclinical drug evaluation. Rhim, J.A., Sandgren, E.P., Palmiter, R.D., Brinster, R.L., 1995. Complete
Hepatology 55, 685694. reconstitution of mouse liver with xenogeneic hepatocytes. Proc. Natl.
Mason, W.S., Seal, G., Summers, J., 1980. Virus of Pekin ducks with structural Acad. Sci. U. S. A. 92, 49424946.
and biological relatedness to human hepatitis B virus. J. Virol. 36, Rizzetto, M., 2009. Hepatitis D: thirty years after. J. Hepatol. 50, 10431050.
829836. Roggendorf, M., Tolle, T.K., 1995. The woodchuck: an animal model for
Mason, W.S., Aldrich, C., Summers, J., Taylor, J.M., 1982. Asymmetric hepatitis B virus infection in man. Intervirology 38, 100112.
replication of duck hepatitis B virus DNA in liver cells: free minus- Samuel, D., Zignego, A.L., Reynes, M., Feray, C., Arulnaden, J.L., David, M.F.,
strand DNA. Proc. Natl. Acad. Sci. U. S. A. 79, 39974001. Gigou, M., Bismuth, A., Mathieu, D., Gentilini, P., et al., 1995. Long-term
Mason, W.S., Jilbert, A.R., Summers, J., 2005. Clonal expansion of hepatocytes clinical and virological outcome after liver transplantation for cirrhosis
during chronic woodchuck hepatitis virus infection. Proc. Natl. Acad. Sci. caused by chronic delta hepatitis. Hepatology 21, 333339.
U. S. A. 102, 11391144. Sandgren, E.P., Palmiter, R.D., Heckel, J.L., Daugherty, C.C., Brinster, R.L.,
McCaffrey, A.P., Nakai, H., Pandey, K., Huang, Z., Salazar, F.H., Xu, H., Wieland, Degen, J.L., 1991. Complete hepatic regeneration after somatic deletion
S.F., Marion, P.L., Kay, M.A., 2003. Inhibition of hepatitis B virus in mice of an albumin-plasminogen activator transgene. Cell 66, 245256.
by RNA interference. Nat. Biotechnol. 21, 639644. Sitia, G., Isogawa, M., Iannacone, M., Campbell, I.L., Chisari, F.V., Guidotti, L.G.,
Mederacke, I., Filmann, N., Yurdaydin, C., Bremer, B., Puls, F., Zacher, B.J., 2004. MMPs are required for recruitment of antigen-nonspecific
Heidrich, B., Tillmann, H.L., Rosenau, J., Bock, C.T., Savas, B., Helfritz, F., mononuclear cells into the liver by CTLs. J. Clin. Invest. 113, 11581167.
Lehner, F., Strassburg, C.P., Klempnauer, J., Wursthorn, K., Lehmann, U., Slagle, B.L., Lee, T.H., Medina, D., Finegold, M.J., Butel, J.S., 1996. Increased
Manns, M.P., Herrmann, E., Wedemeyer, H., 2012. Rapid early HDV RNA sensitivity to the hepatocarcinogen diethylnitrosamine in transgenic
decline in the peripheral blood but prolonged intrahepatic hepatitis mice carrying the hepatitis B virus X gene. Mol. Carcinog. 15, 261269.
delta antigen persistence after liver transplantation. J. Hepatol. 56, Song, X., Guo, Y., Duo, S., Che, J., Wu, C., Ochiya, T., Ding, M., Deng, H., 2009. A
115122. mouse model of inducible liver injury caused by tet-on regulated
Mercer, D.F., Schiller, D.E., Elliott, J.F., Douglas, D.N., Hao, C., Rinfret, A., urokinase for studies of hepatocyte transplantation. Am. J. Pathol. 175,
Addison, W.R., Fischer, K.P., Churchill, T.A., Lakey, J.R., Tyrrell, D.L., 19751983.
Kneteman, N.M., 2001. Hepatitis C virus replication in mice with Sprinzl, M.F., Oberwinkler, H., Schaller, H., Protzer, U., 2001. Transfer of
chimeric human livers. Nat. Med. 7, 927933. hepatitis B virus genome by adenovirus vectors into cultured cells and
Meuleman, P., Leroux-Roels, G., 2008. The human liver-uPA-SCID mouse: a mice: crossing the species barrier. J. Virol. 75, 51085118.
model for the evaluation of antiviral compounds against HBV and HCV. Summers, J., Mason, W.S., 2004. Residual integrated viral DNA after
Antivir. Res. 80, 231238. hepadnavirus clearance by nucleoside analog therapy. Proc. Natl. Acad.
Meuleman, P., Catanese, M.T., Verhoye, L., Desombere, I., Farhoudi, A., Jones, Sci. U. S. A. 101, 638640.
C.T., Sheahan, T., Grzyb, K., Cortese, R., Rice, C.M., Leroux-Roels, G., Sureau, C., 2006. The role of the HBV envelope proteins in the HDV
Nicosia, A., 2012. A human monoclonal antibody targeting scavenger replication cycle. Curr. Top. Microbiol. Immunol. 307, 113131.
receptor class B type I precludes hepatitis C virus infection and viral Tateno, C., Yoshizane, Y., Saito, N., Kataoka, M., Utoh, R., Yamasaki, C.,
spread in vitro and in vivo. Hepatology 55, 364372. Tachibana, A., Soeno, Y., Asahina, K., Hino, H., Asahara, T., Yokoi, T.,
Moriyama, T., Guilhot, S., Klopchin, K., Moss, B., Pinkert, C.A., Palmiter, R.D., Furukawa, T., Yoshizato, K., 2004. Near completely humanized liver in
Brinster, R.L., Kanagawa, O., Chisari, F.V., 1990. Immunobiology and mice shows human-type metabolic responses to drugs. Am. J. Pathol.
pathogenesis of hepatocellular injury in hepatitis B virus transgenic 165, 901912.
mice. Science 248, 361364. Tesfaye, A., Stift, J., Maric, D., Cui, Q., Dienes, H.P., Feinstone, S.M., 2013.
Nakagawa, S., Hirata, Y., Kameyama, T., Tokunaga, Y., Nishito, Y., Hirabayashi, Chimeric mouse model for the infection of hepatitis B and C viruses.
K., Yano, J., Ochiya, T., Tateno, C., Tanaka, Y., Mizokami, M., Tsukiyama- PLoS One 8, e77298.
Kohara, K., Inoue, K., Yoshiba, M., Takaoka, A., Kohara, M., 2013. Targeted Tsuge, M., Hiraga, N., Takaishi, H., Noguchi, C., Oga, H., Imamura, M.,
induction of interferon-lambda in humanized chimeric mouse liver Takahashi, S., Iwao, E., Fujimoto, Y., Ochi, H., Chayama, K., Tateno, C.,
abrogates hepatotropic virus infection. PLoS One 8, e59611. Yoshizato, K., 2005. Infection of human hepatocyte chimeric mouse
Nassal, M., 2008. Hepatitis B viruses: reverse transcription a different way. with genetically engineered hepatitis B virus. Hepatology 42,
Virus Res. 134, 235249. 10461054.
Nkongolo, S., Ni, Y., Lempp, F.A., Kaufman, C., Lindner, T., Esser-Nobis, K., Tsuge, M., Hiraga, N., Akiyama, R., Tanaka, S., Matsushita, M., Mitsui, F., Abe,
Lohmann, V., Mier, W., Mehrle, S., Urban, S., 2013. Cyclosporin A inhibits H., Kitamura, S., Hatakeyama, T., Kimura, T., Miki, D., Mori, N., Imamura,
hepatitis B and hepatitis D virus entry by cyclophilin-independent M., Takahashi, S., Hayes, C.N., Chayama, K., 2010. HBx protein is
interference with the NTCP receptor. J. Hepatol. http://dx.doi.org/10. indispensable for development of viraemia in human hepatocyte
1016/j.jhep.2013.11.022 (Epub ahead of print). chimeric mice. J. Gen. Virol. 91, 18541864.
Ohira, M., Ishiyama, K., Tanaka, Y., Doskali, M., Igarashi, Y., Tashiro, H., Hiraga, Volz, T., Lutgehetmann, M., Allweiss, L., Warlich, M., Bierwolf, J., Pollok, J.M.,
N., Imamura, M., Sakamoto, N., Asahara, T., Chayama, K., Ohdan, H., 2009. Petersen, J., Matthes, E., Dandri, M., 2012. Strong antiviral activity of the
Adoptive immunotherapy with liver allograft-derived lymphocytes new l-hydroxycytidine derivative, l-Hyd4FC, in HBV-infected human
induces anti-HCV activity after liver transplantation in humans and chimeric uPA/SCID mice. Antivir. Ther. 17, 623631.
humanized mice. J. Clin. Invest. 119, 32263235. Volz, T., Allweiss, L., MB, M.B., Warlich, M., Lohse, A.W., Pollok, J.M.,
Okamoto, Y., Shinjo, K., Shimizu, Y., Sano, T., Yamao, K., Gao, W., Fujii, M., Alexandrov, A., Urban, S., Petersen, J., Lutgehetmann, M., Dandri, M.,
Osada, H., Sekido, Y., Murakami, S., Tanaka, Y., Joh, T., Sato, S., Takahashi, 2013. The entry inhibitor Myrcludex-B efficiently blocks intrahepatic
S., Wakita, T., Zhu, J., Issa, J.P., Kondo, Y., 2014. Hepatitis virus infection virus spreading in humanized mice previously infected with hepatitis B
affects DNA methylation in mice with humanized livers. Gastroenterol- virus. J. Hepatol. 58, 861867.
ogy 146, 562572. von Weizsacker, F., Kock, J., MacNelly, S., Ren, S., Blum, H.E., Nassal, M., 2004.
Okazaki, A., Hiraga, N., Imamura, M., Hayes, C.N., Tsuge, M., Takahashi, S., Aikata, H., The tupaia model for the study of hepatitis B virus: direct infection and
Abe, H., Miki, D., Ochi, H., Tateno, C., Yoshizato, K., Ohdan, H., Chayama, K., HBV genome transduction of primary tupaia hepatocytes. Methods Mol.
2012. Severe necroinflammatory reaction caused by natural killer cell- Med. 96, 153161.
mediated Fas/Fas ligand interaction and dendritic cells in human hepatocyte Walter, E., Keist, R., Niederost, B., Pult, I., Blum, H.E., 1996. Hepatitis B virus
chimeric mouse. Hepatology 56, 555566. infection of tupaia hepatocytes in vitro and in vivo. Hepatology 24, 15.
Petersen, J., Dandri, M., Gupta, S., Rogler, C.E., 1998. Liver repopulation with Washburn, M.L., Bility, M.T., Zhang, L., Kovalev, G.I., Buntzman, A., Frelinger,
xenogenic hepatocytes in B and T cell-deficient mice leads to chronic J.A., Barry, W., Ploss, A., Rice, C.M., Su, L., 2011. A humanized mouse
hepadnavirus infection and clonal growth of hepatocellular carcinoma. model to study hepatitis C virus infection, immune response, and liver
Proc. Natl. Acad. Sci. U. S. A. 95, 310315. disease. Gastroenterology 140, 13341344.
Petersen, J., Dandri, M., Mier, W., Lutgehetmann, M., Volz, T., von Watanabe, T., Tanigawa, T., Shiba, M., Nadatani, Y., Nagami, Y., Sugimori, S.,
Weizsacker, F., Haberkorn, U., Fischer, L., Pollok, J.M., Erbes, B., Seitz, Yamagami, H., Watanabe, K., Tominaga, K., Fujiwara, Y., Koike, T.,
S., Urban, S., 2008. Prevention of hepatitis B virus infection in vivo Arakawa, T., 2014 Mar. Anti-tumour necrosis factor agents reduce non-
by entry inhibitors derived from the large envelope protein. Nat. steroidal anti-inflammatory drug-induced small bowel injury in rheu-
Biotechnol. 26, 335341. matoid arthritis patients. Gut 63 (3), 409414.
M. Dandri, M. Ltgehetmann / Journal of Immunological Methods 410 (2014) 3949 49

Weber, O., Schlemmer, K.H., Hartmann, E., Hagelschuer, I., Paessens, A., is a functional receptor for human hepatitis B and D virus. eLife 1,
Graef, E., Deres, K., Goldmann, S., Niewoehner, U., Stoltefuss, J., Haebich, e00049.
D., Ruebsamen-Waigmann, H., Wohlfeil, S., 2002. Inhibition of human Yan, H., Peng, B., He, W., Zhong, G., Qi, Y., Ren, B., Gao, Z., Jing, Z., Song, M., Xu,
hepatitis B virus (HBV) by a novel non-nucleosidic compound in a G., Sui, J., Li, W., 2013. Molecular determinants of hepatitis B and D virus
transgenic mouse model. Antivir. Res. 54, 6978. entry restriction in mouse sodium taurocholate cotransporting poly-
Wieland, S.F., Vega, R.G., Muller, R., Evans, C.F., Hilbush, B., Guidotti, L.G., peptide. J. Virol. 87, 79777991.
Sutcliffe, J.G., Schultz, P.G., Chisari, F.V., 2003. Searching for interferon- Yang, P.L., Althage, A., Chung, J., Chisari, F.V., 2002. Hydrodynamic injection
induced genes that inhibit hepatitis B virus replication in transgenic of viral DNA: a mouse model of acute hepatitis B virus infection. Proc.
mouse hepatocytes. J. Virol. 77, 12271236. Natl. Acad. Sci. U. S. A. 99, 1382513830.
Yan, H., Zhong, G., Xu, G., He, W., Jing, Z., Gao, Z., Huang, Y., Qi, Y., Peng, B., Zoulim, F., Saputelli, J., Seeger, C., 1994. Woodchuck hepatitis virus X protein
Wang, H., Fu, L., Song, M., Chen, P., Gao, W., Ren, B., Sun, Y., Cai, T., Feng, is required for viral infection in vivo. J. Virol. 68, 20262030.
X., Sui, J., Li, W., 2012. Sodium taurocholate cotransporting polypeptide

Anda mungkin juga menyukai