You are on page 1of 19

REVIEW ARTICLE

Plasmodium meets AAV—the (un)likely marriage of


parasitology and virology, and how to make the match
Franziska Hentzschel1,2,3, Anne-Kathrin Herrmann2,3, Ann-Kristin Mueller1 and Dirk Grimm2,3,4
1 Department of Parasitology, Center for Infectious Diseases, Heidelberg University Hospital, Germany
2 Department of Virology, Center for Infectious Diseases, Heidelberg University Hospital, Germany
3 Cluster of Excellence CellNetworks, Heidelberg, Germany
4 German Center for Infection Research (DZIF), Heidelberg, Germany

Correspondence The increasing use of screening technologies in malaria research has substan-
D. Grimm, Heidelberg University Hospital, tially expanded our knowledge on cellular factors hijacked by the Plasmod-
BioQuant BQ0030, Im Neuenheimer Feld
ium parasite in the infected host, including those that participate in the
267, D-69120 Heidelberg, Germany
clinically silent liver stage. This rapid gain in our understanding of the hep-
Fax: +49 6221 5451481
Tel: +49 6221 5451339 atic interaction partners now requires a means to validate and further disen-
E-mail: dirk.grimm@bioquant.uni-heidelberg. tangle parasite–host networks in physiologically relevant liver model systems.
de Here, we outline seminal work that contributed to our present knowledge on
the intrahepatic Plasmodium host factors, followed by a discussion of surro-
The first two authors contributed equally gate models of mammalian livers or hepatocytes. We finally describe how
Adeno-associated viruses could be engineered and used as hepatotropic tools
(Received 3 March 2016, revised 24 March
to dissect Plasmodium–host interactions, and to deliberately control these
2016, accepted 21 April 2016, available
online 9 May 2016) networks for antimalaria vaccination or therapy.

doi:10.1002/1873-3468.12187 Keywords: 3D culture; AAV; adeno-associated virus; liver; malaria;


Plasmodium
Edited by Wilhelm Just

Malaria is a devastating infectious disease that kills While a pivotal step in the field and a significant scien-
438 000 people a year worldwide [1], mainly children tific achievement, the enthusiasm is unfortunately
—one every 30 s. An effective vaccine is widely dampened by clinical data implying that Mosquirix
regarded as an essential tool for sustainable malaria only yields modest protection against malaria infection
control. To date, attenuated whole parasites and wild- [8–10]. In fact, it was concluded that a bed net remains
type parasites under drug cover have been shown to more effective than this vaccine.
induce complete sterilizing immunity [2–6], but numer- In general, a seminal reason for the persisting lack
ous practical issues make it unlikely that this vaccine of better vaccines is the gaps in our understanding of
type could be licensed for common use in humans. the interactions of the malaria parasite, Plasmodium,
This may differ for the latest subunit of malaria vac- with its human host in vivo. This is especially the case
cine candidates, which are best represented by Mos- for the first site in the infected body, namely, the liver
quirix/RTS,S and which have recently obtained market —the bridgehead of malaria infection where Plasmod-
authorization by the European Medicines Agency [7]. ium develops further after the bite of an infected

Abbreviations
AAV, Adeno-associated viral; CSP, circumsporozoite protein; ER, endoplasmic reticulum; FGF, fibroblast growth factor; HBV, Hepatitis B
virus; HCV, Hepatitis C virus; HGF, hepatocyte growth factor; HO-1, heme oxygenase 1; HSPGs, heparan sulfate proteogylcans; IFN, inter-
feron; iPSCs, induced pluripotent stem cells; ISGs, interferon-stimulated genes; L-FABP, liver-fatty acid-binding protein; LSEC, liver sinusoidal
endothelial cells; MAVS, mitochondrial antiviral signaling protein; MDA5, melanoma differentiation-associated gene 5; miRNA, microRNA;
MPCC, micropatterned cocultivation; MSC, mesenchymal stem cells; ORFs, open reading frames; PV, parasitophorous vacuole; ss, single-
stranded; TRAP, thrombospondin-related adhesion protein; UIS3, up-regulated in sporozoites 3.

FEBS Letters 590 (2016) 2027–2045 ª 2016 Federation of European Biochemical Societies 2027
Plasmodium meets AAV F. Hentzschel et al.

Anopheles mosquito. In principle, a single infectious instead, malaria pathology starts with the transition of
sporozoite reaching the liver and completing its intra- parasites from the liver into the blood where they
hepatic development suffices to give rise to up to repeatedly infect red blood cells. Hence, only interven-
10 000 merozoites capable of invading erythrocytes tions that target the clinically silent pre-erythrocytic
and causing malaria pathology [11,12]. Before produc- stages, that is, before the parasite enters the blood, can
tively invading a final host hepatocyte, sporozoites mediate complete sterilizing protection and thereby pre-
transmigrate through multiple hepatocytes, whereby vent malaria disease. In turn, this creates an urgent need
the parasites breach the cell plasma membrane and to fully dissect the molecular repertoire of intrahepatic
glide through the cytosol before exiting the cell [13] Plasmodium stages and their interplay with the host
(Fig. 1). Eventually sporozoites initiate a transforma- cells. It is, thus, even more puzzling how poorly these
tion process in their final host cell and develop into factors and mechanisms have been characterized to
liver trophozoites. Multiple rounds of DNA replication date, as compared to the pathogenic erythrocytic Plas-
and nuclear divisions without subsequent mitosis (i.e., modium stages and also considering that the intrahepatic
schizogony) then lead to the formation of a multinu- stages were discovered more than 60 years ago.
clear syncytium, the so-called schizont. Subsequent In this review, we first summarize recent pivotal
repeated invaginations of the schizont plasma mem- advances in identifying host factors that promote or
brane result in the generation of individual first-gen- restrict parasite liver-stage development, and point out
eration merozoites. Finally, the infected host cell remaining gaps in our knowledge of host contributions
detaches from its surroundings, and merozoites pack- to hepatic malaria infection. Subsequently, we high-
aged in vesicles called merosomes bud off the detached light newly emerging key technologies and strategies
cell and are released into the blood stream, to initiate that should ultimately permit dissection of parasite–
the intraerythrocytic stage [14]. host interactions within hepatocytes in much greater
Curiously, as noted, parasite propagation within hep- detail. Our specific focus is on (a) novel surrogate
atocytes is not accompanied by disease symptoms; models of whole livers that could replace animal

Fig. 1. Intrahepatic stages in the Plasmodium life cycle. (1) Traversal through Kupffer cell(s); (2) traversal through hepatocyte(s); (3)
attachment to final hepatocyte; (4) invasion via moving junctions; (5) dedifferentiation; (6) schizogony; (7) invagination of parasitophorous
vacuolar membrane; (8) egress. Indicated are major interacting host factors and, where occurring, their dysregulation (arrows up or down).
CSP, circumsporozoite protein; FABP, fatty acid binding protein; HSPG, heparan sulfate proteoglycans; IFN, interferon; ISG, interferon-
stimulated genes; LSEC, liver sinusoidal endothelial cell; PVM, parasitophorous vacuolar membrane; TRAP, thrombospondin-related
adhesion protein; UIS3, up-regulated in sporozoites 3.

2028 FEBS Letters 590 (2016) 2027–2045 ª 2016 Federation of European Biochemical Societies
F. Hentzschel et al. Plasmodium meets AAV

experimentation in the future, as well as on (b) recom- A limitation of these approaches, which focus on
binant Adeno-associated viruses as a powerful, versa- single factors, is that the assessment of individual pro-
tile, and ideally suited tool to study or perturb teins is labor-intensive and requires a priori knowledge
Plasmodium–host interactions ex or in vivo. of promising candidates. Therefore, a number of
groups conducted high-throughput screens with the
aim to evaluate the contribution of a wider variety of
Wanted ex or in vivo—host factors
potential host factors to Plasmodium liver-stage devel-
involved in intrahepatic Plasmodium
opment. Typically, these screens were based on siRNA
development
knock-down of a set of genes in human hepatoma cell
Sporozoite infection follows a very specific path in the lines prior to infection with the rodent parasite P. ber-
mammalian host, largely dictated by interactions with ghei. In one such comprehensive approach, 727 genes
distinct cellular factors. It is initiated by the extravascu- belonging to the human kinome were assessed individ-
lar inoculation of infectious single-celled sporozoites ually and stringently, resulting in the identification of
into the skin by anopheline mosquito bites [15,16]. Once five kinases that are required for P. berghei liver stages
deposited into the skin, they reach and invade the vascu- [29]. One of the hits was PKCf, a member of the PKC
lature to be passively transported by the blood flow until family. More specifically, it belongs to the atypical
they eventually arrive at the liver [17]. There, the infec- PKCs that are involved in various cellular processes
tious malarial sporozoites arrest at the liver endothelium such as cell growth and survival as well as regulation
due to a specific interaction between both, their major of NFΚB [30]. Further in vitro experiments revealed
surface proteins, circumsporozoite protein (CSP) and that PKCf is required for sporozoite invasion into
thrombospondin-related adhesion protein (TRAP), with hepatocytes, but not for liver-stage development. Inter-
highly sulfated heparan sulfate proteogylcans (HSPGs) estingly, mice that had received a siRNA-targeting
presented by stellate cells and hepatocytes [18–20]. PKCf had reduced liver burdens and exhibited a delay
Within the infected hepatocyte, the highly differentiat- in the onset of blood-stage parasitemia [29].
ing and replicative parasite resides in a membranous In a similar approach, all lipoprotein-related host
compartment. In addition to its own plasma membrane, factors—a total of 53 genes—were screened for their
this so-called parasitophorous vacuole (PV) physically contribution to Plasmodium infection, leading to the
separates the parasite from the host cell cytoplasm identification of the class B, type I scavenger receptor
[11,21] (see also Fig. 1). (SR-BI) as an important candidate [31]. The authors
To date, only few studies have investigated which of this work could show that SR-BI is required for
host-specific factors next to HSPGs are exploited by both, invasion and intracellular development of P. ber-
the parasite, and thereby contribute to the establish- ghei and P. falciparum, respectively. A lack of SR-BI
ment and maintenance of its intrahepatic develop- impedes liver-stage development in vitro and in vivo,
ment. The first host factors involved in Plasmodium albeit studies with blocking antibodies revealed that no
sporozoite invasion were identified either as a direct direct interaction is needed between SR-BI and the
protein interaction partner of a Plasmodium-specific infectious sporozoite [31–33]. It is intriguing to specu-
protein, or because they were already known to be late that the cholesterol, which is provided by SR-BI
important for infection with other hepatotropic patho- and incorporated into the plasma membrane, is impor-
gens such as Hepatitis C virus (HCV). In this manner, tant for parasite invasion; perhaps it may also serve as
the tetraspanin CD81, a surface molecule on hepato- a lipid source during growth. Notably, HCV entry
cytes and one of the main receptors for HCV [22,23], depends on SR-BI as well, and cholesterol and antago-
was identified as a crucial host factor for hepatocyte nists against this receptor are currently under clinical
invasion of both, P. falciparum and P. yoelii sporo- investigation [34–37].
zoites [23–25]. Similarly, Cha and coworkers deter- In the aforementioned studies, the assays were
mined a critical role of host CD68 as a putative restricted to a preselected group of genes due to the
receptor for sporozoite invasion and traversal of limitations of RNAi-based screening platforms. Also,
Kupffer cells [26]. A final relevant example is the host only loss-of-function effects on parasite development
protein L-FABP (liver-fatty acid-binding protein) that could be assessed, but not the natural response of the
was found to specifically interact with the PV mem- host cell to infection. In fact, transcriptional changes
brane-resident protein, UIS3 (up-regulated in sporo- upon productive hepatocyte infection are not restricted
zoites 3, [27]) within the infected hepatocyte, and to the Plasmodium parasite, but the host cell, too,
which is presumably required for fatty acid uptake responds with numerous alterations in gene expression.
into the parasite [28]. To begin to unravel these, Albuquerque and colleagues

FEBS Letters 590 (2016) 2027–2045 ª 2016 Federation of European Biochemical Societies 2029
Plasmodium meets AAV F. Hentzschel et al.

performed an unbiased assessment of the global host measurement of transcriptome changes in the murine
response to Plasmodium infection, using high-through- liver upon Plasmodium infection has revealed a strong
put microarray technology to measure genome-wide induction of 89 interferon-stimulated genes (ISGs)
transcriptional changes in murine hepatoma cells upon linked to the type I interferon (IFN) signaling pathway
infection with P. berghei [38]. Strikingly, they found [45,46]. It was demonstrated that the host hepatocyte
1064 host genes to be expressed differentially at one or senses intrahepatic parasite infection by pattern recog-
more of the selected time points (6, 12, 18, or 24 h nition of cytoplasmic Plasmodium RNA via the
post infection), and noted that transcriptional changes MDA5 (melanoma differentiation-associated gene 5)/
followed a coordinated and timed pattern. Based on MAVS (mitochondrial antiviral signaling protein)
their data, it seems that once infected, the host cell ini- pathway. This in turn leads to the transcription of
tially mounts a stress response to the presence of the IFN-a/b that finally triggers a type I IFN response in
parasite. This is followed by subsequent engagement of hepatocytes and leukocytes, in an autocrine and para-
diverse metabolic pathways, presumably to cope with crine manner. Knock-out of this type I IFN response
the parasite’s rapid proliferation and energy needs. At increased parasite liver infection, while an induction of
all times, genes involved in cell death and apoptosis type I IFN prior to Plasmodium infection led to a sig-
are regulated differentially in order to maintain viabil- nificant reduction in liver parasite load. While it
ity of the host cell [39–41]. Furthermore, the develop- remains unclear how parasite RNA reaches the hepa-
ing intrahepatic parasite is highly metabolically active tocyte cytoplasm, the study by Liehl et al. demon-
and up-regulates various pathways, such as redox, strates that, in contrast to previous assumptions, the
mitochondrial citric acid cycle, apicoplastic FAS II host innate immune response indeed recognizes and
pathway, and glycolysis, among several others [42]. acts against Plasmodium liver-stage infection [45].
Notably, some of the hits from the work mentioned Importantly, not all host responses to Plasmodium
above have already been further validated in follow-up infection can be measured as changes in mRNA levels.
studies in vivo. For example, In acio and colleagues Instead, the host cell might also respond with alter-
showed that parasite infection induces an ER (endo- ations on the translational or post-translational level,
plasmic reticulum) stress response in hepatocytes, acti- for example, by adjusting the phosphorylation status
vating proteins of the unfolded protein response of members of different signaling cascades. This vital
pathway [43]. Exogenous pharmaceutical activation of type of host responses would therefore be missed when
the ER stress response in vivo prior to infection with only quantifying transcript levels. Consequently, one
P. berghei sporozoites yielded a higher number of liver study by Kaushansky et al. addressed perturbations on
stages, but no size change (i.e., maturation status), at the protein level after Plasmodium infection using
24 h after infection. Another example is heme oxyge- reverse-phase protein microarray technology [47].
nase 1 (HO-1) which was identified as a main host fac- Although restricted to a subset of proteins by the anti-
tor required for rodent Plasmodium infection [44]. body selection, the authors were able to detect a gener-
Interestingly, while the initial screening that identified ally antiapoptotic, proliferative, and antiautophagic
HO-1 to be up-regulated upon infection was per- environment in the host cell. Specifically, the tumor
formed in hepatoma cell lines, immunohistochemical suppressor, p53, was largely decreased in infected hep-
stainings of liver sections revealed that HO-1 is atocytes, while the phosphorylated form of Bcl-2,
induced even more robustly in Kupffer cells (liver-resi- involved in antiapoptotic signaling, was elevated. A
dent macrophages). Congruently, mechanistic studies deliberate increase in p53 levels, either by overexpres-
revealed that HO-1 plays an anti-inflammatory role sion or by application of the drug, nutlin-3, that inhi-
protecting liver stages from the immune response of bits p53 degradation, reduced liver-stage burden
the host, and siRNA-mediated knock-down of this fac- in vitro and in vivo. Of note, drug-mediated increase in
tor in vivo nearly abolished liver-stage infection. This p53 is currently being evaluated in several clinical trials
example nicely illustrates that while in vitro screenings as a potential treatment for a variety of cancers [48].
in single cell types can provide clues on which factors This exemplifies how the study of malaria host factors
might be involved, it is key to dissect the parasite–host as therapeutic targets might benefit from synergisms
interactions in a more physiologically relevant system with other research areas sharing the same focus.
that encompasses all cell types, to fully understand the Next to proteins, an additional class of important
host’s contribution(s). host factors are microRNA (miRNA), roughly 22-
Accordingly, it should be rewarding to try and iden- nucleotide-long RNA that are master regulators of at
tify host factors related to Plasmodium infection least 60% of all mammalian genes and as such play
directly in vivo in the mouse liver. Indeed, a recent central roles in pathological processes, including cancer

2030 FEBS Letters 590 (2016) 2027–2045 ª 2016 Federation of European Biochemical Societies
F. Hentzschel et al. Plasmodium meets AAV

and pathogen infections [49]. Recently, we dissected deliberately, specifically, and robustly control the
the effects of Plasmodium sporozoite infection on host expression of putative host factors in live animals and/
miRNA expression under physiologically relevant con- or in these new surrogate systems, including gene over-
ditions in livers of adult mice [50]. Combined microar- expression or inhibition, in order to fully dissect their
ray and PCR screens using total liver RNA obtained role in the hepatic Plasmodium stages. Above, we have
after infection with either wild-type or attenuated Plas- already briefly mentioned AAV vectors as one possible
modium strains revealed not only an up-regulation of solution for this second challenge. We further make a
IFN-associated genes (confirming and extending previ- case for these particular tools below, by highlighting
ous work, see above and [2,3]) but also a dysregulation most recent progress in their successful use in livers in
of 11 of 698 studied miRNA. In particular, by focus- in vivo or in ex vivo organotypic models, including
ing on known immunoregulatory miRNA, we found their engineering toward functionality in human hepa-
miR-155 to be highly up-regulated upon Plasmodium tocytes. First, however, we discuss selected examples
liver-stage infection. Moreover, we could identify of promising liver replacement systems for the study of
Kupffer cells as the main source of this particular intrahepatic Plasmodium stages (Fig. 2).
miRNA in the adult mouse liver. Intriguingly, miR-
155 is one of the best characterized proinflammatory
Back to the future—From whole livers
miRNA and plays a fundamental role in mammalian
to organotypic models and back to
host defense mechanisms against pathogen challenge
chimeric organs
[51]. In the context of Plasmodium, we thus concluded
that miR-155 boosts the immune response against liver At first glance, it may appear straight-forward to use
stages. Additional evidence was that exogenous overex- established hepatic cell lines as a liver surrogate, con-
pression of this miRNA using hepatotropic Adeno- sidering the benefits of indefinite growth and limited
associated viral (AAV) gene delivery vectors improved costs that are inherent to transformed cell lines. Yet,
the vaccination of mice with genetically attenuated there are numerous reasons that argue against this par-
parasites [3,4]. In turn, this reduced the number of ticular strategy, most notably the fact that the histo-
immunizations needed to achieve sterile protection typic and phenotypic characteristics of cell lines as
against rechallenge with wild-type parasites from three well as their gene/miRNA expression profiles differ
to one [50]. substantially from their cellular counterparts within an
Particularly noteworthy about this study is that it intact liver. Adding to the seminal differences is that
illustrates two major persisting challenges in the field cells in culture typically grow in two dimensions (2D),
of malaria liver-stage research that need to be over- whereas in the liver, hepatocytes and all the other cell
come and that are in the focus of the remainder of this types are embedded in a three-dimensional (3D) envi-
article. (1) The first is related to the facts that (i) the ronment providing special architecture and hemody-
infection rate of liver stages is typically very low, namic properties. Using primary liver cells instead of
implying that subtle changes around the local infection cell lines increases the physiological relevance but does
site are easily overlooked among the vast majority of not solve the problem of an unnatural and inadequate
uninfected cells, and that (ii) there are differences microenvironment, comprising the lack of an extracel-
between rodent and human Plasmodium strains that lular matrix. Moreover, primary cells are difficult to
have to be accounted for. For example, P. berghei obtain and culture in a differentiated state, particularly
infection seems to be supported by the hepatocyte for longer periods. The latter, however, may be
receptor c-Met that is activated by the hepatocyte required to fully mimic processes that can take days or
growth factor, while this is not the case for P. falci- weeks in the intact liver, such as the establishment of
parum infections [52,53]. In addition, (iii) although an anti-Plasmodium immune response.
rodent models are physiologically highly relevant, their For all these reasons, major efforts have been under-
routine use is hampered by ethical, financial, and logis- taken and are currently intensifying to create more
tical concerns that are inherently associated with ani- sophisticated, multicellular, and organotypic ex vivo
mal experimentation. Consequently, there is a strong liver models that are as close as possible to the natural
desire to implement and harness novel surrogate environment, and that are ideally also compatible with
ex vivo models of an intact liver or of specific hepatic medium- to high-throughput experimental strategies.
cell types that circumvent the use of live animals, and Examples for such systems that have been developed
that can be customized for a given Plasmodium strain and tested before include 3D sandwich cell cultures,
and/or experimental question. (2) The second challenge microfluidic perfusion arrays, bioreactors, biochips, or
in the field is to concurrently devise means to hanging droplets, to name a few. These systems find

FEBS Letters 590 (2016) 2027–2045 ª 2016 Federation of European Biochemical Societies 2031
Plasmodium meets AAV F. Hentzschel et al.

Fig. 2. Surrogate liver models to study Plasmodium liver stages. (A) Types of ex vivo cell culture systems and cell sources. See text for
details. (B) In vivo systems. Top: Creation of humanized mice by partial depletion of murine (mu) hepatocytes, followed by transplantation
of, and liver repopulation with, human (hu) hepatocytes. Bottom: Isolated livers can be decellularized using combinations of physical and
chemical methods, followed by recellularization of the remaining liver scaffold with human hepatocytes (and other cell types).

broad utility in biomedical research, from hepatotoxic- (both rodent), falciparum, and vivax (both human).
ity testing of pharmaceutical compounds, to investiga- This was in line with the typical hepatocyte morphol-
tions into liver cell physiology and pathology. For ogy of the iHLCs as well as their expression of proto-
more details on these various systems, we refer the typical hepatocyte markers such as albumin and
reader to excellent previous review articles (e.g., [54– alpha-1-antitrypsin. Moreover, they expressed SR-B1
56]). In the following, we will specifically highlight two and CD81, which are both host entry factors for Plas-
ex vivo cell culture models that have recently been modium (as well as for HCV). Further interesting was
shown to support Plasmodium liver-stage infections that the cells acquired Plasmodium susceptibility
and that, in our opinion, hold great promise for future already at the hepatoblast stage on day 15 in the dif-
malaria research for reasons explained below. ferentiation protocol, albeit liver-stage development
The first of these two systems, reported by Ng and seemed to be delayed or perturbed as compared to the
colleagues in 2015 [57], capitalizes on the possibility to more mature cells on day 20. Finally noteworthy is
differentiate induced pluripotent stem cells (iPSCs, that additional treatment of the cells with the small
[58,59]) into so-called iPSC-derived hepatocyte-like molecule FPH1 advanced the maturation process and
cells or, for short, iHLCs (for review see [60]) eventually rendered the cells sensitive to the anti-
(Fig. 2A). This was achieved using a 20-day differenti- malaria drug primaquine, exemplifying the potential of
ation protocol involving iPSC culturing in a mono- this system for future phenotypic drug screens.
layer on matrigel and exposure to various agents As compared to established hepatocyte cell lines, the
including fibroblast growth factor (FGF), hepatocyte use of iHLCs provides a variety of advantages, most
growth factor (HGF), and oncostatin M. Remarkably, important of which is that they more accurately reca-
as demonstrated via immunostaining for specific mark- pitulate parasite–host interactions. At least from a
ers of parasite liver stages (HSP70 and MSP-1), the technical standpoint, the iHLC system is also superior
iHLCs were susceptible to infection with sporozoites to primary human cells since the iPSC-derived cells are
from four different Plasmodium species—berghei, yoelii renewable in culture. Another unique trait and benefit

2032 FEBS Letters 590 (2016) 2027–2045 ª 2016 Federation of European Biochemical Societies
F. Hentzschel et al. Plasmodium meets AAV

is that iPSCs and thus iHLCs can be derived from hepatocytes. However, the liver comprises a variety of
somatic cells from any human donor, offering the pos- other cell types that are most likely also involved in
sibility to include a broad spectrum of the population Plasmodium infection and establishment of liver stages,
in future assays with this system. This is essential as it especially Kupffer cells, stellate cells, and liver sinu-
will permit to assess the effects of different genotypes soidal endothelial cells (LSEC). In fact, there is evi-
related to highly polymorphic genetic variants and dence that prior to hepatocyte infection, the parasite
diverse ethnic groups. In turn, this equips these cells traverses Kupffer cells and LSECs, and that this may
with a much higher predictive value in drug screens or be essential for subsequent liver-stage formation
biological assays than primary cells derived from a sin- [63,64].
gle donor. On top of the natural heterogeneity, it In this respect, a second novel system should be
should be possible to deliberately modulate the gen- highlighted that has recently been reported by March
ome of iPSC/iHLCs, by exploiting easily accessible and colleagues [65,66]. This study is noteworthy as it
and customizable CRISPR gene-editing technology. describes a robust and multicellular ex vivo platform
Notably, the two essential CRISPR components (guide that recapitulates the complete liver stage of Plasmod-
RNA and Cas9 nuclease) can be delivered by recombi- ium sporozoite infection, including progression to the
nant AAV vectors (see also below), which makes the schizont and merozoite stages. In more detail, the hall-
combination of these powerful technologies—CRISPR, mark of this platform is micropatterned cocultivation
iHLCs, and AAV—very promising. Support for this (MPCC, [67]) of cryopreserved primary human hepato-
enthusiasm comes from data provided by us and cytes together with supportive stromal cells (murine
others (A-K. Herrmann & D. Grimm, unpublished embryonic fibroblasts) (Fig. 2A). Notably, the hepato-
results and [61]) showing that iPSC/iHLCs can be cytes grown in this system polarize, exhibit typical
transduced very efficiently with AAV vectors, depend- drug and energy metabolism, do not proliferate, and
ing on the capsid variant used for vector generation. maintain a functional phenotype for up to 6 weeks. In
In this context, a recent study by Egan et al. [62] is addition, high reproducibility is guaranteed through
worth highlighting as well, in which the authors used the use of cryopreserved human cells, which minimizes
a forward shRNA screen to identify host factors for donor-to-donor interexperimental variability, and like-
P. falciparum blood-stage development within the ery- wise through the use of cryopreserved, purified, vialed,
throcyte. As these cells are denucleated and therefore and aseptic sporozoites (P. falciparum or vivax). Using
impervious to genetic modification, the shRNA were a battery of assays, March and coworkers were able to
instead delivered to hematopoietic progenitor cells, validate the viability of these sporozoites and to prove
which were subsequently matured into erythroblasts their ability to mature into hepatic schizonts. Remark-
and then infected with the parasite. The fact that this ably, the latter were also shown to release merozoites
permitted the identification of CD55 as an essential that were capable of infecting human red blood cells,
factor for parasite invasion into the erythrocyte further which were added to the MPCC culture.
exemplifies the power of stem cell-based strategies and In comparison to regular cultures of primary human
thus complements the work by Ng and colleagues [57]. liver cells, an advantage of this system and a promis-
Taken together, the key features of the iHLC system ing feature for its future use in malaria research is that
—natural genetic diversity, possibility of deliberate it combines high reproducibility, robustness, and relia-
mutation, and susceptibility to Plasmodium sporozoite bility, with the possibility to maintain and study func-
infection during iPSC-to-iHLC differentiation—suggest tional cellular phenotypes over extended periods of
a large potential for the study and dissection of para- time. Equally remarkable is the ability of this platform
site–host interactions. To even further enhance these to support the full Plasmodium liver-stage infection,
prospects, it would be beneficial to standardize the dif- from sporozoite invasion to schizont/merozoite release.
ferentiation protocol to facilitate its rapid implementa- In this regard, it may even surpass the capabilities of
tion in other laboratories. Additionally, efforts should the iHLC system highlighted above (albeit a direct
be undertaken to improve the degree of cell matura- comparison has not yet been performed). However, a
tion, to ensure that the iHLCs truly mimic the host potential drawback of the MPCC approach may be
factor repertoire of primary hepatocytes and thus even the lower amenability to stable genetic engineering for
more accurately recapitulate the situation in an intact it is based on nonproliferating primary hepatocytes,
liver. rather than on expandable stem cells. In fact, the
Importantly, even with all these improvements in authors had to prescreen various batches of cryo-pre-
place, a limitation of the current iHLC system remains served cells in order to eventually identify two which
its concentration on a single cell type, that is, mediated high-level sporozoite infection; similar

FEBS Letters 590 (2016) 2027–2045 ª 2016 Federation of European Biochemical Societies 2033
Plasmodium meets AAV F. Hentzschel et al.

variations in efficiency were also noted for three differ- moreover able to demonstrate potent transduction of
ent frozen parasite batches. Together, this suggests the engrafted cells with AAV vectors. This again exem-
that it may be more challenging with this particular plifies the great potential of these vectors for func-
system to cover the natural genotypic variation in the tional studies in ex vivo liver models, as already noted
human population, and hence the predictive value may above in the context of the iHLC strategy and as fur-
be lower than that of the iHLC approach. Moreover, ther discussed in the next chapter. Akin to the other
to fully mimic an intact liver, the MPCC system will organotypic systems, it will now be important to
need to be expanded by other physiologically relevant include additional cell types that represent the physio-
cell types, especially Kupffer cells, which are likely logical situation better, especially Kupffer cells and
required for Plasmodium liver infection [26,63]. LSECs. To this end, it may again be beneficial to com-
In conclusion, there are numerous promising ave- bine the best of multiple strategies, for example, to
nues in the field of ex vivo liver surrogate models for replace the HepG2 cells in the rat liver scaffold with
malaria research, two of which were highlighted above MSC or iPSC, and to then differentiate them into vari-
—use of iPSC as an unlimited cell source and multicel- ous hepatic cell populations.
lular coculture systems. An intriguing and rewarding Last but not least, we wish to point out another
next step should be to try and juxtapose the best fea- related strategy, which is in vivo repopulation of livers
tures of the two strategies, that is, to establish robust with human hepatocytes in a living mouse (Fig. 2B
protocols for iPSC differentiation into other relevant top). Therefore, a subpopulation of the murine hepato-
hepatic cell types, such as Kupffer cells, and to then cytes is destroyed deliberately (e.g., by overexpression
combine all the different cell types in a single of a suicide gene), triggering liver regeneration and
micropatterned culture vessel. Ideally, this will occur creating a short window during which human hepato-
in a 3D format comprising an adequate extracellular cytes can be transplanted and engrafted. The resulting
matrix and fully mimicking the architecture and hemo- ‘humanized’ mice support the life cycle of the human
dynamics in an intact liver. In this respect, we also Plasmodium strains P. falciparum and P. vivax [71–74],
wish to note a recent series of encouraging reports on thus complementing wild-type mice that are routinely
newly developed bioartificial 3D scaffolds (Fig. 2B used to propagate the rodent parasite strains. How-
bottom). This includes a study by Schanz et al. [68] ever, such human-chimeric mice are costly and labori-
who extracted small bowel segments from pigs, chemi- ous to generate. Moreover, one should consider that
cally removed the residual porcine cells, seeded the without further transplantation of, for instance,
vascular structures with human endothelial cells, and human immune cells or hematopoietic stem cells, all
eventually populated this vascularized scaffold with the other cell types in these mice remain of murine ori-
primary human hepatocytes. While not tested yet for gin. Importantly, as described in more detail below, it
susceptibility to Plasmodium infection, the fact that the is possible to engineer AAV vectors that readily trans-
engrafted hepatocytes were morphologically intact as duce the human hepatocytes within these chimeric liv-
well as functionally and metabolically highly active is ers, again illustrating the potential of this particular
certainly auspicious. Even more promising may be, vector for malaria research in various ex or in vivo
however, to engineer decellularized liver scaffolds liver models.
instead of bowel tissue as a biomimetic 3D matrix pro-
viding optimal gas and nutrient diffusion. In one
Fighting fire with fire—engineering of
example, Jiang et al. [69] used a combination of physi-
hepatotropic viruses to dissect and
cal and chemical methods to decellularize whole
control Plasmodium liver stages
extracted livers of mice, which were subsequently
repopularized with mesenchymal stem cells (MSC) and Adeno-associated viruses are a large group of viruses
attached to a perfusion culture system. Notably, the that are endemic in many species including humans,
MSC efficiently and rapidly differentiated into HLC and that have been studied intensely for the past five
showing typical hepatocyte morphology, gene expres- decades. One reason for their attractiveness is their
sion, and functionality that was maintained even after unique biology, since AAVs strictly depend on coin-
transplantion of the entire recellularized scaffold into fection with a second unrelated, so-called helper
recipient mice. Similarly, the Kurreck group has more virus, typically Adenovirus or Herpes simplex virus.
recently reported successful decellularization of iso- This is because AAV has only two open reading
lated rat liver 3D scaffolds and subsequent repopula- frames (ORFs; Fig. 3A left) whose differential
tion with HepG2 cells, a human hepatoma cell line expression results in eight proteins which play essen-
[70]. Particularly noteworthy is that the authors were tial roles in viral gene expression, replication, and

2034 FEBS Letters 590 (2016) 2027–2045 ª 2016 Federation of European Biochemical Societies
F. Hentzschel et al. Plasmodium meets AAV

Fig. 3. Adaptation of the AAV vector system for use in malaria research. (A) Left: Wild-type genome consisting of two open reading frames
rep and cap, which are flanked by inverted terminal repeats (ITR, typically derived from AAV2). Right: Three possible recombinant vector
configurations, that is, (i) a conventional single-stranded vector containing a transgene expression cassette of up to 4.5 kb, (ii) a self-
complementary genome for expression of shRNA (usually together with a second cassette, for example, a gfp reporter), and (iii) a similar
variant of self-complementary AAVs for gRNA expression (including a Cas9-specific gRNA scaffold, shown in orange). Note that the self-
complementary genotype in (ii) and (iii) is created through specific mutation of one of the ITRs (indicated with the asterisk, see main text for
details). (B) Four options to retarget AAV vectors to specific liver cell types of potential interest for malaria research. Specificity can be
achieved through either (i) modification of the target cell surface, resulting in de novo binding of a selected AAV capsid; (ii) vice versa,
genetic modulation of the AAV capsid surface, creating novel binding sites for particular cellular receptors; (iii) selection of cell type-specific
promoters (arrows); and/or (iv) inclusion of binding sites for miRNA (red triangle) that are abundant in off-target cells where gene expression
is unwanted, but missing in the actual on-target cell(s). HC, hepatocyte; KC, Kupffer cell; LSEC, liver sinusoidal endothelial cell.

capsid formation, but which fail to mediate the cloning techniques [78]. Thirdly, there are over 100
entire AAV life cycle. To compensate for this defi- naturally occurring isolates of AAV which differ in
ciency, AAV hijacks multiple proteins and RNA (in cell specificity and infection efficiency, providing a
the case of Adenovirus) from its helper virus, to fos- vast toolkit for vector engineering [79]. To even fur-
ter its own transcription, translation, and particle ther expand on this natural diversity, we and others
egress [75]. Yet, even more appealing than this implemented a variety of methods for molecular evo-
intriguing biology is an exclusive combination of fea- lution of ‘designer’ AAV capsids that are tailored for
tures that makes AAV highly attractive and impor- a given application, for example, transduction of a
tant as a basis for recombinant vectors for human specific cell type in the presence of neutralizing anti-
gene therapy. First and foremost, AAV is believed to AAV antibodies [80–82].
be apathogenic in humans, and hence AAV vectors In the following sections, we discuss why we believe
are considered as the safest of all recombinant that AAV vectors are ideally suited as a tool to study
viruses currently under (pre-) clinical investigation. In Plasmodium liver stages in ex or in vivo model systems,
fact, there has not been a single case of serious based on accumulating data from the recent literature
adverse events in any human patient treated with and from our own research.
AAV vectors in over 100 clinical trials thus far
[76,77]. Secondly, the viral genome is very easy to
Versatility of the genome
engineer due its small size (roughly 4.7 kilobases)
and due to the availability of AAV vector plasmids In principle, there are at least three possibilities to
that can be readily customized using standard identify, validate, or further dissect Plasmodium host

FEBS Letters 590 (2016) 2027–2045 ª 2016 Federation of European Biochemical Societies 2035
Plasmodium meets AAV F. Hentzschel et al.

factors and associated networks in liver cells: (a) over- begin with, we strongly recommend the use of scAAV
expression, (b) RNAi-mediated inhibition (knock- vector genomes.
down) on the mRNA level, and (c) nuclease-mediated Unlike the situation with cDNA that vary in size,
suppression (knock-out) on the DNA level (Fig. 3A scAAV vectors are always the preferred option for
right). Importantly, all three are fully compatible with strategies requiring knock-down or knock-out of Plas-
AAV vectors, as extensively demonstrated by us and modium host factors. This is because the expression
others in the past. Their implementation is facilitated cassettes to trigger RNAi with shRNA, or CRISPR
by the aforementioned fact that there are widely avail- with g(uide)RNA, respectively, are typically in a range
able, user-friendly, high-copy number AAV vector of 100–700 bp, depending on the promoter used for
plasmids. They contain the only viral element which is small RNA expression (e.g., H1: 100 bp, or U6:
required for vector generation in cis, namely, the 600 bp) [85]. This is far below the packaging limit of
inverted terminal repeats or ITRs. These are short scAAV vectors and thus even permits the inclusion of
(about 145 bp) hairpin sequences that flank the viral a second expression cassette, for instance encoding a
genes or the transgene expression cassette, respectively, fluorescence marker to track the targeted cells
and mediate its replication and encapsidation. Since (Fig. 3A[ii/iii]). In fact, we have recently created a
the two AAV ORFs can be replaced entirely with for- toolbox of scAAV vector templates that allow for
eign DNA, and since the capsid can package slightly coexpression of any shRNA or gRNA together with
oversized DNA (up to 120% of the length of the wild- various fluorescence reporters, and that can be easily
type genome), an AAV vector construct can accommo- customized for any desired Plasmodium host factor
date roughly 5.2 kb of DNA. Considering a typical ([86], and F. Schmidt, K. B€ orner & D. Grimm, unpub-
size of promoter and polyadenylation elements in a lished results). This vector collection, which is freely
range of 1 kb, this leaves about 4 to 4.5 kb for the available upon request, should largely foster and accel-
cDNA of choice, which is probably sufficient for the erate the future dissection of parasite–host interactions
majority of mammalian cDNA of interest (Fig. 3A[i]). ex or in vivo.
Notably, in case the cDNA is sufficiently short (less Notably, CRISPR-based knock-out of Plasmodium
than 2.4 kb including promoter and polyadenylation host factors will also entail expression of the Cas9
signal), there is also the option to embed it in a ‘self- nuclease, whose currently known orthologs are in a
complementary’ (sc)AAV vector genome. This is a size range of 3.1–4.2 kb [87,88]. Since all of these
variant of standard AAV vectors in which one of the cDNA exceed the capacity of scAAV vectors, coex-
ITRs is mutated by deleting the so-called ‘terminal res- pression of Cas9 from an AAV vector requires the use
olution site’, that is, a short sequence that is normally of ss genomes which, as noted before, express more
nicked during replication of the AAV vector genome slowly and less efficiently than sc vectors (encoding the
from single-stranded (ss) to double-stranded DNA. gRNA). Still, this is no major limitation as recently
[83]. Consequently, scAAV genomes package as a demonstrated by a variety of reports describing very
molecule consisting of two inverted copies of the trans- efficient AAV-mediated Cas9 expression and gRNA-
gene; hence the reduction in the capacity for the for- induced gene editing in cultured cells and livers of
eign DNA cassette to about 50% of the wild-type adult mice [86,89–91]. In addition, the increasing avail-
genome (Fig. 3A[ii/iii]; the mutated ITR is labeled ability of stable Cas9-expressing cell lines or of Cas9-
with an asterisk). Importantly, scAAV vectors express transgenic mice [90] will soon entirely alleviate the
more rapidly and more robustly than conventional need for vector-mediated Cas9 expression, at least in
AAV vectors, because the latter package as ssDNA an exploratory context.
which has to be converted into a double-stranded
molecule prior to transcription. It may thus take a
Efficiency in liver cells
ssAAV vector up to 2 weeks to reach the peak of
transgene expression in cultured cells or in the liver, as Adeno-associated viral vectors are predestined for use
compared to a few days for a scAAV counterpart in the liver or in isolated liver cells due to the fact that
[83,84]. Therefore, one should carefully weigh the pros this organ is one of the main natural targets for the
and cons of the two possible vector configurations wild-type virus, in particular for the most commonly
when designing an experiment involving overexpres- used AAV serotypes including the AAV2 prototype.
sion of a (putative) Plasmodium host factor from AAV This explains why vectors derived from these serotypes
vectors—larger capacity (ss vectors) versus more potent can readily transduce 100% of the hepatocytes in an
and faster transgene expression, at the cost of size (sc intact mouse liver, as consistently found with, for
vectors). Clearly, if the latter has no limitation to example, AAV8 or AAV9 vectors by us and many

2036 FEBS Letters 590 (2016) 2027–2045 ª 2016 Federation of European Biochemical Societies
F. Hentzschel et al. Plasmodium meets AAV

others [84,92,93]. Of note, this complete liver transduc- Glybera, is derived from a recombinant AAV serotype
tion is observed after peripheral vector application via 1 vector [97].
tail vein injection, which is a simple and rapid proce- In addition to their great potential in vivo, AAV
dure that can be quickly adapted in any laboratory. vectors are likewise highly efficient in liver cell lines or
One should also not get discouraged by the AAV par- primary cells that are cultured ex vivo. This comprises
ticle numbers that have to be administered to achieve numerous hepatoma cell lines, such as Huh7 or
100% transduction in vivo, and that are typically in a HepG2, all of which can be transduced very robustly
range of 1 9 1011 per mouse or higher. In fact, thanks with, for example, AAV2 or AAV6 vectors. Impor-
to the highly advanced AAV vector production tech- tantly, these cell lines retain their high susceptibility to
nology, it is feasible to generate stocks with titers of AAV transduction even when grown in a 3D environ-
greater than 1 9 1013 particles per mL within 2 weeks, ment rather than a 2D monolayer. This has recently
using standard cell culture techniques that are already been exemplified in the aforementioned study by Wag-
available in the majority of laboratories [78]. ner and colleagues [70] who engrafted HepG2 cells
Next to the mouse, AAV vectors are also very effi- onto decellularized rat liver scaffolds and then demon-
cient at transducing livers of larger animal species, strated wide-spread and efficient transduction with an
such as dogs and nonhuman primates. While not all AAV6 vector. Comparable findings were made with
of these animals are already used in malaria research, primary mouse or human hepatocytes that can be
it is important to realize that, in principle, the same transduced with various AAV serotypes when cultured
vector constructs exploited in mice to identify or vali- in both, 2D or 3D structures such as spheroids [80,98]
date parasite host factors can also be translated into (K. Börner & D. Grimm, unpublished results).
these larger animals. This is particularly beneficial for In view of these attractive features, it is not surpris-
candidates that hold biomedical potential as malaria ing that one can find an exhaustive list of published
therapeutics or vaccines, and hence require extensive examples for biomedical AAV vector applications in
preclinical investigation in different small and large cultured liver cells or whole livers in vivo, including in
animal species. Likewise, important in this context is humans as mentioned above. Particularly noteworthy
to again highlight the extensive clinical experience with with respect to the topic of the current article are
AAV vectors in humans, which has been gathered in studies showing the successful development and use of
over 100 patient trials thus far and which is utmost AAV vectors expressing shRNA or shmiRNA (differ-
encouraging. Most notable with respect to malaria ent types of RNAi triggers) directed against liver
research on Plasmodium liver stages are the latest clini- pathogens. For instance, we and several other groups
cal trials with scAAV8 vectors in patients suffering have previously engineered scAAV vectors from hepa-
from hemophilia B, a bleeding disorder caused by totropic serotypes, such as AAV7, 8, or 9, to express
defects in the human blood clotting factor IX. Follow- shRNA against Hepatitis B virus (HBV) and then
ing a single peripheral AAV vector administration, the proved their efficacy in cultured liver cell lines as well
majority of treated patients expressed stable factor IX as in livers of HBV-transgenic mice [84,99,100]. Best
levels above the critical threshold needed to provide results include the robust and persistent suppression
therapeutic benefit (50 ng per mL, that is, 1% of nor- of HBV gene expression in adult mice for over 1 year,
mal) [94,95]. None of the patients, neither in this trial achieved with a scAAV8 vector that was optimized
nor in any other AAV-based clinical study experienced for hepatocyte-specific shRNA expression from a
serious adverse events related to the vector, again illus- RNA polymerase II promoter [99]. An important les-
trating the safety of the recombinant AAV system. It son learned in this and other in vivo studies using
is finally also essential in this context to understand AAV/RNAi vectors is that vector-mediated shRNA
that a recent report claiming an association of AAV expression needs to be tightly controlled, in order to
infection with hepatocellular carcinoma in humans avoid adverse oversaturation of the endogenous cellu-
attributed this effect to wild-type AAV2, but not to lar miRNA pathway and ensuing cytotoxicity. This
AAV vectors [96]. Moreover, this study has been risk was first discovered by us in a 2006 study in
highly criticized within the AAV community for which we had inadvertently triggered liver damage
potentially overstating the results [76,77], and based and animal fatalities during our evaluation of a bat-
on the aforementioned clinical evidence that AAV tery of anti-HBV AAV/shRNA vectors in livers of
vectors are exceptionally safe in human patients. HBV-transgenic mice [84]. As these effects are pre-
Besides, further evidence for the safety and promise of dominantly dose-dependent, we and others subse-
AAV vectors in humans is that the first gene therapy quently designed optimized AAV vectors that use
product approved in the Western world in 2012, weaker and/or hepatocyte-specific promoters (see

FEBS Letters 590 (2016) 2027–2045 ª 2016 Federation of European Biochemical Societies 2037
Plasmodium meets AAV F. Hentzschel et al.

above) for shRNA expression and thus alleviate the Adaptability of the capsid
oversaturation concern. In addition, concurrent over-
As described in the previous two paragraphs, there are
expression of Exportin-5 and Argonaute-2, two major
a variety of naturally occurring AAV serotypes that
rate-limiting factors in the mammalian RNAi path-
robustly transduce cultured liver cell lines (e.g., AAV2
way, can further improve AAV/RNAi vector safety
or AAV6) or livers of newborn or adult mice (e.g.,
and efficiency in cells and mouse livers [84,101,102].
AAV8 or AAV9). Important to point out, the hepatic
Yet another critical parameter is the on- versus off-
cell type that is predominantly targeted by all these
target specificity of the encoded shRNA, which has to
capsid variants are hepatocytes (or hepatocyte-derived
be considered for both shRNA strands, that is, the
cell lines, respectively). In contrast, other, non-
antisense strand directed against the actual target as
parenchymal liver cell types that are relevant and
well as the passenger strand. In fact, we have found
interesting in the malaria liver-stage context, that is,
recently that both shRNA strands are usually active,
Kupffer cells, stellate cells, and LSECs, are largely
and that deliberate suppression of passenger strand
refractory to transduction with wild-type AAV capsids.
activity using a ‘tough decoy’ inhibitor improves
In our previous work, we did observe AAV8 activity
potency and efficiency of AAV/shRNA vectors target-
in Kupffer cells in mouse livers following systemic vec-
ing HCV or HBV [103] (T. Michler, S. Grosse,
tor administration, but the efficiency was clearly below
M. Heikenwalder, U. Protzer & D. Grimm, unpub-
that in hepatocytes in the same animals [50]. We have
lished results). Altogether, these findings yield a vari-
recently made similar findings for another wild-type
ety of options to fine-tune the activity and in
serotype, AAV6, and stellate cells in mouse liver
particular the safety of shRNA-expressing AAV vec-
(D. Grimm & H. Willenbring, unpublished results).
tors for use in animal or human liver in vivo, which
Fortunately, there are at least two major possibili-
provides a very solid basis for their future develop-
ties to improve AAV targeting to nonhepatocyte cell
ment in the context of malaria research. To this end,
types in the liver (Fig 3B). One is to alter the host
it is highly beneficial that AAV/RNAi vectors can
environment, or, more specifically, to modify the sur-
also be evaluated and optimized in ex vivo 3D organ-
face composition of the cells of interest, in order to
otypic liver models, as recently exemplified by up to
increase the binding avidity of AAV serotypes (Fig 3B
90% knock-down of human cyclophilin B gene
[i]). However, a drawback of this strategy, especially
expression in a recellularized rat liver scaffold [70].
when used in combination with in vivo Plasmodium
Next to RNAi, similarly encouraging data are also
infection, is that it globally alters the configuration of
already available for the combination of AAV vectors
cells in the liver and is thus prone to creating artifacts.
and the CRISPR gene editing system in liver cell lines
Nonetheless, a notable example was reported by the
or mouse livers in vivo, although CRISPR technology
Asokan group who treated mice with intravenously
is much younger than RNAi. Most notable results
administered recombinant sialidase, knowing that this
thus far include a report of over 40% in vivo modifica-
would lead to cell surface exposure of N-terminal
tion of the Pcsk9 gene, resulting in significant reduc-
galactose, a receptor for AAV9. Indeed, this resu-
tions in serum cholesterol [89], as well as a recent
lted in a marked change in the overall tropism of
study achieving functional correction of a mutation in
AAV9, including a sequestration in Kupffer cells and
the ornithine transcarbamylase gene in livers of new-
LSECs [105].
born mice [104]. In both cases, appropriate gRNA
Because of the practical restrictions of this
were delivered with an AAV8 vector together with
approach, it is more adequate and more promising for
Cas9 from Staphylococcus aureus, the smallest of all
malaria research to pursue a second strategy to retar-
currently known Cas9 variants (3.1 kb); the latter was
get AAV vectors to nonparenchymal liver cells,
either encoded in a second [104] or on the same [89]
namely, genetic modification of the viral capsid
vector. Moreover, as noted above, our own groups
(Fig 3B[ii]). For details on the underlying complex
have reported a user-friendly AAV vector toolbox for
technology, we refer the reader to a set of excellent
cloning and expression of custom gRNA, alone or
recent reviews [81,106–108]. Here, suffice it to highlight
from an ‘all-in-one’ vector also comprising the Cas9
what we consider as the most versatile strategy for
cDNA [86]. Collectively, these first in vivo data and
AAV capsid diversification, that is, ‘shuffling’ of the
the availability of customizable gRNA templates
capsid genes from multiple different AAV serotypes
should pave the way for the looming application of
through enzymatic fragmentation and subsequent
the CRISPR system as a novel, powerful, and versatile
reassembly based on partial homologies. In 2008, we
tool to further dissect Plasmodium liver stages and
have introduced this technology into the AAV field
associated host factors.

2038 FEBS Letters 590 (2016) 2027–2045 ª 2016 Federation of European Biochemical Societies
F. Hentzschel et al. Plasmodium meets AAV

and used it to molecularly evolve the capsid AAV-DJ, fusions with, for example, C3d3 [115] or CTLA4-Ig
a unique chimera between wild-type serotypes AAV2, [116]. Curiously, all these efforts resulted in antigen-
8, and 9 that combines high activity in both, cultured specific immunity but failed to elicit protection against
cells including hepatoma lines and livers (hepatocytes) Plasmodium infection, implying that further improve-
of adult mice [80]. In addition, more recently [109], ments to this particular approach are required. Sec-
Lisowski and colleagues reported the capsid LK03 ondly, Huang and colleagues exemplified the
which had been enriched from a shuffled AAV capsid usefulness of AAV9 vectors to foster reconstitution of
library by iterative screening in livers of human-chi- immunodeficient mice with a human immune system
meric mice. Interestingly, this capsid appears to have a [117]. Therefore, the vectors were engineered to deliver
higher efficiency in human hepatocytes than AAV8, a cocktail of HLA class II genes, genes encoding vari-
which was tested in parallel as the gold standard ous human cytokines and human B-cell activation fac-
(based on its high efficiency in mouse hepatocytes). tor. Vector administration followed by engraftment of
Most likely, the elevated potency of LK03 is related to hematopoietic stem cells resulted in humanized mice
the fact over 95% of its sequence are identical to wild- carrying human CD4+ T- and B-cells. Notably, these
type AAV3, which others also found to work well in animals could be immunized with recombinant P. fal-
human hepatocytes [110,111]. ciparum CSP and were subsequently protected from
Because these data clearly illustrate the tremendous in vivo challenge with sporozoites. Also, their sera ini-
potential of DNA shuffling technology, we consider it hibited parasite invasion into HepG2 hepatoma cells
highly rewarding to now apply the same capsid evolu- in vitro, further validating the power of this mouse
tion and selection strategy to other cell types in the model to investigate human immune mechanisms of
liver as well. Ideally, this will eventually result in an antibody-based malaria vaccines. Thirdly, Deal et al.
AAV vector toolbox permitting to specifically target studied a vectored immunoprophylaxis (VIP) strategy
any given cell population, either in an intact liver or in by exploiting intramuscularly injected AAV8 vectors
complex cell culture systems. This will in turn allow to to express antibodies against P. falciparum CSP [118].
overexpress or inhibit any desired putative Plasmodium Remarkably, up to 70% of treated mice were pro-
host factor (provided its cDNA size is compatible with tected long-term from either intravenous or mosquito
AAV, see above) and, ultimately, to thus fully unravel bite challenge with sporozoites, in an antibody dose-
the intricate parasite–host interactions in all relevant dependent manner. This clearly shows the great poten-
liver cell types. Toward this aim, it should be useful to tial of AAV-based VIP approaches to confer immunity
further juxtapose retargeting strategies based on AAV to infection, albeit a number of parameters still need
capsid modification with synergistic other approaches, to be elucidated and optimized prior to clinical transla-
such as the use of cell type-specific promoters (Fig 3B tion in humans, such as the effective antibody levels
[iii]), or cellular detargeting through incorporation of and the best sporozoite and/or pre-erythrocytic antigen
selected miRNA-binding sites into the vector (Fig 3B targets.
[iv]). For instance, one candidate miRNA that could Lastly, our own two groups have previously har-
be exploited for this purpose is miR-122, as it is exclu- nessed AAV8 vectors to dysregulate miR-155 in
sively present in hepatocytes and can thus be har- mouse livers prior to vaccination with genetically
nessed to detarget AAV vectors from this cell type attenuated P. berghei parasites (GAP, [3,4]), following
[86,112,113]. our discovery that this immunoregulatory host
miRNA is highly elevated upon Plasmodium liver-
stage infection [50]. Our concomitant result that GAP
Experience in malaria context
infusion stimulates expression of TNFa (tumor necro-
In light of the numerous benefits and features of AAV sis factor alpha) and IFNc shed light on the underly-
vectors outlined above, it should be evident why they ing molecular mechanism, considering that these two
are increasingly used in malaria research. Already, one cytokines are known upstream regulators of miR-155
can distinguish at least four distinct applications in the expression. Most notable was, however, our finding
literature, and we can readily anticipate that many that AAV8 vector-mediated miR-155 overexpression
more will be tested soon. Firstly, several reports in hepatocytes and Kupffer cells boosts the protective
described the use of AAV1 vectors to express different capacity of the GAP vaccine. This exemplifies how
malarial antigens—MSP4 (merozoite surface protein 4, AAV vector technology can be used not only to bet-
P. falciparum) or MSP4/5 (P. yoelii)—in the muscle of ter dissect host–parasite interactions in the liver, but
mice. These antigens were either expressed alone [114], concurrently also to develop novel biomedically rele-
or, in efforts to augment the immune response, as vant tools and strategies.

FEBS Letters 590 (2016) 2027–2045 ª 2016 Federation of European Biochemical Societies 2039
Plasmodium meets AAV F. Hentzschel et al.

Concluding remarks Schaijk B, Teelen K, Arens T et al. (2009) Protection


against a malaria challenge by sporozoite inoculation.
Over the past decade, there has been tremendous and N Engl J Med 361, 468–477.
impressive progress in all research areas that were in 6 Roestenberg M, Teirlinck AC, McCall MB, Teelen
the center of this article, culminating in an improved K, Makamdop KN, Wiersma J, Arens T, Beckers P,
understanding of Plasmodium–host interactions, an van Gemert G, van de Vegte-Bolmer M et al. (2011)
arsenal of novel surrogate models of mammalian liv- Long-term protection against malaria after
ers, and an ever expanding repertoire of technologies experimental sporozoite inoculation: an open-label
for AAV vector engineering. In view of the rapid pace follow-up study. Lancet 377, 1770–1776.
at which these advances were made, we can readily 7 http://www.path.org/news/press-room/724/.
anticipate that the continued optimization and combi- 8 Agnandji ST, Lell B, Fernandes JF, Abossolo BP,
nation of these synergistic avenues holds great poten- Methogo BG, Kabwende AL, Adegnika AA,
tial to propel both basic and applied malaria research Mordmuller B, Issifou S, Kremsner PG et al. (2012) A
fast forward, and to hopefully result in the eventual phase 3 trial of RTS, S/AS01 malaria vaccine in
eradication of this devastating disease. African infants. N Engl J Med 367, 2284–2295.
9 Agnandji ST, Lell B, Soulanoudjingar SS, Fernandes
JF, Abossolo BP, Conzelmann C, Methogo BG,
Acknowledgements Doucka Y, Flamen A, Mordmuller B et al. (2011)
First results of phase 3 trial of RTS, S/AS01 malaria
All four authors appreciate the support of their
vaccine in African children.
research by the DFG (German Research Foundation)
N Engl J Med 365, 1863–1875.
through the Collaborative Research Center SFB1129.
10 RTS,S Clinical Trials Partnership (2015) Efficacy and
F.H., A.-K.H & D.G. are grateful for support from
safety of RTS,S/AS01 malaria vaccine with or without
the DFG Cluster of Excellence CellNetworks a booster dose in infants and children in Africa: final
(EXC81), and A.-K.M. is thankful for support from results of a phase 3, individually randomised,
the DFG SPP1580 program. Finally, we appreciate controlled trial. Lancet 386, 31–45.
critical reading of the manuscript by several members 11 Prudencio M, Rodriguez A and Mota MM (2006) The
of our two (A.-K.M. & D.G.) laboratories. The figures silent path to thousands of merozoites: the Plasmodium
in this article use clipart from Servier Medical Art. liver stage. Nat Rev Microbiol 4, 849–856.
12 Rodrigues T, Prudencio M, Moreira R, Mota MM
and Lopes F (2012) Targeting the liver stage of
Author contributions
malaria parasites: a yet unmet goal. J Med Chem 55,
A-KM and DG conceived this review. All four authors 995–1012.
jointly wrote and edited the manuscript. 13 Mota MM and Rodriguez A (2001) Migration through
host cells by apicomplexan parasites. Microbes Infect
3, 1123–1128.
References 14 Sturm A, Amino R, van de Sand C, Regen T, Retzlaff
1 WHO Malaria Report 2015 (http:// S, Rennenberg A, Kruger A, Pollok JM, Menard R
www.who.int/malaria/publications/world-malaria- and Heussler VT (2006) Manipulation of host
report-2015/report/en/). hepatocytes by the malaria parasite for delivery into
2 Nussenzweig RS, Vanderberg J, Most H and Orton C liver sinusoids. Science 313, 1287–1290.
(1967) Protective immunity produced by the injection 15 Amino R, Thiberge S, Martin B, Celli S, Shorte S,
of x-irradiated sporozoites of Plasmodium berghei. Frischknecht F and Menard R (2006) Quantitative
Nature 216, 160–162. imaging of Plasmodium transmission from mosquito to
3 Mueller AK, Camargo N, Kaiser K, Andorfer C, mammal. Nat Med 12, 220–224.
Frevert U, Matuschewski K and Kappe SH (2005) 16 Yamauchi LM, Coppi A, Snounou G and Sinnis P
Plasmodium liver stage developmental arrest by (2007) Plasmodium sporozoites trickle out of the
depletion of a protein at the parasite-host interface. injection site. Cell Microbiol 9, 1215–1222.
Proc Natl Acad Sci USA 102, 3022–3027. 17 Douglas RG, Amino R, Sinnis P and Frischknecht F
4 Mueller AK, Labaied M, Kappe SH and (2015) Active migration and passive transport of
Matuschewski K (2005) Genetically modified malaria parasites. Trends Parasitol 31, 357–362.
Plasmodium parasites as a protective experimental 18 Pradel G, Garapaty S and Frevert U (2002)
malaria vaccine. Nature 433, 164–167. Proteoglycans mediate malaria sporozoite targeting to
5 Roestenberg M, McCall M, Hopman J, Wiersma J, the liver. Mol Microbiol 45, 637–651.
Luty AJ, van Gemert GJ, van de Vegte-Bolmer M, van

2040 FEBS Letters 590 (2016) 2027–2045 ª 2016 Federation of European Biochemical Societies
F. Hentzschel et al. Plasmodium meets AAV

19 Pradel G, Garapaty S and Frevert U (2004) Kupffer 31 Rodrigues CD, Hannus M, Prudencio M, Martin C,
and stellate cell proteoglycans mediate malaria Goncalves LA, Portugal S, Epiphanio S, Akinc A,
sporozoite targeting to the liver. Comp Hepatol 3 Hadwiger P, Jahn-Hofmann K et al. (2008) Host
(Suppl. 1), S47. scavenger receptor SR-BI plays a dual role in the
20 Coppi A, Tewari R, Bishop JR, Bennett BL, establishment of malaria parasite liver infection. Cell
Lawrence R, Esko JD, Billker O and Sinnis P (2007) Host Microbe 4, 271–282.
Heparan sulfate proteoglycans provide a signal to 32 Foquet L, Hermsen CC, Verhoye L, van Gemert GJ,
Plasmodium sporozoites to stop migrating and Cortese R, Nicosia A, Sauerwein RW, Leroux-Roels G
productively invade host cells. Cell Host Microbe 2, and Meuleman P (2015) Anti-CD81 but not anti-SR-
316–327. BI blocks Plasmodium falciparum liver infection in a
21 Lingelbach K and Joiner KA (1998) The humanized mouse model. J Antimicrob Chemother 70,
parasitophorous vacuole membrane surrounding 1784–1787.
Plasmodium and Toxoplasma: an unusual 33 Yalaoui S, Huby T, Franetich JF, Gego A, Rametti A,
compartment in infected cells. J Cell Sci 111 (Pt 11), Moreau M, Collet X, Siau A, van Gemert GJ,
1467–1475. Sauerwein RW et al. (2008) Scavenger receptor BI
22 Zona L, Tawar RG, Zeisel MB, Xiao F, Schuster C, boosts hepatocyte permissiveness to Plasmodium
Lupberger J and Baumert TF (2014) CD81-receptor infection. Cell Host Microbe 4, 283–292.
associations–impact for hepatitis C virus entry and 34 Kapadia SB, Barth H, Baumert T, McKeating JA and
antiviral therapies. Viruses 6, 875–892. Chisari FV (2007) Initiation of hepatitis C virus
23 Zhang J, Randall G, Higginbottom A, Monk P, Rice infection is dependent on cholesterol and cooperativity
CM and McKeating JA (2004) CD81 is required for between CD81 and scavenger receptor B type I.
hepatitis C virus glycoprotein-mediated viral infection. J Virol 81, 374–383.
J Virol 78, 1448–1455. 35 Sulkowski MS, Kang M, Matining R, Wyles D,
24 Cormier EG, Tsamis F, Kajumo F, Durso RJ, Johnson VA, Morse GD, Amorosa V, Bhattacharya
Gardner JP and Dragic T (2004) CD81 is an entry D, Coughlin K, Wong-Staal F et al. (2014) Safety and
coreceptor for hepatitis C virus. Proc Natl Acad Sci antiviral activity of the HCV entry inhibitor ITX5061
USA 101, 7270–7274. in treatment-naive HCV-infected adults: a randomized,
25 Silvie O, Rubinstein E, Franetich JF, Prenant M, double-blind, phase 1b study. J Infect Dis 209,
Belnoue E, Renia L, Hannoun L, Eling W, Levy S, 658–667.
Boucheix C et al. (2003) Hepatocyte CD81 is required 36 Rowe IA, Tully DC, Armstrong MJ, Parker R, Guo
for Plasmodium falciparum and Plasmodium yoelii K, Barton D, Morse GD, Venuto CS, Ogilvie CB,
sporozoite infectivity. Nat Med 9, 93–96. Hedegaard DL et al. (2016) Effect of scavenger
26 Cha SJ, Park K, Srinivasan P, Schindler CW, van receptor class B type I antagonist ITX5061 in patients
Rooijen N, Stins M and Jacobs-Lorena M (2015) with hepatitis C virus infection undergoing liver
CD68 acts as a major gateway for malaria transplantation. Liver Transpl 22, 287–297.
sporozoite liver infection. J Exp Med 212, 1391– 37 Xiao F, Fofana I, Thumann C, Mailly L, Alles R,
1403. Robinet E, Meyer N, Schaeffer M, Habersetzer F,
27 Matuschewski K, Ross J, Brown SM, Kaiser K, Doffoel M et al. (2015) Synergy of entry inhibitors
Nussenzweig V and Kappe SH (2002) Infectivity- with direct-acting antivirals uncovers novel
associated changes in the transcriptional repertoire of combinations for prevention and treatment of hepatitis
the malaria parasite sporozoite stage. J Biol Chem 277, C. Gut 64, 483–494.
41948–41953. 38 Albuquerque SS, Carret C, Grosso AR, Tarun AS, Peng
28 Mikolajczak SA, Jacobs-Lorena V, MacKellar DC, X, Kappe SH, Prudencio M and Mota MM (2009) Host
Camargo N and Kappe SH (2007) L-FABP is a cell transcriptional profiling during malaria liver stage
critical host factor for successful malaria liver stage infection reveals a coordinated and sequential set of
development. Int J Parasitol 37, 483–489. biological events. BMC Genom 10, 270.
29 Prudencio M, Rodrigues CD, Hannus M, Martin C, 39 Leiriao P, Albuquerque SS, Corso S, van Gemert GJ,
Real E, Goncalves LA, Carret C, Dorkin R, Rohl I, Sauerwein RW, Rodriguez A, Giordano S and Mota
Jahn-Hoffmann K et al. (2008) Kinome-wide RNAi MM (2005) HGF/MET signalling protects
screen implicates at least 5 host hepatocyte kinases in Plasmodium-infected host cells from apoptosis. Cell
Plasmodium sporozoite infection. PLoS Pathog 4, Microbiol 7, 603–609.
e1000201. 40 Leiriao P, Mota MM and Rodriguez A (2005)
30 Hirai T and Chida K (2003) Protein kinase Czeta Apoptotic Plasmodium-infected hepatocytes provide
(PKCzeta): activation mechanisms and cellular antigens to liver dendritic cells. J Infect Dis 191,
functions. J Biochem 133, 1–7. 1576–1581.

FEBS Letters 590 (2016) 2027–2045 ª 2016 Federation of European Biochemical Societies 2041
Plasmodium meets AAV F. Hentzschel et al.

41 van de Sand C, Horstmann S, Schmidt A, Sturm A, infection is not conserved among Plasmodium species.
Bolte S, Kruger A, Lutgehetmann M, Pollok JM, Nat Med 17, 1180–1181; author reply 1181.
Libert C and Heussler VT (2005) The liver stage of 53 Carrolo M, Giordano S, Cabrita-Santos L, Corso S,
Plasmodium berghei inhibits host cell apoptosis. Mol Vigario AM, Silva S, Leiriao P, Carapau D, Armas-
Microbiol 58, 731–742. Portela R, Comoglio PM et al. (2003) Hepatocyte
42 Tarun AS, Peng X, Dumpit RF, Ogata Y, Silva- growth factor and its receptor are required for malaria
Rivera H, Camargo N, Daly TM, Bergman LW and infection. Nat Med 9, 1363–1369.
Kappe SH (2008) A combined transcriptome and 54 Godoy P, Hewitt NJ, Albrecht U, Andersen ME,
proteome survey of malaria parasite liver stages. Proc Ansari N, Bhattacharya S, Bode JG, Bolleyn J, Borner
Natl Acad Sci USA 105, 305–310. C, Bottger J et al. (2013) Recent advances in 2D and
43 Inacio P, Zuzart-Luis V, Ruivo MT, Falkard B, 3D in vitro systems using primary hepatocytes,
Nagaraj N, Rooijers K, Mann M, Mair G, Fidock DA alternative hepatocyte sources and non-parenchymal
and Mota MM (2015) Parasite-induced ER stress liver cells and their use in investigating mechanisms of
response in hepatocytes facilitates Plasmodium liver hepatotoxicity, cell signaling and ADME. Arch Toxicol
stage infection. EMBO Rep 16, 955–964. 87, 1315–1530.
44 Epiphanio S, Mikolajczak SA, Goncalves LA, 55 LeCluyse EL, Witek RP, Andersen ME and Powers
Pamplona A, Portugal S, Albuquerque S, Goldberg M, MJ (2012) Organotypic liver culture models: meeting
Rebelo S, Anderson DG, Akinc A et al. (2008) Heme current challenges in toxicity testing. Crit Rev Toxicol
oxygenase-1 is an anti-inflammatory host factor that 42, 501–548.
promotes murine plasmodium liver infection. Cell Host 56 Fitzgerald KA, Malhotra M, Curtin CM, O’Brien FJ
Microbe 3, and O’Driscoll CM (2015) Life in 3D is never flat: 3D
331–338. models to optimise drug delivery. J Control Release
45 Liehl P, Zuzarte-Luis V, Chan J, Zillinger T, Baptista 215, 39–54.
F, Carapau D, Konert M, Hanson KK, Carret C, 57 Ng S , Schwartz RE, March S, Galstian A, Gural N,
Lassnig C et al. (2014) Host-cell sensors for Shan J, Prabhu M, Mota MM and Bhatia SN (2015)
Plasmodium activate innate immunity against liver- Human iPSC-derived hepatocyte-like cells support
stage infection. Nat Med 20, 47–53. Plasmodium liver-stage infection in vitro. Stem Cell
46 Portugal S, Carret C, Recker M, Armitage AE, Reports 4, 348–359.
Goncalves LA, Epiphanio S, Sullivan D, Roy C, 58 Inoue H, Nagata N, Kurokawa H and Yamanaka S
Newbold CI, Drakesmith H et al. (2011) Host- (2014) iPS cells: a game changer for future medicine.
mediated regulation of superinfection in malaria. Nat EMBO J 33, 409–417.
Med 17, 732–737. 59 Gonzalez F, Boue S and Izpisua Belmonte JC (2011)
47 Kaushansky A, Ye AS, Austin LS, Mikolajczak SA, Methods for making induced pluripotent stem cells:
Vaughan AM, Camargo N, Metzger PG, Douglass reprogramming a la carte. Nat Rev Genet 12, 231–242.
AN, MacBeath G and Kappe SH (2013) Suppression 60 Schwartz RE, Fleming HE, Khetani SR and Bhatia
of host p53 is critical for Plasmodium liver-stage SN (2014) Pluripotent stem cell-derived hepatocyte-like
infection. Cell Rep 3, 630–637. cells. Biotechnol Adv 32, 504–513.
48 Teoh PJ and Chng WJ (2014) p53 abnormalities and 61 Asuri P, Bartel MA, Vazin T, Jang JH, Wong TB and
potential therapeutic targeting in multiple myeloma. Schaffer DV (2012) Directed evolution of adeno-
Biomed Res Int 2014, 717919. associated virus for enhanced gene delivery and gene
49 Tili E, Michaille JJ and Croce CM (2013) targeting in human pluripotent stem cells. Mol Ther
MicroRNAs play a central role in molecular 20, 329–338.
dysfunctions linking inflammation with cancer. 62 Egan ES, Jiang RH, Moechtar MA, Barteneva NS,
Immunol Rev 253, 167–184. Weekes MP, Nobre LV, Gygi SP, Paulo JA,
50 Hentzschel F, Hammerschmidt-Kamper C, Borner K, Frantzreb C, Tani Y et al. (2015) Malaria. A
Heiss K, Knapp B, Sattler JM, Kaderali L, Castoldi forward genetic screen identifies erythrocyte CD55 as
M, Bindman JG, Malato Y et al. (2014) AAV8- essential for Plasmodium falciparum invasion. Science
mediated in vivo overexpression of miR-155 348, 711–714.
enhances the protective capacity of genetically 63 Frevert U, Engelmann S, Zougbede S, Stange J, Ng B,
attenuated malarial parasites. Mol Ther 22, 2130– Matuschewski K, Liebes L and Yee H (2005)
2141. Intravital observation of Plasmodium berghei
51 Mashima R (2015) Physiological roles of miR-155. sporozoite infection of the liver. PLoS Biol 3, e192.
Immunology 145, 323–333. 64 Tavares J, Formaglio P, Thiberge S, Mordelet E, Van
52 Kaushansky A and Kappe SH (2011) The crucial role Rooijen N, Medvinsky A, Menard R and Amino R
of hepatocyte growth factor receptor during liver-stage (2013) Role of host cell traversal by the malaria

2042 FEBS Letters 590 (2016) 2027–2045 ª 2016 Federation of European Biochemical Societies
F. Hentzschel et al. Plasmodium meets AAV

sporozoite during liver infection. J Exp Med 210, 77 Berns KI, Byrne KI, Flotte TR, Gao G, Hauswirth
905–915. WW, Herzog RW, Muzyczka N, VandenDriessche T,
65 March S, Ramanan V, Trehan K, Ng S, Galstian A, Xiao X, Zolotukhin S et al. (2015) Adeno-associated
Gural N, Scull MA, Shlomai A, Mota MM, Fleming virus type 2 and hepatocellular carcinoma? Hum Gene
HE et al. (2015) Micropatterned coculture of primary Ther 26, 779–781.
human hepatocytes and supportive cells for the study 78 Grimm D (2002) Production methods for gene transfer
of hepatotropic pathogens. Nat Protoc 10, 2027–2053. vectors based on adeno-associated virus serotypes.
66 March S, Ng S, Velmurugan S, Galstian A, Shan J, Methods 28, 146–157.
Logan DJ, Carpenter AE, Thomas D, Sim BK, Mota 79 Asokan A, Schaffer DV and Samulski RJ (2012) The
MM et al. (2013) A microscale human liver platform AAV vector toolkit: poised at the clinical crossroads.
that supports the hepatic stages of Plasmodium Mol Ther 20, 699–708.
falciparum and vivax. Cell Host Microbe 14, 104–115. 80 Grimm D, Lee JS, Wang L, Desai T, Akache B,
67 Khetani SR and Bhatia SN (2008) Microscale culture Storm TA and Kay MA (2008) In vitro and in vivo
of human liver cells for drug development. Nat gene therapy vector evolution via multispecies
Biotechnol 26, 120–126. interbreeding and retargeting of adeno-associated
68 Schanz J, Pusch J, Hansmann J and Walles H (2010) viruses. J Virol 82, 5887–5911.
Vascularised human tissue models: a new approach for 81 Grimm D and Zolotukhin S (2015) E Pluribus Unum:
the refinement of biomedical research. J Biotechnol 50 years of research, millions of viruses, and one goal-
148, 56–63. tailored acceleration of AAV evolution. Mol Ther 23,
69 Jiang WC, Cheng YH, Yen MH, Chang Y, Yang VW 1819–1831.
and Lee OK (2014) Cryo-chemical decellularization of 82 Kotterman MA and Schaffer DV (2014) Engineering
the whole liver for mesenchymal stem cells-based adeno-associated viruses for clinical gene therapy. Nat
functional hepatic tissue engineering. Biomaterials 35, Rev Genet 15, 445–451.
3607–3617. 83 McCarty DM (2008) Self-complementary AAV
70 Wagner A, Rohrs V, Materne EM, Hiller T, vectors; advances and applications. Mol Ther 16,
Kedzierski R, Fechner H, Lauster R and Kurreck J 1648–1656.
(2015) Use of a three-dimensional humanized liver 84 Grimm D, Streetz K, Jopling CL, Storm TA, Pandey
model for the study of viral gene vectors. J Biotechnol K, Davis CR, Marion P, Salazar F and Kay MA
212, 134–143. (2006) Fatality in mice due to oversaturation of
71 Morosan S, Hez-Deroubaix S, Lunel F, Renia L, cellular microRNA/short hairpin RNA pathways.
Giannini C, Van Rooijen N, Battaglia S, Blanc C, Nature 441, 537–541.
Eling W, Sauerwein R et al. (2006) Liver-stage 85 Schmidt F and Grimm D (2015) CRISPR genome
development of Plasmodium falciparum, in a engineering and viral gene delivery: a case of mutual
humanized mouse model. J Infect Dis 193, 996–1004. attraction. Biotechnol J 10, 258–272.
72 Vaughan AM, Kappe SH, Ploss A and Mikolajczak 86 Senis E, Fatouros C, Grosse S, Wiedtke E, Niopek D,
SA (2012) Development of humanized mouse models Mueller AK, Borner K and Grimm D (2014)
to study human malaria parasite infection. Future CRISPR/Cas9-mediated genome engineering: an
Microbiol 7, 657–665. adeno-associated viral (AAV) vector toolbox.
73 Soulard V, Bosson-Vanga H, Lorthiois A, Roucher C, Biotechnol J 9, 1402–1412.
Franetich JF, Zanghi G, Bordessoulles M, Tefit M, 87 Wright AV, Nunez JK and Doudna JA (2016) Biology
Thellier M, Morosan S et al. (2015) Plasmodium and applications of CRISPR systems: harnessing
falciparum full life cycle and Plasmodium ovale liver nature’s toolbox for genome engineering. Cell 164,
stages in humanized mice. Nat Commun 6, 7690. 29–44.
74 Wijayalath W, Majji S, Villasante EF, Brumeanu TD, 88 Shalem O, Sanjana NE and Zhang F (2015) High-
Richie TL and Casares S (2014) Humanized HLA- throughput functional genomics using CRISPR-Cas9.
DR4.RagKO.IL2RgammacKO.NOD (DRAG) mice Nat Rev Genet 16, 299–311.
sustain the complex vertebrate life cycle of Plasmodium 89 Ran FA, Cong L, Yan WX, Scott DA, Gootenberg
falciparum malaria. Malar J 13, 386. JS, Kriz AJ, Zetsche B, Shalem O, Wu X, Makarova
75 Grimm D and Kay MA (2003) From virus evolution to KS et al. (2015) In vivo genome editing using
vector revolution: use of naturally occurring serotypes of Staphylococcus aureus Cas9. Nature 520, 186–191.
adeno-associated virus (AAV) as novel vectors for 90 Platt RJ, Chen S, Zhou Y, Yim MJ, Swiech L,
human gene therapy. Curr Gene Ther 3, 281–304. Kempton HR, Dahlman JE, Parnas O, Eisenhaure TM,
76 Buning H and Schmidt M (2015) Adeno-associated Jovanovic M et al. (2014) CRISPR-Cas9 knockin mice
vector toxicity-to be or not to be? Mol Ther 23, for genome editing and cancer modeling. Cell 159, 440–
1673–1675. 455.

FEBS Letters 590 (2016) 2027–2045 ª 2016 Federation of European Biochemical Societies 2043
Plasmodium meets AAV F. Hentzschel et al.

91 Swiech L, Heidenreich M, Banerjee A, Habib N, Li Y, enhancement via human Argonaute-2 overexpression


Trombetta J, Sur M and Zhang F (2015) In vivo from plasmids, viral vectors and cell lines. Nucleic
interrogation of gene function in the mammalian brain Acids Res 41, e199.
using CRISPR-Cas9. Nat Biotechnol 33, 102–106. 103 Mockenhaupt S, Grosse S, Rupp D, Bartenschlager
92 Gao G, Vandenberghe LH, Alvira MR, Lu Y, R and Grimm D (2015) Alleviation of off-target
Calcedo R, Zhou X and Wilson JM (2004) Clades of effects from vector-encoded shRNAs via codelivered
adeno-associated viruses are widely disseminated in RNA decoys. Proc Natl Acad Sci USA 112, E4007–
human tissues. J Virol 78, 6381–6388. E4016.
93 Nakai H, Fuess S, Storm TA, Muramatsu S, Nara Y 104 Yang Y, Wang L, Bell P, McMenamin D, He Z,
and Kay MA (2005) Unrestricted hepatocyte White J, Yu H, Xu C, Morizono H, Musunuru K
transduction with adeno-associated virus serotype 8 et al. (2016) A dual AAV system enables the Cas9-
vectors in mice. J Virol 79, 214–224. mediated correction of a metabolic liver disease in
94 Nathwani AC, Reiss UM, Tuddenham EG, Rosales C, newborn mice. Nat Biotechnol 34, 334–338.
Chowdary P, McIntosh J, Della Peruta M, Lheriteau 105 Shen S, Bryant KD, Sun J, Brown SM, Troupes
E, Patel N, Raj D et al. (2014) Long-term safety and A, Pulicherla N and Asokan A (2012) Glycan
efficacy of factor IX gene therapy in hemophilia B. N binding avidity determines the systemic fate of
Engl J Med 371, 1994–2004. adeno-associated virus type 9. J Virol 86, 10408–
95 Nathwani AC, Tuddenham EG, Rangarajan S, 10417.
Rosales C, McIntosh J, Linch DC, Chowdary P, 106 Lisowski L, Tay SS and Alexander IE (2015) Adeno-
Riddell A, Pie AJ, Harrington C et al. (2011) associated virus serotypes for gene therapeutics. Curr
Adenovirus-associated virus vector-mediated gene Opin Pharmacol 24, 59–67.
transfer in hemophilia B. N Engl J Med 365, 2357– 107 Junge N, Mingozzi F, Ott M and Baumann U (2015)
2365. Adeno-associated virus vector-based gene therapy for
96 Nault JC, Datta S, Imbeaud S, Franconi A, Mallet M, monogenetic metabolic diseases of the liver. J Pediatr
Couchy G, Letouze E, Pilati C, Verret B, Blanc JF Gastroenterol Nutr 60, 433–440.
et al. (2015) Recurrent AAV2-related insertional 108 Kienle E, Senis E, Borner K, Niopek D, Wiedtke E,
mutagenesis in human hepatocellular carcinomas. Nat Grosse S and Grimm D (2012) Engineering and
Genet 47, 1187–1193. evolution of synthetic adeno-associated virus (AAV)
97 Salmon F, Grosios K and Petry H (2014) Safety gene therapy vectors via DNA family shuffling. J Vis
profile of recombinant adeno-associated viral vectors: Exp . pii: 3819.
focus on alipogene tiparvovec (Glybera(R)). Expert 109 Lisowski L, Dane AP, Chu K, Zhang Y, Cunningham
Rev Clin Pharmacol 7, 53–65. SC, Wilson EM, Nygaard S, Grompe M, Alexander
98 Mueller S, Huard J, Waldow K, Huang X, IE and Kay MA (2014) Selection and evaluation of
D’Alessandro LA, Bohl S, Borner K, Grimm D, clinically relevant AAV variants in a xenograft liver
Klamt S, Klingmueller U et al. (2015) T160- model. Nature 506, 382–386.
phosphorylated CDK2 defines threshold for HGF 110 Cheng B, Ling C, Dai Y, Lu Y, Glushakova LG, Gee
dependent proliferation in primary hepatocytes. Mol SW, McGoogan KE, Aslanidi GV, Park M, Stacpoole
Syst Biol 11, 795. PW et al. (2012) Development of optimized AAV3
99 Giering JC, Grimm D, Storm TA and Kay MA (2008) serotype vectors: mechanism of high-efficiency
Expression of shRNA from a tissue-specific pol II transduction of human liver cancer cells. Gene Ther
promoter is an effective and safe RNAi therapeutic. 19, 375–384.
Mol Ther 16, 1630–1636. 111 Ling C Wang Y, Zhang Y, Ejjigani A, Yin Z, Lu Y,
100 Chen CC, Sun CP, Ma HI, Fang CC, Wu PY, Xiao X Wang L, Wang M, Li J, Hu Z et al. (2014) Selective
and Tao MH (2009) Comparative study of anti- in vivo targeting of human liver tumors by optimized
hepatitis B virus RNA interference by double-stranded AAV3 vectors in a murine xenograft model. Hum
adeno-associated virus serotypes 7, 8, and 9. Mol Ther Gene Ther 25, 1023–1034.
17, 352–359. 112 Xie J, Xie Q, Zhang H, Ameres SL, Hung JH, Su
101 Grimm D, Wang L, Lee JS, Schurmann N, Gu S, Q, He R, Mu X, Seher Ahmed S, Park S et al.
Borner K, Storm TA and Kay MA (2010) Argonaute (2011) MicroRNA-regulated, systemically delivered
proteins are key determinants of RNAi efficacy, rAAV9: a step closer to CNS-restricted transgene
toxicity, and persistence in the adult mouse liver. expression. Mol Ther 19, 526–535.
J Clin Invest 120, 3106–3119. 113 Geisler A, Jungmann A, Kurreck J, Poller W, Katus
102 Borner K, Niopek D, Cotugno G, Kaldenbach M, HA, Vetter R, Fechner H and Muller OJ (2011)
Pankert T, Willemsen J, Zhang X, Schurmann N, microRNA122-regulated transgene expression
Mockenhaupt S, Serva A et al. (2013) Robust RNAi increases specificity of cardiac gene transfer upon

2044 FEBS Letters 590 (2016) 2027–2045 ª 2016 Federation of European Biochemical Societies
F. Hentzschel et al. Plasmodium meets AAV

intravenous delivery of AAV9 vectors. Gene Ther 18, intramuscular delivery of adeno-associated viruses
199–209. encoding CTLA4-Ig-antigen fusion molecules. Gene
114 Logan GJ, Wang L, Zheng M, Cunningham SC, Ther 16, 200–210.
Coppel RL and Alexander IE (2007) AAV vectors 117 Huang J, Li X, Coelho-Dos-Reis JG, Zhang M,
encoding malarial antigens stimulate antigen-specific Mitchell R, Nogueira RT, Tsao T, Noe AR, Ayala R,
immunity but do not protect from parasite infection. Sahi V et al. (2015) Human immune system mice
Vaccine 25, 1014–1022. immunized with Plasmodium falciparum
115 Logan GJ, Wang L, Zheng M, Coppel RL and circumsporozoite protein induce protective human
Alexander IE (2010) Antigen fusion with C3d3 humoral immunity against malaria. J Immunol
augments or inhibits humoral immunity to AAV Methods 427, 42–50.
genetic vaccines in a transgene-dependent manner. 118 Deal C, Balazs AB, Espinosa DA, Zavala F,
Immunol Cell Biol 88, 228–232. Baltimore D and Ketner G (2014) Vectored antibody
116 Logan GJ, Wang L, Zheng M, Ginn SL, Coppel RL gene delivery protects against Plasmodium falciparum
and Alexander IE (2009) Antigen-specific humoral sporozoite challenge in mice. Proc Natl Acad Sci USA
tolerance or immune augmentation induced by 111, 12528–12532.

FEBS Letters 590 (2016) 2027–2045 ª 2016 Federation of European Biochemical Societies 2045

You might also like