ϟ

Isabelle Coppens

Here are all the papers by Isabelle Coppens that you can download and read on OA.mg.
Isabelle Coppens’s last known institution is . Download Isabelle Coppens PDFs here.

Claim this Profile →
DOI: 10.1084/jem.20061318
2006
Cited 341 times
Vacuolar and plasma membrane stripping and autophagic elimination of <i>Toxoplasma gondii</i> in primed effector macrophages
Apicomplexan protozoan pathogens avoid destruction and establish a replicative niche within host cells by forming a nonfusogenic parasitophorous vacuole (PV). Here we present evidence for lysosome-mediated degradation of Toxoplasma gondii after invasion of macrophages activated in vivo. Pathogen elimination was dependent on the interferon gamma inducible-p47 GTPase, IGTP, required PI3K activity, and was preceded by PV membrane indentation, vesiculation, disruption, and, surprisingly, stripping of the parasite plasma membrane. Denuded parasites were enveloped in autophagosome-like vacuoles, which ultimately fused with lysosomes. These observations outline a series of mechanisms used by effector cells to redirect the fate of a classically nonfusogenic intracellular pathogen toward a path of immune elimination.
DOI: 10.1016/j.cell.2006.01.056
2006
Cited 310 times
Toxoplasma gondii Sequesters Lysosomes from Mammalian Hosts in the Vacuolar Space
The intracellular compartment harboring Toxoplasma gondii satisfies the parasite's nutritional needs for rapid growth in mammalian cells. We demonstrate that the parasitophorous vacuole (PV) of T. gondii accumulates material coming from the host mammalian cell via the exploitation of the host endo-lysosomal system. The parasite actively recruits host microtubules, resulting in selective attraction of endo-lysosomes to the PV. Microtubule-based invaginations of the PV membrane serve as conduits for the delivery of host endo-lysosomes within the PV. These tubular conduits are decorated by a parasite coat, including the tubulogenic protein GRA7, which acts like a garrote that sequesters host endocytic organelles in the vacuolar space. These data define an unanticipated process allowing the parasite intimate and concentrated access to a diverse range of low molecular weight components produced by the endo-lysosomal system. More generally, they identify a unique mechanism for unidirectional transport and sequestration of host organelles.
DOI: 10.1126/science.1165740
2009
Cited 267 times
Rapid Membrane Disruption by a Perforin-Like Protein Facilitates Parasite Exit from Host Cells
Perforin-like proteins are expressed by many bacterial and protozoan pathogens, yet little is known about their function or mode of action. Here, we describe Toxoplasma perforin-like protein 1 (TgPLP1), a secreted perforin-like protein of the intracellular protozoan pathogen Toxoplasma gondii that displays structural features necessary for pore formation. After intracellular growth, TgPLP1-deficient parasites failed to exit normally, resulting in entrapment within host cells. We show that this defect is due to an inability to rapidly permeabilize the parasitophorous vacuole membrane and host plasma membrane during exit. TgPLP1 ablation had little effect on growth in culture but resulted in a reduction greater than five orders of magnitude of acute virulence in mice. Perforin-like proteins from other intracellular pathogens may play a similar role in microbial egress and virulence.
DOI: 10.1074/jbc.ra118.006472
2019
Cited 115 times
Listeria monocytogenes virulence factors, including listeriolysin O, are secreted in biologically active extracellular vesicles
Outer membrane vesicles produced by Gram-negative bacteria have been studied for half a century but the possibility that Gram-positive bacteria secrete extracellular vesicles (EVs) was not pursued until recently due to the assumption that the thick peptidoglycan cell wall would prevent their release to the environment. However, following their discovery in fungi, which also have cell walls, EVs have now been described for a variety of Gram-positive bacteria. EVs purified from Gram-positive bacteria are implicated in virulence, toxin release, and transference to host cells, eliciting immune responses, and spread of antibiotic resistance. Listeria monocytogenes is a Gram-positive bacterium that causes listeriosis. Here we report that L. monocytogenes produces EVs with diameters ranging from 20 to 200 nm, containing the pore-forming toxin listeriolysin O (LLO) and phosphatidylinositol-specific phospholipase C (PI-PLC). Cell-free EV preparations were toxic to mammalian cells, the murine macrophage cell line J774.16, in a LLO-dependent manner, evidencing EV biological activity. The deletion of plcA increased EV toxicity, suggesting PI-PLC reduced LLO activity. Using simultaneous metabolite, protein, and lipid extraction (MPLEx) multiomics we characterized protein, lipid, and metabolite composition of bacterial cells and secreted EVs and found that EVs carry the majority of listerial virulence proteins. Using immunogold EM we detected LLO at several organelles within infected human epithelial cells and with high-resolution fluorescence imaging we show that dynamic lipid structures are released from L. monocytogenes during infection. Our findings demonstrate that L. monocytogenes uses EVs for toxin release and implicate these structures in mammalian cytotoxicity.
DOI: 10.1016/j.ebiom.2022.103812
2022
Cited 46 times
Endothelial thrombomodulin downregulation caused by hypoxia contributes to severe infiltration and coagulopathy in COVID-19 patient lungs
Thromboembolism is a life-threatening manifestation of coronavirus disease 2019 (COVID-19). We investigated a dysfunctional phenotype of vascular endothelial cells in the lungs during COVID-19.We obtained the lung specimens from the patients who died of COVID-19. The phenotype of endothelial cells and immune cells was examined by flow cytometry and immunohistochemistry (IHC) analysis. We tested the presence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the endothelium using IHC and electron microscopy.The autopsy lungs of COVID-19 patients exhibited severe coagulation abnormalities, immune cell infiltration, and platelet activation. Pulmonary endothelial cells of COVID-19 patients showed increased expression of procoagulant von Willebrand factor (VWF) and decreased expression of anticoagulants thrombomodulin and endothelial protein C receptor (EPCR). In the autopsy lungs of COVID-19 patients, the number of macrophages, monocytes, and T cells was increased, showing an activated phenotype. Despite increased immune cells, adhesion molecules such as ICAM-1, VCAM-1, E-selectin, and P-selectin were downregulated in pulmonary endothelial cells of COVID-19 patients. Notably, decreased thrombomodulin expression in endothelial cells was associated with increased immune cell infiltration in the COVID-19 patient lungs. There were no SARS-CoV-2 particles detected in the lung endothelium of COVID-19 patients despite their dysfunctional phenotype. Meanwhile, the autopsy lungs of COVID-19 patients showed SARS-CoV-2 virions in damaged alveolar epithelium and evidence of hypoxic injury.Pulmonary endothelial cells become dysfunctional during COVID-19, showing a loss of thrombomodulin expression related to severe thrombosis and infiltration, and endothelial cell dysfunction might be caused by a pathologic condition in COVID-19 patient lungs rather than a direct infection with SARS-CoV-2.This work was supported by the Johns Hopkins University, the American Heart Association, and the National Institutes of Health.
DOI: 10.1083/jcb.149.1.167
2000
Cited 274 times
<i>Toxoplasma gondii</i> Exploits Host Low-Density Lipoprotein Receptor-Mediated Endocytosis for Cholesterol Acquisition
The obligate intracellular protozoan Toxoplasma gondii resides within a specialized parasitophorous vacuole (PV), isolated from host vesicular traffic. In this study, the origin of parasite cholesterol was investigated. T. gondii cannot synthesize sterols via the mevalonate pathway. Host cholesterol biosynthesis remains unchanged after infection and a blockade in host de novo sterol biosynthesis does not affect parasite growth. However, simultaneous limitation of exogenous and endogenous sources of cholesterol from the host cell strongly reduces parasite replication and parasite growth is stimulated by exogenously supplied cholesterol. Intracellular parasites acquire host cholesterol that is endocytosed by the low-density lipoprotein (LDL) pathway, a process that is specifically increased in infected cells. Interference with LDL endocytosis, with lysosomal degradation of LDL, or with cholesterol translocation from lysosomes blocks cholesterol delivery to the PV and significantly reduces parasite replication. Similarly, incubation of T. gondii in mutant cells defective in mobilization of cholesterol from lysosomes leads to a decrease of parasite cholesterol content and proliferation. This cholesterol trafficking to the PV is independent of the pathways involving the host Golgi or endoplasmic reticulum. Despite being segregated from the endocytic machinery of the host cell, the T. gondii vacuole actively accumulates LDL-derived cholesterol that has transited through host lysosomes.
DOI: 10.1111/j.1550-7408.1987.tb03216.x
1987
Cited 218 times
Receptor‐Mediated Endocytosis in the Bloodstream Form of <i>Trypanosoma brucei</i>1
The uptake of various host plasma proteins by the bloodstream form of Trypanosoma brucei was studied both biochemically, using radiolabeled proteins, and with the electron microscope, using colloidal gold particles as molecular tracers onto which plasma proteins had been adsorbed. Total plasma proteins and serum albumin were taken up by a mechanism of fluid endocytosis with low clearance (0.1 microliter [mg cell protein]-1 h-1), while low-density lipoprotein (LDL) and transferrin were taken up by a receptor-mediated process with a clearance of two to three orders of magnitude higher than that of serum albumin. Binding prior to uptake of LDL and transferrin was saturable, depended on the presence of Ca2+, and the labeled ligand could be displaced by the homologous but not by heterologous protein. Binding of gold-labeled proteins was seen only to the membrane of the flagellar pocket and not elsewhere on the plasma membrane. After 1 h of incubation at 30 degrees C with gold-labeled LDL and transferrin, labeled cellular structures represented respectively half and one-third of the total volume of all single-membrane bounded endocytotic and electron-dense vacuoles within the cell.
DOI: 10.1083/jcb.98.4.1178
1984
Cited 194 times
Purification, morphometric analysis, and characterization of the glycosomes (microbodies) of the protozoan hemoflagellate Trypanosoma brucei.
Trypanosoma brucei glycosomes (microbodies containing nine enzymes involved in glycolysis) have been purified to near homogeneity from bloodstream-form trypomastigotes for the purpose of morphologic and biochemical analysis. Differential centrifugation followed by two isopycnic centrifugations in an isotonic Percoll and in a sucrose gradient, respectively, resulted in 12- to 13-fold purified glycosomes with an overall yield of 31%. These glycosomes appeared to be highly pure and contained less than 1% mitochondrial contamination as judged by morphometric and biochemical analyses. In intact cells, glycosomes displayed a remarkably homogeneous size distribution centered on an average diameter of 0.27 micron with a standard deviation of 0.03 micron. The size distribution of isolated glycosomes differed only slightly from that measured in intact cells. One T. brucei cell contained on average 230 glycosomes, representing 4.3% of the total cell volume. The glycosomes were surrounded by a single membrane and contained as phospholipids only phosphatidyl choline and phosphatidyl ethanolamine in a ratio of 2:1. The purified glycosomal fraction had a very low DNA content of 0.18 microgram/mg protein. No DNA molecules were observed that could not have been derived from contaminating mitochondrial or nuclear debris.
DOI: 10.1038/nature00946
2002
Cited 189 times
Golgi biogenesis in Toxoplasma gondii
DOI: 10.1042/bj20060986
2007
Cited 188 times
The role of neutral lipid nanospheres in <i>Plasmodium falciparum</i> haem crystallization
The intraerythrocytic malaria parasite constructs an intracellular haem crystal, called haemozoin, within an acidic digestive vacuole where haemoglobin is degraded. Haem crystallization is the target of the widely used antimalarial quinoline drugs. The intracellular mechanism of molecular initiation of haem crystallization, whether by proteins, polar membrane lipids or by neutral lipids, has not been fully substantiated. In the present study, we show neutral lipid predominant nanospheres, which envelop haemozoin inside Plasmodium falciparum digestive vacuoles. Subcellular fractionation of parasite-derived haemozoin through a dense 1.7 M sucrose cushion identifies monoacylglycerol and diacylglycerol neutral lipids as well as some polar lipids in close association with the purified haemozoin. Global MS lipidomics detects monopalmitic glycerol and monostearic glycerol, but not mono-oleic glycerol, closely associated with haemozoin. The complex neutral lipid mixture rapidly initiates haem crystallization, with reversible pH-dependent quinoline inhibition associated with quinoline entry into the neutral lipid microenvironment. Neutral lipid nanospheres both enable haem crystallization in the presence of high globin concentrations and protect haem from H2O2 degradation. Conceptually, the present study shifts the intracellular microenvironment of haem crystallization and quinoline inhibition from a polar aqueous location to a non-polar neutral lipid nanosphere able to exclude water for efficient haem crystallization.
DOI: 10.1242/jcs.00194
2003
Cited 186 times
Myosin A tail domain interacting protein (MTIP) localizes to the inner membrane complex of<i>Plasmodium</i>sporozoites
Apicomplexan host cell invasion and gliding motility depend on the parasite's actomyosin system located beneath the plasma membrane of invasive stages. Myosin A (MyoA), a class XIV unconventional myosin, is the motor protein. A model has been proposed to explain how the actomyosin motor operates but little is known about the components, topology and connectivity of the motor complex. Using the MyoA neck and tail domain as bait in a yeast two-hybrid screen we identified MTIP, a novel 24 kDa protein that interacts with MyoA. Deletion analysis shows that the 15 amino-acid C-terminal tail domain of MyoA, rather than the neck domain, specifically interacts with MTIP. In Plasmodium sporozoites MTIP localizes to the inner membrane complex (IMC), where it is found clustered with MyoA. The data support a model for apicomplexan motility and invasion in which the MyoA motor protein is associated via its tail domain with MTIP, immobilizing it at the outer IMC membrane. The head domain of the immobilized MyoA moves actin filaments that, directly or via a bridging protein, connect to the cytoplasmic domain of a transmembrane protein of the TRAP family. The actin/TRAP complex is then redistributed by the stationary MyoA from the anterior to the posterior end of the zoite, leading to its forward movement on a substrate or to penetration of a host cell.
DOI: 10.1111/j.1365-2958.2010.07165.x
2010
Cited 156 times
Characterization of a novel organelle in Toxoplasma gondii with similar composition and function to the plant vacuole
Toxoplasma gondii belongs to the phylum Apicomplexa and is an important cause of congenital disease and infection in immunocompromised patients. Like most apicomplexans, T. gondii possesses several plant-like features, such as the chloroplast-like organelle, the apicoplast. We describe and characterize a novel organelle in T. gondii tachyzoites, which is visible by light microscopy and possesses a broad similarity to the plant vacuole. Electron tomography shows the interaction of this vacuole with other organelles. The presence of a plant-like vacuolar proton pyrophosphatase (TgVP1), a vacuolar proton ATPase, a cathepsin L-like protease (TgCPL), an aquaporin (TgAQP1), as well as Ca(2+)/H(+) and Na(+)/H(+) exchange activities, supports similarity to the plant vacuole. Biochemical characterization of TgVP1 in enriched fractions shows a functional similarity to the respective plant enzyme. The organelle is a Ca(2+) store and appears to have protective effects against salt stress potentially linked to its sodium transport activity. In intracellular parasites, the organelle fragments, with some markers colocalizing with the late endosomal marker, Rab7, suggesting its involvement with the endocytic pathway. Studies on the characterization of this novel organelle will be relevant to the identification of novel targets for chemotherapy against T. gondii and other apicomplexan parasites as well.
DOI: 10.1073/pnas.85.18.6753
1988
Cited 154 times
Receptors for the host low density lipoproteins on the hemoflagellate Trypanosoma brucei: purification and involvement in the growth of the parasite.
The slender bloodstream form of Trypanosoma brucei shows receptor-mediated endocytosis of low density lipoprotein (LDL) particles of its hosts. We have purified the LDL receptor of this species nearly to homogeneity (about 1000-fold purification) and obtained monospecific polyclonal antibodies against it. As analyzed by NaDodSO4/polyacrylamide gel electrophoresis, the purified receptor consists of a single subunit, with an apparent molecular mass of 86 kDa. Its isoelectric point is 5.9. On the average, each cell exposes 52,000 copies of low-affinity receptors (Kd of 250 nM) and 1800 copies of high-affinity receptors (Kd of 5.7 nM). According to indirect en bloc immunolabeling of fixed parasites, the receptor appears to be localized to the flagellar pocket membrane and the flagellar membrane and to be completely absent from the rest of the pericellular membrane. LDL is required for optimal growth of the trypanosome in vitro: cell growth can be inhibited either by removal of LDL from the culture medium or by antibodies against the purified LDL receptors. In both cases, growth is restored by the addition of excess LDL.
DOI: 10.1128/iai.00225-07
2007
Cited 147 times
<i>Plasmodium yoelii</i> Sporozoites with Simultaneous Deletion of P52 and P36 Are Completely Attenuated and Confer Sterile Immunity against Infection
Malaria infection starts when sporozoites are transmitted to the mammalian host during a mosquito bite. Sporozoites enter the blood circulation, reach the liver, and infect hepatocytes. The formation of a parasitophorous vacuole (PV) establishes their intracellular niche. Recently, two members of the 6-Cys domain protein family, P52 and P36, were each shown to play an important albeit nonessential role in Plasmodium berghei sporozoite infectivity for the rodent host. Here, we generated p52/p36-deficient Plasmodium yoelii parasites by the simultaneous deletion of both genes using a single genetic manipulation. p52/p36-deficient parasites exhibited normal progression through the life cycle during blood-stage infection, transmission to mosquitoes, mosquito-stage development, and sporozoite infection of the salivary glands. p52/p36-deficient sporozoites also showed normal motility and cell traversal activity. However, immunofluorescence analysis and electron microscopic observations revealed that p52/p36-deficient parasites did not form a PV within hepatocytes in vitro and in vivo. The p52/p36-deficient parasites localized as free entities in the host cell cytoplasm or the host cell nucleoplasm and did not develop as liver stages. Consequently, they did not cause blood-stage infections even at high sporozoite inoculation doses. Mice immunized with p52/p36-deficient sporozoites were completely protected against infectious sporozoite challenge. Our results demonstrate for the first time the generation of two-locus gene deletion-attenuated parasites that infect the liver but do not progress to blood-stage infection. The study will critically guide the design of Plasmodium falciparum live attenuated malaria vaccines.
DOI: 10.4161/auto.7.2.13310
2011
Cited 146 times
Autophagy in protists
Autophagy is the degradative process by which eukaryotic cells digest their own components using acid hydrolases within the lysosome. Originally thought to function almost exclusively in providing starving cells with nutrients taken from their own cellular constituents, autophagy is in fact involved in numerous cellular events including differentiation, turnover of macromolecules and organelles, and defense against parasitic invaders. During the last 10-20 years, molecular components of the autophagic machinery have been discovered, revealing a complex interactome of proteins and lipids, which, in a concerted way, induce membrane formation to engulf cellular material and target it for lysosomal degradation. Here, our emphasis is autophagy in protists. We discuss experimental and genomic data indicating that the canonical autophagy machinery characterized in animals and fungi appeared prior to the radiation of major eukaryotic lineages. Moreover, we describe how comparative bioinformatics revealed that this canonical machinery has been subject to moderation, outright loss or elaboration on multiple occasions in protist lineages, most probably as a consequence of diverse lifestyle adaptations. We also review experimental studies illustrating how several pathogenic protists either utilize autophagy mechanisms or manipulate host-cell autophagy in order to establish or maintain infection within a host. The essentiality of autophagy for the pathogenicity of many parasites, and the unique features of some of the autophagy-related proteins involved, suggest possible new targets for drug discovery. Further studies of the molecular details of autophagy in protists will undoubtedly enhance our understanding of the diversity and complexity of this cellular phenomenon and the opportunities it offers as a drug target.
DOI: 10.1084/jem.20082108
2009
Cited 137 times
Host ER–parasitophorous vacuole interaction provides a route of entry for antigen cross-presentation in <i>Toxoplasma gondii</i>–infected dendritic cells
Toxoplasma gondii tachyzoites infect host cells by an active invasion process leading to the formation of a specialized compartment, the parasitophorous vacuole (PV). PVs resist fusion with host cell endosomes and lysosomes and are thus distinct from phagosomes. Because the parasite remains sequestered within the PV, it is unclear how T. gondii-derived antigens (Ag's) access the major histocompatibility complex (MHC) class I pathway for presentation to CD8(+) T cells. We demonstrate that recruitment of host endoplasmic reticulum (hER) to the PV in T. gondii-infected dendritic cells (DCs) directly correlates with cross-priming of CD8(+) T cells. Furthermore, we document by immunoelectron microscopy the transfer of hER components into the PV, a process indicative of direct fusion between the two compartments. In strong contrast, no association between hER and phagosomes or Ag presentation activity was observed in DCs containing phagocytosed live or dead parasites. Importantly, cross-presentation of parasite-derived Ag in actively infected cells was blocked when hER retrotranslocation was inhibited, indicating that the hER serves as a conduit for the transport of Ag between the PV and host cytosol. Collectively, these findings demonstrate that pathogen-driven hER-PV interaction can serve as an important mechanism for Ag entry into the MHC class I pathway and CD8(+) T cell cross-priming.
DOI: 10.1371/journal.ppat.1006362
2017
Cited 127 times
Host lipid droplets: An important source of lipids salvaged by the intracellular parasite Toxoplasma gondii
Toxoplasma is an obligate intracellular parasite that replicates in mammalian cells within a parasitophorous vacuole (PV) that does not fuse with any host organelles. One mechanism developed by the parasite for nutrient acquisition is the attraction of host organelles to the PV. Here, we examined the exploitation of host lipid droplets (LD), ubiquitous fat storage organelles, by Toxoplasma. We show that Toxoplasma replication is reduced in host cells that are depleted of LD, or impaired in TAG lipolysis or fatty acid catabolism. In infected cells, the number of host LD and the expression of host LD-associated genes (ADRP, DGAT2), progressively increase until the onset of parasite replication. Throughout infection, the PV are surrounded by host LD. Toxoplasma is capable of accessing lipids stored in host LD and incorporates these lipids into its own membranes and LD. Exogenous addition of oleic acid stimulates LD biogenesis in the host cell and results in the overaccumulation of neutral lipids in very large LD inside the parasite. To access LD-derived lipids, Toxoplasma intercepts and internalizes within the PV host LD, some of which remaining associated with Rab7, which become wrapped by an intravacuolar network of membranes (IVN). Mutant parasites impaired in IVN formation display diminished capacity of lipid uptake from host LD. Moreover, parasites lacking an IVN-localized phospholipase A2 are less proficient in salvaging lipids from host LD in the PV, suggesting a major contribution of the IVN for host LD processing in the PV and, thus lipid content release. Interestingly, gavage of parasites with lipids unveils, for the first time, the presence in Toxoplasma of endocytic-like structures containing lipidic material originating from the PV lumen. This study highlights the reliance of Toxoplasma on host LD for its intracellular development and the parasite's capability in scavenging neutral lipids from host LD.
DOI: 10.1128/mbio.00808-16
2016
Cited 125 times
<i>In Vivo</i> Biotinylation of the <i>Toxoplasma</i> Parasitophorous Vacuole Reveals Novel Dense Granule Proteins Important for Parasite Growth and Pathogenesis
ABSTRACT Toxoplasma gondii is an obligate intracellular parasite that invades host cells and replicates within a unique parasitophorous vacuole. To maintain this intracellular niche, the parasite secretes an array of dense granule proteins (GRAs) into the nascent parasitophorous vacuole. These GRAs are believed to play key roles in vacuolar remodeling, nutrient uptake, and immune evasion while the parasite is replicating within the host cell. Despite the central role of GRAs in the Toxoplasma life cycle, only a subset of these proteins have been identified, and many of their roles have not been fully elucidated. In this report, we utilize the promiscuous biotin ligase BirA* to biotinylate GRA proteins secreted into the vacuole and then identify those proteins by affinity purification and mass spectrometry. Using GRA-BirA* fusion proteins as bait, we have identified a large number of known and candidate GRAs and verified localization of 13 novel GRA proteins by endogenous gene tagging. We proceeded to functionally characterize three related GRAs from this group (GRA38, GRA39, and GRA40) by gene knockout. While Δ gra38 and Δ gra40 parasites showed no altered phenotype, disruption of GRA39 results in slow-growing parasites that contain striking lipid deposits in the parasitophorous vacuole, suggesting a role in lipid regulation that is important for parasite growth. In addition, parasites lacking GRA39 showed dramatically reduced virulence and a lower tissue cyst burden in vivo . Together, the findings from this work reveal a partial vacuolar proteome of T. gondii and identify a novel GRA that plays a key role in parasite replication and pathogenesis. IMPORTANCE Most intracellular pathogens reside inside a membrane-bound vacuole within their host cell that is extensively modified by the pathogen to optimize intracellular growth and avoid host defenses. In Toxoplasma , this vacuole is modified by a host of secretory GRA proteins, many of which remain unidentified. Here we demonstrate that in vivo biotinylation of proximal and interacting proteins using the promiscuous biotin ligase BirA* is a powerful approach to rapidly identify vacuolar GRA proteins. We further demonstrate that one factor identified by this approach, GRA39, plays an important role in the ability of the parasite to replicate within its host cell and cause disease.
DOI: 10.1111/j.1365-2958.2010.07181.x
2010
Cited 124 times
Cathepsin L occupies a vacuolar compartment and is a protein maturase within the endo/exocytic system of Toxoplasma gondii
Summary Regulated exocytosis allows the timely delivery of proteins and other macromolecules precisely when they are needed to fulfil their functions. The intracellular parasite Toxoplasma gondii has one of the most extensive regulated exocytic systems among all unicellular organisms, yet the basis of protein trafficking and proteolytic modification in this system is poorly understood. We demonstrate that a parasite cathepsin protease, TgCPL, occupies a newly recognized vacuolar compartment (VAC) that undergoes dynamic fragmentation during T. gondii replication. We also provide evidence that within the VAC or late endosome this protease mediates the proteolytic maturation of proproteins targeted to micronemes, regulated secretory organelles that deliver adhesive proteins to the parasite surface during cell invasion. Our findings suggest that processing of microneme precursors occurs within intermediate endocytic compartments within the exocytic system, indicating an extensive convergence of the endocytic and exocytic pathways in this human parasite.
DOI: 10.1016/j.molbiopara.2011.02.003
2011
Cited 115 times
Autophagy in parasitic protists: Unique features and drug targets
Eukaryotic cells can degrade their own components, cytosolic proteins and organelles, using dedicated hydrolases contained within the acidic interior of their lysosomes. This degradative process, called autophagy, is used under starvation conditions to recycle redundant or less important macromolecules, facilitates metabolic re-modeling in response to environmental cues, and is also often important during cell differentiation. In this review, we discuss the role played by autophagy during the life cycles of the major parasitic protists. To provide context, we also provide an overview of the different forms of autophagy and the successive steps in the autophagic processes, including the proteins involved, as revealed in recent decades by studies using the model organism Saccharomyces cerevisiae, methylotrophic yeasts and mammalian cells. We describe for trypanosomatid parasites how autophagy plays a role in the differentiation from one life cycle stage to the next one and, in the case of the intracellular parasites, for virulence. For malarial parasites, although only a limited repertoire of canonical autophagy-related proteins can be detected, autophagy seems to play a role in the removal of redundant organelles important for cell invasion, when sporozoites develop into intracellular trophozoites inside the hepatocytes. The complete absence of a canonical autophagy pathway from the microaerophile Giardia lamblia is also discussed. Finally, the essential role of autophagy for differentiation and pathogenicity of some pathogenic protists suggests that the proteins involved in this process may represent new targets for drug development. Opportunities and strategies for drug design targeting autophagy proteins are discussed.
DOI: 10.1371/journal.ppat.1005211
2015
Cited 106 times
Fundamental Roles of the Golgi-Associated Toxoplasma Aspartyl Protease, ASP5, at the Host-Parasite Interface
Toxoplasma gondii possesses sets of dense granule proteins (GRAs) that either assemble at, or cross the parasitophorous vacuole membrane (PVM) and exhibit motifs resembling the HT/PEXEL previously identified in a repertoire of exported Plasmodium proteins. Within Plasmodium spp., cleavage of the HT/PEXEL motif by the endoplasmic reticulum-resident protease Plasmepsin V precedes trafficking to and export across the PVM of proteins involved in pathogenicity and host cell remodelling. Here, we have functionally characterized the T. gondii aspartyl protease 5 (ASP5), a Golgi-resident protease that is phylogenetically related to Plasmepsin V. We show that deletion of ASP5 causes a significant loss in parasite fitness in vitro and an altered virulence in vivo. Furthermore, we reveal that ASP5 is necessary for the cleavage of GRA16, GRA19 and GRA20 at the PEXEL-like motif. In the absence of ASP5, the intravacuolar nanotubular network disappears and several GRAs fail to localize to the PVM, while GRA16 and GRA24, both known to be targeted to the host cell nucleus, are retained within the vacuolar space. Additionally, hypermigration of dendritic cells and bradyzoite cyst wall formation are impaired, critically impacting on parasite dissemination and persistence. Overall, the absence of ASP5 dramatically compromises the parasite's ability to modulate host signalling pathways and immune responses.
DOI: 10.1073/pnas.1103657108
2011
Cited 101 times
<i>Plasmodium</i> ookinetes coopt mammalian plasminogen to invade the mosquito midgut
Ookinete invasion of the mosquito midgut is an essential step for the development of the malaria parasite in the mosquito. Invasion involves recognition between a presumed mosquito midgut receptor and an ookinete ligand. Here, we show that enolase lines the ookinete surface. An antienolase antibody inhibits oocyst development of both Plasmodium berghei and Plasmodium falciparum, suggesting that enolase may act as an invasion ligand. Importantly, we demonstrate that surface enolase captures plasminogen from the mammalian blood meal via its lysine motif (DKSLVK) and that this interaction is essential for midgut invasion, because plasminogen depletion leads to a strong inhibition of oocyst formation. Although addition of recombinant WT plasminogen to depleted serum rescues oocyst formation, recombinant inactive plasminogen does not, thus emphasizing the importance of plasmin proteolytic activity for ookinete invasion. The results support the hypothesis that enolase on the surface of Plasmodium ookinetes plays a dual role in midgut invasion: by acting as a ligand that interacts with the midgut epithelium and, further, by capturing plasminogen, whose conversion to active plasmin promotes the invasion process.
DOI: 10.1091/mbc.e12-11-0827
2013
Cited 98 times
<i>Toxoplasma gondii</i>salvages sphingolipids from the host Golgi through the rerouting of selected Rab vesicles to the parasitophorous vacuole
The obligate intracellular protozoan Toxoplasma gondii actively invades mammalian cells and, upon entry, forms its own membrane-bound compartment, named the parasitophorous vacuole (PV). Within the PV, the parasite replicates and scavenges nutrients, including lipids, from host organelles. Although T. gondii can synthesize sphingolipids de novo, it also scavenges these lipids from the host Golgi. How the parasite obtains sphingolipids from the Golgi remains unclear, as the PV avoids fusion with host organelles. In this study, we explore the host Golgi–PV interaction and evaluate the importance of host-derived sphingolipids for parasite growth. We demonstrate that the PV preferentially localizes near the host Golgi early during infection and remains closely associated with this organelle throughout infection. The parasite subverts the structure of the host Golgi, resulting in its fragmentation into numerous ministacks, which surround the PV, and hijacks host Golgi–derived vesicles within the PV. These vesicles, marked with Rab14, Rab30, or Rab43, colocalize with host-derived sphingolipids in the vacuolar space. Scavenged sphingolipids contribute to parasite replication since alterations in host sphingolipid metabolism are detrimental for the parasite's growth. Thus our results reveal that T. gondii relies on host-derived sphingolipids for its development and scavenges these lipids via Golgi-derived vesicles.
DOI: 10.1038/nmicrobiol.2017.17
2017
Cited 86 times
Plasmodium falciparum CRK4 directs continuous rounds of DNA replication during schizogony
Plasmodium parasites, the causative agents of malaria, have evolved a unique cell division cycle in the clinically relevant asexual blood stage of infection1. DNA replication commences approximately halfway through the intracellular development following invasion and parasite growth. The schizont stage is associated with multiple rounds of DNA replication and nuclear division without cytokinesis, resulting in a multinucleated cell. Nuclei divide asynchronously through schizogony, with only the final round of DNA replication and segregation being synchronous and coordinated with daughter cell assembly2,3. However, the control mechanisms for this divergent mode of replication are unknown. Here, we show that the Plasmodium-specific kinase PfCRK4 is a key cell-cycle regulator that orchestrates multiple rounds of DNA replication throughout schizogony in Plasmodium falciparum. PfCRK4 depletion led to a complete block in nuclear division and profoundly inhibited DNA replication. Quantitative phosphoproteomic profiling identified a set of PfCRK4-regulated phosphoproteins with greatest functional similarity to CDK2 substrates, particularly proteins involved in the origin of replication firing. PfCRK4 was required for initial and subsequent rounds of DNA replication during schizogony and, in addition, was essential for development in the mosquito vector. Our results identified an essential S-phase promoting factor of the unconventional P. falciparum cell cycle. PfCRK4 is required for both a prolonged period of the intraerythrocytic stage of Plasmodium infection, as well as for transmission, revealing a broad window for PfCRK4-targeted chemotherapeutics.
DOI: 10.1182/blood-2017-11-814665
2018
Cited 84 times
Remodeling of the malaria parasite and host human red cell by vesicle amplification that induces artemisinin resistance
Key Points Vesicular system causing artemisinin resistance modifies malaria parasites and host red cells.
DOI: 10.1126/sciadv.abc9955
2020
Cited 83 times
Mitochondria form contact sites with the nucleus to couple prosurvival retrograde response
Mitochondria drive cellular adaptation to stress by retro-communicating with the nucleus. This process is known as mitochondrial retrograde response (MRR) and is induced by mitochondrial dysfunction. MRR results in the nuclear stabilization of prosurvival transcription factors such as the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Here, we demonstrate that MRR is facilitated by contact sites between mitochondria and the nucleus. The translocator protein (TSPO) by preventing the mitophagy-mediated segregation o mitochonria is required for this interaction. The complex formed by TSPO with the protein kinase A (PKA), via the A-kinase anchoring protein acyl-CoA binding domain containing 3 (ACBD3), established the tethering. The latter allows for cholesterol redistribution of cholesterol in the nucleus to sustain the prosurvival response by blocking NF-κB deacetylation. This work proposes a previously unidentified paradigm in MRR: the formation of contact sites between mitochondria and nucleus to aid communication.
DOI: 10.1084/jem.20161702
2017
Cited 77 times
Eosinophil-derived IL-4 drives progression of myocarditis to inflammatory dilated cardiomyopathy
Inflammatory dilated cardiomyopathy (DCMi) is a major cause of heart failure in children and young adults. DCMi develops in up to 30% of myocarditis patients, but the mechanisms involved in disease progression are poorly understood. Patients with eosinophilia frequently develop cardiomyopathies. In this study, we used the experimental autoimmune myocarditis (EAM) model to determine the role of eosinophils in myocarditis and DCMi. Eosinophils were dispensable for myocarditis induction but were required for progression to DCMi. Eosinophil-deficient ΔdblGATA1 mice, in contrast to WT mice, showed no signs of heart failure by echocardiography. Induction of EAM in hypereosinophilic IL-5Tg mice resulted in eosinophilic myocarditis with severe ventricular and atrial inflammation, which progressed to severe DCMi. This was not a direct effect of IL-5, as IL-5TgΔdblGATA1 mice were protected from DCMi, whereas IL-5-/- mice exhibited DCMi comparable with WT mice. Eosinophils drove progression to DCMi through their production of IL-4. Our experiments showed eosinophils were the major IL-4-expressing cell type in the heart during EAM, IL-4-/- mice were protected from DCMi like ΔdblGATA1 mice, and eosinophil-specific IL-4 deletion resulted in improved heart function. In conclusion, eosinophils drive progression of myocarditis to DCMi, cause severe DCMi when present in large numbers, and mediate this process through IL-4.
DOI: 10.1038/nmicrobiol.2017.96
2017
Cited 71 times
Toxoplasma depends on lysosomal consumption of autophagosomes for persistent infection
Globally, nearly 2 billion people are infected with the intracellular protozoan Toxoplasma gondii1. This persistent infection can cause severe disease in immunocompromised people and is epidemiologically linked to major mental illnesses2 and cognitive impairment3. There are currently no options for curing this infection. The lack of effective therapeutics is due partly to a poor understanding of the essential pathways that maintain long-term infection. Although it is known that Toxoplasma replicates slowly within intracellular cysts demarcated with a cyst wall, precisely how it sustains itself and remodels organelles in this niche is unknown. Here, we identify a key role for proteolysis within the parasite lysosomal organelle (the vacuolar compartment or VAC) in turnover of autophagosomes and persistence during neural infection. We found that disrupting a VAC-localized cysteine protease compromised VAC digestive function and markedly reduced chronic infection. Death of parasites lacking the VAC protease was preceded by accumulation of undigested autophagosomes in the parasite cytoplasm. These findings suggest an unanticipated function for parasite lysosomal degradation in chronic infection, and identify an intrinsic role for autophagy in the T. gondii parasite and its close relatives. This work also identifies a key element of Toxoplasma persistence and suggests that VAC proteolysis is a prospective target for pharmacological development. Disruption of a cysteine protease that localizes to the vacuolar compartment of Toxoplasma gondii shows that autophagy is required for the intracellular survival of the parasite during chronic infection.
DOI: 10.3389/fcimb.2020.00269
2020
Cited 53 times
Fussing About Fission: Defining Variety Among Mainstream and Exotic Apicomplexan Cell Division Modes
Cellular reproduction defines life, yet our textbook-level understanding of cell division is limited to a small number of model organisms centered around humans. The horizon on cell division variants is expanded here by advancing insights on the fascinating cell division modes found in the Apicomplexa, a key group of protozoan parasites. The Apicomplexa display remarkable variation in offspring number, whether karyokinesis follows each S/M-phase or not, and whether daughter cells bud in the cytoplasm or bud from the cortex. We find that the terminology used to describe the various manifestations of asexual apicomplexan cell division emphasizes either the number of offspring or site of budding, which are not directly comparable features and has led to confusion in the literature. Division modes have been primarily studied in two human pathogenic Apicomplexa, malaria-causing Plasmodium spp. and Toxoplasma gondii, a major cause of opportunistic infections. Plasmodium spp. divide asexually by schizogony, producing multiple daughters per division round through a cortical budding process, though at several life-cycle nuclear amplifications are not followed by karyokinesis. T. gondii divides by endodyogeny producing two internally budding daughters per division round. Here we add to this diversity in replication mechanisms by considering the cattle parasite Babesia bigemina and the pig parasite Cystoisospora suis. B. bigemina produces two daughters per division round by a ‘binary fission’ mechanism whereas C. suis produces daughters through both endodyogeny and multiple internal budding known as endopolygeny. In addition, we provide new data from the causative agent of equine protozoal myeloencephalitis (EPM), Sarcocystis neurona, which also undergoes endopolygeny but differs from C. suis by maintaining a single multiploid nucleus. Overall, we operationally define two principally different division modes: internal budding found in cyst-forming Coccidia (comprising endodyogeny and two forms of endopolygeny) and external budding found in the other parasites studied (comprising the two forms of schizogony, binary fission and multiple fission). Progressive insights into the principles defining the molecular and cellular requirements for internal versus external budding, as well as variations encountered in sexual stages are discussed. The evolutionary pressures and mechanisms underlying apicomplexan cell division diversification carries relevance across Eukaryota.
DOI: 10.1126/science.adf8141
2023
Cited 14 times
<i>Delftia tsuruhatensis</i> TC1 symbiont suppresses malaria transmission by anopheline mosquitoes
Malaria control demands the development of a wide range of complementary strategies. We describe the properties of a naturally occurring, non–genetically modified symbiotic bacterium, Delftia tsuruhatensis TC1, which was isolated from mosquitoes incapable of sustaining the development of Plasmodium falciparum parasites. D. tsuruhatensis TC1 inhibits early stages of Plasmodium development and subsequent transmission by the Anopheles mosquito through secretion of a small-molecule inhibitor. We have identified this inhibitor to be the hydrophobic molecule harmane. We also found that, on mosquito contact, harmane penetrates the cuticle, inhibiting Plasmodium development. D. tsuruhatensis TC1 stably populates the mosquito gut, does not impose a fitness cost on the mosquito, and inhibits Plasmodium development for the mosquito’s life. Contained field studies in Burkina Faso and modeling showed that D. tsuruhatensis TC1 has the potential to complement mosquito-targeted malaria transmission control.
DOI: 10.1038/s41586-023-06821-y
2023
Cited 10 times
In vitro production of cat-restricted Toxoplasma pre-sexual stages
Sexual reproduction of Toxoplasma gondii, confined to the felid gut, remains largely uncharted owing to ethical concerns regarding the use of cats as model organisms. Chromatin modifiers dictate the developmental fate of the parasite during its multistage life cycle, but their targeting to stage-specific cistromes is poorly described1,2. Here we found that the transcription factors AP2XII-1 and AP2XI-2 operate during the tachyzoite stage, a hallmark of acute toxoplasmosis, to silence genes necessary for merozoites, a developmental stage critical for subsequent sexual commitment and transmission to the next host, including humans. Their conditional and simultaneous depletion leads to a marked change in the transcriptional program, promoting a full transition from tachyzoites to merozoites. These in vitro-cultured pre-gametes have unique protein markers and undergo typical asexual endopolygenic division cycles. In tachyzoites, AP2XII-1 and AP2XI-2 bind DNA as heterodimers at merozoite promoters and recruit MORC and HDAC3 (ref. 1), thereby limiting chromatin accessibility and transcription. Consequently, the commitment to merogony stems from a profound epigenetic rewiring orchestrated by AP2XII-1 and AP2XI-2. Successful production of merozoites in vitro paves the way for future studies on Toxoplasma sexual development without the need for cat infections and holds promise for the development of therapies to prevent parasite transmission.
DOI: 10.1091/mbc.e03-06-0355
2003
Cited 156 times
Sites of Interaction between Aldolase and Thrombospondin-related Anonymous Protein in<i>Plasmodium</i>
Gliding motility and host cell invasion by apicomplexan parasites are empowered by an acto-myosin motor located underneath the parasite plasma membrane. The motor is connected to host cell receptors through trans-membrane invasins belonging to the thrombospondin-related anonymous protein (TRAP) family. A recent study indicates that aldolase bridges the cytoplasmic tail of MIC2, the homologous TRAP protein in Toxoplasma, and actin. Here, we confirm these unexpected findings in Plasmodium sporozoites and identify conserved features of the TRAP family cytoplasmic tail required to bind aldolase: a subterminal tryptophan residue and two noncontiguous stretches of negatively charged amino acids. The aldolase substrate and other compounds that bind to the active site inhibit its interaction with TRAP and with F-actin, suggesting that the function of the motor is metabolically regulated. Ultrastructural studies in salivary gland sporozoites localize aldolase to the periphery of the secretory micronemes containing TRAP. Thus, the interaction between aldolase and the TRAP tail takes place during or preceding the biogenesis of the micronemes. The release of their contents in the anterior pole of the parasite upon contact with the target cells should bring simultaneously aldolase, TRAP and perhaps F-actin to the proper subcellular location where the motor is engaged.
DOI: 10.1242/jcs.00683
2003
Cited 153 times
Activation of NF-κB by<i>Toxoplasma gondii</i>correlates with increased expression of antiapoptotic genes and localization of phosphorylated IκB to the parasitophorous vacuole membrane
Mammalian cells infected with Toxoplasma gondii are resistant to apoptosis induced by a variety of stimuli. We have demonstrated that the host transcription factor NF-kappaB plays a pivotal role in the T.-gondii-mediated blockade of apoptosis because inhibition is lost in cells lacking the p65 (RelA) subunit of NF-kappaB (p65-/-). In the present study, we examined the effects of T. gondii infection on NF-kappaB activation and the expression of genes involved in the apoptotic cascade. Infection of wild-type mouse embryonic fibroblasts (MEFs) with T.-gondii-induced nuclear translocation of the p50 and p65 subunits of NF-kappaB as examined by immunoblotting of nuclear extracts, immunofluorescence and electrophoretic mobility shift assays. A comparison of apoptotic gene expression profiles from wild-type and p65-/- MEFs revealed distinct patterns of induction in response to T. gondii infection. In particular, the differences seen in the Bcl-2 and IAP families are consistent with the antiapoptotic responses observed in the resistant wild-type cells compared with the sensitive p65-/- fibroblasts. Consistent with NF-kappaB activation, T. gondii infection promoted phosphorylation of the inhibitor IkappaB. Interestingly, phosphorylated IkappaB was concentrated on the parasitophorous vacuole membrane (PVM), suggesting a parasite-directed event. Results from this study suggest that activation of NF-kappaB plays an important role in stimulation of antiapoptotic gene expression by T. gondii. Furthermore, recruitment of phosphorylated IkappaB to the PVM implies the presence of intrinsic factor(s) in T. gondii that might be used to manipulate the NF-kappaB signaling pathway in the host to elicit a survival response during infection.
DOI: 10.1091/mbc.e02-12-0830
2003
Cited 139 times
Host but Not Parasite Cholesterol Controls<i>Toxoplasma</i>Cell Entry by Modulating Organelle Discharge
Host cell cholesterol is implicated in the entry and replication of an increasing number of intracellular microbial pathogens. Although uptake of viral particles via cholesterol-enriched caveolae is increasingly well described, the requirement of cholesterol for internalization of eukaryotic pathogens is poorly understood and is likely to be partly organism specific. We examined the role of cholesterol in active host cell invasion by the protozoan parasite Toxoplasma gondii. The parasitophorous vacuole membrane (PVM) surrounding T. gondii contains cholesterol at the time of invasion. Although cholesterol-enriched parasite apical organelles termed rhoptries discharge at the time of cell entry and contribute to PVM formation, surprisingly, rhoptry cholesterol is not necessary for this process. In contrast, host plasma membrane cholesterol is incorporated into the forming PVM during invasion, through a caveolae-independent mechanism. Unexpectedly, depleting host cell plasma membrane cholesterol blocks parasite internalization by reducing the release of rhoptry proteins that are necessary for invasion. Cholesterol back-addition into host plasma membrane reverses this inhibitory effect of depletion on parasite secretion. These data define a new mechanism by which host cholesterol specifically controls entry of an intracellular pathogen.
DOI: 10.1074/jbc.m800681200
2008
Cited 116 times
Translation Regulation by Eukaryotic Initiation Factor-2 Kinases in the Development of Latent Cysts in Toxoplasma gondii
A key problem in the treatment of numerous pathogenic eukaryotes centers on their development into latent forms during stress. For example, the opportunistic protist Toxoplasma gondii converts to latent cysts (bradyzoites) responsible for recrudescence of disease. We report that Toxoplasma eukaryotic initiation factor-2alpha (TgIF2alpha) is phosphorylated during stress and establish that protozoan parasites utilize translation control to modulate gene expression during development. Importantly, TgIF2alpha remains phosphorylated in bradyzoites, explaining how these cells maintain their quiescent state. Furthermore, we have characterized novel eIF2 kinases; one in the endoplasmic reticulum and a likely regulator of the unfolded protein response (TgIF2K-A) and another that is a probable responder to cytoplasmic stresses (TgIF2K-B). Significantly, our data suggest that 1) the regulation of protein translation through eIF2 kinases is associated with development, 2) eIF2alpha phosphorylation is employed by cells to maintain a latent state, and 3) endoplasmic reticulum and cytoplasmic stress responses evolved in eukaryotic cells before the early diverging Apicomplexa. Given its importance to pathogenesis, eIF2 kinase-mediated stress responses may provide opportunities for novel therapeutics.
DOI: 10.1016/j.pt.2003.10.011
2004
Cited 116 times
Apicomplexan gliding motility and host cell invasion: overhauling the motor model
<h2>Abstract</h2> A unique actomyosin motor powers both host cell invasion and locomotion of apicomplexan invasive stages. This article integrates a revised model of some of the recent advances in our understanding of the components and the overall molecular architecture of this intriguing motion system. Many questions remain, however, regarding the dynamics and regulation of the motor complex.
DOI: 10.1016/j.ijpara.2007.04.005
2007
Cited 109 times
Cellular interactions of Plasmodium liver stage with its host mammalian cell
The Plasmodium liver forms are bridgehead stages between the mosquito sporozoite stages and mammalian blood stages that instigate the malaria disease. In hepatocytes, Plasmodium achieves one of the fastest growth rates among eukaryotic cells. However, nothing is known about host hepatic cell interactions, e.g. nutrient scavenging and/or subversion of cellular functions necessary for Plasmodium development and replication. Plasmodium usually invades hepatocytes by establishing a parasitophorous vacuole wherein it undergoes multiple nuclear division cycles. We show that Plasmodium preferentially develops in the host juxtanuclear region. By comparison with the parasitophorous vacuole of other apicomplexan parasites which associate with diverse host organelles, the Plasmodium parasitophorous vacuole only forms an association with the host endoplasmic reticulum. Intrahepatic Plasmodium actively modifies the permeability of its vacuole to allow the transfer of a large variety of molecules from the host cytosol to the vacuolar space through open channels. In contrast with malaria blood stages, the pores within the parasitophorous vacuole membrane of the liver stage display a smaller size as they restrict the passage of solutes to less than 855Da. These pores are stably maintained during parasite karyokinesis until complete cellularisation. Host-derived cholesterol accumulated at the parasitophorous vacuole membrane may modulate the channel activity. These observations define the parasitophorous vacuole of the Plasmodium liver stage as a dynamic and highly permeable compartment that can ensure the sustained supply of host molecules to support parasite growth in the nutrient-rich environment of liver cells.
DOI: 10.1111/j.1462-5822.2010.01555.x
2010
Cited 104 times
Plasmodium salvages cholesterol internalized by LDL and synthesized de novo in the liver
Our previous morphological studies illustrated the association of sterols with Plasmodium infecting hepatocytes. Because malaria parasites cannot synthesize sterols, they must scavenge these lipids from the host. In this paper, we have examined the source/s of sterols for intrahepatic Plasmodium and evaluated the importance of sterols for liver stage development. We show that Plasmodium continuously diverts cholesterol from hepatocytes until release of merozoites. Removal of plasma lipoproteins from the medium results in a 70% reduction of cholesterol content in hepatic merozoites but these parasites remain infectious in animals. Plasmodium salvages cholesterol that has been internalized by low-density lipoprotein but reduced expression of host low-density lipoprotein receptors by 70% does not influence liver stage burden. Plasmodium is also able to intercept cholesterol synthesized by hepatocytes. Pharmacological blockade of host squalene synthase or downregulation of the expression of this enzyme by 80% decreases by twofold the cholesterol content of merozoites without further impacting parasite development. These data enlighten that, on one hand, malaria parasites have moderate need of sterols for optimal development in hepatocytes and, on the other hand, they can adapt to survive in cholesterol-restrictive conditions by exploitation of accessible sterols derived from alternative sources in hepatocytes to maintain proper infectivity.
DOI: 10.1111/j.1365-2958.2008.06271.x
2008
Cited 102 times
Targeted deletion of <i>SAP1</i> abolishes the expression of infectivity factors necessary for successful malaria parasite liver infection
Malaria parasite sporozoites prepare for transmission to a mammalian host by upregulation of UIS (Upregulated in Infectious Sporozoites) genes. A number of UIS gene products are essential for the establishment of the intrahepatocytic niche. However, the factors that regulate the expression of genes involved in gain of infectivity for the liver are unknown. Herein, we show that a conserved Plasmodium sporozoite low-complexity asparagine-rich protein, SAP1 (Sporozoite Asparagine-rich Protein 1), has an essential role in malaria parasite liver infection. Targeted deletion of SAP1 in the rodent malaria parasite Plasmodium yoelii generated mutant parasites that traverse and invade hepatocytes normally but cannot initiate liver-stage development in vitro and in vivo. Moreover, immunizations with Pysap1(-) sporozoites confer long-lasting sterile protection against wild-type sporozoite infection. Strikingly, lack of SAP1 abolished expression of essential UIS genes including UIS3, UIS4 and P52 but not the constitutively expressed genes encoding, among others, sporozoite proteins CSP and TRAP. SAP1 localization to the cell interior but not the nucleus of sporozoites suggests its involvement in a post-transcriptional mechanism of gene expression control. These findings demonstrate that SAP1 is essential for liver infection possibly by functioning as a selective regulator controlling the expression of infectivity-associated parasite effector genes.
DOI: 10.1091/mbc.e06-01-0064
2006
Cited 99 times
A Cleavable Propeptide Influences<i>Toxoplasma</i>Infection by Facilitating the Trafficking and Secretion of the TgMIC2–M2AP Invasion Complex
Propeptides regulate protein function and trafficking in many eukaryotic systems and have emerged as important features of regulated secretory proteins in parasites of the phylum Apicomplexa. Regulated protein secretion from micronemes and host cell invasion are inextricably linked and essential processes for the apicomplexan parasite Toxoplasma gondii. TgM2AP is a propeptide-containing microneme protein found in a heterohexameric complex with the microneme protein TgMIC2, a protein that has a demonstrated fundamental role in gliding motility and invasion. TgM2AP function is also central to these processes, because disruption of TgM2AP (m2apKO) results in secretory retention of TgMIC2, leading to reduced TgMIC2 secretion from the micronemes and impaired invasion. Because the TgM2AP propeptide is predicted to be processed in an intracellular site near where TgMIC2 is retained in m2apKO parasites, we hypothesized that the propeptide and its proteolytic removal influence trafficking and secretion of the complex. We found that proTgM2AP traffics through endosomal compartments and that deletion of the propeptide leads to defective trafficking of the complex within or near this site, resulting in aberrant processing and decreased secretion of TgMIC2, impaired invasion, and reduced virulence in vivo, mirroring the phenotypes observed in m2apKO parasites. In contrast, mutation of several cleavage site residues resulted in normal localization, but it affected the stability and secretion of the complex from the micronemes. Therefore, the propeptide and its cleavage site influence distinct aspects of TgMIC2-M2AP function, with both impacting the outcome of infection.
DOI: 10.1111/j.1462-5822.2005.00628.x
2006
Cited 98 times
The host cell transcription factor hypoxia-inducible factor 1 is required for Toxoplasma gondii growth and survival at physiological oxygen levels
Toxoplasma gondii is an obligate intracellular protozoan pathogen. We previously found that genes mediating cellular responses to hypoxia were upregulated in Toxoplasma -infected cells but not in cells infected with another intracellular pathogen, Trypanosoma cruzi. The inducible expression of these genes is controlled by the hypoxia-inducible factor 1 (HIF1) transcription factor, which is the master regulator of cells exposed to low oxygen. Because this response may be important for parasites to grow at physiological oxygen levels, we tested the hypothesis that HIF1 is important for Toxoplasma growth. Here, we demonstrate that Toxoplasma infection rapidly increased the abundance of the HIF1α subunit and activated HIF1 reporter gene expression. In addition, we found that Toxoplasma growth and survival was severely reduced in HIF1α knockout cells at 3% oxygen. While HIF1α was not required for parasite invasion, we determined that HIF1 was required for parasite cell division and organelle maintenance at 3% oxygen. These data indicate that Toxoplasma activates HIF1 and requires HIF1 for growth and survival at physiologically relevant oxygen levels.
DOI: 10.1128/ec.00081-08
2008
Cited 91 times
A Thioredoxin Family Protein of the Apicoplast Periphery Identifies Abundant Candidate Transport Vesicles in <i>Toxoplasma gondii</i>
Toxoplasma gondii, which causes toxoplasmic encephalitis and birth defects, contains an essential chloroplast-related organelle to which proteins are trafficked via the secretory system. This organelle, the apicoplast, is bounded by multiple membranes. In this report we identify a novel apicoplast-associated thioredoxin family protein, ATrx1, which is predominantly soluble or peripherally associated with membranes, and which localizes primarily to the outer compartments of the organelle. As such, it represents the first protein to be identified as residing in the apicoplast intermembrane spaces. ATrx1 lacks the apicoplast targeting sequences typical of luminal proteins. However, sequences near the N terminus are required for proper targeting of ATrx1, which is proteolytically processed from a larger precursor to multiple smaller forms. This protein reveals a population of vesicles, hitherto unrecognized as being highly abundant in the cell, which may serve to transport proteins to the apicoplast.
DOI: 10.4161/auto.25832
2013
Cited 82 times
<i><i>Plasmodium falciparum</i></i>ATG8 implicated in both autophagy and apicoplast formation
Amino acid utilization is important for the growth of the erythrocytic stages of the human malaria parasite Plasmodium falciparum, however the molecular mechanism that permits survival of the parasite during conditions of limiting amino acid supply is poorly understood. We provide data here suggesting that an autophagy pathway functions in P. falciparum despite the absence of a typical lysosome for digestion of the autophagosomes. It involves PfATG8, which has a C-terminal glycine which is absolutely required for association of the protein with autophagosomes. Amino acid starvation provoked increased colocalization between PfATG8- and PfRAB7-labeled vesicles and acidification of the colabeled structures consistent with PfRAB7-mediated maturation of PfATG8-positive autophagosomes; this is a rapid process facilitating parasite survival. Immuno-electron microscopic analyses detected PfRAB7 and PfATG8 on double-membrane-bound vesicles and also near or within the parasite's food vacuole, consistent with autophagosomes fusing with the endosomal system before being routed to the food vacuole for digestion. In nonstarved parasites, PfATG8, but not PfRAB7, was found on the intact apicoplast membrane and on apicoplast-targeted vesicles and apicoplast remnants when the formation of the organelle was disrupted; a localization also requiring the C-terminal glycine. These findings suggest that in addition to a classical role in autophagy, which involves the PfRAB7-endosomal system and food vacuole, PfATG8 is associated with apicoplast-targeted vesicles and the mature apicoplast, and as such contributes to apicoplast formation and maintenance. Thus, PfATG8 may be unique in having such a second role in addition to the formation of autophagosomes required for classical autophagy.
DOI: 10.1371/journal.ppat.1003162
2013
Cited 80 times
A Toxoplasma Palmitoyl Acyl Transferase and the Palmitoylated Armadillo Repeat Protein TgARO Govern Apical Rhoptry Tethering and Reveal a Critical Role for the Rhoptries in Host Cell Invasion but Not Egress
Apicomplexans are obligate intracellular parasites that actively penetrate their host cells to create an intracellular niche for replication. Commitment to invasion is thought to be mediated by the rhoptries, specialized apical secretory organelles that inject a protein complex into the host cell to form a tight-junction for parasite entry. Little is known about the molecular factors that govern rhoptry biogenesis, their subcellular organization at the apical end of the parasite and subsequent release of this organelle during invasion. We have identified a Toxoplasma palmitoyl acyltransferase, TgDHHC7, which localizes to the rhoptries. Strikingly, conditional knockdown of TgDHHC7 results in dispersed rhoptries that fail to organize at the apical end of the parasite and are instead scattered throughout the cell. While the morphology and content of these rhoptries appears normal, failure to tether at the apex results in a complete block in host cell invasion. In contrast, attachment and egress are unaffected in the knockdown, demonstrating that the rhoptries are not required for these processes. We show that rhoptry targeting of TgDHHC7 requires a short, highly conserved C-terminal region while a large, divergent N-terminal domain is dispensable for both targeting and function. Additionally, a point mutant lacking a key residue predicted to be critical for enzyme activity fails to rescue apical rhoptry tethering, strongly suggesting that tethering of the organelle is dependent upon TgDHHC7 palmitoylation activity. We tie the importance of this activity to the palmitoylated Armadillo Repeats-Only (TgARO) rhoptry protein by showing that conditional knockdown of TgARO recapitulates the dispersed rhoptry phenotype of TgDHHC7 knockdown. The unexpected finding that apicomplexans have exploited protein palmitoylation for apical organelle tethering yields new insight into the biogenesis and function of rhoptries and may provide new avenues for therapeutic intervention against Toxoplasma and related apicomplexan parasites.
DOI: 10.1038/nrmicro3139
2013
Cited 78 times
Targeting lipid biosynthesis and salvage in apicomplexan parasites for improved chemotherapies
DOI: 10.1083/jcb.201701108
2017
Cited 70 times
The parasite Toxoplasma sequesters diverse Rab host vesicles within an intravacuolar network
Many intracellular pathogens subvert host membrane trafficking pathways to promote their replication. Toxoplasma multiplies in a membrane-bound parasitophorous vacuole (PV) that interacts with mammalian host organelles and intercepts Golgi Rab vesicles to acquire sphingolipids. The mechanisms of host vesicle internalization and processing within the PV remain undefined. We demonstrate that Toxoplasma sequesters a broad range of Rab vesicles into the PV. Correlative light and electron microscopy analysis of infected cells illustrates that intravacuolar Rab1A vesicles are surrounded by the PV membrane, suggesting a phagocytic-like process for vesicle engulfment. Rab11A vesicles concentrate to an intravacuolar network (IVN), but this is reduced in Δgra2 and Δgra2Δgra6 parasites, suggesting that tubules stabilized by the TgGRA2 and TgGRA6 proteins secreted by the parasite within the PV contribute to host vesicle sequestration. Overexpression of a phospholipase TgLCAT, which is localized to the IVN, results in a decrease in the number of intravacuolar GFP-Rab11A vesicles, suggesting that TgLCAT controls lipolytic degradation of Rab vesicles for cargo release.
DOI: 10.1016/j.ijpara.2013.09.003
2014
Cited 62 times
Exploitation of auxotrophies and metabolic defects in Toxoplasma as therapeutic approaches
Like any obligate intracellular pathogen, the parasite Toxoplasma gondii has lost its capacity for living independently of another organism. Toxoplasma lacks many genes that encode for entire metabolic pathways and has, in return, expanded genes that promote nutrient scavenging to meet its basic metabolic requirements. Although sequestrated in a parasitophorous vacuole and thus insulated from the nutrient-rich host cytosol and organelles by a membrane, T. gondii has evolved efficient strategies to acquire essential metabolites from mammalian cells. This review explores the natural auxotrophies and nutrient scavenging activities of the parasite, emphasising unique transport systems and salvage pathways. We describe the mechanisms deployed by Toxoplasma to modify its parasitophorous vacuole to gain access to host cytosolic molecules and to hijack host organelles to retrieve their nutrient content. From a therapeutic perspective, we survey the different possibilities to starve T. gondii by nutrient depletion or disruption of salvage pathways.
DOI: 10.1016/j.celrep.2019.06.007
2019
Cited 43 times
The Cardiac Microenvironment Instructs Divergent Monocyte Fates and Functions in Myocarditis
<h2>Summary</h2> Two types of monocytes, Ly6C<sup>hi</sup> and Ly6C<sup>lo</sup>, infiltrate the heart in murine experimental autoimmune myocarditis (EAM). We discovered a role for cardiac fibroblasts in facilitating monocyte-to-macrophage differentiation of both Ly6C<sup>hi</sup> and Ly6C<sup>lo</sup> cells, allowing these macrophages to perform divergent functions in myocarditis progression. During the acute phase of EAM, IL-17A is highly abundant. It signals through cardiac fibroblasts to attenuate efferocytosis of Ly6C<sup>hi</sup> monocyte-derived macrophages (MDMs) and simultaneously prevents Ly6C<sup>lo</sup> monocyte-to-macrophage differentiation. We demonstrated an inverse clinical correlation between heart IL-17A levels and efferocytic receptor expressions in humans with heart failure (HF). In the absence of IL-17A signaling, Ly6C<sup>hi</sup> MDMs act as robust phagocytes and are less pro-inflammatory, whereas Ly6C<sup>lo</sup> monocytes resume their differentiation into MHCII<sup>+</sup> macrophages. We propose that MHCII<sup>+</sup>Ly6C<sup>lo</sup> MDMs are associated with the reduction of cardiac fibrosis and prevention of the myocarditis sequalae.
DOI: 10.3389/fcimb.2021.670049
2021
Cited 33 times
The Modular Circuitry of Apicomplexan Cell Division Plasticity
The close-knit group of apicomplexan parasites displays a wide variety of cell division modes, which differ between parasites as well as between different life stages within a single parasite species. The beginning and endpoint of the asexual replication cycles is a ‘zoite’ harboring the defining apical organelles required for host cell invasion. However, the number of zoites produced per division round varies dramatically and can unfold in several different ways. This plasticity of the cell division cycle originates from a combination of hard-wired developmental programs modulated by environmental triggers. Although the environmental triggers and sensors differ between species and developmental stages, widely conserved secondary messengers mediate the signal transduction pathways. These environmental and genetic input integrate in division-mode specific chromosome organization and chromatin modifications that set the stage for each division mode. Cell cycle progression is conveyed by a smorgasbord of positively and negatively acting transcription factors, often acting in concert with epigenetic reader complexes, that can vary dramatically between species as well as division modes. A unique set of cell cycle regulators with spatially distinct localization patterns insert discrete check points which permit individual control and can uncouple general cell cycle progression from nuclear amplification. Clusters of expressed genes are grouped into four functional modules seen in all division modes: 1. mother cytoskeleton disassembly; 2. DNA replication and segregation (D&amp;amp;S); 3. karyokinesis; 4. zoite assembly. A plug-and-play strategy results in the variety of extant division modes. The timing of mother cytoskeleton disassembly is hard-wired at the species level for asexual division modes: it is either the first step, or it is the last step. In the former scenario zoite assembly occurs at the plasma membrane (external budding), and in the latter scenario zoites are assembled in the cytoplasm (internal budding). The number of times each other module is repeated can vary regardless of this first decision, and defines the modes of cell division: schizogony, binary fission, endodyogeny, endopolygeny.
DOI: 10.1016/0166-6851(92)90111-v
1992
Cited 96 times
Characterization of carbohydrate metabolism and demonstration of glycosomes in a Phytomonas sp. isolated from Euphorbia characias
Phytomonas sp. isolated from Euphorbia characias was adapted to SDM-79 medium. Cells isolated in the early stationary phase of growth were analyzed for their capacity to utilize plant carbohydrates for their energy requirements. The cellulose-degrading enzymes amylase, amylomaltase, invertase, carboxymethylcellulase, and the pectin-degrading enzymes polygalacturonase and oligo-D-galactosiduronate lyase were present in Phytomonas sp. and were all, except for amylomaltase, excreted into the external medium. Glucose, fructose and mannose served as the major energy substrates. Catabolism of carbohydrates occurred mainly via aerobic glycolysis according to the Embden-Meyerhof pathway, of which all the enzymes were detected. Likewise, the end-products of glycolysis, acetate and pyruvate, glycerol, succinate and ethanol were detected in the culture medium, as were the enzymes responsible for their production. Mitochondria were incapable of oxidizing succinate, 2-oxoglutarate, pyruvate, malate and proline, but had a high capacity to oxidize glycerol 3-phosphate. This oxidation was completely inhibited by salicylhydroxamic acid. No cytochromes could be detected either in intact mitochondria or in sub-mitochondrial particles. Mitochondrial respiration was not inhibited by antimycin, azide or cyanide. The glycolytic enzymes, from hexokinase to phosphoglycerate kinase, and the enzymes glycerol kinase, glycerol-3-phosphate dehydrogenase, phosphoenolpyruvate carboxykinase, malate dehydrogenase and adenylate kinase, were all associated with glycosomes that had a buoyant density of about 1.24 g cm-1 in sucrose. Cytochemical staining revealed the presence of catalase in these organelles. The cytosolic enzyme pyruvate kinase was activated by fructose 2,6-bisphosphate, typical of all other pyruvate kinases from Kinetoplastida. The energy metabolism of the plant parasite Phytomonas sp. isolated from E. characias resembled that of the bloodstream form of the mammalian parasite Trypanosoma brucei.
DOI: 10.1074/jbc.m411465200
2005
Cited 94 times
A Surface Phospholipase Is Involved in the Migration of Plasmodium Sporozoites through Cells
<i>Plasmodium</i> sporozoites, injected by mosquitoes into the skin of the host, traverse cells during their migration to hepatocytes where they continue their life cycle. The mechanisms used by the parasite to rupture the plasma membrane of the host cells are not known. Here we report the presence of a phospholipase on the surface of <i>Plasmodium berghei</i> sporozoites (<i>P. berghei</i> phospholipase; Pb PL) and demonstrate that it is involved in the establishment of a malaria infection <i>in vivo</i>. Pb PL is highly conserved among the <i>Plasmodium</i> species. The protein is about 750 amino acids, with a predicted signal sequence and a carboxyl terminus that is 32% identical to the vertebrate lecithin:cholesterol acyltransferase, a secreted phospholipase. Pb PL contains a motif characteristic of lipases and a catalytic triad of a serine, aspartate, and histidine that is found in several phospholipases. We have verified its lipase and membrane lytic activity <i>in vitro</i>, using recombinant baculovirus-expressed protein. To study its role <i>in vivo</i>, we have disrupted the <i>P. berghei</i> PL open reading frame and generated mutants in its active site. During an infection through mosquito bite, the infectivity of the knock-out parasites in the liver is decreased by ∼90%. The prepatent period of the resulting blood infection is 1 day longer as compared with wild type. Further, the mutant sporozoites are impaired in their ability to cross epithelial cell layers. Thus, the Pb PL functions as a lipase to damage cell membranes and facilitates sporozoite passage through cells during their migration from the skin to the bloodstream.
DOI: 10.1074/jbc.m501523200
2005
Cited 91 times
Selective Disruption of Phosphatidylcholine Metabolism of the Intracellular Parasite Toxoplasma gondii Arrests Its Growth
<i>Toxoplasma gondii</i> is an intracellular protozoan parasite capable of causing devastating infections in immunocompromised and immunologically immature individuals. In this report, we demonstrate the relative independence of <i>T. gondii</i> from its host cell for aminoglycerophospholipid synthesis. The parasite can acquire the lipid precursors serine, ethanolamine, and choline from its environment and use them for the synthesis of its major lipids, phosphatidylserine (PtdSer), phosphatidylethanolamine (PtdEtn), and phosphatidylcholine (PtdCho), respectively. Dimethylethanolamine (Etn(Me)<sub>2</sub>), a choline analog, dramatically interfered with the PtdCho metabolism of <i>T. gondii</i> and caused a marked inhibition of its growth within human foreskin fibroblasts. In tissue culture medium supplemented with 2 mm Etn(Me)<sub>2</sub>, the parasite-induced lysis of the host cells was dramatically attenuated, and the production of parasites was inhibited by more than 99%. The disruption of parasite growth was paralleled by structural abnormalities in its membranes. In contrast, no negative effect on host cell growth and morphology was observed. The data also reveal that the Etn(Me)<sub>2</sub>-supplemented parasite had a time-dependent decrease in its PtdCho content and an equivalent increase in phosphatidyldimethylethanolamine, whereas other major lipids, PtdSer, PtdEtn, and PtdIns, remained largely unchanged. Relative to host cells, the parasites incorporated more than 7 times as much Etn(Me)<sub>2</sub> into their phospholipid. These findings reveal that Etn(Me)<sub>2</sub> selectively alters parasite lipid metabolism and demonstrate how selective inhibition of PtdCho synthesis is a powerful approach to arresting parasite growth.
DOI: 10.1111/j.1462-5822.2005.00518.x
2005
Cited 82 times
Host cell lipids control cholesteryl ester synthesis and storage in intracellular Toxoplasma
Cellular MicrobiologyVolume 7, Issue 6 p. 849-867 Free Access Host cell lipids control cholesteryl ester synthesis and storage in intracellular Toxoplasma Yoshifumi Nishikawa, Yoshifumi Nishikawa Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA. Both authors contributed equally to this work.Search for more papers by this authorFriederike Quittnat, Friederike Quittnat Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA. Both authors contributed equally to this work.Search for more papers by this authorTimothy T. Stedman, Timothy T. Stedman Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.Search for more papers by this authorDennis R. Voelker, Dennis R. Voelker Department of Medicine, National Jewish Medical and Research Center, Denver, CO 80206, USA.Search for more papers by this authorJae-Yeon Choi, Jae-Yeon Choi Department of Medicine, National Jewish Medical and Research Center, Denver, CO 80206, USA.Search for more papers by this authorMatt Zahn, Matt Zahn Department of Medicine, National Jewish Medical and Research Center, Denver, CO 80206, USA.Search for more papers by this authorMei Yang, Mei Yang Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.Search for more papers by this authorMarc Pypaert, Marc Pypaert Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA.Search for more papers by this authorKeith A. Joiner, Keith A. Joiner Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.Search for more papers by this authorIsabelle Coppens, Corresponding Author Isabelle Coppens Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA. *E-mail [email protected]; Tel. (+1) 443 287 1589; Fax (+1) 410 955 0105.Search for more papers by this author Yoshifumi Nishikawa, Yoshifumi Nishikawa Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA. Both authors contributed equally to this work.Search for more papers by this authorFriederike Quittnat, Friederike Quittnat Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA. Both authors contributed equally to this work.Search for more papers by this authorTimothy T. Stedman, Timothy T. Stedman Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.Search for more papers by this authorDennis R. Voelker, Dennis R. Voelker Department of Medicine, National Jewish Medical and Research Center, Denver, CO 80206, USA.Search for more papers by this authorJae-Yeon Choi, Jae-Yeon Choi Department of Medicine, National Jewish Medical and Research Center, Denver, CO 80206, USA.Search for more papers by this authorMatt Zahn, Matt Zahn Department of Medicine, National Jewish Medical and Research Center, Denver, CO 80206, USA.Search for more papers by this authorMei Yang, Mei Yang Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.Search for more papers by this authorMarc Pypaert, Marc Pypaert Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA.Search for more papers by this authorKeith A. Joiner, Keith A. Joiner Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.Search for more papers by this authorIsabelle Coppens, Corresponding Author Isabelle Coppens Department of Internal Medicine, Yale University School of Medicine, New Haven, CT 06520, USA. *E-mail [email protected]; Tel. (+1) 443 287 1589; Fax (+1) 410 955 0105.Search for more papers by this author First published: 31 March 2005 https://doi.org/10.1111/j.1462-5822.2005.00518.xCitations: 72 AboutSectionsPDF ToolsRequest permissionExport citationAdd to favoritesTrack citation ShareShare Give accessShare full text accessShare full-text accessPlease review our Terms and Conditions of Use and check box below to share full-text version of article.I have read and accept the Wiley Online Library Terms and Conditions of UseShareable LinkUse the link below to share a full-text version of this article with your friends and colleagues. Learn more.Copy URL Share a linkShare onFacebookTwitterLinkedInRedditWechat Summary The intracellular protozoan Toxoplasma gondii lacks a de novo mechanism for cholesterol synthesis and therefore must scavenge this essential lipid from the host environment. In this study, we demonstrated that T. gondii diverts cholesterol from low-density lipoproteins for cholesteryl ester synthesis and storage in lipid bodies. We identified and characterized two isoforms of acyl-CoA:cholesterol acyltransferase (ACAT)-related enzymes, designated TgACAT1α and TgACAT1β in T. gondii. Both proteins are coexpressed in the parasite, localized to the endoplasmic reticulum and participate in cholesteryl ester synthesis. In contrast to mammalian ACAT, TgACAT1α and TgACAT1β preferentially incorporate palmitate into cholesteryl esters and present a broad sterol substrate affinity. Mammalian ACAT-deficient cells transfected with either TgACAT1α or TgACAT1β are restored in their capability of cholesterol esterification. TgACAT1α produces steryl esters and forms lipid bodies after transformation in a Saccharomyces cerevisiae mutant strain lacking neutral lipids. In addition to their role as ACAT substrates, host fatty acids and low-density lipoproteins directly serve as Toxoplasma ACAT activators by stimulating cholesteryl ester synthesis and lipid droplet biogenesis. Free fatty acids significantly increase TgACAT1α mRNA levels. Selected cholesterol esterification inhibitors impair parasite growth by rapid disruption of plasma membrane. Altogether, these studies indicate that host lipids govern neutral lipid synthesis in Toxoplasma and that interference with mechanisms of host lipid storage is detrimental to parasite survival in mammalian cells. Introduction Sterols are found in all eukaryotic organisms, and are membrane components that regulate the fluidity and the permeability of phospholipid bilayers. Sterol homeostasis is a complex event under subtle regulatory controls. Esterification of sterols by fatty acids, one of the critical homeostatic responses, is upregulated upon elevated cellular cholesterol or fatty acid levels (Chang et al., 1997). The esterification reaction is mediated by O-acyltransferases providing an important steryl ester storage depot, which overcomes any membrane perturbations that accrue from excessive sterol or free fatty acid levels (Warner et al., 1995; Tabas, 1997; Kellner-Weibel et al., 1998). In mammalian cells, two acyl-CoA:cholesterol acyltransferases (ACAT; EC 2.3.2.26), ACAT1 and ACAT2, are involved in cholesteryl ester (CE) biosynthesis (summarized in Buhman et al., 2000; Chang et al., 2001). Both enzymes are mainly located to the endoplasmic reticulum (ER). The expression of ACAT1 is ubiquitous whereas that of ACAT2 is tissue-restricted (small intestine and liver). Nevertheless, the enzymatic properties of human ACAT1 and ACAT2 are quite similar (Chang et al., 2000). Mammalian ACAT1 and ACAT2 descended from ancestral genes in yeast (Yang et al., 1996). Surprisingly, these mammalian enzymes do not have the same intron–exon structures despite they share nearly 40% identity at the amino acid level (Buhman et al., 2000). This implies that the two enzymes diverged quite early during evolution, possibly to perform different cellular functions. Based on ACAT-knockout mice observations (Meiner et al., 1996), CE synthesis mediated by ACAT1 may be preferentially coupled with ester storage in cytosolic lipid bodies while ACAT2 may be functionally linked to the lipoprotein-mediated secretion of CE (reviewed in Buhman et al., 2000). Conversely, in Saccharomyces cerevisiae, two ACAT-related enzymes are present within the same cell, and both yield steryl ester pools in yeast (Yang et al., 1996). In eukaryotic cells, steryl esters are stored in cytoplasmic lipid droplets, which are metabolically active inclusions (reviewed in Sandager et al., 2002). In mammalian cells, impairment in lipid body functions leads to several serious human diseases (summarized in Murphy, 2001). Less clear are the functions of lipid droplets as well as the importance of neutral lipid storage in primitive eukaryotes. We have recently characterized the activity of triacylglycerol biosynthetic enzymes homologues to mammalian acyl-CoA:diacylglycerol acyltransferase 1 (DGAT1) in two related apicomplexan parasites: Plasmodium falciparum, aetiologic agent of malaria (Vielemeyer et al., 2004) and Toxoplasma gondii (Quittnat et al., 2004). The latter is an obligate intracellular parasite capable of infecting nearly all types of nucleated cells and is responsible for fatal diseases in neonates and immunocompromised individuals (Luft and Remington, 1992). An ACAT activity has been formerly detected in the free living ciliate Tetrahymena pyriformis (Billheimer et al., 1989) and in T. gondii (Sonda et al., 2001; Charron and Sibley, 2002). Interestingly, two different sites of cholesterol accumulation have been identified in T. gondii: (i) the apical secretory organelle rhoptries implicated in the biogenesis of the parasitophorous vacuole (PV) membrane during parasite invasion and (ii) lipid inclusions similar to mammalian lipid bodies (Charron and Sibley, 2002; Coppens and Joiner, 2003). We have previously shown that T. gondii is auxotrophic from low-density lipoproteins (LDL)-derived cholesterol and that interfering with host cholesterol acquisition by T. gondii impairs parasite growth (Coppens et al., 2000). As this parasite has no capability to synthesize sterols when needed, we therefore hypothesize that impairing cholesterol storage will also be detrimental to parasite viability. To test this hypothesis, we characterized the sterol esterification reaction in both functional and molecular terms, emphasizing on the unique features in steryl ester synthesis developed by T. gondii. Namely, the parasite expresses two unusual ACAT isoforms. Compared with mammalian ACAT, the parasite homologues differ in their substrate affinity and specificity, as well as in their mechanisms of regulation. In addition, some inhibitors of mammalian ACAT are particularly cytotoxic to Toxoplasma, reflecting differences in the steryl ester cycle between mammalian cells and this protozoan. Results Toxoplasma is competent to synthesize cholesteryl esters using host cell-derived cholesterol We assayed ACAT activity and quantified the synthesis of CE in Toxoplasma. Intracellular parasites were incubated with labelled substrates, including [3H]-cholesterol incorporated into LDL, [3H]-oleate associated with albumin, or [14H]-phosphaditylcholine (PC) labelled on the fatty acid moiety for different times. After isolation of parasite lipids by chromatography, we demonstrated that T. gondii was able to incorporate exogenous cholesterol and fatty acid into the CE fraction (Table 1), which confirms previous studies using radioactive oleate (Sonda et al., 2001). The production of CE was proportional to the amount of lipid taken up by the parasite over the time. Incubation in the presence of the lysosomotropic agent chloroquine that impairs the release of cholesterol within LDL (Brown et al., 1975), abolished cholesterol availability for the parasite (Coppens et al., 2000), and therefore CE synthesis. Table 1. Lipid uptake and esterification activity in T. gondii. Pulse conditions at 37°C Lipid uptake (cpm per µg cell protein) CE formed (cpm per µg cell protein) 1 h, [3H]-C-LDL 523 ± 22 3140 ± 110 (n = 4) 1 h + chloroquine, [3H]-C-LDL 56 ± 12 ND (n = 3) 3 h, [3H]-C-LDL 850 ± 44 4910 ± 165 (n = 4) 24 h, [3H]-C-LDL 1310 ± 62 10 890 ± 180 (n = 5) 30 min, [3H]-oleate-BSA 390 ± 32 2099 ± 123 (n = 3) 3 h, [14C]-PC-BSA 877 ± 54 3388 ± 230 (n = 3) 3 h, [14C]-PC-LDL 243 ± 30 989 ± 55 (n = 3) Intravacuolar parasites have been pulse-labelled with 1 mg ml−1 tritiated cholesterol incorporated into LDL (C-LDL), tritiated oleate at 0.2 mM bound to albumin or 1 mM [14C]-PC for the indicated times. For chloroquine treatment, infected monolayers were incubated for 10 min with 200 µM chloroquine before the pulse and then maintained in the presence of 100 µM chloroquine during the 1 h pulse with radioactive LDL. Results of lipid uptake and CE production expressed in cpm per µg cell protein are means ± SD of three to five independent experiments. ND, not detected. The fatty acid substrate specificity for Toxoplasma was then examined. Intravacuolar parasites were incubated with radioactive oleate, palmitate, arachidonate, stearate or linoleate for 2 h. All the tested free fatty acids (FFA) were incorporated into parasite CE, suggestive of broad fatty acid specificity (not shown). However, in contrast to mammalian cells, T. gondii showed a nearly twofold higher incorporation of palmitate into CE as compared with other FFA. This palmitate specificity has been previously observed for triacylglycerol synthesis in T. gondii and was ascribed to a preferential uptake of palmitate over other FFA by the parasite (Quittnat et al., 2004). To circumvent the consequences of FFA access or metabolism when added to infected cells, T. gondii was incubated axenically in the presence of the same radioactive FFA for 2 h. A higher incorporation of palmitate into parasite CE was also observed in extracellular parasites (not shown). Identification of two isoforms of a T. gondii ACAT-related enzyme Through homology searches of the T. gondii EST databases using coding sequences from O-acyltransferases, we identified an EST clone that shared homology with ACAT C-terminus sequences. The full cDNA corresponding to this EST was 1950 bp in length and contained an open reading frame (ORF) encoding a 650 aa protein (Fig. 1A). Northern blot analysis on total RNA revealed a mature transcript of ∼2.6 kb (Fig. 1B). Based on phylogenetic analysis, the full length of the predicted protein was more closely related to various ACAT1 (∼18% identical) than to ACAT2 (∼14% identical), although the differences were minor (not shown). However, based on the ACAT functional motifs described below, the parasite enzyme was closely related to yeast homologues (Fig. 1C), which preferentially use ergosterol as substrate (Yang et al., 1997). Figure 1Open in figure viewerPowerPoint Sequence alignment for TgACTA1α and TgACAT1β, transcriptional analysis and phylogeny. A. Alignments were performed using the clustalw program. The deduced protein sequences of TgACAT1α and TgACAT1β were aligned with the following sequences: HsACAT1, the Homo sapiens ACAT1 (Accession No. L21934; Chang et al., 1993), HsACAT2, the Homo sapiens ACAT2 (Accession No. AF059202; Delker et al., 1998), MmACAT1, the Mus musculusACAT1 (Accession No. I49454; Uelmen et al., 1998), MmACAT2, the M. musculusACAT2 (Accession No. AF078751; Cases et al., 1998), ScARE1, the S. cerevisiaeACAT1-related enzyme (Accession No. P25628; Yang et al., 1996) and ScARE2, the S. cerevisiaeACAT2-related enzyme (Accession No. U51790; Yang et al., 1996). Identical amino acids are shaded in black; similar and conserved amino acids are shaded in light and dark grey respectively. Signs are placed under the important amino acids or highly conserved motives (see their description in the text). The potential sites of N-glycosylation are located on Asn162, Asn407 and Asn520 for TgACAT1α and on Asn160 and Asn273 for TgACAT1β. The predicted membrane helix spanning domains are situated between amino acids 264–286, 305–329, 344–376, 446–466, 476–496, 555–578, 572–599, 608–626 for TgACAT1α and between amino acids 17–38, 58–82, 97–129, 199–219, 229–249, 308–331, 325–352, 361–379 for TgACAT1β. B. Analysis of the TgACAT1α and TgACAT1β transcripts and translated products. Northern blotting of total RNA probed with radiolabelled TgACAT1α cDNA. Twenty micrograms of total RNA were resolved on a denaturing gel, blotted onto a nylon membrane and hybridized with a [32P]-dCTP-TgACAT1α probe (lane 1: full length: 1–1950 bp; lane 2: 1–336 bp). The size of RNA markers are indicated at the right margin. The arrows denote the TgACAT1α 2.6 kb and TgACAT1β 1.3 kb transcripts. C. Phylogeny of TgACAT1. Neighbour-joining phylogenetic tree constructed from the conserved domains in the protein sequences of TgACAT1 (starting at Lys373 for TgACAT1α) and the indicated homologues. In the course of reverse transcription polymerase chain reaction (RT-PCR) cloning of T. gondii ACAT1-related enzyme, we observed that half of the clones had a smaller size than expected. Sequence analysis revealed that the smaller transcript of ∼1.3 kb (Fig. 1B) was identical to the parasite ACAT1-related enzyme sequence, but lacked 738 nt encoding a predicted hydrophilic serine-rich region at the N-terminus. Of interest, the transcript also differed in the first 45 nucleotides of the truncated ORF. The full cDNA corresponding to this ACAT1-related enzyme was 1209 bp in length and contained an ORF encoding a 403 aa protein (Fig. 1B). We termed these isoforms TgACAT1α (650 aa) and TgACAT1β (403 aa). Both isoforms shared common features of the superfamily of membrane-bound O-acyltransferases (Cases et al., 1998a,b; Hofmann, 2000) and had an invariant histidine positioned within a long hydrophobic region (His570 and His323 for TgACAT1α and TgACAT1β respectively; Fig. 1A). Two signatures shared between all ACATs and DGAT1 (Bouvier et al., 2000), have been identified in TgACAT1α and TgACAT1β: (i) a putative invariant serine (Ser376 and Ser136 for TgACAT1α and TgACAT1β respectively) and (ii) the remarkably conserved motif [AExxRFGDRxFYxDWWN] potentially ascribed to include the fatty acid binding site, which corresponds to [AEITNFANRNFYDDWWN] in the TgACAT1α and TgACAT1β sequences, starting at Ala604 and Ala357 respectively. Finally, the TgACAT1 sequences had the putative cholesterol binding site [H/YSF] including a serine surrounded by an aromatic and/or basic amino acids (Guo et al., 2001) present in all the identified ACATs so far (His375SY and His135SY in TgACAT1α and TgACAT2β respectively). Analysis by a transmembrane region prediction program (http://www.ch.embmet.org/) favours eight membrane helix spanning domains for both isoforms. Hence, the two parasite TgACAT1 isoforms have the major hallmarks of the ACAT family previously found in various species. Coexpression and endoplasmic reticulum localization TgACAT1α and TgACAT1β Parasite lysates probed on immunoblots with anti-TgACAT1 antibodies against the C-terminus of TgACAT1α revealed two bands, one at Mr 73 kDa and the other at 42.5 kDa (Fig. 2A, lane 1). On immunoblots using the same parasite lysates incubated with antibodies against the specific N-terminus of TgACAT1α, only the 73 kDa band was observed (Fig. 2A, lane 3), leading to the conclusion that the upper band probably corresponds to TgACAT1α and the lower one to TgACAT1β. This indicates that both parasite isoforms were fully expressed in T. gondii. Nevertheless, quantitative immunoblot analysis revealed that parasites expressed 3.5-fold lower levels of TgACAT1β protein compared with TgACAT1α. Metabolic labelling of TgACAT1α-HA and TgACAT1β-HA expressing cells with radioactive methionine followed by immunoprecipitation of the two enzymes with anti-HA antibodies showed that the half-lives of TgACAT1α and TgACAT1β enzymes were 8 h and 30 min respectively (data not shown). Figure 2Open in figure viewerPowerPoint Coexpression and localization of TgACAT1α and TgACAT1β in T. gondii. A. Immunoblots of parasite lysates. After lysis of the parasites in a solution of deoxycholate/PC, resolution of the proteins on gel and transfer to a nitrocellulose membrane, the Western blot analysis of the gel was probed with anti-TgACAT1α474−650 antibodies (lane 1) and preimmune (lane 2), or anti-TgACAT1α14−117 antibodies (lane 3) and preimmune (lane 4). The molecular weight markers are indicated at the left margin. B. Fluorescence microscopy of stably expressed Toxoplasma with TgACAT1α-HA and TgACAT1β-HA. Intravacuolar parasites were stained with anti-HA antibodies, followed by FITC-conjugated goat anti-mouse IgG, revealing an ER staining. The parasite nuclei were stained with DAPI. C. Immunogold labelling of TgACAT1α-HA and TgACAT1β-HA. Cryosections of stably expressed Toxoplasma with TgACAT1α-HA and TgACAT1β-HA were incubated with anti-HA antibodies, followed by incubation with protein A adsorbed to 10 nm gold particles. The gold particles were concentrated in the parasite ER area. Control cryosections on wild-type parasites with equivalent anti-HA antibody dilution were completely free of immunogold label (not shown). G, Golgi; n, nucleus. Scale bars are 0.150 µm. To visualize the distribution of these enzymes in T. gondii, parasite lines expressing either TgACAT1α or TgACAT1β fused with a C-terminal nine-residue HA epitope tag were engineered. In either transiently or stably transfected Toxoplasma, TgACAT1α-HA and TgACAT1β-HA were predominantly localized to a network of membranes dispersed throughout the parasite cytoplasm and around the nucleus (Fig. 2B), which was identified as the ER by immunoelectron microscopy (Fig. 2C). Both the cortical and perinuclear ER were intensely labelled. No labelling was observed on other parasite structures such as at the surface of lipid bodies or in rhoptries. In addition, anti-TgACAT1α or -β antibodies colocalized with antibodies against TgDAGT1 to the parasite ER (Quittnat et al., 2004). To confirm that the serine-rich region at the N-terminus of TgACAT1α was fully translated, a c-myc tag was inserted in the middle of the N-terminal sequence (aa 110). In parasites expressing this construct, a clear co-labelling of the ER with antibodies against c-myc and HA was observed (not shown). Both TgACAT1α and TgACAT1β can restore cholesteryl ester production in mammalian ACAT-deficient cells and present broad sterol substrate specificity The presence of two ACAT isoforms is likely to have functional implications for CE production in T. gondii. To investigate the relative function of TgACAT1α and TgACAT1β, mammalian ACAT-deficient cell lines were transiently transfected with expression plasmids encoding each isoform of TgACAT1-HA. Both constructs localized to the ER in mammalian cells and exhibited extensive colocalization with calnexin, an endogenous ER marker (TgACAT1α, Fig. 3A; TgACAT1β, not shown). We first monitored oleate uptake and CE formation in MEF–/–, an ACAT-deficient cell line (Meiner et al., 1996), after transfection with pTgACAT1α or pTgACAT1β (Fig. 3B). The uptake of oleate by the mutant cells expressing one isoform of TgACAT1 was similar over a period of 2 h. As a result of TgACAT1α or TgACAT1β expression in both cell lines, a significant ACAT activity was reconstituted, leading to the production of CE. The addition of exogenous cholesterol to the cells greatly increased esterification by approximately twofold in TgACAT1α- and TgACAT1β-expressing cell lines. This indicates that the two Toxoplasma isoforms are functional ACAT enzymes, although a threefold lower incorporation of oleate into CE was observed in TgACAT1β-expressing cells as compared with TgACAT1α-expressing cells. This observation might be ascribed to a lower percentage of MEF–/– expressing the β compared with the α isoform. A lower CE activity for TgACAT1β compared with TgACAT1α was also detected in mammalian AC29 cells, another cell line lacking ACAT activity (Cadigan et al., 1988; data not shown). Additionally, parasites transiently transfected with pTgACAT1α or pTgACAT1β showed an increase in CE production by 230 ± 73% and 146 ± 45%, respectively, compared with the wild-type strain upon incubation with tritiated oleate (not shown). Figure 3Open in figure viewerPowerPoint TgACAT1α and TgACAT1β distribution and function in mammalian ACAT-deficient cells and yeast lacking neutral lipid synthesis. A. Fluorescence microscopy of TgACAT1α transiently expressed in ACAT–/– MEF. Twenty-four hours after transfection, mammalian cells were first stained with mouse anti-HA antibodies, followed by rhodamin-conjugated goat anti-mouse IgG, then incubated with rabbit anti-calnexin antibodies, revealed by FITC-conjugated goat anti-mouse IgG. The merge picture shows colocalization of TgACAT1α and calnexin along the ER. B. Oleate uptake and incorporation into CE by ACAT-deficient cells expressing TgACAT1α or TgACAT1β. Twenty-four hours after transfection with pCI-neo, pTgACAT1α or TgACAT1β, the MEF–/– have been incubated for 2 h with 0.2 mM tritiated oleate. Results of oleate uptake or CE production expressed in nmol FFA per mg cell protein or nmol FFA incorporated into CE per mg cell protein, respectively, are means ± SD of three separate experiments. Differences between values of oleate incorporated into CE in MEF–/– between vector controls and TgACAT1α or TgACAT1β are statistically significant in the presence of endogenous or exogenous cholesterol (100 mg per ml LDl) (*P < 0.005; **P < 0.0005). By comparison, values of oleate incorporation into CE in the parental MEF+/+ were 934 ± 210 pmol FFA per mg cell protein. The transfection percentage (monitored by counting fluorescent cells revealed by immunofluorescence assay using anti-HA antibodies) for MEF–/– cells is 5.12 ± 2.7% for TgACAT1α and 1.19 ± 0.9% for TgACAT1β. C. In vitro sterol substrate specificity. The indicated fatty acyl acceptors were incubated in the presence of parasites or CHO cell homogenates solubilized in a deoxycholate/PC solution for 10 min at 37°C after the addition of radioactive oleate to monitor the production of steryl oleate. Results are expressed in percentage control values in the presence of cholesterol, which were 630 and 955 nmol of cholesteryl oleate formed per mg protein per minute for Toxoplasma and CHO cells. 7-dehydrochol, 7-dehydrocholesterol; 25-OH-chol, 25-hydroxycholesterol. D. TLC of mammalian and T. gondii sterols. Lanes 1 and 2 represent sterols isolated from 1 mg of fibroblasts and intravacuolar parasites, respectively, after cultivation in medium containing 10% complete serum plus 200 µg ml−1 human LDL. Lane 3 shows sterols purified from 1 mg of intravacuolar parasites maintained for 36 h in lipoprotein-deficient serum (LPDS). Accordingly, lanes 1, 2 and 3 show exclusively cholesterol. STD, standard. E. Reconstitution of lipid body formation and steryl ester formation in yeast mutants expressing TgACAT1α. Wild-type (WT), mutant yeast (Mut.) transformed or not with either TgACAT1α (Mut. +TgACAT1α) or TgACAT1β (Mut. +TgACAT1β) were stained with Nile Red and examined by fluorescence microscopy for lipid body formation (left). The arrows pinpoint the filamentous form of TgACAT1α-transformed mutant yeast. The heterogeneity of the yeast phenotype after transformation with the isoform resulted from a non-synchronization of yeast population. Quantification of CE synthesis in TgACAT1α- and β-expressing mutant yeast in the presence of 100 µg ml−1 cholesterol (right). The wild-type and the mutant strain transformed with either TgACAT1α or TgACAT1β were grown under inducing conditions, harvested and processed for measuring enzyme activity. Cell extracts were assayed for the CE synthesis using radioactive palmitoyl-CoA and cholesterol before lipid analysis by TLC. Results expressed in pmoles CE per mg cell protein per minute are representative of three separate experiments. Having demonstrated the activity of the Toxoplasma ACAT1 cDNA in ACAT-mammalian cells, we extended these observations to assess the affinity of the parasite ACAT enzymes for various sterols in comparison to mammalian ACAT (Fig. 3C). An in vitro system was utilized to measure ACAT activity in the presence of radioactive oleate and different potential fatty acyl acceptors for the esterification reaction. Both Toxoplasma and mammalian enzymes esterified the cholesterol precursor 7-dehydrocholesterol, but not lanosterol. The β-sitosterol, a specific substrate for the plasma lecithin:cholesterol acyltransferase (LCAT), was esterifiable by the parasite ACAT while mammalian enzymes had a limited cellular synthesis of sitosteryl esters. TgACAT1 also showed an esterification activity for 25-hydroxycholesterol, a hydroxylated derivative of cholesterol known to both inhibit cholesterol synthesis (Adams et al., 2005) and activate ACAT by post-translational mechanisms (Cheng et al., 1995; Chang et al., 1997), in the same extent as mammalian ACAT. In contrast to mammalian enzyme, the parasite ACAT were competent to readily esterify ergosterol. Nevertheless, we determined that Toxoplasma was unable to synthesize ergosterol in physiological conditions (Fig. 3D) or under growth condition in the absence of lipoproteins for 36 h. Only TgACAT1α expressed in a yeast mutant strain lacking neutral lipid synthesis can form lipid bodies and produce cholesteryl esters The capability of T. gondii ACAT to esterify ergosterol has been directly addressed in a yeast strain devoid of DGAT and ACAT activities (Sandager et al., 2002). This strain was transformed with the parasite ACAT enzymes under the control of a yeast GAL1 promoter and expressed under induced and non-induced conditions. Wild-type yeast accumulated lipid droplets in stationary phase as visualized by Nile Red staining and fluorescence microscopy (Fig. 3E). In contrast, the JCY500 strain with null alleles for LRO1, DGA1, ARE1 and ARE2 genes failed to accumulate lipid droplets. When th
DOI: 10.1111/j.1462-5822.2005.00647.x
2006
Cited 75 times
Contribution of host lipids to Toxoplasma pathogenesis
As an actively dividing organism, the intracellular parasite Toxoplasma gondii must adjust the size and composition of its membranes in order to accommodate changes due to housekeeping activities, to commit division and in fine to produce new viable progenies. Lipid inventory of T. gondii reveals that the biological membranes of this parasite are composed of a complex mixture of neutral and polar lipids. After examination of the origin of T. gondii membrane lipids, three categories of lipids can be described: (i) lipids scavenged by T. gondii from the host cell; (ii) lipids synthesized in large amounts by the parasite, independently from its host cell; and (iii) lipids produced de novo by the parasite, but whose synthesis does not come close to satisfying the entire parasite's needs. These latter must be adeptly acquired from the host environment. To this end, T. gondii diverts a large variety of lipid precursors from host cytoplasm and efficiently manufacture them into complex lipids. This rather remarkable reliance on host lipid resources for parasite survival opens new avenues to restrict parasite growth. Indeed, parasite starvation can be induced upon deprivation from essential host lipids. Lipid analogues with anti-proliferative properties are voraciously taken up by the parasites, which results in parasite membrane defects, and ultimately death.
DOI: 10.1111/j.1365-2958.2007.05619.x
2007
Cited 71 times
Cell cycle‐regulated vesicular trafficking of <i>Toxoplasma</i> APT1, a protein localized to multiple apicoplast membranes
The apicoplast is a relict plastid essential for viability of the apicomplexan parasites Toxoplasma and Plasmodium. It is surrounded by multiple membranes that proteins, substrates and metabolites must traverse. Little is known about apicoplast membrane proteins, much less their sorting mechanisms. We have identified two sets of apicomplexan proteins that are homologous to plastid membrane proteins that transport phosphosugars or their derivatives. Members of the first set bear N-terminal extensions similar to those that target proteins to the apicoplast lumen. While Toxoplasma gondii lacks this type of translocator, the N-terminal extension from the Plasmodium falciparum sequence was shown to be functional in T. gondii. The second set of translocators lacks an N-terminal targeting sequence. This translocator, TgAPT1, when tagged with HA, localized to multiple apicoplast membranes in T. gondii. Contrasting with the constitutive targeting of luminal proteins, the localization of the translocator varied during the cell cycle. Early-stage parasites showed circumplastid distribution, but as the plastid elongated in preparation for division, vesicles bearing TgAPT1 appeared adjacent to the plastid. After plastid division, the protein resumes a circumplastid colocalization. These studies demonstrate for the first time that vesicular trafficking likely plays a role in the apicoplast biogenesis.
DOI: 10.1038/cr.2010.88
2010
Cited 71 times
Metamorphosis of the malaria parasite in the liver is associated with organelle clearance
Malaria parasites encounter diverse conditions as they cycle between their vertebrate host and mosquito vector. Within these distinct environments, the parasite undergoes drastic transformations, changing both its morphology and metabolism. Plasmodium species that infect mammals must first take up residence in the liver before initiating red blood cell infection. Following penetration into hepatocytes, the parasite converts from an invasion-competent, motile, elongated sporozoite to a metabolically active, round trophozoite. Relatively little is known about the cellular events involved in sporozoite metamorphosis. Our data uncover the early cellular events associated with these transformations. We illustrate that the beginning of metamorphosis is marked by the disruption of the membrane cytoskeleton beneath the plasma membrane, which results in a protruding area around the nucleus. As this bulbous region expands, the two distal ends of the sporozoite gradually retract and disappear, leading to cell sphericalization. This shape change is associated with major interior renovations and clearance of superfluous organelles, e.g. micronemes involved in invasion. The membrane cytoskeleton is reorganized into dense lamellar arrays within the cytoplasm and is partially expulsed by converting parasites. Simultaneously, micronemes are compartmentalized into large exocytic vesicles and are then discharged into the environment. At the completion of metamorphosis, the parasites only retain organelles necessary for replication. These observations lay the groundwork for further investigations on the developmental pathways implicated in the metamorphosis of the malaria parasite.
DOI: 10.1186/1475-2875-8-179
2009
Cited 66 times
Plasmodium falciparum enolase: stage-specific expression and sub-cellular localization
In an earlier study, it was observed that the vaccination with Plasmodium falciparum enolase can confer partial protection against malaria in mice. Evidence has also build up to indicate that enolases may perform several non-glycolytic functions in pathogens. Investigating the stage-specific expression and sub-cellular localization of a protein may provide insights into its moonlighting functions.Sub-cellular localization of P. falciparum enolase was examined using immunofluorescence assay, immuno-gold electron microscopy and western blotting.Enolase protein was detected at every stage in parasite life cycle examined. In asexual stages, enolase was predominantly (>or=85-90%) present in soluble fraction, while in sexual stages it was mostly associated with particulate fraction. Apart from cytosol, enolase was found to be associated with nucleus, food vacuole, cytoskeleton and plasma membrane.Diverse localization of enolase suggests that apart from catalyzing the conversion of 2-phosphoglycericacid into phosphoenolpyruvate in glycolysis, enolase may be involved in a host of other biological functions. For instance, enolase localized on the merozoite surface may be involved in red blood cell invasion; vacuolar enolase may be involved in food vacuole formation and/or development; nuclear enolase may play a role in transcription.
DOI: 10.1111/j.1365-2958.2010.07081.x
2010
Cited 59 times
A high‐affinity putrescine‐cadaverine transporter from <i>Trypanosoma cruzi</i>
Whereas mammalian cells and most other organisms can synthesize polyamines from basic amino acids, the protozoan parasite Trypanosoma cruzi is incapable of polyamine biosynthesis de novo and therefore obligatorily relies upon putrescine acquisition from the host to meet its nutritional requirements. The cell surface proteins that mediate polyamine transport into T. cruzi, as well as most eukaryotes, however, have by-in-large eluded discovery at the molecular level. Here we report the identification and functional characterization of two polyamine transporters, TcPOT1.1 and TcPOT1.2, encoded by alleles from two T. cruzi haplotypes. Overexpression of the TcPOT1.1 and TcPOT1.2 genes in T. cruzi epimastigotes revealed that TcPOT1.1 and TcPOT1.2 were high-affinity transporters that recognized both putrescine and cadaverine but not spermidine or spermine. Furthermore, the activities and subcellular locations of both TcPOT1.1 and TcPOT1.2 in intact parasites were profoundly influenced by extracellular putrescine availability. These results establish TcPOT1.1 and TcPOT1.2 as key components of the T. cruzi polyamine transport pathway, an indispensable nutritional function for the parasite that may be amenable to therapeutic manipulation.
DOI: 10.1016/j.jsb.2012.09.001
2012
Cited 50 times
Structural characterization and inhibition of the Plasmodium Atg8–Atg3 interaction
The autophagy-related proteins are thought to serve multiple functions in Plasmodium and are considered essential to parasite survival and development. We have studied two key interacting proteins, Atg8 and Atg3, of the autophagy pathway in Plasmodium falciparum. These proteins are vital for the formation and elongation of the autophagosome and essential to the process of macroautophagy. Autophagy may be required for conversion of the sporozoite into erythrocytic-infective merozoites and may be crucial for other functions during asexual blood stages. Here we describe the identification of an Atg8 family interacting motif (AIM) in Plasmodium Atg3, which binds Plasmodium Atg8. We determined the co-crystal structure of PfAtg8 with a short Atg3103–110 peptide, corresponding to this motif, to 2.2 Å resolution. Our in vitro interaction studies are in agreement with our X-ray crystal structure. Furthermore they suggest an important role for a unique Apicomplexan loop absent from human Atg8 homologues. Prevention of the protein–protein interaction of full length PfAtg8 with PfAtg3 was achieved at low micromolar concentrations with a small molecule, 1,2,3-trihydroxybenzene. Together our structural and interaction studies represent a starting point for future antimalarial drug discovery and design for this novel protein–protein interaction.
DOI: 10.1074/jbc.m115.671974
2016
Cited 48 times
A Lipolytic Lecithin:Cholesterol Acyltransferase Secreted by Toxoplasma Facilitates Parasite Replication and Egress
The protozoan parasite <i>Toxoplasma gondii</i> develops within a parasitophorous vacuole (PV) in mammalian cells, where it scavenges cholesterol. When cholesterol is present in excess in its environment, the parasite expulses this lipid into the PV or esterifies it for storage in lipid bodies. Here, we characterized a unique <i>T. gondii</i> homologue of mammalian lecithin:cholesterol acyltransferase (LCAT), a key enzyme that produces cholesteryl esters via transfer of acyl groups from phospholipids to the 3-OH of free cholesterol, leading to the removal of excess cholesterol from tissues. TgLCAT contains a motif characteristic of serine lipases "AHSLG" and the catalytic triad consisting of serine, aspartate, and histidine (SDH) from LCAT enzymes. TgLCAT is secreted by the parasite, but unlike other LCAT enzymes it is cleaved into two proteolytic fragments that share the residues of the catalytic triad and need to be reassembled to reconstitute enzymatic activity. TgLCAT uses phosphatidylcholine as substrate to form lysophosphatidylcholine that has the potential to disrupt membranes. The released fatty acid is transferred to cholesterol, but with a lower transesterification activity than mammalian LCAT. TgLCAT is stored in a subpopulation of dense granule secretory organelles, and following secretion, it localizes to the PV and parasite plasma membrane. LCAT-null parasites have impaired growth <i>in vitro</i>, reduced virulence in animals, and exhibit delays in egress from host cells. Parasites overexpressing LCAT show increased virulence and faster egress. These observations demonstrate that TgLCAT influences the outcome of an infection, presumably by facilitating replication and egress depending on the developmental stage of the parasite.
DOI: 10.1002/ana.25351
2018
Cited 42 times
<i>KCTD7</i> deficiency defines a distinct neurodegenerative disorder with a conserved autophagy‐lysosome defect
Objective Several small case series identified KCTD7 mutations in patients with a rare autosomal recessive disorder designated progressive myoclonic epilepsy (EPM3) and neuronal ceroid lipofuscinosis (CLN14). Despite the name KCTD (potassium channel tetramerization domain), KCTD protein family members lack predicted channel domains. We sought to translate insight gained from yeast studies to uncover disease mechanisms associated with deficiencies in KCTD7 of unknown function. Methods Novel KCTD7 variants in new and published patients were assessed for disease causality using genetic analyses, cell‐based functional assays of patient fibroblasts and knockout yeast, and electron microscopy of patient samples. Results Patients with KCTD7 mutations can exhibit movement disorders or developmental regression before seizure onset, and are distinguished from similar disorders by an earlier age of onset. Although most published KCTD7 patient variants were excluded from a genome sequence database of normal human variations, most newly identified patient variants are present in this database, potentially challenging disease causality. However, genetic analysis and impaired biochemical interactions with cullin 3 support a causal role for patient KCTD7 variants, suggesting deleterious alleles of KCTD7 and other rare disease variants may be underestimated. Both patient‐derived fibroblasts and yeast lacking Whi2 with sequence similarity to KCTD7 have impaired autophagy consistent with brain pathology. Interpretation Biallelic KCTD7 mutations define a neurodegenerative disorder with lipofuscin and lipid droplet accumulation but without defining features of neuronal ceroid lipofuscinosis or lysosomal storage disorders. KCTD7 deficiency appears to cause an underlying autophagy‐lysosome defect conserved in yeast, thereby assigning a biological role for KCTD7. Ann Neurol 2018;84:774–788
DOI: 10.1371/journal.ppat.1005647
2016
Cited 40 times
Na+ Influx Induced by New Antimalarials Causes Rapid Alterations in the Cholesterol Content and Morphology of Plasmodium falciparum
Among the several new antimalarials discovered over the past decade are at least three clinical candidate drugs, each with a distinct chemical structure, that disrupt Na+ homeostasis resulting in a rapid increase in intracellular Na+ concentration ([Na+]i) within the erythrocytic stages of Plasmodium falciparum. At present, events triggered by Na+ influx that result in parasite demise are not well-understood. Here we report effects of two such drugs, a pyrazoleamide and a spiroindolone, on intraerythrocytic P. falciparum. Within minutes following the exposure to these drugs, the trophozoite stage parasite, which normally contains little cholesterol, was made permeant by cholesterol-dependent detergents, suggesting it acquired a substantial amount of the lipid. Consistently, the merozoite surface protein 1 and 2 (MSP1 and MSP2), glycosylphosphotidylinositol (GPI)-anchored proteins normally uniformly distributed in the parasite plasma membrane, coalesced into clusters. These alterations were not observed following drug treatment of P. falciparum parasites adapted to grow in a low [Na+] growth medium. Both cholesterol acquisition and MSP1 coalescence were reversible upon the removal of the drugs, implicating an active process of cholesterol exclusion from trophozoites that we hypothesize is inhibited by high [Na+]i. Electron microscopy of drug-treated trophozoites revealed substantial morphological changes normally seen at the later schizont stage including the appearance of partial inner membrane complexes, dense organelles that resemble "rhoptries" and apparent nuclear division. Together these results suggest that [Na+]i disruptor drugs by altering levels of cholesterol in the parasite, dysregulate trophozoite to schizont development and cause parasite demise.
DOI: 10.1128/aac.00347-18
2018
Cited 38 times
Novel Approaches To Kill Toxoplasma gondii by Exploiting the Uncontrolled Uptake of Unsaturated Fatty Acids and Vulnerability to Lipid Storage Inhibition of the Parasite
Toxoplasma gondii , an obligate intracellular parasite replicating in mammalian cells within a parasitophorous vacuole (PV), is an avid scavenger of lipids retrieved from the host cell. Following lipid uptake, this parasite stores excess lipids in lipid droplets (LD).
DOI: 10.1128/mbio.01510-18
2018
Cited 38 times
A Member of the Ferlin Calcium Sensor Family Is Essential for Toxoplasma gondii Rhoptry Secretion
Invasion of host cells by apicomplexan parasites such as Toxoplasma gondii is critical for their infectivity and pathogenesis. In Toxoplasma, secretion of essential egress, motility, and invasion-related proteins from microneme organelles is regulated by oscillations of intracellular Ca2+ Later stages of invasion are considered Ca2+ independent, including the secretion of proteins required for host cell entry and remodeling from the parasite's rhoptries. We identified a family of three Toxoplasma proteins with homology to the ferlin family of double C2 domain-containing Ca2+ sensors. In humans and model organisms, such Ca2+ sensors orchestrate Ca2+-dependent exocytic membrane fusion with the plasma membrane. Here we focus on one ferlin that is conserved across the Apicomplexa, T. gondii FER2 (TgFER2). Unexpectedly, conditionally TgFER2-depleted parasites secreted their micronemes normally and were completely motile. However, these parasites were unable to invade host cells and were therefore not viable. Knockdown of TgFER2 prevented rhoptry secretion, and these parasites failed to form the moving junction at the parasite-host interface necessary for host cell invasion. Collectively, these data demonstrate the requirement of TgFER2 for rhoptry secretion in Toxoplasma tachyzoites and suggest a possible Ca2+ dependence of rhoptry secretion. These findings provide the first mechanistic insights into this critical yet poorly understood aspect of apicomplexan host cell invasion.IMPORTANCE Apicomplexan protozoan parasites, such as those causing malaria and toxoplasmosis, must invade the cells of their hosts in order to establish a pathogenic infection. Timely release of proteins from a series of apical organelles is required for invasion. Neither the vesicular fusion events that underlie secretion nor the observed reliance of the various processes on changes in intracellular calcium concentrations is completely understood. We identified a group of three proteins with strong homology to the calcium-sensing ferlin family, which are known to be involved in protein secretion in other organisms. Surprisingly, decreasing the amounts of one of these proteins (TgFER2) did not have any effect on the typically calcium-dependent steps in invasion. Instead, TgFER2 was essential for the release of proteins from organelles called rhoptries. These data provide a tantalizing first look at the mechanisms controlling the very poorly understood process of rhoptry secretion, which is essential for the parasite's infection cycle.
DOI: 10.1016/j.devcel.2020.05.014
2020
Cited 32 times
Endolysosomal Targeting of Mitochondria Is Integral to BAX-Mediated Mitochondrial Permeabilization during Apoptosis Signaling
Mitochondrial outer membrane permeabilization (MOMP) is a core event in apoptosis signaling. However, the underlying mechanism of BAX and BAK pore formation remains incompletely understood. We demonstrate that mitochondria are globally and dynamically targeted by endolysosomes (ELs) during MOMP. In response to pro-apoptotic BH3-only protein signaling and pharmacological MOMP induction, ELs increasingly form transient contacts with mitochondria. Subsequently, ELs rapidly accumulate within the entire mitochondrial compartment. This switch-like accumulation period temporally coincides with mitochondrial BAX clustering and cytochrome c release. Remarkably, interactions of ELs with mitochondria control BAX recruitment and pore formation. Knockdown of Rab5A, Rab5C, or USP15 interferes with EL targeting of mitochondria and functionally uncouples BAX clustering from cytochrome c release, while knockdown of the Rab5 exchange factor Rabex-5 impairs both BAX clustering and cytochrome c release. Together, these data reveal that EL-mitochondrial inter-organelle communication is an integral regulatory component of functional MOMP execution during cellular apoptosis signaling.
DOI: 10.7554/elife.59384
2021
Cited 27 times
Toxoplasma TgATG9 is critical for autophagy and long-term persistence in tissue cysts
Many of the world's warm-blooded species are chronically infected with Toxoplasma gondii tissue cysts, including an estimated one-third of the global human population. The cellular processes that permit long-term persistence within the cyst are largely unknown for T. gondii and related coccidian parasites that impact human and animal health. Herein, we show that genetic ablation of TgATG9 substantially reduces canonical autophagy and compromises bradyzoite viability. Transmission electron microscopy revealed numerous structural abnormalities occurring in ∆atg9 bradyzoites. Intriguingly, abnormal mitochondrial networks were observed in TgATG9-deficient bradyzoites, some of which contained numerous different cytoplasmic components and organelles. ∆atg9 bradyzoite fitness was drastically compromised in vitro and in mice, with very few brain cysts identified in mice 5 weeks post-infection. Taken together, our data suggests that TgATG9, and by extension autophagy, is critical for cellular homeostasis in bradyzoites and is necessary for long-term persistence within the cyst of this coccidian parasite.
DOI: 10.1371/journal.ppat.1010818
2022
Cited 17 times
The inclusion membrane protein IncS is critical for initiation of the Chlamydia intracellular developmental cycle
All Chlamydia species are obligate intracellular bacteria that undergo a unique biphasic developmental cycle strictly in the lumen of a membrane bound compartment, the inclusion. Chlamydia specific Type III secreted effectors, known as inclusion membrane proteins (Inc), are embedded into the inclusion membrane. Progression through the developmental cycle, in particular early events of conversion from infectious (EB) to replicative (RB) bacteria, is important for intracellular replication, but poorly understood. Here, we identified the inclusion membrane protein IncS as a critical factor for Chlamydia development. We show that a C. trachomatis conditional mutant is impaired in transition from EB to RB in human cells, and C. muridarum mutant bacteria fail to develop in a mouse model of Chlamydia infection. Thus, IncS represents a promising target for therapeutic intervention of the leading cause of sexually transmitted infections of bacterial origin.
DOI: 10.1128/mbio.01442-22
2022
Cited 14 times
Dense Granule Protein GRA64 Interacts with Host Cell ESCRT Proteins during<i>Toxoplasma gondii</i>Infection
The intracellular parasite Toxoplasma gondii adapts to diverse host cell environments within a replicative compartment that is heavily decorated by secreted proteins. In an attempt to identify novel parasite secreted proteins that influence host cell activity, we identified and characterized a transmembrane dense granule protein dubbed GRA64 (TGME49_202620). We found that GRA64 is on the parasitophorous vacuolar membrane (PVM) and is partially exposed to the host cell cytoplasm in both tachyzoite and bradyzoite parasitophorous vacuoles. Using co-immunoprecipitation and proximity-based biotinylation approaches, we demonstrated that GRA64 appears to interact with components of the host endosomal sorting complexes required for transport (ESCRT). Genetic disruption of GRA64 does not affect acute Toxoplasma virulence or encystation in mice, as observed via tissue cyst burdens in mice during chronic infection. However, ultrastructural analysis of Δgra64 tissue cysts using electron tomography revealed enlarged vesicular structures underneath the cyst membrane, suggesting a role for GRA64 in organizing the recruitment of ESCRT proteins and subsequent intracystic vesicle formation. This study uncovers a novel host-parasite interaction that contributes to an emerging paradigm in which specific host ESCRT proteins are recruited to the limiting membranes (PVMs) of tachyzoite and bradyzoite vacuoles formed during acute and chronic Toxoplasma infection. IMPORTANCE Toxoplasma gondii is a widespread foodborne parasite that causes congenital disease and life-threatening complications in immunocompromised individuals. Part of this parasite's success lies in its ability to infect diverse organisms and host cells and to persist as a latent infection within parasite-constructed structures called tissue cysts. In this study, we characterized a protein that is secreted by T. gondii into its parasitophorous vacuole during intracellular infection, which we dub GRA64. On the vacuolar membrane, this protein is exposed to the host cell cytosol and interacts with specific host ESCRT proteins. Parasites lacking the GRA64 protein exhibit ultrastructural changes in tissue cysts during chronic infection. This study lays the foundation for future studies on the mechanics and consequences of host ESCRT-parasite protein interactions.
DOI: 10.1101/2023.01.16.524187
2023
Cited 6 times
<i>In vitro</i>production of cat-restricted<i>Toxoplasma</i>pre-sexual stages by epigenetic reprogramming
Summary paragraph Sexual reproduction of Toxoplasma gondii , which is restricted to the small intestine of felids, is sparsely documented, due to ethical concerns surrounding the use of cats as model organisms. Chromatin modifiers dictate the developmental fate of the parasite during its multistage life cycle, but their targeting to stage-specific cistromes is poorly described 1 . In this study, we found that transcription factors AP2XII-1 and AP2XI-2, expressed in tachyzoite stage that causes acute toxoplasmosis, can silence genes necessary for merozoites, a developmental stage critical for sexual commitment and transmission to the next host, including humans. Their conditional and simultaneous depletion leads to a drastic change in the transcriptional program, promoting a complete transition from tachyzoites to merozoites. Pre-gametes produced in vitro under these conditions are characterized by specific protein markers and undergo typical asexual endopolygenic division cycles. In tachyzoites, AP2XII-1 and AP2XI-2 bind DNA as heterodimers at merozoite promoters and recruit the epigenitors MORC and HDAC3 1 , which in turn restrict the accessibility of chromatin to the transcriptional machinery. Thus, the commitment to merogony stems from a profound epigenetic rewiring orchestrated by AP2XII-1 and AP2XI-2. This effective in vitro culture of merozoites paves the way to explore Toxoplasma sexual reproduction without the need to infect kittens and has potential for the development of therapeutics to block parasite transmission.
DOI: 10.1371/journal.pbio.3002634
2024
Systematic characterization of all Toxoplasma gondii TBC domain-containing proteins identifies an essential regulator of Rab2 in the secretory pathway
Toxoplasma gondii resides in its intracellular niche by employing a series of specialized secretory organelles that play roles in invasion, host cell manipulation, and parasite replication. Rab GTPases are major regulators of the parasite's secretory traffic that function as nucleotide-dependent molecular switches to control vesicle trafficking. While many of the Rab proteins have been characterized in T. gondii, precisely how these Rabs are regulated remains poorly understood. To better understand the parasite's secretory traffic, we investigated the entire family of Tre2-Bub2-Cdc16 (TBC) domain-containing proteins, which are known to be involved in vesicle fusion and secretory protein trafficking. We first determined the localization of all 18 TBC domain-containing proteins to discrete regions of the secretory pathway or other vesicles in the parasite. Second, we use an auxin-inducible degron approach to demonstrate that the protozoan-specific TgTBC9 protein, which localizes to the endoplasmic reticulum (ER), is essential for parasite survival. Knockdown of TgTBC9 results in parasite growth arrest and affects the organization of the ER and mitochondrial morphology. TgTBC9 knockdown also results in the formation of large lipid droplets (LDs) and multi-membranous structures surrounded by ER membranes, further indicating a disruption of ER functions. We show that the conserved dual-finger active site in the TBC domain of the protein is critical for its GTPase-activating protein (GAP) function and that the Plasmodium falciparum orthologue of TgTBC9 can rescue the lethal knockdown. We additionally show by immunoprecipitation and yeast 2 hybrid analyses that TgTBC9 preferentially binds Rab2, indicating that the TBC9-Rab2 pair controls ER morphology and vesicular trafficking in the parasite. Together, these studies identify the first essential TBC protein described in any protozoan and provide new insight into intracellular vesicle trafficking in T. gondii.
DOI: 10.1016/0001-706x(93)90096-t
1993
Cited 77 times
The uptake of the trypanocidal drug suramin in combination with low-density lipoproteins by Trypanosoma brucei and its possible mode of action
In plasma, a significant part of suramin circulates in tight association with low-density lipoproteins (LDL). At therapeutically obtainable concentrations (100 μM) of suramin, about 85% of the total amount of the drug was bound to proteins, approximately 15% of which was bound to LDL. The molar ratio of suramin bound to LDL in serum was 7.5. The capacity of the high-affinity binding sites of LDL were 6.6 × 106 M−1, both in Tris buffer and in ultrafiltrate of serum. Suramin (100 μM) decreased the uptake of host LDL through receptor-mediated endocytosis by Trypanosoma brucei, with approximately 50%. LDL served as the only carrier for suramin uptake. Serum albumin, another important carrier for suramin in blood, was not able to promote suramin uptake, neither was delipidified plasma. The suramin taken up by T. brucei was recovered, in part, in the lysosomal fractions. It is suggested that deprivation of the parasite from cholesterol and phospholipids by an inhibition of the uptake of LDL, contributes to the mode of action of suramin, in addition to the many other effects that the drug may exert on the parasite. The toxic side-effects of suramin on the host are discussed in the light of its association with circulating lipoproteins.
DOI: 10.1074/jbc.270.11.5736
1995
Cited 77 times
Host Plasma Low Density Lipoprotein Particles as an Essential Source of Lipids for the Bloodstream Forms of Trypanosoma brucei
In contrast to mammalian cells, bloodstream forms of Trypanosoma brucei show no activity for fatty acid and sterol synthesis and critically depend on plasma low density lipoprotein (LDL) particles for their rapid growth. We report here that these parasites acquire such lipids by receptor-mediated endocytosis of LDL, subsequent lysosomal degradation of apoprotein B-LDL, and utilization of these lipids. Uptake of LDL-associated [3H]sphingomyelin and of LDL-associated [3H]cholesteryl oleate paralleled each other, and that of 125I-apoprotein B-LDL showed saturation and could be inhibited by unlabeled LDL or by anti-LDL receptor antibodies. Metabolism of lipids carried by LDL was abolished by chloroquine and by the thiol protease inhibitor, leupeptin. Sphingomyelin was cleaved by an acid sphingomyelinase to yield ceramide, which was itself split up into sphingosine and fatty acids. The latter were further incorporated into phosphatidylcholine, triacylglycerols, or cholesteryl esters. Similarly, cholesteryl oleate was hydrolyzed by an acid lipase to yield free cholesterol, which was reesterified with fatty acids, presumably in the cytosol. Like free cholesterol, LDL provided substrate for cholesterol esterification. In the culture-adapted procyclic form of T. brucei, which is capable of sterol synthesis, exogenous LDL-cholesterol rather than endogenously synthesized sterol was utilized for sterol esterification. Interference with exogenous supply of lipids via receptor-mediated endocytosis of LDL should be explored to fight against trypanosomiasis. In contrast to mammalian cells, bloodstream forms of Trypanosoma brucei show no activity for fatty acid and sterol synthesis and critically depend on plasma low density lipoprotein (LDL) particles for their rapid growth. We report here that these parasites acquire such lipids by receptor-mediated endocytosis of LDL, subsequent lysosomal degradation of apoprotein B-LDL, and utilization of these lipids. Uptake of LDL-associated [3H]sphingomyelin and of LDL-associated [3H]cholesteryl oleate paralleled each other, and that of 125I-apoprotein B-LDL showed saturation and could be inhibited by unlabeled LDL or by anti-LDL receptor antibodies. Metabolism of lipids carried by LDL was abolished by chloroquine and by the thiol protease inhibitor, leupeptin. Sphingomyelin was cleaved by an acid sphingomyelinase to yield ceramide, which was itself split up into sphingosine and fatty acids. The latter were further incorporated into phosphatidylcholine, triacylglycerols, or cholesteryl esters. Similarly, cholesteryl oleate was hydrolyzed by an acid lipase to yield free cholesterol, which was reesterified with fatty acids, presumably in the cytosol. Like free cholesterol, LDL provided substrate for cholesterol esterification. In the culture-adapted procyclic form of T. brucei, which is capable of sterol synthesis, exogenous LDL-cholesterol rather than endogenously synthesized sterol was utilized for sterol esterification. Interference with exogenous supply of lipids via receptor-mediated endocytosis of LDL should be explored to fight against trypanosomiasis.
DOI: 10.1128/iai.00551-07
2007
Cited 65 times
Protective Properties and Surface Localization of<i>Plasmodium falciparum</i>Enolase
The enolase protein of the human malarial parasite Plasmodium falciparum has recently been characterized. Apart from its glycolytic function, enolase has also been shown to possess antigenic properties and to be present on the cell wall of certain invasive organisms, such as Candida albicans. In order to assess whether enolase of P. falciparum is also antigenic, sera from residents of a region of Eastern India where malaria is endemic were tested against the recombinant P. falciparum enolase (r-Pfen) protein. About 96% of immune adult sera samples reacted with r-Pfen over and above the seronegative controls. Rabbit anti-r-Pfen antibodies inhibited the growth of in vitro cultures of P. falciparum. Mice immunized with r-Pfen showed protection against a challenge with the 17XL lethal strain of the mouse malarial parasite Plasmodium yoelii. The antibodies raised against r-Pfen were specific for Plasmodium and did not react to the host tissues. Immunofluorescence as well as electron microscopic examinations revealed localization of the enolase protein on the merozoite cell surface. These observations establish malaria enolase to be a potential protective antigen.
DOI: 10.1371/journal.ppat.1000302
2009
Cited 57 times
The Glutathione Biosynthetic Pathway of Plasmodium Is Essential for Mosquito Transmission
Infection of red blood cells (RBC) subjects the malaria parasite to oxidative stress. Therefore, efficient antioxidant and redox systems are required to prevent damage by reactive oxygen species. Plasmodium spp. have thioredoxin and glutathione (GSH) systems that are thought to play a major role as antioxidants during blood stage infection. In this report, we analyzed a critical component of the GSH biosynthesis pathway using reverse genetics. Plasmodium berghei parasites lacking expression of gamma-glutamylcysteine synthetase (gamma-GCS), the rate limiting enzyme in de novo synthesis of GSH, were generated through targeted gene disruption thus demonstrating, quite unexpectedly, that gamma-GCS is not essential for blood stage development. Despite a significant reduction in GSH levels, blood stage forms of pbggcs(-) parasites showed only a defect in growth as compared to wild type. In contrast, a dramatic effect on development of the parasites in the mosquito was observed. Infection of mosquitoes with pbggcs(-) parasites resulted in reduced numbers of stunted oocysts that did not produce sporozoites. These results have important implications for the design of drugs aiming at interfering with the GSH redox-system in blood stages and demonstrate that de novo synthesis of GSH is pivotal for development of Plasmodium in the mosquito.
DOI: 10.1074/jbc.m110.122275
2010
Cited 52 times
Glutathione Reductase-null Malaria Parasites Have Normal Blood Stage Growth but Arrest during Development in the Mosquito
Malaria parasites contain a complete glutathione (GSH) redox system, and several enzymes of this system are considered potential targets for antimalarial drugs. Through generation of a gamma-glutamylcysteine synthetase (gamma-GCS)-null mutant of the rodent parasite Plasmodium berghei, we previously showed that de novo GSH synthesis is not critical for blood stage multiplication but is essential for oocyst development. In this study, phenotype analyses of mutant parasites lacking expression of glutathione reductase (GR) confirmed that GSH metabolism is critical for the mosquito oocyst stage. Similar to what was found for gamma-GCS, GR is not essential for blood stage growth. GR-null parasites showed the same sensitivity to methylene blue and eosin B as wild type parasites, demonstrating that these compounds target molecules other than GR in Plasmodium. Attempts to generate parasites lacking both GR and gamma-GCS by simultaneous disruption of gr and gamma-gcs were unsuccessful. This demonstrates that the maintenance of total GSH levels required for blood stage survival is dependent on either de novo GSH synthesis or glutathione disulfide (GSSG) reduction by Plasmodium GR. Our studies provide new insights into the role of the GSH system in malaria parasites with implications for the development of drugs targeting GSH metabolism.
DOI: 10.4161/auto.27166
2013
Cited 38 times
Characterization of the ATG8-conjugation system in 2<i>Plasmodium</i>species with special focus on the liver stage
Plasmodium parasites successfully colonize different habitats within mammals and mosquitoes, and adaptation to various environments is accompanied by changes in their organelle composition and size. Previously, we observed that during hepatocyte infection, Plasmodium discards organelles involved in invasion and expands those implicated in biosynthetic pathways. We hypothesized that this process is regulated by autophagy. Plasmodium spp. possess a rudimentary set of known autophagy-related proteins that includes the ortholog of yeast Atg8. In this study, we analyzed the activity of the ATG8-conjugation pathway over the course of the lifecycle of Plasmodium falciparum and during the liver stage of Plasmodium berghei. We engineered a transgenic P. falciparum strain expressing mCherry-PfATG8. These transgenic parasites expressed mCherry-PfATG8 in human hepatocytes and erythrocytes, and in the midgut and salivary glands of Anopheles mosquitoes. In all observed stages, mCherry-PfATG8 was localized to tubular structures. Our EM and colocalization studies done in P. berghei showed the association of PbATG8 on the limiting membranes of the endosymbiont-derived plastid-like organelle known as the apicoplast. Interestingly, during parasite replication in hepatocytes, the association of PbATG8 with the apicoplast increases as this organelle expands in size. PbATG3, PbATG7 and PbATG8 are cotranscribed in all parasitic stages. Molecular analysis of PbATG8 and PbATG3 revealed a novel mechanism of interaction compared with that observed for other orthologs. This is further supported by the inability of Plasmodium ATG8 to functionally complement atg8Δ yeast or localize to autophagosomes in starved mammalian cells. Altogether, these data suggests a unique role for the ATG8-conjugation system in Plasmodium parasites.
DOI: 10.1128/ec.00262-14
2015
Cited 38 times
Neospora caninum Recruits Host Cell Structures to Its Parasitophorous Vacuole and Salvages Lipids from Organelles
ABSTRACT Toxoplasma gondii and Neospora caninum , which cause the diseases toxoplasmosis and neosporosis, respectively, are two closely related apicomplexan parasites. They have similar heteroxenous life cycles and conserved genomes and share many metabolic features. Despite these similarities, T. gondii and N. caninum differ in their transmission strategies and zoonotic potential. Comparative analyses of the two parasites are important to identify the unique biological features that underlie the basis of host preference and pathogenicity. T. gondii and N. caninum are obligate intravacuolar parasites; in contrast to T. gondii , events that occur during N. caninum infection remain largely uncharacterized. We examined the capability of N. caninum (Liverpool isolate) to interact with host organelles and scavenge nutrients in comparison to that of T. gondii (RH strain). N. caninum reorganizes the host microtubular cytoskeleton and attracts endoplasmic reticulum (ER), mitochondria, lysosomes, multivesicular bodies, and Golgi vesicles to its vacuole though with some notable differences from T. gondii . For example, the host ER gathers around the N. caninum parasitophorous vacuole (PV) but does not physically associate with the vacuolar membrane; the host Golgi apparatus surrounds the N. caninum PV but does not fragment into ministacks. N. caninum relies on plasma lipoproteins and scavenges cholesterol from NPC1-containing endocytic organelles. This parasite salvages sphingolipids from host Golgi Rab14 vesicles that it sequesters into its vacuole. Our data highlight a remarkable degree of conservation in the intracellular infection program of N. caninum and T. gondii . The minor differences between the two parasites related to the recruitment and rearrangement of host organelles around their vacuoles likely reflect divergent evolutionary paths.
DOI: 10.1111/cmi.12749
2017
Cited 33 times
A novel dense granule protein, GRA41, regulates timing of egress and calcium sensitivity in<i>Toxoplasma gondii</i>
Toxoplasma gondii is an obligate intracellular apicomplexan parasite with high seroprevalence in humans. Repeated lytic cycles of invasion, replication, and egress drive both the propagation and the virulence of this parasite. Key steps in this cycle, including invasion and egress, depend on tightly regulated calcium fluxes and, although many of the calcium-dependent effectors have been identified, the factors that detect and regulate the calcium fluxes are mostly unknown. To address this knowledge gap, we used a forward genetic approach to isolate mutants resistant to extracellular exposure to the calcium ionophore A23187. Through whole genome sequencing and complementation, we have determined that a nonsense mutation in a previously uncharacterised protein is responsible for the ionophore resistance of one of the mutants. The complete loss of this protein recapitulates the resistance phenotype and importantly shows defects in calcium regulation and in the timing of egress. The affected protein, GRA41, localises to the dense granules and is secreted into the parasitophorous vacuole where it associates with the tubulovesicular network. Our findings support a connection between the tubulovesicular network and ion homeostasis within the parasite, and thus a novel role for the vacuole of this important pathogen.
DOI: 10.1371/journal.ppat.1006893
2018
Cited 31 times
Hostile intruder: Toxoplasma holds host organelles captive
DOI: 10.1038/s41467-021-25987-5
2021
Cited 20 times
A plastid two-pore channel essential for inter-organelle communication and growth of Toxoplasma gondii
Two-pore channels (TPCs) are a ubiquitous family of cation channels that localize to acidic organelles in animals and plants to regulate numerous Ca2+-dependent events. Little is known about TPCs in unicellular organisms despite their ancient origins. Here, we characterize a TPC from Toxoplasma gondii, the causative agent of toxoplasmosis. TgTPC is a member of a novel clad of TPCs in Apicomplexa, distinct from previously identified TPCs and only present in coccidians. We show that TgTPC localizes not to acidic organelles but to the apicoplast, a non-photosynthetic plastid found in most apicomplexan parasites. Conditional silencing of TgTPC resulted in progressive loss of apicoplast integrity, severely affecting growth and the lytic cycle. Isolation of TPC null mutants revealed a selective role for TPCs in replication independent of apicoplast loss that required conserved residues within the pore-lining region. Using a genetically-encoded Ca2+ indicator targeted to the apicoplast, we show that Ca2+ signals deriving from the ER but not from the extracellular space are selectively transmitted to the lumen. Deletion of the TgTPC gene caused reduced apicoplast Ca2+ uptake and membrane contact site formation between the apicoplast and the ER. Fundamental roles for TPCs in maintaining organelle integrity, inter-organelle communication and growth emerge.
DOI: 10.1101/2023.03.05.531216
2023
Cited 5 times
Functional profiling of the<i>Toxoplasma</i>genome during acute mouse infection
Within a host, pathogens encounter a diverse and changing landscape of cell types, nutrients, and immune responses. Examining host-pathogen interactions in animal models can therefore reveal aspects of infection absent from cell culture. We use CRISPR-based screens to functionally profile the entire genome of the model apicomplexan parasite Toxoplasma gondii during mouse infection. Barcoded gRNAs were used to track mutant parasite lineages, enabling detection of bottlenecks and mapping of population structures. We uncovered over 300 genes that modulate parasite fitness in mice with previously unknown roles in infection. These candidates span multiple axes of host-parasite interaction, including determinants of tropism, host organelle remodeling, and metabolic rewiring. We mechanistically characterized three novel candidates, including GTP cyclohydrolase I, against which a small-molecule inhibitor could be repurposed as an antiparasitic compound. This compound exhibited antiparasitic activity against T. gondii and Plasmodium falciparum, the most lethal agent of malaria. Taken together, we present the first complete survey of an apicomplexan genome during infection of an animal host, and point to novel interfaces of host-parasite interaction that may offer new avenues for treatment.
DOI: 10.1016/j.molbiopara.2004.08.004
2004
Cited 61 times
On the biogenesis of lipid bodies in ancient eukaryotes: synthesis of triacylglycerols by a Toxoplasma DGAT1-related enzyme
In mammalian cells, the main stored neutral lipids are triacylglycerol and cholesteryl esters, which are produced by two related enzymes, acyl-CoA:diacylglycerol acyltransferase (DGAT) and acyl-CoA:cholesterol acyltransferase (ACAT), respectively. Very little is known about the metabolism, intracellular storage and function of neutral lipids in many pathogenic lower eukaryotes. In this paper, we have characterized the activity of an important triacylglycerol synthetic enzyme in the protozoan Toxoplasma gondii. A full-length cDNA and gene encoding a T. gondii DGAT1-related enzyme were identified and designated TgDGAT1. The gene is composed of 15 exons and 14 introns, and encodes a protein with a predicted Mr 63.5 kDa, containing signature motifs characteristic of the DGAT1 family. The native protein migrates at 44 kDa under reducing conditions. TgDGAT1 is an integral membrane protein localized to the parasite cortical and perinuclear endoplasmic reticulum, with the C-terminus oriented to the lumen of the organelle. When a Saccharomyces cerevisiae mutant strain lacking neutral lipid production is transformed with TgDGAT1 cDNA, a significant DGAT activity is reconstituted, resulting in triacylglycerol synthesis and biogenesis of cytosolic lipid inclusions, resembling lipid bodies in T. gondii. No production of steryl esters is observed upon TgDGAT1 expression in yeast. In contrast to human DGAT1 lacking fatty acid specificity, TgDGAT1 preferentially incorporates palmitate. Our results indicate that parasitic protozoa are also neutral lipid accumulators and illustrate the first example of the existence of a functional DGAT gene in an ancient eukaryote, demonstrating that diacylglycerol esterification is evolutionarily conserved.
DOI: 10.1016/j.ijpara.2005.01.009
2005
Cited 60 times
Insights into unique physiological features of neutral lipids in Apicomplexa: from storage to potential mediation in parasite metabolic activities
The fast intracellular multiplication of apicomplexan parasites including Toxoplasma and Plasmodium, requires large amounts of lipids necessary for the membrane biogenesis of new progenies. Hence, the study of lipids is fundamental in order to understand the biology and pathogenesis of these deadly organisms. Much has been reported on the importance of polar lipids, e.g. phospholipids in Plasmodium. Comparatively, little attention has been paid to the metabolism of neutral lipids, including sterols, steryl esters and acylglycerols. In eukaryotic cells, free sterols are membrane components whereas steryl esters and acylglycerols are stored in cytosolic lipid inclusions. The first part of this review describes the recent advances in neutral lipid synthesis and storage in Toxoplasma and Plasmodium. New potential pharmacological targets in the pathways producing neutral lipids are outlined. In addition to lipid bodies, Apicomplexa contain unique secretory organelles involved in parasite invasion named rhoptries. These compartments appear to sequester most of the cholesterol found in the exocytic pathway. The second part of the review focuses on rhoptry cholesterol and its potential roles in the biogenesis, structural organisation and function of these unique organelles among eukaryotes.
DOI: 10.1046/j.1462-5822.2002.00178.x
2002
Cited 57 times
Toxoplasma gondii Rab5 enhances cholesterol acquisition from host cells
The role of endocytosis in nutrient uptake by Toxoplasma gondii is unknown. To explore this issue, we characterized an endosomal compartment by identifying a T. gondii Rab5 homologue, a molecular marker for early endosomes in eukaryotic cells. The deduced amino acid sequence of the T. gondii Rab5 gene encodes a protein of 240 amino acids, which we termed TgRab51. TgRab51 was epitope-tagged at the N-terminus, expressed in the parasite, and localized by immunofluorescence and immunoelectron microscopy to tubulovesicular structures anterior to the parasite nucleus and adjacent to, but distinct from the Golgi. By immunofluorescence analysis, TgRab51wt-HA staining partially overlapped with Golgi/TGN markers, but not with the T. gondii secretory organelles. A dominant positive mutant, TgRab51Q103L-HA, enhanced uptake of exogenous cholesterol analogues in intracellular parasites, augmented formation of lipid droplets and accelerated parasite growth. Brefeldin A disrupted the TgRab51 compartment, and altered the distribution of fluorescent exogenous cholesterol in cells expressing TgRab51Q103L-HA. These results suggest that TgRab51 facilitates sterol uptake, possibly through a Golgi-dependent pathway.
DOI: 10.1016/j.molbiopara.2003.08.017
2004
Cited 53 times
Neutral lipid synthesis and storage in the intraerythrocytic stages of Plasmodium falciparum
In eukaryotic cells the neutral lipids, steryl esters and triacylglycerol, are synthesized by membrane-bound O-acyltransferases and stored in cytosolic lipid bodies. We show here that the intraerythrocytic stages of Plasmodium falciparum produce triacylglycerol using oleate and diacylglycerol as substrates. Parasite membrane preparations reveal a synthesis rate of 4.5 +/- 0.8 pmol x min(-1)mg(-1) of protein with maximal production occurring in the mid- and late-trophozoite stages in both, membrane preparations and live parasites. In contrast to other eukaryotic cells, no discernable amounts of steryl esters are produced, and the parasite is insensitive to cholesterol esterification inhibitors. Synthesized neutral lipids are stored as lipid bodies in the parasite cytosol in a stage specific manner. Their biogenesis is not modified upon incubation with excess fatty acids or lipoproteins or after lipoprotein depletion of the culture medium. We investigated on the enzymes involved in neutral lipid synthesis and found that only one gene with significant homology to known members of the membrane-bound O-acyltransferase family is present in the P. falciparum genome. It encodes a microsomal transmembrane protein with a predicted size of 78.1 kDa, which we named PfDGAT because of its close identity with various known acyl-CoA:diacylglycerol acyltransferases. PfDGAT is expressed in a stage specific manner as documented by Western blotting and immunoprecipitation assays using antibodies against Toxoplasma DGAT, suggesting that PfDGAT is the most likely candidate for plasmodial triacylglycerol synthesis.
DOI: 10.1111/j.1600-0854.2007.00637.x
2007
Cited 50 times
A Membrane Protease is Targeted to the Relict Plastid of <i>Toxoplasma </i>via an Internal Signal Sequence
The apicoplast is a secondary plastid found in Toxoplasma gondii, Plasmodium species and many other apicomplexan parasites. Although the apicoplast is essential to parasite survival, little is known about the protein constituents of the four membranes surrounding the organelle. Luminal proteins are directed to the endoplasmic reticulum (ER) by an N-terminal signal sequence and from there to the apicoplast by a transit peptide domain. We have identified a membrane-associated AAA protease in T. gondii, FtsH1. Although the protein lacks a canonical bipartite-targeting sequence, epitope-tagged FtsH1 colocalizes with the recently identified apicoplast membrane marker APT1 and immunoelectron microscopy confirms the residence of FtsH1 on plastid membranes. Trafficking appears to occur via the ER because deletion mutants lacking the peptidase domain are retained in the ER. When extended to include the peptidase domain, the protein trafficks properly. The transmembrane domain is required for localization of the full-length protein to the apicoplast and a truncation mutant to the ER. Thus, at least two distinct regions of FtsH1 are required for proper trafficking, but they differ from those of luminal proteins and would not be detected by the algorithms currently used to identify apicoplast proteins.
DOI: 10.1111/mmi.12142
2013
Cited 36 times
Characterization of a second sterol‐esterifying enzyme in <i><scp>T</scp>oxoplasma</i> highlights the importance of cholesterol storage pathways for the parasite
Summary Lipid bodies are eukaryotic structures for temporary storage of neutral lipids such as acylglycerols and steryl esters. Fatty acyl‐ CoA and cholesterol are two substrates for cholesteryl ester ( CE ) synthesis via the ACAT reaction. The intracellular parasite T oxoplasma gondii is incapable of sterol synthesis and unremittingly scavenges cholesterol from mammalian host cells. We previously demonstrated that the parasite expresses a cholesteryl ester‐synthesizing enzyme, TgACAT 1. In this article, we identified and characterized a second ACAT ‐like enzyme, TgACAT 2, which shares 56% identity with TgACAT 1. Both enzymes are endoplasmic reticulum‐associated and contribute to CE formation for storage in lipid bodies. While TgACAT 1 preferentially utilizes palmitoyl‐ CoA , TgACAT 2 has broader fatty acid specificity and produces more CE . Genetic ablation of each individual ACAT results in parasite growth impairment whereas dual ablation of ACAT1 and ACAT2 is not tolerated by T oxoplasma . Δ ACAT 1 and Δ ACAT 2 parasites have reduced CE levels, fewer lipid bodies, and accumulate free cholesterol, which causes injurious membrane effects. Mutant parasites are particularly vulnerable to ACAT inhibitors. This study underlines the important physiological role of ACAT enzymes to store cholesterol in a sterol‐auxotrophic organism such as T oxoplasma , and furthermore opens up possibilities of exploiting TgACAT as targets for the development of antitoxoplasmosis drugs.
DOI: 10.1111/cmi.12107
2013
Cited 36 times
<i>Cryptosporidium parvum</i>scavenges LDL-derived cholesterol and micellar cholesterol internalized into enterocytes
Cryptosporidium spp. are responsible for devastating diarrhoea in immunodeficient individuals. In the intestinal tract, the developmental stages of the parasite are confined to the apical surfaces of epithelial cells. Upon invasion, Cryptosporidium incorporates the microvillous membrane of the enterocyte to form the parasitophorous vacuole (PV) and sequesters itself from the host cytoplasm by rearranging the host cytoskeleton. Cryptosporidium parvum has minimal anabolic capabilities and relies on transporters and salvage pathways to meet its basic metabolic requirements. The cholesterol salvage pathway is crucial for the development of protozoan parasites. In this study, we have examined the sources of cholesterol from C. parvum infecting enterocytes. We illustrated that the intracellular stages of Cryptosporidium as well as the oocysts shed by the host, contain cholesterol. Incubation of infected enterocytes in lipoprotein-free medium impairs parasite development and results in substantial decrease in cholesterol content associated with the PV. Among lipoproteins, LDL constitutes an important source of cholesterol for Cryptosporidium. Dietary cholesterol incorporated into micelles is internalized into enterocytes by the NPC1L1 transporter. We showed that C. parvum also obtains cholesterol from micelles in enterocytes.Pharmacological blockade of NPC1L1 function by ezetimibe or moderate downregulation of NPC1L1 expression decreases parasite infectivity. These observations indicate that, despite its dual sequestration from the intestinal lumen and the host cytoplasm, C. parvum can, in fact, obtain cholesterol both from the gut's lumen and the host cell. This study highlights the evolutionary advantages for epicellular pathogens to access to nutrients from the outside and inside of the host cell.
DOI: 10.1074/jbc.m112.443697
2013
Cited 36 times
Non-canonical Maturation of Two Papain-family Proteases in Toxoplasma gondii
Proteases regulate key events during infection by the pervasive intracellular parasite Toxoplasma gondii. Understanding how parasite proteases mature from an inactive zymogen to an active enzyme is expected to inform new strategies for blocking their actions. Herein, we show that T. gondii cathepsin B protease (TgCPB) does not undergo self-maturation but instead requires the expression of a second papain-family cathepsin protease, TgCPL. Using recombinant enzymes we also show that TgCPL is capable of partially maturing TgCPB in vitro. Consistent with this interrelationship, antibodies with validated specificity detected TgCPB in the lysosome-like vacuolar compartment along with TgCPL. Our findings also establish that TgCPB does not localize to the rhoptries as previously reported. Accordingly, rhoptry morphology and rhoptry protein maturation are normal in TgCPB knock-out parasites. Finally, we show that although maturation of TgCPL is independent of TgCPB, it may involve an additional protease(s) in conjunction with self-maturation.
DOI: 10.1371/journal.pone.0087695
2014
Cited 33 times
Plasmodium falciparum Rab5B Is an N-Terminally Myristoylated Rab GTPase That Is Targeted to the Parasite's Plasma and Food Vacuole Membranes
Plasmodium falciparum (Pf) has a family of 11 Rab GTPases to regulate its vesicular transport. However, PfRab5B is unique in lacking a C-terminal geranyl-geranylation motif, while having N-terminal palmitoylation and myristoylation motifs. We show that the N-terminal glycine is required for PfRab5B myristoylation in vitro and when an N-terminal PfRab5B fragment possessing both acylation motifs is fused to GFP and expressed in transgenic P. falciparum parasites, the chimeric PfRab5B protein localizes to the plasma membrane. Upon substitution of the modified glycine by alanine the staining becomes diffuse and GFP is found in soluble subcellular fractions. Immuno-electron microscopy shows endogenous PfRab5B decorating the parasite's plasma and food vacuole membranes. Using reverse genetics rab5b couldn't be deleted from the haploid genome of asexual blood stage P. berghei parasites. The failure of PbRab5A or PbRab5C to complement for loss of PbRab5B function indicates non-overlapping roles for the three Plasmodium Rab5s, with PfRab5B involved in trafficking MSP1 to the food vacuole membrane and CK1 to the plasma membrane. We discuss similarities between Plasmodium Rab5B and Arabidopsis thaliana ARA6, a similarly unusual Rab5-like GTPase of plants.
DOI: 10.1128/mbio.00682-16
2016
Cited 29 times
Overexpression of Plasmodium berghei ATG8 by Liver Forms Leads to Cumulative Defects in Organelle Dynamics and to Generation of Noninfectious Merozoites
Plasmodium parasites undergo continuous cellular renovation to adapt to various environments in the vertebrate host and insect vector. In hepatocytes, Plasmodium berghei discards unneeded organelles for replication, such as micronemes involved in invasion. Concomitantly, intrahepatic parasites expand organelles such as the apicoplast that produce essential metabolites. We previously showed that the ATG8 conjugation system is upregulated in P. berghei liver forms and that P. berghei ATG8 (PbATG8) localizes to the membranes of the apicoplast and cytoplasmic vesicles. Here, we focus on the contribution of PbATG8 to the organellar changes that occur in intrahepatic parasites. We illustrated that micronemes colocalize with PbATG8-containing structures before expulsion from the parasite. Interference with PbATG8 function by overexpression results in poor development into late liver stages and production of small merosomes that contain immature merozoites unable to initiate a blood infection. At the cellular level, PbATG8-overexpressing P. berghei exhibits a delay in microneme compartmentalization into PbATG8-containing autophagosomes and elimination compared to parasites from the parental strain. The apicoplast, identifiable by immunostaining of the acyl carrier protein (ACP), undergoes an abnormally fast proliferation in mutant parasites. Over time, the ACP staining becomes diffuse in merosomes, indicating a collapse of the apicoplast. PbATG8 is not incorporated into the progeny of mutant parasites, in contrast to parental merozoites in which PbATG8 and ACP localize to the apicoplast. These observations reveal that Plasmodium ATG8 is a key effector in the development of merozoites by controlling microneme clearance and apicoplast proliferation and that dysregulation in ATG8 levels is detrimental for malaria infectivity.Malaria is responsible for more mortality than any other parasitic disease. Resistance to antimalarial medicines is a recurring problem; new drugs are urgently needed. A key to the parasite's successful intracellular development in the liver is the metabolic changes necessary to convert the parasite from a sporozoite to a replication-competent, metabolically active trophozoite form. Our study reinforces the burgeoning concept that organellar changes during parasite differentiation are mediated by an autophagy-like process. We have identified ATG8 in Plasmodium liver forms as an important effector that controls the development and fate of organelles, e.g., the clearance of micronemes that are required for hepatocyte invasion and the expansion of the apicoplast that produces many metabolites indispensable for parasite replication. Given the unconventional properties and the importance of ATG8 for parasite development in hepatocytes, targeting the parasite's autophagic pathway may represent a novel approach to control malarial infections.
DOI: 10.1016/j.mib.2017.10.009
2017
Cited 27 times
How Toxoplasma and malaria parasites defy first, then exploit host autophagic and endocytic pathways for growth
Infections caused by the apicomplexan parasites Plasmodium and Toxoplasma are wide-spread, life-threatening and therapeutically challenging. These pathogens are obligate intracellular microorganisms that invade mammalian cells by forming a self-made niche, the parasitophorous vacuole that is impervious to host lysosomal fusion. Shortly after invasion, a noncanonical xenophagic pathway resembling LC3-associated phagocytosis is activated by the host cell to control infections. However, Plasmodium and Toxoplasma have elaborated strategies to avoid clearance by the sentinel activities of the host autophagic system. After this initial confrontation, replicating Plasmodium and Toxoplasma adeptly usurp, for their own benefit, host autophagic and endocytic structures by attracting these organelles to their vacuole, likely to access their nutrient-rich content. The pleomorphic function of the autophagy system, from microbial defense to nutrient supply, is reflected by its ambivalent role during the intracellular development of these apicomplexan parasites.
DOI: 10.3389/fcimb.2022.882166
2022
Cited 11 times
Toxoplasma gondii’s Basal Complex: The Other Apicomplexan Business End Is Multifunctional
The Apicomplexa are famously named for their apical complex, a constellation of organelles at their apical end dedicated to invasion of their host cells. In contrast, at the other end of the cell, the basal complex (BC) has been overshadowed since it is much less prominent and specific functions were not immediately obvious. However, in the past decade a staggering array of functions have been associated with the BC and strides have been made in understanding its structure. Here, these collective insights are supplemented with new data to provide an overview of the understanding of the BC in Toxoplasma gondii . The emerging picture is that the BC is a dynamic and multifunctional complex, with a series of (putative) functions. The BC has multiple roles in cell division: it is the site where building blocks are added to the cytoskeleton scaffold; it exerts a two-step stretch and constriction mechanism as contractile ring; and it is key in organelle division. Furthermore, the BC has numerous putative roles in ‘import’, such as the recycling of mother cell remnants, the acquisition of host-derived vesicles, possibly the uptake of lipids derived from the extracellular medium, and the endocytosis of micronemal proteins. The latter process ties the BC to motility, whereas an additional role in motility is conferred by Myosin C. Furthermore, the BC acts on the assembly and/or function of the intravacuolar network, which may directly or indirectly contribute to the establishment of chronic tissue cysts. Here we provide experimental support for molecules acting in several of these processes and identify several new BC proteins critical to maintaining the cytoplasmic bridge between divided parasites. However, the dispensable nature of many BC components leaves many questions unanswered regarding its function. In conclusion, the BC in T. gondii is a dynamic and multifunctional structure at the posterior end of the parasite.
DOI: 10.7554/elife.74535
2022
Cited 11 times
Phosphoregulation accommodates Type III secretion and assembly of a tether of ER-Chlamydia inclusion membrane contact sites
Membrane contact sites (MCS) are crucial for nonvesicular trafficking-based interorganelle communication. Endoplasmic reticulum (ER)–organelle tethering occurs in part through the interaction of the ER resident protein VAP with FFAT motif-containing proteins. FFAT motifs are characterized by a seven amino acidic core surrounded by acid tracks. We have previously shown that the human intracellular bacterial pathogen Chlamydia trachomatis establishes MCS between its vacuole (the inclusion) and the ER through expression of a bacterial tether, IncV, displaying molecular mimicry of eukaryotic FFAT motif cores. Here, we show that multiple layers of host cell kinase-mediated phosphorylation events govern the assembly of the IncV–VAP tethering complex and the formation of ER-Inclusion MCS. Via a C-terminal region containing three CK2 phosphorylation motifs, IncV recruits CK2 to the inclusion leading to IncV hyperphosphorylation of the noncanonical FFAT motif core and serine-rich tracts immediately upstream of IncV FFAT motif cores. Phosphorylatable serine tracts, rather than genetically encoded acidic tracts, accommodate Type III-mediated translocation of IncV to the inclusion membrane, while achieving full mimicry of FFAT motifs. Thus, regulatory components and post-translational modifications are integral to MCS biology, and intracellular pathogens such as C. trachomatis have evolved complex molecular mimicry of these eukaryotic features.
DOI: 10.1242/jcs.260159
2023
Cited 4 times
<i>Toxoplasma gondii</i>scavenges mammalian host organelles through the usurpation of host ESCRT-III and Vps4A
ABSTRACT Intracellular pathogens exploit cellular resources through host cell manipulation. Within its nonfusogenic parasitophorous vacuole (PV), Toxoplasma gondii targets host nutrient-filled organelles and sequesters them into the PV through deep invaginations of the PV membrane (PVM) that ultimately detach from this membrane. Some of these invaginations are generated by an intravacuolar network (IVN) of parasite-derived tubules attached to the PVM. Here, we examined the usurpation of host ESCRT-III and Vps4A by the parasite to create PVM buds and vesicles. CHMP4B associated with the PVM/IVN, and dominant-negative (DN) CHMP4B formed many long PVM invaginations containing CHMP4B filaments. These invaginations were shorter in IVN-deficient parasites, suggesting cooperation between the IVN and ESCRT. In infected cells expressing Vps4A-DN, enlarged intra-PV structures containing host endolysosomes accumulated, reflecting defects in PVM scission. Parasite mutants lacking T. gondii (Tg)GRA14 or TgGRA64, which interact with ESCRT, reduced CHMP4B-DN-induced PVM invaginations and intra-PV host organelles, with greater defects in a double knockout, revealing the exploitation of ESCRT to scavenge host organelles by Toxoplasma.
DOI: 10.1146/annurev.micro.54.1.129
2000
Cited 57 times
The Adaptative Mechanisms of<i>Trypanosoma Brucei</i>for Sterol Homeostasis in Its Different Life-Cycle Environments
Bloodstream forms of Trypanosoma brucei do not synthesize sterols de novo and therefore cannot survive in medium devoid of lipoproteins. Growth of parasites is essentially supported by receptor-mediated endocytosis of low-density lipoproteins (LDLs), which carry phospholipids and cholesteryl esters. These lipids are released from internalized LDL after apoprotein B-100 is degraded by acidic thiol-proteases in the endolysosomal apparatus and then metabolized, as in mammalian cells. The LDL receptor is recycled and its expression is regulated by the sterol stores. Documented pharmacological and immunological interferences with LDL receptor-mediated lipid supply to the bloodstream forms are summarized, and the potential for new approaches to fight against these parasites is evaluated. In contrast to bloodstream forms, cultured procyclic forms can acquire sterols from both exogenous (lipoprotein endocytosis) and endogenous (biosynthesis of ergosterol) sources. The rate-limiting steps of both endocytosis (surface LDL receptor expression) and biosynthesis (3-hydroxy-3-methylglutaryl coenzyme A reductase activity) are regulated by the cellular content of sterol. These two pathways thus complement each other to yield a balanced sterol supply, which demonstrates adaptative capacities to survive in totally different environments and fine regulatory mechanisms of sterol homeostasis.
DOI: 10.1242/jcs.00382
2003
Cited 52 times
Pleiotropic effect due to targeted depletion of secretory rhoptry protein ROP2 in<i>Toxoplasma gondii</i>
Long after their discovery, the function and biogenesis of rhoptries remain enigmatic. In Apicomplexan parasites, these organelles discharge and their contents are exocytosed at the time of host cell invasion, and are thus proposed to play an essential role in establishing the parasitophorous vacuole. In Toxoplasma gondii, ROP2 is suspected to serve as the molecular link between host cell mitochondria and parasitophorous vacuole membrane. In this study we addressed the function of ROP2. Targeted depletion of ROP2 using a ribozyme-modified antisense RNA strategy resulted in multiple effects on parasite morphology because of a disruption in the formation of mature rhoptries, and an arrest in cytokinesis. The association of host cell mitochondria with the parasitophorous vacuole membrane was abolished and the ROP2-deficient parasites had a reduced uptake of sterol from the host cell. Furthermore, these parasites invaded human fibroblasts poorly and had markedly attenuated virulence in mice. We conclude that rhoptry discharge, and in particular release of ROP2, are essential for parasite invasion, replication and host cell-parasite interaction.
DOI: 10.1016/0166-6851(95)00114-g
1995
Cited 51 times
Exogenous and endogenous sources of sterols in the culture-adapted procyclic trypomastigotes of Trypanosoma brucei
The growth of the culture-adapted procyclic forms of Trypanosoma brucei (procyclics) is accelerated by supplementation of the medium with low-density lipoprotein (LDL) particles. This effect can be attributed to receptor-mediated endocytosis of LDL, followed by utilization of lipids carried by the lipoproteins. Indeed, procyclics that normally contain ergosterol synthesized de novo, also incorporate exogenous cholesterol in their membranes. In turn, import of exogenous lipids down-regulates the isoprenoid biosynthetic machinery as measured by a approx. 3-fold decrease of [14C]acetate incorporation into sterols and a approx. 2-fold decrease of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase activity, compared with cells grown in lipoprotein-depleted medium. Synvinolin, a specific inhibitor of HMG-CoA reductase that slows down the procyclic growth in vitro and decreases [14C]acetate incorporation into sterols, produces striking morphological modifications, including an arrest at cytokinesis and an extensive swelling of the kinetoplast-mitochondrion system. These cytotoxic effects are amplified in the absence of lipoprotein supply. In conclusion, procyclics may acquire sterols from both exogenous and endogenous sources. To a large extent, these two pathways compensate each other, illustrating adaptation of the parasites to survive in extremely different environments.
DOI: 10.1111/j.1600-0854.2005.00348.x
2005
Cited 48 times
Peculiarities of Host Cholesterol Transport to the Unique Intracellular Vacuole Containing <i>Toxoplasma</i>
The intracellular protozoan Toxoplasma gondii is auxotrophic for low‐density lipoprotein (LDL)‐derived cholesterol (C). We previously showed that T . gondii scavenges this essential lipid from host endolysosomal compartments and that C delivery to the parasitophorous vacuole (PV) does not require transit through host Golgi or endoplasmic reticulum. In this study, we explore the itinerary of C from the host endolysosomes to the PV. Labeled C incorporated into LDL is rapidly detected in intravacuolar parasites and partially esterified by the parasites. In contrast to diverse mammalian organelles, the post‐endolysosomal transfer of C to the PV does not involve the host plasma membrane as an intermediate. Nevertheless, the PV membrane is accessible to extracellular sterol acceptors, suggesting C trafficking from intracellular parasites to host plasma membrane. C movement to the PV requires temperatures permissive for vesicular transport, metabolic energy and functional microtubules. Host caveolae vesicles and the sterol carrier protein‐2 do not participate in this process. Proteolytic treatment of purified PV or free parasites abolishes C acquisition by the parasites. Altogether, these results support a vesicular transport system from host endolysosomes to the PV, and a requirement for PV membrane and parasite plasma membrane proteins in C delivery to T. gondii .
DOI: 10.1242/jcs.01237
2004
Cited 45 times
The <i>Plasmodium falciparum Vps4</i> homolog mediates multivesicular body formation
Members of the apicomplexan family of parasites contain morphologically unique secretory organelles termed rhoptries that are essential for host cell invasion. Rhoptries contain internal membranes, and thus resemble multivesicular bodies. To determine whether multivesicular body endosomal intermediates are formed in Apicomplexa, we used the Plasmodium falciparum homolog of the class E gene, Vps4, as a probe. Endogenous P. falciparum Vps4 (PfVps4) localized to the cytoplasm of P. falciparum trophozoites, and transgenic PfVps4 localized to the cytosol in P. falciparum, in the related parasite Toxoplasma gondii and in COS cells. When mutated to block ATP hydrolysis, transiently expressed PfVps4 localized instead to large vesicular structures in P. falciparum. The same construct, and another mutant blocked in ATP binding, generated large cholesterol-enriched multivesicular bodies in both COS cells and T. gondii. Mutant PfVps4 structures in T. gondii co-localized with markers for early endosomes. These results demonstrate a conservation of Vps4 function across wide phylogenetic boundaries, and indicate that endosomal multivesicular bodies form in both P. falciparum and T. gondii.