<<

Article

Toxoplasma gondii transmembrane and their modular design

SHEINER, Lilach, et al.

Abstract

Summary Host invasion by the critically relies on regulated secretion of transmembrane micronemal proteins (TM-MICs). possesses functionally non-redundant MICs complexes that participate in gliding , host cell attachment, moving junction formation, rhoptry secretion and invasion. The TM-MICs are released onto the parasite's surface as complexes capable of interacting with host cell receptors. Additionally, TgMIC2 simultaneously connects to the actomyosin system via binding to aldolase. During invasion these adhesive complexes are shed from the surface notably via intramembrane cleavage of the TM-MICs by a rhomboid . Some TM-MICs act as escorters and assure trafficking of the complexes to the . We have investigated the properties of TgMIC6, TgMIC8, TgMIC8.2, TgAMA1 and the new micronemal TgMIC16 with respect to interaction with aldolase, susceptibility to rhomboid cleavage and presence of trafficking signals. We conclude that several TM-MICs lack targeting information within their C-terminal domains, indicating that trafficking depends on yet unidentified [...]

Reference

SHEINER, Lilach, et al. Toxoplasma gondii transmembrane microneme proteins and their modular design. Molecular microbiology, 2010, vol. 77, no. 4, p. 912-929

DOI : 10.1111/j.1365-2958.2010.07255.x PMID : 20545864

Available at: http://archive-ouverte.unige.ch/unige:12352

Disclaimer: layout of this document may differ from the published version.

1 / 1 Molecular Microbiology (2010) ᭿ doi:10.1111/j.1365-2958.2010.07255.x

Toxoplasma gondii transmembrane microneme proteins and

their modular designmmi_7255 1..18

Lilach Sheiner,1†‡ Joana M. Santos,1*‡ essential for TgAMA1 but dispensable for TgMIC6 Natacha Klages,1‡ Fabiola Parussini,2 function during invasion. Noelle Jemmely,1 Nikolas Friedrich,1 Gary E. Ward2 1 and Dominique Soldati-Favre Introduction 1Department of Microbiology and Molecular Medicine, CMU, University of Geneva, 1 rue Michel-Servet, 1211 Toxoplasma gondii is an obligate intracellular parasite of Geneva 4, Switzerland. the phylum Apicomplexa, which also includes the deadly 2Department of Microbiology and Molecular Genetics, agent of , falciparum. Host cell inva- University of Vermont, Burlington, VT 05405, USA. sion by these parasites is a multi-step process (Carruthers and Boothroyd, 2007) propelled by the gliding motility machinery. It is initiated by apical attachment of the para- Summary site to the host cell, followed by reorientation, formation of Host cell invasion by the Apicomplexa critically relies a junction between the parasite and host cell membranes, on regulated secretion of transmembrane micronemal penetration and, finally, sealing of the parasitophorous proteins (TM-MICs). Toxoplasma gondii possesses vacuolar membrane. functionally non-redundant MIC complexes that par- Some of the proteins implicated in invasion are sequen- ticipate in gliding motility, host cell attachment, tially released from two types of secretory organelles, moving junction formation, rhoptry secretion and named micronemes and rhoptries (Carruthers and Sibley, invasion. The TM-MICs are released onto the para- 1997). In T. gondii, four complexes composed of soluble site’s surface as complexes capable of interacting and transmembrane microneme proteins or including with host cell receptors. Additionally, TgMIC2 simul- rhoptry neck proteins (RONs) have been investigated and taneously connects to the actomyosin system via shown to perform non-overlapping functions during inva- binding to aldolase. During invasion these adhesive sion (Fig. 1A). More complexes are, however, likely to complexes are shed from the surface notably via contribute to invasion as additional uncharacterized trans- intramembrane cleavage of the TM-MICs by a rhom- membrane microneme proteins (TM-MICs) are encoded boid protease. Some TM-MICs act as escorters in the genome. and assure trafficking of the complexes to the The selective participation of each of the four com- micronemes. We have investigated the properties of plexes in the invasion process has been uncovered by TgMIC6, TgMIC8, TgMIC8.2, TgAMA1 and the new generating conventional or conditional knockouts of the micronemal protein TgMIC16 with respect to interac- genes encoding components of the complexes. The tion with aldolase, susceptibility to rhomboid cleav- TM-MIC TgMIC2 forms a multimeric complex with the age and presence of trafficking signals. We conclude soluble partner TgM2AP (Rabenau et al., 2001; Jewett that several TM-MICs lack targeting information and Sibley, 2004). Parasites depleted in TgMIC2 are within their C-terminal domains, indicating that traf- markedly deficient in host cell attachment, motility and ficking depends on yet unidentified proteins interact- hence unable to invade host cells (Huynh and Carruthers, ing with their ectodomains. Most TM-MICs serve as 2006). Another complex, composed of the transmem- substrates for a and some of them brane protein TgMIC6, is interacting with two soluble mol- are able to bind to aldolase. We also show that the ecules TgMIC1 and TgMIC4. Genetic disruption of any of residues responsible for binding to aldolase are the three encoding genes is still compatible with parasite survival (Reiss et al., 2001) even if the complex has been demonstrated to play an important role in invasion in vitro Accepted 1 June, 2010. *For correspondence. E-mail joana. and to contribute to in vivo (Cerede et al., 2005; [email protected]; Tel. (+41) 2 2379 5656; Fax (+41) 2 Blumenschein et al., 2007; Sawmynaden et al., 2008). A 2379 5702. †Present address: Center for Tropical & Emerging Global Diseases, University of Georgia, Athens, GA, USA. ‡These authors third complex is composed of the TM-MIC TgMIC8 and contributed equally to this work. the soluble protein TgMIC3. Genetic disruption of TgMIC8

© 2010 Blackwell Publishing Ltd 2 L. Sheiner et al. ᭿

Fig. 1. A. Schematic representation of the four major microneme complexes in T. gondii, as well as of TgMIC12 and TgMIC16, when in the micronemes (top) and on the parasite’s surface (bottom). Represented is the currently known composition of the complexes, including the various proteolytic cleavage events as demonstrated in Carruthers et al. (2000) and Harper et al. (2006) for TgMIC2/M2AP, in Meissner et al. (2002), Opitz et al. (2002) and Sawmynaden et al. (2008) for TgMIC6/MIC1/MIC4, in Meissner et al. (2002) and Cerede et al. (2005) for TgMIC8/MIC3, in Hehl et al. (2000), Alexander et al. (2005), Straub et al. (2008) and Besteiro et al. (2009) for TgAMA1/RON2/RON4/RON5/RON8, in Opitz et al. (2002) for TgMIC12 and in this study for TgMIC16. B. Amino acid sequence alignment of the TMDs and CTDs of TgMIC2, TgMIC6, TgMIC12, TgAMA1, TgMIC16, TgMIC8 and TgMIC8.2. Boxed in grey are the TMDs, in green the putative rhomboid cleavage sites and in pink the motifs for traffic to the micronemes. The tryptophan signature residue boxed in yellow and the acidic residues boxed in blue are both involved in binding to aldolase. interferes with rhoptry secretion and, consequently, pre- to invade host cells (Mital et al., 2005). Gliding motility is vents formation of the moving junction (MJ) and comple- not significantly altered in the absence of TgAMA1 or tion of invasion (Kessler et al., 2008). A fourth complex, TgMIC8 (Mital et al., 2005; Kessler et al., 2008). which uniquely localizes to the MJ (Alexander et al., So far, TgMIC2 and TgMIC6 are the only TM-MICs 2005), contains the rhoptry proteins TgRON2, TgRON4, shown to play a crucial role as force-transducers during TgRON5 and TgRON8 (Alexander et al., 2005; Straub motility and invasion. TgMIC2 binds to receptor(s) on the et al., 2008; Besteiro et al., 2009) and the TM-MIC host cell surface and establishes simultaneously a con- TgAMA1, which anchors the complex to the parasite nection, via its C-terminal cytoplasmic domain (CTD), with plasma membrane (PM). Parasites lacking TgAMA1 effi- the parasite’s actomyosin system, hence powering para- ciently attach to host cells but are defective in rhoptry site motility. The CTDs of TgMIC2, TgMIC6 and other secretion, fail to create a MJ and are consequently unable members of the thrombospondin-related anonymous

© 2010 Blackwell Publishing Ltd, Molecular Microbiology Transmembrane microneme proteins modular function 3 protein (TRAP) family in Plasmodium (TRAP, CTRP and for correct trafficking of the entire complex (Harper et al., TLP) interact with aldolase, a glycolytic also 2006; Brydges et al., 2008; El Hajj et al., 2008). capable of binding to filamentous-actin (F-actin) (Busca- To gain insight into the mechanistic contribution of each glia et al., 2003; Jewett and Sibley, 2003; Heiss et al., of the MIC complexes to invasion, we have undertaken a 2008; Zheng et al., 2009). It is unknown whether other T. detailed analysis of the TM-MICs currently identified in T. gondii TM-MICs, that are part of adhesive complexes and gondii and included a new member, TgMIC16. We have exhibiting crucial functions in invasion, can as well interact searched for the presence of trafficking determinants, with aldolase and thus act as bridge molecules. assessed their susceptibility to intramembrane cleavage At the end of the penetration process, the tight interac- and their ability to interact with aldolase. The results indi- tions formed between the different MIC complexes and the cate that in contrast to TgMIC2, TgMIC6 and TgMIC12, the host cell receptors have to be disengaged to let the para- CTDs of TgMIC8, TgMIC8.2, TgAMA1 and TgMIC16 do not site freely replicate. This has been proposed to occur by carry the information for proper trafficking to the micron- proteolytic shedding of the MIC complexes from the para- emes and cannot therefore be considered as escorters. All site’s surface. Cell-based cleavage assays and studies these TM-MICs, apart from TgMIC8.2, appear to be sus- on parasites have demonstrated that critical cleavage ceptible to intramembrane cleavage and the CTDs of events takes place at a conserved motif within the luminal TgMIC6, TgMIC12 and TgAMA1 can bind to aldolase in part of the transmembrane domains of TgMIC2, TgMIC6, pull-down assays. Additionally, we have identified specific TgMIC12 and TgAMA1. The protease responsible for this residues within the CTD of TgAMA1 that are required for intramembrane cleavage was named microneme protein both association with aldolase and host cell invasion. protease 1 (MPP1) and likely corresponds to a PM Collectively these data support a model describing the rhomboid-like protease (Opitz et al., 2002; Brossier et al., involvement of TM-MICs, as part of complexes with distinct 2003; Urban and Freeman, 2003; Zhou et al., 2004; Howell and non-overlapping functions during invasion. et al., 2005). Prime candidates for this shedding activity are TgROM4 and TgROM5, which are found at the PM of the parasite (Brossier et al., 2005; Dowse et al., 2005). Results More recently, parasites depleted in TgROM4 indicate that TgMIC16 is a conserved Coccidia TM-MIC containing this protease acts as sheddase for TgMIC2 and TgAMA1 six thrombospondin repeat (TSR) domains and hence critically contributes to the creation of an apical- posterior gradient of adhesins necessary for an apical A search in the T. gondii genome database for putative new orientation of the parasite during invasion (Buguliskis microneme proteins containing TRAP family-like trans- et al., 2010).At the end of the penetration process, the tight membrane sequences led to the identification of a gene interactions formed between the different MIC complexes encoding a hypothetical protein (TGME49_089630) of 669 and the host cell receptors have to be disengaged to let the amino acids. This gene model (80.m00085) has also been parasite freely replicate. This has been proposed to occur identified by a recent in silico screen for secretory proteins by proteolytic shedding of the MIC complexes from the and was proposed to reside in an apical compartment parasite’s surface by the TgROM5 activity. (Chen et al., 2008). The amino acid sequence of the A prerequisite for successful invasion is the correct protein includes an N-terminal predicted signal , six trafficking of the MIC complexes, from the endoplasmic putative TSR type 1 domains (Fig. S1) and one TMD reticulum, where they are pre-assembled, to the micron- (transmembrane domain). This TMD is located close to the emes, where they are stored prior to invasion. Similar to C-terminal end, contains a motif reminiscent of a rhomboid other eukaryotic sorting mechanisms, some TM-MICs are cleavage site and delimits a very short C-terminal tail accurately targeted to the micronemes via recognition of a (Fig. 2). Another TMD is also predicted at the N-terminal tyrosine-based motif in the cytoplasmic CTDs (Sheiner end of the protein (TMHMM prediction programme), but and Soldati-Favre, 2008). TgMIC2 and TgMIC6 CTDs with a low probability and therefore it is not depicted as contain such a microneme targeting motif (EIEYE) and such in the schemes. A search of the available apicom- have been shown to serve as escorters for the soluble plexan genomes revealed that homologues of MICs that are part of the respective complexes (Di Cris- TGME49_089630 are present in the genomes of Neospora tina et al., 2000; Reiss et al., 2001; Opitz et al., 2002). caninum and Eimeria tenella but are absent in Hemospo- Recent studies have also revealed an important contribu- ridia, suggesting that this gene is restricted to the Coccidia. tion of some of the soluble MICs to trafficking. TgMIC1 Alignment of the amino acid sequences of these genes was shown to promote folding of TgMIC6 by serving as a (Fig. 2A) uncovered a very similar domain structure. quality control mechanism (Saouros et al., 2005) and Transient expression of this new protein carrying a Ty other soluble MICs contain pro- that act as epitope at the C-terminus revealed a micronemal localiza- luminal forward targeting elements and are indispensable tion in T. gondii tachyzoites (Fig. 2B). Reflective of this

© 2010 Blackwell Publishing Ltd, Molecular Microbiology 4 L. Sheiner et al. ᭿

Fig. 2. A. Amino acid sequences alignment of the T. gondii (T. g.)TgMIC16 (EU791458) and the homologous gene in E. tenella (E. t.) (SNAP00000003913). The predicted annotations of N. caninum and E. tenella genes await experimental confirmation. Boxed in light blue is the putative signal peptide, in grey the putative TMD and in light green the rhomboid cleavage motif. The strictly conserved residues are boxed in yellow. The six putative TSR domains are indicated. The TMD prediction was performed with the programme TMHMM. B. Double immunofluorescence analysis by confocal microscopy of intracellular parasites transiently expressing MIC16-Ty carried out with anti-Ty-1 (green) and anti-MIC4 (red), a micronemal marker. Scale bars indicates 10 mm.

localization and the nomenclature status, this protein has TgMIC8 ectodomain fused to the TM-CTD portion of been named TgMIC16 (accession number EU791458). TgMIC8.2 was able to functionally complement mic8ko Given its predicted domain structure and the presence of parasites, indicating that the TM-CTDs of these proteins a putative rhomboid cleavage site in the TMD, this protein are functionally equivalent (Kessler et al., 2008). Finally, was included in this study along with the TM-MICs that are TgMIC12, the homologue of the Eimeria TM-MICs EtMIC4 part of the four major T. gondii MIC complexes (TgAMA1, and EmTFP250 (Witcombe et al., 2004; Periz et al., TgMIC2, TgMIC6 and TgMIC8). We also included one of 2009), shown before to be susceptible to rhomboid cleav- the homologues of TgMIC8, TgMIC8.2, previously known age (Opitz et al., 2002), was also included in the com- as MIC8-like 1 (Kessler et al., 2008). A chimera of the parative analysis.

© 2010 Blackwell Publishing Ltd, Molecular Microbiology Transmembrane microneme proteins modular function 5

Multiple motifs and functions are conserved in the surface antigen 1 protein (TgSAG1), lacking its GPI TM-CTDs of the TM-MICs anchor signal (Di Cristina et al., 2000; Reiss et al., 2001; Opitz et al., 2002). From these studies it was concluded Several lines of evidence indicate that the TM-CTDs of that these three TM-MICs act as escorters, bringing the the TM-MICs play an essential role in supporting the soluble components of their respective complexes to the functionality of their respective complexes (information organelle. TgMIC8 was initially suspected to act as an regarding the composition and susceptibility to pro- escorter based on the ability of a GPI anchored TgMIC8 teolytic cleavage of these complexes is recapitulated in construct to bring TgMIC3 to the PM (Meissner et al., Fig. 1A), and therefore an alignment of the amino acid 2002). However this experiment only demonstrated that sequences of these domains was performed and care- TgMIC3 and TgMIC8 were part of the same complex fully examined (Fig. 1B). and the escorter hypothesis had to be revisited in the Previous studies on the aldolase binding capacity of the light of a recent study, which established that the soluble CTDs of TgMIC2 and other TRAP-related TM-MICs have partner TgMIC3 was correctly targeted to the micron- demonstrated the importance of both a stretch of acidic emes, even in the absence of TgMIC8 (Kessler et al., residues and a penultimate tryptophan residue in the 2008). extreme C-terminal sequence (Buscaglia et al., 2003; To assess if TgMIC8, TgMIC8.2 and TgMIC16 contain Starnes et al., 2006). TgMIC6 and TgMIC12 possess both trafficking information, their TM-CTDs were C-terminally the acidic stretch and a tryptophan residue near the fused to the SAG1 coding sequence, lacking its GPI C-terminus (Fig. 1B). The CTDs of the two TgMIC8 homo- anchoring signal, and to a Ty-1 tag epitope, and logues possess a penultimate tryptophan residue but are expressed under the control of the TgMIC2 promoter (the not of acidic nature. Conversely, TgAMA1 contains the constructs are depicted in Fig. S2). Depending on the C-terminal acidic residues, but the most C-terminal tryp- information carried by the CTDs, these chimeras were tophan is 21 residues in from the C-terminus (W520). This expected to travel through the secretory pathway and to residue lies within a FW motif that is highly conserved be secreted onto the parasite surface, either via the in the AMA1 homologues of different apicomplexans micronemes by extracellular parasites at the time of inva- (Donahue et al., 2000; Hehl et al., 2000) and is known to sion, or via the dense granules (DGs) in a constitutive be essential for invasion in P. falciparum (Treeck et al., fashion. 2009). The short TgMIC16 CTD does not exhibit any In parasites expressing pMS1tyMIC16TM-CTD or feature of the aldolase-binding motifs. pMS1tyMIC8TM-CTD, the fusion proteins accumulated in the TgAMA1, TgMIC2, TgMIC6 and TgMIC12 possess a DGs and were delivered to the parasitophorous vacuole rhomboid cleavage site, IAGG or IAGL, at a conserved (PV), as shown by colocalization with the DG marker position within the TMD, and were previously shown to be GRA3 (Fig. 3). An identical SAG1 fusion with the TM-CTD cleaved in the parasite and in in vitro cleavage assays of TgMIC8.2 (pMS1tyMIC8.2TM-CTD) accumulated in the (Opitz et al., 2002; Urban and Freeman, 2003; Brossier trans-Golgi, as it accumulated in a compartment in the et al., 2005; Dowse et al., 2005; Howell et al., 2005; Bug- proximity of cis-Golgi as shown by staining with the uliskis et al., 2010). A very similar motif is found at the marker GRASP-YFP (Pelletier et al., 2002) (Fig. 3). corresponding position within the TMD of TgMIC16 and in Because of the presence of a TMD, the chimeric proteins a different position within the TMD of TgMIC8. No rhom- were expected to accumulate at the PM. The absence of boid cleavage motif could be identified in the TMD of PM staining suggests that these proteins are cleaved TgMIC8.2. once delivered to the PM, and what is being detected is From the two motifs shown to be essential for TgMIC2 the processed form. targeting to the micronemes (Di Cristina et al., 2000), the Insight into the trafficking of TgAMA1 was performed by sequence SYHYY is not conserved in any of the CTDs of expressing its TM-CTD C-terminally fused to the SAG1 the TM-MICs analysed, whereas the motif EIEYE is and Ty-1 tag epitope, under the control of the endogenous strictly conserved in the tail of TgMIC6. A sequence promoter. The fusion pAS1tyAMA1TM-CTD localized to the resembling EIEYE is similarly positioned in the TgMIC12 DGs, and not to the PM, as previously shown for and TgAMA1 CTDs but the critical last glutamine residue pMS1tyMIC16TM-CTD and pMS1tyMIC8TM-CTD, suggesting is only present in TgMIC12. No sequence reminiscent of that this protein also undergoes proteolysis (Fig. 4). A such a targeting motif could be identified in the CTDs of series of truncated variants of TgAMA1 with an N-terminal the TgMIC8 family members or TgMIC16. His tag, under the control of the endogenous pro- moter, were also expressed. Unlike pAS1tyAMA1TM-CTD, Several TM-MICs lack trafficking signals in their CTDs pAhisAMA1DTM-CTD, encoding the ectodomain only, or The TM-CTDs of TgMIC2, TgMIC6 and TgMIC12 were pAhisAMA1DCTD, encoding the ectodomain and TMD, shown to be able to target to the micronemes the were predominantly targeted to the micronemes, as

© 2010 Blackwell Publishing Ltd, Molecular Microbiology 6 L. Sheiner et al. ᭿

Fig. 3. Localization of several stably transfected chimeras in the parasite using double immunofluorescence analysis and confocal microscopy. Anti-GRA3 or GRASP-YFP were used as dense granules and Golgi markers respectively. Scale bars indicate 5 mm. pMS1tyMIC16TM-CTD and pMS1tyMIC8TM-CTD expressing parasites were stained with anti-Ty-1 (in green) and anti-GRA3 (in red). pMS1tyMIC8.2TM-CTD was stained with anti-Ty-1 (in red) and colocalized with expression of GRASP-YFP (in green). The nucleus was stained with DAPI (in blue).

shown by colocalization with TgMIC4. The same localiza- made on the Plasmodium AMA1 and other micronemal tion was obtained for the full-length protein, pAhisAMA1 proteins (Healer et al., 2002; Treeck et al., 2006; 2009). (Fig. 4). These data suggest that the ectodomain, but not These results indicate that unlike TgMIC2, TgMIC6 and the CTD, of TgAMA1 assures correct trafficking to the TgMIC12, none of the other TM-MICs analysed here carry micronemes potentially via interaction with a yet uniden- the necessary signal in their CTD to travel to the tified protein. This is in accordance with the observations micronemes.

Fig. 4. Localization of several stably transfected chimeras in the parasite using double immunofluorescence analysis and confocal microscopy and schemes of the different constructs. Anti-MIC4 was used as micronemal marker. Scale bars indicate 5mm. pAS1tyAMA1TM-CTD expressing parasites were stained with anti-Ty-1 (in green) and colocalized with anti-GRA3 (in red). pAhisAMA1DTM-CTD, pAhisAMA1DCTD and pAhisAMA1 expressing parasites were stained with anti-his (in green) and colocalized with anti-MIC4 (in red).

© 2010 Blackwell Publishing Ltd, Molecular Microbiology Transmembrane microneme proteins modular function 7

Several TM-MICs are susceptible to cleavage within the to occur by proteolytic cleavage at a precise site within the membrane-spanning domain TMD (Howell et al., 2005; Buguliskis et al., 2010). These results indicate that pAS1tyAMA1TM-CTD, To determine if the SAG1-ty-TM-CTD chimeras were pMS1tyMIC8TM-CTD and pMS1tyMIC16TM-CTD are cleaved serving as substrates for intramembrane cleavage we likely by a rhomboid protease at the PM. generated parasites expressing SAG1 fusion constructs mutated in the predicted rhomboid cleavage sites (the Several TM-MICs bind to aldolase mutated residues are boxed in the schemes depicted in Fig. 5B–E). Analysis by indirect immunofluorescence To determine whether other TM-MICs besides TgMIC2 assay (IFA) of the corresponding transgenic parasite can interact with aldolase, we examined the ability of lines revealed that there was a dramatic change in the bacterially expressed GST–CTD fusions to bind to aldo- subcellular localization when compared with the wild- lase by in vitro pull-down assays. Purified recombinant type chimeras (Fig. 3). The mutant fusion proteins accu- rabbit aldolase was used as source of aldolase and GST- mulated at the PM and residually at the DGs (Fig. 5A), MIC2CTD and GST alone served as positive and negative suggesting that they were indeed subject to intramem- controls respectively. The sequences of the TM-MICs brane proteolysis and introduction of the mutations con- used to generate the GST-fusions are listed in Fig. S3. ferred resistance to cleavage and accumulation at the The experiment was repeated several times using inde- parasite surface. pendent purifications of each GST-fusion and reproduc- To confirm that the changes in localization coincided ibly showed that GST-MIC8.2CTD and GST-MIC16CTD were with abrogation of cleavage, Western blot analyses were unable to bind to aldolase. In contrast, significant binding performed on total lysates from transgenic parasites was monitored with GST-MIC6CTD, GST-MIC12CTD and expressing wild-type or mutated SAG1-ty-TM-CTD chi- GST-AMA1CTD (Fig. 6A), confirming previous results with meras (an additional blot can be seen on Fig. S2). TgMIC6 (Zheng et al., 2009). In the case of GST-MIC8CTD, pMS1tyMIC16TM-CTD is detectable as a processed form no conclusions could be taken regarding binding, that is no longer detected in the mutant chimera, because of aberrant migration of the protein on the gel, pMS1tyMIC16mTM-CTD, when the putative rhomboid possibly result of protein instability. cleavage site AGGI was mutated to VVLV. The size dif- It is known that mutation of the conserved tryptophan ference between the processed and non-processed residue at the C-terminus of PbTRAP, PfTRAP, PfTLP and forms suggests that this cleavage is occurring down- TgMIC2 abrogates interaction with aldolase (Buscaglia stream of the Ty-1 epitope within the TMD (Fig. 5B). et al., 2003; Jewett and Sibley, 2003; Heiss et al., 2008). Similarly, when the motif IAGG in pMS1tyMIC8TM-CTD was TgMIC6 possesses a tryptophan residue in the same mutated to IILV in pMS1tyMIC8mTM-CTD, there was a position as the one in TgMIC2 (Fig. 1B), suggesting that change in the migration pattern, compatible with the this is the residue responsible for binding to aldolase. occurrence of a proteolytic cleavage downstream of the Indeed a GST-MIC6mCTD, in which W348 was replaced by an Ty-1 epitope, within the putative cleavage motif alanine residue (MIC6W/A) (Fig. S3), showed a significant (Fig. 5C). In sharp contrast to all the other rhomboid reduction in binding to aldolase (Fig. 6B). Although there is cleavage sites identified to date in apicomplexan sub- not a tryptophan residue at the extreme C-terminus of strates, this potential cleavage motif lies close to the TgAMA1, site-directed mutagenesis was performed to cytoplasmic side of the TgMIC8 TMD (Fig. 1A). mutate F519W520, which is more distal to the C-terminus but Expression of pMS1tyMIC8.2TM-CTD led to the generation represents a highly conserved motif in all apicomplexan of two products suggesting that the protein undergoes AMA1 proteins and precedes a stretch of acidic residues in proteolytic maturation (Fig. 5D). The smaller product the TgAMA1 CTD (Fig. S3). Intriguingly, the replacement shows the same migration behaviour on SDS-PAGE as of F519W520 by AA (AMA1FW/AA) led to a significant reduction the intramembrane cleavage product observed for in the binding of GST-AMA1mCTD to aldolase (Fig. 6A). pMS1tyMIC16TM-CTD and other fusions (Fig. S2), suggest- ing that the cleavage occurs within or close to the TMD but Mutations in the CTD of TgAMA1 that block binding to there is no recognizable rhomboid cleavage site in the aldolase inhibit invasion TMD of TgMIC8.2 and therefore we could not test for rhomboid cleavage. The availability of mutant parasite strains in which the pAS1ty1AMA1TM-CTD was mainly detected in the DGs TgMIC6 and TgAMA1 genes have been disrupted by and was subject to proteolysis at a site compatible with double homologous recombination offered the opportunity intramembrane cleavage as determined by Western blot to examine the importance of the tryptophan residue in (Figs 4 and 5E). In fact shedding of TgAMA1 from the TgMIC6 and TgAMA1 for invasion (Reiss et al., 2001; parasite surface, during invasion, was previously reported Mital et al., 2005).

© 2010 Blackwell Publishing Ltd, Molecular Microbiology 8 L. Sheiner et al. ᭿

© 2010 Blackwell Publishing Ltd, Molecular Microbiology Transmembrane microneme proteins modular function 9

Fig. 5. A. Subcellular distribution of the SAG1-TM-CTD mutant chimeras by IFA and documented by confocal microscopy. Anti-GAP45 antibodies (in red), Anti-GRA3 (in red) and GRASP-YFP (in green) were used as IMC, DGs and Golgi markers respectively. The nucleus was stained with DAPI (blue). pMS1tyMIC16mTM-CTD, pMS1tyMIC8mTM-CTD (in green) and pAS1tyAMA1mTM-CTD (in red) expressing parasites were stained with anti-Ty-1. Scale bars indicate 5 mm. B–E. Analysis of the cleavage events in the different chimeras. On the top is shown the TMD of each TM-MIC in the original (wt) and mutated chimera (mut) and the residues mutated in the rhomboid cleavage motif are boxed. Below is Western blot analysis of lysates from parasites stably expressing the different pMS1tyMICTM-CTD fusion constructs. Indicated by arrows are the migration of the full (f) and shed (s) forms with indication of the molecular weight. (B) Migration of pMS1tyMIC16TM-CTD (wt) and pMS1tyMIC16mTM-CTD (mut). (C) Migration of pMS1tyMIC8TM-CTD (wt) and pMS1tyMIC8mTM-CTD (mut). (D) Migration of pMS1tyMIC8.2TM-CTD. (E) Migration of pAS1tyAMA1TM-CTD. The proteins were detected with anti-Ty-1. Molecular weight markers are indicated in kDa.

A mutant of TgMIC6, TgMIC6W/A-Ty, was generated in MIC6Ty or MIC6W/ATy restored the invasion phenotype to which the residue W348, lying in a similar position as the a level comparable to wild-type level. Gliding assays tryptophan residue involved in TgMIC2 binding to aldo- showed that mic6ko parasites are not defective in gliding lase, was converted to an alanine residue (Fig. 1B). and, as expected, the MIC6W/ATy complemented para- TgMIC6-Ty and TgMIC6W/A-Ty expressing vectors were sites also glide normally (Fig. 7C). These results suggest used to complement the mic6ko strain and the resulting that the residue W348, and therefore aldolase binding, is proteins were shown to localize to the micronemes not critical for the function of the TgMIC4–MIC1–MIC6 (Fig. 7A). Given that TgMIC6 is acting as escorter, in the complex during invasion. absence of the protein, the soluble partners of its adhe- To study whether the residues F519W520 contribute to sive complex, TgMIC1 and TgMIC4, are mistargeted to the function of the TgAMA1 during invasion we used the DGs and hence unable to participate in the invasion a previously reported TgAMA1 conditional knockout process (Reiss et al., 2001). In consequence, the parasite (ama1koi; Mital et al., 2005), in which the mic6ko mutant is virtually comparable to a triple- expression of wild-type (myc-tagged) TgAMA1 can be knockout of TgMIC6, TgMIC1 and TgMIC4 (Reiss et al., controlled by the addition of anhydrotetracycline (ATc). In 2001), in a situation parallel to the mic1ko strain, where the absence of ATc, AMA1myc is expressed in these TgMIC4 and TgMIC6 fail to traffic to the micronemes. parasites and they are fully invasive; in the presence of Consistent with the invasiveness of mic1ko (Cerede ATc, AMA1myc expression is repressed and the para- et al., 2005), mic6ko shows about a 50% reduction of sites are severely defective in invasion (Mital et al., invasion efficiency compared with the RH-2YFP strain, 2005). The ama1koi parasites were transfected with which was used as an internal standard for parasite plasmids encoding Flag-tagged wild-type or mutant fitness (Fig. 7B). Complementation of mic6ko with either TgAMA1 (AMA1WTFlag and AMA1FW/AAFlag, respec-

Fig. 6. In vitro aldolase (ADL) binding assay. A. GST pull-down assays were performed with GST alone or with the GST-fusion constructs GST-MIC2CTD, GST-MIC6CTD, GST-MIC8CTD, GST-MIC8.2CTD, GST-MIC12CTD, GST-AMA1CTD and aldolase alone. GST-MIC16CTD and GST-MIC6W/ACTD were tested separately with GST-MIC2CTD. Note that GST-MIC8CTD migration is aberrant, as indicated by an asterisk. B. GST pull-down assays were performed with GST alone or with the GST-fusion constructs GST-MIC2CTD, GST-MIC6CTD, GST-MIC6W/ACTD and aldolase alone. SDS-PAGE gel was stained with Coomassie blue. The migration of aldolase, GST and GST-MIC2CTD is indicated by arrows.

© 2010 Blackwell Publishing Ltd, Molecular Microbiology 10 L. Sheiner et al. ᭿

Fig. 7. Invasion and gliding assays of mic6ko and complemented strains. The penultimate tryptophan residue in the CTD of TgMIC6 is not critical for productive invasion. A. IFA documented by confocal microscopy of intracellular parasites deficient in TgMIC6 (mic6ko) and complemented with TgMIC6-Ty (mic6ko/MIC6Ty) or MIC6W/ATy (mic6ko/MIC6W/ATy). Colocalization with the micronemal marker TgMIC4 was assessed using antibodies anti-TgMIC4 (red) and anti-Ty-1 (green). Scale bars indicate 5 mm. B. Quantification of the relative invasion efficiency of the four parasite strains as determined by a cell invasion assay normalized to RH-YFP strain co-cultivated and used as internal standard for parasite fitness. Error bars indicate standard deviations. C. Gliding assays of mic6ko and mic6ko/MIC6W/ATy parasites. The trails were stained with anti-SAG1. The arrow indicates a trail.

tively), and independent clones expressing similar levels AMA1WTFlag was able to complement the ATc-induced WT FW/AA FW/AA of AMA1 Flag and AMA1 Flag in the presence of invasion defect in the ama1koi parasites, AMA1 Flag ATc were isolated. Both the wild-type and mutant pro- was not (Fig. 8B). These data demonstrate that the teins localized to the apical end of the parasite, as hydrophobic residues F519W520 within the CTD of shown by colocalization with M2AP, indicating proper TgAMA1 are essential for both aldolase binding and localization (Fig. 8A and data not shown). While host cell invasion.

© 2010 Blackwell Publishing Ltd, Molecular Microbiology Transmembrane microneme proteins modular function 11

Fig. 8. Invasion assay of TgAMA1-depleted parasites and complemented strains. Residues F519W520 within the CTD of TgAMA1 are critical for productive invasion. A. IFA analysis of AMA1FW/AAFlag expressed in

the AMA1 conditional knockout (ama1koi) parasites shows proper colocalization of the mutant protein with the microneme marker protein, M2AP. Two independent AMA1FW/AAFlag-expressing clones are shown (AMA1FW/AAFlag-1 and 2), as are the corresponding DIC images (left panels). Scale bars indicate 5 mm. B. Invasion assay. Host cell invasion by AMA1WTFlag, AMA1FW/AAFlag-1, FW/AA AMA1 Flag-2 and ama1koi parasites, each grown in the presence of ATc for 36 h, was measured 1 h post infection using the laser scanning cytometer-based assay. The average number of intracellular parasites per scan area is presented (two independent experiments, two replicates within each experiment), with error bars representing the SD of the mean between experiments. The invasion levels for each parasite population (expressed relative to the AMA1WTFlag parasites) are shown. The total number of intracellular parasites counted is also listed below each sample. *P < 0.05, relative to AMA1WTFlag.

Discussion ectodomains, on one hand, recruit microneme or rhoptry proteins to the complex and, in several instances also MIC complexes serve essential roles during host cell inva- interact directly with host cell receptors; and the sion, by mediating parasite attachment, MJ formation and TM-CTDs, on the other hand, contribute to targeting, pro- bridging of the host cell receptors to the actomyosin teolytic shedding and connection to the actomyosin system, hence promoting gliding and invasion. The system of the parasite. TgMIC2 and other members of the smooth transition through the various steps of the inva- TRAP family are suited to carry out these multiple tasks sion process requires a high level of coordination not only (Morahan et al., 2009). between the different MIC complexes but also between In this study, we have investigated and compared with each component of a given complex. The TM-MICs, TgMIC2, the biological properties of the TM-MICs asso- in particular are multitaskers and execute distinct func- ciated with the three other major MIC complexes known tions that are specified by their modular design. The to be involved in invasion, as well as of TgMIC12,

© 2010 Blackwell Publishing Ltd, Molecular Microbiology 12 L. Sheiner et al. ᭿

TgMIC8.2 and TgMIC16, whose functions remain to be like protease. Interestingly, while TgMIC8 appears to be established. cleaved by a rhomboid, this proteolysis occurs much Targeting to the micronemes, as demonstrated for closer to the cytoplasmic region of the TMD than in all the the rhoptries (Ngo et al., 2003; Richard et al., 2009), other TM-MICs. This may allow the direct release of the resembles post-TGN targeting in other CTD into the cytoplasm, where it can initiate a signalling (Sheiner and Soldati-Favre, 2008). Complexes of soluble cascade, as previously proposed (Kessler et al., 2008). and TM-MICs are formed in the endoplasmic reticulum Given the absence of a recognizable rhomboid cleavage and travel through the secretory pathway until they are motif in the TMD of TgMIC8.2, we could not assess the finally secreted (Reiss et al., 2001; Huynh et al., 2003). nature of the processing event. However, the cleavage Some TM-MICs have been shown to act as escorters, product runs at a size compatible with intramembrane implying that their CTDs are recognized by components of processing and TgMIC8.2-CTD can replace that of the vesicular sorting machinery (Meissner et al., 2002). TgMIC8 (Kessler et al., 2008), which is susceptible to Consistent with this idea, two micronemal targeting motifs, intramembrane cleavage. It is consequently still plausible SYHYY and EIEYE, were identified in TgMIC2 (Di Cristina that the TgMIC8.2 chimera is as well processed within the et al., 2000). The apparent absence of such motifs in the TMD. CTDs of TgAMA1, TgMIC16 and the TgMIC8 family Preventing the proteolytic cleavage by mutagenesis members is in agreement with the findings here that the had an anticipated impact on the localization of the SAG1- respective SAG1-ty-TM-CTD chimeras fail to traffic to the ty-TM-CTD chimeras. All the mutant chimeras showed a micronemes. Consequently, these TM-MICs do not func- dramatic change in subcellular localization, accumulating tion as escorters and are likely to interact, via their at the parasite’s surface, indicative of cleavage ectodomains, with other proteins that carry a determinant abrogation. In a prior study, a similar accumulation at for micronemal targeting. Consistent with this hypothesis, the parasite PM was observed for the uncleaved the chimera the chimera MIC8 fused to the CTD of Plas- SAG1TgMIC12TM-CTD mutant, which was mistargeted to modium berghei TRAP localizes to the micronemes the DGs (Opitz et al., 2002). This observation suggested (Kessler et al., 2008) although the PbTRAP TM-CTD does that MPP1 is a constitutively active rhomboid-like pro- not confer trafficking to micronemes in T. gondii (Di Cris- tease at the PM of the parasite. We cannot discriminate tina et al., 2000). Similarly, the refined analysis of between cleavage of the SAG1-ty-TM-CTD constructs at TgAMA1 clearly established that it is the ectodomain of the PM or inside the parasite, but it is likely that TgMIC16 the protein that carries the necessary traffic information to and TgMIC8 fusion constructs are cleaved by MPP1, the micronemes. Studies on AMA1 in P. falciparum led to because the corresponding non-cleaved mutants accu- the same conclusion (Healer et al., 2002; Treeck et al., mulate at the parasite’s surface. The unambiguous 2006; 2009). These observations imply that TgAMA1, assignment of each of the TM-MIC substrates to a given TgMIC8, TgMIC8.2 and TgMIC16 may belong to com- protease awaits further investigations. plexes that are composed of more than one type of Several Plasmodium TM-MIC proteins have been TM-MIC. reported to interact with the F-actin binding protein aldo- The SAG1-ty-TM-CTDs chimeras localized either to the lase and in this way bridge the host cell surface with the DGs and PV (TgMIC8, TgMIC16 and TgAMA1), or were actomyosin motor of the parasite. These proteins share retained in the Golgi (TgMIC8.2). An alignment of the the structural features characteristic of TRAP, namely an TM-CTDs of the selected TM-MICs predicted the pres- N-terminal secretion signal, a van Willebrand A-domain, ence of a rhomboid cleavage motif similar to IAGG in the one or more TSR domains, a TMD with a rhomboid cleav- TMDs of TgMIC2, TgMIC6, TgMIC12, TgAMA1 and age motif and an acidic CTD with a unique tryptophan TgMIC16. An IAGG motif is also present within the TMD of residue close to the C-terminus (Morahan et al., 2009). In TgMIC8, but it is significantly shifted within the TMD, T. gondii, TgMIC6 and TgMIC2 are the only TM-MICs closer to the CTD. No apparent rhomboid cleavage site shown to bind to aldolase (Jewett and Sibley, 2003; signature could be identified in the TMD of TgMIC8.2. Starnes et al., 2006; Zheng et al., 2009), in a model com- Consistent with cleavage at the rhomboid cleavage motif, patible with TgMIC2 redistribution along the parasite’s proteolytic processing at the expected position was surface upon invasion (Carruthers and Sibley, 1999) and observed for pMS1tyMIC8TM-CTD, pMS1tyMIC16TM-CTD and demonstrated role in motility and invasion (Huynh and pAS1tyAMA1TM-CTD chimeric proteins. To provide further Carruthers, 2006). The patches of acidic amino acids evidence for intramembrane processing by a rhomboid, constituting the aldolase within the TgMIC2 point mutations were introduced in the identified cleavage CTD (Starnes et al., 2006) are not strictly conserved in the motifs. The majority of the mutations introduced abro- TgMIC6, TgMIC12 and TgAMA1 CTDs (Fig. S4) but as gated processing and thus provided strong evidence that shown in this study, these proteins are able to bind to the chimeras are cleaved within their TMD by a rhomboid- aldolase in an in vitro pull-down assay. This suggests that

© 2010 Blackwell Publishing Ltd, Molecular Microbiology Transmembrane microneme proteins modular function 13 the composition in acidic amino acids and their precise Consistent with these findings mic6ko showed no defect location within the CTD can accommodate a level of in gliding motility, suggesting that the function of TgMIC1– variation. The second prominent feature of aldolase MIC4–MIC6 complex in invasion might be assisted via the binding is the presence of a conserved tryptophan residue formation of a macrocomplex by another TM-MIC that at the extreme C-terminus of the CTD. All the TM-MICs connects to the actomyosin system. studied here possess this residue except TgAMA1 and It remains to be determined for TgMIC12, if aldolase TgMIC16, and as shown by mutation of the residue in plays a role in its functions in vivo and if it reflects a direct TgMIC6, the residue mediates binding to aldolase. interaction with the actomyosin motor or another biologi- Although the TgMIC8 family members possess a tryp- cal role. tophan residue at the extreme C-terminus, the acidic Taken together, there is an excellent correlation patch is absent and none of these CTDs bind to aldolase between the predictions made from sequence analysis in the in vitro assay. This is in accordance with functional and the three biological properties examined experimen- analysis showing no motility defect in TgMIC8-depleted tally: presence of trafficking determinants, susceptibility to parasites (Kessler et al., 2008). In contrast, TgAMA1 is rhomboid protease cleavage and binding to aldolase. able to bind to aldolase without an extreme C-terminal Moreover the assessed properties of the TM-MICs are in tryptophan, although TgAMA1 does contain a tryptophan good accordance with their functional contribution to the just N-terminal to a patch of acidic residues (W520; Fig. S4) individual steps of the invasion process (Table 1). within an FW motif that is well conserved among AMA1 homologues in other Apicomplexans (Hehl et al., 2000). Experimental procedures Mutation of this FW motif in TgAMA1 disrupted both aldo- lase binding and invasion. This result suggests that Reagents and parasite culture TgAMA1 serves as a bridging protein that physically con- Restriction were purchased from New England nects the glideosome (via its CTD) to other components of Biolabs and secondary antibodies for Western blots and the MJ complex (via its ectodomain) and thus plays a IFA from Molecular Probes. T. gondii tachyzoites (RH critical role in the posterior translocation of the MJ strain wild-type and RHhxgprt -) were grown in human complex during invasion. However, during invasion the foreskin fibroblasts (HFF) or Vero cells in Dulbecco’s majority of the TgAMA1 is not restricted to the MJ but is modified Eagle’s medium (DMEM, Gibco) supplemented found over the parasite’s surface (Alexander et al., 2005; with 5% fetal calf serum (FCS), 2 mM glutamine and Howell et al., 2005). This suggests that there may be two 25 mgml-1 gentamicin. ama1ko parasites were cultured in pools of TgAMA1 at the parasite surface, one of which is i DMEM supplemented with 1% fetal bovine serum, bound to aldolase and is responsible for anchoring the MJ 25 mgml-1 mycophenolic acid, 50 mgml-1 xanthine and complex to the actomyosin motor. Whether the remaining 6.8 mgml-1 chloramphenicol (Mital et al., 2005). Down- fraction of TgAMA1 serves a distinct function or is simply regulation of AMA1-myc expression in intracellular available to be recruited to the MJ is unknown, but it is ama1ko parasites was achieved by incubation of infected possible that the two pools of TgAMA1 are distinguished i cells for 36 h in medium containing 1,5 mgml-1 anhy- by different post-translational modifications of their CTDs, drotetracycline (ATc, Clontech). such as phosphorylation (Treeck et al., 2009). Disappoint- ingly, our repeated efforts to monitor TgAMA1, or even TgMIC2, interaction with aldolase in the parasite by Cloning of DNA constructs co-immunoprecipitation were unfruitful. The extreme C-terminus of TgMIC12 shares a nearly For determination of MIC16 localization in T. gondii, the strictly conserved amino acid sequence with TgMIC2, and full-length gene was amplified from tachyzoite cDNA by this reflects its comparable propensity to bind to aldolase PCR using the primers 1969 and 1971 (Table S1). The in pull-down experiments. Given the fact that no functional PCR product was purified, digested with EcoRI and NsiI data are available to date on TgMIC12, the physiological and cloned into the corresponding sites in pTUB8Ty relevance of these observations is not known. (Meissner et al., 2002). TgMIC6 binds less efficiently to aldolase compared with For expression in T. gondii as N-terminal SAG1-Ty the CTDs of TgMIC2 or TgMIC12, and this can simply fusions, DNA fragments coding for the TMD and CTD of reflect the fact that fewer acidic residues are present at its MIC8, MIC8.2 and MIC16 were amplified from tachyzoite extreme C-terminus. TgMIC6 wild type or TgMIC6 carry- cDNA by PCR using the primers 1887 and 1888, 1889 ingaW348/A mutation are both able to functionally comple- and 1890, 1970 and 1972 (Table S1). MIC8TMCTD, MIC8. ment the invasion defect in parasites depleted of TgMIC6, 2TMCTD and MIC16TMCTD were digested with SalI and PacI, suggesting that the tryptophan residue may not be crucial and each fragment was cloned into the corresponding for the function of the TgMIC1–MIC4–MIC6 complex. sites in pMSAG1Ty vector, which drives expression

© 2010 Blackwell Publishing Ltd, Molecular Microbiology 14 L. Sheiner et al. ᭿

under control of the TgMIC2 promoter, originating TMCTD TMCTD TS TS TS pMSAG1tyMIC8 , pMSAG1tyMIC8.2 and ND - - + pMSAG1tyMIC16TMCTD (Di Cristina et al., 2000).

., For expression of the different AMA1 constructs under

et al control of its endogenous promoter in RH strain T. gondii, DNA fragments coding for TMD and CTD, the full-length protein, only the ectodomain (amino acids 1–456) or the Rhoptry secretion 2005) (Howell TS TS MJ formation, - + + ectodomain and TMD (amino acids 1–479) were amplified from tachyzoite cDNA by PCR using the primers 1079 and 1080, 2225 and 1080, 2247 and 2249 and 2247 and 2248 respectively (Table S1). The three last pair of primers added a 8His tag immediately after amino acid 25.

complements MIC8 TMCTD TS TS TS AMA1 was digested with XhoI and PacI and cloned in CTD - - - pMSAG1Ty vector, originating pMSAG1tyAMA1TMCTD, and the other PCR products were digested with Nsil and PacI and cloned into the corresponding sites in pROP1 vector,

secretion originating pROPhisAMA1, pROPhisAMA1DTM-CTD and TS TS TS CTD - - + pROPhisAMA1D (Soldati et al., 1998). The AMA1 pro- moter was amplified with primers 2462 and 2463 ., and cloned between KpnI and NsiI sites in the pROP1

et al vectors, originating pAhisAMA1, pAhisAMA1DTM-CTD and pAhisAMA1DCTD, or between KpnI and NsiI sites in the pMSAG1Ty vector expressing AMA1 TM and CTD, origi- ., 2001) ., 2002; Brossier nating pAS1tyAMA1TM-CTD. et al et al Generation of plasmid pSK+A/AMA1-Flag for expres- ., 2003; Urban and ., 2005; Dowse

sion of Flag-tagged TgAMA1 in the ama1koi parasites has et al Freeman, 2003; Brossier et al 2005) (Reiss (Opitz TS + + + been described elsewhere (F. Parussini, submitted). For bacterial expression, DNA fragments corresponding

., to TM-CTDs of TgMIC2, TgMIC12, TgMIC8, TgMIC8.2, W/A

et al TgMIC6, TgMIC6 , TgAMA1 and TgMIC16 were ampli- fied from tachyzoite cDNA by PCR using the primers 176 and 177, 1599 and 710, 325 and 326, 1832 and 1833, 211 ., 2002) ., 2002; Brossier and 212, 211 and 3100, 1948 and 1949 and 2001 and et al et al 2002 respectively (Table S1). PCR products were purified ., 2003; Urban and ., 2005; Dowse using the Easy Pure-DNA Purification Kit (Biozym). et al Freeman, 2003; Brossier et al 2005) TMCTD TMCTD W/ATMCTD (Opitz (Opitz TS MIC2 , MIC6 and MIC6 were digested + + + with EcoRI and SalI, MIC12 TMCTD and AMA1 TMCTD with EcoRI and XhoI, MIC8TMCTD with BamHI and MIC8.2TMCTD TMCTD ., and MIC16 with BamHI and XhoI. Each fragment

et al was cloned into the corresponding sites in the pGEX4T1

., 2000) vector to generate N-terminal GST-fusions. ., 2003; Jewett et al et al ., 2002; Brossier Mutated constructs et al ., 2008) ., 2003; Urban and ., 2005; Dowse To mutate the motif FW to AA on the AMA1CTD of the (Di Cristina and Sibley, 2003; Heiss et al et al Freeman, 2003; Brossier et al 2005) TMCTD (Buscaglia (Opitz plasmid pGEX4T1-AMA1 the primers 1830 and 1831 Motility, Host cell binding ND Host cell binding Rhoptry ++ + + were used in a site-directed mutagenesis reaction using the commercial QuikChange II Site-Directed Mutagenesis Kit (Stratagen) according to the manufacturer’s instruc- tions. Similarly, the residues AGG, YTG, AG or AGGI were

List of primers used in this study. mutated to ILV, VL or VVLV in the TMDs of MIC8, MIC8.2 and MIC16 in the plasmids pMSAG1tyMIC8TMCTD, pMSAG1tyMIC8.2TMCTD and pMSAG1tyMIC16TMCTD, invasion cleavage Table 1. TM-MICRole in motility and MIC2ND, not determined; TS, this study. MIC12 MIC6 MIC8 MIC8.2 AMA1 MIC16 Trafficking determinant Binding to aldolase for rhomboid respectively, using the primers 2003 and 2004, 2005 and

© 2010 Blackwell Publishing Ltd, Molecular Microbiology Transmembrane microneme proteins modular function 15

2006 or 2226 and 2227 respectively. To mutate F519W520 to (Roos et al., 1997) plasmids. Parasites were electropo- AA on the AMA1 cytosolic tail in pSK+A/AMA1-Flag, rated at 2 kV, 25 mF, 48 V using a BTX electroporator primers TgAMA1FW/AA.f and TgAMA1FW/AA.r (Table S1) were (Harvard biosciences, Holliston, MA, USA) before being used for site-directed mutagenesis as described above, added to a monolayer of HFF cells in the presence generating the vector pSK+A/AMA1FW/AAFlag. of mycophenolic acid/xanthine. Selection with 1 mM pyrimethamine was initiated 24 h later and continued for 7–10 days, after which resistant clones were isolated by Protein expression and purification limiting dilution. pGEX-4T1 vectors encoding the MIC2, MIC12, MIC6, MIC8, MIC8.2, MIC16 or AMA1 TMCTD GST-fusion Western blotting proteins were transformed into the BL21 strain (Novagen, Madison, WI). Protein expres- For AMA1 Western blots, 108 parasites were harvested sion was induced using 1 mM isopropyl-beta-d- after complete lysis of the host cell monolayer and thiogalactopyranoside (IPTG) for 4 h at 37°C. Bacterial extracted for 30 min at 4°C in 1 ml of RIPA buffer [50 mM pellets were resuspended in 1¥ PBS supplemented with Tris-HCl, pH 7.5; 1% (vol/vol) Triton X-100; 0.5% (vol/vol) -1 -1 -1 1mgml of Lysozym, 10 mgml DNAse, 20 mgml sodium deoxycholate; 0.2% (wt/vol) SDS; 100 mM NaCl2; RNAse and 1 mM PMSF, and were allowed to homog- 5 mM EDTA] containing protease inhibitors (Sigma enize for 30 min at 4°C, following which, cells were dis- P8340), added directly to SDS-PAGE-loading buffer and rupted by five consecutive cycles freeze/thaw. After boiled for 10 min. For all other samples, extracts from centrifugation (30 min, 30 000 r.p.m.), the supernatant 2 ¥ 107 parasites were prepared in 1¥ PBS by five con- containing the soluble GST-fusions was collected, and secutive freeze/thaw cycles with intermediate homogeni- purified using GSH-beads (Glutathione Sepharose 4 zation, following two consecutive sonications, and the Fast Flow, Amersham) according to the manufactures suspension was boiled in SDS-PAGE-loading buffer con- advice. taining 100 mM DTT. SDS-PAGE was performed using standard methods. Separated proteins were trans- ferred to a nitrocellulose membrane using a semidry GST-fusion protein pull-down experiment electroblotter. Western blots were performed using anti- Glutathione-sepharose beads were incubated with 0.5 mg Ty1 mAb (Bastin et al., 1996), anti-AMA1 mAb (B3.90, of GST-fusion proteins or GST for 1 h at 4°C. Beads were Donahue et al., 2000), anti-myc mAb (Mital et al., 2005) washed twice with PBS and once with buffer XB (50 mM and anti-Flag mAb (Sigma F3165) in 5% non-fat milk

KCL, 20 mM HEPES, 2 mM MgCL2, 0.2 mM EDTA, 0.2% powder in 1¥ PBS. As secondary antibody, a peroxidase- Tween 2, pH 7.7), prior to the addition of approximately conjugated goat anti-mouse or anti-rabbit antibody was 400 mg of recombinant aldolase (Sigma). Aldolase was used (Molecular Probes, Paisley, UK). Bound antibodies incubated with the GST-fusion purified proteins for 4 h at were visualized using either the ECL system (Amersham 4°C, washed five times with buffer XB, and bound proteins Corp) or with SuperSignalTM West Pico chemiluminescent were eluted using SDS-PAGE-loading buffer supple- substrates (Pierce). mented with 100 mM DTT.

IFA and confocal microscopy Parasite transfection and selection of clonal stable lines All manipulations were carried out at room temperature. Parasites transfection was performed by electroporation Intracellular parasites grown in HFF seeded on glass cov- as previously described (Soldati and Boothroyd, 1993). erslips were fixed with 4% paraformaldehyde (PFA) for The HXGPRT gene was used as a positive selectable 10 min or 4% PFA/0.002% glutaraldehyde for 10 min. Fol- marker in the presence of mycophenolic acid (25 mg ml-1) lowing fixation, slides were quenched in 1¥ PBS-0.1 M and xanthine (50 mg ml-1) as described previously glycine. Cells were then permeabilized in 1¥ PBS/0.2% (Donald et al., 1996). Briefly, freshly released parasites Triton X-100 (PBS/Triton) for 20 min and blocked in the (5 ¥ 107) of the RHhxgprt strain were resuspended in same buffer supplemented with 2% FCS (PBS/Triton/ cytomix buffer in the presence of 50–80 mg of linearized FCS). Slides were incubated for 60 min with the primary plasmid carrying the selectable marker gene and the antibodies anti-GAP45, anti-MIC4 (Brecht et al., 2001), expression cassette containing the DNA sequences. anti-Myc, anti-GRA3 (kindly provided by JF Dubremetz) or

Transfection of ama1koi conditional knockout parasites anti-Ty1 diluted in PBS/Triton/FCS, washed and incu- was carried out by electroporation using 2.6 ¥ 107 freshly bated for 40 min with Alexa488- or Alexa594-conjugated released parasites, 100 mgofpSK+A/AMA1-Flag or goat anti-mouse or goat anti-rabbit IgGs diluted in PBS/ pSK+A/AMA1FW/AAFlag, and 6 mg of pDHFR*.Tsc3ABP Triton/FCS. DAPI staining was performed with a concen-

© 2010 Blackwell Publishing Ltd, Molecular Microbiology 16 L. Sheiner et al. ᭿ tration of 0.1 mg of DAPI per millilitre of 1¥ PBS for 5 min fixed, blocked and labelled with an anti-SAG1 antibody incubation before slides were mounted in Fluoromount G (mAb GII-9; Argene, North Massapequa, USA) followed (Southern Biotech) and stored at 4°C in the dark. Micro- by an R-phycoerythrin-conjugated secondary antibody graphs were obtained on a Zeiss Axioskop 2 equipped (‘orange,’ DAKO, Carpenteria, USA). Samples were then with an Axiocam colour CCD camera. Images where permeabilized, blocked and labelled with anti-SAG1 fol- recorded and treated on computer through the AxioVi- lowed by an Alexa647-conjugated secondary antibody sion™ software. Confocal images were collected with a (‘red,’ Molecular Probes). Samples were analysed on a Leica laser scanning confocal microscope (TCS-NTDM/ CompuCyte Laser Scanning Cytometer equipped with IRB) using a 63 Plan-Apo objective with NA 1.40. Optical a BX50 upright fluorescence microscope (Olympus sections were recorded at 250 nm per vertical step and America, Melville, USA), 20¥ objective (N.A. 0.5), argon four times averaging. ion (488 nm) and helium/neon (633 nm) lasers and three filter blocks/photomultiplier tubes [530–555 nm (green), 600–640 nm (orange), and 650 nm (long-red)]. Data were Cell invasion assays acquired and analysed using Wincyte 3.4 Software (Com- Comparison of different T. gondii strains for invasion effi- puCyte, Cambridge, USA). Red parasites were counted to ciency was done using an RH-2YFP strain as internal determine the total number of parasites per field. The standard. A confluent 60 mm dish of HFF was heavily number of orange, extracellular parasites was counted infected with a mixture of the strain of interest and and subtracted from the total to calculate the number of RH-2YFP parasites. Some hours later the dish was invaded parasites per field. One-way ANOVA and Dun- washed to remove any non-invaded parasites. Two days nett’s multiple comparison post test were used to deter- later parasites egressed from their host cells and were mine the significance of differences between groups. collected by centrifugation at 240 g, room temperature for P-values of less than 0.05 were considered significant. 10 min and resuspended in 5 ml of culture medium (DMEM complemented with 2 mM L-glutamine, 5% FCS and Gliding motility assay 25 mgml-1 gentamicin) preheated to 37°C. From this sus- pension a 1:10 dilution was made in preheated medium, Freshly egressed tachyzoites were filtered, pelleted and the ratio of non-YFP to YFP parasites was determined in a resuspended in calcium-saline containing 1 mM of ionomy- Neubauer chamber (ratio between 0.8 and 7), and 500 ml cin. The suspension was deposited on coverslips previ- were inoculated into a well on a 24-well IFA plate. Invasion ously coated with poly-L-lysine (3 h at room temperature). was allowed to take place for 1 h at 37°C; then the wells Parasites were fixed with PAF/GA, and IFA using the were washed two times in CM-PBS (1 mM CaCl2, 0.5 mM a-SAG1 antibody was performed to visualize the trails.

MgCl2 in PBS) and refilled with fresh medium. The plate was incubated for another 24–32 h in order for the para- sites to devide. Afterwards cells were fixed with 4% Acknowledgements paraformaldehyde for 20 min, followed by 3 min incubation We thank Anne Kelsen for providing expert technical support. with 0.1 M glycine in PBS to quench the reaction and This work is part of the activities of the BioMalPar European subjected to an IFA. Fixed cells were permeabilized with Network of Excellence supported by a European grant 0.2% Triton X-100 in PBS for 20 min and blocked in 2% (LSHP-CT-2004-503578) from the Priority 1 ‘Life Sciences, bovine serum albumin, 0.2% Triton X-100 in PBS for Genomics and Biotechnology for Health’ in the sixth Frame- work Program, from supports to DS by the Swiss National 20 min. The cells were then stained with rabbit anti-TgMLC Foundation and the Howard Hughes Medical Institute. Addi- antibodies followed by Alexa594 goat anti-rabbit antibod- tional funding was provided by USPHS grant AI063276 ies (Molecular Probes). Total number of parasite vacuoles (GEW). JMS is a recipient of the EU-funded Marie Curie and RH-2YFP parasite vacuoles were counted on 20 Action MalParTraining (MEST-CT-2005-020492), ‘The chal- microscopic fields on each IFA-slide with a minimum of 750 lenge of malaria in the post-genomic Era’. vacuoles in total per slide. Only vacuoles containing at least two parasites were counted. The ratio of YFP to References non-YFP vacuoles was calculated and compared with the ratio obtained from live parasites at the beginning of the Alexander, D.L., Mital, J., Ward, G.E., Bradley, P., and Boo- experiment. Each experiment has been repeated six times. throyd, J.C. (2005) Identification of the moving junction Alternatively, host cell invasion was measured using a complex of Toxoplasma gondii: a collaboration between distinct secretory organelles. PLoS Pathog 1: e17. laser scanning cytometer-based assay (Mital et al., 2006). Bastin, P., Bagherzadeh, Z., Matthews, K.R., and Gull, K. Briefly, parasites grown for 36 h in the presence of ATc (1996) A novel epitope tag system to study protein target- were harvested, added to HFF monolayers and incubated ing and organelle biogenesis in brucei. Mol at 37°C. One hour post infection, the coverslips were Biochem Parasitol 77: 235–239.

© 2010 Blackwell Publishing Ltd, Molecular Microbiology Transmembrane microneme proteins modular function 17

Besteiro, S., Michelin, A., Poncet, J., Dubremetz, J.F., and (2000). The Toxoplasma homolog of Plasmodium apical Lebrun, M. (2009) Export of a Toxoplasma gondii rhoptry membrane antigen-1 (AMA-1) is a microneme protein neck protein complex at the host to form the secreted in response to elevated intracellular calcium moving junction during invasion. PLoS Pathog 5: levels. Mol Biochem Parasitol 111: 15–30. e1000309. Donald, R.G., Carter, D., Ullman, B., and Roos, D.S. (1996) Blumenschein, T.M., Friedrich, N., Childs, R.A., Saouros, S., Insertional tagging, cloning, and expression of the Toxo- Carpenter, E.P., Campanero-Rhodes, M.A., et al. (2007) plasma gondii hypoxanthine-xanthine-guanine phosphori- Atomic resolution insight into host cell recognition by Toxo- bosyltransferase gene. Use as a selectable marker for plasma gondii. EMBO J 26: 2808–2820. stable transformation. J Biol Chem 271: 14010–14019. Brecht, S., Carruthers, V.B., Ferguson, D.J., Giddings, O.K., Dowse, T.J., Pascall, J.C., Brown, K.D., and Soldati, D. Wang, G., Jakle, U., et al. (2001) The toxoplasma micron- (2005) Apicomplexan rhomboids have a potential role in emal protein MIC4 is an adhesin composed of six con- microneme protein cleavage during host cell invasion. Int J served apple domains. J Biol Chem 276: 4119–4127. Parasitol 35: 747–756. Brossier, F., Jewett, T.J., Lovett, J.L., and Sibley, L.D. (2003) El Hajj, H., Papoin, J., Cerede, O., Garcia-Reguet, N., Soete, C-terminal processing of the toxoplasma protein MIC2 is M., Dubremetz, J.F., and Lebrun, M. (2008) Molecular essential for invasion into host cells. J Biol Chem 278: signals in the trafficking of the Toxoplasma gondii protein 6229–6234. MIC3 to the micronemes. Eukaryot Cell 7: 1019–1028. Brossier, F., Jewett, T.J., Sibley, L.D., and Urban, S. (2005) A Harper, J.M., Huynh, M.H., Coppens, I., Parussini, F., spatially localized rhomboid protease cleaves cell surface Moreno, S., and Carruthers, V.B. (2006) A cleavable adhesins essential for invasion by Toxoplasma. Proc Natl propeptide influences Toxoplasma infection by facilitating Acad Sci USA 102: 4146–4151. the trafficking and secretion of the TgMIC2-M2AP invasion Brydges, S.D., Harper, J.M., Parussini, F., Coppens, I., and complex. Mol Biol Cell 17: 4551–4563. Carruthers, V.B. (2008) A transient forward-targeting Healer, J., Crawford, S., Ralph, S., McFadden, G., and element for microneme-regulated secretion in Toxoplasma Cowman, A.F. (2002). Independent translocation of two gondii. Biol Cell 100: 253–264. micronemal proteins in developing Plasmodium falciparum Buguliskis, J.S., Brossier, F., Shuman, J., and Sibley, L.D. merozoites. Infect Immun 70: 5751–5758. (2010) Rhomboid 4 (ROM4) affects the processing of Hehl, A.B., Lekutis, C., Grigg, M.E., Bradley, P.J., Dubremetz, surface adhesins and facilitates host cell invasion by Toxo- J.F., Ortega-Barria, E., and Boothroyd, J.C. (2000) Toxo- plasma gondii. PLoS Pathog 6: e1000858. plasma gondii homologue of plasmodium apical membrane Buscaglia, C.A., Coppens, I., Hol, W.G., and Nussenzweig, V. antigen 1 is involved in invasion of host cells. Infect Immun (2003) Sites of interaction between aldolase and 68: 7078–7086. thrombospondin-related anonymous protein in Heiss, K., Nie, H., Kumar, S., Daly, T.M., Bergman, L.W., and plasmodium. Mol Biol Cell 14: 4947–4957. Matuschewski, K. (2008) Functional characterization of a Carruthers, V., and Boothroyd, J.C. (2007) Pulling together: redundant Plasmodium TRAP-family invasin, TRAP-like an integrated model of Toxoplasma cell invasion. Curr Opin protein (TLP), by aldolase binding and a genetic comple- Microbiol 10: 83–89. mentation test. Eukaryot Cell 7: 1062–1070. Carruthers, V.B., and Sibley, L.D. (1997) Sequential protein Howell, S.A., Hackett, F., Jongco, A.M., Withers-Martinez, C., secretion from three distinct organelles of Toxoplasma Kim, K., Carruthers, V.B., and Blackman, M.J. (2005) Dis- gondii accompanies invasion of human fibroblasts. Eur J tinct mechanisms govern proteolytic shedding of a key Cell Biol 73: 114–123. invasion protein in apicomplexan pathogens. Mol Microbiol Carruthers, V.B., and Sibley, L.D. (1999) Mobilization of intra- 57: 1342–1356. cellular calcium stimulates microneme discharge in Toxo- Huynh, M.H., and Carruthers, V.B. (2006) Toxoplasma MIC2 plasma gondii. Mol Microbiol 31: 421–428. is a major determinant of invasion and virulence. PLoS Carruthers, V.B., Sherman, G.D., and Sibley, L.D. (2000) The Pathog 2: e84. Toxoplasma adhesive protein MIC2 is proteolytically pro- Huynh, M.H., Rabenau, K.E., Harper, J.M., Beatty, W.L., cessed at multiple sites by two parasite-derived . Sibley, L.D., and Carruthers, V.B. (2003) Rapid invasion of J Biol Chem 275: 14346–14353. host cells by Toxoplasma requires secretion of the MIC2- Cerede, O., Dubremetz, J.F., Soete, M., Deslee, D., Vial, H., M2AP adhesive protein complex. EMBO J 22: 2082–2090. Bout, D., and Lebrun, M. (2005) Synergistic role of micron- Jewett, T.J., and Sibley, L.D. (2003) Aldolase forms a bridge emal proteins in Toxoplasma gondii virulence. J Exp Med between cell surface adhesins and the actin cytoskeleton 201: 453–463. in apicomplexan parasites. Mol Cell 11: 885–894. Chen, Z., Harb, O.S., and Roos, D.S. (2008) In silico identi- Jewett, T.J., and Sibley, L.D. (2004) The toxoplasma proteins fication of specialized secretory-organelle proteins in api- MIC2 and M2AP form a hexameric complex necessary for complexan parasites and in vivo validation in Toxoplasma intracellular survival. J Biol Chem 279: 9362–9369. gondii. PLoS ONE 3: e3611. Kessler, H., Herm-Götz, A., Hegge, S., Rauch, M., Soldati- Di Cristina, M., Spaccapelo, R., Soldati, D., Bistoni, F., and Favre, D., Frischknecht, F., and Meissner, M. (2008) Crisanti, A. (2000) Two conserved amino acid motifs Microneme protein 8 is a new essential invasion factor in mediate protein targeting to the micronemes of the apicom- Toxoplasma gondii. J Cell Sci 121: 947–956. plexan parasite Toxoplasma gondii. Mol Cell Biol 20: 7332– Meissner, M., Reiss, M., Viebig, N., Carruthers, V.B., Toursel, 7341. C., Tomavo, S., et al. (2002) A family of transmembrane Donahue, C.G., Carruthers, V.B., Gilk, S.D., and Ward, G.E. microneme proteins of Toxoplasma gondii contain EGF-like

© 2010 Blackwell Publishing Ltd, Molecular Microbiology 18 L. Sheiner et al. ᭿

domains and function as escorters. J Cell Sci 115: 563– Soldati, D., Lassen, A., Dubremetz, J.-F., and Boothroyd, J.C. 574. (1998) Processing of Toxoplasma ROP1 protein in nascent Mital, J., Meissner, M., Soldati, D., and Ward, G.E. (2005) rhoptries. Mol Biochem Parasitol 96: 37–48. Conditional expression of Toxoplasma gondii apical mem- Starnes, G.L., Jewett, T.J., Carruthers, V.B., and Sibley, L.D. brane antigen-1 (TgAMA1) demonstrates that TgAMA1 (2006) Two separate, conserved acidic amino acid plays a critical role in host cell invasion. Mol Biol Cell 16: domains within the Toxoplasma gondii MIC2 cytoplasmic 4341–4349. tail are required for parasite survival. J Biol Chem 281: Mital, J., Schwarz, J., Taatjes, D.J., and Ward, G.E. (2006) 30745–30754. Laser scanning cytometer-based assays for measuring Straub, K.W., Cheng, S.J., Sohn, C.S., and Bradley, P.J. host cell attachment and invasion by the human pathogen (2008) Novel components of the Apicomplexan moving Toxoplasma gondii. Cytometry A 69: 13–19. junction reveal conserved and coccidia-restricted Morahan, B.J., Wang, L., and Coppel, R.L. (2009) No TRAP, elements. Cell Microbiol 4: 590–603. no invasion. Trends Parasitol 25: 77–84. Tan, K., Duquette, M., Liu, J.H., Lawler, J., and Wang, J.H. Ngo, H.M., Yang, M., Paprotka, K., Pypaert, M., Hoppe, H., (2008) The crystal structure of the heparin-binding -N and Joiner, K.A. (2003) AP-1 in Toxoplasma gondii medi- domain of f-spondin. J Mol Biol 381: 1213–1223. ates biogenesis of the rhoptry secretory organelle from a Tossavainen, H., Pihlajamaa, T., Huttunen, T.K., Raulo, E., post-Golgi compartment. J Biol Chem 278: 5343–5352. Rauvala, H., Permi, P., and Kilpelainen, I. (2006) The Opitz, C., Cristina, M.D., Reiss, M., Ruppert, T., Crisanti, A., layered fold of the TSR domain of P. falciparum TRAP and Soldati, D. (2002) Intramembrane cleavage of micron- contains a heparin binding site. Protein Sci 15: 1760– eme proteins at the surface of the apicomplexan parasite 1768. Toxoplasma gondii. EMBO J 21: 1577–1585. Treeck, M., Struck, N.S., Haase, S., Langer, C., Herrmann, Pelletier, L., Stern, C.A., Pypaert, M., Sheff, D., Ngo, H.M., S., Healer, J., et al. (2006) A conserved region in the EBL Roper, N., et al. (2002) Golgi biogenesis in Toxoplasma proteins is implicated in microneme targeting of the malaria gondii. Nature 418: 548–552. parasite Plasmodium falciparum. J Biol Chem 281: 31995– Periz, J., Ryan, R., Blake, D.P., and Tomley, F.M. (2009) 32003. Eimeria tenella microneme protein EtMIC4: capture of the Treeck, M., Zacherl, S., Herrmann, S., Cabrera, A., Kono, M., full-length transcribed sequence and comparison with Struck, N.S., et al. (2009) Functional analysis of the other microneme proteins. Parasitol Res 104: 717–721. leading malaria vaccine candidate AMA-1 reveals an Rabenau, K.E., Sohrabi, A., Tripathy, A., Reitter, C., Ajioka, essential role for the cytoplasmic domain in the invasion J.W., Tomley, F.M., and Carruthers, V.B. (2001) TgM2AP process. PLoS Pathog 5: e1000322. participates in Toxoplasma gondii invasion of host cells and Urban, S., and Freeman, M. (2003) Substrate specificity of is tightly associated with the adhesive protein TgMIC2. Mol rhomboid intramembrane proteases is governed by helix- Microbiol 41: 537–547. breaking residues in the substrate transmembrane domain. Reiss, M., Viebig, N., Brecht, S., Fourmaux, M.N., Soete, M., Mol Cell 11: 1425–1434. Cristina, M.D., et al. (2001) Identification and characteriza- Witcombe, D.M., Ferguson, D.J., Belli, S.I., Wallach, M.G., tion of an escorter for two secretory adhesins in Toxo- and Smith, N.C. (2004) Eimeria maxima TRAP family plasma gondii. J Cell Biol 152: 563–578. protein EmTFP250: subcellular localisation and induction Richard, D., Kats, L.M., Langer, C., Black, C.G., Mitri, K., of immune responses by immunisation with a recombi- Boddey, J.A., et al. (2009) Identification of rhoptry traffick- nant C-terminal derivative. Int J Parasitol 34: 861– ing determinants and evidence for a novel sorting mecha- 872. nism in the malaria parasite Plasmodium falciparum. PLoS Zheng, B., He, A., Gan, M., Li, Z., He, H., and Zhan, X. (2009) Pathog 5: e1000328. MIC6 associates with aldolase in host cell invasion by Roos, D.S., Sullivan, W.J., Striepen, B., Bohne, W., and Toxoplasma gondii. Parasitol Res 2: 441–445. Donald, R.G. (1997) Tagging genes and trapping promot- Zhou, X.W., Blackman, M.J., Howell, S.A., and Carruthers, ers in Toxoplasma gondii by insertional mutagenesis. V.B. (2004) Proteomic analysis of cleavage events reveals Methods 13: 112–122. a dynamic two-step mechanism for proteolysis of a key Saouros, S., Edwards-Jones, B., Reiss, M., Sawmynaden, parasite adhesive complex. Mol Cell Proteomics 3: 565– K., Cota, E., Simpson, P., et al. (2005) A novel galectin-like 576. domain from Toxoplasma gondii micronemal protein 1 assists the folding, assembly, and transport of a cell adhe- sion complex. J Biol Chem 280: 38583–38591. Supporting information Sawmynaden, K., Saouros, S., Friedrich, N., Marchant, J., Simpson, P., Bleijlevens, B., et al. (2008) Structural insights Additional supporting information may be found in the into microneme protein assembly reveal a new mode of online version of this article. EGF domain recognition. EMBO Rep 9: 1149–1155. Please note: Wiley-Blackwell are not responsible for the Sheiner, L., and Soldati-Favre, D. (2008) Protein trafficking inside Toxoplasma gondii. Traffic 9: 636–646. content or functionality of any supporting materials sup- Soldati, D., and Boothroyd, J.C. (1993) Transient transfection plied by the authors. Any queries (other than missing and expression in the obligate intracellular parasite Toxo- material) should be directed to the corresponding author plasma gondii. Science (New York, NY) 260: 349–352. for the article.

© 2010 Blackwell Publishing Ltd, Molecular Microbiology