Skip to main content
Erschienen in: Malaria Journal 1/2018

Open Access 01.12.2018 | Research

A novel immortalized hepatocyte-like cell line (imHC) supports in vitro liver stage development of the human malarial parasite Plasmodium vivax

verfasst von: Yongyut Pewkliang, Siriwan Rungin, Kaewta Lerdpanyangam, Apisak Duangmanee, Phongthon Kanjanasirirat, Phichaya Suthivanich, Khanit Sa-ngiamsuntorn, Suparerk Borwornpinyo, Jetsumon Sattabongkot, Rapatbhorn Patrapuvich, Suradej Hongeng

Erschienen in: Malaria Journal | Ausgabe 1/2018

Abstract

Background

Eradication of malaria is difficult because of the ability of hypnozoite, the dormant liver-stage form of Plasmodium vivax, to cause relapse in patients. Research efforts to better understand the biology of P. vivax hypnozoite and design relapse prevention strategies have been hampered by the lack of a robust and reliable model for in vitro culture of liver-stage parasites. Although the HC-04 hepatoma cell line is used for culturing liver-stage forms of Plasmodium, these cells proliferate unrestrictedly and detach from the culture dish after several days, which limits their usefulness in a long-term hypnozoite assay.

Methods

A novel immortalized hepatocyte-like cell line (imHC) was evaluated for the capability to support P. vivax sporozoite infection. First, expression of basic hepatocyte markers and all major malaria sporozoite-associated host receptors in imHC was investigated. Next, in vitro hepatocyte infectivity and intracellular development of sporozoites in imHC were determined using an indirect immunofluorescence assay. Cytochrome P450 isotype activity was also measured to determine the ability of imHC to metabolize drugs. Finally, the anti-liver-stage agent primaquine was used to test this model for a drug sensitivity assay.

Results

imHCs maintained major hepatic functions and expressed the essential factors CD81, SR-BI and EphA2, which are required for host entry and development of the parasite in the liver. imHCs could be maintained long-term in a monolayer without overgrowth and thus served as a good, supportive substrate for the invasion and growth of P. vivax liver stages, including hypnozoites. The observed high drug metabolism activity and potent responses in liver-stage parasites to primaquine highlight the potential use of this imHC model for antimalarial drug screening.

Conclusions

imHCs, which maintain a hepatocyte phenotype and drug-metabolizing enzyme expression, constitute an alternative host for in vitro Plasmodium liver-stage studies, particularly those addressing the biology of P. vivax hypnozoite. They potentially offer a novel, robust model for screening drugs against liver-stage parasites.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12936-018-2198-4) contains supplementary material, which is available to authorized users.
Abkürzungen
BSA
bovine serum albumin
CYP450s
cytochrome P450 isotypes
DAPI
4′,6-diamidino-2-phenylindole
EEs
exoerythrocytic stages
FBS
fetal bovine serum
IC50
50% inhibitory concentration
iHLCs
induced pluripotent stem cells-derived hepatocyte-like cells
MEM
minimal essential medium
MSCs
mesenchymal stem cells
PBS
phosphate-buffered saline
PQ
primaquine
PVM
parasitophorous vacuole membrane

Background

Malaria remains a major health problem worldwide, with an estimated 212 million disease cases and hundreds of thousands of related deaths in 2015 alone [1]. Malarial parasites are transmitted to humans via bites of infected female Anopheles mosquitoes; specifically, these parasites reside in the sporozoite form in mosquito salivary glands and are injected into human skin. The sporozoites rapidly leave the injection site and migrate through the bloodstream to the liver, where they invade hepatocytes and develop into liver-stage schizonts. A mature schizont contains thousands of daughter forms, termed merozoites, which are eventually released into the bloodstream where they invade erythrocytes and trigger the clinical symptoms of the disease. Of the five Plasmodium species that infect humans, Plasmodium falciparum is thought to cause the majority deaths, of which most cases (70%) involve children aged < 5 years in sub-Saharan Africa [1]. Plasmodium vivax is the most widespread Plasmodium species outside Africa. This organism is recognized as a major obstacle in malaria eradication campaigns because it can hide in a form called a hypnozoite [2, 3] in the human liver, and can reactivate weeks, months or years after the primary infection to cause a relapse [48].
The Plasmodium liver stage has become an attractive target for the development of anti-malarial drugs and vaccines because it is the precursor to blood-stage disease and also serves as a reservoir of the hypnozoites [9]. To date, only one approved drug, primaquine (PQ), has been shown to effectively eliminate liver-stage parasites, including P. vivax hypnozoites [10, 11]; however, this drug induces haemolytic toxicity in glucose-6-phosphate dehydrogenase-deficient individuals [12] and drug tolerance and therapeutic failures have compromised its clinical use [1316]. Avoidance of these toxic effects have been considered in the development of an alternative drug, tafenoquine (TQ) [17, 18], which targets hypnozoites and is currently in Phase III clinical trials [19]. However, the development of more safe and efficacious anti-liver stage drugs, particularly those that target hypnozoites, remains essential [20].
The lack of a robust, reliable in vitro model of the Plasmodium liver stage has limited understanding of the biology of Plasmodium liver stage and consequently, hampered development of drug and discovery programmes [19]. Current in vitro models of human liver-stage parasites (mainly P. falciparum and P. vivax) use both hepatoma cell lines, such as HepG2-A16 and HC-04 [2124], and primary human hepatocytes [2528]. The major advantages of the hepatoma cell lines include reliability and reduced variability between infection batches. Nonetheless, these cells exhibit abnormal cell regulation and proliferation and thus do not accurately recapitulate the biology of a liver-stage infection. Primary human hepatocytes are considered an ideal model for liver-stage cultures because these are the natural hosts. Nevertheless, primary hepatocytes are rarely used because of their limited availability and gradual loss of hepatic functions over time under conventional culture conditions [29]. Several groups have attempted to establish models that could prolong functional phenotypes of primary hepatocytes in culture. The primary hepatocyte and human hepatoma HepaRG cell co-culture model has been shown to not only retain hepatic functions but also to help maintain the susceptibility of primary hepatocytes to P. falciparum infection [30]. Another advanced micropatterned co-culture (MPCC) model, in which primary hepatocytes are organized among supportive stromal cells (3T3-J2 fibroblast cells), has been shown to retain functional hepatocytes for up to 4–6 weeks [28, 3134] and thus, allow the establishment of P. falciparum and P. vivax liver stage forms [28]. Recently, a microfluidic bilayer device was developed to promote long-term stability of primary hepatocytes and offer another platform for human liver-stage culture in vitro [35].
A ‘continuous non-tumorous cell line’ in which hepatocyte phenotypes are maintained could be a necessary substitute for primary hepatocytes. Some alternative sources of human cells that mimic the phenotypes of hepatocytes have been developed. In recent advances, induced pluripotent stem cell-derived hepatocyte-like cells (iHLCs) have been shown to support Plasmodium liver stages [36]. Nevertheless, these iHLCs are functionally immature and their hepatic functions must be induced and extensively characterized before each use [37]. Moreover, iHLCs that have been induced to differentiate usually exhibit limited life spans. Accordingly, iHLCs do not seem to be a simple or robust option for an in vitro model.
A novel ‘immortalized’ hepatocyte-like cell line (imHC) derived from human bone marrow mesenchymal stem cells (hMSCs) has been established [38]. These imHCs maintain the production of hepatocyte-specific markers, including albumin (ALB), urea, glycogen, alpha-fetoprotein (AFP), tyrosine aminotransferase, hepatocyte nuclear factor-4-alpha (HNF-4ɑ), glucose-6-phosphate dehydrogenase and all major cytochrome P450 isotypes (CYP450s). In this report, the feasibility of imHCs as a model for establishing a malaria infection was demonstrated. imHCs support the growth of P. vivax liver stages. This study also highlights the potential use of imHCs as a model for drug screening applications.

Methods

Ethical approval

In this study, human blood was collected and patient samples were used in strict accordance with the human use protocol TMEC 11-033, approved by the Institute Ethical Review Committee of the Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand. Written informed consent was obtained from the patient for the publication of this report.

Plasmodium sporozoite preparation

Laboratory-reared female Anopheles dirus mosquitoes were maintained in a colony at the Mahidol Vivax Research Unit in Bangkok, Thailand. The mosquitoes were membrane-fed with blood samples collected from Plasmodium-infected patients in Kanchanaburi and Tak Provinces, Thailand. Briefly, 5 ml of patient blood was collected in a heparinized tube and centrifuged at 1500×g for 10 min to remove plasma. The resulting pellet was washed with 10 ml of phosphate-buffered saline (PBS) and reconstituted to the original volume with naïve-type AB serum for mosquito feeding [39]. Each feeder contained approximately 0.5 ml of infected blood and was used to feed approximately 100 5–7-day-old mosquitoes during a 30-min period. Mosquitoes were subsequently maintained on a 10% sucrose solution under a controlled environment at 26 °C and 75% humidity with a 12-h light/dark cycle. Midgut oocysts of P. vivax were monitored on dissected mosquitoes using a mercurochrome staining method on day 7 post feeding [40]. The salivary gland sporozoites were examined on day 14 post feeding and dissected from the infected mosquitoes using a standard protocol [41]. In brief, salivary glands of 50 infected mosquitoes were dissected, placed in 50 μl of ice-cold RPMI 1640 medium (Gibco, Grand Island, NY, USA), pH 8.2 [42], supplemented with 200 U/ml penicillin (Invitrogen, Carlsbad, CA, USA), 200 µg/ml streptomycin (Invitrogen), and 0.25 µg/ml amphotericin B (Invitrogen), and ground with a sterile pestle. The released sporozoites were counted in a hemocytometer and kept on ice until used, but for no more than 1 h to avoid a reduction in parasite infectivity (Patrapuvich R, unpublished data and [43]).

Hepatocyte culture

imHCs were cultured in 1:1 DMEM:Ham’s F12 media (Invitrogen) supplemented with 10% heat-inactivated fetal bovine serum (FBS) (Gibco), 100 U/ml penicillin, and 100 µg/ml streptomycin. The HC-04 hepatoma cell line (ATCC patent deposit no. PTA-3441: ATCC, Manassas, VA, USA) was cultured in 1:1 minimal essential medium (MEM):Ham’s F12 media (Invitrogen) supplemented with 10% heat-inactivated FBS, 100 U/ml penicillin, and 100 µg/ml streptomycin. Both cell lines were maintained at 37 °C in a humidified atmosphere containing 5% CO2. Cells were subcultured every 2–4 days or once they had reached approximately 80% confluence, after detachment with 0.125% trypsin–EDTA (Invitrogen).

Growth curve

Hepatocytes were seeded at a density of 2 × 104 cells per well in a six-well plate (Thermo Scientific™, Nunc™; Waltham, MA, USA) and maintained as described above for 2 weeks with daily changes in medium. Viable cells were monitored and counted daily using a hemocytometer and an Olympus IX71 inverted phase/fluorescence microscope (Olympus, Tokyo, Japan). Growth curves were generated using GraphPad Prism software, version 7.0 (GraphPad Inc, La Jolla, CA, USA).

In vitro liver-stage infection

Hepatocytes were seeded at a density of 3 × 105 cells per well in a Matrigel (Corning Corp, Corning, NY, USA)-coated Millicell EZ SLIDE eight-well glass slide (Millipore, Billerica, MA, USA) and maintained in complete medium (1:1 MEM:Ham F12 supplemented with 10% FBS, 100 U/ml penicillin, and 100 µg/ml streptomycin) at 37 °C. After incubation for 18–20 h, the medium was aspirated, and an aliquot of sporozoite suspension (3 × 105 in 200 µl of complete medium) was added to each well. After a 4-h inoculation at 37 °C, free sporozoites were removed by aspiration and a 400-µl aliquot of fresh infection medium (1:1 MEM:Ham F12 supplemented with 10% FBS, 100 U/ml of penicillin, 100 µg/ml of streptomycin, and 0.25 µg/ml amphotericin B) was added. The infected hepatocyte culture was maintained at 37 °C with daily changes in medium until the liver-stage parasites or exoerythocytic forms (EEs) were established and visualized using an indirect immunofluorescence assay. The total number of EEs in each well was manually quantified. Per cent sporozoite infectivity was determined by comparing the number of infected hepatocytes to the total number of inoculated sporozoites [42].

Indirect immunofluorescence assay

After removing the culture medium, the infected hepatocyte cells were washed with PBS buffer thrice, fixed with 4% paraformaldehyde for 20 min, and permeabilized by exposure to 0.1% Triton X-100 for 3–5 min at room temperature (20–25 °C). The cells were then treated with 3% BSA (in PBS) solution, followed by incubation with mouse primary monoclonal anti-PbHSP70 antibodies (clone 4C9, kindly provided by Dr. Fidel P. Zavala [44]) and rabbit primary polyclonal anti-PvUIS4 antibodies (1:500 dilution, kindly provided by Dr. Sebastian A. Mikolajczak [45]). Subsequently, goat secondary IgG Alexa Fluor® 488-conjugated anti-mouse antibodies (1:500 dilution, Invitrogen), goat secondary IgG Alexa Fluor® 568-conjugated anti-rabbit 2nd antibodies (1:500 dilution, Invitrogen) and 0.1 μg/ml 4′,6-diamidino-2-phenylindole (DAPI) (Invitrogen) were added to the cells. Incubation with primary and secondary antibodies was performed at 37 °C for 1 h and 30 min, respectively. In some experiments, rabbit anti-acetylated histone H3K9 antibodies (1:200 dilution, Millipore) and mouse anti-acyl carrier protein (ACP) antibodies (1:200 dilution, kindly provided by Dr. Sebastian A. Mikolajczak [45]) were used for detecting nuclear histones and apicoplasts of P. vivax EEs, respectively. Finally, samples were covered with ProLong Gold antifade reagent (Invitrogen), sealed under coverslips, and viewed under a fluorescent microscope (400× magnification; AXIO Scope. A1 equipped with AxioVision Rel 4.8 Software; Carl Zeiss AG, Oberkochen, Germany). Fluorescence images were acquired using an Olympus FluoView™ FV1000 confocal laser scanning microscope (Olympus, Tokyo, Japan) equipped with a 60× oil objective. Images were captured using FV10-ASW 3.0 viewer software and prepared for publication using Adobe Illustrator CC (Adobe Systems, San Jose, CA, USA).
In some experiments, uninfected hepatocyte cells were grown in 96-well CellCarrier-96 optic black plates (PerkinElmer, Waltham, MA, USA) and stained with antibodies against the following hepatocyte markers: ALB (1:100 dilution, ab10241, Abcam, Cambridge, UK), AFP (1:100 dilution, SC8399, Santa Cruz Biotechnology, Dallas, TX, USA), low-density lipoprotein receptor (LDLR; 1:100 dilution, SC373830, Santa Cruz Biotechnology), Na+-taurocholate cotransporting polypeptide (NTCP; 1:100 dilution, ab131084, Abcam, Cambridge, UK), and HNF-4ɑ (1:100 dilution, SC6556, Santa Cruz Biotechnology). Cells were also stained with antibodies against the following cell surface receptors: CD81 (1:100 dilution, ab79559, Abcam), scavenger receptor type B class I (SR-BI) (1:100 dilution, NB400-104, Novus Biologicals, Littleton, CO, USA), and EphA2 (1:100 dilution, 37-4400, Thermo Fisher Scientific). Samples were then incubated with a goat anti-mouse Alexa Fluor® 488-conjugated (1:500 dilution, Invitrogen), goat anti-rabbit Alexa Fluor® 488-conjugated (1:500 dilution, Invitrogen), or donkey anti-goat Cy3-conjugated secondary antibody (1:500 dilution, BioLegend, San Diego, CA, USA), as appropriate; hepatocyte nuclei were stained with 2 µM Hoechst 33342 (Thermo Fisher Scientific). Mouse IgG2a, mouse IgG1, rabbit IgG and goat IgG were used as negative control for staining. Fluorescence images were captured and analysed using an Operetta High-Content Imaging System (PerkinElmer) with a 40× objective lens.

Primaquine treatment

PQ (Sigma-Aldrich, St Louis, MO, USA) was added to infected hepatocytes at concentrations ranging from 0.1 to 10 μM 2 h post sporozoite infection. The cultures were maintained for up to 5 days with daily changes in medium (including drug supplementation) and harvested on day 6 to determine sporozoite infectivity by immunofluorescence assay, as described above. Numbers and sizes of EEs were manually examined using a fluorescent microscope (ZEISS AXIO Scope.A1). The relative proportions of small EEs (diameter ≤ 5 µm) and schizonts (diameter > 5 µm) were determined relative to the 0.1% dimethyl sulfoxide untreated controls. Plasmodium vivax assays were conducted over three independent experiments using three batches of sporozoites generated from different P. vivax isolates.

Quantitative real-time PCR

Total RNA was extracted from the hepatocytes using a RNeasy Plus Mini Kit (Qiagen, Hilden, Germany). The isolated total RNA was then treated with DNase I (Thermo Scientific, USA), according to the manufacturer’s instructions. The quantity and quality of total RNA were determined using a NanoDrop spectrophotometer (Thermo Scientific). For the CYP450 qRT-PCR analysis, 500 ng of total RNA was converted to cDNA using the SuperScript® III First-Strand Synthesis System (Invitrogen). The primer sets and conditions used for CYP450 amplification have been described previously [38]. Briefly, 2 μl of cDNA was diluted in a 10-μl reaction mixture containing 0.4 μM of each primer and 5 μl of Luminaris Color HiGreen qPCR Master Mix (Thermo Scientific, USA). Each reaction was run in a CFX96 Touch™ Real-Time PCR Detector (Bio-Rad, Hercules, CA, USA) with the following conditions: 50 °C for 2 min; 95 °C for 10 min; and 40 cycles of amplification at 95 °C for 15 s and 60 °C for 45 s. Reverse transcriptase- and template-negative controls were used for each gene-specific primer pair. The number of cycles required for the fluorescent signal to cross the threshold (Ct value) was determined for each CYP450 isotype. Obtained Ct values were subtracted from Ct of the respective house-keeping gene (GAPDH) obtained from the same cells to generate a relative gene expression value ΔCt. In some experiments, hepatocytes were treated with PQ for 6 days with daily changes in medium prior to RNA extraction.

Statistical analysis

At least three independent experiments were conducted in triplicate. Each individual sporozoite infection experiment was performed using a different parasite isolate. Differences in the results were determined using a standard Student’s t test, and a p value of < 0.05 was considered to indicate statistical significance. For non-normally distributed data, the Mann–Whitney U test was used in place of the paired t test.

Results

imHCs expressed hepatic phenotypes

hMSCs, precursors of imHCs, were immortalized via lentiviral transduction with the human telomerase reverse transcriptase gene (hTERT) and Bmi-1 [46], according to previously described protocol [38]. After induction of differentiation, resulting imHCs were examined for hepatocyte phenotype and metabolic function maintenance, and for continuous cell line property using population doubling level (PDL) [38]. imHCs exhibited a typical homogenous hepatocyte morphology, polygonal shape, and cord-like structure (Fig. 1a), and expressed basic hepatocyte-specific markers, including ALB, AFP, HNF-4ɑ, LDLR, and NTCP, as demonstrated by immunofluorescence staining (Fig. 1b–f, respectively). As shown in Fig. 1g, more than 80% of the population of imHCs contained ALB, AFP, HNF-4ɑ, and NTCP and approximately 42% of the cells appeared positive for LDLR. The expression level of LDLR in imHCs was comparable with that observed in human hepatocyte cell line HepG2 [47]. The expression of HNF-4ɑ, a hepatic maturation marker, indicated that imHCs had fully matured into hepatocytes. This result was consistent with the previous study, which also showed that the expression levels of the hepatic marker in imHCs were comparable to those in primary hepatocytes [38].

imHCs expressed essential receptors for malaria infection

To investigate the potential use of imHCs in malaria infection studies, the expression of essential molecules required for Plasmodium liver-stage infection on the putative host cell surfaces was examined. imHCs expressed all major malaria sporozoite-associated receptors known to be essential for parasite entry and intracellular development in the liver, including TAPA-1 (CD81) [48] (Fig. 2a), EphA2 [49] (Fig. 2b) and SR-BI [50] (Fig. 2c). CD81 and EphA2 were expressed at higher levels in imHCs than in HC-04 cells, a cell line most commonly used for malaria sporozoite infection (Fig. 2d, e). The numbers of CD81- and EphA2-positive cells were significant higher in imHCs than in HC-04 (Fig. 2g, h; p < 0.0001 for both cases). HC-04 cells exhibited clear inter-cellular variations in levels of EphA2, similar to the findings reported from hepatoma Hepa1-6 cultures [49]. Both imHCs and HC-04 cells expressed SR-BI at similar levels, with 87 ± 2.5% and 95 ± 1.4% of cells, respectively (Fig. 2i), although the protein localization differed slightly between the cells (Fig. 2c, f). Given these findings, imHCs could possibly be infected by Plasmodium malarial parasites and may be superior to the classical host (HC-04 cells).

imHCs were suitable for long-term culture

imHCs grew more slowly than HC-04 cells, as demonstrated by their doubling times (Fig. 3a). The mean doubling time of imHCs was 45 h versus 40 h of HC-04 cells (Fig. 3a). The batch of HC-04 cells used in this study had a similar growth rate to the reported value, with a doubling time of approximately 37 h [23]. HC-04 cells were round in shape and tended to aggregate and clump together (Fig. 3b). While a reduction in HC-04 cell growth once the culture reached confluency was observed, HC-04 cells exhibited non-stop growth and a loss of cell–cell contact inhibition and formed a multi-layer arrangement of cells (Fig. 3b) that gradually detached from the culture plate after 3 weeks of culture. In contrast, imHCs displayed an epithelial-like morphology and clearly exhibited contact inhibition after reaching confluence (Fig. 3b). This property allowed imHC cells to remain in a monolayer without overgrowth for up to 6 weeks of culture (Additional file 1: Figure S1). This finding suggested that imHCs may be suitable for long-term culture of P. vivax hypnozoites. In addition, the flat imHC monolayer may not only help in prolonging the culture but also in improving the ability to visualize and detect liver-stage parasites using immunofluorescence microscopy.

imHCs supported Plasmodium vivax liver-stage infection in vitro

Next, the feasibility of using imHCs for P. vivax infection was examined. First, imHCs were infected with P. vivax sporozoites and EEs were determined by immunostaining with an antibody specific for Plasmodium HSP70. EEs were evaluated on day 4 post sporozoite infection because this is the earliest day when the developing schizonts and small EEs of P. vivax could be differentiated. Consistent with the hypothesis, imHCs could be infected by P. vivax sporozoites. The total number of P. vivax EEs obtained from 3 × 105 inoculated sporozoites was in the range of 30–1000, and this number did not significantly differ from the number observed in the HC-04 culture (p > 0.05). Infection efficiencies of the sporozoites were 0.14 ± 0.16% and 0.15 ± 0.19% in imHCs and HC-04 cells, respectively (Fig. 4).
To investigate whether imHCs could fully support further development of P. vivax sporozoites, the rate at which parasites developed into schizont forms (diameter > 10 μm) between days 4 and 7 post infection was monitored. Although imHCs and HC-04 cells were infected by different batches of sporozoites, similar patterns of development of EEs in these cells were observed (Fig. 5a, b). All EEs obtained during the 4-day period measured < 15 μm in diameter; however, on day 7, approximately 40% of EEs had progressed to the schizont stage (> 10 μm in diameter) of the liver-stage life cycle, with approximate maximum sizes of 40–45 μm (Fig. 5a, b). However, imHCs allow a better Plasmodium liver stage than HC-04 cells because larger EEs were found in the former (30–45 μm) relative to the latter (20–25 μm). Representative immunofluorescence images of P. vivax EEs on days 4 and 7 post sporozoite infection are shown in Fig. 5c and Additional file 2: Figure S2. The normal growth of P. vivax EEs in the imHC model was confirmed by the expression of parasitophorous vacuole membrane (PVM)-localized PvUIS4, a known liver-stage-specific marker [45]. Importantly, day 4 EEs exhibited a PvUIS4-positive PVM feature known as a ‘prominence’, which has been reported as a potential indicator of hypnozoites [45] (Fig. 5c, upper panel). Furthermore, small forms (< 10 μm) were also observed in imHCs after day 7 post-infection (Fig. 5a, b and Additional file 2: Figure S2).
Then, the maturation of P. vivax EEs in an imHC culture was monitored. EEs increased in size from an average of 5 μm on day 4 to a wide range of sizes up to 50 μm by day 10 post-infection (Fig. 6a, b and Additional file 3: Figure S3). The gradual decline in the numbers of EEs after day 10 was possibly attributable to the rupture of some mature schizonts and release of merozoites, indicating the completion of its life cycle in the liver (Fig. 6a). Asynchronous development is a normal characteristic of in vitro EE culture [23, 51] and was clearly observed in P. vivax-infected imHCs after day 7 (Fig. 6a). Representative images of day 10 EEs and asynchronous features of EEs, which ranged in size from < 10 μm to up to 50 μm, are shown in Fig. 6b. In imHCs, P. vivax EEs also displayed another typical liver-stage-specific marker, apicoplast-localized ACP [45], during a 10-day infection period, thus confirming the normal development of P. vivax in these cells (Fig. 6b). The presence of acetylated lysine 9 of histone H3-positive small EEs on day 14 post-infection (Fig. 6c) might indicate the existence of the hypnozoites in the culture; however, these small EEs in imHCs require further characterization. Moreover, two variants of Thai P. vivax sporozoites with the genotypes CSP-VK210 and CSP-VK247 were used in this study, and imHCs were found to be susceptible to infection by both variants. On day 7 post infection, the CSP-VK210 and CSP-VK247 variants yielded a small form frequency of approximately 36 and 77%, respectively.

Plasmodium-infected imHCs were sensitive to PQ treatment

Successful action of a drug such as primaquine required activation by oxidative enzymes [52, 53]. To determine whether imHCs could be used for anti-liver-stage drug screening, the expression of CYP450s in these cells was examined. imHCs had a basal expression of eight major CYP450 isotypes (CYP1A1, CYP1A2, CYP2B6, CYP2C9, CYP2C19, CYP2D6, CYP2E1, CYP3A4), as determined by RT-PCR (Fig. 7a). CYP1A1, CYP2C9, CYP2D6, CYP2E1 and CYP3A4 were expressed at relatively higher levels than CYP1A2, CYP2B6 and CYP2C19 (Fig. 7a). In contrast, only CYP1A1, CYP2D6 and CYP2E1 were expressed in HC-04 cells, and the expression levels were relatively lower than those in imHCs (Fig. 7b).
Even higher expression of six CYP450 isotypes (CYP1A1, CYP1A2, CYP2B6, CYP2C9, CYP2D6, CYP2E1) was induced in imHCs after PQ treatment (Fig. 7a). PQ easily induced the expression of CYP2C9, CYP2D6 and CYP2E1 in a dose–response manner. The expression of CYP1A1, CYP2C9, CYP2D6 and CYP2E1 were significantly upregulated after 10 μM treatment, compared to the untreated controls (p < 0.05 in all cases). No significant upregulation was observed in the expression of CYP1A2, CYP2B6, CYP2C19, and CYP3A4 in response to PQ. Among the three CYP450 isotypes in HC-04 cells, the expression of only CYP1A1 was induced by PQ treatment (Fig. 7b). Altogether, imHCs would serve as the site of PQ bioactivation and anti-liver-stage drug activity would be more potent in these cells than in HC-04 cells.
To test PQ sensitivity, hepatocytes were infected with P. vivax sporozoites 2 h prior to PQ exposure for a total of 6 days. Plasmodium vivax-infected imHCs were sensitive to PQ (Fig. 7c), whereas very low responses to PQ were observed in parasite-infected HC-04 cells (Fig. 7d). In imHCs, PQ activity was detected against schizonts and small forms (presumably hypnozoites), with an IC50 value < 0.1 μM for both forms (Fig. 7c). Plasmodium vivax-infected HC-04 cells were less responsive to PQ (IC50: approximately 10 μM) compared with their imHC counterparts (Fig. 7d). In the liver, PQ is known to produce active metabolites via bioactivation [53]. Therefore, the potent activity of PQ against P. vivax parasites in imHCs may be attributed to the higher level of CYP450s in the imHC culture.

Discussion

Urgent strategies aimed at achieving the current worldwide goal of eradication of malaria have included approaches for discovering drugs that target the Plasmodium liver stage, particularly dormant hypnozoites [20]. To date, however, biological features of P. vivax hypnozoites remain largely unknown, mainly because of the difficulty of producing large amounts of P. vivax sporozoites [42] and the lack of a robust and reliable culture system for this purpose [19]. Currently, humanized liver mouse models appear to be a powerful tool for obtaining human liver-stage parasites in vivo [45, 5456]. In the context of drug discovery, the cost and technical challenges associated with this animal model render it largely infeasible for early-stage drug screening, although it remains valuable during preclinical testing [56]. Although in vitro primary human hepatocyte models have advanced the ability to identify potential molecules for liver-stage targeting [28, 30, 34], the loss of hepatic functions over time and donor-to-donor variability [28] remains a major challenge when using primary hepatocytes for in vitro assays. Therefore, most studies on liver-stage parasites heavily rely on human hepatoma cell lines as hosts for sporozoite infection [5759]. However, these hepatoma cells may not be suitable for assessment of drug metabolism and drug interactions because they usually lack various functional CYP450s and other phase I, II, and III drug-metabolizing enzymes. HC-04 cells tend to overgrow in culture, leading to the detachment of host cells, and thus, limiting long-term monitoring of EEs and hindering the establishment of hypnozoites. The reduction in HC-04 cell growth upon reaching confluency might also affect the development of EEs. In addition, infection rates among HC-04 cells alone are relatively low [23] but, as in the present study, a Matrigel co-culture improved this parameter [30]. Infection rates were 4–6 times higher in the Matrgel-HC-04 co-culture than in the original untreated HC-04 model (Patrapuvich R, unpublished data).
To overcome the noted deficiencies of HC-04 cells with respect to supporting liver-stage culture of malaria parasites, a novel hepatocyte cell line, imHC, was therefore tested the ability to support the development of P. vivax liver-stage.
imHCs displayed essential liver functions comparable with those of mature hepatocytes [38], ceased proliferation upon reaching a high cell density, and remained in a monolayer even after months of culture. In addition, imHCs expressed the host factors CD81 [48, 60], EphA2 [49] and SR-BI [50], which may be important for sporozoite invasion. No significant difference in infectivity between imHCs (CD81++) and HC-04 (CD81+) cells was observed, suggesting that CD81 was not the primary determinant of P. vivax infection. This observation is consistent with the recent study from Manzoni et al. showing that antibodies against SR-BI but not against CD81 inhibit P. vivax infection of primary human hepatocyte cultures [61]. The results affirm the potential role of SR-BI during hepatocyte infection by P. vivax sporozoites. Manzoni et al. also reported that P. falciparum, another major human malarial parasite, relies on CD81 but not SR-BI for host entry [61]. Given these data, it will be interesting to examine whether imHCs can better support P. falciparum infection than HC-04 cells. The same study also showed that parasite protein P36 is essential for both SR-BI- and CD81-dependent sporozoite invasion [61]. No direct interaction between P36-dependent invasion and EphA2 has been demonstrated [49, 61, 62]. The role of EphA2 in P. vivax infection remains to be further investigated.
In the present study, P. vivax infection rate in imHCs (0.14%) was comparable or even higher than that observed in HC-04 cells [23] and in a primary hepatocyte MPCC model [28]. Nevertheless, these infection rates were relatively low in comparison to those observed in in vivo [63, 64]. No significant difference in the development rates of P. vivax EEs in imHCs versus those in HC-04 cells based on the percentages of EEs that developed into schizonts (> 10 μm) was observed. However, on day 7, there were many larger EEs among the schizonts growing in imHCs (> 20–45 μm); whereas most EEs in HC-04 cells were smaller (approximately 20–25 μm). The sizes of these day-7 P. vivax schizonts in imHCs were similar to those recently reported in MPCC on day 6 (approximately 30 μm) [28] and in a humanized mice model (approximately 40–50 μm) [45]. Larger mature EEs were clearly visible on day 10 (up to approximately 50 μm), but remained smaller than those (60–80 μm) observed in vivo [45]. In imHCs, antibodies specific for PVMs and apicoplasts of EEs revealed a complex cellular structure similar to that observed in vivo [45]. In this study, while MSP-1 expression was not used as a late liver-stage marker for determining parasite maturation, the presence of large multi-nuclei parasites on day 10 confirmed the establishment of mature P. vivax schizonts in the imHC culture. Importantly, the long-term culture of P. vivax-infected imHCs yielded a population of small (< 10 μm in diameter) forms, presumably hypnozoites, that maintained a single-nuclear structure as detected by histone-acetylation staining of the parasite nuclei [45]. This finding demonstrates the capacity of imHCs to establish P. vivax hypnozoites. These presumed hypnozoites in imHCs will continue to require further characterization. It was very interesting to note that a small form observed on day 14 did not exhibit the ‘UIS4-positive prominence’, previously considered as a unique feature of P. vivax hypnozoites described by Mikolajczak et al. [45]. In contrast, the young parasite showed on day 4 clearly displayed the prominence pattern. The reliability of this marker as a hypnozoite-specific trait should be considered with care. Here, CSP-VK247 sporozoites produced larger numbers of small forms (77% of EEs) than those of the CSP-VK210 genotype (36% of EEs). This result is in agreement with the recent report in humanized mice model showing that a greater proportion of CSP-VK247 sporozoites (~ 40% of EEs) form hypnozoites compared with CSP-VK210 sporozoites (~ 5% of EEs) [45]. The higher percentage of small EEs obtained in the cultured imHCs compared to infections in in vivo mice may reflect the presence of a sub-population of slow-growing parasites in culture.
The responses of malaria parasites in imHCs to PQ, a drug known to target Plasmodium liver stages, were tested to demonstrate the potential use of imHCs as a platform for drug screening. imHCs, which exhibit a cellular physiology closer to that of human hepatocytes compared to HC-04 cells (including CYP450 drug metabolic activity), provide a physiologically relevant platform for drug screening and, thus, would be expected to catch agents that cause drug-induced liver injury [65], particularly during the initial stage of drug discovery. PQ more potently affected EEs in imHCs than in HC-04 cells. Increased PQ sensitivity was associated with higher levels of CYP450 activity in imHCs. CYP2C19, CYP3A4 and CYP2D6 have been identified as the three major enzymes involved in PQ metabolism [53]. The observation that CYP2C19 and CYP3A4 were not induced in imHCs after PQ exposure suggests the essentiality of CYP2D6 in PQ metabolism [52, 66].
Taken together, the findings suggest that imHCs could be an improved hepatoma cell line for study P. vivax liver stage parasites. This finding is expected to lead to an array of imHC applications that will facilitate understanding the biology of P. vivax hypnozoites and help discover anti-relapse drug, particularly in high-throughput screening formats.

Conclusions

Previous in vitro cultures of liver-stage malarial parasites have relied on hepatocyte cell lines, particularly the HC-04 line. In short experiments, HC-04 cells support the growth of human malarial parasites P. falciparum and P. vivax. However, HC-04 cell proliferation is not inhibited by cell–cell contact and host cells detach after a long period. HC-04 cells are not well suitable for long-term hypnozoite cultures that need to study the biology of hypnozoites, identification of putative regulators of parasite dormancy, and drug screening. imHCs, which comprise an immortalized human hepatocyte cell line generated from mesenchymal stem cells, offer an alternative host platform for the culture of human malarial liver stages in vitro. imHCs are superior to HC-04 cells for long-term culture because the former can be maintained for months without overgrowth and detachment, thereby facilitating the development of an in vitro hypnozoite system. imHCs also retain the functions of CYP450s enzymes responsible for drug metabolism and provide a more physiologically relevant model for drug testing. Overall, imHCs potentially serve as a new, robust system for analyzing human P. vivax liver-stage parasites, particularly hypnozoites. The resulting models will facilitate a better understanding of the biology of parasites and promote discovery of drugs and vaccines against the liver stage.

Authors’ contributions

SH provided imHCs. JS provided P. vivax sporozoites. AD prepared and dissected the mosquitoes. YP, SR and KL maintained hepatocyte cultures, prepared sporozoites, and conducted the sporozoite infection experiments. YP, RP, SR and KL performed the immunofluorescence assay and identified liver-stage parasites. YP, PS and PK participated in fluorescence imaging and analysis. KS evaluated the expression of CYP450s. YP, RP and SB conceived the study, analysed data, and drafted the manuscript. JS and SH supervised the research. All authors read and approved the final manuscript.

Acknowledgements

The authors thank the study participants and field team staff at the Mahidol Vivax Research Unit (MVRU) for their assistance with patient blood collection. Dr. Sebastian A. Mikolajczak (Center for Infectious Disease Research, Seattle, WA, USA) provided antibodies against P. vivax EEs. Dr. Fidel P. Zavala (Johns Hopkins University, Baltimore, MD, USA) provided anti-PbHSP70 antibodies. RP is supported by the Office of the Higher Education Commission and Mahidol University under the National Research Universities Initiative. KS is supported by Mahidol University and the Thailand Research Fund (TRG5880087). SB is supported by the Research Chair Grant of the National Science and Technology Development Agency (NSTDA), Thailand. SB and SH are also supported by the research support grant of the Excellent Center for Drug Discovery (ECDD). The ECDD is supported by the Faculty of Science and Faculty of Medicine Ramathibodi Hospital, Mahidol University and the Thailand Centre of Excellence for Life Sciences (TCELS).

Competing interests

The authors declare that they have no competing interests.

Availability of data and materials

All data generated during this study are included in this published article and the additional files.
In this study, human blood was collected and patient samples were used in strict accordance with the human use protocol TMEC 11-033, approved by the Institute Ethical Review Committee of the Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand. Written informed consent was obtained from the patient for the publication of this report.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Literatur
1.
Zurück zum Zitat WHO. World malaria report 2016. Geneva: World Health Organization; 2016. WHO. World malaria report 2016. Geneva: World Health Organization; 2016.
3.
Zurück zum Zitat Krotoski WA, Collins WE, Bray RS, Garnham PC, Cogswell FB, Gwadz RW, et al. Demonstration of hypnozoites in sporozoite-transmitted Plasmodium vivax infection. Am J Trop Med Hyg. 1982;31:1291–3.CrossRefPubMed Krotoski WA, Collins WE, Bray RS, Garnham PC, Cogswell FB, Gwadz RW, et al. Demonstration of hypnozoites in sporozoite-transmitted Plasmodium vivax infection. Am J Trop Med Hyg. 1982;31:1291–3.CrossRefPubMed
6.
Zurück zum Zitat Dao NV, Cuong BT, Ngoa ND, le Thuy TT, The ND, Duy DN, et al. Vivax malaria: preliminary observations following a shorter course of treatment with artesunate plus primaquine. Trans R Soc Trop Med Hyg. 2007;101:534–9.CrossRefPubMed Dao NV, Cuong BT, Ngoa ND, le Thuy TT, The ND, Duy DN, et al. Vivax malaria: preliminary observations following a shorter course of treatment with artesunate plus primaquine. Trans R Soc Trop Med Hyg. 2007;101:534–9.CrossRefPubMed
7.
Zurück zum Zitat Durante Mangoni E, Severini C, Menegon M, Romi R, Ruggiero G, Majori G. Case report: an unusual late relapse of Plasmodium vivax malaria. Am J Trop Med Hyg. 2003;68:159–60.PubMed Durante Mangoni E, Severini C, Menegon M, Romi R, Ruggiero G, Majori G. Case report: an unusual late relapse of Plasmodium vivax malaria. Am J Trop Med Hyg. 2003;68:159–60.PubMed
8.
Zurück zum Zitat Garnham PC, Bray RS, Bruce-Chwatt LJ, Draper CC, Killick-Kendrick R, Sergiev PG, et al. A strain of Plasmodium vivax characterized by prolonged incubation: morphological and biological characteristics. Bull World Health Organ. 1975;52:21–32.PubMedPubMedCentral Garnham PC, Bray RS, Bruce-Chwatt LJ, Draper CC, Killick-Kendrick R, Sergiev PG, et al. A strain of Plasmodium vivax characterized by prolonged incubation: morphological and biological characteristics. Bull World Health Organ. 1975;52:21–32.PubMedPubMedCentral
9.
Zurück zum Zitat Mazier D, Renia L, Snounou G. A pre-emptive strike against malaria’s stealthy hepatic forms. Nat Rev Drug Discov. 2009;8:854–64.CrossRefPubMed Mazier D, Renia L, Snounou G. A pre-emptive strike against malaria’s stealthy hepatic forms. Nat Rev Drug Discov. 2009;8:854–64.CrossRefPubMed
10.
12.
Zurück zum Zitat Alving AS, Carson PE, Flanagan CL, Ickes CE. Enzymatic deficiency in primaquine-sensitive erythrocytes. Science. 1956;124:484–5.PubMed Alving AS, Carson PE, Flanagan CL, Ickes CE. Enzymatic deficiency in primaquine-sensitive erythrocytes. Science. 1956;124:484–5.PubMed
13.
Zurück zum Zitat Chiang TY, Lin WC, Kuo MC, Ji DD, Fang CT. Relapse of imported vivax malaria despite standard-dose primaquine therapy: an investigation with molecular genotyping analyses. Clin Microbiol Infect. 2012;18:E232–4.CrossRefPubMed Chiang TY, Lin WC, Kuo MC, Ji DD, Fang CT. Relapse of imported vivax malaria despite standard-dose primaquine therapy: an investigation with molecular genotyping analyses. Clin Microbiol Infect. 2012;18:E232–4.CrossRefPubMed
14.
Zurück zum Zitat Townell N, Looke D, McDougall D, McCarthy JS. Relapse of imported Plasmodium vivax malaria is related to primaquine dose: a retrospective study. Malar J. 2012;11:214.CrossRefPubMedPubMedCentral Townell N, Looke D, McDougall D, McCarthy JS. Relapse of imported Plasmodium vivax malaria is related to primaquine dose: a retrospective study. Malar J. 2012;11:214.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Bright AT, Alenazi T, Shokoples S, Tarning J, Paganotti GM, White NJ, et al. Genetic analysis of primaquine tolerance in a patient with relapsing vivax malaria. Emerg Infect Dis. 2013;19:802–5.CrossRefPubMedPubMedCentral Bright AT, Alenazi T, Shokoples S, Tarning J, Paganotti GM, White NJ, et al. Genetic analysis of primaquine tolerance in a patient with relapsing vivax malaria. Emerg Infect Dis. 2013;19:802–5.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Thomas D, Tazerouni H, Sundararaj KG, Cooper JC. Therapeutic failure of primaquine and need for new medicines in radical cure of Plasmodium vivax. Acta Trop. 2016;160:35–8.CrossRefPubMed Thomas D, Tazerouni H, Sundararaj KG, Cooper JC. Therapeutic failure of primaquine and need for new medicines in radical cure of Plasmodium vivax. Acta Trop. 2016;160:35–8.CrossRefPubMed
17.
Zurück zum Zitat Llanos-Cuentas A, Lacerda MV, Rueangweerayut R, Krudsood S, Gupta SK, Kochar SK, et al. Tafenoquine plus chloroquine for the treatment and relapse prevention of Plasmodium vivax malaria (DETECTIVE): a multicentre, double-blind, randomised, phase 2b dose-selection study. Lancet. 2014;383:1049–58.CrossRefPubMed Llanos-Cuentas A, Lacerda MV, Rueangweerayut R, Krudsood S, Gupta SK, Kochar SK, et al. Tafenoquine plus chloroquine for the treatment and relapse prevention of Plasmodium vivax malaria (DETECTIVE): a multicentre, double-blind, randomised, phase 2b dose-selection study. Lancet. 2014;383:1049–58.CrossRefPubMed
18.
Zurück zum Zitat Marcsisin SR, Sousa JC, Reichard GA, Caridha D, Zeng Q, Roncal N, et al. Tafenoquine and NPC-1161B require CYP 2D metabolism for anti-malarial activity: implications for the 8-aminoquinoline class of anti-malarial compounds. Malar J. 2014;13:2.CrossRefPubMedPubMedCentral Marcsisin SR, Sousa JC, Reichard GA, Caridha D, Zeng Q, Roncal N, et al. Tafenoquine and NPC-1161B require CYP 2D metabolism for anti-malarial activity: implications for the 8-aminoquinoline class of anti-malarial compounds. Malar J. 2014;13:2.CrossRefPubMedPubMedCentral
19.
Zurück zum Zitat Campo B, Vandal O, Wesche DL, Burrows JN. Killing the hypnozoite–drug discovery approaches to prevent relapse in Plasmodium vivax. Pathog Glob Health. 2015;109:107–22.CrossRefPubMedPubMedCentral Campo B, Vandal O, Wesche DL, Burrows JN. Killing the hypnozoite–drug discovery approaches to prevent relapse in Plasmodium vivax. Pathog Glob Health. 2015;109:107–22.CrossRefPubMedPubMedCentral
20.
Zurück zum Zitat Wells TN, Burrows JN, Baird JK. Targeting the hypnozoite reservoir of Plasmodium vivax: the hidden obstacle to malaria elimination. Trends Parasitol. 2010;26:145–51.CrossRefPubMed Wells TN, Burrows JN, Baird JK. Targeting the hypnozoite reservoir of Plasmodium vivax: the hidden obstacle to malaria elimination. Trends Parasitol. 2010;26:145–51.CrossRefPubMed
21.
Zurück zum Zitat Hollingdale MR, Collins WE, Campbell CC, Schwartz AL. In vitro culture of two populations (dividing and nondividing) of exoerythrocytic parasites of Plasmodium vivax. Am J Trop Med Hyg. 1985;34:216–22.CrossRefPubMed Hollingdale MR, Collins WE, Campbell CC, Schwartz AL. In vitro culture of two populations (dividing and nondividing) of exoerythrocytic parasites of Plasmodium vivax. Am J Trop Med Hyg. 1985;34:216–22.CrossRefPubMed
22.
Zurück zum Zitat Hollingdale MR, Collins WE, Campbell CC. In vitro culture of exoerythrocytic parasites of the North Korean strain of Plasmodium vivax in hepatoma cells. Am J Trop Med Hyg. 1986;35:275–6.CrossRefPubMed Hollingdale MR, Collins WE, Campbell CC. In vitro culture of exoerythrocytic parasites of the North Korean strain of Plasmodium vivax in hepatoma cells. Am J Trop Med Hyg. 1986;35:275–6.CrossRefPubMed
23.
Zurück zum Zitat Sattabongkot J, Yimamnuaychoke N, Leelaudomlipi S, Rasameesoraj M, Jenwithisuk R, Coleman RE, et al. Establishment of a human hepatocyte line that supports in vitro development of the exo-erythrocytic stages of the malaria parasites Plasmodium falciparum and P. vivax. Am J Trop Med Hyg. 2006;74:708–15.PubMed Sattabongkot J, Yimamnuaychoke N, Leelaudomlipi S, Rasameesoraj M, Jenwithisuk R, Coleman RE, et al. Establishment of a human hepatocyte line that supports in vitro development of the exo-erythrocytic stages of the malaria parasites Plasmodium falciparum and P. vivax. Am J Trop Med Hyg. 2006;74:708–15.PubMed
24.
Zurück zum Zitat Chattopadhyay R, Velmurugan S, Chakiath C, Andrews Donkor L, Milhous W, Barnwell JW, et al. Establishment of an in vitro assay for assessing the effects of drugs on the liver stages of Plasmodium vivax malaria. PLoS ONE. 2010;5:e14275.CrossRefPubMedPubMedCentral Chattopadhyay R, Velmurugan S, Chakiath C, Andrews Donkor L, Milhous W, Barnwell JW, et al. Establishment of an in vitro assay for assessing the effects of drugs on the liver stages of Plasmodium vivax malaria. PLoS ONE. 2010;5:e14275.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Mazier D, Landau I, Druilhe P, Miltgen F, Guguen-Guillouzo C, Baccam D, et al. Cultivation of the liver forms of Plasmodium vivax in human hepatocytes. Nature. 1984;307:367–9.CrossRefPubMed Mazier D, Landau I, Druilhe P, Miltgen F, Guguen-Guillouzo C, Baccam D, et al. Cultivation of the liver forms of Plasmodium vivax in human hepatocytes. Nature. 1984;307:367–9.CrossRefPubMed
26.
Zurück zum Zitat Mazier D, Beaudoin RL, Mellouk S, Druilhe P, Texier B, Trosper J, et al. Complete development of hepatic stages of Plasmodium falciparum in vitro. Science. 1985;227:440–2.CrossRefPubMed Mazier D, Beaudoin RL, Mellouk S, Druilhe P, Texier B, Trosper J, et al. Complete development of hepatic stages of Plasmodium falciparum in vitro. Science. 1985;227:440–2.CrossRefPubMed
27.
Zurück zum Zitat van Schaijk BC, Janse CJ, van Gemert GJ, van Dijk MR, Gego A, Franetich JF, et al. Gene disruption of Plasmodium falciparum p52 results in attenuation of malaria liver stage development in cultured primary human hepatocytes. PLoS ONE. 2008;3:e3549.CrossRefPubMedPubMedCentral van Schaijk BC, Janse CJ, van Gemert GJ, van Dijk MR, Gego A, Franetich JF, et al. Gene disruption of Plasmodium falciparum p52 results in attenuation of malaria liver stage development in cultured primary human hepatocytes. PLoS ONE. 2008;3:e3549.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat March S, Ng S, Velmurugan S, Galstian A, Shan J, Logan DJ, et al. A microscale human liver platform that supports the hepatic stages of Plasmodium falciparum and vivax. Cell Host Microbe. 2013;14:104–15.CrossRefPubMedPubMedCentral March S, Ng S, Velmurugan S, Galstian A, Shan J, Logan DJ, et al. A microscale human liver platform that supports the hepatic stages of Plasmodium falciparum and vivax. Cell Host Microbe. 2013;14:104–15.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Bhatia SN, Balis UJ, Yarmush ML, Toner M. Effect of cell-cell interactions in preservation of cellular phenotype: cocultivation of hepatocytes and nonparenchymal cells. FASEB J. 1999;13:1883–900.CrossRefPubMed Bhatia SN, Balis UJ, Yarmush ML, Toner M. Effect of cell-cell interactions in preservation of cellular phenotype: cocultivation of hepatocytes and nonparenchymal cells. FASEB J. 1999;13:1883–900.CrossRefPubMed
30.
Zurück zum Zitat Dembele L, Franetich JF, Lorthiois A, Gego A, Zeeman AM, Kocken CH, et al. Persistence and activation of malaria hypnozoites in long-term primary hepatocyte cultures. Nat Med. 2014;20:307–12.CrossRefPubMed Dembele L, Franetich JF, Lorthiois A, Gego A, Zeeman AM, Kocken CH, et al. Persistence and activation of malaria hypnozoites in long-term primary hepatocyte cultures. Nat Med. 2014;20:307–12.CrossRefPubMed
31.
Zurück zum Zitat Khetani SR, Bhatia SN. Microscale culture of human liver cells for drug development. Nat Biotechnol. 2008;26:120–6.CrossRefPubMed Khetani SR, Bhatia SN. Microscale culture of human liver cells for drug development. Nat Biotechnol. 2008;26:120–6.CrossRefPubMed
32.
Zurück zum Zitat Ploss A, Khetani SR, Jones CT, Syder AJ, Trehan K, Gaysinskaya VA, et al. Persistent hepatitis C virus infection in microscale primary human hepatocyte cultures. Proc Natl Acad Sci USA. 2010;107:3141–5.CrossRefPubMedPubMedCentral Ploss A, Khetani SR, Jones CT, Syder AJ, Trehan K, Gaysinskaya VA, et al. Persistent hepatitis C virus infection in microscale primary human hepatocyte cultures. Proc Natl Acad Sci USA. 2010;107:3141–5.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Wang WW, Khetani SR, Krzyzewski S, Duignan DB, Obach RS. Assessment of a micropatterned hepatocyte coculture system to generate major human excretory and circulating drug metabolites. Drug Metab Dispos. 2010;38:1900–5.CrossRefPubMed Wang WW, Khetani SR, Krzyzewski S, Duignan DB, Obach RS. Assessment of a micropatterned hepatocyte coculture system to generate major human excretory and circulating drug metabolites. Drug Metab Dispos. 2010;38:1900–5.CrossRefPubMed
34.
Zurück zum Zitat March S, Ramanan V, Trehan K, Ng S, Galstian A, Gural N, et al. Micropatterned coculture of primary human hepatocytes and supportive cells for the study of hepatotropic pathogens. Nat Protoc. 2015;10:2027–53.CrossRefPubMed March S, Ramanan V, Trehan K, Ng S, Galstian A, Gural N, et al. Micropatterned coculture of primary human hepatocytes and supportive cells for the study of hepatotropic pathogens. Nat Protoc. 2015;10:2027–53.CrossRefPubMed
35.
Zurück zum Zitat Maher SP, Crouse RB, Conway AJ, Bannister EC, Achyuta AK, Clark AY, et al. Microphysical space of a liver sinusoid device enables simplified long-term maintenance of chimeric mouse-expanded human hepatocytes. Biomed Microdevices. 2014;16:727–36.CrossRefPubMedPubMedCentral Maher SP, Crouse RB, Conway AJ, Bannister EC, Achyuta AK, Clark AY, et al. Microphysical space of a liver sinusoid device enables simplified long-term maintenance of chimeric mouse-expanded human hepatocytes. Biomed Microdevices. 2014;16:727–36.CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Ng S, Schwartz RE, March S, Galstian A, Gural N, Shan J, et al. Human iPSC-derived hepatocyte-like cells support Plasmodium liver-stage infection in vitro. Stem Cell Reports. 2015;4:348–59.CrossRefPubMedPubMedCentral Ng S, Schwartz RE, March S, Galstian A, Gural N, Shan J, et al. Human iPSC-derived hepatocyte-like cells support Plasmodium liver-stage infection in vitro. Stem Cell Reports. 2015;4:348–59.CrossRefPubMedPubMedCentral
37.
38.
Zurück zum Zitat Sa-ngiamsuntorn K, Wongkajornsilp A, Kasetsinsombat K, Duangsa-ard S, Nuntakarn L, Borwornpinyo S, et al. Upregulation of CYP 450s expression of immortalized hepatocyte-like cells derived from mesenchymal stem cells by enzyme inducers. BMC Biotechnol. 2011;11:89.CrossRefPubMedPubMedCentral Sa-ngiamsuntorn K, Wongkajornsilp A, Kasetsinsombat K, Duangsa-ard S, Nuntakarn L, Borwornpinyo S, et al. Upregulation of CYP 450s expression of immortalized hepatocyte-like cells derived from mesenchymal stem cells by enzyme inducers. BMC Biotechnol. 2011;11:89.CrossRefPubMedPubMedCentral
39.
Zurück zum Zitat Sattabongkot J, Maneechai N, Phunkitchar V, Eikarat N, Khuntirat B, Sirichaisinthop J, et al. Comparison of artificial membrane feeding with direct skin feeding to estimate the infectiousness of Plasmodium vivax gametocyte carriers to mosquitoes. Am J Trop Med Hyg. 2003;69:529–35.PubMed Sattabongkot J, Maneechai N, Phunkitchar V, Eikarat N, Khuntirat B, Sirichaisinthop J, et al. Comparison of artificial membrane feeding with direct skin feeding to estimate the infectiousness of Plasmodium vivax gametocyte carriers to mosquitoes. Am J Trop Med Hyg. 2003;69:529–35.PubMed
40.
Zurück zum Zitat Usui M, Fukumoto S, Inoue N, Kawazu S. Improvement of the observational method for Plasmodium berghei oocysts in the midgut of mosquitoes. Parasit Vectors. 2011;4:118.CrossRefPubMedPubMedCentral Usui M, Fukumoto S, Inoue N, Kawazu S. Improvement of the observational method for Plasmodium berghei oocysts in the midgut of mosquitoes. Parasit Vectors. 2011;4:118.CrossRefPubMedPubMedCentral
41.
Zurück zum Zitat Kennedy M, Fishbaugher ME, Vaughan AM, Patrapuvich R, Boonhok R, Yimamnuaychok N, et al. A rapid and scalable density gradient purification method for Plasmodium sporozoites. Malar J. 2012;11:421.CrossRefPubMedPubMedCentral Kennedy M, Fishbaugher ME, Vaughan AM, Patrapuvich R, Boonhok R, Yimamnuaychok N, et al. A rapid and scalable density gradient purification method for Plasmodium sporozoites. Malar J. 2012;11:421.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Patrapuvich R, Lerdpanyangam K, Jenwithisuk R, Rungin S, Boonhok R, Duangmanee A, et al. Viability and infectivity of cryopreserved Plasmodium vivax sporozoites. Southeast Asian J Trop Med Public Health. 2016;47:171–81.PubMed Patrapuvich R, Lerdpanyangam K, Jenwithisuk R, Rungin S, Boonhok R, Duangmanee A, et al. Viability and infectivity of cryopreserved Plasmodium vivax sporozoites. Southeast Asian J Trop Med Public Health. 2016;47:171–81.PubMed
43.
Zurück zum Zitat Lupton EJ, Roth A, Patrapuvich R, Maher SP, Singh N, Sattabongkot J, Adams JH. Enhancing longevity of Plasmodium vivax and P. falciparum sporozoites after dissection from mosquito salivary glands. Parasitol Int. 2015;64:211–8.CrossRefPubMed Lupton EJ, Roth A, Patrapuvich R, Maher SP, Singh N, Sattabongkot J, Adams JH. Enhancing longevity of Plasmodium vivax and P. falciparum sporozoites after dissection from mosquito salivary glands. Parasitol Int. 2015;64:211–8.CrossRefPubMed
44.
Zurück zum Zitat Tsuji M, Mattei D, Nussenzweig RS, Eichinger D, Zavala F. Demonstration of heat-shock protein 70 in the sporozoite stage of malaria parasites. Parasitol Res. 1994;80:16–21.CrossRefPubMed Tsuji M, Mattei D, Nussenzweig RS, Eichinger D, Zavala F. Demonstration of heat-shock protein 70 in the sporozoite stage of malaria parasites. Parasitol Res. 1994;80:16–21.CrossRefPubMed
45.
Zurück zum Zitat Mikolajczak SA, Vaughan AM, Kangwanrangsan N, Roobsoong W, Fishbaugher M, Yimamnuaychok N, et al. Plasmodium vivax liver stage development and hypnozoite persistence in human liver-chimeric mice. Cell Host Microbe. 2015;17:526–35.CrossRefPubMedPubMedCentral Mikolajczak SA, Vaughan AM, Kangwanrangsan N, Roobsoong W, Fishbaugher M, Yimamnuaychok N, et al. Plasmodium vivax liver stage development and hypnozoite persistence in human liver-chimeric mice. Cell Host Microbe. 2015;17:526–35.CrossRefPubMedPubMedCentral
46.
Zurück zum Zitat Unger C, Gao S, Cohen M, Jaconi M, Bergstrom R, Holm F, et al. Immortalized human skin fibroblast feeder cells support growth and maintenance of both human embryonic and induced pluripotent stem cells. Hum Reprod. 2009;24:2567–81.CrossRefPubMed Unger C, Gao S, Cohen M, Jaconi M, Bergstrom R, Holm F, et al. Immortalized human skin fibroblast feeder cells support growth and maintenance of both human embryonic and induced pluripotent stem cells. Hum Reprod. 2009;24:2567–81.CrossRefPubMed
47.
Zurück zum Zitat Gupta N, Fisker N, Asselin MC, Lindholm M, Rosenbohm C, Orum H, et al. A locked nucleic acid antisense oligonucleotide (LNA) silences PCSK9 and enhances LDLR expression in vitro and in vivo. PLoS ONE. 2010;5:e10682.CrossRefPubMedPubMedCentral Gupta N, Fisker N, Asselin MC, Lindholm M, Rosenbohm C, Orum H, et al. A locked nucleic acid antisense oligonucleotide (LNA) silences PCSK9 and enhances LDLR expression in vitro and in vivo. PLoS ONE. 2010;5:e10682.CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Silvie O, Rubinstein E, Franetich JF, Prenant M, Belnoue E, Renia L, et al. Hepatocyte CD81 is required for Plasmodium falciparum and Plasmodium yoelii sporozoite infectivity. Nat Med. 2003;9:93–6.CrossRefPubMed Silvie O, Rubinstein E, Franetich JF, Prenant M, Belnoue E, Renia L, et al. Hepatocyte CD81 is required for Plasmodium falciparum and Plasmodium yoelii sporozoite infectivity. Nat Med. 2003;9:93–6.CrossRefPubMed
49.
Zurück zum Zitat Kaushansky A, Douglass AN, Arang N, Vigdorovich V, Dambrauskas N, Kain HS, et al. Malaria parasites target the hepatocyte receptor EphA2 for successful host infection. Science. 2015;350:1089–92.CrossRefPubMedPubMedCentral Kaushansky A, Douglass AN, Arang N, Vigdorovich V, Dambrauskas N, Kain HS, et al. Malaria parasites target the hepatocyte receptor EphA2 for successful host infection. Science. 2015;350:1089–92.CrossRefPubMedPubMedCentral
50.
Zurück zum Zitat Rodrigues CD, Hannus M, Prudencio M, Martin C, Goncalves LA, Portugal S, et al. Host scavenger receptor SR-BI plays a dual role in the establishment of malaria parasite liver infection. Cell Host Microbe. 2008;4:271–82.CrossRefPubMed Rodrigues CD, Hannus M, Prudencio M, Martin C, Goncalves LA, Portugal S, et al. Host scavenger receptor SR-BI plays a dual role in the establishment of malaria parasite liver infection. Cell Host Microbe. 2008;4:271–82.CrossRefPubMed
51.
Zurück zum Zitat Karnasuta C, Pavanand K, Chantakulkij S, Luttiwongsakorn N, Rassamesoraj M, Laohathai K, et al. Complete development of the liver stage of Plasmodium falciparum in a human hepatoma cell line. Am J Trop Med Hyg. 1995;53:607–11.CrossRefPubMed Karnasuta C, Pavanand K, Chantakulkij S, Luttiwongsakorn N, Rassamesoraj M, Laohathai K, et al. Complete development of the liver stage of Plasmodium falciparum in a human hepatoma cell line. Am J Trop Med Hyg. 1995;53:607–11.CrossRefPubMed
52.
Zurück zum Zitat Pybus BS, Marcsisin SR, Jin X, Deye G, Sousa JC, Li Q, et al. The metabolism of primaquine to its active metabolite is dependent on CYP 2D6. Malar J. 2013;12:212.CrossRefPubMedPubMedCentral Pybus BS, Marcsisin SR, Jin X, Deye G, Sousa JC, Li Q, et al. The metabolism of primaquine to its active metabolite is dependent on CYP 2D6. Malar J. 2013;12:212.CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat Pybus BS, Sousa JC, Jin X, Ferguson JA, Christian RE, Barnhart R, et al. CYP450 phenotyping and accurate mass identification of metabolites of the 8-aminoquinoline, anti-malarial drug primaquine. Malar J. 2012;11:259.CrossRefPubMedPubMedCentral Pybus BS, Sousa JC, Jin X, Ferguson JA, Christian RE, Barnhart R, et al. CYP450 phenotyping and accurate mass identification of metabolites of the 8-aminoquinoline, anti-malarial drug primaquine. Malar J. 2012;11:259.CrossRefPubMedPubMedCentral
54.
Zurück zum Zitat Vaughan AM, Mikolajczak SA, Wilson EM, Grompe M, Kaushansky A, Camargo N, et al. Complete Plasmodium falciparum liver-stage development in liver-chimeric mice. J Clin Invest. 2012;122:3618–28.CrossRefPubMedPubMedCentral Vaughan AM, Mikolajczak SA, Wilson EM, Grompe M, Kaushansky A, Camargo N, et al. Complete Plasmodium falciparum liver-stage development in liver-chimeric mice. J Clin Invest. 2012;122:3618–28.CrossRefPubMedPubMedCentral
55.
Zurück zum Zitat Soulard V, Bosson-Vanga H, Lorthiois A, Roucher C, Franetich JF, Zanghi G, et al. Plasmodium falciparum full life cycle and Plasmodium ovale liver stages in humanized mice. Nat Commun. 2015;6:7690.CrossRefPubMedPubMedCentral Soulard V, Bosson-Vanga H, Lorthiois A, Roucher C, Franetich JF, Zanghi G, et al. Plasmodium falciparum full life cycle and Plasmodium ovale liver stages in humanized mice. Nat Commun. 2015;6:7690.CrossRefPubMedPubMedCentral
56.
Zurück zum Zitat Ng S, March S, Galstian A, Gural N, Stevens KR, Mota MM, Bhatia SN. Towards a humanized mouse model of liver stage malaria using ectopic artificial livers. Sci Rep. 2017;7:45424.CrossRefPubMedPubMedCentral Ng S, March S, Galstian A, Gural N, Stevens KR, Mota MM, Bhatia SN. Towards a humanized mouse model of liver stage malaria using ectopic artificial livers. Sci Rep. 2017;7:45424.CrossRefPubMedPubMedCentral
57.
Zurück zum Zitat da Cruz FP, Martin C, Buchholz K, Lafuente-Monasterio MJ, Rodrigues T, Sonnichsen B, et al. Drug screen targeted at Plasmodium liver stages identifies a potent multistage antimalarial drug. J Infect Dis. 2012;205:1278–86.CrossRefPubMedPubMedCentral da Cruz FP, Martin C, Buchholz K, Lafuente-Monasterio MJ, Rodrigues T, Sonnichsen B, et al. Drug screen targeted at Plasmodium liver stages identifies a potent multistage antimalarial drug. J Infect Dis. 2012;205:1278–86.CrossRefPubMedPubMedCentral
58.
Zurück zum Zitat Meister S, Plouffe DM, Kuhen KL, Bonamy GM, Wu T, Barnes SW, et al. Imaging of Plasmodium liver stages to drive next-generation antimalarial drug discovery. Science. 2011;334:1372–7.CrossRefPubMedPubMedCentral Meister S, Plouffe DM, Kuhen KL, Bonamy GM, Wu T, Barnes SW, et al. Imaging of Plasmodium liver stages to drive next-generation antimalarial drug discovery. Science. 2011;334:1372–7.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Derbyshire ER, Prudencio M, Mota MM, Clardy J. Liver-stage malaria parasites vulnerable to diverse chemical scaffolds. Proc Natl Acad Sci USA. 2012;109:8511–6.CrossRefPubMedPubMedCentral Derbyshire ER, Prudencio M, Mota MM, Clardy J. Liver-stage malaria parasites vulnerable to diverse chemical scaffolds. Proc Natl Acad Sci USA. 2012;109:8511–6.CrossRefPubMedPubMedCentral
60.
Zurück zum Zitat Risco-Castillo V, Topcu S, Son O, Briquet S, Manzoni G, Silvie O. CD81 is required for rhoptry discharge during host cell invasion by Plasmodium yoelii sporozoites. Cell Microbiol. 2014;16:1533–48.CrossRefPubMed Risco-Castillo V, Topcu S, Son O, Briquet S, Manzoni G, Silvie O. CD81 is required for rhoptry discharge during host cell invasion by Plasmodium yoelii sporozoites. Cell Microbiol. 2014;16:1533–48.CrossRefPubMed
61.
Zurück zum Zitat Manzoni G, Marinach C, Topcu S, Briquet S, Grand M, Tolle M, et al. Plasmodium P36 determines host cell receptor usage during sporozoite invasion. Elife. 2017;6:e25903.CrossRefPubMedPubMedCentral Manzoni G, Marinach C, Topcu S, Briquet S, Grand M, Tolle M, et al. Plasmodium P36 determines host cell receptor usage during sporozoite invasion. Elife. 2017;6:e25903.CrossRefPubMedPubMedCentral
62.
Zurück zum Zitat Naudin C, Sirvent A, Leroy C, Larive R, Simon V, Pannequin J, et al. SLAP displays tumour suppressor functions in colorectal cancer via destabilization of the SRC substrate EPHA2. Nat Commun. 2014;5:3159.CrossRefPubMed Naudin C, Sirvent A, Leroy C, Larive R, Simon V, Pannequin J, et al. SLAP displays tumour suppressor functions in colorectal cancer via destabilization of the SRC substrate EPHA2. Nat Commun. 2014;5:3159.CrossRefPubMed
63.
Zurück zum Zitat Shortt HE, Garnham PC. Pre-erythrocytic stage in mammalian malaria parasites. Nature. 1948;161:126.CrossRefPubMed Shortt HE, Garnham PC. Pre-erythrocytic stage in mammalian malaria parasites. Nature. 1948;161:126.CrossRefPubMed
64.
Zurück zum Zitat Shortt HE, Fairley NH, Covell G, Shute PG, Garnham PC. The pre-erythrocytic stage of Plasmodium falciparum. Trans R Soc Trop Med Hyg. 1951;44:405–19.CrossRefPubMed Shortt HE, Fairley NH, Covell G, Shute PG, Garnham PC. The pre-erythrocytic stage of Plasmodium falciparum. Trans R Soc Trop Med Hyg. 1951;44:405–19.CrossRefPubMed
65.
Zurück zum Zitat McDonnell ME, Braverman LE. Drug-related hepatotoxicity. N Engl J Med. 2006;354:2191–3 (author reply 2191–3).CrossRefPubMed McDonnell ME, Braverman LE. Drug-related hepatotoxicity. N Engl J Med. 2006;354:2191–3 (author reply 2191–3).CrossRefPubMed
66.
Zurück zum Zitat Fasinu PS, Tekwani BL, Nanayakkara NP, Avula B, Herath HM, Wang YH, et al. Enantioselective metabolism of primaquine by human CYP2D6. Malar J. 2014;13:507.CrossRefPubMedPubMedCentral Fasinu PS, Tekwani BL, Nanayakkara NP, Avula B, Herath HM, Wang YH, et al. Enantioselective metabolism of primaquine by human CYP2D6. Malar J. 2014;13:507.CrossRefPubMedPubMedCentral
Metadaten
Titel
A novel immortalized hepatocyte-like cell line (imHC) supports in vitro liver stage development of the human malarial parasite Plasmodium vivax
verfasst von
Yongyut Pewkliang
Siriwan Rungin
Kaewta Lerdpanyangam
Apisak Duangmanee
Phongthon Kanjanasirirat
Phichaya Suthivanich
Khanit Sa-ngiamsuntorn
Suparerk Borwornpinyo
Jetsumon Sattabongkot
Rapatbhorn Patrapuvich
Suradej Hongeng
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Malaria Journal / Ausgabe 1/2018
Elektronische ISSN: 1475-2875
DOI
https://doi.org/10.1186/s12936-018-2198-4

Weitere Artikel der Ausgabe 1/2018

Malaria Journal 1/2018 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.