Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 556
Filter
1.
Parasite ; 31: 23, 2024.
Article in English | MEDLINE | ID: mdl-38759153

ABSTRACT

Eimeria tenella is an obligate intracellular parasite which causes great harm to the poultry breeding industry. Protein phosphorylation plays a vital role in host cell-E. tenella interactions. However, no comprehensive phosphoproteomic analyses of host cells at various phases of E. tenella infection have been published. In this study, quantitative phosphoproteomic analysis of chicken embryo DF-1 fibroblasts that were uninfected (UI) or infected with E. tenella for 6 h (PI6, the early invasion phase) or 36 h (PI36, the trophozoite development phase) was conducted. A total of 10,122 phosphopeptides matched to 3,398 host cell phosphoproteins were identified and 13,437 phosphorylation sites were identified. Of these, 491, 1,253, and 275 differentially expressed phosphorylated proteins were identified in the PI6/UI, PI36/UI, and PI36/PI6 comparisons, respectively. KEGG pathway enrichment analysis showed that E. tenella modulated host cell processes through phosphorylation, including focal adhesion, regulation of the actin cytoskeleton, and FoxO signaling to support its early invasion phase, and modulating adherens junctions and the ErbB signaling pathway to favor its trophozoite development. These results enrich the data on the interaction between E. tenella and host cells and facilitate a better understanding of the molecular mechanisms underlying host-parasite relationships.


Title: Analyse phosphoprotéomique quantitative de cellules DF-1 de poulet infectées par Eimeria tenella, par spectrométrie de masse avec marqueur de masse en tandem (TMT) et surveillance des réactions parallèles (PRM). Abstract: Eimeria tenella est un parasite intracellulaire obligatoire qui cause de graves dommages à l'industrie de l'élevage de volailles. La phosphorylation des protéines joue un rôle essentiel dans les interactions entre la cellule hôte et E. tenella. Cependant, aucune analyse phosphoprotéomique complète des cellules hôtes à différentes phases de l'infection par E. tenella n'a été publiée. Dans cette étude, une analyse phosphoprotéomique quantitative de fibroblastes DF-1 d'embryon de poulet non infectés (NI) ou infectés par E. tenella pendant 6 h (PI6, la phase d'invasion précoce) ou 36 h (PI36, la phase de développement des trophozoïtes) a été réalisée. Un total de 10 122 phosphopeptides correspondant à 3 398 phosphoprotéines de cellules hôtes ont été identifiés et 13 437 sites de phosphorylation ont été identifiés. Parmi celles-ci, 491, 1 253 et 275 protéines différentiellement phosphorylées exprimées ont été identifiées respectivement dans les comparaisons PI6/NI, PI36/NI et PI36/PI6. L'analyse d'enrichissement de la voie KEGG a montré qu'E. tenella modulait les processus de la cellule hôte par phosphorylation, y compris l'adhésion focale, la régulation du cytosquelette d'actine et la signalisation FoxO, pour aider sa phase d'invasion précoce, et la modulation des jonctions adhérentes et de la voie de signalisation ErbB pour favoriser le développement de son trophozoïte. Ces résultats enrichissent les données sur l'interaction entre E. tenella et les cellules hôtes et facilitent une meilleure compréhension des mécanismes moléculaires sous-jacents aux relations hôtes­parasites.


Subject(s)
Chickens , Eimeria tenella , Fibroblasts , Phosphoproteins , Proteomics , Tandem Mass Spectrometry , Animals , Eimeria tenella/physiology , Chickens/parasitology , Proteomics/methods , Phosphoproteins/analysis , Phosphoproteins/metabolism , Phosphorylation , Fibroblasts/parasitology , Cell Line , Poultry Diseases/parasitology , Host-Parasite Interactions , Coccidiosis/parasitology , Coccidiosis/veterinary , Chick Embryo , Signal Transduction
2.
J Biol Chem ; 300(1): 105582, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38141762

ABSTRACT

The intracellular parasite, Toxoplasma gondii, has developed sophisticated molecular strategies to subvert host processes and promote growth and survival. During infection, T. gondii replicates in a parasitophorous vacuole (PV) and modulates host functions through a network of secreted proteins. Of these, Mitochondrial Association Factor 1b (MAF1b) recruits host mitochondria to the PV, a process that confers an in vivo growth advantage, though the precise mechanisms remain enigmatic. To address this knowledge gap, we mapped the MAF1b interactome in human fibroblasts using a commercial Yeast-2-hybrid (Y2H) screen, which revealed several previously unidentified binding partners including the GAP domain of Ral GTPase Accelerating Protein α1 (RalGAPα1(GAP)). Recombinantly produced MAF1b and RalGAPα1(GAP) formed as a stable binary complex as shown by size exclusion chromatography with a Kd of 334 nM as measured by isothermal titration calorimetry (ITC). Notably, no binding was detected between RalGAPα1(GAP) and the structurally conserved MAF1b homolog, MAF1a, which does not recruit host mitochondria. Next, we used hydrogen deuterium exchange mass spectrometry (HDX-MS) to map the RalGAPα1(GAP)-MAF1b interface, which led to identification of the "GAP-binding loop" on MAF1b that was confirmed by mutagenesis and ITC to be necessary for complex formation. A high-confidence Alphafold model predicts the GAP-binding loop to lie at the RalGAPα1(GAP)-MAF1b interface further supporting the HDX-MS data. Mechanistic implications of a RalGAPα1(GAP)-MAF1b complex are discussed in the context of T. gondii infection and indicates that MAF1b may have evolved multiple independent functions to increase T. gondii fitness.


Subject(s)
GTPase-Activating Proteins , Mitochondria , Protein Interaction Maps , Protozoan Proteins , Toxoplasma , Humans , Binding Sites , Calorimetry , Chromatography, Gel , Fibroblasts/metabolism , Fibroblasts/parasitology , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Hydrogen Deuterium Exchange-Mass Spectrometry , Mitochondria/metabolism , Mitochondria/parasitology , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Toxoplasma/chemistry , Toxoplasma/genetics , Toxoplasma/metabolism , Two-Hybrid System Techniques
3.
Open Biol ; 11(10): 210053, 2021 10.
Article in English | MEDLINE | ID: mdl-34610266

ABSTRACT

Toxoplasma gondii is a eukaryotic parasite that forms latent cysts in the brain of immunocompetent individuals. The latent parasite infection of the immune-privileged central nervous system is linked to most complications. With no drug currently available to eliminate the latent cysts in the brain of infected hosts, the consequences of neurons' long-term infection are unknown. It has long been known that T. gondii specifically differentiates into a latent form (bradyzoite) in neurons, but how the infected neuron responds to the infection remains to be elucidated. We have established a new in vitro model resulting in the production of mature bradyzoite cysts in brain cells. Using dual, host and parasite RNA-seq, we characterized the dynamics of differentiation of the parasite, revealing the involvement of key pathways in this process. Moreover, we identified how the infected brain cells responded to the parasite infection revealing the drastic changes that take place. We showed that neuronal-specific pathways are strongly affected, with synapse signalling being particularly affected, especially glutamatergic synapse signalling. The establishment of this new in vitro model allows investigating both the dynamics of parasite differentiation and the specific response of neurons to long-term infection by this parasite.


Subject(s)
Foreskin/cytology , Gene Expression Profiling/methods , Gene Regulatory Networks , Neurons/cytology , Protozoan Proteins/genetics , Toxoplasma/pathogenicity , Toxoplasmosis, Cerebral/pathology , Animals , Cells, Cultured , Disease Models, Animal , Fibroblasts/cytology , Fibroblasts/parasitology , Foreskin/parasitology , High-Throughput Nucleotide Sequencing , Humans , Male , Mice , Neurons/parasitology , Primary Cell Culture , Rats , Sequence Analysis, RNA , Toxoplasma/genetics , Toxoplasmosis, Cerebral/genetics
4.
Parasit Vectors ; 14(1): 450, 2021 Sep 06.
Article in English | MEDLINE | ID: mdl-34488852

ABSTRACT

BACKGROUND: Alveolar echinococcosis (AE) is a chronic zoonosis caused by the larval form of Echinococcus multilocularis (E. multilocularis). Current chemotherapy against AE has relied on albendazole and mebendazole, which only exhibit parasitostatic and not parasiticidal efficacy. Therefore, novel compounds for the treatment of this disease are needed. METHODS: Phosphoglucose isomerase (PGI) assays were used for compound screening of seven neonicotinoids. The anti-parasitic effects of thiacloprid were then evaluated on E. multilocularis metacestode vesicles, germinal cells and protoscoleces in vitro. Human foreskin fibroblasts (HFF) and Reuber rat hepatoma (RH) cells were used to assess cytotoxicity. Glucose consumption in E. multilocularis protoscoleces and germinal cells was assessed by measuring uptake of 2-deoxyglucose (2-DG). Molecular docking was used to evaluate the potential binding sites of thiacloprid to acetylcholine receptors. In vivo efficacy of thiacloprid was evaluated in mice by secondary infection with E. multilocularis. In addition, ELISA and flow cytometry were used to evaluate the effects of cytokines and T lymphocyte subsets after thiacloprid treatment. Furthermore, collagen deposition and degradation in the host lesion microenvironment were evaluated. RESULTS: We found that thiacloprid is the most promising compound, with an IC50 of 4.54 ± 1.10 µM and 2.89 ± 0.34 µM, respectively, against in vitro-cultured E. multilocularis metacestodes and germinal cells. Thiacloprid was less toxic for HFF and RH mammalian cell lines than for metacestodes. In addition, thiacloprid inhibited the acetylcholinesterase activity in protoscoleces, metacestodes and germinal cells. Thiacloprid inhibited glucose consumption by protoscoleces and germinal cells. Subsequently, transmission electron microscopy revealed that treatment with thiacloprid damaged the germinal layer. In vivo, metacestode weight was significantly reduced following oral administration of thiacloprid at 15 and 30 mg/kg. The level of CD4+ T lymphocytes in metacestodes and spleen increased after thiacloprid treatment. Anti-echinococcosis-related cytokines (IL-2, IL-4, IL-10) were significantly increased. Furthermore, thiacloprid inhibited the expression of matrix metalloproteinases (MMPs 1, 3, 9, 13) and promoted collagen deposition in the host lesion microenvironment. CONCLUSIONS: The results demonstrated that thiacloprid had parasiticidal activity against E. multilocularis in vitro and in vivo, and could be used as a novel lead compound for the treatment of AE.


Subject(s)
Anthelmintics/pharmacology , Anthelmintics/therapeutic use , Echinococcus multilocularis/drug effects , Neonicotinoids/pharmacology , Neonicotinoids/therapeutic use , Thiazines/pharmacology , Thiazines/therapeutic use , Animals , Anthelmintics/metabolism , Echinococcosis/drug therapy , Female , Fibroblasts/drug effects , Fibroblasts/parasitology , Foreskin/cytology , Humans , Inhibitory Concentration 50 , Male , Mice, Inbred BALB C , Molecular Docking Simulation , Neonicotinoids/metabolism , Receptors, Cholinergic/metabolism , Specific Pathogen-Free Organisms , Thiazines/metabolism
5.
Biol Open ; 10(9)2021 09 15.
Article in English | MEDLINE | ID: mdl-34590698

ABSTRACT

Most intracellular pathogens replicate in a vacuole to avoid the defense system of the host. A few pathogens recruit host mitochondria around those vacuoles, but the molecules responsible for mitochondrial recruitment remain unidentified. It is only in the apicomplexan parasite Toxoplasma gondii, that mitochondrial association factor 1b (MAF1b) has been identified as an association factor for host mitochondria. Here, we show that rhoptry kinase family protein 39 (ROP39) induces host mitochondrial recruitment in T. gondii. We found that the abundance of ROP39 was increased on host mitochondria extracted from human foreskin fibroblasts (HFFs) infected with T. gondii. ROP39 expressed exogenously in HFFs localized on host mitochondria, indicating that it has the potential to bind to host mitochondria without assistance from other parasite factors. Confocal microscopy revealed that ROP39 colocalized with host mitochondria on the membrane of parasitophorous vacuoles, in which the parasites reside. Moreover, we observed about a 10% reduction in the level of mitochondrial association in rop39-knockout parasites compared with a parental strain.


Subject(s)
Fibroblasts/parasitology , Mitochondria/parasitology , Protein Kinases/physiology , Protozoan Proteins/physiology , Toxoplasma/physiology , Vacuoles/parasitology , Host-Parasite Interactions , Humans
6.
J Med Chem ; 64(17): 12691-12704, 2021 09 09.
Article in English | MEDLINE | ID: mdl-34427442

ABSTRACT

1,2,3-Triazole is one of the most flexible chemical scaffolds broadly used in various fields. Here, we report the antileishmanial activity of 1,2,3-triazole derivatives, the ultrastructural alterations induced by their treatment, and the nitric oxide (NO) modulation effect on their efficacy against Leishmania amazonensis in vitro infection. After the screening of eleven compounds, compound 4 exhibited better results against L. amazonensis promastigotes (IC50 = 15.52 ± 3.782 µM) and intracellular amastigotes (IC50 = 4.10 ± 1.136 µM), 50% cytotoxicity concentration at 84.01 ± 3.064 µM against BALB/c peritoneal macrophages, and 20.49-fold selectivity for the parasite over the cells. Compound 4 induced ultrastructural mitochondrial alterations and lipid inclusions in L. amazonensis promastigotes, upregulated tumor necrosis factor α, interleukin (IL)-1ß, IL-6, IL-12, and IL-10 messenger RNA expressions, and enhanced the NO production, verified by nitrite (p = 0.0095) and inducible nitric oxide synthase expression (p = 0.0049) quantification, which played an important role in its activity against intramacrophagic L. amazonensis. In silico prediction in association with antileishmanial activity results showed compound 4 as a hit compound with promising potential for further studies of new leishmaniasis treatment options.


Subject(s)
Antiprotozoal Agents/pharmacology , Leishmania/drug effects , Macrophages, Peritoneal/drug effects , Macrophages, Peritoneal/parasitology , Nitric Oxide/metabolism , Triazoles/pharmacology , Animals , Antiprotozoal Agents/chemistry , Cell Line , Cell Survival/drug effects , Female , Fibroblasts/drug effects , Fibroblasts/parasitology , Gene Expression Regulation, Enzymologic/drug effects , Humans , Mice , Mice, Inbred BALB C , Molecular Structure , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/metabolism , Triazoles/chemistry
7.
Parasit Vectors ; 14(1): 400, 2021 Aug 12.
Article in English | MEDLINE | ID: mdl-34384491

ABSTRACT

BACKGROUND: Metacaspases are multifunctional proteins found in plants, fungi and protozoa, and are involved in processes such as insoluble protein aggregate clearance and cell proliferation. Our previous study demonstrated that metacaspase-1 (MCA1) contributes to parasite apoptosis in Toxoplasma gondii. Deletion of MCA1 from T. gondii has no effect on the growth and virulence of the parasites. Three metacaspases were identified in the ToxoDB Toxoplasma Informatics Resource, and the function of metacaspase-2 (MCA2) and metacaspase-3 (MCA3) has not been demonstrated. METHODS: In this study, we constructed MCA1, MCA2 and MCA1/MCA2 transgenic strains from RHΔku80 (Δku80), including overexpressing strains and knockout strains, to clarify the function of MCA1 and MCA2 of T. gondii. RESULTS: MCA1 and MCA2 were distributed in the cytoplasm with punctuated aggregation, and part of the punctuated aggregation of MCA1 and MCA2 was localized on the inner membrane complex of T. gondii. The proliferation of the MCA1/MCA2 double-knockout strain was significantly reduced; however, the two single knockout strains (MCA1 knockout strain and MCA2 knockout strain) exhibited normal growth rates as compared to the parental strain, Δku80. In addition, endodyogeny was impaired in the tachyzoites whose MCA1 and MCA2 were both deleted due to multiple nuclei and abnormal expression of IMC1. We further found that IMC1 of the double-knockout strain was detergent-soluble, indicating that MCA1 and MCA2 are associated with IMC1 maturation. Compared to the parental Δku80 strain, the double-knockout strain was more readily induced from tachyzoites to bradyzoites in vitro. Furthermore, the double-knockout strain was less pathogenic in mice and was able to develop bradyzoites in the brain, which formed cysts and established chronic infection. CONCLUSION: MCA1 and MCA2 are important factors which participate in IMC1 maturation and endodyogeny of T. gondii. The double-knockout strain has slower proliferation and was able to develop bradyzoites both in vitro and in vivo.


Subject(s)
Caspases/metabolism , Protozoan Proteins/metabolism , Toxoplasma/enzymology , Toxoplasma/pathogenicity , Animals , Caspases/classification , Caspases/genetics , Chlorocebus aethiops , Female , Fibroblasts/parasitology , Foreskin/cytology , Humans , Male , Mice , Mice, Inbred BALB C , Protozoan Proteins/genetics , Toxoplasma/genetics , Toxoplasma/physiology , Vero Cells , Virulence
8.
PLoS Pathog ; 17(8): e1009835, 2021 08.
Article in English | MEDLINE | ID: mdl-34432856

ABSTRACT

Intracellular parasites of the phylum Apicomplexa are dependent on the scavenging of essential amino acids from their hosts. We previously identified a large family of apicomplexan-specific plasma membrane-localized amino acid transporters, the ApiATs, and showed that the Toxoplasma gondii transporter TgApiAT1 functions in the selective uptake of arginine. TgApiAT1 is essential for parasite virulence, but dispensable for parasite growth in medium containing high concentrations of arginine, indicating the presence of at least one other arginine transporter. Here we identify TgApiAT6-1 as the second arginine transporter. Using a combination of parasite assays and heterologous characterisation of TgApiAT6-1 in Xenopus laevis oocytes, we demonstrate that TgApiAT6-1 is a general cationic amino acid transporter that mediates both the high-affinity uptake of lysine and the low-affinity uptake of arginine. TgApiAT6-1 is the primary lysine transporter in the disease-causing tachyzoite stage of T. gondii and is essential for parasite proliferation. We demonstrate that the uptake of cationic amino acids by TgApiAT6-1 is 'trans-stimulated' by cationic and neutral amino acids and is likely promoted by an inwardly negative membrane potential. These findings demonstrate that T. gondii has evolved overlapping transport mechanisms for the uptake of essential cationic amino acids, and we draw together our findings into a comprehensive model that highlights the finely-tuned, regulated processes that mediate cationic amino acid scavenging by these intracellular parasites.


Subject(s)
Amino Acid Transport Systems, Basic/metabolism , Amino Acids, Essential/metabolism , Fibroblasts/metabolism , Oocytes/metabolism , Protozoan Proteins/metabolism , Toxoplasmosis/metabolism , Amino Acid Transport Systems, Basic/genetics , Animals , Arginine/metabolism , Biological Transport , Fibroblasts/parasitology , Humans , Lysine/metabolism , Oocytes/parasitology , Protozoan Proteins/genetics , Toxoplasma/physiology , Toxoplasmosis/parasitology , Xenopus laevis
9.
Mol Biochem Parasitol ; 244: 111384, 2021 07.
Article in English | MEDLINE | ID: mdl-34051228

ABSTRACT

A tetracycline-responsive transcription system (Tet-Off) adapted for use in Toxoplasma gondii (nicknamed TATi) is useful for molecular biological studies of this organism. Previous studies using TATi incorporated minimal promoters derived from the gene promoters for TgSAG1 or TgSAG4. The present study achieves improved activation and suppression of an integrated reporter gene in the absence and presence of anhydrotetracycline, respectively (p < 0.0001), by use of a newly derived minimal promoter based on the core promoter of TgGRA2. In comparison with the SAG1 minimal promoter, use of the GRA2 minimal promoter in stable transfectants has a 23-fold higher Signal to Noise Ratio for EYFP fluorescence in the absence or presence of anhydrotetracycline. We conclude that the performance of TATi for both activation and suppression of transcription can be markedly enhanced by incorporating a GRA2 minimal promoter.


Subject(s)
Antigens, Protozoan/genetics , Bacterial Proteins/genetics , Gene Expression Regulation/drug effects , Luminescent Proteins/genetics , Promoter Regions, Genetic , Protozoan Proteins/genetics , Tetracyclines/pharmacology , Toxoplasma/drug effects , Antigens, Protozoan/metabolism , Bacterial Proteins/metabolism , Cells, Cultured , Cloning, Molecular/methods , Fibroblasts/parasitology , Gene Expression , Genes, Reporter , Humans , Luminescent Proteins/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Plasmids/chemistry , Plasmids/metabolism , Protozoan Proteins/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Toxoplasma/genetics , Toxoplasma/metabolism , Transcription, Genetic , Transfection
10.
Parasit Vectors ; 14(1): 253, 2021 May 13.
Article in English | MEDLINE | ID: mdl-33985552

ABSTRACT

BACKGROUND: The predominant genotype of Toxoplasma in China is the Chinese 1 (ToxoDB#9) lineage. TgCtwh3 and TgCtwh6 are two representative strains of Chinese 1, exhibiting high and low virulence to mice, respectively. Little is known regarding the virulence mechanism of this non-classical genotype. Our previous RNA sequencing data revealed differential mRNA levels of TgMIC1 in TgCtwh3 and TgCtwh6. We aim to further confirm the differential expression of TgMIC1 and its significance in this atypical genotype. METHODS: Quantitative real-time PCR was used to verify the RNA sequencing data; then, polyclonal antibodies against TgMIC1 were prepared and identified. Moreover, the invasion and proliferation of the parasite in HFF cells were observed after treatment with TgMIC1 polyclonal antibody or not. RESULTS: The data showed that the protein level of TgMIC1 was significantly higher in high-virulence strain TgCtwh3 than in low-virulence strain TgCtwh6 and that the invasion and proliferation of TgCtwh3 were inhibited by TgMIC1 polyclonal antibody. CONCLUSION: Differential expression of TgMIC1 in TgCtwh3 and TgCtwh6 may explain, at least partly, the virulence mechanism of this atypical genotype.


Subject(s)
Cell Adhesion Molecules/genetics , Genotype , Protozoan Proteins/genetics , Toxoplasma/genetics , Toxoplasma/pathogenicity , Animals , China , Fibroblasts/parasitology , Humans , Mice , Toxoplasma/classification , Toxoplasma/isolation & purification , Virulence
11.
mSphere ; 6(3)2021 05 19.
Article in English | MEDLINE | ID: mdl-34011689

ABSTRACT

Toxoplasma gondii is a widespread apicomplexan parasite that causes severe disease in immunocompromised individuals and the developing fetus. Like other apicomplexans, T. gondii uses an unusual form of substrate-dependent gliding motility to invade cells of its hosts and to disseminate throughout the body during infection. It is well established that a myosin motor consisting of a class XIVa heavy chain (TgMyoA) and two light chains (TgMLC1 and TgELC1/2) plays an important role in parasite motility. The ability of the motor to generate force at the parasite periphery is thought to be reliant upon its anchoring and immobilization within a peripheral membrane-bound compartment, the inner membrane complex (IMC). The motor does not insert into the IMC directly; rather, this interaction is believed to be mediated by the binding of TgMLC1 to the IMC-anchored protein, TgGAP45. Therefore, the binding of TgMLC1 to TgGAP45 is considered a key element in the force transduction machinery of the parasite. TgMLC1 is palmitoylated, and we show here that palmitoylation occurs on two N-terminal cysteine residues, C8 and C11. Mutations that block TgMLC1 palmitoylation completely abrogate the binding of TgMLC1 to TgGAP45. Surprisingly, the loss of TgMLC1 binding to TgGAP45 in these mutant parasites has little effect on their ability to initiate or sustain movement. These results question a key tenet of the current model of apicomplexan motility and suggest that our understanding of gliding motility in this important group of human and animal pathogens is not yet complete.IMPORTANCE Gliding motility plays a central role in the life cycle of T. gondii and other apicomplexan parasites. The myosin motor thought to power motility is essential for virulence but distinctly different from the myosins found in humans. Consequently, an understanding of the mechanism(s) underlying parasite motility and the role played by this unusual myosin may reveal points of vulnerability that can be targeted for disease prevention or treatment. We show here that mutations that uncouple the motor from what is thought to be a key structural component of the motility machinery have little impact on parasite motility. This finding runs counter to predictions of the current, widely held "linear motor" model of motility, highlighting the need for further studies to fully understand how apicomplexan parasites generate the forces necessary to move into, out of, and between cells of the hosts they infect.


Subject(s)
Lipoylation , Membrane Proteins/antagonists & inhibitors , Molecular Motor Proteins/chemistry , Myosin Light Chains/antagonists & inhibitors , Protozoan Proteins/antagonists & inhibitors , Toxoplasma/metabolism , Fibroblasts/parasitology , Foreskin/cytology , Host-Parasite Interactions/physiology , Humans , Life Cycle Stages , Male , Membrane Proteins/genetics , Molecular Motor Proteins/genetics , Molecular Motor Proteins/metabolism , Movement , Mutation , Myosin Light Chains/genetics , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Toxoplasma/genetics
12.
Nat Commun ; 12(1): 2813, 2021 05 17.
Article in English | MEDLINE | ID: mdl-34001876

ABSTRACT

Apicomplexa are obligate intracellular parasites responsible for major human diseases. Their intracellular survival relies on intense lipid synthesis, which fuels membrane biogenesis. Parasite lipids are generated as an essential combination of fatty acids scavenged from the host and de novo synthesized within the parasite apicoplast. The molecular and metabolic mechanisms allowing regulation and channeling of these fatty acid fluxes for intracellular parasite survival are currently unknown. Here, we identify an essential phosphatidic acid phosphatase in Toxoplasma gondii, TgLIPIN, as the central metabolic nexus responsible for controlled lipid synthesis sustaining parasite development. Lipidomics reveal that TgLIPIN controls the synthesis of diacylglycerol and levels of phosphatidic acid that regulates the fine balance of lipids between storage and membrane biogenesis. Using fluxomic approaches, we uncover the first parasite host-scavenged lipidome and show that TgLIPIN prevents parasite death by 'lipotoxicity' through effective channeling of host-scavenged fatty acids to storage triacylglycerols and membrane phospholipids.


Subject(s)
Cell Membrane/metabolism , Lipidomics/methods , Phosphatidate Phosphatase/metabolism , Phospholipids/metabolism , Protozoan Proteins/metabolism , Toxoplasma/metabolism , Cells, Cultured , Cytoplasm/metabolism , Endoplasmic Reticulum/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/parasitology , Foreskin/cytology , Gene Knockdown Techniques , Homeostasis/genetics , Host-Parasite Interactions , Humans , Male , Microscopy, Electron, Transmission , Microscopy, Fluorescence , Phosphatidate Phosphatase/genetics , Protozoan Proteins/genetics , Toxoplasma/genetics , Toxoplasma/ultrastructure
13.
mBio ; 12(2)2021 03 02.
Article in English | MEDLINE | ID: mdl-33653884

ABSTRACT

Polymorphic effector proteins determine the susceptibility of Toxoplasma gondii strains to IFN-γ-mediated clearance mechanisms deployed by murine host cells. However, less is known about the influence of these polymorphic effector proteins on IFN-γ-independent clearance mechanisms. Here, we show that deletion of one such polymorphic effector protein, ROP16, from a type III background leads to a defect in parasite growth and survival in unstimulated human fibroblasts and murine macrophages. Rescue of these defects requires a ROP16 with a functional kinase domain and the ability to activate a specific family of host cell transcription factors (STAT3, 5a, and 6). The growth and survival defects correlate with an accumulation of host cell reactive oxygen species (ROS) and are prevented by treatment with an ROS inhibitor. Exogenous activation of STAT3 and 6 suppresses host cell ROS production during infection with ROP16-deficient parasites and depletion of STAT6, but not STAT3 or 5a, causes an accumulation of ROS in cells infected with wild-type parasites. Pharmacological inhibition of NOX2 and mitochondrially derived ROS also rescues growth and survival of ROP16-deficient parasites. Collectively, these findings reveal an IFN-γ-independent mechanism of parasite restriction in human cells that is subverted by injection of ROP16 by type III parasites.IMPORTANCEToxoplasma gondii is an obligate intracellular parasite that infects up to one-third of the world's population. Control of the parasite is largely accomplished by IFN-γ-dependent mechanisms that stimulate innate and adaptive immune responses. Parasite suppression of IFN-γ-stimulated responses has been linked to proteins that the parasite secretes into its host cell. These secreted proteins vary by T. gondii strain and determine strain-specific lethality in mice. How these strain-specific polymorphic effector proteins affect IFN-γ-independent parasite control mechanisms in human and murine cells is not well known. This study shows that one such secreted protein, ROP16, enables efficient parasite growth and survival by suppressing IFN-γ-independent production of ROS by human and mouse cells.


Subject(s)
Host-Parasite Interactions , Protein-Tyrosine Kinases/genetics , Protozoan Proteins/genetics , Reactive Oxygen Species/antagonists & inhibitors , STAT6 Transcription Factor/genetics , Toxoplasma/growth & development , Toxoplasma/genetics , Animals , Cell Line , Fibroblasts/parasitology , Humans , Immunity, Innate , Macrophages , Mice , Protein-Tyrosine Kinases/metabolism , Protozoan Proteins/metabolism , STAT6 Transcription Factor/metabolism , THP-1 Cells , Toxoplasma/immunology , Toxoplasma/physiology
14.
PLoS Pathog ; 17(2): e1009325, 2021 02.
Article in English | MEDLINE | ID: mdl-33635921

ABSTRACT

Apicomplexan parasites are causative agents of major human diseases. Calcium Dependent Protein Kinases (CDPKs) are crucial components for the intracellular development of apicomplexan parasites and are thus considered attractive drug targets. CDPK7 is an atypical member of this family, which initial characterization suggested to be critical for intracellular development of both Apicomplexa Plasmodium falciparum and Toxoplasma gondii. However, the mechanisms via which it regulates parasite replication have remained unknown. We performed quantitative phosphoproteomics of T. gondii lacking TgCDPK7 to identify its parasitic targets. Our analysis lead to the identification of several putative TgCDPK7 substrates implicated in critical processes like phospholipid (PL) synthesis and vesicular trafficking. Strikingly, phosphorylation of TgRab11a via TgCDPK7 was critical for parasite intracellular development and protein trafficking. Lipidomic analysis combined with biochemical and cellular studies confirmed that TgCDPK7 regulates phosphatidylethanolamine (PE) levels in T. gondii. These studies provide novel insights into the regulation of these processes that are critical for parasite development by TgCDPK7.


Subject(s)
Lipogenesis , Phosphatidylethanolamines/metabolism , Protein Kinases/metabolism , Protozoan Proteins/metabolism , Toxoplasma/enzymology , Toxoplasmosis/metabolism , Transport Vesicles/metabolism , Biological Transport , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/parasitology , Humans , Phosphorylation , Protein Kinases/genetics , Protozoan Proteins/genetics , Toxoplasma/growth & development , Toxoplasmosis/parasitology
15.
mBio ; 12(1)2021 02 16.
Article in English | MEDLINE | ID: mdl-33593973

ABSTRACT

The inner membrane complex (IMC) is a unique organelle of apicomplexan parasites that plays critical roles in parasite motility, host cell invasion, and replication. Despite the common functions of the organelle, relatively few IMC proteins are conserved across the phylum and the precise roles of many IMC components remain to be characterized. Here, we identify a novel component of the Toxoplasma gondii IMC (IMC32) that localizes to the body portion of the IMC and is recruited to developing daughter buds early during endodyogeny. IMC32 is essential for parasite survival, as its conditional depletion results in a complete collapse of the IMC that is lethal to the parasite. We demonstrate that localization of IMC32 is dependent on both an N-terminal palmitoylation site and a series of C-terminal coiled-coil domains. Using deletion analyses and functional complementation, we show that two conserved regions within the C-terminal coiled-coil domains play critical roles in protein function during replication. Together, this work reveals an essential component of parasite replication that provides a novel target for therapeutic intervention of T. gondii and related apicomplexan parasites.IMPORTANCE The IMC is an important organelle that apicomplexan parasites use to maintain their intracellular lifestyle. While many IMC proteins have been identified, only a few central players that are essential for internal budding have been described and even fewer are conserved across the phylum. Here, we identify IMC32, a novel component of the Toxoplasma gondii IMC that localizes to very early daughter buds, indicating a role in the early stages of parasite replication. We then demonstrate that IMC32 is essential for parasite survival and pinpoint conserved regions within the protein that are important for membrane association and daughter cell formation. As IMC32 is unique to these parasites and not present in their mammalian hosts, it serves as a new target for the development of drugs that exclusively affect these important intracellular pathogens.


Subject(s)
Cell Division/genetics , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Toxoplasma/genetics , Toxoplasma/physiology , Cell Membrane , Cells, Cultured , Fibroblasts/parasitology , Foreskin , Humans , Male , Toxoplasma/chemistry , Toxoplasma/cytology
16.
Diagn Microbiol Infect Dis ; 99(4): 115296, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33387894

ABSTRACT

The objective of this pilot study was to examine the activity of iclaprim, a diaminopyrimidine dihydrofolate reducatase inhibitor, in an in vitro infection model of infection with Toxoplasma gondii. Toxoplasma growth was assessed by enzyme linked immunoassay (ELISA) performed directly on the fixed cultures using a peroxidase labeled monoclonal antibody directed against the SAG-l surface protein of T. gondii. For each well, the results were expressed as optical density (OD) values. Iclaprim inhibited T. gondii growth at concentrations between 0.1 and 10 mg/L; the IC50 was estimated at 0.26 mg/L (95% confidence interval 0.22-0.33). Iclaprim was about 10 times more active than trimethoprim, which had an IC50 of 2.3 mg/L. Iclaprim demonstrated synergistic effects at concentrations of 0.02, 0.05 and 0.1 mg/L when combined with subinhibitory concentrations of sulfamethoxazole (0.1 or 0.02 mg/L). These results show that iclaprim is a potent inhibitor of T. gondii growth in vitro. In addition, iclaprim exhibited synergy in vitro when tested in the presence of sulfamethoxazole. Iclaprim should be further investigated as an agent for the treatment or prophylaxis of toxoplasmosis.


Subject(s)
Fibroblasts/parasitology , Folic Acid Antagonists/pharmacology , Pyrimidines/pharmacology , Tetrahydrofolate Dehydrogenase/metabolism , Toxoplasma/drug effects , Toxoplasma/enzymology , Animals , Cell Line , Pilot Projects
17.
mBio ; 12(1)2021 01 26.
Article in English | MEDLINE | ID: mdl-33500345

ABSTRACT

Toxoplasma gondii is an obligate intracellular parasite that persists in its vertebrate hosts in the form of dormant tissue cysts, which facilitate transmission through predation. The parasite must strike a balance that allows it to disseminate throughout its host without killing it, which requires the ability to properly counter host cell defenses. For example, oxidative stress encountered by Toxoplasma is suggested to impair parasite replication and dissemination. However, the strategies by which Toxoplasma mitigates oxidative stress are not yet clear. Among eukaryotes, environmental stresses induce the integrated stress response via phosphorylation of a translation initiation factor, eukaryotic initiation factor 2 (eIF2). Here, we show that the Toxoplasma eIF2 kinase TgIF2K-B is activated in response to oxidative stress and affords protection. Knockout of the TgIF2K-B gene, Δtgif2k-b, disrupted parasite responses to oxidative stresses and enhanced replication, diminishing the ability of the parasite to differentiate into tissue cysts. In addition, parasites lacking TgIF2K-B exhibited resistance to activated macrophages and showed greater virulence in an in vivo model of infection. Our results establish that TgIF2K-B is essential for Toxoplasma responses to oxidative stress, which are important for the parasite's ability to establish persistent infection in its host.IMPORTANCEToxoplasma gondii is a single-celled parasite that infects nucleated cells of warm-blooded vertebrates, including one-third of the human population. The parasites are not cleared by the immune response and persist in the host by converting into a latent tissue cyst form. Development of tissue cysts can be triggered by cellular stresses, which activate a family of TgIF2 kinases to phosphorylate the eukaryotic translation initiation factor TgIF2α. Here, we establish that the TgIF2 kinase TgIF2K-B is activated by oxidative stress and is critical for maintaining oxidative balance in the parasite. Depletion of TgIF2K-B alters gene expression, leading to accelerated growth and a diminished ability to convert into tissue cysts. This study establishes that TgIF2K-B is essential for the parasite's oxidative stress response and its ability to persist in the host as a latent infection.


Subject(s)
Host-Parasite Interactions , Oxidative Stress , Toxoplasma/metabolism , Toxoplasma/pathogenicity , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism , Fibroblasts/parasitology , Foreskin/cytology , Gene Knockout Techniques , Humans , Male , Phosphorylation , Stress, Physiological , Toxoplasma/enzymology , Virulence
18.
mSphere ; 6(1)2021 01 06.
Article in English | MEDLINE | ID: mdl-33408226

ABSTRACT

Toxoplasmosis, a protozoan infection caused by Toxoplasma gondii, is estimated to affect around 2.5 billion people worldwide. Nevertheless, the side effects of drugs combined with the long period of therapy usually result in discontinuation of the treatment. New therapies should be developed by exploring peculiarities of the parasite's metabolic pathways, similarly to what has been well described in cancer cell metabolism. An example is the switch in the metabolism of cancer that blocks the conversion of pyruvate into acetyl coenzyme A in mitochondria. In this context, dichloroacetate (DCA) is an anticancer drug that reverts the tumor proliferation by inhibiting the enzymes responsible for this switch: the pyruvate dehydrogenase kinases (PDKs). DCA has also been used in the treatment of certain symptoms of malaria; however, there is no evidence of how this drug affects apicomplexan species. In this paper, we studied the metabolism of T. gondii and demonstrate that DCA also inhibits T. gondii's in vitro infection with no toxic effects on host cells. DCA caused an increase in the activity of pyruvate dehydrogenase followed by an unbalanced mitochondrial activity. We also observed morphological alterations frequently in mitochondria and in a few apicoplasts, essential organelles for parasite survival. To date, the kinases that potentially regulate the activity of pyruvate metabolism in both organelles have never been described. Here, we confirmed the presence in the genome of two putative kinases (T. gondii PDK [TgPDK] and T. gondii branched-chain α-keto acid dehydrogenase kinase [TgBCKDK]), verified their cellular localization in the mitochondrion, and provided in silico data suggesting that they are potential targets of DCA.IMPORTANCE Currently, the drugs used for toxoplasmosis have severe toxicity to human cells, and the treatment still lacks effective and safer alternatives. The search for novel drug targets is timely. We report here that the treatment of T. gondii with an anticancer drug, dichloroacetate (DCA), was effective in decreasing in vitro infection without toxicity to human cells. It is known that PDK is the main target of DCA in mammals, and this inactivation increases the conversion of pyruvate into acetyl coenzyme A and reverts the proliferation of tumor cells. Moreover, we verified the mitochondrial localization of two kinases that possibly regulate the activity of pyruvate metabolism in T. gondii, which has never been studied. DCA increased pyruvate dehydrogenase (PDH) activity in T. gondii, followed by an unbalanced mitochondrial activity, in a manner similar to what was previously observed in cancer cells. Thus, we propose the conserved kinases as potential regulators of pyruvate metabolism and interesting targets for new therapies.


Subject(s)
Antiprotozoal Agents/pharmacology , Apoptosis/drug effects , Dichloroacetic Acid/pharmacology , Fibroblasts/drug effects , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/antagonists & inhibitors , Pyruvates/metabolism , Toxoplasma/drug effects , Dichloroacetic Acid/chemistry , Fibroblasts/parasitology , Humans , Metabolic Networks and Pathways/drug effects , Mitochondria/metabolism , Oxidation-Reduction , Oxidoreductases , Toxoplasmosis/drug therapy
19.
Front Cell Infect Microbiol ; 11: 788482, 2021.
Article in English | MEDLINE | ID: mdl-35071040

ABSTRACT

Trypanosoma cruzi invades non-professional phagocytic cells by subverting their membrane repair process, which is dependent on membrane injury and cell signaling, intracellular calcium increase, and lysosome recruitment. Cells lacking lysosome-associated membrane proteins 1 and 2 (LAMP1 and LAMP2) are less permissive to parasite invasion but more prone to parasite intracellular multiplication. Several passages through a different intracellular environment can significantly change T. cruzi's gene expression profile. Here, we evaluated whether one single passage through LAMP-deficient (KO) or wild-type (WT) fibroblasts, thus different intracellular environments, could influence T. cruzi Y strain trypomastigotes' ability to invade L6 myoblasts and WT fibroblasts host cells. Parasites released from LAMP2 KO cells (TcY-L2-/-) showed higher invasion, calcium signaling, and membrane injury rates, for the assays in L6 myoblasts, when compared to those released from WT (TcY-WT) or LAMP1/2 KO cells (TcY-L1/2-/-). On the other hand, TcY-L1/2-/- showed higher invasion, calcium signaling, and cell membrane injury rates, for the assays in WT fibroblasts, compared to TcY-WT and TcY-L1/2-/-. Albeit TcY-WT presented an intermediary invasion and calcium signaling rates, compared to the others, in WT fibroblasts, they induced lower levels of injury, which reinforces that signals mediated by surface membrane protein interactions also have a significant contribution to trigger host cell calcium signals. These results clearly show that parasites released from WT or LAMP KO cells are distinct from each other. Additionally, these parasites' ability to invade the cell may be distinct depending on which cell type they interact with. Since these alterations most likely would reflect differences among parasite surface molecules, we also evaluated their proteome. We identified few protein complexes, membrane, and secreted proteins regulated in our dataset. Among those are some members of MASP, mucins, trans-sialidases, and gp63 proteins family, which are known to play an important role during parasite infection and could correlate to TcY-WT, TcY-L1/2-/-, and TcY-L2-/- biological behavior.


Subject(s)
Chagas Disease , Trypanosoma cruzi , Animals , Cells, Cultured , Chagas Disease/pathology , Fibroblasts/parasitology , Lysosomal-Associated Membrane Protein 2/genetics , Lysosomal Membrane Proteins/genetics , Lysosomes , Membrane Proteins , Mice , Myoblasts/parasitology
20.
Cell Microbiol ; 23(2): e13278, 2021 02.
Article in English | MEDLINE | ID: mdl-33040458

ABSTRACT

Toxoplasma gondii infects virtually any nucleated cell and resides inside a non-phagocytic vacuole surrounded by a parasitophorous vacuolar membrane (PVM). Pivotal to the restriction of T. gondii dissemination upon infection in murine cells is the recruitment of immunity regulated GTPases (IRGs) and guanylate binding proteins (GBPs) to the PVM that leads to pathogen elimination. The virulent T. gondii type I RH strain secretes a handful of effectors including the dense granule protein GRA7, the serine-threonine kinases ROP17 and ROP18, and a pseudo-kinase ROP5, that synergistically inhibit the recruitment of IRGs to the PVM. Here, we characterise GRA60, a novel dense granule effector, which localises to the vacuolar space and PVM and contributes to virulence of RH in mice, suggesting a role in the subversion of host cell defence mechanisms. Members of the host cell IRG defence system Irgb10 and Irga6 are recruited to the PVM of RH parasites lacking GRA60 as observed previously for the avirulent RHΔrop5 mutant, with RH preventing such recruitment. Deletion of GRA60 in RHΔrop5 leads to a recruitment of IRGs comparable to the single knockouts. GRA60 therefore represents a novel parasite effector conferring resistance to IRGs in type I parasites, and found associated to ROP18, a member of the virulence complex.


Subject(s)
Protozoan Proteins/immunology , Protozoan Proteins/metabolism , Toxoplasma/immunology , Toxoplasma/metabolism , Toxoplasmosis/immunology , Toxoplasmosis/metabolism , Animals , Antigens, Protozoan/immunology , Antigens, Protozoan/metabolism , DNA, Protozoan , Fibroblasts/parasitology , Foreskin/parasitology , GTP Phosphohydrolases/immunology , GTP Phosphohydrolases/metabolism , Gene Knockout Techniques , Host-Parasite Interactions , Humans , Immunity , Interferon-gamma/metabolism , Male , Mice , Mice, Inbred C57BL , Mutation , Protein Serine-Threonine Kinases/metabolism , Toxoplasma/genetics , Vacuoles/metabolism , Virulence
SELECTION OF CITATIONS
SEARCH DETAIL
...