Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 379, 2024 Jan 09.
Article in English | MEDLINE | ID: mdl-38191574

ABSTRACT

In Apicomplexa, rhoptry discharge is essential for invasion and involves an apical vesicle (AV) docking one or two rhoptries to a macromolecular secretory apparatus. Toxoplasma gondii is armed with 10-12 rhoptries and 5-6 microtubule-associated vesicles (MVs) presumably for iterative rhoptry discharge. Here, we have addressed the localization and functional significance of two intraconoidal microtubule (ICMT)-associated proteins instrumental for invasion. Mechanistically, depletion of ICMAP2 leads to a dissociation of the ICMTs, their detachment from the conoid and dispersion of MVs and rhoptries. ICMAP3 exists in two isoforms that contribute to the control of the ICMTs length and the docking of the two rhoptries at the AV, respectively. This study illuminates the central role ICMTs play in scaffolding the discharge of multiple rhoptries. This process is instrumental for virulence in the mouse model of infection and in addition promotes sterile protection against T. gondii via the release of key effectors inducing immunity.


Subject(s)
Toxoplasma , Animals , Mice , Microtubule-Associated Proteins , Cytoskeleton , Microtubules , Biological Transport
2.
Article in English | MEDLINE | ID: mdl-37004488

ABSTRACT

Toxoplasma gondii is a pervasive apicomplexan parasite that can cause severe disease and death in immunocompromised individuals and the developing foetus. The treatment of toxoplasmosis often leads to serious side effects and novel drugs and drug targets are therefore actively sought. In 2014, Mageed and colleagues suggested that the T. gondii pantothenate synthetase, the enzyme responsible for the synthesis of the vitamin B5 (pantothenate), the precursor of the important cofactor, coenzyme A, is a good drug target. Their conclusion was based on the ability of potent inhibitors of the M. tuberculosis pantothenate synthetase to inhibit the proliferation of T. gondii tachyzoites. They also reported that the inhibitory effect of the compounds could be antagonised by supplementing the medium with pantothenate, supporting their conclusion that the compounds were acting on the intended target. Contrary to these observations, we find that compound SW314, one of the compounds used in the Mageed et al. study and previously shown to be active against M. tuberculosis pantothenate synthetase in vitro, is inactive against the T. gondii pantothenate synthetase and does not inhibit tachyzoite proliferation, despite gaining access into the parasite in situ. Furthermore, we validate the recent observation that the pantothenate synthetase gene in T. gondii can be disrupted without detrimental effect to the survival of the tachyzoite-stage parasite in the presence or absence of extracellular pantothenate. We conclude that the T. gondii pantothenate synthetase is not essential during the tachyzoite stage of the parasite and it is therefore not a target for drug discovery against T. gondii tachyzoites.


Subject(s)
Parasites , Toxoplasma , Toxoplasmosis , Tuberculosis , Humans , Animals , Toxoplasma/genetics , Toxoplasmosis/drug therapy , Coenzyme A
3.
Anal Chem ; 95(2): 668-676, 2023 01 17.
Article in English | MEDLINE | ID: mdl-36548400

ABSTRACT

It is estimated that more than 2 billion people are chronically infected with the intracellular protozoan parasite Toxoplasma gondii (T. gondii). Despite this, there is currently no vaccine to prevent infection in humans, and there is no recognized curative treatment to clear tissue cysts. A major hurdle for identifying effective drug candidates against chronic-stage cysts has been the low throughput of existing in vitro assays for testing the survival of bradyzoites. We have developed a luciferase-based platform for specifically determining bradyzoite survival within in vitro cysts in a 96-well plate format. We engineered a cystogenic type II T. gondii PruΔku80Δhxgpr strain for stage-specific expression of firefly luciferase in the cytosol of bradyzoites and nanoluciferase for secretion into the lumen of the cyst (DuaLuc strain). Using this DuaLuc strain, we found that the ratio of firefly luciferase to nanoluciferase decreased upon treatment with atovaquone or LHVS, two compounds that are known to compromise bradyzoite viability. The 96-well format allowed us to test several additional compounds and generate dose-response curves for calculation of EC50 values indicating relative effectiveness of a compound. Accordingly, this DuaLuc system should be suitable for screening libraries of diverse compounds and defining the potency of hits or other compounds with a putative antibradyzoite activity.


Subject(s)
Toxoplasma , Humans , Luciferases, Firefly/genetics , Luciferases, Firefly/metabolism , Atovaquone/metabolism , Atovaquone/pharmacology , Luciferases/genetics , Luciferases/metabolism
4.
Mol Microbiol ; 118(6): 601-622, 2022 12.
Article in English | MEDLINE | ID: mdl-36210525

ABSTRACT

Virulence and persistence of the obligate intracellular parasite Toxoplasma gondii involve the secretion of effector proteins belonging to the family of dense granule proteins (GRAs) that act notably as modulators of the host defense mechanisms and participate in cyst wall formation. The subset of GRAs residing in the parasitophorous vacuole (PV) or exported into the host cell, undergo proteolytic cleavage in the Golgi upon the action of the aspartyl protease 5 (ASP5). In tachyzoites, ASP5 substrates play central roles in the morphology of the PV and the export of effectors across the translocon complex MYR1/2/3. Here, we used N-terminal amine isotopic labeling of substrates to identify novel ASP5 cleavage products by comparing the N-terminome of wild-type and Δasp5 lines in tachyzoites and bradyzoites. Validated substrates reside within the PV or PVM in an ASP5-dependent manner. Remarkably, Δasp5 bradyzoites are impaired in the formation of the cyst wall in vitro and exhibit a considerably reduced cyst burden in chronically infected animals. More specifically two-photon serial tomography of infected mouse brains revealed a comparatively reduced number and size of the cysts throughout the establishment of persistence in the absence of ASP5.


Subject(s)
Aspartic Acid Proteases , Toxoplasma , Animals , Mice , Toxoplasma/metabolism , Aspartic Acid Proteases/metabolism , Protozoan Proteins/metabolism , Persistent Infection , Vacuoles/metabolism , Aspartic Acid Endopeptidases/metabolism
5.
mBio ; 13(5): e0196522, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36200777

ABSTRACT

Toxoplasma gondii is an intracellular apicomplexan parasite that relies on cyclic GMP (cGMP)-dependent signaling to trigger timely egress from host cells in response to extrinsic and intrinsic signals. A guanylate cyclase (GC) complex, conserved across the Apicomplexa, plays a pivotal role in integrating these signals, such as the key lipid mediator phosphatidic acid and changes in pH and ionic composition. This complex is composed of an atypical GC fused to a flippase-like P4-ATPase domain and assembled with the cell division control protein CDC50.1 and a unique GC organizer (UGO). While the dissemination of the fast-replicating tachyzoites responsible for acute infection is well understood, it is less clear if the cyst-forming bradyzoites can disseminate and contribute to cyst burden. Here, we characterized a novel component of the GC complex recently termed signaling linking factor (SLF). Tachyzoites conditionally depleted in SLF are impaired in microneme exocytosis, conoid extrusion, and motility and hence unable to invade and egress. A stage-specific promoter swap strategy allowed the generation of SLF- and GC-deficient bradyzoites that are viable as tachyzoites but show a reduction in cyst burden during the onset of chronic infection. Upon oral infection, SLF-deficient cysts failed to establish infection in mice, suggesting SLF's importance for the natural route of T. gondii infection. IMPORTANCE Toxoplasma gondii is an obligate intracellular parasite of the phylum Apicomplexa. This life-threatening opportunistic pathogen establishes a chronic infection in human and animals that is resistant to immune attacks and chemotherapeutic intervention. The slow-growing parasites persist in tissue cysts that constitute a predominant source of transmission. Host cell invasion and egress are two critical steps of the parasite lytic cycle that are governed by a guanylate cyclase complex conserved across the Apicomplexa. A signaling linked factor is characterized here as an additional component of the complex that not only is essential during acute infection but also plays a pivotal role during natural oral infection with tissue cysts' dissemination and persistence.


Subject(s)
Toxoplasma , Animals , Humans , Mice , Toxoplasma/metabolism , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Persistent Infection , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Cyclic GMP/metabolism , Phosphatidic Acids/metabolism , Adenosine Triphosphatases/metabolism
6.
Front Microbiol ; 13: 899243, 2022.
Article in English | MEDLINE | ID: mdl-35756016

ABSTRACT

To gain access to the intracellular cytoplasmic niche essential for their growth and replication, apicomplexan parasites such as Toxoplasma gondii rely on the timely secretion of two types of apical organelles named micronemes and rhoptries. Rhoptry proteins are key to host cell invasion and remodeling, however, the molecular mechanisms underlying the tight control of rhoptry discharge are poorly understood. Here, we report the identification and functional characterization of two novel T. gondii thrombospondin-related proteins implicated in rhoptry exocytosis. The two proteins, already annotated as MIC15 and MIC14, were renamed rhoptry discharge factor 1 (RDF1) and rhoptry discharge factor 2 (RDF2) and found to be exclusive of the Coccidia class of apicomplexan parasites. Furthermore, they were shown to have a paralogous relationship and share a C-terminal transmembrane domain followed by a short cytoplasmic tail. Immunofluorescence analysis of T. gondii tachyzoites revealed that RDF1 presents a diffuse punctate localization not reminiscent of any know subcellular compartment, whereas RDF2 was not detected. Using a conditional knockdown approach, we demonstrated that RDF1 loss caused a marked growth defect. The lack of the protein did not affect parasite gliding motility, host cell attachment, replication and egress, whereas invasion was dramatically reduced. Notably, while RDF1 depletion did not result in altered microneme exocytosis, rhoptry discharge was found to be heavily impaired. Interestingly, rhoptry secretion was reversed by spontaneous upregulation of the RDF2 gene in knockdown parasites grown under constant RDF1 repression. Collectively, our results identify RDF1 and RDF2 as additional key players in the pathway controlling rhoptry discharge. Furthermore, this study unveils a new example of compensatory mechanism contributing to phenotypic plasticity in T. gondii.

7.
Nat Commun ; 13(1): 345, 2022 01 17.
Article in English | MEDLINE | ID: mdl-35039477

ABSTRACT

Coenzyme A (CoA) is an essential molecule acting in metabolism, post-translational modification, and regulation of gene expression. While all organisms synthesize CoA, many, including humans, are unable to produce its precursor, pantothenate. Intriguingly, like most plants, fungi and bacteria, parasites of the coccidian subgroup of Apicomplexa, including the human pathogen Toxoplasma gondii, possess all the enzymes required for de novo synthesis of pantothenate. Here, the importance of CoA and pantothenate biosynthesis for the acute and chronic stages of T. gondii infection is dissected through genetic, biochemical and metabolomic approaches, revealing that CoA synthesis is essential for T. gondii tachyzoites, due to the parasite's inability to salvage CoA or intermediates of the pathway. In contrast, pantothenate synthesis is only partially active in T. gondii tachyzoites, making the parasite reliant on its uptake. However, pantothenate synthesis is crucial for the establishment of chronic infection, offering a promising target for intervention against the persistent stage of T. gondii.


Subject(s)
Pantothenic Acid/biosynthesis , Parasites/pathogenicity , Persistent Infection/parasitology , Toxoplasma/pathogenicity , Toxoplasmosis/parasitology , Animals , Biosynthetic Pathways , Cell Differentiation , Cell Membrane/metabolism , Coenzyme A/biosynthesis , Coenzyme A/chemistry , Coenzyme A/metabolism , Cytoplasm/metabolism , Female , Life Cycle Stages , Mice , Pantothenic Acid/chemistry , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Multimerization , Toxoplasma/growth & development
8.
PLoS Pathog ; 17(12): e1010124, 2021 12.
Article in English | MEDLINE | ID: mdl-34969059

ABSTRACT

The Apicomplexa phylum comprises thousands of distinct intracellular parasite species, including coccidians, haemosporidians, piroplasms, and cryptosporidia. These parasites are characterized by complex and divergent life cycles occupying a variety of host niches. Consequently, they exhibit distinct adaptations to the differences in nutritional availabilities, either relying on biosynthetic pathways or by salvaging metabolites from their host. Pantothenate (Pan, vitamin B5) is the precursor for the synthesis of an essential cofactor, coenzyme A (CoA), but among the apicomplexans, only the coccidian subgroup has the ability to synthesize Pan. While the pathway to synthesize CoA from Pan is largely conserved across all branches of life, there are differences in the redundancy of enzymes and possible alternative pathways to generate CoA from Pan. Impeding the scavenge of Pan and synthesis of Pan and CoA have been long recognized as potential targets for antimicrobial drug development, but in order to fully exploit these critical pathways, it is important to understand such differences. Recently, a potent class of pantothenamides (PanAms), Pan analogs, which target CoA-utilizing enzymes, has entered antimalarial preclinical development. The potential of PanAms to target multiple downstream pathways make them a promising compound class as broad antiparasitic drugs against other apicomplexans. In this review, we summarize the recent advances in understanding the Pan and CoA biosynthesis pathways, and the suitability of these pathways as drug targets in Apicomplexa, with a particular focus on the cyst-forming coccidian, Toxoplasma gondii, and the haemosporidian, Plasmodium falciparum.


Subject(s)
Antiparasitic Agents/pharmacology , Apicomplexa/metabolism , Apicomplexa/parasitology , Coenzyme A/biosynthesis , Pantothenic Acid/biosynthesis , Protozoan Infections , Animals , Humans
9.
Metabolites ; 11(8)2021 Jul 23.
Article in English | MEDLINE | ID: mdl-34436417

ABSTRACT

Apicomplexan parasites are responsible for devastating diseases, including malaria, toxoplasmosis, and cryptosporidiosis. Current treatments are limited by emerging resistance to, as well as the high cost and toxicity of existing drugs. As obligate intracellular parasites, apicomplexans rely on the uptake of many essential metabolites from their host. Toxoplasma gondii, the causative agent of toxoplasmosis, is auxotrophic for several metabolites, including sugars (e.g., myo-inositol), amino acids (e.g., tyrosine), lipidic compounds and lipid precursors (cholesterol, choline), vitamins, cofactors (thiamine) and others. To date, only few apicomplexan metabolite transporters have been characterized and assigned a substrate. Here, we set out to investigate whether untargeted metabolomics can be used to identify the substrate of an uncharacterized transporter. Based on existing genome- and proteome-wide datasets, we have identified an essential plasma membrane transporter of the major facilitator superfamily in T. gondii-previously termed TgApiAT6-1. Using an inducible system based on RNA degradation, TgApiAT6-1 was depleted, and the mutant parasite's metabolome was compared to that of non-depleted parasites. The most significantly reduced metabolite in parasites depleted in TgApiAT6-1 was identified as the amino acid lysine, for which T. gondii is predicted to be auxotrophic. Using stable isotope-labeled amino acids, we confirmed that TgApiAT6-1 is required for efficient lysine uptake. Our findings highlight untargeted metabolomics as a powerful tool to identify the substrate of orphan transporters.

10.
Nat Commun ; 12(1): 3788, 2021 06 18.
Article in English | MEDLINE | ID: mdl-34145271

ABSTRACT

Active host cell invasion by the obligate intracellular apicomplexan parasites relies on the formation of a moving junction, which connects parasite and host cell plasma membranes during entry. Invading Toxoplasma gondii tachyzoites secrete their rhoptry content and insert a complex of RON proteins on the cytoplasmic side of the host cell membrane providing an anchor to which the parasite tethers. Here we show that a rhoptry-resident kinase RON13 is a key virulence factor that plays a crucial role in host cell entry. Cryo-EM, kinase assays, phosphoproteomics and cellular analyses reveal that RON13 is a secretory pathway kinase of atypical structure that phosphorylates rhoptry proteins including the components of the RON complex. Ultimately, RON13 kinase activity controls host cell invasion by anchoring the moving junction at the parasite-host cell interface.


Subject(s)
Cell Membrane/parasitology , Protozoan Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Toxoplasma/metabolism , Toxoplasmosis/pathology , Biological Transport/physiology , Cells, Cultured , Host-Parasite Interactions , Humans , Secretory Pathway/physiology , Virulence Factors
12.
Cell Host Microbe ; 27(2): 290-306.e11, 2020 02 12.
Article in English | MEDLINE | ID: mdl-31991093

ABSTRACT

To survive and proliferate in diverse host environments with varying nutrient availability, the obligate intracellular parasite Toxoplasma gondii reprograms its metabolism. We have generated and curated a genome-scale metabolic model (iTgo) for the fast-replicating tachyzoite stage, harmonized with experimentally observed phenotypes. To validate the importance of four metabolic pathways predicted by the model, we have performed in-depth in vitro and in vivo phenotyping of mutant parasites including targeted metabolomics and CRISPR-Cas9 fitness screening of all known metabolic genes. This led to unexpected insights into the remarkable flexibility of the parasite, addressing the dependency on biosynthesis or salvage of fatty acids (FAs), purine nucleotides (AMP and GMP), a vitamin (pyridoxal-5P), and a cofactor (heme) in both the acute and latent stages of infection. Taken together, our experimentally validated metabolic network leads to a deeper understanding of the parasite's biology, opening avenues for the development of therapeutic intervention against apicomplexans.


Subject(s)
Fatty Acids/metabolism , Heme/metabolism , Toxoplasma/metabolism , Vitamin B 6/metabolism , Animals , Clustered Regularly Interspaced Short Palindromic Repeats , Computational Biology , Drug Development/trends , Genomics , Life Cycle Stages/physiology , Metabolic Networks and Pathways , Metabolomics , Mice , Phenotype , Toxoplasma/genetics
13.
J Biol Chem ; 295(3): 701-714, 2020 01 17.
Article in English | MEDLINE | ID: mdl-31767680

ABSTRACT

The Apicomplexa phylum comprises diverse parasitic organisms that have evolved from a free-living ancestor. These obligate intracellular parasites exhibit versatile metabolic capabilities reflecting their capacity to survive and grow in different hosts and varying niches. Determined by nutrient availability, they either use their biosynthesis machineries or largely depend on their host for metabolite acquisition. Because vitamins cannot be synthesized by the mammalian host, the enzymes required for their synthesis in apicomplexan parasites represent a large repertoire of potential therapeutic targets. Here, we review recent advances in metabolic reconstruction and functional studies coupled to metabolomics that unravel the interplay between biosynthesis and salvage of vitamins and cofactors in apicomplexans. A particular emphasis is placed on Toxoplasma gondii, during both its acute and latent stages of infection.


Subject(s)
Apicomplexa/metabolism , Coenzymes/metabolism , Toxoplasmosis/metabolism , Vitamins/metabolism , Apicomplexa/genetics , Coenzymes/genetics , Host-Parasite Interactions/genetics , Humans , Metabolic Networks and Pathways/genetics , Protein Biosynthesis/genetics , Toxoplasma/genetics , Toxoplasma/metabolism , Toxoplasma/pathogenicity , Toxoplasmosis/parasitology , Vitamins/genetics
14.
mBio ; 10(4)2019 08 06.
Article in English | MEDLINE | ID: mdl-31387907

ABSTRACT

Toxoplasma gondii is a ubiquitous pathogen that can cause encephalitis, congenital defects, and ocular disease. T. gondii has also been implicated as a risk factor for mental illness in humans. The parasite persists in the brain as slow-growing bradyzoites contained within intracellular cysts. No treatments exist to eliminate this form of parasite. Although proteolytic degradation within the parasite lysosome-like vacuolar compartment (VAC) is critical for bradyzoite viability, whether other aspects of the VAC are important for parasite persistence remains unknown. An ortholog of Plasmodium falciparum chloroquine resistance transporter (CRT), TgCRT, has previously been identified in T. gondii To interrogate the function of TgCRT in chronic-stage bradyzoites and its role in persistence, we knocked out TgCRT in a cystogenic strain and assessed VAC size, VAC digestion of host-derived proteins and parasite autophagosomes, and the viability of in vitro and in vivo bradyzoites. We found that whereas parasites deficient in TgCRT exhibit normal digestion within the VAC, they display a markedly distended VAC and their viability is compromised both in vitro and in vivo Interestingly, impairing VAC proteolysis in TgCRT-deficient bradyzoites restored VAC size, consistent with a role for TgCRT as a transporter of products of digestion from the VAC. In conjunction with earlier studies, our current findings suggest a functional link between TgCRT and VAC proteolysis. This study provides further evidence of a crucial role for the VAC in bradyzoite persistence and a new potential VAC target to abate chronic Toxoplasma infection.IMPORTANCE Individuals chronically infected with the intracellular parasite Toxoplasma gondii are at risk of experiencing reactivated disease that can result in progressive loss of vision. No effective treatments exist for chronic toxoplasmosis due in part to a poor understanding of the biology underlying chronic infection and a lack of well-validated potential targets. We show here that a T. gondii transporter is functionally linked to protein digestion within the parasite lysosome-like organelle and that this transporter is necessary to sustain chronic infection in culture and in experimentally infected mice. Ablating the transporter results in severe bloating of the lysosome-like organelle. Together with earlier work, this study suggests the parasite's lysosome-like organelle is vital for parasite survival, thus rendering it a potential target for diminishing infection and reducing the risk of reactivated disease.


Subject(s)
Membrane Transport Proteins/metabolism , Protozoan Proteins/metabolism , Toxoplasma/growth & development , Toxoplasma/metabolism , Toxoplasmosis/parasitology , Vacuoles/metabolism , Animals , Autophagosomes/metabolism , Cell Survival , Female , Humans , Life Cycle Stages , Lysosomes/genetics , Lysosomes/metabolism , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Proteolysis , Protozoan Proteins/genetics , Toxoplasma/genetics , Vacuoles/genetics
15.
Nat Microbiol ; 4(3): 420-428, 2019 03.
Article in English | MEDLINE | ID: mdl-30742070

ABSTRACT

Toxoplasma gondii establishes a lifelong chronic infection in humans and animals1. Host cell entry and egress are key steps in the lytic cycle of this obligate intracellular parasite, ensuring its survival and dissemination. Egress is temporally orchestrated, underpinned by the exocytosis of secretory organelles called micronemes. At any point during intracellular replication, deleterious environmental changes such as the loss of host cell integrity can trigger egress2 through the activation of the cyclic guanosine monophosphate-dependent protein kinase G3. Notably, even in the absence of extrinsic signals, the parasites egress from infected cells in a coordinated manner after five to six cycles of endodyogeny multiplication. Here we show that diacylglycerol kinase 2 is secreted into the parasitophorous vacuole, where it produces phosphatidic acid. Phosphatidic acid acts as an intrinsic signal that elicits natural egress upstream of an atypical guanylate cyclase (GC), which is uniquely conserved in alveolates4 and ciliates5, and composed of a P4-ATPase and two GC catalytic domains. Assembly of GC at the plasma membrane depends on two associated cofactors - the cell division control 50.1 and a unique GC organizer. This study reveals the existence of a signalling platform that responds to an intrinsic lipid mediator and extrinsic signals to control programmed and induced egress.


Subject(s)
Guanylate Cyclase/metabolism , Host-Parasite Interactions , Phosphatidic Acids/metabolism , Signal Transduction , Toxoplasma/growth & development , Cell Line , Fibroblasts/parasitology , Humans , Protozoan Proteins/metabolism
16.
Sci Rep ; 7(1): 13822, 2017 10 23.
Article in English | MEDLINE | ID: mdl-29062106

ABSTRACT

Toxoplasma gondii infects a broad range of hosts and can establish chronic infections with the formation of brain cysts. Infected animals show altered risk behaviour which has been suggested to increase capture probability of hosts, and thus enhance parasite transmission. It has been proposed that the ability of Toxoplasma cysts to secrete tyrosine hydroxylase could mediate these behavioural alterations. We tested the involvement of secreted tyrosine hydroxylase, coded by the parasite AaaH2 gene, in the development of alterations in mouse behaviour, by generating an AaaH2 deletion mutant parasite strain and testing its influence on behaviour. We found that both mice infected with wild type or AaaH2 mutant strains showed changes in risk behaviour. We confirmed these findings using factor analysis of the behaviour, which revealed that behavioural changes happened along a single dimension, and were observed in both infected groups. Furthermore, we developed a new behavioural paradigm in which animals are unpredictably trapped, and observed that both groups of infected animals perceive trapping but fail to adjust their behaviour to avoid further trapping. These results demonstrate that parasite-secreted AaaH2 TH is neither necessary for the generation of risky behaviour nor for the increased trappability observed during chronic Toxoplasma infection.


Subject(s)
Behavior, Animal , Brain/parasitology , Host-Parasite Interactions , Toxoplasma/enzymology , Toxoplasmosis/parasitology , Tyrosine 3-Monooxygenase/metabolism , Animals , Brain/pathology , Female , Mice , Mice, Inbred C57BL , Risk-Taking , Toxoplasma/genetics , Tyrosine 3-Monooxygenase/genetics
17.
Nat Microbiol ; 2: 17096, 2017 Jun 19.
Article in English | MEDLINE | ID: mdl-28628099

ABSTRACT

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.


Subject(s)
Autophagosomes/metabolism , Host-Pathogen Interactions , Lysosomes/metabolism , Toxoplasma/physiology , Animals , Cell Survival , Cells, Cultured , Cysteine Proteases/genetics , Cysteine Proteases/metabolism , Fibroblasts/parasitology , Gene Knockout Techniques , Humans , Mice, Inbred C57BL , Neurons/parasitology , Proteolysis , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Toxoplasma/enzymology , Toxoplasma/metabolism
18.
Curr Genet ; 62(1): 31-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26194054

ABSTRACT

Apicomplexan parasites including Toxoplasma gondii and Plasmodium species have complex life cycles that include multiple hosts and differentiation through several morphologically distinct stages requiring marked changes in gene expression. This review highlights emerging evidence implicating regulation of mRNA splicing as a mechanism to prime these parasites for rapid gene expression upon differentiation. We summarize the most important insights in alternative splicing including its role in regulating gene expression by decreasing mRNA abundance via 'Regulated Unproductive Splicing and Translation'. As a related but less well-understood mechanism, we discuss also our recent work suggesting a role for intron retention for precluding translation of stage specific isoforms of T. gondii glycolytic enzymes. We additionally provide new evidence that intron retention might be a widespread mechanism during parasite differentiation. Supporting this notion, recent genome-wide analysis of Toxoplasma and Plasmodium suggests intron retention is more pervasive than heretofore thought. These findings parallel recent emergence of intron retention being more prevalent in mammals than previously believed, thereby adding to the established roles in plants, fungi and unicellular eukaryotes. Deeper mechanistic studies of intron retention will provide important insight into its role in regulating gene expression in apicomplexan parasites and more general in eukaryotic organisms.


Subject(s)
Alternative Splicing , Gene Expression Regulation , RNA Processing, Post-Transcriptional , Animals , Genomics , Humans , Introns , Parasites/genetics , Parasites/metabolism , Protein Biosynthesis , Proteome , Toxoplasma/genetics , Toxoplasma/metabolism
19.
PLoS Pathog ; 11(10): e1005211, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26473595

ABSTRACT

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.


Subject(s)
Aspartic Acid Proteases/metabolism , Golgi Apparatus/enzymology , Host-Parasite Interactions/physiology , Toxoplasma/pathogenicity , Toxoplasmosis/enzymology , Animals , Blotting, Western , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Gene Knockout Techniques , Humans , Mice , Mice, Inbred C57BL , Microscopy, Electron, Transmission , Molecular Sequence Data , Protein Transport , Real-Time Polymerase Chain Reaction , Toxoplasma/enzymology , Transfection
20.
Mol Microbiol ; 96(6): 1159-75, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25777509

ABSTRACT

The intracellular parasite Toxoplasma gondii converts from a rapidly replicating tachyzoite form during acute infection to a quiescent encysted bradyzoite stage that persists inside long-lived cells during chronic infection. Bradyzoites adopt reduced metabolism and slow replication while waiting for an opportunity to recrudesce the infection within the host. Interconversion between these two developmental stages is characterized by expression of glycolytic isoenzymes that play key roles in parasite metabolism. The parasite genome encodes two isoforms of lactate dehydrogenase (LDH1 and LDH2) and enolase (ENO1 and ENO2) that are expressed in a stage-specific manner. Expression of different isoforms of these enzymes allows T. gondii to rapidly adapt to diverse metabolic requirements necessary for either a rapid replication of the tachyzoite stage or a quiescent lifestyle typical of the bradyzoites. Herein we identified unspliced forms of LDH and ENO transcripts produced during transition between these two parasite stages suggestive of an intron retention mechanism to promptly exchange glycolytic isoforms for rapid adaptation to environmental changes. We also identified key regulatory elements in the ENO transcription units, revealing cooperation between the ENO2 5'-untranslated region and the ENO2 intron, along with identifying a role for the ENO1 3'-untranslated region in stage-specific expression.


Subject(s)
Introns , L-Lactate Dehydrogenase/biosynthesis , Phosphopyruvate Hydratase/biosynthesis , Toxoplasma/enzymology , Toxoplasma/genetics , Animals , Gene Expression Regulation, Bacterial , Gene Expression Regulation, Developmental , Genes, Protozoan , Glycolysis , Humans , Isoenzymes , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Mice , Phosphopyruvate Hydratase/genetics , Phosphopyruvate Hydratase/metabolism , Toxoplasma/growth & development , Toxoplasmosis, Animal/parasitology
SELECTION OF CITATIONS
SEARCH DETAIL
...