Pellicle formation in the malaria parasite · SHORT REPORT Pellicle formation in the malaria...

8
SHORT REPORT Pellicle formation in the malaria parasite Maya Kono 1 , Dorothee Heincke 1,2 , Louisa Wilcke 1,2 , Tatianna Wai Ying Wong 1,2 , Caroline Bruns 1, *, Susann Herrmann 1,2, , Tobias Spielmann 1 and Tim W. Gilberger 1,2,3,§ ABSTRACT The intraerythrocytic developmental cycle of Plasmodium falciparum is completed with the release of up to 32 invasive daughter cells, the merozoites, into the blood stream. Before release, the final step of merozoite development is the assembly of the cortical pellicle, a multi- layered membrane structure. This unique apicomplexan feature includes the inner membrane complex (IMC) and the parasites plasma membrane. A dynamic ring structure, referred to as the basal complex, is part of the IMC and helps to divide organelles and abscises in the maturing daughter cells. Here, we analyze the dynamics of the basal complex of P. falciparum. We report on a novel transmembrane protein of the basal complex termed BTP1, which is specific to the genus Plasmodium. It colocalizes with the known basal complex marker protein MORN1 and shows distinct dynamics as well as localization when compared to other IMC proteins during schizogony. Using a parasite plasma membrane marker cell line, we correlate dynamics of the basal complex with the acquisition of the maternal membrane. We show that plasma membrane invagination and IMC propagation are interlinked during the final steps of cell division. KEY WORDS: Malaria, Apicomplexa, Cell division, Inner membrane complex, Plasma membrane, Basal complex INTRODUCTION Apicomplexans are a clade of single-cell eukaryotes, the majority of which are obligate intracellular parasites (Cavelier-Smith, 1993). Infections involving apicomplexan parasites, such as Plasmodium (the causative agent of malaria), represent a severe threat to global health. Apicomplexan parasites multiply within their host cell (Fig. 1A) through internal budding, resulting in either two daughter cells (endodyogeny) or multiple progeny (schizogony). Toxoplasma parasites can reproduce by means of endodyogeny and form only two daughter cells within a still-polarized mother cell. Plasmodium parasites replicate through schizogony, resulting in a first unpolarized multinucleated schizont (reviewed in Francia and Striepen, 2014). The full assembly of organelles, the pellicle and cytoplasma segmentation (cytomere formation) of the nascent daughter cells takes place at the very end of schizogony. In both apicomplexan parasites, the inner membrane complex (IMC) represents a scaffold for assembly of daughter parasites (Hu et al., 2002; Keeley and Soldati, 2004; Striepen et al., 2007; Agop- Nersesian et al., 2010; Kono et al., 2012). The IMC is a membranous structure underlying the parasite plasma membrane (PPM). It is established before the final nuclear division at the apical end of the nascent daughter cell, a process guided by the centrosomes (Striepen et al., 2000, 2007; Hu et al., 2002; Kono et al., 2012). In Toxoplasma gondii, the apical and the basal end of the IMC, termed apical cap and basal complex respectively, represent two specialized cytoskeletal structures that are characterized by specific sets of proteins. So far, at least ten of them, including MORN1 (Gubbels et al., 2006; Hu et al., 2006), DLC (Hu et al., 2006), Centrin2 (Hu, 2008), the alveolins IMC5, IMC8, IMC9 and IMC13 (Anderson-White et al., 2011) and 14-3- 3 and MSC1a (Lorestani et al., 2012) have been found to be located at the basal complex in T. gondii. This structure functions as a contractile ring promoting daughter cell segregation (Ferguson et al., 2008; Hu, 2008; Lorestani et al., 2010). It migrates distally away from the apical end of the daughter cells, marking the basal end of the newly formed parasite at the end of cytokinesis (Gubbels et al., 2006; Hu et al., 2006; Ferguson et al., 2008; Hu, 2008). Ultrastructural studies have revealed that the basal complex comprises two electron-dense structures surrounding the basal end of the IMC (Anderson-White et al., 2011). In contrast, the only data available on the basal complex in Plasmodium is the localization of MORN1 using cross-reacting antibodies from the T. gondii homolog (Gubbels et al., 2006). The coating of the mature merozoite by the PPM is the final step of pellicle assembly and thus cytokinesis. This process starts with the invagination of the PPM before gradually sheathing the cell. This chronology has also been shown in Plasmodium berghei liver- stage schizogony, where the PPM first ingresses around cytomeres before each daughter cell is individually covered and the parasitophorous vacuole membrane (PVM) disintegrates (Graewe et al., 2012). Here, we report on the dynamics of the basal complex in P. falciparum merozoites using a newly identified protein (PFF0570c) termed basal complex transmembrane protein 1 (BTP1), which is unique to the genus Plasmodium. RESULTS AND DISCUSSION BTP1 is a Plasmodium-specific transmembrane protein expressed during schizogony BTP1 (PFF0570c) was initially identified in a screen for proteins implicated in invasion (Hu et al., 2010). The protein has no homologs outside of the genus of Plasmodium and appears to be a recent Plasmodium-specific evolutionary innovation, like the recently described IMC membrane matrix protein MAL13P1.228, which marks transversal structures in the pre-sexual stages of the parasite (Kono et al., 2012). Most apicomplexan sequence Received 1 October 2015; Accepted 31 December 2015 1 Department of Cellular Parasitology, Bernhard-Nocht-Institute for Tropical Medicine, Hamburg 20359, Germany. 2 M.G. DeGroote Institute for Infectious Disease Research, Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario L8N 3Z5, Canada. 3 Center for Structural Systems Biology, Hamburg 22607, Germany. *Present address: Leibniz Institut fu ̈ r Molekulare Pharmakologie (FMP) & Freie Universita ̈ t Berlin, Department of Molecular Pharmacology and Cell Biology, Germany. Present address: Department of Biochemistry and Molecular Biology, Bio21, The University of Melbourne, Australia. § Author for correspondence ([email protected]) This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use, distribution and reproduction in any medium provided that the original work is properly attributed. 673 © 2016. Published by The Company of Biologists Ltd | Journal of Cell Science (2016) 129, 673-680 doi:10.1242/jcs.181230 Journal of Cell Science

Transcript of Pellicle formation in the malaria parasite · SHORT REPORT Pellicle formation in the malaria...

Page 1: Pellicle formation in the malaria parasite · SHORT REPORT Pellicle formation in the malaria parasite Maya Kono1, Dorothee Heincke1,2, Louisa Wilcke1,2, Tatianna Wai Ying Wong1,2,

SHORT REPORT

Pellicle formation in the malaria parasiteMaya Kono1, Dorothee Heincke1,2, Louisa Wilcke1,2, Tatianna Wai Ying Wong1,2, Caroline Bruns1,*,Susann Herrmann1,2,‡, Tobias Spielmann1 and Tim W. Gilberger1,2,3,§

ABSTRACTThe intraerythrocytic developmental cycle of Plasmodium falciparumis completed with the release of up to 32 invasive daughter cells, themerozoites, into the blood stream. Before release, the final step ofmerozoite development is the assembly of the cortical pellicle, amulti-layered membrane structure. This unique apicomplexan featureincludes the inner membrane complex (IMC) and the parasite’splasma membrane. A dynamic ring structure, referred to as the basalcomplex, is part of the IMC and helps to divide organelles andabscises in the maturing daughter cells. Here, we analyze thedynamics of the basal complex of P. falciparum. We report on a noveltransmembrane protein of the basal complex termed BTP1, which isspecific to the genusPlasmodium. It colocalizes with the known basalcomplex marker protein MORN1 and shows distinct dynamics as wellas localization when compared to other IMC proteins duringschizogony. Using a parasite plasma membrane marker cell line,we correlate dynamics of the basal complex with the acquisition of thematernal membrane. We show that plasma membrane invaginationand IMC propagation are interlinked during the final steps of celldivision.

KEY WORDS: Malaria, Apicomplexa, Cell division, Inner membranecomplex, Plasma membrane, Basal complex

INTRODUCTIONApicomplexans are a clade of single-cell eukaryotes, the majority ofwhich are obligate intracellular parasites (Cavelier-Smith, 1993).Infections involving apicomplexan parasites, such as Plasmodium(the causative agent of malaria), represent a severe threat to globalhealth. Apicomplexan parasites multiply within their hostcell (Fig. 1A) through internal budding, resulting in either twodaughter cells (endodyogeny) or multiple progeny (schizogony).Toxoplasma parasites can reproduce by means of endodyogeny andform only two daughter cells within a still-polarized mother cell.Plasmodium parasites replicate through schizogony, resulting in afirst unpolarized multinucleated schizont (reviewed in Francia andStriepen, 2014). The full assembly of organelles, the pellicle andcytoplasma segmentation (cytomere formation) of the nascent

daughter cells takes place at the very end of schizogony. In bothapicomplexan parasites, the inner membrane complex (IMC)represents a scaffold for assembly of daughter parasites (Hu et al.,2002; Keeley and Soldati, 2004; Striepen et al., 2007; Agop-Nersesian et al., 2010; Kono et al., 2012). The IMC is amembranous structure underlying the parasite plasma membrane(PPM). It is established before the final nuclear division at the apicalend of the nascent daughter cell, a process guided by thecentrosomes (Striepen et al., 2000, 2007; Hu et al., 2002; Konoet al., 2012). In Toxoplasma gondii, the apical and the basal end ofthe IMC, termed apical cap and basal complex respectively,represent two specialized cytoskeletal structures that arecharacterized by specific sets of proteins. So far, at least ten ofthem, including MORN1 (Gubbels et al., 2006; Hu et al., 2006),DLC (Hu et al., 2006), Centrin2 (Hu, 2008), the alveolins – IMC5,IMC8, IMC9 and IMC13 (Anderson-White et al., 2011) – and 14-3-3 and MSC1a (Lorestani et al., 2012) have been found to be locatedat the basal complex in T. gondii. This structure functions as acontractile ring promoting daughter cell segregation (Fergusonet al., 2008; Hu, 2008; Lorestani et al., 2010). It migrates distallyaway from the apical end of the daughter cells, marking the basalend of the newly formed parasite at the end of cytokinesis (Gubbelset al., 2006; Hu et al., 2006; Ferguson et al., 2008; Hu, 2008).Ultrastructural studies have revealed that the basal complexcomprises two electron-dense structures surrounding the basal endof the IMC (Anderson-White et al., 2011). In contrast, the only dataavailable on the basal complex in Plasmodium is the localization ofMORN1 using cross-reacting antibodies from the T. gondiihomolog (Gubbels et al., 2006).

The coating of the mature merozoite by the PPM is the final stepof pellicle assembly and thus cytokinesis. This process starts withthe invagination of the PPM before gradually sheathing the cell.This chronology has also been shown in Plasmodium berghei liver-stage schizogony, where the PPM first ingresses around cytomeresbefore each daughter cell is individually covered and theparasitophorous vacuole membrane (PVM) disintegrates (Graeweet al., 2012).

Here, we report on the dynamics of the basal complex in P.falciparummerozoites using a newly identified protein (PFF0570c)termed basal complex transmembrane protein 1 (BTP1), which isunique to the genus Plasmodium.

RESULTS AND DISCUSSIONBTP1 is a Plasmodium-specific transmembrane proteinexpressed during schizogonyBTP1 (PFF0570c) was initially identified in a screen for proteinsimplicated in invasion (Hu et al., 2010). The protein has nohomologs outside of the genus of Plasmodium and appears to be arecent Plasmodium-specific evolutionary innovation, like therecently described IMC membrane matrix protein MAL13P1.228,which marks transversal structures in the pre-sexual stages ofthe parasite (Kono et al., 2012). Most apicomplexan sequenceReceived 1 October 2015; Accepted 31 December 2015

1Department of Cellular Parasitology, Bernhard-Nocht-Institute for TropicalMedicine, Hamburg 20359, Germany. 2M.G. DeGroote Institute for InfectiousDisease Research, Department of Pathology and Molecular Medicine, McMasterUniversity, Hamilton, Ontario L8N 3Z5, Canada. 3Center for Structural SystemsBiology, Hamburg 22607, Germany.*Present address: Leibniz Institut fur Molekulare Pharmakologie (FMP) & FreieUniversitat Berlin, Department of Molecular Pharmacology and Cell Biology,Germany. ‡Present address: Department of Biochemistry and Molecular Biology,Bio21, The University of Melbourne, Australia.

§Author for correspondence ([email protected])

This is an Open Access article distributed under the terms of the Creative Commons AttributionLicense (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use,distribution and reproduction in any medium provided that the original work is properly attributed.

673

© 2016. Published by The Company of Biologists Ltd | Journal of Cell Science (2016) 129, 673-680 doi:10.1242/jcs.181230

Journal

ofCe

llScience

Page 2: Pellicle formation in the malaria parasite · SHORT REPORT Pellicle formation in the malaria parasite Maya Kono1, Dorothee Heincke1,2, Louisa Wilcke1,2, Tatianna Wai Ying Wong1,2,

innovations are associated with adaptation to a specific ecologicalniche and can be connected with parasite survival strategies, such ashost-cell invasion or immune evasion (Wasmuth et al., 2009).BTP1 encompasses 650 amino acids and is predicted to possess

four transmembrane domains (Fig. 1B). To investigate itsexpression and localization during the intraerythrocytic cycle, weendogenously tagged the corresponding gene with GFP using a 3′-replacement strategy (Fig. S1A). Correct integration into theappropriate gene locus was confirmed with PCR analysis(Fig. S1B). Expression was analyzed using tightly synchronized

parasites, revealing a maximum expression of BTP1 atapproximately 42 h post invasion, starting at about 34 h postinvasion and decreasing towards the end of schizogony (Fig. 1C).Even though BTP1–GFP has a predicted molecular weight of105 kDa, the fusion protein BTP1–GFP runs at around 115 kDa,possibly owing to its extensive phosphorylation (Treeck et al.,2011).

Studies of the localization of BTP1–GFP showed that thisprotein marks a dynamic ring structure during daughter cellformation. The ring structure moves along the longitudinal

Fig. 1. Expression and localization of BTP1. (A) A graphic of the two modes of parasite replication depicting either endodyogeny of Toxoplasma sp. orschizogony of Plasmodium sp. N, nucleus; PV, parasitophorous vacuole; PPM, parasite plasmamembrane; IMC, inner membrane complex. (B) Schematic of theoverall structure of BTP1. Blue, predicted transmembrane domains. (C) Stage-specific expression profile of 3D7-PfBTP1–GFP cells using tightly synchronizedparasite material harvested in 4-h intervals commencing 34 h post invasion (hpi). Expression of the transgene from the endogenous locus was shown by westernblot analysis using anti-GFP antibodies (α-GFP, upper panel); the fusion protein is approximately 115 kDa (calculated molecular mass 105 kDa). Anti-BIPantibody (α-BiP) was used as a loading control. (D) BTP1–GFP can be visualized as a contractile ring structure in unfixed late-stage parasites (T1–T4). It firstappears at the apical pole of the nascent merozoite (T1) and ends up at the basal pole of the mature merozoite (T4). Nuclei were stained with DAPI (blue).Enlargement of selected areas are marked with a white square and referred to as ‘zoom’ (a fourfold magnification). (E) Surface-rendering plot of anindicated section, highlighting the spatial relationship between nuclei and the basal-complex ring. (F) Colocalization of BTP1–GFP and the apical membraneantigen 1 (αAMA1; red); AMA1 localizes in the micronomes at the apical pole opposing BPT1, confirming the basal localization of BTP1–GFP at the end ofdaughter cell formation. (G–I) Localization of the BPT1 deletion mutants. (G) Deletion of all four transmembrane domains leads to a nuclear and cytosolicdistribution of BTP1Δ4−GFP. (H) Expression of BTP1 with one (BTP1Δ3−GFP) or two (BTP1Δ2−GFP, I) transmembrane domains results in localization of mutantBTP1 at the food vacuole membrane. DIC, differential interference contrast.

674

SHORT REPORT Journal of Cell Science (2016) 129, 673-680 doi:10.1242/jcs.181230

Journal

ofCe

llScience

Page 3: Pellicle formation in the malaria parasite · SHORT REPORT Pellicle formation in the malaria parasite Maya Kono1, Dorothee Heincke1,2, Louisa Wilcke1,2, Tatianna Wai Ying Wong1,2,

axis from the apical to the basal end of the nascent merozoite(Fig. 1D,E; Movies 1,2). At the end of schizogony, the BTP1 ringstructure is concentrated at the basal end of the daughter cellsdirectly opposing the apical pole, which is defined by secretoryorganelles such as the micronemes (Fig. 1F). Overexpression of thewild-type protein shows the same localization pattern as theendogenously tagged BTP1 (Fig. S2A).Deletion of the C-terminus, including all transmembrane

domains, resulted in a mainly nuclear localization with somecytosolic background staining (Fig. 1G). Interestingly, theinclusion of individual transmembrane domains led tomislocalization of the protein to the food vacuole membrane(Fig. 1H,I). These findings indicate that the truncated protein lackscrucial trafficking determinants or that the transmembrane domaindeletion has an altered membrane topology that interferes withcorrect trafficking.To identify the membrane topology of BTP1, a Proteinase K

protection assay was conducted. First, saponin and digitonin wereused for permeabilization of the erythrocyte membrane, theparasitophorous vacuole membrane and parasite plasmamembrane. Second, Proteinase K was added, leading to thedegradation of all exposed proteins except those proteins orprotein domains that were surrounded by organelle membranes.The assay revealed that the short GFP-tagged C-terminus of BTP1was sensitive to Proteinase K treatment, which suggests that both

C- and N-termini are cytosolic (Fig. S1C,D). Hence, especially thelong N-terminus with its multiple phosphorylation sites couldinteract with other proteins or membranes.

BTP1 marks the basal rim of the nascent IMCIt has been previously shown that the IMC is defined by distinctsubgroups of proteins, which differ in their temporal and spatialassociation with the IMC (Kono et al., 2012). Group A proteins,such as the glideosome-associated protein with multiple membranespans (GAPM2; Bullen et al., 2009), are directly incorporated intoIMC membranes from the onset of the biogenesis of thecompartment. Group B proteins, such as Alveolin 5 (ALV5), arelater recruited to the emerging IMC (Kono et al., 2012). To analyzethe spatio-temporal correlation of BTP1 in more detail,colocalization studies were performed with ALV5 and GAPM2.The BTP1–GFP-defined ring remained distinct from but in closeassociation with both of these IMC markers and exhibited adifferent distribution and dynamic phenotype (Fig. 2). As opposedto ALV5 (Fig. 2A) but similar to GAPM2 (Fig. 2C), BTP1was already associated with the nascent IMC compartment at stageT1 – most likely marking the rim of the longitudinal expandingdouble-membrane structure. Also, although both ALV5 andGAPM2 were dispersed across the entire IMC membrane inmature merozoites, BTP1 remained restricted to the basal end of theIMC.

Fig. 2. Colocalization of BTP1 with ALV5 and GAPM2during IMC biogenesis. (A) Colocalization of BTP1–GFP with ALV5–mCherry (red) in unfixed parasites. Theinitial ring structure marked by BTP1–GFP precedes theappearance of Alveolin 5 (ALV5) (T1). The emergingAlveolin-5-defined ring is associated but distinct fromthat marked by BTP1 (T2). During IMC expansion, BTP1remains rim associated (T3) and, at the end of daughtercell formation, is concentrated at the basal endwith ALV5equally distributed within the IMC (T4). (B) Colocalizationwas quantified within the selected areas (‘zoom’ in A; afourfold magnification) by calculating the intensitycorrelation of the red (y-axis) and green (z-axis) signals.Pearson correlation coefficients (r) are indicated in thecorresponding scatter plots. (C) Colocalization of BTP1–GFP with GAPM2 (red) in fixed parasites. Both proteinsappeared at about the same time, marking the onset ofIMC biogenesis (T1). GAPM2 stains small ring–shapedformations that are encircled by BTP1–GFP (T2). Withelongation and propagation of the IMC from the apicaltowards the basal end of the forming daughter cell,GAPM2 is equally distributed within the forming IMC, butBTP1 is only present at the rim of the nascent IMC (T3)and, at the end of schizogony, is concentrated at thebasal pole of the daughter cell (T4). Nuclei were stainedwith DAPI (blue). Enlargement of selected areas aremarked with a white square and referred to as ‘zoom’ (afourfold magnification). The bottom row shows aschematic representation of the spatio-temporaldistribution of BTP1 and GAPM2. (D) Quantification ofthe colocalization analysis of BTP1–GFP and GAPM2(for explanation, refer to B).

675

SHORT REPORT Journal of Cell Science (2016) 129, 673-680 doi:10.1242/jcs.181230

Journal

ofCe

llScience

Page 4: Pellicle formation in the malaria parasite · SHORT REPORT Pellicle formation in the malaria parasite Maya Kono1, Dorothee Heincke1,2, Louisa Wilcke1,2, Tatianna Wai Ying Wong1,2,

BTP1 colocalizes with MORN1A contractile ring structure associated with the extending IMC wasfirst described in T. gondii with the protein multiple membraneoccupation and recognition nexus (MORN1) as its first markerprotein (Gubbels et al., 2006; Hu et al., 2006). To investigatesimilarities between the dynamics of MORN1 and the non-homologous BTP1, we first established a MORN1–GFP-overexpressing cell line (Fig. 3A–C). As initially described,MORN1 localizes to a focal structure as well as to the contractilering (Fig. 3B, arrowheads). This might represent the centrocone, aspecialized nuclear structure that organizes the mitotic spindle(Gubbels et al., 2006). Colocalization of this additional MORN1foci with the microtubule organizing centers (MTOCs; recognizedusing an antibody against α-tubulin) (Kono et al., 2012) pointstowards a close association of these two structures. We alsoexamined the colocalization of BTP1–GFP and MORN1–mCherryby using a bicistronic expression vector. The respective genes andfluorescent tags were fused together but separated by a sequencecoding for a viral 2A ribosome skip peptide (Szymczak et al., 2004,Straimer et al., 2012). Overexpression was controlled by a schizont-specific promoter (Fig. 3E). Western blot analysis confirmedskipping and the presence of two individual fusion proteins(Fig. 3F). Analysis of the location of both fusion proteins inunfixed parasites revealed their identical localization duringschizogony (Fig. 3G,H), defining BTP1 as a basal complexprotein. The punctate structure in the center of the ring marked byMORN1–mCherry (the putative centrocone) was, however, absentfor BTP1–GFP, providing an additional internal control that bothproteins properly localized when using this approach.In contrast to all established basal complex proteins, including

MORN1, BTP1 contains several transmembrane domains andtherefore is likely to be an integral membrane protein that isembedded in the basal rim of the IMC. Like other IMCtransmembrane proteins such as GAPM2, it is presumably Golgiderived and uses the secretory pathway to reach its final destination.Interestingly, BTP1 remained spatially separated from other IMCproteins during IMCbiogenesis. One explanation for this segregationmight be its specific interaction with other basal complex proteins,excluding other IMC proteins from this defined sub-compartment.The topology of BTP1 deduced by using the Proteinase K detectionassay (Fig. S1D) suggests that C- and N-termini are accessiblewithinthe cytosol, where they could mediate the proposed interaction. Howbasal complex proteins such as MORN1 are recruited and retained atthe basal complex in general is unknown. Although not conserved,BTP1 offers an attractive explanatory approach to basal complexarchitecture, at least in Plasmodium merozoites – as an integralprotein, it could play a role as a membrane-anchored hub in thespecific recruitment of other basal complex proteins to this structure.The multiple phosphorylation sites within the N-terminus of BTP1could help to orchestrate and regulate these interactions.

PPM formation and basal complex dynamics are twointerconnected processes during schizogonyAlthough a detailed picture of the initiation and regulation ofdaughter cell formation is emerging (Suvorova et al., 2015; Franciaand Striepen, 2014), the coordination and assembly of the pellicle asthe last step of budding ismuch less clear. In order to follow the PPMdevelopment through the erythrocytic cycle, we first generated aPPM marker that allows analysis of the PPM in living cells.PFF1210w is annotated as an integral 6-transmembrane domainprotein and as a putative sphingomyelin-synthetase (SMS1,Fig. 4A). Overexpressed as an mCherry fusion protein, it localized

at the periphery of the developing parasite (Fig. 4B; Fig. S3;Movie 3). It also highlights one or more cavities – specific PPMdomains of yet unknown function (Grüring et al., 2011) – andcolocalizes with the membrane marker fluorophore-labeledBODIPY©-FL C5-sphingomyelin lipid (Fig. 4D). Additionally,SMS1–mCherry plasmamembrane associationwas verified throughcolocalization with the merozoite surface protein 1 (MSP1, Fig. 4E).Next, to probe into the correlation between the invagination andpropagation of the plasma membrane and the basal complex, weused BTP1–GFP and the PPM-marker cell line together andperformed live-cell imaging. In the early stages, the basal complexappeared to be unconnected to the smoothly outlined PPM, whereaslater, it was situated at the sites where the PPM ingression around theforming daughter cell takes place (Fig. 4F). From work in T. gondiiwe know that the basal complex is assembled and expands before, inmid-budding, its constriction leads to a tapering of the corticalcytoskeleton (Gubbels et al., 2006; Hu, 2008; Anderson-Whiteet al., 2011). T. gondii Centrin2 has been proposed to be theconstrictive force, whereas MORN1 is thought to be a structural keyelement, upholding the integrity of the basal complex (Hu, 2008).Our data shows that the invagination process of the PPM isconnected and is in synchrony with the distal migration of the basalcomplex and BTP1. The time point of mid-budding is presumablythe moment where the basal-complex ring reaches its maximumwidth (referred to as T3) before it moves further towards the basalpole. Interestingly, this is also the time when the PPM invaginationand interactionwith the IMCbecomes evident. This suggests that thebasal complex, after initiation of the budding process in the malariaschizont, plays a role in linking the IMCwith the PPM to ensure theirpropagation in a convergent manner. BTP1 as a basal complexprotein is situated at the interface of these two membranes (Fig. 4F,time point T3). Future work will help to define the exact role ofBTP1 in this process. As a lineage-specific innovation, BTP1 mightbe also instrumental to dissect genera-specific differences in thebudding processes in the apicomplexa clade.

MATERIALS AND METHODSCell culture and transfection of P. falciparumP. falciparum (3D7) was cultured in human O+ erythrocytes according tostandard procedures using complete Roswell Park Memorial Institute(RPMI) medium (Trager and Jensen, 1976). For transfection, ring-stageparasites (10%) were electroporated with 100 µg of plasmid DNAresuspended in cytomix, as previously described (Fidock and Wellems,1997). Transfectants were selected using 10 nM WR99210 or 30 nMblasticidin. For single crossover recombination resulting in the 3′ tagging ofBTP1, 3D7 parasites were alternatively grown with and without WR99210pressure after transfection (∼2 weeks for each interval off-drug cycle). Aftertwo on–off cycles, all parasites exhibited plasmid integration into the BTP1locus, analyzed using PCR, resulting in 3D7-PfBTP1–GFP. Doubletransgenic parasites were generated by transfecting 100 µg of pBCam-PfSMS1-mCherry into the cell line 3D7-PfBTP1–GFP. For time-courseexperiments, parasites were synchronized twice with 5% sorbitol (Lambrosand Vanderberg, 1979). Late schizonts were then purified using a percollgradient. These schizonts were incubatedwith 50 ml of pre-warmedmediumand 10% hematocrit at 37°C under rolling conditions for 2–3 h to allowreinvasion. The freshly invaded ring parasites were then separated again byusing a percoll gradient, and equal amounts were divided into eight disheswith 5% hematocrit, respectively. Two dishes were harvested every 4 h, andpellets were lysed with saponin for subsequent western blot analysis.

Nucleic acids and constructsOligonucleotides used for plasmid constructions are listed in Table S1. TheGFP replacement construct for tagging the endogenous BTP1 was designedusing the pARL-GFP vector (Treeck et al., 2006). 1 kb of the 3′ end of BTP1

676

SHORT REPORT Journal of Cell Science (2016) 129, 673-680 doi:10.1242/jcs.181230

Journal

ofCe

llScience

Page 5: Pellicle formation in the malaria parasite · SHORT REPORT Pellicle formation in the malaria parasite Maya Kono1, Dorothee Heincke1,2, Louisa Wilcke1,2, Tatianna Wai Ying Wong1,2,

Fig. 3. Colocalization of BTP1 withMORN1. (A) Schematic of the structure ofMORN1. (B) Localization of P. falciparum(pf )MORN1–GFP in unfixed parasites.MORN1–GFP reveals a contractile ringstructure that moves along the longitudinalaxes of the nascent merozoite (T1–T4) withan additional punctate structure that mightrepresent the centrocone (marked witharrowheads). Nuclei were stainedwith DAPI(blue). Enlargement of selected areas aremarked with a white square and referred toas ‘zoom’ (a fourfold magnification).(C) Western blot analysis of MORN1–GFP-expressing parasites using antibodiesagainst GFP shows a fusion protein ofapproximately 70 kDa (calculatedmolecular weight 67 kDa) using late-stageparasite material. (D) Colocalization ofMORN1–GFP with α-tubulin (red) thathighlights the microtubule-organizingcenters (MTOCs) at this stage. Although theMORN1-marked ring structure is clearlydistinct from the MTOC (zoom 1), there is aclose association of the MTOC with theadditional MORN1-marked structure (zoom2). Nuclei were stained with DAPI (blue).(E) Schematic representation of thechimeric (BTP1–GFP)–2A–(MORN1–mCherry) protein that results from thebicistronic expression; the two individualproteins expressed from the entire cassetteare shown. (F) Western blot analysis of the(BTP1–GFP)–2A–(MORN1–mCherry)-expressing cell line using antibodies againstGFP (α-GFP) and mCherry (α-mCherry).2A-mediated skipping results in twoindividual fusion proteins – BTP1–GFP ofapproximately 100 kDa and MORN1–mCherry of approximately 70 kDa, asdetected in extracts from late-stageparasites. (G) Colocalization analysis ofBTP1–GFP (green) with MORN1–mCherry(red) revealed their identical localizationduring schizogony, except for theexclusively MORN1–mCherry-highlightedputative centrocone. (H) Colocalization wasquantified within the selected areas (zoom;a fourfold magnification) by calculating theintensity correlation of the red and greensignals. Pearson correlation coefficients(r) are indicated in the corresponding scatterplots. DIC, differential interference contrast.

677

SHORT REPORT Journal of Cell Science (2016) 129, 673-680 doi:10.1242/jcs.181230

Journal

ofCe

llScience

Page 6: Pellicle formation in the malaria parasite · SHORT REPORT Pellicle formation in the malaria parasite Maya Kono1, Dorothee Heincke1,2, Louisa Wilcke1,2, Tatianna Wai Ying Wong1,2,

was PCR amplified using Phusion DNA polymerase (New EnglandBiolabs), digested with NotI and AvrII, and cloned into the NotI–AvrII-digested pARL-1a-GFP plasmid, leading to a loss of the AMA1-promoter(Treeck et al., 2006). For the BTP1 overexpression constructs, BTP1

deletion constructs and MORN1 were also PCR amplified using Phusionpolymerase, were subsequently digested with KpnI and AvrII, and clonedinto the KpnI–AvrII-digested-pARL-1a-GFP plasmid. The expression wasdriven by the late-stage-specific AMA1-promoter. The bicistronic

Fig. 4. Parasite plasma membrane dynamics and correlation with BTP1. (A) Schematic of the structure of SMS1 (PFF1210w). Predicted transmembranedomains are indicated in blue. (B) Localization of SMS1–mCherry (red) during asexual proliferation in rings (R), trophozoites (TR), schizonts (S) and merozoites(M). Nuclei were stained with DAPI (blue). Enlargement of selected areas are marked with a white square and referred to as ‘zoom’ (a fourfold magnification).Please also refer to Fig. 3 and Movie 3. (C) Expression of SMS1–mCherry was analyzed by western blotting analysis using antibodies against mCherry(α-mCherry), resulting in the detection of a fusion protein of approximately 80 kDa (calculatedmolecular mass 78 kDa). (D) Colocalization of SMS1–mCherry withthe membrane marker fluorophore-labeled BODIPY©-FL C5-sphingomyelin lipid (upper panels) or the plasma membrane surface protein MSP1 (lower panel;αMSP1). (E) Colocalization was quantified within the selected area (zoom) by calculating the intensity correlation of the red (y-axis) and green (z-axis) signals.Pearson correlation coefficients (r) are indicated in the corresponding scatter plots. (F) Colocalization of BTP1–GFP (green) with SMS1–mCherry (red) introphozoites (TR) and during schizogony (T1–T4). Nuclei were stained with DAPI (blue). Enlargement of selected areas are marked with a white square andreferred to as ‘zoom’. (G) Quantification of the colocalization analysis of BTP1–GFP with SMS1–mCherry (please refer to F). (H) Expression of BTP1–GFP andSMS1–mCherry was confirmed by western blotting analysis using antibodies against GFP and mCherry. DIC, differential interference contrast.

678

SHORT REPORT Journal of Cell Science (2016) 129, 673-680 doi:10.1242/jcs.181230

Journal

ofCe

llScience

Page 7: Pellicle formation in the malaria parasite · SHORT REPORT Pellicle formation in the malaria parasite Maya Kono1, Dorothee Heincke1,2, Louisa Wilcke1,2, Tatianna Wai Ying Wong1,2,

expression of BTP1–GFP and PF10_0306-mCherry (MORN1) is based onthe same pARL-1 plasmid backbone used for the overexpression studies.Here, the full-length BTP1 was cloned into the KpnI–AvrII of pARL-1a-GFP. The codon-optimized viral 2A ‘ribosome skip’ peptide (Straimer et al.,2012; Szymczak et al., 2004) was introduced at the 3′ end of the GFPsequence with additional MluI and SpeI digestion sites in front of XhoI.MORN1 (PF10_0306) and mCherry were PCR amplified using Phusionpolymerase and subsequently cloned using MluI–SpeI and SpeI–XhoI,respectively. The overexpression plasmid pBcamR-SMS1-mCherry wasconstructed by amplifying full-length SMS1 from genomic DNA and wascloned into a derivate of pBcamR (Flueck et al., 2010). The 3×HA-tag of theoriginal vector was replaced with mCherry using NotI–SalI cut sites. SMS1was cloned into BamHI–NotI sites of the pBcamR-mCherry vector.Oligonucleotides used for plasmid construction are summarized inTable S1. All plasmids were sequenced and analyzed for accuracy.

Western blot analysisLate-stage schizont P. falciparum cultures were lysed with 0.03% saponin(Sigma-Aldrich). Parasite pellets were resuspended in an adequateamount of PBS and 5× SDS loading dye. Proteins were separated in12% SDS-PAGE gels and transferred onto nitrocellulose membranes(Schleicher & Schuell). Monoclonal mouse anti-GFP (Roche; catalognumber 11814460001) or monoclonal rat anti-mCherry (Chromotek;catalog number 5f8) antibodies were diluted 1:1000 in 2.5% (w/v)skimmed milk. Polyclonal rabbit anti-BIP antibody was diluted 1:3000(Struck et al., 2005), polyclonal rabbit anti-RhopH3 (Topolska et al., 2003)was diluted 1:1000. Polyclonal mouse anti-α-tubulin antibody was diluted1:2000 (Molecular Probes; catalog number A11126). The secondaryantibodies were horseradish peroxidase (HRP)-conjugated goat anti-rabbitimmunoglobulin G (IgG; catalog number G-21234), goat anti-mouse IgG orgoat anti-rat (1:3000, Jackson ImmunoResearch; catalog numbers 115-001-003 and 112-001-003). The immunoblots were developed by usingchemiluminescence with SuperSignal West Pico ChemiluminescentSubstrate (ThermoFisher Scientific) and visualized using the ChemiDocXRS+ System (BioRad).

Imaging and immunofluorescence assaysAll fluorescence staining was observed and captured using either a ZeissAxioskop 2plus microscope with a Hamamatsu digital camera (ModelC4742-95) or an Olympus FV1000 confocal microscope. For confocalimages and 3D reconstructions, 10–32 z-stacks (0.38-µm step size) werecollected using 488- or 594-nm lasers. For long-term observations, parasiteswere arrested using culture-grade 0.5 mg/ml concanavalin A (Sigma-Aldrich) on glass-bottomed 35-mm dishes (Ibidi) and viewed at 37°C usingan Olympus FV1000 confocal microscope equipped with an OlympusCellincubator, as previously described (Grüring et al., 2011). Imagecollection parameters were: a 4-µs dwell time, 512×512 dpi, 16–32 z-stacks(0.2–0.4-µm step size), a zoom level of 1–7 and laser levels of 1–5% for0.37 nm. All confocal images were analyzed and processed in Imaris 8.1.2.Cropping of movies and addition of time stamps were performed in ImageJ(http://rsb.info.nih.gov/ij/). Gauss filters were used with the filter widthsuggested by Imaris. Single images were processed in Adobe PhotoshopCS4. Colocalization analysis was conducted using ‘Just anotherColocalization Plugin’ (JACoP) for ImageJ (Bolte and Cordelieres, 2006).Thresholds were set to the same levels of fluorescence present in theoriginal image. The resulting data was exported to Excel for graphicrepresentation in scatter plots. Immunofluorescence assays were performedusing fixed P. falciparum parasites, as previously described (Tonkin et al.,2004). All primary antibody dilutions were prepared in 3% BSA. 1F9monoclonal anti-AMA1 (Coley et al., 2001) was diluted 1:2000, polyclonalmouse GAPM2 was diluted 1:1000 (Kono et al., 2012). The secondaryantibodies Alexa-Fluor-594 goat anti-rabbit and anti-mouse IgG (MolecularProbes; catalog numbers A-11008 and A-11001, respectively) were diluted1:2000. 1 µg/ml of DAPI (Roche) was used to visualize the nuclei. Formembrane visualization, BODIPY® FL C5-sphingomyelin (ThermoFisherScientific) was used. The dye was prepared according to the manufacturer’sprotocol to obtain a 250 mg/ml in a filter-sterilized BSA solution. For live-cell imaging, cells from the parasite culture were removed, washed once in

PBS and resuspended in an equal volume BODIPY solution and incubatedfor 5 min at 37°C.

Proteinase-K protection assayP. falciparum parasites were extracted using 0.03% saponin; the pellet waswashed with PBS and resuspended in 1.5 ml of cold SoTE (0.6 M sorbitol,20 mMTris-HCl pH 7.5 and 2 mMEDTA). Subsequently it was allocated tothree tubes (0.5 ml each). Cold SoTE was added to tube 1 and kept ascontrol. Parasites in tubes 2 and 3 were permeabilized by adding 0.5 ml ofcold 0.02% digitonin (Sigma-Aldrich) in SoTE. Samples were carefullymixed by inversion and incubated on ice for 5 min. This was followed by a10-min centrifugation at 800 g at 4°C. Supernatants were discarded, and0.5 ml of cold SoTE was added into tubes 1 and 2. 0.5 ml of cold 0.1% µg/µlProteinase K (Sigma) in SoTE was added to tube 3. All tubes were gentlymixed by inversion and incubated on ice for 30 min. Proteinase K wasinactivated by adding cold trichloroacetic acid (TCA) to a finalconcentration of 10% and further incubating on ice for 30 min. Allsamples were centrifuged at full speed for 20 min, washed once withacetone, dried briefly, resuspended in TE buffer and prepared for SDS-PAGE analysis. SDS-PAGE was performed as previously describedrunning samples 1–3 side by side; the samples were probed with eitheranti-GFP or anti-BIP antibodies, and an anti-RhopH3 antibody as control.

AcknowledgementsWe are grateful for WR99210 provided by Jacobus Pharmaceuticals. We would liketo thank Drs Ross Coppel and Brian Cooke (Monash Biomedicine DiscoveryInstitute Department of Microbiology, Melbourne) for providing antibodies againstRhopH3, Dr Michael Foley (La Trobe Institute for Molecular Science, Department ofBiochemistry and Genetics, Melbourne) for the 1F9 antibody against AMA1 andMichael Blackmann (London Schoool of Hygiene and Tropical Medicine, London)for the MSP1 antibody. Special thanks to Dr Klemens Engelberg for his criticalcomments.

Competing interestsThe authors declare no competing or financial interests.

Author contributionsM.K., D.H., L.W., T.W., C.B. and S.H. performed the experiments; M.K. and T.G.coordinated the project; M.K., D.H., T.S. and T.G. analysed the data; M.K. and T.G.wrote the paper.

FundingThis study was supported in part by the Canadian Institutes for Health Research[grant number MOP#111196]; the Canadian Foundation for Innovation (to T.W.G.);and the Natural Sciences and Engineering Research Council. Deposited in PMC forimmediate release.

Supplementary informationSupplementary information available online athttp://jcs.biologists.org/lookup/suppl/doi:10.1242/jcs.181230/-/DC1

ReferencesAgop-Nersesian, C., Egarter, S., Langsley, G., Foth, B. J., Ferguson, D. J. P. and

Meissner, M. (2010). Biogenesis of the innermembrane complex is dependent onvesicular transport by the alveolate specific GTPase Rab11B. PLoS Pathog. 6,e1001029.

Anderson-White, B. R., Ivey, F. D., Cheng, K., Szatanek, T., Lorestani, A.,Beckers, C. J., Ferguson, D. J. P., Sahoo, N. and Gubbels, M.-J. (2011). Afamily of intermediate filament-like proteins is sequentially assembled into thecytoskeleton of Toxoplasma gondii. Cell Microbiol. 13, 18-31.

Bolte, S. and Cordelieres, F. P. (2006). A guided tour into subcellular colocalizationanalysis in light microscopy. J. Microsc. 224, 213-232.

Bullen, H. E., Tonkin, C. J., O’Donnell, R. A., Tham, W.-H., Papenfuss, A. T.,Gould, S., Cowman, A. F., Crabb, B. S. and Gilson, P. R. (2009). A novel familyof Apicomplexan glideosome-associated proteins with an inner membrane-anchoring role. J. Biol. Chem. 284, 25353-25363.

Cavalier-Smith, T. (1993). Kingdom protozoa and its 18 phyla. Microbiol. Rev. 57,953-994.

Coley, A. M., Campanale, N. V., Casey, J. L., Hodder, A. N., Crewther, P. E.,Anders, R. F., Tilley, L. M. and Foley, M. (2001). Rapid and precise epitopemapping of monoclonal antibodies against Plasmodium falciparum AMA1 bycombined phage display of fragments and random peptides. Protein Eng. 14,691-698.

679

SHORT REPORT Journal of Cell Science (2016) 129, 673-680 doi:10.1242/jcs.181230

Journal

ofCe

llScience

Page 8: Pellicle formation in the malaria parasite · SHORT REPORT Pellicle formation in the malaria parasite Maya Kono1, Dorothee Heincke1,2, Louisa Wilcke1,2, Tatianna Wai Ying Wong1,2,

Ferguson, D. J. P., Sahoo, N., Pinches, R. A., Bumstead, J. M., Tomley, F. M. andGubbels, M.-J. (2008). MORN1 has a conserved role in asexual and sexualdevelopment across the apicomplexa. Eukaryot. Cell 7, 698-711.

Fidock, D. A. and Wellems, T. E. (1997). Transformation with human dihydrofolatereductase renders malaria parasites insensitive to WR99210 but does not affectthe intrinsic activity of proguanil. Proc. Natl. Acad. Sci. USA 94, 10931-10936.

Flueck, C., Bartfai, R., Niederwieser, I., Witmer, K., Alako, B. T. F., Moes, S.,Bozdech, Z., Jenoe, P., Stunnenberg, H. G. and Voss, T. S. (2010). Amajor rolefor the Plasmodium falciparum ApiAP2 protein PfSIP2 in chromosome endbiology. PLoS Pathog. 6, e1000784.

Francia, M. E. and Striepen, B. (2014). Cell division in apicomplexan parasites.Nat. Rev. Microbiol. 12, 125-136.

Graewe, S., Stanway, R. R., Rennenberg, A. andHeussler, V. T. (2012). Chronicleof a death foretold: Plasmodium liver stage parasites decide on the fate of the hostcell. FEMS Microbiol. Rev. 36, 111-130.

Gruring, C., Heiber, A., Kruse, F., Ungefehr, J., Gilberger, T.-W. and Spielmann,T. (2011). Development and host cell modifications of Plasmodium falciparumblood stages in four dimensions. Nat. Commun. 2, 165.

Gubbels, M.-J., Vaishnava, S., Boot, N., Dubremetz, J.-F. and Striepen, B.(2006). A MORN-repeat protein is a dynamic component of the Toxoplasmagondii cell division apparatus. J. Cell Sci. 119, 2236-2245.

Hu, K. (2008). Organizational changes of the daughter basal complex during theparasite replication of Toxoplasma gondii. PLoS Pathog. 4, e10.

Hu, K., Mann, T., Striepen, B., Beckers, C. J. M., Roos, D. S. and Murray, J. M.(2002). Daughter cell assembly in the protozoan parasite Toxoplasma gondii.Mol.Biol. Cell 13, 593-606.

Hu, K., Johnson, J., Florens, L., Fraunholz, M., Suravajjala, S., DiLullo, C.,Yates, J., Roos, D. S. and Murray, J. M. (2006). Cytoskeletal components of aninvasion machine—the apical complex of Toxoplasma gondii. PLoS Pathog. 2,e13.

Hu, G., Cabrera, A., Kono, M., Mok, S., Chaal, B. K., Haase, S., Engelberg, K.,Cheemadan, S., Spielmann, T., Preiser, P. R. et al. (2010). Transcriptionalprofiling of growth perturbations of the human malaria parasite Plasmodiumfalciparum. Nat. Biotechnol. 28, 91-98.

Keeley, A. and Soldati, D. (2004). The glideosome: a molecular machine poweringmotility and host-cell invasion by Apicomplexa. Trends Cell Biol. 14, 528-532.

Kono, M., Prusty, D., Parkinson, J. and Gilberger, T. W. (2013). Theapicomplexan inner membrane complex. Front. Biosci. 18, 982-992.

Lambros, C. and Vanderberg, J. P. (1979). Synchronization of Plasmodiumfalciparum erythrocytic stages in culture. J. Parasitol. 65, 418-420.

Lorestani, A., Sheiner, L., Yang, K., Robertson, S. D., Sahoo, N., Brooks, C. F.,Ferguson, D. J. P., Striepen, B. and Gubbels, M.-J. (2010). A ToxoplasmaMORN1 null mutant undergoes repeated divisions but is defective in basalassembly, apicoplast division and cytokinesis. PLoS ONE 5, e12302.

Lorestani, A., Ivey, F. D., Thirugnanam, S., Busby, M. A., Marth, G. T.,Cheeseman, I. M. and Gubbels, M.-J. (2012). Targeted proteomic dissection ofToxoplasma cytoskeleton sub-compartments using MORN1. Cytoskeleton 69,1069-1085.

Straimer, J., Lee, M. C. S., Lee, A. H., Zeitler, B., Williams, A. E., Pearl, J. R.,Zhang, L., Rebar, E. J., Gregory, P. D., Llinas, M. et al. (2012). Site-specificgenome editing in Plasmodium falciparum using engineered zinc-fingernucleases. Nat. Methods 9, 993-998.

Striepen, B., Crawford, M. J., Shaw, M. K., Tilney, L. G., Seeber, F. and Roos,D. S. (2000). The plastid of Toxoplasma gondii is divided by association with thecentrosomes. J. Cell Biol. 151, 1423-1434.

Striepen, B., Jordan, C. N., Reiff, S. and van Dooren, G. G. (2007). Building theperfect parasite: cell division in apicomplexa. PLoS Pathog. 3, e78.

Struck, N. S., de Souza Dias, S., Langer, C., Marti, M., Pearce, J. A., Cowman,A. F. and Gilberger, T. W. (2005). Re-defining the Golgi complex in Plasmodiumfalciparum using the novel Golgi marker PfGRASP. J. Cell Sci. 118, 5603-5613.

Suvorova, E. S., Francia, M., Striepen, B. and White, M. W. (2015). A novelbipartite centrosome coordinates the apicomplexan cell cycle. PLoS Biol. 13,e1002093.

Szymczak, A. L., Workman, C. J., Wang, Y., Vignali, K. M., Dilioglou, S., Vanin,E. F. andVignali, D. A. A. (2004). Correction ofmulti-gene deficiency in vivo usinga single ‘self-cleaving’ 2A peptide–based retroviral vector. Nat. Biotechnol. 22,589-594.

Tonkin, C. J., van Dooren, G. G., Spurck, T. P., Struck, N. S., Good, R. T.,Handman, E., Cowman, A. F. and McFadden, G. I. (2004). Localization oforganellar proteins in Plasmodium falciparum using a novel set of transfectionvectors and a new immunofluorescence fixation method.Mol. Biochem. Parasitol.137, 13-21.

Topolska, A. E., Lidgett, A., Truman, D., Fujioka, H. and Coppel, R. L. (2003).Characterization of a membrane-associated rhoptry protein of Plasmodiumfalciparum. J. Biol. Chem. 279, 4648-4656.

Trager, W. and Jensen, J. B. (1976). Human malaria parasites in continuousculture. Science 193, 673-675.

Treeck, M., Struck, N. S., Haase, S., Langer, C., Herrmann, S., Healer, J.,Cowman, A. F. and Gilberger, T. W. (2006). A conserved region in the EBLproteins is implicated in microneme targeting of the malaria parasite Plasmodiumfalciparum. J. Biol. Chem. 281, 31995-32003.

Treeck, M., Sanders, J. L., Elias, J. E. and Boothroyd, J. C. (2011). Thephosphoproteomes of Plasmodium falciparum and Toxoplasma gondii revealunusual adaptations within and beyond the parasites’ boundaries. Cell HostMicrobe 10, 410-419.

Wasmuth, J., Daub, J., Peregrın-Alvarez, J. M., Finney, C. A. M. and Parkinson,J. (2009). The origins of apicomplexan sequence innovation. Genome Res. 19,1202-1213.

680

SHORT REPORT Journal of Cell Science (2016) 129, 673-680 doi:10.1242/jcs.181230

Journal

ofCe

llScience