ArticlePDF AvailableLiterature Review

Malaria infections: What and how can mice teach us?

Authors:
Research paper
Malaria infections: What and how can mice teach us
Vanessa Zuzarte-Luis
a
, Maria M. Mota
a,
, Ana M. Vigário
a,b,
⁎⁎
a
Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
b
Unidade de Ciências Médicas, Centro de Competência de Ciências da Vida, Universidade da Madeira, Funchal, Portugal
article info abstract
Article history:
Received 18 February 2014
Received in revised form 24 April 2014
Accepted 1 May 2014
Available online 14 May 2014
Malaria imposes a horrific public health burden hundreds of millions of infections and
millions of deaths on large parts of the world. While this unacceptable health burden and its
economic and social impact have made it a focal point of the international development
agenda, it became consensual that malaria control or elimination will be difficult to attain prior
to gain a better understanding of the complex interactions occurring between its main players:
Plasmodium, the causative agent of disease, and its hosts. Practical and ethical limitations exist
regarding the ability to carry out research with human subjects or with human samples. In this
review, we highlight how rodent models of infection have contributed significantly during the
past decades to a better understanding of the basic biology of the parasite, host response and
pathogenesis.
© 2014 Elsevier B.V. All rights reserved.
Keywords:
Plasmodium
Malaria
Host
Rodent models
Pathology
Infection
1. Introduction
Malaria still imposes a significant health and economic
burden in large parts of the world, particularly in sub-Saharan
Africa and Southeast Asia, whereat least 200 million infections
and over 600000 deaths are registered annually (WHO, 2013).
The disease is caused by protozoan parasites of the genus
Plasmodium, which are transmitted by female Anopheline
mosquitoes. During a blood meal the infected female mosqui-
toes deposit Plasmodium sporozoites in the mammalian skin.
Within minutes to few hours after inoculation (Sinnis and
Zavala, 2012) these highly motile forms enter the circulatory
system and reach the liver where they infect hepatocytes
establishing the so-called pre-erythrocytic phase of malaria
infection. This phase of infection is completely asymptomatic
and lasts in humans 517 days (the length varies according to
Plasmodium species (Coatney et al., 1971)). Each sporozoite
that infects the liver replicates into thousands of new parasites
in a process called schizogony. Once parasite replication and
cellularization are completed, the newly formed parasites,
called merozoites, are released into the bloodstream and infect
erythrocytes, initiating the erythrocytic stage of malaria
infection (Prudencio et al., 2006). The cycles of parasite
multiplication inside erythrocytes are shorter (24, 48 or 72 h,
depending on parasite species), and causative of the classic
symptoms of the disease (Coatney et al., 1971). When left
untreated, the disease can eventually progress to severe
syndromes and cause death (Haldar et al., 2007).
Human malaria can be caused by five Plasmodium species:
Plasmodium falciparum,Plasmodium vivax,Plasmodium ovale,
Plasmodium malariae and Plasmodium knowlesi.Ofthese,P.
falciparum and P. vivax are the focus of intense research and
targeting strategies due to the high mortality and/or morbidity
they cause. P. falciparum is the most virulent species and is
responsible for the vast majority of deaths in sub-Saharan Africa,
primarily of young children and pregnant women.
P. vivax malaria is the most widespread and was
previously considered a benign disease but is emerging as a
potentially lethal condition outside of Africa (Baird, 2013),
as current control measures are successfully reducing P.
Journal of Immunological Methods 410 (2014) 113122
Corresponding author.
⁎⁎ Correspondence to: A.M. Vigário, Instituto de Medicina Molecular, Faculdade
de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal.
E-mail addresses: mmota@fm.ul.pt (M.M. Mota), avigario@fm.ul.pt
(A.M. Vigário).
http://dx.doi.org/10.1016/j.jim.2014.05.001
0022-1759/© 2014 Elsevier B.V. All rights reserved.
Contents lists available at ScienceDirect
Journal of Immunological Methods
journal homepage: www.elsevier.com/locate/jim
falciparum transmission (Cotter et al., 2013). Additionally, P.
vivax is capable of forming cryptic forms called hyponozoites
during the pre-erythrocytic stage that cause relapses months
and even years after blood stage parasite clearance, contrib-
uting to the complexity of understanding and treating P.
vivax malaria (Shanks and White, 2013; Kondrashin et al.,
2014).
Despite major advances in the development and implemen-
tation of novel intervention strategies, the scientific community
is still limited by substantial gaps in understanding the biology of
Plasmodium and its complex interaction with the human host
(The malERA Consultative Group on Basic Science and Enabling
Technologies, 2011). Further studies addressing fundamental
hostparasite interactions, as well as patho-physiological fea-
tures of infection are necessary, but such studies are difficult to
perform in humans.
2. The importance of addressing malaria
infection experimentally
The study of human malaria involves a myriad of methods
such as epidemiological analysis, population genetics, clinical
studies of patients, both in field research studies and in hospital
settings, as well as analyses of post-mortem biopsies. However,
limitations exist regarding t he ability to carry out research with
human subjects or with human samples. For example, access to
post-mortem tissues is hindered due to religious and cultural
objection to autopsy and the lack of proper control subjects for
most studies such as samples from infected patients that do
not develop the pathology or die are some of the reasons that
make research difficult. Moreover, the data obtained from
post-mortem studies only represents the end stage of a long
process, and the analysis of the sequence of events leading to
pathology through monitoring the internal organs and envi-
ronment is very limited; e.g. the study of the liver during the
first phase of infection or the brain during cerebral malaria.
Despite their limitation and controversy on replicability
of human disease, mouse models of malaria infection have
been used for decades and have contributed significantly to
a better understanding of the basic biology of the parasite,
host response and pathogenesis. Several rodent-infectious
Plasmodium parasites are available, Plasmodium berghei,
Plasmodium yoelii,Plasmodium chabaudi and Plasmodium
vinckei, each including several strains, which lead to
distinct courses and outcomes of infection, depending on
the host-mouse strain combination (see Box 1), raising
questions about which, if any, of the mouse models can be
extrapolated to understand human disease or diseases.
Concerns exist about the translational utility of animal
models in pathogenesis, immunity, vaccine development
and drug discovery due to the heterogeneity observed with
different parasite and mouse combinations (White et al.,
2010; Craig et al., 2012). However, one can argue that the
range of disease manifestations in the different mouse
models should be considered as a reflection of the diversity
of the human disease rather than a limitation (Langhorne
et al., 2011). Undoubtedly, the availability of inbred/
congenic/transgenic animals and the ability to manipulate
and control different aspects of the host, including the
immune system, make the mouse model a precious tool.
Still, mice are not humans and the Plasmodium spp. that
infect rodents are distinct from the ones that infect
humans. As such, results arising from studies using rodent
models should be interpreted with caution (White et al.,
2010; Craig et al., 2012; Langhorne et al., 2011). Aware of
the importance and simultaneously the limitation of the
mouse models, there has been a constant search for mouse
models that better reflect the different field situations (see
Boxes 1 and 2).
2.1. How to address malaria infection experimentally
In addition to studying disease mechanisms, an advantage
of using rodent models is the ease of maintaining the entire
life cycle of the parasite in controlled and optimized
laboratory conditions. The establishment and maintenance
of laboratory Anopheles stephensi (as well as Anopheles
gambiae) vector colonies and the development of transgenic
parasite lines have allowed the dissection of processes
occurring during transmission from the mosquito vector to
the mammalian host, as well as studies of transmission from
the mammalian host to the mosquito vector. Controlled
infections can be initiated directly by mosquito bite or,
alternatively, by intra-dermal or intra-venous injection of
sporozoites. The infection can then be analyzed in the liver or
be allowed to progress into the blood, and disease outcome
can be monitored. It is also possible to bypass the skin and
liver stages of infection by directly injecting parasitized red
blood cells (pRBCs) intra-peritoneally or intravenously. The
careful choice of transgenic parasite line determines the
possibilities of analysis; e.g. the use of fluorescent parasites
allows monitoring the infection and the parasite's interaction
with host cells in vivo and in real-time (Gomes-Santos et al.,
2012). When using chemiluminescent parasites, infection
can be analyzed longitudinally over time (within the same
infected animal) in a non-invasive (or minimally invasive)
manner throughout liver stage into blood stage infection,
where bioluminescence is correlated with the level of liver
infection and with blood parasitemia (Ploemen et al., 2009;
Zuzarte-Luis et al., 2014). Additionally, chemiluminescent
parasites have also facilitated the study of infected erythro-
cyte sequestration (Franke-Fayard et al., 2006). Moreover,
the combination of transgenic parasites (lacking or overex-
pressing parasite or exogenous molecules) with genetically
engineered mice lacking key molecules (e.g. immune medi-
ators or their receptors) has proved useful in deciphering the
host response to parasite infection throughout the latter's life
cycle.
Further technological progress such as the development of
high-throughput Omics technologies employed to study Plasmo-
dium infection at the DNA, RNA, protein, and metabolite levels
has prompted further advances in our understanding of both
host as well as parasite biology (Tarun et al., 2008; Albuquerque
et al., 2009; Olszewski et al., 2009). Equally important was the
recent development of clinical diagnostic techniques for small
animals, including non-invasive imaging techniques such as
computer tomography (CT) and magnetic resonance imaging
(MRI), as well as monitoring systems for cardio-vascular and
respiratory function (Penet et al., 2005; Martins et al., 2013).
Such advances are crucial for the detailed characterization of
malaria pathology in animal models and help determine the
degree of similitude with the human pathology.
114 V. Zuzarte-Luis et al. / Journal of Immunological Methods 410 (2014) 113122
3. Plasmodium's journey inside the host
3.1. In the skin on the way to the liver
Until recently, details regarding the initiation of malaria
infection when an infected female mosquito injects sporozoite-
containing saliva as it probes the dermis for blood, remained
inscrutable. Questions such as where are sporozoites deposit-
ed?or how long do sporozoites stay at the injection site?
remained unsolved until recently. The small number of sporo-
zoites injected and their highly motile behavior hindered the
study of the dermal phase of the infection. The notion that
sporozoites are deposited in the skin, rather than directly into
the blood circulation, was first suggested in the 1930s upon
histological analysis of skin tissue of volunteers exposed to P.
vivax-infected mosquitoes (Boyd and S.F., 1939). Nevertheless, it
was the use of rodent models that allowed the experimental
confirmation of such results. Experiments of excision of the
bitten tissue (Sidjanski and Vanderberg, 1997)andinterruption
of mosquito feeding (Matsuoka et al., 2002; Ponnudurai et al.,
1991) resulted in a delayed onset of blood parasitemia, indirectly
suggesting that sporozoites are indeed deposited in the skin. The
direct observation of this phenomenon was possible due to
advances in molecular biology and imaging technologies, once
again using rodent models of infection. Using intravital fluores-
cence microscopy and genetically engineered parasites express-
ing fluorescent tags, researchers directly observed sporozoites
being injected in the host skin when the mosquito ejects saliva
(Vanderberg and Frevert, 2004). Similarly, the study of the
dynamics of sporozoites in the skin has significantly evolved
with the advances in technologies that enabled the direct
observation, in real-time, of the interactions between parasites
and their hosts. Upon injection, and contrary to the initial
assumption, sporozoites may spend several hours at the
inoculation site. The first experimental evidence of this fact
came from skin transplantation experiments in monkeys (Lloyd
and Sommerville, 1949). Later use of rodent models, in which
sporozoite load determination in excised tissue was performed
through histological analysis or quantitative molecular methods
(PCR), further supported the concept that the transit to the
bloodstream can take several hours (reviewed in Sinnis and
Zavala, 2012).
Nevertheless, insights on sporozoite numbers, character-
istics of movement and positional information could only be
gathered using high-resolution and high-speed microscopy.
The combination of in vivo imaging of rodent models with
genetically engineered P. berghei parasites enabled the determi-
nation of sporozoite numbers and parasite release rate through
the mosquito proboscis (Amino et al., 2006) and evidenced
the randomness of movement of sporozoites in the dermis
until contacting dermal blood vessels (Frischknecht, 2007).
Finally, this technology allowed the quantification of the
proportion of blood vessels (Amino et al., 2006,reviewed
in Sinnis and Zavala, 2012; Menard et al., 2013). Sporozo-
ites that remain at the inoculum site are likely destroyed by
the innate immune cells contributing to the initiation of the
immune response. Surgical excision of local lymph nodes or
pharmacological inhibition of T-cell egress from these organs
demonstrated, in a vaccination rodent model, that sporozoites
that reach lymphatic circulation and the draining lymph nodes
(1520%) are critical for priming CD8 + T cell response against
liver stage probably by being captured and their antigens
presented by dendritic cells (reviewed in Sinnis and Zavala,
2012).
3.2. In the liver replicating at full speed
Sporozoites that successfully invade blood vessels rapidly
home to the liver where they arrest in the liver sinusoids
through specific interactions between parasite surface pro-
teins and host molecules. These molecular interactions have
been extensively characterized in the 1990s using biochem-
ical and molecular approaches (Ejigiri and Sinnis, 2009), but
the details of liver invasion were still unclear. Early
observations of sporozoites going in and out of cells
(Vanderberg et al., 1990) inspired the discovery that
sporozoites have the ability to traverse cells prior to
establishing infection in the liver (Mota et al., 2001), which
in turn strongly impacted the view on how parasites reach
the liver from the mosquito bite site (Ishino et al., 2004;
Amino et al., 2008; Tavares et al., 2013). Imaging techniques,
both in vitro (using cell lines) and in vivo (using rodent
models), were pivotal to these discoveries (Mota et al., 2001;
Frevert et al., 2005).
Once inside hepatocytes, sporozoites undergo a remarkable
process of transformation and intense replication that lasts 517
days in humans and ~2 days in rodents. The complex interac-
tions that occur between the host cell and Plasmodium parasites
during this phase of infection only recently have begun to be
elucidated. The application of high-throughput technologies,
namely genomics, proteomics or lipidomics to the study of
isolated infected hepatocytes was and still is, fundamental to the
understanding of how the host cell responds to the presence of a
developing and highly replicative parasite and which host and
parasite pathways are engaged during the successful establish-
ment of infection (Tarun et al., 2008; Albuquerque et al., 2009).
Several relevant and novel questions regarding host-Plasmodium
interactions in the liver have emerged from the analysis of such
complex data sets (Epiphanio et al., 2008; Vaughan et al., 2009).
The similarly unbiased analysis of whole-liver samples has
further contributed to the understanding of the responses at
the organ/organism level (Portugal et al., 2011; Liehl et al., 2014).
In fact, such a recent analysis using rodent models of infection
enabled us to demonstrate the engagement of a type I interferon
(IFN) response during Plasmodium replication in the liver (Liehl
et al., 2014), a stage that until now was thought to be undetected
by the host (Liehl and Mota, 2012). The use of genetic mouse
models in combination with classical immunology techniques,
such as flow cytometry for the ex vivo analysis of liver immune
cell populations, immunohistochemistry analysis for positional
information, and molecular techniques (such as PCR or Western
blot), has been critical for the characterization of the immune
populations and molecular pathways involved in the immune
response elicited by the immunization with radiation-attenuated
Plasmodium spp. sporozoites or other protocols known to induce
sterile protective immunity against parasite challenge (reviewed
in Doolan and Martinez-Alier, 2006). Complete sterile protection
against liver stage, first demonstrated following immunization
with radiation-attenuated sporozoites in mice (Nussenzweig et
al., 1967), and latter in non-human primates (Gwadz et al., 1979)
and humans (Clyde, 1975), can also be achieved in mice with
genetically-attenuated sporozoites (Mueller et al., 2005a, 2005b;
115V. Zuzarte-Luis et al. / Journal of Immunological Methods 410 (2014) 113122
van Dijk et al., 2005) or with sporozoites in combination with
chemoprophylaxis in mice and humans (Belnoue et al., 2004;
Roestenberg et al., 2009).
After an intense period of liver schizogny, the newly formed
merozoites are released into the blood stream, where they
initiate the cycles of erythrocyte infection. The use of time-lapse
Box 1. Animal models to study severe malaria
Blood stage malaria infection has variable clinical outcomes, ranging from mild or uncomplicated malaria, usually non-lethal,
to severe or complicated malaria, with a mortality rate of 2030%. Severe and complicated malaria includes different clinical
features with different organs being affected (reviewed in Haldar et al., 2007 and see references in Fig. 1 for individual
syndromes Trang et al., 1992; Joshi et al., 1986; Rogerson et al., 2007; Douglas et al., 2012; Perkins et al., 2011; Taylor et
al., 2012), which have been modeled in rodents using different parasite/mouse strain combinations.
Cerebral malaria is responsible for most of deaths occurring due to malaria infection especially in children in endemic areas
(Haldar et al., 2007). CM can also develop in non-immune adults but with some symptomatological differences (Mishra and
Wiese, 2009). Even in children it is clear that CM is not a homogenous syndrome and 3 patterns of histopathological
changes (based on presence or absence of brain pRBC sequestration and/or microvascular pathology) have been described
in children dying with clinically defined CM (Dorovini-Zis et al., 2011). The reasons for this heterogeneity are unclear but are
probably due to host or parasite genetic variations as well as environmental factors. Notably, different parasite/mouse strain
combinations also show some heterogeneity on histopathological patterns (reviewed in Brian de Souza et al. 2009).
Nevertheless, while different combinations of parasite/mouse strains have been used in the past, most recent studies on
experimental cerebral malaria (ECM) have been conducted using C57BL/6 mice following intraperitoneal injection of P.
berghei ANKA-parasitized red blood cells (pRBC). C57BL/6 mice infected with P. berghei ANKA-related parasite isolates,
such as P. berghei NK65 or P. berghei K173, do not develop neurological symptoms and, as such, have been used as controls
on ECM studies. Similarly, mouse strains shown to be resistant to ECM when injected with P. berghei ANKA, like BALB/c
mice, have been used as controls.
Placental malaria (PM), a major malaria complication occurring during pregnancy, is estimated to be the cause of up to 200
000 infant deaths per year (WHO, 2013). It is usually associated with fetal growth restriction and/or preterm delivery,
stillbirths and maternal anemia. Different parasite/mouse strain combinations have been used to study PM. Early P. berghei
ANKA infection of pregnant BALB/c mice generally causes abortion, while infection at around gestational day 13 induces a
syndrome that resembles severe PM in women, with clear signs of pathology in the mother as well as the offspring (Neres et
al., 2008; Oduola et al., 1982; Hioki et al., 1990). To take advantage of the available C57BL/6 mutant strains, experimental
models of C57BL/6 mice infected, either early in gestation with P. chabaudi (Poovassery and Moore, 2006), or at gestational
day 13 with different lines of P. berghei (such as PbNK65 or PbK173) (Rodrigues-Duarte et al., 2012) were also developed.
Given that in areas where malaria is endemic, women generally develop considerable clinical immunity to malaria before
reproductive age, PM has also been studied in mice that had been immunized prior to mating (Megnekou et al., 2009;
Marinho et al., 2009; van Zon and Eling, 1980).
Malaria-associated acute lung injury (ALI) and its more severe form, malaria-associated acute respiratory distress
syndrome (ARDS) often occur in association with other severe forms of malaria infection (reviewed in Taylor et al., 2012).
Although, pulmonary pathology has been described in early studies of P. berghei experimental severe/cerebral malaria prior to
the development of ECM, only recently was it used as a model with the specific objective of studying ALI/ARDS (Lovegrove
et al., 2008). P. berghei ANKA-infected DBA/2 mice (Epiphanio et al., 2010) and either P. berghei K173- (Hee et al., 2011)or
P. berghei NK65-infected C57BL/6 mice (Van den Steen et al., 2010) were also described as models to investigate the
mechanisms leading to lung disease but without any associated cerebral complication.
Acute kidney injury (AKI) is a malaria-associated complication observed especially in adults. Although no specific mice
model exists, acute kidney injury has been studied in P. berghei infected BALB/c (Elias et al., 2012) or C57BL/6 (Sinniah et
al., 1999) mice.
Severe malarial anemia (SMA) is a life-threatening complication of malaria, highly prevalent in regions with high malaria
transmission. Several mouse/parasite combinations have been used to study this severe complication. However, in the
majority of these models severe anemia is associated with hyperparasitemia, which is not a characteristic of human SMA.
As such, other models like P. berghei ANKA infection in semi-immune BALB/c mice (Evans et al., 2006) or sequential
infection with two different Plasmodium species (Harris et al., 2012) have been described.
Liver injury (LI) can happen as consequence of malaria infection. Immune-mediated liver damage was first described on P.
berghei NK65 infection of C57BL/6 and BALB/c mice (Yoshimoto et al., 1998; Adachi et al., 2001), but other models were
also described such as P. chabaudi infection in DBA/2 mice (Seixas et al., 2001) and P. berghei ANKA in C57BL/6 mice
(Haque et al., 2011b).
Often, different disease mechanisms have been described for the same malaria complication based on different animal/
parasite combination models. In spite of being used as an argument against animal models this can also be seen as an
advantage. In fact, differences in the mechanism or outcome of the human disease exist, either related to host differences
(age, genetic variability, previous exposure and environment) or parasite variations. Human severe malaria is complex and
very likely implicates several of these mechanisms. Undoubtedly the use of models has significantly contributed to dissect
the mechanisms of severe pathology. However, their input for therapeutic interventions has been limited possibly because
most studies test the intervention strategies during a controlled infection and prior or at early stage of symptoms onset,
disregarding the fact that patients only present to the hospital in an advanced stage of disease.
116 V. Zuzarte-Luis et al. / Journal of Immunological Methods 410 (2014) 113122
intravital, confocal and electron microscopy allowed the visual-
ization of the mechanism by which hepatic merozoites reach
blood circulation, escaping the immune system (Sturm et al.,
2006; Baer et al., 2007). At the end of the liver stage, merozoites
are packed into merosomes, covered by host cell membrane, and
released into the bloodstream (Sturm et al., 2006). Once in
circulation, merosomes can reach the lung capillaries where they
rupture releasing the erythrocyte-infectious merozoites (Baer et
al., 2007). The timing of appearance/detection of infected
erythrocytes in circulation can be used as an indirect measure
Fig. 1. Table summarizing the possible severe malaria syndromes developed depending on the rodent host-parasite
combinations (top). Pictures representing three different outcomes of P. berghei ANKA infection in different host-mouse
strains (bottom). Left: Brains illustrating blood-brain barrier disruption (Evans blue) and parasite sequestration
(luminescence) in infected C57BL/6 mice that developed ECM. Middle: Impaired fetal development in infected pregnant
Balb/c females with PM (image from Neres et al., 2008). Right: Histological analysis of lung tissue evidencing hemorrhage
and edema in DBA/2 mice that developed ALI / ARDS.
117V. Zuzarte-Luis et al. / Journal of Immunological Methods 410 (2014) 113122
of viable parasite load in the previous stage (the liver). The
accurate and fast detection of the first generation of infected
erythrocytes is therefore important to evaluate interventions
targeting the liver stage. This can be done by analysis of blood
smears, by qRT-PCR or by a luciferase assay using transgenic
chemiluminescent parasite lines (Zuzarte-Luis et al., 2014).
Altogether, the use of rodent models has been absolutely
critical to the current understanding of Plasmodium liver
stage, recently recognized as the ideal target for the
development of novel anti-malarial strategies, such as drug
development or vaccines (Derbyshire et al., 2011; Duffy et al.,
2012; Rodrigues et al., 2012). Still, not all aspects of human
Plasmodium spp. biology can be modeled using rodent
malaria. The recent engineering of chimeric humanized
mouse models to study P. falciparum and P. vivax infections
constitutes a great evolution in the study of the human
parasites, enabling the study of parasite biology in vivo, but
also the evaluation of specific anti-malarial interventions in a
more physiological setting (Vaughan et al., 2012). The
technology is still evolving and while it is already possible
to investigate separately P. falciparum liver and blood-stage
development in vivo, the critical step of merozoite release
Box 2. From conventional rodent models of infection to humanized mice
The liver forms of Plasmodium parasites were first identified in 1948 (Cox, 2010). Still, our understanding of the biology of
Plasmodium hepatic stages remains limited. Despite being asymptomatic, the hepatic stage is an obligatory phase of
Plasmodium development. During this phase parasite numbers increase by four orders of magnitude (Prudencio et al., 2006)
but the load of parasites in the liver is significantly lower than in blood stage. This feature makes the liver stage an attractive
target for the development of malaria prophylaxis strategies.
Humans ultimately constitute the ideal system to study infection by Plasmodium parasites. However, given the difficulties in
obtaining human liver samples, animal models remain the closest surrogates available to researchers. In fact, the vast
majority of our knowledge has emerged by using P. berghei- and P. yoelii-infected mice. However, not all aspects of P.
falciparum biology can be modeled using rodent malaria, for example, the vaccine candidate LSA-1 has no ortholog in rodent
malaria species (Frech and Chen, 2011). Moreover, the human parasites P. ovale and P. vivax, are able to generate cryptic
forms called hypnozoites, never identified in rodent parasites. These dormant forms may remain in the liver for long periods
and cause disease relapses when re-activated. Therefore, intervention during this stage of infection is essential to achieve
complete parasite elimination. As such, an understanding the biology of this enigmatic form of the parasite and the
development of effective hypnozoiticides are crucial for the goal of malaria eradication.
The development of a small-animal model capable of efficiently supporting human liver-stage development in vivo became
an important challenge. The proof-of-concept that P. falciparum infected hepatocytes could develop in a small animal model
was demonstrated with the transplantation of human hepatocytes into immunosuppressed severe combined immunode-
ficiency (SCID) mice (Sacci et al., 1992). To promote human hepatocyte expansion by giving them a competitive growth
advantage over the endogenous murine hepatocytes, genetic approaches were employed. SCID mice homozygous for the
urokinase type plasminogen activator transgene (SCID/Alb-uPA), known to cause murine liver injury, were engrafted with
human hepatocytes (Mercer et al., 2001; Meuleman et al., 2005). These humanized liver-chimeric mice are able to support
complete development of P. falciparum liver stage (Morosan et al., 2006; Sacci et al., 2006), culminating with the release of
merozoites capable of invading hRBCs ex-vivo (Sacci et al., 2006), and were shown to be useful to study the phenotype of P.
falciparum gene knockouts (VanBuskirk et al., 2009; Mikolajczak et al., 2011). Nevertheless, this model cannot be used to
study the parasite transition from the liver stage to the blood within the mouse. When SCID/Alb-uPA mice are depleted in NK
cells and macrophages, the level of human hepatocytes chimerism and consequently of P. falciparum liver-stage
development improves (Morosan et al., 2006).
Nevertheless, the human liver chimeric SCID/Alb-uPA mouse model presents some drawbacks, mainly due to the severe
liver injury induced by the uPA transgene expression and to mice hypofertility (Vaughan et al., 2012). These weaknesses,
and the need for high-quality adult human hepatocytes for transplantation, make this model extremely costly and time-
consuming, which drastically limits the number of experimental mice and consequently the strength of the conclusions.
To overcome the above-mentioned weakness of the SCID/Alb-uPA liver injury model, an alternative mouse model of human
hepatocyte engraftment was developed (Azuma et al., 2007; Bissig et al., 2007). In this new model, the liver injury is
caused by the ablations of fumarylacetoacetate hydrolase (FAH) gene and mice are rescued from death by providing them 2-
(2-nitro-4-fluoromethylbenzoyl)-1,3-cyclohexanedione (NTBC). FAH
/
mice can be crossed with extremely immunocom-
promised mice, without B, T and NK cells (due to disruption of the Rag2 and the IL2Rγgenes). These FAH
/
Rag2
/
IL2Rγ
null
(FRG) mice can then be efficiently engrafted with human adult hepatocytes (Bissig et al., 2010). These mice breed efficiently, do
not show liver injury, and have a long lifespan. Moreover, human hepatocytes from a donor FRG mouse can be transplanted into
recipient mice, reducing the need of adult human hepatocytes (Azuma et al., 2007). There is no doubt that liver-humanized mice
are an important tool to understand particular aspects of the human hepatic infection such the biology of the human parasite, its
sensitivity to new drugs, and may be critical for future studies of Plasmodium hypnozoite forms. Importantly, not all aspects of
liver infectioncan be addressedwith these models, in particular the immune responseto the parasite,either in primaryinfection or
in vaccine studies. Moreover, the high cost of these models also hampers their regular use, thus rodent models of infection
together with novel models of in vitro infection developed recently for P. falciparum and P. vivax infection (March et al., 2013; Ng
et al., 2014) will always be critical to improve our still scarce knowledge of the liver stage of infection.
118 V. Zuzarte-Luis et al. / Journal of Immunological Methods 410 (2014) 113122
from hepatocytes and transition to RBC invasion still awaits
further elucidation. Please refer to Box 2 for further details on
the current models to study liver stage infection and for a
summary of humanized models to study pre-erythrocytic
stage malaria infection.
3.3. Reaching the blood to cause malaria
The possibility of maintaining P. falciparum blood stage cycles
in vitro using human RBCs has contributed to the fact that the
majority of studies attempting to establish the parasite and RBC
determinants of infection have been performed in vitro without
the need of animal models (Bei and Duraisingh, 2012;
Cowman et al., 2012). Similarly, while the recent develop-
ment of humanized mouse models for blood stage of P.
falciparum has allowed to answer several questions related
with in vivo multiplication of the human parasite P.
falciparum and contributed to drug development (discussed
elsewhere in this issue), the majority of these studies have
been performed using P. falciparum in vitro cultures.
Nevertheless, animal models have been critical in dissecting
key processes and players of the immune response mounted
during the blood stage of infection. In particular, the use of
genetically engineered mice lacking different molecules of
the immune system, in combination with depleting antibodies,
unveiled some of the complex immune mechanisms and
immuno-pathogenesis of Plasmodium infection (Schofield,
2007). These studies contributed to establish potential bio-
markers to predict risks of malaria related mortality (such as
IP-10 or CXCL10). The influence of genetic polymorphisms for
several of these molecules has also been studied on susceptibil-
ity/resistance to P. falciparum malaria (reviewed in Driss et al.,
2011).
If left untreated the infection may eventually progress to
severe disease. In the case of some Plasmodium strains, pRBCs
have the ability to cytoadhere to the endothelium of different
organs, which has been associated with the development of
severe pathology in human malaria. Hence, a detailed analysis of
the organs during disease progression is of major importance.
Similarly to the studies of liver stage, approaches from classical
histopathology, high-throughput Omics techniques and in vivo
imaging (for whole-organ or specific cell population analysis),
provided additional contributions to understanding pathogene-
sis at the tissue level. Histological and ultra-structural analysis of
brains from animals developing experimental cerebral malaria
(ECM) evidenced the sequestration of leukocytes (Polder et al.,
1992; Hearn et al., 2000; Vigario et al., 2007) but also platelets
(Combes et al., 2004) and some infected erythrocytes (Hearn et
al., 2000). It was later demonstrated, using flow cytometry
analysis of isolated brain leucocytes, that although very low in
number, CD8 T cells are the key leukocytes and the terminal
effector cells associated with ECM (Belnoue et al., 2002; Nitcheu
Fig. 2. Table summarizing the possibility of studies when using wild-type mice models infected with rodent Plasmodium
parasites (left) versus liver humanized mice models infected with human Plasmodium parasites (right).
119V. Zuzarte-Luis et al. / Journal of Immunological Methods 410 (2014) 113122
et al., 2003; Haque et al., 2011a). This important technical
advance contributed to the recognition of the importance of
these cells in the development of the pathology, an aspect that
remained unnoticed until recently due to their low frequency in
the brain. Still, they have occasionally been detected by
microscopy on both mouse models (Belnoue et al., 2002)as
well as on the brain of Malawian children who died of CM
(Dorovini-Zis et al., 2011). A similar strategy of isolating and
analyzing immune cells from an entire organ has also been used
to study lung and placental pathology in mouse models in order
to dissect the players on the immune response occurring in these
organs (Hee et al., 2011; Van den Steen et al., 2010). Despite the
advances in the understanding of immunopathology, these
approaches to study ECM have failed to detect sequestration of
pRBCs within the brain vasculature, a hallmark of severe P.
falciparum malaria. The recent availability of transgenic chemi-
luminescent parasite lines allowed the in vivo real-time imaging
of pRBCs, bringing valuable information on the dynamic of
parasite biomass/sequestration in different organs, including
placenta of pregnant mice, brain, lungs and its association with
pathogenesis (Franke-Fayard et al., 2006; Franke-Fayard et al.,
2005). Ex vivo imaging revealed random parasite accumula-
tion/sequestration in the brain of mice developing ECM
(Baptista et al., 2010). The engineering of transgenic
parasites expressing luciferase under the control of a
schizont-specific promoter allowed the analysis of the
organ-specific distribution of these mature forms of the P.
berghei parasite (reviewed in Franke-Fayard et al., 2010).
The development of imaging technologies has also signifi-
cantly contributed to the characterization of malaria pathology.
The recent availability of high performance computer tomog-
raphy (CT) and magnetic resonance imaging (MRI) equipment
for small animals, allowed in vivo monitoring of neuropathol-
ogy during ECM (Penetetal.,2005). These and related
non-invasive imaging techniques, applied for the first time to
the study of ECM in 2005 (Penet et al., 2005), have huge
potential since they can provide a large spectrum of informa-
tion such as localization of structural lesions, edema, hemor-
rhages, and thrombosis. Importantly, magnetic resonance
techniques also revealed differences in the blood flow and
metabolic changes in the brains of infected but ECM-resistant
mice (Penet et al., 2007).
Intravital microscopy (IVM) enables the in vivo analysis of
parasite-host interaction at the cellular level, allowing the
measure of blood flow, vascular leakage, cellular adhesion to
the endothelium, leukocyte recruitment and migration and it
has been used mainly for ECM (reviewed in Frevert et al.,
2014). Moreover, the use of this technique in combination
with the newly available fluorescent parasite lines (Graewe
et al., 2009) increases the possibilities of analyses. The use of
these imaging technologies can also be of great importance to
understand other severe malaria complications, such as
placental malaria (de Moraes et al., 2013; Conroy et al.,
2013).
Acknowledgements
The authors gratefully acknowledge Sabrina Epiphanio
and Cláudio Marinho for providing pictures used in this
review, and Iset Vera, Ana Pamplona and Miguel Prudêncio
for critical reading of the manuscript. V.Z.-L. is funded by
FCT fellowship (SFRH/BPD/81953/2011). We apologize to
all authors whose work was not cited due to constraints on
the number of references.
References
Adachi, K., Tsutsui, H., Kashiwamura, S., Seki, E., Nakano, H., Takeuchi, O.,
Takeda, K., Okumura, K., Van Kaer, L., Okamura, H., Akira, S., Nakanishi, K.,
2001. Plasmodium berghei infection in mice induces liver injury by an IL-
12- and toll-like receptor/myeloid differentiation factor 88-dependent
mechanism. J. Immunol. 167, 5928.
Albuquerque, S.S., Carret, C., Grosso, A.R., Tarun, A.S., Peng, X., Kappe, S.H.,
Prudencio, M., Mota, M.M., 2009. Host cell transcriptional profiling
during malaria liver stage infection reveals a coordinated and sequential
set of biological events. BMC Genomics 10, 270.
Amino, R., Thiberge, S., Martin, B., Celli, S., Shorte, S., Frischknecht, F.,
Menard, R., 2006. Quantitative imaging of Plasmodium transmission
from mosquito to mammal. Nat. Med. 12, 220.
Amino, R., Giovannini, D., Thiberge, S., Gueirard, P., Boisson, B., Dubremetz, J.F.,
Prevost, M.C., Ishino, T., Yuda, M., Menard, R., 2008. Host cell traversal is
importantfor progression of themalaria parasite throughthe dermis to the
liver. Cell Host Microbe 3, 88.
Azuma, H., Paulk, N., Ranade, A., Dorrell, C., Al-Dhalimy, M., Ellis, E., Strom, S.,
Kay, M.A., Finegold, M., Grompe, M., 2007. Robust expansion of human
hepatocytes in Fah-/-/Rag2-/-/Il2rg-/- mice. Nat. Biotechnol. 25, 903.
Baer, K., Klotz, C., Kappe, S.H., Schnieder, T., Frevert, U., 2007. Release of
hepatic Plasmodium yoelii merozoites into the pulmonary microvascu-
lature. PLoS Pathog. 3, e171.
Baird, J.K., 2013. Evidence and implications of mortality associated with
acute Plasmodium vivax malaria. Clin. Microbiol. Rev. 26, 36.
Baptista, F.G., Pamplona, A., Pena, A.C., Mota, M.M., Pied, S., Vigario, A.M.,
2010. Accumulation of Plasmodium berghei-infected red blood cells in
the brain is crucial for the development of cerebral malaria in mice.
Infect. Immun. 78, 4033.
Bei, A.K., Duraisingh, M.T., 2012. Functional analysis of erythrocyte
determinants of Plasmodium infection. Int. J. Parasitol. 42, 575.
Belnoue, E., Kayibanda, M., Vigario, A.M., Deschemin, J.C., van Rooijen, N.,
Viguier, M., Snounou, G., Renia, L., 2002. On the pathogenic role of brain-
sequestered alphabeta CD8+ T cells in experimental cerebral malaria. J.
Immunol. 169, 6369.
Belnoue, E., Costa, F.T., Frankenberg, T., Vigario, A.M., Voza, T., Leroy, N.,
Rodrigues, M.M., Landau, I., Snounou, G., Renia, L., 2004. Protective T cell
immunity against malaria liver stage after vaccination with live
sporozoites under chloroquine treatment. J. Immunol. 172, 2487.
Bissig, K.D., Le, T.T., Woods, N.B., Verma, I.M., 2007. Repopulation of adult
and neonatal mice with human hepatocytes: a chimeric animal model.
Proc. Natl. Acad. Sci. U. S. A. 104, 20507.
Bissig, K.D., Wieland,S.F., Tran, P., Isogawa,M., Le, T.T., Chisari, F.V., Verma, I.M.,
2010. Human liver chimeric mice provide a model for hepatitis B and C
virus infection and treatment. J. Clin. Invest. 120, 924.
Boyd, M.F.K., S.F., 1939. The demonstration of sporozoites in human tissues.
Am. J. Trop. Med. Hyg. 19, 27.
Clyde, D.F., 1975. Immunization of man against falciparum and vivax malaria
by use of attenuated sporozoites. Am. J. Trop. Med. Hyg. 24, 397.
Coatney, G.R., Collins, W.E., Warren, M., Contacos, P.G., 1971. The primate
malarias. p. 1.
Combes, V.,Rosenkranz, A.R.,Redard, M., Pizzolato, G., Lepidi,H., Vestweber, D.,
Mayadas, T.N., Grau, G.E., 2004. Pathogenic role of P-selectin in experi-
mental cerebral malaria: importance of theendothelial compartment. Am.
J. Pathol. 164, 781.
Conroy, A.L., Silver, K.L., Zhong, K., Rennie, M., Ward, P., Sarma, J.V.,
Molyneux, M.E., Sled, J., Fletcher, J.F., Rogerson, S., Kain, K.C., 2013.
Complement activation and the resulting placental vascular insufficien-
cy drives fetal growth restriction associated with placental malaria. Cell
Host Microbe 13, 215.
Cotter, C., Sturrock, H.J., Hsiang, M.S., Liu, J., Phillips, A.A., Hwang, J., Gueye, C.S.,
Fullman, N., Gosling, R.D., Feachem, R.G., 2013. The changing epidemi-
ology of malaria elimination: new strategies for new challenges. Lancet
382, 900.
Cowman, A.F., Berry, D., Baum, J., 2012. The cellular and molecular basis for
malaria parasite invasion of the humanred blood cell. J. Cell Biol. 198, 961.
Cox, F.E., 2010. History of the discovery of the malaria parasites and their
vectors. Parasitol. Vectors 3, 5.
Craig, A.G., Grau, G.E., Janse, C., Kazura, J.W., Milner, D., Barnwell, J.W.,
Turner, G., Langhorne, J., 2012. The role of animal models for research on
severe malaria. PLoS Pathog. 8, e1002401.
de Moraes, L.V., Tadokoro, C.E., Gomez-Conde, I., Olivieri, D.N., Penha-
Goncalves, C., 2013. Intravital placenta imaging reveals microcirculatory
120 V. Zuzarte-Luis et al. / Journal of Immunological Methods 410 (2014) 113122
dynamics impact on sequestration and phagocytosis of Plasmodium-
infected erythrocytes. PLoS Pathog. 9, e1003154.
Derbyshire, E.R., Mota, M.M., Clardy, J., 2011. The next opportunity in anti-
malaria drug discovery: the liver stage. PLoS Pathog. 7, e1002178.
Doolan, D.L., Martinez-Alier, N., 2006. Immune response to pre-erythrocytic
stages of malaria parasites. Curr. Mol. Med. 6, 169.
Dorovini-Zis, K., Schmidt, K., Huynh, H., Fu, W., Whitten, R.O., Milner, D.,
Kamiza, S., Molyneux, M., Taylor, T.E., 2011. The neuropathology of fatal
cerebral malaria in malawian children. Am. J. Pathol. 178, 2146.
Douglas, N.M.,Anstey, N.M., Buffet, P.A.,Poespoprodjo, J.R., Yeo, T.W., White, N.J.,
Price, R.N., 2012. The anaemia of Plasmodium vivax malaria. Malar. J. 11, 135.
Driss, A., Hibbert, J.M., Wilson, N.O., Iqbal, S.A., Adamkiewicz, T.V., S tiles, J.K., 2011.
Genetic polymorphisms linked to susceptibility to malaria. Malar. J. 10, 271.
Duffy, P.E., Sahu, T., Akue, A., Milman, N., Anderson, C., 2012. Pre-erythrocytic
malaria vaccines: identifying the targets. Expert Rev. Vaccin. 11, 1261.
Ejigiri, I., Sinnis, P., 2009. Plasmodium sporozoite-host interactions from the
dermis to the hepatocyte. Curr. Opin. Microbiol. 12, 401.
Elias, R.M., Correa-Costa, M., Barreto, C.R., Silva, R.C., Hayashida, C.Y., Castoldi,
A., Goncalves, G.M., Braga, T.T., Barboza, R., Rios, F.J., Keller, A.C., Cenedeze,
M.A., Hyane, M.I., D'Imperio-Lima, M.R., Figueiredo-Neto, A.M., Reis, M.A.,
Marinho, C.R., Pacheco-Silva, A., Camara, N.O., 2012. Oxidative stress and
modification of renal vascular permeability are associated with acute
kidney injury during P. berghei ANKA infection. PLoS One 7, e44004.
Epiphanio, S., Mikolajczak, S.A., Goncalves, L.A., Pamplona, A., Portugal, S.,
Albuquerque, S., Goldberg, M., Rebelo, S., Anderson, D.G., Akinc, A.,
Vornlocher, H.P., Kappe, S.H., Soares, M.P., Mota, M.M., 2008. Heme
oxygenase-1 is an anti-inflammatory host factor that promotes murine
plasmodium liver infection. Cell Host Microbe 3, 331.
Epiphanio, S., Campos, M.G., Pamplona, A., Carapau, D., Pena, A.C., Ataide, R.,
Monteiro, C.A., Felix, N., Costa-Silva, A., Marinho, C.R., Dias, S., Mota, M.M.,
2010. VEGF promotes malaria-associated acute lung injury in mice. PLoS
Pathog. 6, e1000916.
Evans, K.J., Hansen, D.S., van Rooijen, N., Buckingham, L.A., Schofield, L., 2006.
Severe malarial anemia of low parasite burden in rodent models results
from accelerated clearance of uninfected erythrocytes. Blood 107, 1192.
Franke-Fayard, B., Janse, C.J., Cunha-Rodrigues, M., Ramesar, J., Buscher, P.,
Que, I., Lowik, C., Voshol, P.J., denBoer, M.A., van Duinen, S.G., Febbraio, M.,
Mota, M.M., Waters, A.P., 2005. Murine malaria parasite sequestration:
CD36 is the major receptor, but cerebral pathology is unlinked to
sequestration. Proc. Natl. Acad. Sci. U. S. A. 102, 11468.
Franke-Fayard, B., Waters, A.P., Janse, C.J., 2006. Real-time in vivo imaging of
transgenic bioluminescent blood stages of rodent malaria parasites in
mice. Nat. Protoc. 1, 476.
Franke-Fayard,B.,Fonager,J.,Braks,A.,Khan,S.M.,Janse,C.J.,2010.Sequestration
and tissue accumulation of human malaria parasites: can we learn anything
from rodent models of malaria? PLoS Pathog. 6, e1001032.
Frech, C., Chen, N., 2011. Genome comparison of human and non-human
malaria parasites reveals species subset-specific genes potentially linked
to human disease. PLoS Comput. Biol. 7, e1002320.
Frevert, U., Engelmann, S., Zougbede, S., Stange, J., Ng, B., Matuschewski, K.,
Liebes, L., Yee, H., 2005. Intravital observation of Plasmodium berghei
sporozoite infection of the liver. PLoS Biol. 3, e192.
Frevert, U., Nacer, A., Cabrera, M., Movila, A., Leberl, M., 2014. Imaging
Plasmodium immunobiology in the liver, brain, and lung. Parasitol. Int.
63, 171.
Frischknecht, F., 2007. The skin as interface in the transmission of arthropod-
borne pathogens. Cell. Microbiol. 9, 1630.
Gomes-Santos, C.S., Itoe, M.A., Afonso, C., Henriques, R., Gardner, R.,
Sepulveda, N., Simoes, P.D., Raquel, H., Almeida, A.P., Moita, L.F.,
Frischknecht, F., Mota, M.M., 2012. Highly dynamic host actin reorgani-
zation around developing Plasmodium inside hepatocytes. PLoS One 7,
e29408.
Graewe, S., Retzlaff, S., Struck, N., Janse, C.J., Heussler, V.T., 2009. Going live: a
comparative analysis of the suitability of the RFP derivatives RedStar,
mCherry and tdTomato for intravital and in vitro live imaging of
Plasmodium parasites. Biotechnol. J. 4, 895.
Gwadz, R.W., Cochrane, A.H., Nussenzweig, V., Nussenzweig, R.S., 1979.
Preliminary studies on vaccination of rhesus monkeys with irradiated
sporozoites of Plasmodium knowlesi and characterization of surface
antigens of these parasites. Bull. World Health Organ. 57 (Suppl. 1),
165.
Haldar, K., Murphy, S.C., Milner, D.A., Taylor, T.E., 2007. Malaria: mechanisms
of erythrocytic infection and pathological correlates of severe disease.
Annu. Rev. Pathol. 2, 217.
Haque, A., Best, S.E., Unosson, K., Amante, F.H., de Labastida, F., Anstey, N.M.,
Karupiah, G., Smyth, M.J., Heath, W.R., Engwerda, C.R., 2011a. Granzyme
B expression by CD8+ T cells is required for the development of
experimental cerebral malaria. J. Immunol. 186, 6148.
Haque, A., Best, S.E., Amante, F.H., Ammerdorffer, A., de Labastida, F., Pereira, T.,
Ramm, G.A., Engwerda, C.R., 2011b. High parasite burdens cause liver damage
in mice following Plasmodium berghei ANKA infection independently of
CD8(+) T cell-mediated immune pathology. Infect. Immun. 79, 1882.
Harris, J.V., Bohr, T.M., Stracener, C., Landmesser, M.E., Torres, V., Mbugua, A.,
Moratz, C., Stoute, J.A., 2012. Sequential Plasmodium chabaudi and
Plasmodium berghei infections provide a novel model of severe malarial
anemia. Infect. Immun. 80, 2997.
Hearn, J., Rayment, N., Landon, D.N., Katz, D.R., de Souza, J.B., 2000.
Immunopathology of cerebral malaria: morphological evidence of parasite
sequestration in murine brain microvasculature. Infect. Immun. 68, 5364.
Hee, L., Dinudom, A., Mitchell, A.J., Grau, G.E., Cook, D.I., Hunt, N.H., Ball, H.J.,
2011. Reduced activity of the epithelial sodium channel in malaria-
induced pulmonary oedema in mice. Int. J. Parasitol. 41, 81.
Hioki, A., Hioki, Y., Ohtomo, H., 1990. Influenceofpregnancyonthecourseof
malaria in mice infected with Plasmodium berghei. J. Protozool. 37, 163.
Ishino, T., Yano, K., Chinzei, Y., Yuda, M., 2004. Cell-passage activity is
required for the malarial parasite to cross the liver sinusoidal cell layer.
PLoS Biol. 2, E4.
Joshi, Y.K., Tandon, B.N., Acharya, S.K., Babu, S., Tandon, M., 1986. Acute
hepatic failure due to Plasmodium falciparum liver injury. Liver 6, 357.
Kondrashin, A., Baranova, A.M., Ashley, E.A., Recht, J., White, N.J., Sergiev, V.P.,
2014. Massprimaquine treatmentto eliminate vivax malaria: lessons from
the past. Malar. J. 13, 51.
Langhorne,J.,Buffet,P.,Galinski,M.,Good,M.,Harty,J.,Leroy,D.,Mota,M.M.,
Pasini,E.,Renia,L.,Riley,E.,Stins,M.,Duffy,P.,2011.The relevance of non-
human primate and rodent malaria models for humans. Malar. J. 10, 23.
Liehl, P., Mota, M.M., 2012. Innate recognition of malarial parasites by
mammalian hosts. Int. J. Parasitol. 42, 557.
Liehl,P.,Zuzarte-Luis,V.,Chan,J.,Zillinger,T.,Baptista,F.,Carapau,D.,Konert,M.,
Hanson, K.K., Carret, C., Lassnig, C., Muller, M., Kalinke, U., Saeed,M., Chora,
A.F., Golenbock, D.T., Strobl, B., Prudencio, M., Coelho, L.P., Kappe, S.H.,
Superti-Furga, G., Pichlmair, A., Vigario, A.M., Rice, C.M., Fitzgerald, K.A.,
Barchet, W., Mota, M.M., 2014. Host-cell sensors for Plasmodium activate
innate immunity against liver-stage infection. Nat. Med. 20, 47.
Lloyd, O.C., Sommerville, T., 1949. The fate of sporozoites of Plasmodium
cynomolgi injected into the skin of rhesus monkeys. Proc. Path. Soc. 61, 144.
Lovegrove, F.E., Gharib, S.A., Pena-Castillo, L., Patel, S.N., Ruzinski, J.T.,
Hughes, T.R., Liles, W.C., Kain, K.C., 2008. Parasite burden and CD36-
mediated sequestration are determinants of acute lung injury in an
experimental malaria model. PLoS Pathog. 4, e1000068.
March, S., Ng, S., Velmurugan, S., Galstian, A., Shan, J., Logan, D.J., Carpenter,
A.E., Thomas, D., Sim, B.K., Mota, M.M., Hoffman, S.L., Bhatia, S.N., 2013. A
microscale human liver platform that supports the hepatic stages of
Plasmodium falciparum and vivax. Cell Host Microbe 14, 104.
Marinho, C.R., Neres, R., Epiphanio, S., Goncalves, L.A., Catarino, M.B., Penha-
Goncalves, C., 2009. Recrudescent Plasmodium berghei from pregnant
mice displays enhanced binding to the placenta and induces protection
in multigravida. PLoS One 4, e5630.
Martins, Y.C., Clemmer, L., Orjuela-Sanchez, P., Zanini, G.M., Ong, P.K.,
Frangos, J.A., Carvalho, L.J., 2013. Slow and continuous delivery of a low
dose of nimodipine improves survival and electrocardiogram parame-
ters in rescue therapy of mice with experimental cerebral malaria.
Malar. J. 12, 138.
Matsuoka, H., Yoshida, S., Hirai, M., Ishii, A., 2002. A rodent malaria,
Plasmodium berghei, is experimentally transmitted to mice by merely
probing of infective mosquito, Anopheles stephensi. Parasitol. Int. 51, 17.
Megnekou, R., Hviid, L., Staalsoe, T., 2009. Variant-specific immunity to
Plasmodium berghei in pregnant mice. Infect. Immun. 77, 1827.
Menard, R., Tavares, J., Cockburn, I., Markus, M., Zavala, F., Amino, R., 2013.
Looking under the skin: the first steps in malarial infection and
immunity. Nat. Rev. Microbiol. 11, 701.
Mercer, D.F., Schiller, D.E., Elliott, J.F., Douglas, D.N., Hao, C., Rinfret, A.,
Addison, W.R., Fischer, K.P., Churchill, T.A., Lakey, J.R., Tyrrell, D.L.,
Kneteman, N.M., 2001. Hepatitis C virus replication in mice with
chimeric human livers. Nat. Med. 7, 927.
Meuleman, P., Libbrecht, L., De Vos, R., de Hemptinne, B., Gevaert, K.,
Vandekerckhove, J., Roskams, T., Leroux-Roels, G., 2005. Morphological
and biochemical characterization of a human liver in a uPA-SCID mouse
chimera. Hepatology 41, 847.
Mikolajczak, S.A., Sacci Jr., J.B., De La Vega, P., Camargo, N., VanBuskirk, K.,
Krzych, U., Cao, J., Jacobs-Lorena, M., Cowman, A.F., Kappe, S.H., 2011.
Disruption of the Plasmodium falciparum liver-stage antigen-1 locus
causes a differentiation defect in late liver-stage parasites. Cell.
Microbiol. 13, 1250.
Mishra, S.K., Wiese, L., 2009. Advances in the management of cerebral
malaria in adults. Curr. Opin. Neurol. 22, 302.
Morosan, S., Hez-Deroubaix, S., Lunel, F., Renia, L., Giannini, C., Van Rooijen,
N., Battaglia, S., Blanc, C., Eling, W., Sauerwein, R., Hannoun, L., Belghiti, J.,
Brechot, C., Kremsdorf, D., Druilhe, P., 2006. Liver-stage development of
Plasmodium falciparum, in a humanized mouse model. J. Infect. Dis. 193,
996.
121V. Zuzarte-Luis et al. / Journal of Immunological Methods 410 (2014) 113122
Mota, M.M., Pradel, G., Vanderberg, J.P., Hafalla, J.C., Frevert, U., Nussenzweig,
R.S., Nussenzweig, V., Rodriguez, A., 2001. Migration of Plasmodium
sporozoites through cells before infection. Science 291, 141.
Mueller, A.K., Camargo, N., Kaiser, K., Andorfer, C., Frevert, U., Matuschewski,
K., Kappe, S.H., 2005a. Plasmodium liver stage developmental arrest by
depletion of a protein at the parasite-host interface. Proc. Natl. Acad. Sci.
U. S. A. 102, 3022.
Mueller, A.K., Labaied, M., Kappe, S.H., Matuschewski, K., 2005b. Genetically
modified Plasmodium parasites as a protective experimental malaria
vaccine. Nature 433, 164.
Neres, R., Marinho, C.R., Goncalves, L.A., Catarino, M.B., Penha-Goncalves, C.,
2008. Pregnancy outcome and placenta pathology in Plasmodium
berghei ANKA infected mice reproduce the pathogenesis of severe
malaria in pregnant women. PLoS One 3, e1608.
Ng, S., March, S., Galstian, A., Hanson, K., Carvalho, T., Mota, M.M., Bhatia, S.N.,
2014. Hypoxia promotes liver stage malaria infection in primary human
hepatocytes in vitro. Dis. Model. Mech. 7, 215.
Nitcheu, J., Bonduelle, O., Combadiere, C., Tefit, M., Seilhean, D., Mazier, D.,
Combadiere, B., 2003. Perforin-dependent brain-infiltrating cytotoxic
CD8+ T lymphocytes mediate experimental cerebral malaria pathogen-
esis. J. Immunol. 170, 2221.
Nussenzweig, R.S., Vanderberg, J., Most, H., Orton, C., 1967. Protective
immunity produced by the injection of x-irradiated sporozoites of
plasmodium berghei. Nature 216, 160.
Oduola, A.M., Holbrook, T.W., Galbraith, R.M., Bank, H., Spicer, S.S., 1982.
Effects of malaria (Plasmodium berghei) on the maternal-fetal relation-
ship in mice. J. Protozool. 29, 77.
Olszewski, K.L., Morrisey, J.M., Wilinski, D., Burns, J.M., Vaidya, A.B.,
Rabinowitz, J.D., Llinas, M., 2009. Host-parasite interactions revealed
by Plasmodium falciparum metabolomics. Cell Host Microbe 5, 191.
Penet, M.F., Viola, A., Confort-Gouny, S., Le Fur, Y., Duhamel, G., Kober, F.,
Ibarrola, D., Izquierdo, M., Coltel, N., Gharib, B., Grau, G.E., Cozzone, P.J.,
2005. Imaging experimental cerebral malaria in vivo: significant role of
ischemic brain edema. J. Neurosci. 25, 7352.
Penet, M.F., Kober, F., Confort-Gouny, S., Le Fur, Y., Dalmasso, C., Coltel, N.,
Liprandi, A., Gulian, J.M., Grau, G.E., Cozzone, P.J., Viola, A., 2007.
Magnetic resonance spectroscopy reveals an impaired brain metabolic
profile in mice resistant to cerebral malaria infected with Plasmodium
berghei ANKA. J. Biol. Chem. 282, 14505.
Perkins, D.J., Were, T., Davenport, G.C., Kempaiah, P., Hittner, J.B., Ong'echa, J.M.,
2011. Severe malarial anemia: innate immunity and pathogenesis. Int. J.
Biol. Sci. 7, 1427.
Ploemen, I.H., Prudencio, M., Douradinha, B.G., Ramesar, J., Fonager, J., van
Gemert, G.J., Luty, A.J., Hermsen, C.C., Sauerwein, R.W., Baptista, F.G.,
Mota, M.M., Waters, A.P., Que, I., Lowik, C.W., Khan, S.M., Janse, C.J.,
Franke-Fayard, B.M., 2009. Visualisation and quantitative analysis of the
rodent malaria liver stage by real time imaging. PLoS One 4, e7881.
Polder, T.W., Eling, W.M., Curfs, J.H., Jerusalem, C.R., Wijers-Rouw, M., 1992.
Ultrastructural changes in the blood-brain barrier of mice infected with
Plasmodium berghei. Acta Leiden 60, 31.
Ponnudurai, T., Lensen, A.H., van Gemert, G.J., Bolmer, M.G., Meuwissen, J.H.,
1991. Feeding behaviour and sporozoite ejection by infected Anopheles
stephensi. Trans. R. Soc. Trop. Med. Hyg. 85, 175.
Poovassery, J., Moore, J.M., 2006. Murine malaria infection induces fetal loss
associated with accumulation of Plasmodium chabaudi AS-infected
erythrocytes in the placenta. Infect. Immun. 74, 2839.
Portugal, S., Carret, C., Recker, M., Armitage, A.E., Goncalves, L.A., Epiphanio, S.,
Sullivan, D., Roy, C., Newbold, C.I., Drakesmith, H., Mota, M.M., 2011. Host-
mediated regulation of superinfection in malaria. Nat. Med. 17, 732.
Prudencio, M., Rodriguez, A., Mota, M.M., 2006. The silent path to thousands
of merozoites: the Plasmodium liver stage. Nat. Rev. Microbiol. 4, 849.
Rodrigues, T., Prudencio, M., Moreira, R., Mota, M.M., Lopes, F., 2012.
Targeting the liver stage of malaria parasites: a yet unmet goal. J.
Med. Chem. 55, 995.
Rodrigues-Duarte, L., de Moraes, L.V., Barboza, R., Marinho, C.R., Franke-
Fayard, B., Janse, C.J., Penha-Goncalves, C., 2012. Distinct placental
malaria pathology caused by different Plasmodium berghei lines that
fail to induce cerebral malaria in the C57BL/6 mouse. Malar. J. 11,
231.
Roestenberg, M., McCall, M., Hopman, J., Wiersma, J., Luty, A.J., van Gemert,
G.J., van de Vegte-Bolmer, M., van Schaijk, B., Teelen, K., Arens, T.,
Spaarman, L., de Mast, Q., Roeffen, W., Snounou, G., Renia, L., van der
Ven, A., Hermsen, C.C., Sauerwein, R., 2009. Protection against a malaria
challenge by sporozoite inoculation. N. Engl. J. Med. 361, 468.
Rogerson, S.J., Hviid, L., Duffy, P.E., Leke, R.F., Taylor, D.W., 2007. Malaria in
pregnancy: pathogenesis and immunity. Lancet Infect. Dis. 7, 105.
Sacci Jr., J.B., Schriefer, M.E., Resau, J.H., Wirtz, R.A., Detolla Jr., L.J., Markham,
R.B., Azad, A.F., 1992. Mouse model for exoerythrocytic stages of
Plasmodium falciparum malaria parasite. Proc. Natl. Acad. Sci. U. S. A.
89, 3701.
SacciJr.,J.B.,Alam,U.,Douglas,D.,Lewis,J.,Tyrrell,D.L.,Azad,A.F.,Kneteman,N.M.,
2006. Plasmodium falciparum infection and exoerythrocytic development in
mice with chimeric human livers. Int. J. Parasitol. 36, 353.
Schofield, L., 2007. Intravascular infiltrates and organ-specific inflammation
in malaria pathogenesis. Immunol. Cell Biol. 85, 130.
Seixas, E., Cross, C., Quin, S., Langhorne, J., 2001. Direct activation of dendritic
cells by the malaria parasite, Plasmodium chabaudi chabaudi. Eur. J.
Immunol. 31, 2970.
Shanks, G.D., White, N.J., 2013. The activation of vivax malaria hypnozoites
by infectious diseases. Lancet Infect. Dis. 13, 900.
Sidjanski, S., Vanderberg, J.P., 1997. Delayed migration of Plasmodium sporozoites
from the mosquito bite site to the blood. Am. J. Trop. Med. Hyg. 57, 426.
Sinniah, R., Rui-Mei, L., Kara, A., 1999. Up-regulation of cytokines in
glomerulonephritis associated with murine malaria infection. Int. J.
Exp. Pathol. 80, 87.
Sinnis, P., Zavala, F., 2012. The skin: where malaria infection and the host
immune response begin. Semin. Immunopathol. 34, 787.
Sturm, A., Amino, R., van de Sand, C., Regen, T., Retzlaff, S., Rennenberg, A.,
Krueger, A., Pollok, J.M., Menard, R., Heussler, V.T., 2006. Manipulation of
host hepatocytes by the malaria parasite for delivery into liver sinusoids.
Science 313, 1287.
Tarun, A.S., Peng, X., Dumpit, R.F., Ogata, Y., Silva-Rivera, H., Camargo, N.,
Daly, T.M., Bergman, L.W., Kappe, S.H., 2008. A combined transcriptome
and proteome survey of malaria parasite liver stages. Proc. Natl. Acad.
Sci. U. S. A. 105, 305.
Tavares, J., Formaglio, P., Thiberge, S., Mordelet, E., Van Rooijen, N.,
Medvinsky, A., Menard, R., Amino, R., 2013. Role of host cell traversal
by the malaria sporozoite during liver infection. J. Exp. Med. 210, 905.
Taylor, W.R., Hanson, J., Turner, G.D., White, N.J., Dondorp, A.M., 2012.
Respiratory manifestations of malaria. Chest 142, 492.
The malERA Consultative Group on Basic Science and Enabling Technologies,
2011. A research agenda for malaria eradication: basic science and
enabling technologies. PLoS Med. 8, e1000399.
Trang, T.T., Phu, N.H., Vinh, H., Hien, T.T., Cuong, B.M., Chau, T.T., Mai, N.T.,
Waller, D.J., White, N.J., 1992. Acute renal failure in patients with severe
falciparum malaria. Clin. Infect. Dis. 15, 874.
Van den Steen, P.E., Geurts, N., Deroost, K., Van Aelst, I., Verhenne, S.,
Heremans, H., Van Damme, J., Opdenakker, G., 2010. Immunopathology
and dexamethasone therapy in a new model for malaria-associated
acute respiratory distress syndrome. Am. J. Respir. Crit. Care Med. 181,
957.
van Dijk, M.R., Douradinha, B.,Franke-Fayard, B., Heussler,V., van Dooren, M.W.,
van Schaijk, B., van Gemert, G.J., Sauerwein, R.W., Mota, M.M., Waters, A.P.,
Janse, C.J., 2005. Genetically attenuated, P36p-deficient malarial sporozoites
induce protective immunity and apoptosis of infected liver cells. Proc. Natl.
Acad. Sci. U. S. A. 102, 12194.
van Zon, A.A., Eling, W.M., 1980. Depressed malarial immunity in pregnant
mice. Infect. Immun. 28, 630.
VanBuskirk, K.M., O'Neill, M.T., De La Vega, P., Maier, A.G., Krzych, U.,
Williams, J., Dowler, M.G., Sacci Jr., J.B., Kangwanrangsan, N., Tsuboi, T.,
Kneteman, N.M., Heppner Jr., D.G., Murdock, B.A., Mikolajczak, S.A., Aly,
A.S., Cowman, A.F., Kappe, S.H., 2009. Preerythrocytic, live-attenuated
Plasmodium falciparum vaccine candidates by design. Proc. Natl. Acad.
Sci. U. S. A. 106, 13004.
Vanderberg, J.P., Frevert, U., 2004. Intravital microscopy demonstrating
antibody-mediated immobilisation of Plasmodium berghei sporozoites
injected into skin by mosquitoes. Int. J. Parasitol. 34, 991.
Vanderberg, J.P., Chew, S., Stewart, M.J., 1990. Plasmodium sporozoite
interactions with macrophages in vitro: a videomicroscopic analysis. J.
Protozool. 37, 528.
Vaughan, A.M., O'Neill, M.T., Tarun, A.S., Camargo, N., Phuong, T.M., Aly, A.S.,
Cowman, A.F., Kappe, S.H., 2009. Type II fatty acid synthesis is essential
only for malaria parasite late liver stage development. Cell. Microbiol.
11, 506.
Vaughan, A.M., Kappe, S.H., Ploss, A., Mikolajczak, S.A., 2012. Development of
humanized mouse models to study human malaria parasite infection.
Future Microbiol 7, 657.
Vigario, A.M., Belnoue, E., Gruner, A.C., Mauduit, M., Kayibanda, M.,
Deschemin, J.C., Marussig, M., Snounou, G., Mazier, D., Gresser, I.,
Renia, L., 2007. Recombinant human IFN-alpha inhibits cerebral malaria
and reduces parasite burden in mice. J. Immunol. 178, 6416.
White, N.J., Turner, G.D., Medana, I.M., Dondorp, A.M., Day, N.P., 2010. The
murine cerebral malaria phenomenon. Trends Parasitol. 26, 11.
WHO, 2013. World Malaria Report: 2013, p. 284.
Yoshimoto, T., Takahama, Y., Wang, C.R., Yoneto, T., Waki, S., Nariuchi, H.,
1998. A pathogenic role of IL-12 in blood-stage murine malaria lethal
strain Plasmodium berghei NK65 infection. J. Immunol. 160, 5500.
Zuzarte-Luis, V., Sales-Dias, J., Mota, M.M., 2014. Simple, sensitive and
quantitative bioluminescence assay for determination of malaria pre-
patent period. Malar. J. 13, 15.
122 V. Zuzarte-Luis et al. / Journal of Immunological Methods 410 (2014) 113122
... There are many different mouse malaria models, with very different characteristics dependening on the combination of parasite species (and strain) and mouse strain which are used (Lamb et al., 2006). Superficially these models can, between them, reproduce almost all the clinical manifestations of human SM, such as coma, seizures, respiratory distress, and severe anemia (SA) (Zuzarte-Luis et al., 2014). Nevertheless, there are also notable differences to human disease, such as the lack of the pathognomonic cytoadhesive sequestration of large numbers of parasite-infected red cells in the cerebral microvasculature in mice with cerebral malaria-like illness (experimental cerebral malaria, ECM) (Ghazanfari et al., 2018). ...
... In C57BL/6 mice infected with Plasmodium berghei ANKA, ECM is dependent on recruitment of CD8+ T cells to the brain, a phenomenon that was recently shown to also occur in human cerebral malaria (Riggle et al., 2020). C57BL/6 mice infected with P. berghei NK65 develop acute lung injury with similarities to malaria-associated acute respiratory distress syndrome (MA-ARDS), associated with hemozoin accumulation (Deroost et al., 2013), endothelial activation, and alveolar edema (Zuzarte-Luis et al., 2014;Van den Steen et al., 2010;Claser et al., 2019). SA can occur in C57BL/6 mice infected with all of the most commonly used mouse malaria parasite species (P. ...
Article
Full-text available
Recent initiatives to improve translation of findings from animal models to human disease have focussed on reproducibility but quantifying the relevance of animal models remains a challenge. Here, we use comparative transcriptomics of blood to evaluate the systemic host response and its concordance between humans with different clinical manifestations of malaria and five commonly used mouse models. Plasmodium yoelii 17XL infection of mice most closely reproduces the profile of gene expression changes seen in the major human severe malaria syndromes, accompanied by high parasite biomass, severe anemia, hyperlactatemia, and cerebral microvascular pathology. However, there is also considerable discordance of changes in gene expression between the different host species and across all models, indicating that the relevance of biological mechanisms of interest in each model should be assessed before conducting experiments. These data will aid the selection of appropriate models for translational malaria research, and the approach is generalizable to other disease models.
... yoelii), and Plasmodium vinckei (P. vinckei), are the most common species used to establish malaria infection in rodent model (Wykes et al., 2009;Zuzarte-Luis et al., 2014). Amongst all, P. berghei is the only species that develops almost all severe malaria syndromes, including cerebral malaria, placental malaria, acute lung injury / acute respiratory distress syndrome, acute kidney injury, and severe malarial anemia in the rodent model (Craig et al., 2012;Zuzarte-Luis et al., 2014). ...
... vinckei), are the most common species used to establish malaria infection in rodent model (Wykes et al., 2009;Zuzarte-Luis et al., 2014). Amongst all, P. berghei is the only species that develops almost all severe malaria syndromes, including cerebral malaria, placental malaria, acute lung injury / acute respiratory distress syndrome, acute kidney injury, and severe malarial anemia in the rodent model (Craig et al., 2012;Zuzarte-Luis et al., 2014). P. berghei can readily infect mice, young rats and hamsters but seems to be refractory in adult rats. ...
Article
Malaria infection still remains as one of the most prominent parasitic diseases afflicting mankind in tropical and subtropical regions. The severity of malaria infection has often been associated to exuberant host immune inflammatory responses that could possibly lead to severe immunopathological conditions and subsequent death of host tissues. Activin A is a protein belonging to the transforming growth factor-beta (TGF-β) family that regulates multiple physiological processes and pathological-associated diseases. The biological roles of activin A have been associated with manipulation of inflammation-related processes and modulation of host immune responses. This implies that activin A protein could play a role in malaria pathogenesis since malaria infection has been closely linked to severe immune responses leading to death, However, the actual in vivo role of activin A in malaria infection remains elusive. Hence, this study was undertaken to investigate the involvement of activin A in malaria infection as well as to assess the modulating effects of activin A on the cytokine releases (TNF-α, IFN-γ and IL-10) and histopathological changes in major affected organs (kidney, liver, lung, brain and spleen) in malarial mice infected with Plasmodium berghei ANKA. Our results showed that the concentrations of plasma activin A were significantly increased in malarial mice throughout the study periods. Also. the systemic activin A level was positively correlated with malaria parasitemia. This indicates that activin A could play a role in malaria pathogenesis and malaria parasitemia development. Plasma TNF-α, IFN-γ and IL-10 cytokine levels were significantly increased in malarial mice at day-5 post infection, suggesting that these cytokines attributed to severe malaria pathogenesis. Histopathological features such as sequestration of parasitized red blood cells (pRBCs) and hemozoin formation were amongst the most common pathological conditions observed in tissues of major affected organs (kidney, liver, lung, brain and spleen) in malarial mice. Neutralization of activin A production via recombinant mouse activin RIIA Fc chimera (rmActivin RIIA Fc chimera) had significantly reduced the parasitemia levels in malarial mice. The release of TNF-α cytokine was significantly reduced as well as the sequestration of parasitized pRBCs and hemozoin formation in major affected organs in malarial mice were also alleviated following inhibition of activin A production. Overall, this preliminary study suggests that activin A could play an immune modulation role in malaria pathogenesis through modulation of TNF-α release that benefits host from severe pathological destructions provoked by intensified inflammatory responses. Further studies are warranted to elucidate the precise mechanism of immune modulation mediated by activin A and its associated immune-modulation mediators in regulating the inflammatory responses elicited during the course of malaria infection.
... Some species of Plasmodium such as P. berghei, P. yoelii, and P. chabaudi, have been used to investigate distinct aspects of malaria infections in rodents that mimic certain aspects of human disease, for example, cerebral malaria, placental malaria, liver injury, and blood stage infection. The importance of murine models is widely appreciated, and several reviews have addressed how mouse models have aided our understanding of human malaria infections (Craig et al., 2012;Zuzarte-Luis et al., 2014). Forward genetic screens in murine models have primarily used P. chabaudi and P. yoelii to understand blood-stage infections, and P. berghei to uncover determinants of resistance to cerebral malaria. ...
Article
Full-text available
Forward genetic approaches have been widely used in parasitology and have proven their power to reveal the complexities of host-parasite interactions in an unbiased fashion. Many aspects of the parasite’s biology, including the identification of virulence factors, replication determinants, antibiotic resistance genes, and other factors required for parasitic life, have been discovered using such strategies. Forward genetic approaches have also been employed to understand host resistance mechanisms to parasitic infection. Here, we will introduce and review all forward genetic approaches that have been used to identify host factors involved with Apicomplexa infections, which include classical genetic screens and QTL mapping, GWAS, ENU mutagenesis, overexpression, RNAi and CRISPR-Cas9 library screens. Collectively, these screens have improved our understanding of host resistance mechanisms, immune regulation, vaccine and drug designs for Apicomplexa parasites. We will also discuss how recent advances in molecular genetics give present opportunities to further explore host-parasite relationships.
... Due to obvious challenges of studying human malaria parasites, mouse models of malaria have always been an important part of malaria research (Zuzarte-Luis et al., 2014;De Niz and Heussler, 2018). There is some degree of conservation in the invasion pathways across different human and rodent malaria species, in terms of utilization of homologous parasite ligands (Iyer et al., 2007;Gunalan et al., 2013) and erythrocyte receptors (Swardson-Olver et al., 2002). ...
Article
Full-text available
Circulating red blood cells consist of young erythrocytes (early and late reticulocytes) and mature erythrocytes (normocytes). The human malaria parasites, Plasmodium falciparum and P. vivax , have a preference to invade reticulocytes during blood-stage infection. Rodent malaria parasites that also prefer reticulocytes could be useful tools to study human malaria reticulocyte invasion. However, previous tropism studies of rodent malaria are inconsistent from one another, making it difficult to compare cell preference of different parasite species and strains. In vivo measurements of cell tropism are also subjected to many confounding factors. Here we developed an ex vivo tropism assay for rodent malaria with highly purified fractions of murine reticulocytes and normocytes. We measured invasion into the different erythrocyte populations using flow cytometry and evaluated the tropism index of the parasite strains. We found that P. berghei ANKA displayed the strongest reticulocyte preference, followed by P. yoelii 17X1.1, whereas P. chabaudi AS and P. vinckei S67 showed mixed tropism. These preferences are intrinsic and were maintained at different reticulocyte and normocyte availabilities. Our study shed light on the true erythrocyte preference of the parasites and paves the way for future investigations on the receptor-ligand interactions mediating erythrocyte tropism.
... There are four species of rodent malaria parasites are used in laboratories worldwide; Plasmodium chabaudi, Plasmodium vinckei, Plasmodium yoelii and Plasmodium berghei (Huang et al., 2015) and these are maintained in a large variety of mouse and rat strains. Rodent malaria parasites have informed studies on malaria pathology, cerebral malaria, antimalarial chemotherapy, malaria immunology, and vaccine development (Zuzarte-Luis et al., 2014;Moussa et al., 2018;Sala et al., 2018). ...
Article
Full-text available
The purification of parasite-infected erythrocytes from whole blood containing leucocytes is crucial for many downstream genetic and molecular assays in parasitology. Current methodologies to achieve this are often costly and time consuming. Here, we demonstrate the successful application of a cheap and simple Non-Woven Fabric (NWF) filter for the purification of parasitized red blood cells from whole blood. NWF filtration was applied to the malaria-parasitized blood of three strains of mice, and one strain of rat, and to Babesia gibsoni parasitized dog blood. Before and after filtration, the white blood cell (WBC) removal rates and red blood cell (RBC) recovery rates were measured. After NWF filter treatment of rodent malaria-infected blood, the WBC removal rates and RBC recovery rates were, for Kunming mice: 99.51%±0.30% and 86.12%±8.37%; for BALB/C mice: 99.61%±0.15% and 80.74%±7.11%; for C57 mice: 99.71%±0.12% and 84.87%±3.83%; for Sprague-Dawley rats: 99.93%±0.03% and 83.30%±2.96%. Microscopy showed WBCs were efficiently removed from infected dog blood samples, and there was no obvious morphological change of B. gibsoni parasites. NWF filters efficiently remove leukocytes from malaria parasite-infected mouse and rat blood, and are also suitable for filtration of B. gibsoni-infected dog blood.
... Previous observation showed the occurrence of endothelial dysregulation in CM as a result of upregulation of ICAM-1, V-CAM, and E-Selectin in the brain endothelial cells. 12 Although there were differences in the brain histopathology between human cerebral malaria and in the mouse model of cerebral malaria in which the infected erythrocyte sequestration was absent, but the significant leucocyte accumulation in the brain of the CM C57BL/6 mice was similar to the one found in the human brain with CM. 4,13 In this study, the experimental CM mice infected with malaria were sacrificed on Day 6 or 7 as they demonstrated the CM signs. According to Jennings et al, the mice died on day 7 were usually due to the encephalitis process caused by the increase of proinflammatory cytokines. ...
Article
Malaria is one of transmittable diseases caused by Plasmodium spp. infection. Severe malaria, including cerebral malaria can lead to deaths. Involvement of cytokines and other molecular markers have been shown in development of cerebral malaria signs. The aims of the present study is to determine the effect of Plasmodium berghei ANKA to the HIF-1α and CD11b expressions, and the correlation between HIF-1α CD11b with cerebral malaria symptoms in mice model. C57BL/6 mice were divided into two groups, control group and group that was infected by Plasmodium berghei ANKA. Parasitemia and clinical score were observed until 7th day, then brains were collected and stained by immunohistochemistry to determine HIF-1α and CD11b expression in both groups. The HIF-1α and CD11b expressions were observed under 1000x magnification in light microscope. There was a significant differences between each group (p=0.001) of HIF-1α expression in response to Plasmodium berghei
Article
Full-text available
Coronavirus disease 2019 (COVID-19) and malaria, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and Plasmodium parasites, respectively, share geographical distribution in regions where the latter disease is endemic, leading to the emergence of co-infections between the two pathogens. Thus far, epidemiologic studies and case reports have yielded insufficient data on the reciprocal impact of the two pathogens on either infection and related diseases. We established novel co-infection models to address this issue experimentally, employing either human angiotensin-converting enzyme 2 (hACE2)-expressing or wild-type mice, in combination with human- or mouse-infective variants of SARS-CoV-2, and the P. berghei rodent malaria parasite. We now show that a primary infection by a viral variant that causes a severe disease phenotype partially impairs a subsequent liver infection by the malaria parasite. Additionally, exposure to an attenuated viral variant modulates subsequent immune responses and provides protection from severe malaria-associated outcomes when a blood stage P. berghei infection was established. Our findings unveil a hitherto unknown host-mediated virus-parasite interaction that could have relevant implications for disease management and control in malaria-endemic regions. This work may contribute to the development of other models of concomitant infection between Plasmodium and respiratory viruses, expediting further research on co-infections that lead to complex disease presentations.
Article
Malaria varies in severity, with complications ranging from uncomplicated to severe malaria. Severe malaria could be attributed to peripheral hyperparasitemia or cerebral malaria. The metabolic interactions between the host and Plasmodium species are yet to be understood during these infections of varied pathology and severity. An untargeted metabolomics approach utilizing the liquid chromatography-mass spectrometry platform has been used to identify the affected host metabolic pathways and associated metabolites in the serum of murine malaria models with uncomplicated malaria, hyperparasitemia, and experimental cerebral malaria. We report that mice with malaria share similar metabolic attributes like higher levels of bile acids, bile pigments, and steroid hormones that have been reported for human malaria infections. Moreover, in severe malaria, upregulated levels of metabolites like phenylalanine, histidine, valine, pipecolate, ornithine, and pantothenate, with decreased levels of arginine and hippurate, were observed. Metabolites of sphingolipid metabolism were upregulated in experimental cerebral malaria. Higher levels of 20-hydroxy-leukotriene B4 and epoxyoctadecamonoenoic acids were found in uncomplicated malaria, with lower levels observed for experimental cerebral malaria. Our study provides insights into host biology during different pathological stages of malaria disease and would be useful for the selection of animal models for evaluating diagnostic and therapeutic interventions against malaria. The raw data files are available via MetaboLights with the identifier MTBLS4387.
Article
Full-text available
Malaria control measures have been in use for years but have not completely curbed the spread of infection. Ultimately, global elimination is the goal. A major playmaker in the various approaches to reaching the goal is the issue of proper diagnosis. Various diagnostic techniques were adopted in different regions and geographical locations over the decades, and these have invariably produced diverse outcomes. In this review, we looked at the various approaches used in malaria diagnostics with a focus on methods favorably used during pre-elimination and elimination phases as well as in endemic regions. Microscopy, rapid diagnostic testing (RDT), loop-mediated isothermal amplification (LAMP), and polymerase chain reaction (PCR) are common methods applied depending on prevailing factors, each with its strengths and limitations. As the drive toward the elimination goal intensifies, the search for ideal, simple, fast, and reliable point-of-care diagnostic tools is needed more than ever before to be used in conjunction with a functional surveillance system supported by the ideal vaccine.
Article
Full-text available
Today's malaria control efforts are limited by our incomplete understanding of the biology of Plasmo-dium and of the complex relationships between human populations and the multiple species of mosquito and parasite. Research priorities include the development of in vitro culture systems for the complete life cycle of P. falciparum and P. vivax and the development of an appropriate liver culture system to study hepatic stages. In addition, genetic technologies for the manipulation of Plasmodium need to be improved, the entire parasite metabolome needs to be characterized to identify new druggable targets, and improved information systems for monitoring the changes in epidemiology, pathology, and host-parasite-vector interactions as a result of intensified control need to be established to bridge the gap between bench, preclinical, clinical, and population-based sciences.
Article
Full-text available
Recent successes in malaria control have put malaria eradication back on the public health agenda. A significant obstacle to malaria elimination in Asia is the large burden of Plasmodium vivax, which is more difficult to eliminate than Plasmodium falciparum. Persistent P. vivax liver stages can be eliminated only by radical treatment with a > seven-day course of an 8-aminoquinoline, with the attendant risk of acute haemolytic anaemia in individuals with glucose-6-phosphate dehydrogenase (G6PD) deficiency. Primaquine is the only generally available 8-aminoquinoline. Testing for G6PD deficiency is not widely available, and so whilst it is widely recommended, primaquine is often not prescribed. In the past, some countries aiming for vivax malaria eradication deployed mass treatments with primaquine on a massive scale, without G6PD testing. In Azerbaijan, Tajikistan (formerly USSR), North Afghanistan and DPR Korea 8,270,185 people received either a 14-day "standard" or a 17-day "interrupted" primaquine treatment to control post-eradication malaria epidemics. These mass primaquine preventive treatment campaigns were conducted by dedicated teams who administered the drugs under supervision and then monitored the population for adverse events. Despite estimated G6PD prevalences up to 38.7%, the reported frequency of severe adverse events related to primaquine was very low. This experience shows that with careful planning and implementation of mass treatment strategies using primaquine and adequate medical support to manage haemolytic toxicity, it is possible to achieve high population coverage, substantially reduce malaria transmission, and manage the risk of severe acute haemolytic anaemia in communities with a relatively high prevalence of G6PD deficiency safely.
Article
Full-text available
The first phase of malaria infection occurs in the liver and is clinically silent. Inside hepatocytes each Plasmodium sporozoite replicate into thousands of erythrocyte-infectious merozoites that when released into the blood stream result in clinical symptoms of the disease. The time between sporozoite inoculation and the appearance of parasites in the blood is defined as the pre-patent period, which is classically analysed by time-consuming and labor-intensive techniques, such as microscopy and PCR. Luciferase-expressing Plasmodium berghei parasites were used to measure pre-patent period of malaria infection in rodents using a bioluminescence assay that requires only one microliter of blood collected from the tail-vein. The accuracy and sensitivity of this new method was compared with conventional microscopy and PCR based techniques, and its capacity to measure the impact of anti-malarial interventions against the liver evaluated. The described method is very sensitive allowing the detection of parasites during the first cycles of blood stage replication. It accurately translates differences in liver load due to inoculation of different sporozoite doses as well as a result of treatment with different primaquine regimens. A novel, simple, fast, and sensitive method to measure pre-patent period of malaria infection in rodents is described here. The sensitivity and accuracy of this new method is comparable to standard PCR and microscopy-based techniques, respectively.
Article
Full-text available
Before they infect red blood cells and cause malaria, Plasmodium parasites undergo an obligate and clinically silent expansion phase in the liver that is supposedly undetected by the host. Here, we demonstrate the engagement of a type I interferon (IFN) response during Plasmodium replication in the liver. We identified Plasmodium RNA as a previously unrecognized pathogen-associated molecular pattern (PAMP) capable of activating a type I IFN response via the cytosolic pattern recognition receptor Mda5. This response, initiated by liver-resident cells through the adaptor molecule for cytosolic RNA sensors, Mavs, and the transcription factors Irf3 and Irf7, is propagated by hepatocytes in an interferon-α/β receptor-dependent manner. This signaling pathway is critical for immune cell-mediated host resistance to liver-stage Plasmodium infection, which we find can be primed with other PAMPs, including hepatitis C virus RNA. Together, our results show that the liver has sensor mechanisms for Plasmodium that mediate a functional antiparasite response driven by type I IFN.
Article
The exoerythrocytic stage of Plasmodium falciparum has remained a difficult phase of the parasite life-cycle to study. The host and tissue specificity of the parasite requires the experimental infection of humans or non-human primates and subsequent surgical recovery of parasite-infected liver tissue to analyze this stage of the parasites development. This type of study is impossible in humans due to obvious ethical considerations and the cost and complexity in working with primate models has precluded their use for extensive studies of the exoerythrocytic stage. In this study we assessed, for the first time, the use of transgenic, chimeric mice containing functioning human hepatocytes as an alternative for modeling the in vivo interaction of P. falciparum parasites and human hepatocytes. Infection of these mice with P. falciparum sporozoites produced morphologically and antigenically mature liver stage schizonts containing merozoites capable of invading human red blood cells. Additionally, using microdissection, highly enriched P. falciparum liver stage parasites essentially free of hepatocyte contamination, were recovered for molecular studies. Our results establish a stable murine model for P. falciparum that will have a wide utility for assessing the biology of the parasite, potential anti-malarial chemotherapeutic agents and vaccine design. (c) 2005 Australian Society for Parasitology Inc. Published by Elsevier Ltd. All rights reserved.
Article
Plasmodium berghei infection was more severe in pregnant than in nonpregnant mice. Infection initiated on gestation day 7 resulted in rapidly increasing parasitemia and deaths of all pregnant mice within 12 days, while some nonpregnant mice survived until day 21 postinfection. When mice were infected on gestation day 12 or 14, a proportion of mice died before parturition; but some animals survived to deliver living pups. Reduced birthweights and increased spleen weight to body weight ratios were seen in pups from infected mice as compared with pups from uninfected animals. Histopathological abnormalities of placentae from infected animals included degeneration of the normal labyrinthine architecture and thickening of the trophobast separating maternal and fetal blood vessels.