Next Article in Journal
Astrocytes in Neurodegenerative Diseases: A Perspective from Tauopathy and α-Synucleinopathy
Next Article in Special Issue
Secreted Effectors Modulating Immune Responses to Toxoplasma gondii
Previous Article in Journal
The Epidemiology and Global Burden of Atopic Dermatitis: A Narrative Review
Previous Article in Special Issue
Do All Coccidia Follow the Same Trafficking Rules?
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Protein Sorting in Plasmodium Falciparum

by
D.C. Ghislaine Mayer
Department of Biology, Manhattan College, Riverdale, New York, NY 10471, USA
Submission received: 30 June 2021 / Revised: 4 September 2021 / Accepted: 4 September 2021 / Published: 9 September 2021

Abstract

:
Plasmodium falciparum is a unicellular eukaryote with a very polarized secretory system composed of micronemes rhoptries and dense granules that are required for host cell invasion. P. falciparum, like its relative T. gondii, uses the endolysosomal system to produce the secretory organelles and to ingest host cell proteins. The parasite also has an apicoplast, a secondary endosymbiotic organelle, which depends on vesicular trafficking for appropriate incorporation of nuclear-encoded proteins into the apicoplast. Recently, the central molecules responsible for sorting and trafficking in P. falciparum and T. gondii have been characterized. From these studies, it is now evident that P. falciparum has repurposed the molecules of the endosomal system to the secretory pathway. Additionally, the sorting and vesicular trafficking mechanism seem to be conserved among apicomplexans. This review described the most recent findings on the molecular mechanisms of protein sorting and vesicular trafficking in P. falciparum and revealed that P. falciparum has an amazing secretory machinery that has been cleverly modified to its intracellular lifestyle.

1. Introduction

Plasmodium falciparum is a protozoan parasite that is responsible for millions of infections resulting in malaria, a devastating disease currently prevalent in sub-Saharan Africa. Malaria continues to be endemic in 87 countries with approximately 229 million cases and 2–3 million deaths [1]. Plasmodium falciparum is an obligate intracellular microbe that infects human erythrocytes. Obligate intracellular microbes use diverse mechanisms to hijack their host cellular processes. Following uptake, most intracellular microbes take over the host organelles to create distinctive and unique microenvironments. Unlike other intracellular microbes, P. falciparum has the unique challenge of residing and developing within a cell devoid of organelles.
Inside the erythrocyte, while the parasite resides within the parasitophorous vacuole, it orchestrates the drastic modification of the erythrocyte. After erythrocyte invasion, the composition of the erythrocyte membrane as well as its permeability and rigidity are radically altered [2]. All the symptoms associated with malaria are due to the invasion, intracellular development, and egress of P. falciparum merozoites into the host’s erythrocytes [3]. Once inside the erythrocyte, the parasite undergoes asexual reproduction producing 8–26 daughter merozoites that are released to invade fresh erythrocytes [4]. The life cycle of the parasite requires the trafficking of a large number of proteins to the erythrocyte, which are necessary for the complete takeover of the erythrocyte. To enable interactions with the erythrocyte, P. falciparum transports virulence proteins using de novo specialized secretion systems. The trafficking of virulence factors and their insertions into the erythrocyte membrane at the knob structures are key events in the development of the pathologies associated with severe malaria, both cerebral and placental [5]. Therefore, the pathogenesis P. falciparum is dependent on the mechanism of protein sorting and trafficking during the intraerythrocytic stage.
Protein sorting is the process by which proteins are moved to their proper destinations inside and outside the cell [6]. In eukaryotes, proteins are directed to the lumen of specific organelles, diverse intracellular membranes, the plasma membrane, or outside of the cell by the process of secretion [6]. The protein targeting process is controlled by specific sequences within the protein itself [7,8]. In order to successfully replicate inside the erythrocyte, P. falciparum needs to correctly sort and traffic its newly-made proteins to all of its organelles. In addition to trafficking proteins within its cell, the parasite ships proteins beyond its plasma membrane, into both the parasitophorous vacuole and into the host erythrocyte cytoplasm and plasma membrane [9,10]. Moreover, the parasite possesses a food vacuole, an acidic organelle where the host cell hemoglobin is digested to amino acids. The food vacuole is also the site of action of the antimalarials chloroquine and quinoline [11]. In P. faclciparum, additional organelles named mononemes and exonemes have also been described [12]. The latter seems to play a role in the parasite egress [12].

2. Protein Sorting to ER and Golgi

Akin to all eukaryotes, P. falciparum proteins that are trafficked to the various organelles contain a canonical signal peptide, a short sequence of hydrophobic amino acids located at the amino terminus. The signal peptide indicates the co-translational transport of these proteins into the endoplasmic reticulum (ER). In P. falciparum, the ER is located near the nucleus and appears as ‘‘horn-like’’ projections [8]. Except for the apicoplast-bound proteins, the parasite organellar proteins are transported to the Golgi apparatus [13]. In most eukaryotes, the Golgi apparatus is organized into biochemically well-defined compartments called cisternae termed the cis-, medial-, and trans-Golgi. In mammalian cells, these cisternae are stacked or in close apposition to each other. However, in the budding yeast, Saccharomyces cerevisiae, the Golgi cisternae are unstacked [14]. Plasmodium falciparum seems to have a simplified, unstacked Golgi apparatus. N-linked glycosylation, a process that is finalized in the cis-Golgi, does not occur in P. falciparum [15]. Several ultrastructural studies involving serial sectioning and 3-D reconstruction analysis of P. falciparum-infected erythrocytes have not determined the presence of stacked Golgi cisternae in the parasite [16,17,18]. In addition, indirect immunofluorescent studies using antibodies against cis- and trans-Golgi resident proteins of P. falciparum also support the absence of stacked cisternae in P. falciparum [19,20]. Therefore, in P. falciparum, the protein sorting apparatus from the ER to Golgi appears to be reduced.

3. Protein Sorting to the Apicoplast

In the mid-1990s, a unique organelle characterized by the presence of a vestigial, non-photosynthetic plastid was discovered in P. falciparum [21,22,23]. The apicoplast in P. falciparum seems to be positioned on a small projection of the ER, but it is not clear whether it is part of the ER [13]. However, current evidence suggests that the apicoplast is separate from the cis-Golgi [13]. In addition, the apicoplast appears to be located upstream of the cis-Golgi (Figure 1). The apicoplast is surrounded by four membranes and is thought to be the result of secondary endosymbiosis [24]. It is the site of quite a few metabolic pathways, such as heme, isoprenoids, and fatty acids syntheses [25]. It is thought that the outermost membrane of the apicoplast is part of the endomembrane system [24]. Most of the resident apicoplast proteins are encoded by genes located in the parasite nucleus [26,27]. These proteins are translocated across the four membranes by means of a two-part N-terminal extension that has been shown to be both necessary and sufficient for sorting to the apicoplast [24]. The first segment of the N-terminal exten sion sequence is similar to a classical signal peptide. It is responsible for the translocation into the secretory pathway. On the other hand, the second segment shares homology to chloroplast transit peptides and is needed for entering the apicoplast [28].
Previous experiments using green fluorescent reporter in which the apicoplast signal peptide protein was replaced by the signal peptide of a micronemal and a knob-associated protein indicated that targeting proteins to the apicoplast begins at the general secretory pathway of P. falciparum [13]. In P. falciparum, the signal peptide in apicoplast-resident proteins, was shown to facilitate translocation across the rough ER (RER) membrane through the Sec machinery releasing the protein into the ER lumen [29,30]. This is followed by the removal of the signal peptide by signal peptidase [31,32]. Once in the ER, the transit peptide directs the transport into the apicoplast [28]. This mechanism of trafficking seems to be shared by other secondary plastids [31,32,33,34,35]. Furthermore, it has been validated in Toxoplasma gondii [36,37,38,39]. On the other hand, apicoplast transmembrane proteins that are encoded by the nucleus are trafficked via the Golgi apparatus [40].

4. Protein Sorting to Food Vacuole

The intraerythrocytic stage of P. falciparum depends heavily on the host cytosolic proteins, which are funneled to the parasite by endocytosis. Hemoglobin from the erythrocyte cytosol is shuttled across the parasitophorous vacuole membrane (PVM), the plasma membrane of the parasite, via the cytostome or micropore [41,42]. Their final destination is the food vacuole, a lysosomal-like compartment in P. falciparum that is specialized for degradation. In Toxoplasma gondii, this organelle is called the lysosomal-like vacuolar compartment or VAC [43,44]. During the intraerythrocytic stage, P. falciparum takes up a considerable portion of the host cell cytosol. The erythrocyte cytosol containing mostly hemoglobin is carried to the food vacuole where it is digested, providing the necessary amino acids required for growth and development [45,46,47]. There, the ingested hemoglobin is acidified and broken down to amino acids. In the food vacuole, peptides are transported and heme is polymerized and detoxified. The food vacuole is also the site of action of antimalarials such as chloroquine. The host cell cytosol uptake (HCCU) that leads to the formation of the food vacuole is essential for the parasite’s survival.
Although many of the resident food vacuole proteins have been well-characterized, the molecular mechanism of protein sorting to the food vacuole is not well understood [12]. Protein trafficking from the host cytosol to the food vacuole of P. falciparum and the vacuole (VAC) of T. gondii remain elusive. It was previously thought that the cytostome was the main driver for the formation of the HCCU [48]. Experiments using inhibitors of the cytoskeletal proteins actin and myosin, as well as inhibitors of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) and dynamin, had suggested that they could play a critical role in the formation of HCCU [49,50,51,52]. However, data from a recent study did not seem to implicate these cytoskeletal proteins. Instead, the P. falciparum vacuolar protein sorting-associated protein 45, PfVPS45, was shown to be required for the formation of HCCU [53]. Conditional mutants of the pfvpd45 gene resulted in the buildup of vesicles packed with the host cell cytoplasm [53]. These vesicles seem to be connected to the host cell [53]. VPS45 was further shown to be needed for trafficking from the host cell cytosol to the food vacuole [53]. In addition, hemoglobin transport to the food vacuole was inhibited in the mutants, halting parasite growth [53]. This function is conserved in T. gondii where TgVPS45 mediate VAC digestion of host-endocytosed proteins [54].
Interestingly, many of the HCCU vesicles seem to have endosomal features since they contain phosphatidylinositol 3-phosphate [53]. In other organisms, such as the budding yeast, VPS45 plays a role in endo-lysosomal transport [55,56]. It appears that P. falciparum is similar to its apicomplexan counterpart, T. gondii, in using the machinery of the endo-lysosomal system for secreting protein [57,58,59,60,61,62,63,64,65,66]. Rab5 plays a central role in early endocytosis where it moves its target to the early endosomes, transports it to the endosomes, and mediates internalization of receptors. However, T. gondii Rab5 paralogs have not been shown to be involved in endocytosis [61]. Rather, they are involved in protein secretion [61]. In summary, the endosome-like compartment (ELC) is where endocytosis and exocytosis overlap in apicomplexans [62]. Protein sorting at the ELC is coordinated by several tethering proteins, such as clathrin [67], HOPS, CORVET [68], DrpB [57], and the Rab5 paralogs in T. gondii [54,61].

5. P. falciparum Protein Export

Following infection, P. falciparum releases the contents of its apical organelles, leading to the formation of the parasitophorous vacuolar membrane (PVM), within which it inhabits. Although the PVM provides protection to the parasite, it is also an obstacle for the transfer and distribution of the host-targeted effectors. These effector proteins can either be shipped to or across the parasite-plasma membrane (PM), as well as the lumen of the PVM. These effectors comprise P. falciparum erythrocyte membrane protein 1 (PfEMP1) that is secreted to the erythrocyte membrane. It is estimated that P. falciparum secretes over 400 proteins into the erythrocyte [69]. These proteins alter the structure and physiology of the erythrocyte membrane, leading to its rigidity and changing its permeability [70]. The proteins that the parasite ships to the erythrocyte membrane are needed for nutrient uptake, evasion of the host immune, and for release from the infected erythrocytes [71,72,73].
During the erythrocytic cycle, P. falciparum causes significant and drastic changes to the erythrocyte cytoplasm and plasma membrane, leading to the formation of the Maurer’s clefts and protrusions in the membrane called knobs. Maurer’s clefts are disc-shaped cisternae that are moveable in the cytoplasm during the ring intraerythrocytic stage but later become attached to the erythrocyte membrane skeleton [74]. The Maurer’s clefts are bound to the erythrocyte membrane by a complex of proteins composed of the host actin filaments and parasite-encoded proteins [75,76]. The knobs function as a scaffold for the display of the virulence antigen P. falciparum erythrocyte membrane protein 1 (PfEMP1) on the erythrocyte plasma membrane [76].
PfEMP1 mediates the sequestration of the infected erythrocytes to tissues and to uninfected erythrocytes, leading to anemia and severe symptoms [74]. PfEMP1 is responsible for the attachment of infected erythrocytes to endothelial lining of the blood vessels, causing the infected erythrocytes to sequester away, thus avoiding clearance from the spleen [2]. PfEMP1 is shipped into the erythrocyte cytoplasm through the Plasmodium translocon of exported proteins (PTEX) located on the parasitophorous vacuole membrane [77,78]. The present model proposes that PfEMP1 is first trafficked to the Maurer’s clefts in association with the chaperone complex made up of hsp70/hsp40 in a soluble state [79,80]. Recently, two PfEMP1-interacting complexes were described [81]. The P. falciparum GEXP07/CX3CL1-binding protein 2 (CBP2) was shown to be necessary for PfEMP1 trafficking to the erythrocyte membrane [81]. Deletion of GEXP07 results in parasite with distorted Maurer’s clefts, faulty PfEMP1 trafficking, abnormal knob formation, and failure of infected erythrocytes to attach to the endothelial lining [81]. The mutant parasites seem to grow faster than the wild-type parasites. Moreover, erythrocytes infected with the mutant parasite are less rigid and more vulnerable to osmotic pressure [81]. It has been hypothesized that vesicle-containing PfEMP1 bud from the Maurer’s clefts and travel along the host actin filaments [82]. However, the precise mechanism of PfEMP1 trafficking remains to be elucidated.
The export of P. falciparum proteins into the erythrocyte begins in the parasite ER. A subgroup of parasite-exported proteins contains an N-terminal export signal named Plasmodium Export Element (PEXEL), or the Host-Targeting motif [83,84]. The PEXEL motif is a pentameric sequence (RxLxE/Q/D) that is cut during translation at the conserved leucine residue by plasmepsin V, an ER-resident endoprotease [85,86,87]. It was recently demonstrated that early recognition of exported proteins takes place during transit across the ER via interactions of plasmepsin V and molecules of the translocation machinery [88]. This indicates that plasmepsin V plays the role of signal peptidase for PEXEL-positive exported proteins. However, the following steps responsible for export have not been defined. A large number of proteins which lack the PEXEL/HT motif are exported to the erythrocyte [89,90,91]. These so-called PEXEL-negative exported proteins (PNEPs) are thought to use some of the same export machinery as the PEXEL-positive proteins [77,78,89,90]. The mechanism by which parasite proteins are exported and refolded in the host cells remains largely unknown. Current data suggest that parasite-exported chaperones as well as chaperones from the host erythrocyte are involved in the process. Indeed, P. falciparum-exported Hsp40 co-chaperones have been shown to play a critical role in remodeling the erythrocyte [92]. Moreover, the localization of erythrocyte Hsp70 was demonstrated to be altered following infection with P. falciparum, since it is soluble in uninfected red blood cells, but is associated with detergent-resistant fractions after infection [93].
Exported P. falciparum proteins must also go through the PVM. This is accomplished by the Plasmodium translocon of exported proteins (PTEX) [94,95]. The translocon is a multi-complex structure composed of EXP2, an integral membrane protein, PTEX150, and the AAA+ ATPase Hsp101, a chaperone [96,97,98]. As an AAA+ ATPase, Hsp101 couples ATP hydrolysis with protein unfolding as the exported protein moves through the translocon [98]. It plays a critical role in the export of parasite proteins [98]. It has been shown that exported proteins that are kept in the folded state remain stuck in the parasite vacuole and block the PTEX translocon [77,95,99,100,101]. These data indicate that PTEX functions as a gateway for P. falciparum proteins to the host cell, and that unfolding by chaperones play a crucial role in the export process.

6. Vesicular Trafficking in P. falciparum

In eukaryotes, communication between organelles is carried out by budding vesicles that move along the cytoskeleton to attach and fuse with their target membranes [102]. The accuracy of the process depends on three molecules that act as distinctive identifiers: the SNARE proteins, the small GTPases, and the phosphatidylinositol phospholipids [103,104]. The assembly of the SNARE complex is regulated by Sec1/Munc18-like proteins in coordination with the syntaxin (Stx)-like SNARE protein [105]. The role of Sec1/Munc18 and SNARE proteins in P. falciparum and T. gondii was recently examined in order to discover the molecules involved in vesicular trafficking in these two apicomplexan parasites. The genome of apicomplexan parasites encodes for four Sec1/Munc18 proteins, including the orthologue of the suppressor of loss of YPT1 function (SLY1), which was shown to be necessary for the movement of vesicles from the ER to the Golgi [53]. SLY1 controls the secretion of all proteins from the Golgi. In apicomplexans, it was determined that Sec1/Munc18 interacts with SNARE proteins to trigger vesicle targeting to numerous organelles, including the pre-rhoptries, the micronemes, the vacuolar compartment, the apicoplast, as well as the inner membrane complex (IMC), a membrane-cytoskeletal scaffold made of flattened vesicles thought to have originated from the trans-Golgi [53,106,107]. Sec1/Munc18-controlled vesicles are thought to originate either from the ER, the Golgi apparatus, or the endosomal-like compartment (ELC) [53]. The ELC is where proteins targeted to the micronemes and the rhoptries are sorted and it is also where micronemal and rhoptry proteins are processed by aspartyl protease 3 (ASP3) [59,108]. It has recently been demonstrated that Vps45 in both P. falciparum and T. gondii is responsible for the biogenesis of the IMC [53]. It is hypothesized that Stx16 and Stx6 work with Vps45 in the formation of the IMC [53]. Additionally, the sorting of the rhoptry, micronemal, and apicoplast proteins to their respective organelles was shown to be dependent on Stx12, an ELC resident protein [53].
The current model of vesicular trafficking in apicomplexan parasites suggest that secreted proteins are first synthesized in the rough endoplasmic reticulum [54,62]. The proteins are then transported to the cis-Golgi, likely by SLY1. The fusion of these vesicles is thought to be mediated by Stx5, a protein known to interact with SLY1 [109]. On the other hand, the inner membrane complex was shown to be derived from the trans-Golgi compartment, where Vsp45 in coordination with Stx16 and Stx6 is involved in the re-processing of the trafficking factors used during biogenesis of the inner membrane complex [53]. Likewise, Vsp45, in association with Stx16 and Stx6, is also responsible for trafficking between the ELC and the trans-Golgi [110]. Although homeostasis of the apicoplast requires Stx12, the SNARE proteins responsible for transport of apicoplast proteins to the ER have not yet been identified [53]. In summary, the identity of specific proteins responsible for trafficking to the various P. falciparum organelles have not fully been uncovered. The current data suggest that endocytosis takes place either in the micropore or the cytostome [41,42,49], followed by transport into sub-compartments of the ELC [62]. Vsp45 and most likely Stx16 are involved in this process [53]. The SNARE proteins involved in the endocytic process have not been identified.

7. The Role of Prenylation in P. falciparum Vesicular Trafficking

The rhoptries and micronemes of apicomplexan parasites are thought to originate from the post-Golgi vesicles. The adaptor protein (AP) complex has been shown to play an important role in protein trafficking to the rhoptry [111]. This complex is responsible for cargo recognition during post-Golgi trafficking in eukaryotic cells [112]. It is a heterotetrameric complex composed of subunits of varying sizes that have distinct roles in the trafficking process [113,114,115]. They are organized into two large subunits (γ, α, δ, ε, ζ, and β1–5), a medium (μ1–5), and a small subunit (σ1–5). Five adaptor protein complexes have been described [108]. AP-1 and AP-2 are known to recognize clathrin, whereas AP-4 and AP-5 do not; the interaction of AP-3 with clathrin has not been clearly established [116]. In higher eukaryotes, the AP-1 complex mediates trafficking between the trans-Golgi network and the endosome, while AP-2 is responsible in clathrin-mediated endocytosis [113]. In contrast, AP-3 directs trafficking to the lysosomes [113]. The roles of the AP-4 and AP-5 complexes remain unclear, although some studies suggest that they might be involved in the shipment of cargo to the endosomes [112,117]. In P. falciparum, the AP-1 complex was shown to be necessary in the trafficking of rhoptry proteins [118]. Indeed, the medium subunit of P. falciparum, Pfμ1, was shown to colocalize with Golgi/ER markers in the trophozoite stage, but Pfμ1 was later associated with the rhoptry makers, RAP1 and Clag3.1 [111]. Stx12 has been identified as the SNARE protein responsible for trafficking of both microneme and rhoptry proteins to their appropriate destination [62].

8. Vesicular Trafficking in P. falciparum

In eukaryotic cells, vesicle fusion depends on the attachment of the SNARE receptors to specific organelles [118,119,120]. Although most SNARE proteins have a C-terminal transmembrane domain that mediates their attachment to their target membranes, some lack these transmembrane domains. The latter rely on lipid modification in order to be membrane-anchored. Protein prenylation is the attachment of either a farnesyl (15-carbon) or a geranylgeranyl (20-carbon) isoprenoid group to cysteine residue of CAAX motif-containing proteins [121,122,123,124]. Protein prenylation is carried out by three groups of protein prenyltransferases: farnesyltransferase (FT), geranylgeranyl- transferase 1 (GGT1), and Rab geranylgeranyltransferase (RabGGT) [125]. The P. falciparum prenylated proteome has been characterized [126]. Interestingly, several prenylation candidates were uncovered [126]. Specifically, The SNARE protein PfYkt6p was characterized in P. falciparum and shown to be both prenylated and surprisingly geranylgeranylated [127]. Importantly, the data from this study indicate that the transport of Ykt6 in P. falciparum depends on prenylation. It is also the first evidence of protein geranylgeranyltransferase activity on SNARE proteins [127]. Taken together, prenylation might play an important role in P. falciparum vesicular trafficking. More studies are needed to uncover the role of prenylation in the endocytic and secretory pathway of P. falciparum.

9. Conclusions

As in other eukaryotes, vesicular trafficking in P. falciparum is essential for the creation of organelles, as well as for the communication between organelles. The pathogenesis of P. falciparum depends heavily on vesicular trafficking processes. This process is mediated by Rab GTPases [54], phospholipids [72,110], and the SNARE proteins [53]. The ELC plays a central role in Plasmodium protein sorting and trafficking. It is now evident that the ELC represents the junction of the endocytic and exocytic, controlling the formation of numerous organelles that are part of these two opposite pathways. It accepts and ships vesicles from the Golgi apparatus to the micronemes, the prorhoptries, and the food vacuole (Figure 1). It is also involved in endocytosis of host proteins. It has recently been shown that Vps45, in both P. falciparum and T. gondii, is responsible for shipping both the endosomal and secretory vesicles. Additionally, it is required in the formation of the inner membrane complex. In summary, P. falciparum, like T. gondii, repurposes a portion of the endosomal system to the secretory pathway particularly for the creation of the secretory organelles such as the micronemes and the rhoptries, which are required during the process of erythrocyte invasion. The SNARE and Sec1/Munc18 proteins in apicomplexans are responsible for creating the ZIP code required for vesicular trafficking. Finally, P. falciparum vesicular trafficking appears to be very similar to that of T. gondii, suggesting that the trafficking and sorting pathways are conserved in apicomplexans [52]. Although significant progress has been made in elucidating vesicular trafficking in P. falciparum, there still remain many unanswered questions. The tethering molecules responsible for sorting to each of the parasite’s respective organelles have yet to be identified.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. World Health Organization. World Malaria Report: 20 Years of Global Progress and Challenges; World Health Organization: Geneva, Switzerland, 2020. [Google Scholar]
  2. Miller, L.H.; Ackerman, H.C.; Su, X.; Wellems, T.E. Malaria biology and disease pathogenesis: Insights for new treatments. Nat. Med. 2013, 19, 156–167. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Yeoh, S.; O’Donnell, R.A.; Koussis, K.; Dluzewski, A.R.; Ansell, K.H.; Osborne, S.A.; Hackett, F.; Withers-Martinez, C.; Mitchell, G.H.; Bannister, L.H.; et al. Subcellular discharge of a serine protease mediates release of invasive malaria parasites from host erythrocytes. Cell 2007, 131, 1072–1083. [Google Scholar] [CrossRef] [Green Version]
  4. Reilly, H.B.; Wang, H.; Steuter, J.A.; Marx, A.M.; Ferdig, M.T. Quantitative dissection of clone-specific growth rates in cultured malaria parasites. Int. J. Parasitol. 2007, 37, 1599–1607. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Wahlgren, M.; Goel, S.; Akhouri, R.R. Variant surface antigens of Plasmodium falciparum and their roles in severe malaria. Nat. Rev. Microbiol. 2017, 15, 479–491. [Google Scholar] [CrossRef] [PubMed]
  6. Pfeffer, S.R.; Rothman, J.E. Biosynthetic protein transport and sorting by the endoplasmic reticulum and Golgi. Ann. Rev. Biochem. 1987, 56, 829–852. [Google Scholar] [CrossRef] [PubMed]
  7. Blobel, G. Intracellular protein topogenesis. Proc. Natl. Acad. Sci. USA 1980, 77, 1496–1500. [Google Scholar] [CrossRef] [Green Version]
  8. von Heijne, G. Signal sequences: The limits of variation. J. Mol. Biol. 1985, 184, 99–105. [Google Scholar] [CrossRef]
  9. Gubbels, M.J.; Duraisingh, M.T. Evolution of apicomplexan secretory organelles. Int. J. Parasitol. 2012, 42, 1071–1081. [Google Scholar] [CrossRef] [Green Version]
  10. Maier, A.G.; Cooke, B.M.; Cowman, A.F.; Tilley, L. Malaria parasite proteins that remodel the host erythrocyte. Nat. Rev. Microbiol. 2009, 7, 341–354. [Google Scholar] [CrossRef]
  11. Banerjee, R.; Goldberg, D.E. The Plasmodium food vacuole. In Antimalarial Chemotherapy; Humana Press: Totowa, NJ, USA, 2001; pp. 43–63. [Google Scholar]
  12. Singh, S.; Plassmeyer, M.; Gaur, D.; Miller, L.H. Mononeme: A new secretory organelle in Plasmodium falciparum merozoites identified by localization of rhomboid-1 protease. Proc. Natl. Acad. Sci. USA 2007, 104, 20043–20048. [Google Scholar] [CrossRef] [Green Version]
  13. Tonkin, C.J.; Struck, N.S.; Mullin, K.A.; Stimmler, L.M.; McFadden, G.I. Evidence for Golgi-independent transport from the early secretory pathway to the plastid in malaria parasites. Mol. Microbiol. 2006, 61, 614–630. [Google Scholar] [CrossRef]
  14. Glick, B.S.; Nakano, A. Membrane traffic within the Golgi apparatus. Annu. Rev. Cell Dev. Biol. 2009, 25, 113–132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Lee, M.C.; Moura, P.A.; Miller, E.A.; Fidock, D.A. Plasmodium falciparum Sec24 marks transitional ER that exports a model cargo via a diacidic motif. Mol. Microbiol. 2008, 68, 1535–1546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Bannister, L.H.; Hopkins, J.M.; Fowler, R.E.; Krishna, S.; Mitchell, G.H. Ultrastructure of rhoptry development in Plasmodium falciparum erythrocytic schizonts. Parasitol. Today 2000, 121, 273–287. [Google Scholar] [CrossRef]
  17. Bannister, L.H.; Hopkins, J.M.; Margos, G.; Dluzewski, A.R.; Mitchell, G.H. Three-dimensional ultrastructure of the ring stage of Plasmodium falciparum: Evidence for export pathways. Microsc. Microanal. 2004, 10, 551–562. [Google Scholar] [CrossRef]
  18. Langreth, S.G.; Jensen, J.B.; Reese, R.T.; Trager, W. Fine structure of human malaria in vitro. J. Protozool. 1978, 25, 443–452. [Google Scholar] [CrossRef] [PubMed]
  19. Adisa, A.; Frankland, S.; Rug, M.; Jackson, K.; Maier, A.G.; Walsh, P.; Lithgow, T.; Klonis, N.; Gilson, P.R.; Cowman, A.F.; et al. Re-assessing the locations of components of the classical vesicle-mediated trafficking machinery in transfected Plasmodium falciparum. Int. J. Parasitol. 2007, 37, 1127–1141. [Google Scholar] [CrossRef]
  20. Van Wye, J.; Ghori, N.; Webster, P.; Mitschler, R.R.; Elmendorf, H.G.; Haldar, K. Identification and localization of rab6, separation of rab6 from ERD2 and implications for an ‘unstacked’ Golgi, in Plasmodium falciparum. Mol. Biochem. Parasitol. 1996, 83, 107–120. [Google Scholar] [CrossRef]
  21. McFadden, G.I.; Reith, M.; Munholland, J.; Lang Unnasch, N. Plastid in human parasites. Nature 1996, 381, 482. [Google Scholar] [CrossRef]
  22. Wilson, R.J.M.; Denny, P.W.; Preiser, P.R.; Rangachari, K.; Roberts, K.; Roy, A.; Whyte, A.; Strath, M.; Moore, D.J.; Moore, P.W.; et al. Complete gene map of the plastid-like DNA of the malaria parasite Plasmodium falciparum. J. Mol. Biol. 1996, 261, 155–172. [Google Scholar] [CrossRef] [PubMed]
  23. Köhler, S.; Delwiche, C.F.; Denny, P.W.; Tilney, L.G.; Webster, P.; Wilson, R.J.M.; Palmer, J.D.; Roos, D.S. A plastid of probable green algal origin in apicomplexan parasites. Science 1997, 275, 1485–1489. [Google Scholar] [CrossRef] [Green Version]
  24. van Dooren, G.G.; Schwartzbach, S.D.; Osafune, T.; McFadden, G.I. Translocation of proteins across the multiple membranes of complex plastids. Biochim. Biophys. Acta 2001, 1541, 34–53. [Google Scholar] [CrossRef] [Green Version]
  25. Seeber, F.; Soldati-Favre, D. Metabolic pathways in the apicoplast of Apicomplexa. Int. Rev. Cell Mol. Biol. 2010, 281, 161–228. [Google Scholar] [CrossRef] [PubMed]
  26. Deane, J.; Fraunholz, M.; Su, V.; Maier, U.-G.; Martin, W.; Durnford, D.; McFadden, G. Evidence for nucleomorph to host nucleus gene transfer: Light harvesting complex proteins from cryptomonads and chlorarachniophytes. Protist 2000, 151, 239–252. [Google Scholar] [CrossRef] [Green Version]
  27. McFadden, G.I.; van Dooren, G.G. Evolution: Red algal genome affirms a common origin of all plastids. Curr. Biol. 2004, 14, R514–R516. [Google Scholar] [CrossRef] [Green Version]
  28. Waller, R.F.; Reed, M.B.; Cowman, A.F.; McFadden, G.I. Protein trafficking to the plastid of Plasmodium falciparum is via the secretory pathway. EMBO J. 2000, 19, 1794–1802. [Google Scholar] [CrossRef] [Green Version]
  29. van Dooren, G.G.; Waller, R.F.; Joiner, K.A.; Roos, D.S.; McFadden, G.I. Traffic jams: Protein transport in Plasmodium falciparum. Parasitol. Today 2000, 16, 421–427. [Google Scholar] [CrossRef]
  30. van Dooren, G.G.; Su, V.; D’Ombrain, M.C.; McFadden, G.I. Processing of an apicoplast leader sequence in Plasmodium falciparum and the identification of a putative leader cleavage enzyme. J. Biol. Chem. 2002, 277, 23612–23619. [Google Scholar] [CrossRef] [Green Version]
  31. Sulli, C.; Fang, Z.; Muchhal, U.; Schwartzbach, S.D. Topology of Euglena chloroplast protein precursors within endoplasmic reticulum to Golgi to chloroplast trans- port vesicles. J. Biol. Chem. 1999, 274, 457–463. [Google Scholar] [CrossRef] [Green Version]
  32. Wastl, J.; Maier, U.G. Transport of proteins into cryptomonads complex plastids. J. Biol. Chem. 2000, 275, 23194–23198. [Google Scholar] [CrossRef] [Green Version]
  33. Apt, K.E.; Zaslavkaia, L.; Lippmeier, J.C.; Lang, M.; Kilian, O.; Wetherbee, R.; Grossman, A.R.; Kroth, P. In vivo characterization of diatom multipartite plastid targeting signals. J. Cell Sci. 2002, 115, 4061–4069. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Nassoury, N.; Cappadocia, M.; Morse, D. Plastid ultrastructure defines the protein import pathway in dinoflagellates. J. Cell Sci. 2003, 116, 2867–2874. [Google Scholar] [CrossRef] [Green Version]
  35. Patron, N.J.; Waller, R.F.; Archibald, J.M.; Keeling, P.J. Complex protein targeting to dinoflagellate plastids. J. Mol. Biol. 2005, 348, 1015–1024. [Google Scholar] [CrossRef]
  36. DeRocher, A.; Hagen, C.B.; Froehlich, J.E.; Feagin, J.E.; Parsons, M. Analysis of targeting sequences demonstrates that trafficking to the Toxoplasma gondii plastid branches of the secretory system. J. Cell Sci. 2000, 113, 3969–3977. [Google Scholar] [CrossRef]
  37. DeRocher, A.; Gilbert, B.; Feagin, J.E.; Parsons, M. Dissection of brefeldin A-sensitive and -insensitive steps in apicoplast protein targeting. J. Cell Sci. 2005, 118, 565–574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Yung, S.; Unnasch, T.R.; Lang-Unnasch, N. Analysis of apicoplast targeting and transit peptide processing in Toxoplasma gondii by deletional and insertional mutagenesis. Mol. Biochem. Parasitol. 2001, 118, 11–21. [Google Scholar] [CrossRef]
  39. Harb, O.S.; Chatterjee, B.; Fraunholz, M.J.; Crawford, M.J.; Nishi, M.; Roos, D.S. Multiple functionally redundant signals mediate targeting to the apicoplast in the apicomplexan parasite Toxoplasma gondii. Eukaryot. Cell 2004, 3, 663–674. [Google Scholar] [CrossRef] [Green Version]
  40. Chaudhari, R.; Dey, V.; Narayan, A.; Sharma, S.; Patankar, S. Membrane and luminal proteins reach the apicoplast by different trafficking pathways in the malaria parasite Plasmodium falciparum. PeerJ 2017, 5, e3128. [Google Scholar] [CrossRef] [Green Version]
  41. Nichols, B.A.; Chiappino, M.L.; Pavesio, C.E. Endocytosis at the micro pore of Toxoplasma gondii. Parasitol. Res. 1994, 80, 91–98. [Google Scholar] [CrossRef]
  42. Spielmann, T.; Gras, S.; Sabitzki, R.; Meissner, M. Endocytosis in Plasmodium and Toxoplasma parasites. Trends Parasitol. 2020, 36, 520–532. [Google Scholar] [CrossRef]
  43. Miranda, K.; Pace, D.A.; Cintron, R.; Rodrigues, J.C.; Fang, J.; Smith, A.; Rohloff, P.; Coelho, E.; de Haas, F.; de Souza, W.; et al. Characterization of a novel organelle in Toxoplasma gondii with similar composition and function to the plant vacuole. Mol. Microbiol. 2010, 76, 1358–1375. [Google Scholar] [CrossRef] [Green Version]
  44. Parussini, F.; Coppens, I.; Shah, P.P.; Diamond, S.L.; Carruthers, V.B. Cathepsin L occupies a vacuolar compartment and is a protein maturase within the endo/exocytic system of Toxoplasma gondii. Mol. Microbiol. 2010, 76, 1340–1357. [Google Scholar] [CrossRef] [Green Version]
  45. Sherman, I.W. Amino acid metabolism and protein synthesis in malarial parasites. Bull. World Health Organ. 1977, 55, 265–276. [Google Scholar]
  46. Hanssen, E.; Knoechel, C.; Dearnley, M.; Dixon, M.W.; Le Gros, M.; Larabell, C.; Tilley, L. Soft X-ray microscopy analysis of cell volume and hemoglobin content in erythrocytes infected with asexual and sexual stages of Plasmodium falciparum. J. Struct. Biol. 2012, 177, 224–232. [Google Scholar] [CrossRef] [Green Version]
  47. Lew, V.L.; Tiffert, T.; Ginsburg, H. Excess hemoglobin digestion and the osmotic stability of Plasmodium falciparum-infected red blood cells. Blood 2003, 101, 4189–4194. [Google Scholar] [CrossRef]
  48. Aikawa, M.; Hepler, P.K.; Huff, C.G.; Sprinz, H. The feeding mechanism of avian malarial parasites. J. Cell Biol. 1966, 28, 355–373. [Google Scholar] [CrossRef] [Green Version]
  49. Lazarus, M.D.; Schneider, T.G.; Taraschi, T.F. A new model for hemoglobin ingestion and transport by the human malaria parasite Plasmodium falciparum. J. Cell Sci. 2008, 121, 1937–1949. [Google Scholar] [CrossRef] [Green Version]
  50. Milani, K.J.; Schneider, T.G.; Taraschi, T.F. Defining the morphology and mechanism of the hemoglobin transport pathway in Plasmodium falciparum infected erythrocytes. Eukaryot. Cell 2015, 14, 415–426. [Google Scholar] [CrossRef] [Green Version]
  51. Smythe, W.A.; Joiner, K.A.; Hoppe, H.C. Actin is required for endocytic trafficking in the malaria parasite Plasmodium falciparum. Cell. Microbiol. 2008, 10, 452–464. [Google Scholar] [CrossRef]
  52. Zhou, H.C.; Gao, Y.H.; Zhong, X.; Wang, H. Dynamin like protein 1 participated in the hemoglobin uptake pathway of Plasmodium falciparum. Chin. Med. J. 2009, 122, 1686–1691. [Google Scholar]
  53. Bisio, H.; Chaabene, R.B.; Sabitzki, R.; Maco, B.; Marq, J.B.; Gilberger, T.W.; Spielmann, T.; Soldati-Favre, D. The ZIP code of vesicle trafficking in apicomplexa: Sec1/Munc18 and SNARE proteins. Mol. Biol. Physiol. 2020, 11, e02092-20. [Google Scholar] [CrossRef]
  54. Venugopal, K.; Marion, S. Secretory organelle trafficking in Toxoplasma gondii: A long story for a short travel. Int. J. Med. Microbiol. 2018, 308, 751–760. [Google Scholar] [CrossRef]
  55. Cowles, C.R.; Emr, S.D.; Horazdovsky, B.F. Mutations in the VPS45 gene, a Sec1 homologue, result in vacuolar protein sorting defects and accumulation of membrane vesicles. J. Cell Sci. 1994, 107, 3449–3459. [Google Scholar] [CrossRef] [PubMed]
  56. Piper, R.C.; Whitters, E.A.; Stevens, T.H. Yeast Vps45p is a Sec1p-like protein required for the consumption of vacuole-targeted, post-Golgi transport vesicles. Eur. J. Cell Biol. 1994, 65, 305–318. [Google Scholar]
  57. Breinich, M.S.; Ferguson, D.J.; Foth, B.J.; van Dooren, G.G.; Lebrun, M.; Quon, D.V.; Striepen, B.; Bradley, P.J.; Frischknecht, F.; Carruthers, V.B.; et al. A dynamin is required for the biogenesis of secretory organelles in Toxoplasma gondii. Curr. Biol. 2009, 19, 277–286. [Google Scholar] [CrossRef] [Green Version]
  58. Hallee, S.; Counihan, N.A.; Matthews, K.; de Koning-Ward, T.F.; Richard, D. The malaria parasite Plasmodium falciparum Sortilin is essential for merozoite formation and apical complex biogenesis. Cell. Microbiol. 2018, 20, e12844. [Google Scholar] [CrossRef]
  59. Jimenez-Ruiz, E.; Morlon-Guyot, J.; Daher, W.; Meissner, M. Vacuolar protein sorting mechanisms in apicomplexan parasites. Mol. Biochem. Parasitol. 2016, 209, 18–25. [Google Scholar] [CrossRef]
  60. Krai, P.; Dalal, S.; Klemba, M. Evidence for a Golgi-to-endosome protein sorting pathway in Plasmodium falciparum. PLoS ONE 2014, 9, e89771. [Google Scholar] [CrossRef] [Green Version]
  61. Kremer, K.; Kamin, D.; Rittweger, E.; Wilkes, J.; Flammer, H.; Mahler, S.; Heng, J.; Tonkin, C.J.; Langsley, G.; Hell, S.W.; et al. An overexpression screen of Toxoplasma gondii Rab-GTPases reveals distinct transport routes to the micronemes. PLoS Pathog. 2013, 9, e1003213. [Google Scholar] [CrossRef]
  62. McGovern, O.L.; Rivera-Cuevas, Y.; Kannan, G.; Narwold, A.J., Jr.; Carruthers, V.B. Intersection of endocytic and exocytic systems in Toxoplasma gondii. Traffic 2018, 19, 336–353. [Google Scholar] [CrossRef] [Green Version]
  63. Pieperhoff, M.S.; Schmitt, M.; Ferguson, D.J.; Meissner, M. The role of clathrin in post-Golgi trafficking in Toxoplasma gondii. PLoS ONE 2013, 8, e77620. [Google Scholar] [CrossRef] [Green Version]
  64. Sangare, L.O.; Alayi, T.D.; Westermann, B.; Hovasse, A.; Sindikubwabo, F.; Callebaut, I.; Werkmeister, E.; Lafont, F.; Slomianny, C.; Hakimi, M.A.; et al. Unconventional endosome-like compartment and retromer complex in Toxoplasma gondii govern parasite integrity and host infection. Nat. Commun. 2016, 7, 11191. [Google Scholar] [CrossRef] [Green Version]
  65. Tomavo, S. Evolutionary repurposing of endosomal systems for apical organelle biogenesis in Toxoplasma gondii. Int. J. Parasitol. 2014, 44, 133–138. [Google Scholar] [CrossRef] [PubMed]
  66. Tomavo, S.; Slomianny, C.; Meissner, M.; Carruthers, V.B. Protein trafficking through the endosomal system prepares intracellular parasites for a home invasion. PLoS Pathog. 2013, 9, e1003629. [Google Scholar] [CrossRef] [Green Version]
  67. Venugopal, K.; Werkmeister, E.; Barois, N.; Saliou, J.M.; Poncet, A.; Huot, L.; Sindikubwabo, F.; Hakimi, M.A.; Langsley, G.; Lafont, F.; et al. Dual role of the Toxoplasma gondii clathrin adaptor AP1 in the sorting of rhoptry and microneme proteins and in parasite division. PLoS Pathog. 2017, 13, e1006331. [Google Scholar] [CrossRef] [Green Version]
  68. Morlon-Guyot, J.; Pastore, S.; Berry, L.; Lebrun, M.; Daher, W. Toxoplasma gondii Vps11, a subunit of HOPS and CORVET tethering complexes, is essential for the biogenesis of secretory organelles. Cell. Microbiol. 2015, 17, 1157–1178. [Google Scholar] [CrossRef] [PubMed]
  69. Cowman, A.F.; Healer, J.; Marapana, D.; Marsh, K. Malaria: Biology and disease. Cell 2016, 167, 610–624. [Google Scholar] [CrossRef] [Green Version]
  70. Spillman, N.J.; Beck, J.R.; Goldberg, D.E. Protein export into malaria parasite-infected erythrocytes: Mechanisms and functional consequences. Ann. Rev. Biochem. 2015, 84, 813–841. [Google Scholar] [CrossRef] [PubMed]
  71. Nguitragool, W.; Bokhari, A.A.B.; Pillai, A.D.; Rayavara, K.; Sharma, P.; Turpin, B.; Aravind, L.; Desai, S.A. Malaria parasite clag3 genes determine channel-mediated nutrient uptake by infected red blood cells. Cell 2011, 145, 665–677. [Google Scholar] [CrossRef] [Green Version]
  72. Smith, J.D.; Rowe, J.A.; Higgins, M.K.; Lavstsen, T. Malaria’s deadly grip: Cytoadhesion of Plasmodium falciparum-infected erythrocytes. Cell. Microbiol. 2013, 15, 1976–1983. [Google Scholar] [CrossRef] [Green Version]
  73. Raj, D.K.; Nixon, C.P.; Nixon, C.E.; Dvorin, J.D.; DiPetrillo, C.G.; Pond-Tor, S.; Wu, H.-W.; Jolly, G.; Pischel, L.; Lu, A.; et al. Antibodies to PfSEA-1 block parasite egress from RBCs and protect against malaria infection. Science 2014, 344, 871–877. [Google Scholar] [CrossRef] [Green Version]
  74. McMillan, P.J.; Millet, C.; Batinovic, S.; Maiorca, M.; Hanssen, E.; Kenny, S.; Muhle, R.A.; Melcher, M.; Fidock, D.A.; Smith, J.D.; et al. Spatial and temporal mapping of the PfEMP1 export pathway in Plasmodium falciparum. Cell. Microbiol. 2013, 15, 1401–1418. [Google Scholar] [CrossRef] [Green Version]
  75. Cyrklaff, M.; Sanchez, C.P.; Kilian, N.; Bisseye, C.; Simpore, J.; Frischknecht, F.; Lanzer, M. Hemoglobins S and C interfere with actin remodeling in Plasmodium falciparum-infected erythrocytes. Science 2011, 334, 1283–1286. [Google Scholar] [CrossRef] [PubMed]
  76. Pachlatko, E.; Rusch, S.; Muller, A.; Hemphill, A.; Tilley, L.; Hanssen, E.; Beck, H.P. MAHRP2, an exported protein of Plasmodium falciparum, is an essential component of Maurer’s cleft tethers. Mol. Microbiol. 2010, 77, 1136–1152. [Google Scholar] [CrossRef]
  77. Beck, J.R.; Muralidharan, V.; Oksman, A.; Goldberg, D.E. PTEX component HSP101 mediates export of diverse malaria effectors into host erythrocytes. Nature 2014, 511, 592–595. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Elsworth, B.; Sanders, P.R.; Nebl, T.; Batinovic, S.; Kalanon, M.; Nie, C.Q.; Charnaud, S.C.; Bullen, H.E.; Ward, T.F.K.; Tilley, L.; et al. Proteomic analysis reveals novel proteins associated with the Plasmodium protein exporter PTEX and a loss of complex stability upon truncation of the core PTEX component, PTEX150. Cell. Microbiol. 2016, 18, 1551–1569. [Google Scholar] [CrossRef]
  79. Kulzer, S.; Charnaud, S.; Dagan, T.; Riedel, J.; Mandal, P.; Pesce, E.R.; Blatch, G.L.; Crabb, B.S.; Gilson, P.R.; Przyborski, J.M. Plasmodium falciparum-encoded exported hsp70/hsp40 chaperone/co-chaperone complexes within the host erythrocyte. Cell. Microbiol. 2012, 14, 1784–1795. [Google Scholar] [CrossRef]
  80. Knuepfer, E.; Rug, M.; Klonis, N.; Tilley, L.; Cowman, A.F. Trafficking of the major virulence factor to the surface of transfected P falciparum-infected erythrocytes. Blood 2005, 105, 4078–4087. [Google Scholar] [CrossRef]
  81. McHugh, E.; Carmo, O.M.S.; Blanch, A.; Looker, O.; Liu, B.; Tiash, S.; Andrew, D.; Batinovic, S.; Low, A.J.Y.; Cho, H.J.; et al. Role of Plasmodium falciparum protein GEXP07 in Maurer’s cleft morphology, knob architecture, and P. falciparum EMP1 trafficking. mBio 2020, 11, e03320-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. de Koning-Ward, T.F.; Dixon, M.W.; Tilley, L.; Gilson, P.R. Plasmodium species: Master renovators of their host cells. Nat. Rev. Microbiol. 2016, 14, 494–507. [Google Scholar] [CrossRef]
  83. Hiller, N.L.; Bhattacharjee, S.; van Ooij, C.; Liolios, K.; Harrison, T.; Lopez-Estraño, C.L.C.; Haldar, K. A host-targeting signal in virulence proteins reveals a secretome in malarial infection. Science 2004, 306, 1934–1937. [Google Scholar] [CrossRef]
  84. Marti, M.; Good, R.T.; Rug, M.; Knuepfer, E.; Cowman, A.F. Targeting malaria virulence and remodeling proteins to the host erythrocyte. Science 2004, 306, 1930–1933. [Google Scholar] [CrossRef] [PubMed]
  85. Boddey, J.A.; Hodder, A.N.; Günther, S.; Gilson, P.R.; Patsiouras, H.; Kapp, E.A.; Pearce, J.A.; de Koning-Ward, T.; Simpson, R.; Crabb, B.S.; et al. An aspartyl protease directs malaria effector proteins to the host cell. Nature 2010, 463, 627–631. [Google Scholar] [CrossRef] [PubMed]
  86. Russo, I.; Babbitt, S.; Muralidharan, V.; Butler, T.; Oksman, A.; Goldberg, D.E. Plasmepsin V licenses Plasmodium proteins for export into the host erythrocyte. Nature 2010, 463, 632–636. [Google Scholar] [CrossRef]
  87. Sleebs, B.E.; Lopaticki, S.; Marapana, D.S.; O’Neill, M.T.; Rajasekaran, P.; Gazdik, M.; Günther, S.; Whitehead, L.W.; Lowes, K.; Barfod, L.; et al. Inhibition of Plasmepsin V activity demonstrates its essential role in protein export, PfEMP1 display, and survival of malaria parasites. PLoS Biol. 2014, 12, e1001897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Marapana, D.S.; Dagley, L.F.; Sandow, J.J.; Nebl, T.; Triglia, T.; Pasternak, M.; Cowman, A.F. Plasmepsin V cleaves malaria effector proteins in a distinct endoplasmic reticulum translocation interactome for export to the erythrocyte. Nat. Microbiol. 2018, 3, 1010–1022. [Google Scholar] [CrossRef] [PubMed]
  89. Grüring, C.; Heiber, A.; Kruse, F.; Flemming, S.; Franci, G.; Colombo, S.F.; Fasana, E.; Schoeler, H.; Borgese, N.; Stunnenberg, H.G.; et al. Uncovering common principles in protein export of malaria parasites. Cell Host Microbe 2012, 12, 717–729. [Google Scholar] [CrossRef] [Green Version]
  90. Heiber, A.; Kruse, F.; Pick, C.; Grüring, C.; Flemming, S.; Oberli, A.; Schoeler, H.; Retzlaff, S.; Mesén-Ramírez, P.; Hiss, J.A.; et al. Identification of new PNEPs indicates a substantial non-PEXEL exportome and underpins common features in Plasmodium falciparum protein export. PLoS Pathog. 2013, 9, e1003546. [Google Scholar] [CrossRef]
  91. Spielmann, T.; Hawthorne, P.L.; Dixon, M.W.A.; Hannemann, M.; Klotz, K.; Kemp, D.J.; Klonis, N.; Tilley, L.; Trenholme, K.R.; Gardiner, D.L. A cluster of ring stage-specific genes linked to a locus implicated in cytoadherence in Plasmodium falciparum codes for PEXEL-negative and PEXEL-positive proteins exported into the host cell. Mol. Biol. Cell 2006, 17, 3613–3624. [Google Scholar] [CrossRef] [Green Version]
  92. Maier, A.G.; Rug, M.; O’Neill, M.T.; Brown, M.; Chakravorty, S.; Szestak, T.; Chesson, J.; Wu, Y.; Hughes, K.; Coppel, R.; et al. Exported Proteins required for virulence and rigidity of Plasmodium falciparum-infected human erythrocytes. Cell 2008, 134, 48–61. [Google Scholar] [CrossRef] [Green Version]
  93. Banumathy, G.; Singh, V.; Tatu, U. Host chaperones are recruited in membrane-bound complexes by Plasmodium falciparum. J. Biol. Chem. 2002, 277, 3902–3912. [Google Scholar] [CrossRef] [Green Version]
  94. de Koning-Ward, T.F.; Gilson, P.R.; Boddey, J.A.; Rug, M.; Smith, B.J.; Papenfuss, A.T.; Sanders, P.R.; Lundie, R.J.; Maier, A.; Cowman, A.F.; et al. A newly discovered protein export machine in malaria parasites. Nature 2009, 459, 945–949. [Google Scholar] [CrossRef] [Green Version]
  95. Riglar, D.T.; Rogers, K.L.; Hanssen, E.; Turnbull, L.; Bullen, H.E.; Charnaud, S.C.; Przyborski, J.; Gilson, P.R.; Whitchurch, C.; Crabb, B.S.; et al. Spatial association with PTEX complexes defines regions for effector export into Plasmodium falciparum-infected erythrocytes. Nat. Comm. 2013, 4, 1415. [Google Scholar] [CrossRef] [PubMed]
  96. Bullen, H.E.; Charnaud, S.C.; Kalanon, M.; Riglar, D.T.; Dekiwadia, C.; Kangwanrangsan, N.; Torii, M.; Tsuboi, T.; Baum, J.; Ralph, S.; et al. Biosynthesis, localization, and macromolecular arrangement of the Plasmodium falciparum translocon of exported proteins (PTEX). J. Biol. Chem. 2012, 287, 7871–7884. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Charnaud, S.C.; Dixon, M.W.A.; Nie, C.Q.; Chappell, L.; Sanders, P.R.; Nebl, T.; Hanssen, E.; Berriman, M.; Chan, J.-A.; Blanch, A.; et al. The exported chaperone Hsp70-x supports virulence functions for Plasmodium falciparum blood stage parasites. PLoS ONE 2017, 12, e0181656. [Google Scholar] [CrossRef] [Green Version]
  98. Ho, C.M.; Beck, J.R.; Lai, M.; Cui, Y.; Goldberg, D.E.; Egea, P.F.; Zhou, Z.H. Malaria parasite translocon structure and mechanism of effector export. Nature 2018, 561, 70–75. [Google Scholar] [CrossRef]
  99. Gehde, N.; Hinrichs, C.; Montilla, I.; Charpian, S.; Lingelbach, K.; Przyborski, J.M. Protein unfolding is an essential requirement for transport across the parasitophorous vacuolar membrane of Plasmodium falciparum. Mol. Microbiol. 2009, 71, 613–628. [Google Scholar] [CrossRef]
  100. Mesén-Ramírez, P.; Reinsch, F.; Soares, A.B.; Bergmann, B.; Ullrich, A.-K.; Tenzer, S.; Spielmann, T. Stable translocation intermediates jam global protein export in Plasmodium falciparum parasites and link the PTEX component EXP2 with translocation activity. PLoS Pathog. 2016, 12, e1005618. [Google Scholar] [CrossRef]
  101. Garten, M.; Nasamu, A.S.; Niles, J.C.; Zimmerberg, J.; Goldberg, D.E.; Beck, J.R. EXP2 is a nutrient-permeable channel in the vacuolar membrane of Plasmodium and is essential for protein export via PTEX. Nat. Microbiol. 2018, 3, 1090–1098. [Google Scholar] [CrossRef] [Green Version]
  102. Bonifacino, J.S.; Glick, B.S. The mechanisms of vesicle budding and fusion. Cell 2004, 116, 153–166. [Google Scholar] [CrossRef] [Green Version]
  103. Koike, S.; Jahn, R. SNAREs define targeting specificity of trafficking vesicles by combinatorial interaction with tethering factors. Nat. Commun. 2019, 10, 1608. [Google Scholar] [CrossRef] [PubMed]
  104. Sudhof, T.C.; Rothman, J.E. Membrane fusion: Grappling with SNARE and SM proteins. Science 2009, 323, 474–477. [Google Scholar] [CrossRef] [Green Version]
  105. Toonen, R.F.; Verhage, M. Vesicle trafficking: Pleasure and pain from SM genes. Trends Cell Biol. 2003, 13, 177–186. [Google Scholar] [CrossRef]
  106. Ouologuem, D.T.; Roos, D.S. Dynamics of the Toxoplasma gondii inner membrane complex. J. Cell Sci. 2014, 127, 3320–3330. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Hu, K.; Mann, T.; Striepen, B.; Beckers, C.J.; Roos, D.S.; Murray, J.M. Daughter cell assembly in the protozoan parasite Toxoplasma gondii. Mol. Biol. Cell 2002, 13, 593–606. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Dogga, S.K.; Mukherjee, B.; Jacot, D.; Kockmann, T.; Molino, L.; Hammoudi, P.M.; Hartkoorn, R.C.; Hehl, A.B.; Soldati-Favre, D. A druggable secretory protein maturase of Toxoplasma essential for invasion and egress. Elife 2017, 6, e27480. [Google Scholar] [CrossRef] [Green Version]
  109. Peng, R.; Gallwitz, D. Sly1 protein bound to Golgi syntaxin Sed5p allows assembly and contributes to specificity of SNARE fusion complexes. J. Cell Biol. 2002, 157, 645–655. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  110. Jonscher, E.; Flemming, S.; Schmitt, M.; Sabitzki, R.; Reichard, N.; Birnbaum, J.; Bergmann, B.; Katharina Hohn, K.; Spielmann, T. PfVPS45 is required for host cell cytosol uptake by malaria blood stage parasites. Cell Host Microbe 2019, 25, 166–173. [Google Scholar] [CrossRef] [Green Version]
  111. Kibria, K.M.K.; Rawat, K.; Klinger, C.M.; Datta, G.G.; Panchal, M.; Singh, S.; Iyer, G.R.; Kaur, I.; Sharma, V.; Dacks, J.B.; et al. A role for adaptor protein complex 1 in protein targeting to rhoptry organelles in Plasmodium falciparum. Biochem. Biophys. Acta 2015, 1853, 699–710. [Google Scholar] [CrossRef] [Green Version]
  112. Hirst, J.; Barlow, L.D.; Francisco, G.C.; Sahlender, D.A.; Seaman, M.N.; Dacks, J.B.; Robinson, M.S. The fifth adaptor protein complex. PLoS Biol. 2011, 9, e1001170. [Google Scholar] [CrossRef]
  113. Robinson, M.S. Adaptable adaptors for coated vesicles. Trends Cell Biol. 2014, 14, 167–174. [Google Scholar] [CrossRef]
  114. Collins, B.M.; McCoy, A.J.; Kent, H.M.; Evans, P.R.; Owen, D.J. Molecular architecture and functional model of the endocytic AP2 complex. Cell 2002, 109, 523–535. [Google Scholar] [CrossRef]
  115. Kelly, B.T.; McCoy, A.J.; Spate, K.; Miller, S.E.; Evans, P.R.; Honing, S.; Owen, D.J. A structural explanation for the binding of endocytic dileucine motifs by the AP2 complex. Nature 2008, 456, 976–979. [Google Scholar] [CrossRef] [Green Version]
  116. Hirst, J.; Borner, G.H.; Edgar, J.; Hein, M.Y.; Mann, M.; Buchholz, F.; Antrobus, R.; Robinson, M.S. Interaction between AP-5 and the hereditary spastic paraplegia proteins SPG11 and SPG15. Mol. Biol. Cell. 2013, 24, 2558–2569. [Google Scholar] [CrossRef] [PubMed]
  117. Hirst, J.; Irving, C.; Borner, G.H. Adaptor protein complexes AP-4 and AP-5: New players in endosomal trafficking and progressive spastic paraplegia. Traffic 2013, 14, 153–164. [Google Scholar] [CrossRef]
  118. Pelham, H.R. SNAREs and the specificity of membrane fusion. Trends Cell Biol. 2001, 11, 99–101. [Google Scholar] [CrossRef]
  119. Hong, W. SNAREs and traffic. Biochim. Biophys. Acta 2005, 1744, 120–144. [Google Scholar] [CrossRef] [Green Version]
  120. Jahn, R.; Scheller, R.H. SNAREs—Engines for membrane fusion. Nat. Rev. Mol. Cell. Biol. 2006, 7, 631–643. [Google Scholar] [CrossRef] [PubMed]
  121. Resh, M.D. Trafficking and signaling by fatty-acylated and prenylated proteins. Nat. Chem. Biol. 2006, 2, 584–590. [Google Scholar] [CrossRef]
  122. Michaelson, D.; Ali, W.; Chiu, V.K.; Bergo, M.; Silletti, J.; Wright, L.; Young, S.G.; Philips, M. Postprenylation CAAX processing is required for proper localization of Ras but not Rho GTPases. Mol. Biol. Cell 2005, 16, 1606–1616. [Google Scholar] [CrossRef] [Green Version]
  123. Yalovsky, S.; Rodr Guez-Concepcion, M.; Gruissem, W. Lipid modifications of proteins—Slipping in and out of membranes. Trends Plant Sci. 1999, 4, 439–445. [Google Scholar] [CrossRef]
  124. Perez-Sala, D. Protein isoprenylation in biology and disease: General overview and perspectives from studies with genetically engineered animals. Front. Biosci. 2007, 12, 4456–4472. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Wright, M.H.; Clough, B.; Rackham, M.D.; Rangachari, K.; Brannigan, J.A.; Grainger, M.; Moss, D.K.; Bottrill, A.R.; Heal, W.P.; Broncel, M.; et al. Validation of N-myristoyltransferase as an antimalarial drug target using an integrated chemical biology approach. Nat. Chem. 2014, 6, 112–121. [Google Scholar] [CrossRef] [Green Version]
  126. Gisselberg, J.E.; Zhang, L.; Elias, J.E.; Yeh, E. The prenylated proteome of Plasmodium falciparum reveals pathogen-specific prenylation activity and drug mechanism of action. Mol. Cell. Proteom. 2017, 16, S54–S64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Ayong, L.; DaSilva, T.T.; Mauser, J.; Allen, C.M.; Chakrabarti, D. Evidence for prenylation dependent-targeting of a Ykt6 SNARE in Plasmodium falciparum. Mol. Biochem. Parasitol. 2011, 175, 162–168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. Schematic representation of protein trafficking directed by P. falciparum merozoite-infected erythrocyte. The model outlined in this figure is a summary of the recent data on protein sorting and trafficking in P. falciparum and its relative, T. gondii. Secreted proteins are made in the rough ER (RER). Secretory vesicles from the RER are thought to be shipped to the cis-Golgi with the help of SLY1 and Stx5. Bidirectional sorting between the endo-lysosomal compartment (ELC) and the trans-Golgi network (TGN) is also mediated by Vsp45. Transport from the ER to the apicoplast is facilitated by unidentified SNARE proteins. Proteins destined to the micronemes and rhoptries are sorted by the adaptor Stx12. Endocytosis, which leads to the host cell cytosol uptake (HCCU), is likely to occur via the cytostome or the micropore (not shown). These vesicles are shuttled to ELC. The SNARE proteins involved in this process are unknown. Vsp45 is necessary for endocytosis and release of the cargo to the food vacuole. The knobs and some of the proteins involved in PfEMP1 trafficking are shown.
Figure 1. Schematic representation of protein trafficking directed by P. falciparum merozoite-infected erythrocyte. The model outlined in this figure is a summary of the recent data on protein sorting and trafficking in P. falciparum and its relative, T. gondii. Secreted proteins are made in the rough ER (RER). Secretory vesicles from the RER are thought to be shipped to the cis-Golgi with the help of SLY1 and Stx5. Bidirectional sorting between the endo-lysosomal compartment (ELC) and the trans-Golgi network (TGN) is also mediated by Vsp45. Transport from the ER to the apicoplast is facilitated by unidentified SNARE proteins. Proteins destined to the micronemes and rhoptries are sorted by the adaptor Stx12. Endocytosis, which leads to the host cell cytosol uptake (HCCU), is likely to occur via the cytostome or the micropore (not shown). These vesicles are shuttled to ELC. The SNARE proteins involved in this process are unknown. Vsp45 is necessary for endocytosis and release of the cargo to the food vacuole. The knobs and some of the proteins involved in PfEMP1 trafficking are shown.
Life 11 00937 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Mayer, D.C.G. Protein Sorting in Plasmodium Falciparum. Life 2021, 11, 937. https://0-doi-org.brum.beds.ac.uk/10.3390/life11090937

AMA Style

Mayer DCG. Protein Sorting in Plasmodium Falciparum. Life. 2021; 11(9):937. https://0-doi-org.brum.beds.ac.uk/10.3390/life11090937

Chicago/Turabian Style

Mayer, D.C. Ghislaine. 2021. "Protein Sorting in Plasmodium Falciparum" Life 11, no. 9: 937. https://0-doi-org.brum.beds.ac.uk/10.3390/life11090937

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop