1. Introduction
In 2020 alone, more than 240 million cases of malaria were reported leading to 627,000 deaths. These values represent a substantial increase in the number of malaria case incidence and deaths estimated globally, fueled by the disruptions caused by the COVID-19 pandemic [
1]. In 2021, the World Health Organization recommended for the first time a malaria vaccine, RTS,S/AS01, for use in children living in endemic areas with moderate to high transmission [
1]. However, this vaccine only confers moderate protection against clinical disease by
Plasmodium falciparum, the most dangerous human malaria parasite [
2]. RTS,S/AS01 targets the circumsporozoite protein (CSP), the protein that densely coats the surface of sporozoites, the parasite stage deposited in the skin of the mammalian host by infected mosquitoes. Sporozoites actively migrate in the skin and invade blood vessels to complete their development in the liver. Inside hepatocytes, a single sporozoite will transform and multiply into thousands of merozoites, the red blood cells infective forms. Sporozoites and ensuing liver stages, called the pre-erythrocytic phase, represent an attractive target for immune interventions [
3].
Sera from individuals immunized with radiation-attenuated
P. falciparum sporozoites, the gold standard malaria vaccine, contain antibodies against multiple pre-erythrocytic antigens highly associated with sporozoite-induced protection [
4]. In an attempt to find novel pre-erythrocytic antigens, Peng and colleagues screened a library of
P. falciparum antigens with sera from volunteers immunized by mosquito bite under chemoprophylaxis with chloroquine [
5]. One of the antigens recognized by the sera from most of the individuals was the membrane-associated erythrocyte binding-like protein (MAEBL) [
5].
MAEBL is a large type I transmembrane protein composed of two N-terminal cysteine-rich adhesion domains homologous to the apical membrane antigen 1 (AMA-1), named M1 and M2, and a C-terminal cysteine-rich region (C-cys) structurally related with
Plasmodium Duffy binding-like family of erythrocyte binding proteins [
6]. Conserved among
Plasmodium species [
7], MAEBL was initially reported as an erythrocytic-binding protein present in blood-stage parasites [
6,
8], but was later found to be expressed in sporozoites and late liver stages [
9,
10,
11,
12]. Although dispensable for asexual blood-stage growth [
13,
14,
15,
16], immunization with MAEBL M2 domain protects animals from dying of a challenge with the lethal
Plasmodium yoelii YM strain infected red blood cells [
17].
MAEBL is required for the colonization of the mosquito salivary glands by sporozoites [
13,
14,
16]. Two main
maebl transcripts are expressed in sporozoites as a result of the alternative splicing in 3′ exons, encoding a canonical transmembrane and a putative soluble MAEBL isoform [
12]. However, only the transmembrane isoform is essential for
P. falciparum sporozoite infection of salivary glands [
16].
In sporozoites, MAEBL is found associated with the micronemes [
13,
14]. However, immunolabelling studies indicate that its subcellular localization is developmentally regulated during parasite maturation, as it changes from being restricted to the apical pole in immature sporozoites, to covering the surface of mature parasites colocalizing with CSP [
11]. In salivary gland sporozoites, the protein was detected both internally and on the parasites surface [
11,
18]. Nevertheless, antibodies generated against MAEBL domains often recognize multiple bands on western blot analysis of parasite extracts that might hinder conclusions on the localization, particularly when sera reactivity is not evaluated also in a knockout line [
18].
While MAEBL was suggested to be dispensable for liver infection by
P. berghei sporozoites collected from the midgut of mosquitoes [
13], MAEBL-deficient
P. falciparum sporozoites from the hemolymph have been shown to exhibit impaired hepatocyte wounding and invasion capacities along with reduced liver infection of humanized chimeric mice [
14]. Indeed, antibodies against MAEBL partially inhibit hepatocyte invasion by sporozoites and/or liver-stage development [
5,
18], supporting a role for MAEBL in sporozoite infectivity in the mammalian host. In this study, and using the rodent malaria model
P. berghei, we aimed at understanding the contribution of this protein in the sequence of events that lead to a successful establishment of liver infection by sporozoites.
3. Discussion
While MAEBL is dispensable for the asexual growth of parasites in the blood [
13,
14,
15,
16], MAEBL-deficient
P. falciparum sporozoites show impairment in hepatocyte wounding and invasion in vitro, as well as decreased infectivity to humanized chimeric mice [
14]. Since previous studies indicate that antibodies against MAEBL can inhibit sporozoites invasion of hepatocytes and/or liver stage development [
5,
18], its exact contribution to the sequence of events that precedes sporozoite hepatocyte infection is worth exploring. To that end, GFP:luciferase-expressing
P. berghei parasites were genetically modified to generate several MAEBL mutant lines (
Figures S1 and S3) and their phenotype in both the vertebrate and invertebrate hosts analyzed.
Our results indicate that in the absence of MAEBL,
P. berghei sporozoites show reduced infectivity to the mammalian host (
Figure 2A,B), in contrast to previous work [
13]. These contradictory findings probably result from the distinct experimental approaches used in both studies, as our experiments were performed with sporozoites collected from the mosquito’s hemolymph instead of parasites collected from the oocysts. Since
maebl- sporozoites can successfully egress from oocysts but fail to colonize the salivary glands (
Figure 1A), we used parasites collected from the mosquito hemocoel, as this transient sporozoite population shows intermediate infectivity to the mammalian host [
24]. Noteworthy, we failed every attempt to infect animals with
maebl- salivary gland-associated sporozoites (
Table S1). Considering that we do not find mutant parasites inside salivary glands of mosquitoes (
Figure 1B), it is possible that we inoculated mice with hemolymph sporozoites that were collected with the salivary glands. Such low numbers of sporozoites are most likely insufficient to reliably yield productive infections. In addition to the inherently reduced infectivity of hemolymph parasites,
maebl- sporozoites are 10- to 100-fold less infectious than control parasites, as delays of 1 to 2 days in the prepatent period are frequently observed in mice. Yet, we cannot exclude the possibility of other proteins being up or downregulated in
maebl- sporozoites, thus, also contributing to the observed phenotype of this line. However, genetic complementation of
maebl- parasites, rescued the defective phenotype of sporozoites both in the mosquito and the mammalian host (
Figure 1A and
Figure 2). This confirms that the major impairments associated with
maebl- sporozoites result from the loss of MAEBL and not from the disarrangement of the
maebl locus.
On the other hand, the defective phenotype of the
maebl- line in this study, i.e., loss of cell wounding activity (
Figure 3D) and decreased infectivity in vitro (
Figure 3A–C) and in vivo (
Figure 2A,B), resembles that of MAEBL-deficient
P. falciparum sporozoites [
14], suggesting that the function of this protein is preserved in rodent and human
Plasmodium infecting species. MAEBL is a conserved protein that predates
Plasmodium speciation and contains two extracellular N-terminal cysteine-rich domains, named M1 and M2. Each of these domains contains two APPLE domains that are found in bacterial and eukaryotic adhesion molecules [
6,
25]. Sequence analysis shows that both the number and location of all cysteine residues present in the M1 and M2 domains are evolutionary conserved, suggesting that both regions have significant and similar functions across different
Plasmodium species [
7,
10,
26]. In blood-stages, it was suggested that MAEBL localizes to the rhoptries and surface of merozoites [
7,
8] and was shown to possess erythrocyte-binding capacity mainly through the M2 domain [
6]. Moreover, it was suggested that MAEBL also participates in the binding of sporozoites to the vector salivary glands [
13]. Based on the nature of the M1 and M2 domains and in the multiple defects of
maebl- sporozoites exhibit in vitro (
Figure 3 and
Figure 4A,B), we hypothesized that MAEBL contributes to sporozoite adhesion. To test whether MAEBL could be involved in adhesion to host cells, we evaluated the binding of mutant sporozoites to HepG2 using a flow cytometry-based assay. Our data shows that in the absence of MAEBL, sporozoites adhere less to HepG2 cells, both in the presence and absence of cytochalasin D (
Figure 4C). These observations, together with the increased percentage of floating
maebl- sporozoites observed in the gliding assays (
Figure 4A), suggest that MAEBL may contribute to the overall adhesion of sporozoites.
As reported previously we did not observe a change in the proportion of motile hemolymph sporozoites in the absence of MAEBL (
Figure 4A). Nonetheless,
maebl- sporozoites glided at a lower average speed compared to controls (
Figure 4B), challenging previous conclusions [
13]. This could indicate that, for example, the loss of MAEBL may disturb the normal dynamics of discrete adhesion sites formed by sporozoites, as it could participate directly in their formation and/or the turnover, or indirectly, by interfering with the function of other adhesins, such as TRAP or TRAP-related proteins present in such sites [
27].
It is likely that the decreased
maebl- sporozoite infectivity, observed in vitro (
Figure 3) and in vivo (
Figure 2A,B), results from multiple adhesion-dependent defects. Nevertheless, we cannot exclude a possible role for MAEBL in other steps of hepatocyte invasion, through the interaction with members of the rhoptry neck protein (RON) complex, for example. However, to our knowledge, unlike the structurally related AMA-1, MAEBL was not found associated with RONs [
28], suggesting that it acts independently of these proteins during host cell invasion.
We also generated a complemented parasite line expressing a myc-tagged MAEBL (
Figure S3). As complementation rescued the phenotype of
maebl- sporozoites, both in the vector and in the mammalian host (
Figure 5), proving it is fully functional, the
maebl:myc line was used for protein quantification and immunolocalization studies.
In this study, the myc-tag epitope coding sequences were inserted at the C-terminus of
maebl ORF right before the stop codon. This means that, theoretically, our tagging strategy only allows the detection of the transmembrane isoform (
Figure S3C). However, we cannot exclude that due to the alternative splicing other myc-tagged putative soluble and processed forms are also being detected [
12]. Nevertheless, our data indicate that MAEBL levels peak in midgut and hemolymph sporozoites (
Figure 6C). These observations are in agreement with the crucial function of MAEBL in sporozoite colonization of the vector salivary glands as well as with previous transcriptomic and proteomic studies [
22,
23,
29]. Furthermore, not all salivary gland sporozoites are positive for myc-tagged MAEBL immunolabeling and importantly, protein expression varies within the positive population. Whether MAEBL is expressed de novo in the salivary glands or is carried over from hemolymph parasites remains unknown. Interestingly, recent data from single-cell RNA sequencing reveals extensive transcription heterogeneity among the sporozoite from the same anatomical compartment [
29,
30]. This could conceivably be an explanation for the fact that we were unable to detect myc-tagged MAEBL in some sporozoites residing in the salivary glands (
Figure 6D).
In terms of protein localization, myc-tagged MAEBL frequently colocalized with TRAP (
Figure 7A), in agreement with the literature that MAEBL is associated with the micronemes of sporozoites [
13,
14]. Notably, our data unequivocally indicate that MAEBL was found not only in the micronemes but also associated to the surface of salivary gland sporozoites (
Figure 7C), a finding that might be relevant for the design of future immune interventions against
Plasmodium sporozoites.
4. Materials and Methods
4.1. Mice, Mosquitoes and Parasites
The
Plasmodium berghei ANKA strain clone 676cl1 expressing a GFP-Luciferase fusion gene via the
pbef1α promoter [
31], henceforth referred to as control line, was used to generate all mutant lines. C57BL/6, NMRI, and CD1 mice were purchased from Charles River (France) or obtained from the IBMC/i3S animal facility.
Anopheles stephensi mosquitoes (Sda500 strain) were reared in the Centre for Production and Infection of
Anopheles (CEPIA) at the Pasteur Institute using standard procedures.
4.2. Generation of Transfection Vectors
PCR reactions were performed using a high-fidelity
Taq DNA polymerase with proofreading activity (Takara Bio, Otsu, Japan) and genomic DNA of control parasites as the template. Primers used for the generation and genotyping of all mutant lines are shown in
Table S2. All PCR products were cloned into the pGEM-T Easy vector (Promega, Madison, WI, USA; unless stated otherwise), sequenced (LightRun, Eurofins Genomics, Ebersberg, Germany), and verified against the
P. berghei genome database (PlasmoDB,
http://plasmodb.org/plasmo/, accessed on 22 September 2015) using the Basic Local Alignment Search Tool (BLAST). As a matter of convenience, the intergenic regions upstream and downstream of the
maebl gene (PBANKA_0901300.2) will be referred to as 5′ and 3′ UTR, respectively.
For the generation of the maebl knockout line (maebl-), the maebl open reading frame (ORF), along with the last 462 bp of the maebl 5′UTR, were replaced by the Toxoplasma gondii dihydrofolate reductase–thymidylate synthase gene (TgDHFR/ts) selectable marker, by a double cross-over homologous recombination event. Part of the maebl 5′ (499 bp) and 3′ (493 bp) UTRs were used as homology regions and were amplified using the primer pairs P1/P2 and P3/P4, respectively. The PCR products were subcloned into the plasmid pL0001 (MRA-770; MR4), on each side of the selectable marker, using the restriction sites KpnI/ClaI or EcoRI/BamHI. The final vector was digested with KpnI and BamHI before transfection.
Genetic complementation of maebl- clone G3 parasites was achieved by reinserting the wild type coding sequence of maebl (maebl_comp) or the same gene fused with a sequence encoding two tandem myc tag epitopes (maebl:myc), along with the last 462 bp of the maebl 5′UTR, into the recombinant locus by a single cross-over homologous recombination event.
The transfection vector containing the untagged
maebl ORF was obtained as follows. The
maebl 3′UTR (510 bp) and a 1907 bp DNA fragment corresponding to the first 87 bp of
maebl ORF, the complete
maebl 5′UTR and the last 621 nucleotides of the gene upstream of
maebl (PBANKA_0901400), henceforth referred to as 5′ fragment, were amplified using the primer pairs P5/P6 and P7/P8, respectively. The 3′UTR and the 5′ fragment were inserted into the pGEM-T Easy vector (Promega) or the pCR
®-TOPO-XL
® vector (Invitrogen, Thermo Fisher Scientific, Waltham, MA, USA), respectively, and subcloned into the XhoI/NheI or XmaI/EcoRI sites of a pL0007 vector (MRA-776; MR4). The resulting plasmid was digested with HincII/BsgI to allow the insertion of a 6664 bp fragment that includes the complete coding sequence of
maebl, flanked by the last 140 bp of the
maebl 5′UTR and the first 387 bp of the 3′UTR, obtained through the digestion of the
P. berghei artificial chromosome PbAC02-99h11 (PlasmoGem, Wellcome Sanger Institute, Hinxton, Cambridge, UK) [
32,
33] with the same restriction enzymes. Finally, the entire DNA sequence ranging from the beginning of the 5′ fragment to the end of the
maebl 3′UTR (8446 bp) was inserted into a new pL0007 vector digested with HindIII, originating to the final transfection vector pL0007_MAEBLcomp. The correct orientation of the insert was confirmed by EcoRI/HincII digestion. pL0007_MAEBLcomp was linearized with PmeI before transfection.
The transfection vector containing the tagged maebl ORF was obtained through modification of the pL0007_MAEBLcomp plasmid by inserting a sequence encoding 2 copies of the myc tag epitope (2× EQKLISEEDL) right before the stop codon. The maebl 3′UTR (497 bp) and the last 669 bp of the maebl ORF (excluding the stop codon) were amplified using the primer pairs P5/P9 and P10/P11, respectively; the latter primer including the coding sequence of the tag. Both fragments were subcloned into the XhoI/EcoRV and EcoRV/EcoRI sites of a pL0007 vector, originating the plasmid pL0007_MAEBLmyc_3′UTR. The pL0007_MAEBLcomp plasmid was digested with the BstBI restriction enzyme to remove a 6689 bp fragment corresponding to the last 13 bp of the PBANKA_0901400 gene sequence, the entire maebl 5′UTR and the first 5475 bp of the maebl ORF, henceforth named 5′UTR_ORF fragment. After religation, the resultant plasmid was digested with BstBI/HincII to replace the maebl 3′UTR and the final portion of the maebl ORF by a myc-tagged version, obtained through the digestion of pL0007_MAEBLmyc_3′UTR with the same restriction enzymes. Finally, the plasmid was digested with BstBI to allow the insertion of the 5′UTR_ORF fragment, originating the final transfection vector. The correct orientation of the insert and the presence of the myc tag epitope coding sequence in the transfection vector were confirmed by DNA sequencing. The plasmid was linearized with PmeI before transfection.
4.3. Transfection and Cloning of Mutant Lines
Transfection of schizonts was performed as previously reported [
34]. Immediately after electroporation, parasites were injected intravenously into 2 mice (parental populations) and selected with the appropriate drug, starting the day after parasite inoculation. Pyrimethamine (0.07 mg/mL) was given in drinking water, to select
maebl- parasites. Once parasitemia was above 1%, blood from each animal was transferred into 2 naïve mice (transfer populations) for another round of selection. Selection of
maebl_comp and
maebl:myc parasites was performed with WR99210 (Jacobus Pharmaceutical Company, Inc., Princeton, NJ, USA). WR99210 (3.2 mg/mL) was dissolved in dH
2O 40% (
v/
v) ethanol, 3% (
v/
v) benzyl alcohol [
34], and administrated subcutaneously (16 mg/Kg) for 3 successive days. Once parasitemia was above 1%, blood from each animal was transferred into a naïve mouse (transfer population). The treatment was repeated, starting from the day of infection. Cloning populations were obtained by limiting dilution [
35].
4.4. Genotypic Analysis of Mutant Parasites by PCR and Southern Blot
Blood from infected mice was collected, filtered through a Plasmodipur filter (EuroProxima, Arnhem, The Netherlands), and lysed with 0.15% (v/v) saponin. Genomic DNA extraction and purification were done using the QIAamp DNA Blood Mini kit (Qiagen, Hilden, Germany). The integration of the constructs in the expected loci (primers P14/P15 and P7/P18, for the maebl- and maebl_myc genotyping strategies, respectively), the presence or absence of the maebl ORF in the genome of parasites (primers P12/P13), and the presence of the myc tag epitope in the final portion of maebl coding sequence (primers P19/P20), were evaluated by PCR using a high-fidelity DNA polymerase (Phusion®, New England Biolabs, Ipswich, MA, USA).
For Southern blot analysis, 2.3 to 10 μg of genomic DNA were digested with HindIII/NruI, separated by 0.8% (w/v) agarose gel electrophoresis, and transferred to a Nytran-N membrane (Amersham Hybond N+, Cytiva, Marlborough, MA, USA). The hybridization probe was obtained by PCR amplification of control DNA, using the primers P1/P2. Labelling of the probe and signal generation were performed using the AlkPhos Direct™ Labeling and Detection System with CDP-Star chemiluminescent detection reagent (Cytiva), respectively.
4.5. Evaluation of Gene Expression by Reverse Transcription PCR (RT-PCR)
Total RNA was isolated from midgut sporozoites using the NucleoSpin RNA II kit (Macherey-Nagel, Düren, Germany) and converted into cDNA using the NZY First-Strand cDNA Synthesis kit (NZYTech, Lisbon, Portugal). Detection of the
maebl cDNA by PCR was done using a high-fidelity DNA polymerase (Phusion
®, New England Biolabs) and the primers P12/P13. A region of the tubulin beta chain (PBANKA_1206900) was amplified using the primers P16/P17 and used as an internal control. Primer sequences are given in
Table S2.
4.6. Mosquito Infections and Isolation of Sporozoites
Female mosquitoes were fed on infected NMRI or CD1 mice as described elsewhere [
36]. Sporozoites were isolated from mosquitoes 17 to 27 days after the infectious bloodmeal. Midguts and salivary glands were collected into cold Dulbecco’s Phosphate Buffered Saline (DPBS; Gibco, Thermo Fisher Scientific) and disrupted with a pestle immediately before use. Hemolymph sporozoites were isolated by flushing the mosquitos with 10 to 15 µL DPBS and left on ice until use. The total number of sporozoites obtained was determined using a plastic slide with a grid (KOVA
® Glasstic
® Slides, Kova International, Inc., Garden Grove, CA, USA) and a light microscope.
4.7. Sporozoite In Vitro Assays
4.7.1. Viability Assay
Hemolymph sporozoites, collected from mosquitoes on days 18 and 19 post-infection, were incubated for 15 min in DPBS on ice or at room temperature (RT), or at RT after dilution with an equal volume of Dulbecco’s Modification of Eagle’s Medium (DMEM;Lonza, Basel, Switzerland) supplemented with 10% (v/v) fetal bovine serum (FBS; Biowest, Nuaillé, France). Propidium iodide (PI; 5 µg/mL; Sigma, Merck, Darmstadt, Germany) was added to the suspensions, which were loaded into Ibidi 18-well µ-Slides (Ibidi GmbH, Gräfelfing, Germany), at a density of 5 × 103 to 1 × 104 parasites per well, and immediately imaged using IN Cell Analyzer 2000 (Cytiva). Based on PI incorporation, sporozoites were manually classified as dead or viable (PI+ or PI− sporozoites, respectively), using ImageJ/Fiji analysis software version 1.53f51 (ImageJ, National Institutes of Health, Bethesda, MD, USA). At least 150 sporozoites were analyzed per well and the percentage of viable sporozoites was calculated by dividing the number of PI− sporozoites by the total number of analyzed sporozoites.
4.7.2. Invasion and Liver Stage Development Assays
Host cell invasion and development assays were performed with sporozoites collected from mosquitoes on days 19 to 21 post-infection. The 8-well Lab-Tek chamber slides (Thermo Fisher Scientific) were precoated with 10 µg/cm2 of collagen type I from rat tail (Sigma), overnight at 4 °C, if required. HepG2 cells (ATCC) were seeded at 1 × 105 cells per well in DMEM high glucose supplemented with 10% FBS (v/v) and cultured at 37 °C, 5% CO2, for 24 h. Infections of hepatoma cells were performed with 1.4 × 104 to 2.0 × 104 sporozoites in DMEM supplemented with 5% FBS (v/v), penicillin–streptomycin (100 U/mL; Lonza), for 2 or 48 h at 37 °C, 5% CO2, to assess sporozoite invasion and liver stage development, respectively. Preparations were fixed with 4% paraformaldehyde (PFA) (w/v) in DPBS, for 30 min, and stored at 4 °C until use.
Processing of samples was performed at RT, unless stated otherwise, and the incubation time of all antibodies was 1 h. The percentage of invaded cells was accessed using a double staining strategy [
37]. Briefly, samples were blocked with 5% FBS (
v/
v) in DPBS, for 30 min, and extracellular sporozoites were labeled with the anti-CSP 3D11 mouse monoclonal antibody (2 μg/mL; MR4) and a goat anti-mouse Alexa Fluor 568 antibody (4 μg/mL; Invitrogen). Following cell permeabilization with 1% (
v/
v) Triton X-100 (Sigma), for 4 min, sporozoites were labeled with the same primary antibody in combination with goat anti-mouse Alexa Fluor 488 antibodies (4 μg/mL; Invitrogen™). Cell nuclei were stained with DAPI. Antifade mounting medium [90% (
v/
v) glycerol (Alfa Aesar, Thermo Fisher Scientific), 0.5% (
w/
v) n-propyl gallate (Sigma), 20 mM Tris-HCl (Sigma), pH 8.0] was added to the preparations and slides were stored at 4 °C until use. Image acquisition was performed using IN Cell Analyzer 2000 (Cytiva). The numbers of sporozoites and HepG2 cell nuclei were determined using the ImageJ/Fiji analysis software (ImageJ, National Institutes of Health) or using an automated counting system, as previously described [
38]. The percentage of infected cells was calculated by dividing the total number of intracellular sporozoites by the total number of HepG2 cell nuclei.
To evaluate the development of parasites, slides were blocked with 5% FBS (
v/
v) in DPBS, for 30 min, permeabilized with 1% (
v/
v) Triton X-100, for 4 min, and labeled with an anti-CSP 3D11 mouse monoclonal antibody (2 μg/mL; MR4) and an anti-GFP rabbit antibody (1:250; MBL, Tokyo, Japan) in combination with secondary antibodies goat anti-mouse Alexa Fluor 568 (4 μg/mL; Invitrogen™) and goat anti-rabbit Alexa Fluor 488 (4 μg/mL; Invitrogen). Nuclei were stained with DAPI. Antifade mounting medium were added to the preparations and slides were stored at 4 °C until use. EEFs were counted by microscopic visualization using a Zeiss Axio Imager Z1 microscope (Carl Zeiss, Oberkochen, Germany) and AxioVision software version 4.9 (Carl Zeiss, Germany), or using an automated counting system, as described previously [
38]. To quantify the size of EEFs, images were taken of random EEFs using a Zeiss Axio Imager Z1 microscope (Carl Zeiss, Germany) and the area was manually determined based on the CSP staining, using the ImageJ/Fiji analysis software (ImageJ, National Institutes of Health).
4.7.3. Cell Wounding Assay
The capacity of sporozoites to wound cells was addressed using a standard flow cytometry-based cell-wounding assay [
20]. In brief, HepG2 cells were seeded on a 96-well plate at a density of 8 × 10
4 cells per well in DMEM supplemented with 10% FBS (
v/
v) and cultured at for 24 h at 37 °C, 5% CO
2. The cells were then incubated with ~3 × 10
4 sporozoites, isolated from mosquitoes on day 19 post-infection, in the presence of 5 μg/mL of PI for 60 min at 37 °C, 5% CO
2. Uninfected cells, incubated with or without PI, were used as controls. Cells were washed twice with warm DPBS and trypsinized. At least 7.8 × 10
3 events were analyzed with a FACS Canto II flow cytometer (BD Biosciences, Franklin Lakes, NJ, USA). Data analysis was performed using the FlowJo software version 10.7.1 (FlowJo LLC, Ashland, OR, USA).
4.7.4. Motility Assay
Sporozoites collected from mosquitoes on day 24 post-infection into DPBS were mixed with an equal volume of DMEM supplemented with 10% FBS (v/v) and transferred into a 384-microwell plate with an optical bottom (Greiner AG, Kremsmünster, Austria). After centrifugation for 5 min at 500× g, the plate was placed into the temperature-controlled microscope chamber held at 37 °C. Bright-field images were acquired every second for 1 min, using a widefield inverted Leica DMI6000 (Leica Microsystems GmbH, Wetzlar, Germany) microscope and LAS X software version 3.7.4.23463 (Leica Microsystems GmbH, Germany). Image analysis was performed using the ImageJ/Fiji analysis software (ImageJ, National Institutes of Health). Sporozoites were classified as follows: (i) attached, defined as sporozoites that were completely immobilized at the bottom of the well during the entire video; (ii) waving, defined as sporozoites that were attached only by a portion of the body; (iii) floating or (iv) motile. Motile sporozoites were further subclassified based on the completion or not of a full circle. The average speed was calculated by manually tracking at the apical end on sporozoites that glide at least one complete circle.
4.7.5. Adhesion Assay
Cell adhesion assays were performed with sporozoites collected from mosquitoes on days 19 to 21 post-infection. HepG2 cells were seeded in 96-well plates at a density of 5.0 × 104 to 1.50 × 105 cells per well, in DMEM supplemented with 10% FBS (v/v) and 1× MEM non-essential amino acids (Sigma), and cultured at 37 °C, 5% CO2 until reach confluency. Sporozoites (1.25 × 104), diluted in an equal volume of DMEM supplemented with 10% FBS (v/v), 1× MEM non-essential amino acids solution (v/v) and penicillin–streptomycin (200 U/mL; Sigma), were incubated with cells for 30 min at 37 °C, 5% CO2 under static conditions, in the presence of 1 µM cytochalasin D (Sigma) or DMSO (Sigma). Following incubation, the supernatant was removed, and cells were washed twice with warm DPBS. Unattached sporozoites, defined as the number of GFP+-parasites present in the supernatants, were quantified by flow cytometry. Subsequently, cells were trypsinized and analyzed by flow cytometry, to determine the number of extracellular GFP+-sporozoites that were attached to cells. The high levels of autofluorescence of HepG2 cells precluded the quantification of intracellular sporozoites. Data were acquired using a CytoFLEX S flow cytometer (Beckman Coulter, Inc., Brea, CA, USA) and analyzed with the CytExpert version 2.0 (Beckman Coulter, Inc.). The numbers of attached and unattached sporozoites were determined based on sample volume and cell concentration. The percentage of the attached sporozoites was calculated by dividing the number of attached parasites by the total number of sporozoites recovered (attached and unattached).
4.7.6. Quantification and Subcellular Localization of Myc-Tagged MAEBL by Immunofluorescence
Sporozoites were collected from mosquitoes and transferred to an Ibidi 18-well µ-Slides (Ibidi GmbH). Sample processing was performed at RT, unless stated otherwise. Preparations were fixed with 4% PFA (w/v) in DPBS, for 30 min, permeabilized with 1% (v/v) Triton X-100, for 4 min, and blocked with 5% FBS (v/v) in DPBS, for 30 min. Sporozoites were stained with a mouse monoclonal anti-myc tag antibody clone 4A6 (5 μg/mL, Merck), overnight at 4 °C, and with goat anti-mouse Alexa Fluor 568 antibodies (2 μg/mL; Invitrogen™), for 30 min. Between each step (except after blocking), wells were washed five times with DPBS. Antifade mounting medium was added to the preparations and slides were immediately imaged using an inverted epifluorescence Leica DMI6000 microscope (Leica Microsystems) and LAS X software version 3.7.4.23463 (Leica Microsystems). Sporozoite signal intensity was quantified as integrated density (the product of the sporozoite area and the mean grey value), using ImageJ/Fiji software (ImageJ, National Institutes of Health). The background fluorescence was subtracted from the integrated density value for every sporozoite. For each day and condition, control sporozoites were used as negative control. The percentage of myc-positive sporozoites was calculated by dividing the number of maebl:myc sporozoites with a fluorescence intensity superior to highest value detected for control sporozoites. For illustrative purpose only, the Smooth filter of the ImageJ/Fiji software was applied to the GFP channel in the representative sporozoite images.
To study of the subcellular localization of myc-tagged MAEBL, maebl:myc sporozoites were stained with anti-myc tag antibodies as described above. Additionally, sporozoites were probed with a rabbit polyclonal anti-TRAP repeats antibody (1:10,000), overnight at 4 °C, stained with goat Alexa Fluor® 647 anti-rabbit IgG antibodies (2 μg/mL; Life Technologies, Thermo Fisher Scientific), for 30 min at RT, and incubated with DAPI (1:5000), for 10 min at RT. The wells were washed with DPBS between each step. Finally, preparations were mounted with antifade solution and immediately imaged. Images were acquired using an inverted microscope Leica TCS SP5 II (Leica Microsystems) and LAS AF software version 2.6.3.8173 (Leica Microsystems), and processed using ImageJ/Fiji software (ImageJ, National Institutes of Health) by projecting the maximum intensity of 7 to 12 contiguous z-stacks, separated by 0.17 to 0.25 µm. For illustrative purpose only, the Smooth filter of the ImageJ/Fiji software was applied to the GFP channel in the representative images of sporozoites.
4.7.7. Transmission Electron Microscopy
Infected salivary glands were collected 27 days post-infections and fixed in 2.5% (w/v) glutaraldehyde (Electron Microscopy Sciences, Hatfield, PA, USA) and 2% (w/v) formaldehyde (Electron Microscopy Sciences) in 0.1 M sodium cacodylate buffer (pH 7.4) for 24 h. Samples were washed in buffer, postfixed with 2% (w/v) osmium tetroxide (Electron Microscopy Sciences) in 0.1 M sodium cacodylate buffer (pH 7.4) for 2 h, washed with water and incubated with 1% (w/v) uranyl acetate (Electron Microscopy Sciences) overnight. Subsequently, samples were dehydrated with ethanol and embedded in EPON resin (Electron Microscopy sciences). Ultrathin sections of 50 nm thickness were cut using an ultramicrotome (RMC PowerTome XL, Boeckeler Instruments, Inc., Tucson, AZ, USA), mounted on mesh copper grids, and stained with uranyl acetate substitute (Electron Microscopy sciences) and lead citrate (Electron Microscopy sciences) for 5 min each. Samples were visualized using a JEOL JEM 1400 transmission electron microscope (JEOL Ltd., Tokyo, Japan). Images were digitally recorded using a CCD digital camera Orius 1100 W (Japan) and analyzed using ImageJ/Fiji software (ImageJ, National Institutes of Health).
For the detection of myc-tagged MAEBL in salivary gland sporozoites by immunoelectron microscopy, salivary glands of mosquitoes were collected on day 18 post-infection, fixed in 0.05% (w/v) glutaraldehyde, 2% (w/v) PFA, 4% (w/v) sucrose in 0.1 M PBS, for 1 h, and washed with PBS. Samples were sequentially postfixed with 2% (w/v) osmium tetroxide in 0.1 M sodium cacodylate buffer (pH 7.4) for 1 h, washed with water, incubated with 1% (w/v) uranyl acetate for 45 min, dehydrated with ethanol and embedded in EPON resin. Ultrathin sections of 60 nm thickness were mounted on mesh nickel grids and processed as follows. Sections washed with Tris-buffered saline (TBS), incubated with 14.4% (w/v) sodium metaperiodate (Merck) for 1 h, washed with TBS, immersed with 10–20 mM glycine (±0.15%; w/v) for 5 min, blocked with 2% (w/v) BSA (AURION BSA-c™, Wageningen, The Netherlands) in TBS for 30 min and incubated with a mouse monoclonal anti-myc tag antibody clone 4A6 (100 μg/mL, Merck) in 2% (w/v) BSA 3% (w/v) NaCl in TBS, overnight at 4 °C. Sections were washed with 0.1% (w/v) BSA in TBS, incubated with 1% (w/v) BSA in TBS for 20 min, and then with goat anti-mouse secondary antibodies conjugated to 6 nm gold particles (1:20, Abcam, Cambridge, UK) diluted in 1% (w/v) BSA in TBS, for 1 h. Finally, sections were washed with TBS, post-fixed in 1% (w/v) glutaraldehyde in TBS for 5 min, washed with water and stained with uranyl acetate substitute and lead citrate for 1 min each. Samples were viewed using a JEOL JEM 1400 transmission electron microscope (JEOL Ltd.). Images were digitally recorded using a CCD digital camera Orius 1100 W (Japan) and analyzed using ImageJ/Fiji software (ImageJ, National Institutes of Health).
4.7.8. Western Blot Analysis
Sporozoites were collected from the midgut of mosquitos on days 17 and 18, mechanically liberated from oocysts, filtered using a 35 μm cell strainer cap (Falcon), and stored at −80 °C until use. Lysates of 8.0 × 104 sporozoites supplemented with cOmplete™, EDTA-free Protease Inhibitor Cocktail (Roche, Basel, Switzerland) were denatured in 2× Laemmli buffer (0.25 M Tris-HCl, pH 6.8, 5% SDS, 20% glycerol 0.02% bromophenol blue, 2.5% β-Mercaptoethanol), for 10 min at 95 °C. Samples were diluted to 1× Laemmli buffer and separated on an 8% (w/v) acrylamide gel by SDS-PAGE. Proteins were allowed to transfer to a PVDF membrane using a wet transfer system, for 16 h at 20 V, in 1× Towbin buffer [25 mM Tris, 192 mM Glycine, 20% (v/v) methanol] with 0.025% (w/v) SDS. After transfer, the membrane was rinsed with PBS and blocked with 5% (w/v) skim milk, 0.1% Tween 20 (Sigma) in PBS, for at least 1 h at RT. Incubations with a mouse monoclonal anti-myc tag antibody clone 4A6 (5 µg/mL, Merck) or with anti-CSP 3D11 mouse monoclonal antibody (0.3 μg/mL; MR4), diluted in blocking solution, were performed overnight at 4 °C or for 1 h at RT, respectively. The membranes were washed, probed with horseradish peroxidase-conjugated goat anti-mouse secondary antibodies (1:5000; SouthernBiotech, Birmingham, AL, USA) diluted in blocking buffer, for 1 h at RT, and washed again. Signal detection was performed using SuperSignal West Pico Chemiluminescent Substrate (Thermo Scientific, Thermo Fisher Scientific) and Amersham Hyperfilm ECL (Cytiva). Films were revealed using the Fujifilm FPM-100A film processor (Fujifilm, Tokyo, Japan).
4.8. Sporozoite In Vivo Assays
To assess mutant sporozoite infectivity and liver-stage development in vivo, C57BL/6 mice were injected i.v. with hemolymph or salivary gland sporozoites, isolated from mosquitoes at day 20 to 25 post-infection.
4.8.1. Bioluminescence Imaging
Bioluminescence imaging was performed as previously described [
39], using the IVIS Lumina LT System (PerkinElmer, Inc., Waltham, MA, USA). Mice were imaged 1- and 2-days post-infection to quantify parasite loads in the liver. Before image acquisition, the ventral fur of mice was depilated with an appropriate clipper. Animals were anesthetized with isoflurane and injected subcutaneously with 2.4 mg of D-luciferin potassium salt (PerkinElmer, Inc.) dissolved in DPBS, 5 min before image acquisition. A non-infected mouse was routinely imaged in parallel to evaluate background noise signal. Quantitative analysis in the anatomical region of interest (ROI) encompassing the liver was performed using the Living Image software version 4.4 (PerkinElmer, Inc.), as previously described [
39].
4.8.2. Parasitemia
Parasitemia was assessed daily by analysis of Giemsa-stained thin blood smears, starting on day 3 or 4 post-inoculation. The prepatent period was defined as the number of days until mice reached 0.1% parasitemia.
4.9. Statistical Analysis
Statistical analyses were performed using the GraphPad Prism Software (version 9.3.0). Statistical significance: p < 0.05 (*), p < 0.01 (**), p < 0.001 (***), p < 0.0001 (****).