Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.358
1.
Parasitol Res ; 123(5): 217, 2024 May 22.
Article En | MEDLINE | ID: mdl-38772951

Toxoplasmosis poses a global health threat, ranging from asymptomatic cases to severe, potentially fatal manifestations, especially in immunocompromised individuals and congenital transmission. Prior research suggests that oregano essential oil (OEO) exhibits diverse biological effects, including antiparasitic activity against Toxoplasma gondii. Given concerns about current treatments, exploring new compounds is important. This study was to assess the toxicity of OEO on BeWo cells and T. gondii tachyzoites, as well as to evaluate its effectiveness in in vitro infection models and determine its direct action on free tachyzoites. OEO toxicity on BeWo cells and T. gondii tachyzoites was assessed by MTT and trypan blue methods, determining cytotoxic concentration (CC50), inhibitory concentration (IC50), and selectivity index (SI). Infection and proliferation indices were analyzed. Direct assessments of the parasite included reactive oxygen species (ROS) levels, mitochondrial membrane potential, necrosis, and apoptosis, as well as electron microscopy. Oregano oil exhibited low cytotoxicity on BeWo cells (CC50: 114.8 µg/mL ± 0.01) and reduced parasite viability (IC50 12.5 ± 0.06 µg/mL), demonstrating 9.18 times greater selectivity for parasites than BeWo cells. OEO treatment significantly decreased intracellular proliferation in infected cells by 84% after 24 h with 50 µg/mL. Mechanistic investigations revealed increased ROS levels, mitochondrial depolarization, and lipid droplet formation, linked to autophagy induction and plasma membrane permeabilization. These alterations, observed through electron microscopy, suggested a necrotic process confirmed by propidium iodide labeling. OEO treatment demonstrated anti-T. gondii action through cellular and metabolic change while maintaining low toxicity to trophoblastic cells.


Autophagy , Oils, Volatile , Origanum , Reactive Oxygen Species , Toxoplasma , Oils, Volatile/pharmacology , Oils, Volatile/chemistry , Toxoplasma/drug effects , Toxoplasma/growth & development , Origanum/chemistry , Humans , Autophagy/drug effects , Reactive Oxygen Species/metabolism , Cell Line , Antiprotozoal Agents/pharmacology , Inhibitory Concentration 50 , Necrosis/drug therapy , Cell Survival/drug effects , Apoptosis/drug effects , Membrane Potential, Mitochondrial/drug effects
2.
Mol Biochem Parasitol ; 258: 111615, 2024 Jun.
Article En | MEDLINE | ID: mdl-38354788

Toxoplasma has high host flexibility, infecting all nucleated cells of mammals and birds. This implies that during its infective process the parasite must constantly adapt to different environmental situations, which in turn leads to modifications in its metabolism, regulation of gene transcription, translation of mRNAs and stage specific factors. There are conserved pathways that support these adaptations, which we aim to elucidate in this review. We begin by exploring the widespread epigenetic mechanisms and transcription regulators, continue with the supportive role of Heat Shock Proteins (Hsp), the translation regulation, stress granules, and finish with the emergence of contingency genes in highly variable genomic domains, such as subtelomeres. Within epigenetics, the discovery of a new histone variant of the H2B family (H2B.Z), contributing to T. gondii virulence and differentiation, but also gene expression regulation and its association with the metabolic state of the parasite, is highlighted. Associated with the regulation of gene expression are transcription factors (TFs). An overview of the main findings on TF and development is presented. We also emphasize the role of Hsp90 and Tgj1 in T. gondii metabolic fitness and the regulation of protein translation. Translation regulation is also highlighted as a mechanism for adaptation to conditions encountered by the parasite as well as stress granules containing mRNA and proteins generated in the extracellular tachyzoite. Another important aspect in evolution and adaptability are the subtelomeres because of their high variability and gene duplication rate. Toxoplasma possess multigene families of membrane proteins and contingency genes that are associated with different metabolic stresses. Among them parasite differentiation and environmental stresses stand out, including those that lead tachyzoite to bradyzoite conversion. Finally, we are interested in positioning protozoa as valuable evolution models, focusing on research related to the Extended Evolutionary Synthesis, based on models recently generated, such as extracellular adaptation and ex vivo cyst recrudescence.


Adaptation, Physiological , Epigenesis, Genetic , Toxoplasma , Toxoplasma/genetics , Toxoplasma/metabolism , Toxoplasma/growth & development , Animals , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Gene Expression Regulation , Humans , Biological Evolution , Transcription Factors/genetics , Transcription Factors/metabolism , Evolution, Molecular
3.
Nature ; 625(7994): 366-376, 2024 Jan.
Article En | MEDLINE | ID: mdl-38093015

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.


Cats , In Vitro Techniques , Life Cycle Stages , Toxoplasma , Animals , Cats/parasitology , Humans , Chromatin/genetics , Chromatin/metabolism , Disease Models, Animal , Epigenesis, Genetic , In Vitro Techniques/methods , Life Cycle Stages/genetics , Merozoites/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic/genetics , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Toxoplasma/genetics , Toxoplasma/growth & development , Toxoplasma/physiology , Toxoplasmosis/genetics , Toxoplasmosis/parasitology , Toxoplasmosis/transmission , Transcription, Genetic
4.
Arch Microbiol ; 205(9): 314, 2023 Aug 21.
Article En | MEDLINE | ID: mdl-37603130

Manipulative neuroparasites are a fascinating group of organisms that possess the ability to hijack the nervous systems of their hosts, manipulating their behavior in order to enhance their own survival and reproductive success. This review provides an overview of the different strategies employed by manipulative neuroparasites, ranging from viruses to parasitic worms and fungi. By examining specific examples, such as Toxoplasma gondii, Leucochloridium paradoxum, and Ophiocordyceps unilateralis, we highlight the complex mechanisms employed by these parasites to manipulate their hosts' behavior. We explore the mechanisms through which these parasites alter the neural processes and behavior of their hosts, including the modulation of neurotransmitters, hormonal pathways, and neural circuits. This review focuses less on the diseases that neuroparasites induce and more on the process of their neurological manipulation. We also investigate the fundamental mechanisms of host manipulation in the developing field of neuroparasitology, which blends neuroscience and parasitology. Finally, understanding the complex interaction between manipulative neuroparasites and their hosts may help us to better understand the fundamentals of behavior, neurology, and host-parasite relationships.


Hypocreales , Nervous System , Toxoplasma , Trematoda , Toxoplasma/growth & development , Toxoplasma/physiology , Trematoda/growth & development , Trematoda/physiology , Hypocreales/growth & development , Hypocreales/physiology , Rabies virus/physiology , Animals , Nervous System/microbiology , Nervous System/parasitology , Humans , Host-Pathogen Interactions
5.
Microbiol Spectr ; 10(1): e0149521, 2022 02 23.
Article En | MEDLINE | ID: mdl-35196797

The apicoplast, which harbors key pathways involved in biosynthesis of vital metabolites, is a unique and essential nonphotosynthetic plastid organelle in apicomplexan parasites. Intriguingly, autophagy-related protein 8 (Atg8), a highly conserved eukaryotic protein, can localize to the outermost membrane of the apicoplast and modulate its inheritance in both Toxoplasma and Plasmodium parasites. The Atg8-Atg3 interaction plays a key role in Atg8 lipidation and localization, and our previously work in Toxoplasma has suggested that the core Atg8-family interacting motif (AIM) in TgAtg3, 239FADI242, and the R27 residue of TgAtg8 contribute to TgAtg8-TgAtg3 interaction in vitro. However, little is known about the function of this interaction or its importance in tachyzoite growth in Toxoplasma gondii. Here, we generated two complemented cell lines, TgAtg3F239A/I242A and TgAtg8R27E, based on the TgAtg3 and TgAtg8 conditional knockdown cell lines, respectively. We found that both mutant complemented cell lines were severely affected in terms of tachyzoite growth and displayed delayed death upon conditional knockdown of endogenous TgAtg3 or TgAtg8. Intriguingly, both complemented lines appeared to be defective in TgAtg8 lipidation and apicoplast inheritance. Moreover, we showed that the interaction of TgAtg8 and TgAtg3 is critical for TgAtg8 apicoplast localization. In addition, we found that the TgAtg3F239A/I242A complemented line exhibits an integral mitochondrial network upon ablation of endogenous TgAtg3, which is distinct from TgAtg3-depleted parasites with a fragmented mitochondrial network. Taken together, this work solidifies the contribution of the TgAtg8-TgAtg3 interaction to apicoplast inheritance and the growth of T. gondii tachyzoites. IMPORTANCEToxoplasma gondiiis a widespread intracellular parasite infecting a variety of warm-blooded animals, including humans. Current frontline treatment of toxoplasmosis suffers many drawbacks, including toxicity, drug resistance, and failure to eradicate tissue cysts, underscoring the need to identify novel drug targets for suppression or treatment of toxoplasmosis. TgAtg8 is thought to serve multiple functions in lipidation and is considered essential to the growth and development of both tachyzoites and bradyzoites. Here, we show that Toxoplasma gondii has adapted a conserved Atg8-Atg3 interaction, required for canonical autophagy in other eukaryotes, to function specifically in apicoplast inheritance. Our finding not only highlights the importance of TgAtg8-TgAtg3 interaction in tachyzoite growth but also suggests that this interaction is a promising drug target for the therapy of toxoplasmosis.


Apicoplasts/metabolism , Protozoan Proteins/metabolism , Toxoplasma/growth & development , Toxoplasma/metabolism , Toxoplasmosis/microbiology , Amino Acid Motifs , Apicoplasts/chemistry , Apicoplasts/genetics , Humans , Mutation , Protein Binding , Protein Transport , Protozoan Proteins/chemistry , Protozoan Proteins/genetics , Toxoplasma/chemistry , Toxoplasma/genetics
6.
Molecules ; 27(3)2022 Feb 02.
Article En | MEDLINE | ID: mdl-35164288

Toxoplasma gondii is an obligate intracellular protozoan of severe threat to humans and livestock, whose life history harbors both gamic and apogamic stages. Chinese 1 (ToxoDB#9) was a preponderant genotype epidemic in food-derived animals and humans in China, with a different pathogenesis from the strains from the other nations of the world. Posttranslational modifications (PTMs) of proteins were critical mediators of the biology, developmental transforms, and pathogenesis of protozoan parasites. The phosphoprotein profiling and the difference between the developmental phases of T. gondii, contributing to development and infectivity, remain unknown. A quantitative phosphoproteomic approach using IBT integrated with TiO2 affinity chromatography was applied to identify and analyze the difference in the phosphoproteomes between the sporulated oocysts and the tachyzoites of the virulent ToxoDB#9 (PYS) strain of T. gondii. A total of 4058 differential phosphopeptides, consisting of 2597 upregulated and 1461 downregulated phosphopeptides, were characterized between sporulated the oocysts and tachyzoites. Twenty-one motifs extracted from the upregulated phosphopeptides contained 19 serine motifs and 2 threonine motifs (GxxTP and TP), whereas 16 motifs identified from downregulated phosphopeptides included 13 serine motifs and 3 threonine motifs (KxxT, RxxT, and TP). Beyond the traditional kinases, some infrequent classes of kinases, including Ab1, EGFR, INSR, Jak, Src and Syk, were found to be corresponding to motifs from the upregulated and downregulated phosphopeptides. Remarkable functional properties of the differentially expressed phosphoproteins were discovered by GO analysis, KEGG pathway analysis, and STRING analysis. S8GFS8 (DNMT1-RFD domain-containing protein) and S8F5G5 (Histone kinase SNF1) were the two most connected peptides in the kinase-associated network. Out of these, phosphorylated modifications in histone kinase SNF1 have functioned in mitosis and interphase of T. gondii, as well as in the regulation of gene expression relevant to differentiation. Our study discovered a remarkable difference in the abundance of phosphopeptides between the sporulated oocysts and tachyzoites of the virulent ToxoDB#9 (PYS) strain of T. gondii, which may provide a new resource for understanding stage-specific differences in PTMs and may enhance the illustration of the regulatory mechanisms contributing to the development and infectivity of T. gondii.


Phosphoproteins/metabolism , Protozoan Proteins/metabolism , Toxoplasma/growth & development , Animals , Humans , Oocysts/chemistry , Oocysts/growth & development , Oocysts/metabolism , Phosphopeptides/analysis , Phosphopeptides/metabolism , Phosphoproteins/analysis , Proteomics , Protozoan Proteins/analysis , Toxoplasma/chemistry , Toxoplasma/metabolism , Toxoplasmosis/parasitology
7.
Science ; 375(6577): eabi4343, 2022 Jan 14.
Article En | MEDLINE | ID: mdl-35025629

The outer mitochondrial membrane (OMM) is essential for cellular homeostasis. Yet little is known of the mechanisms that remodel it during natural stresses. We found that large "SPOTs" (structures positive for OMM) emerge during Toxoplasma gondii infection in mammalian cells. SPOTs mediated the depletion of the OMM proteins mitofusin 1 and 2, which restrict parasite growth. The formation of SPOTs depended on the parasite effector TgMAF1 and the host mitochondrial import receptor TOM70, which is required for optimal parasite proliferation. TOM70 enabled TgMAF1 to interact with the host OMM translocase SAM50. The ablation of SAM50 or the overexpression of an OMM-targeted protein promoted OMM remodeling independently of infection. Thus, Toxoplasma hijacks the formation of SPOTs, a cellular response to OMM stress, to promote its growth.


Mitochondrial Membranes/physiology , Mitochondrial Precursor Protein Import Complex Proteins/metabolism , Protozoan Proteins/metabolism , Toxoplasma/physiology , Animals , Cell Line , GTP Phosphohydrolases/metabolism , Humans , Intracellular Membranes/physiology , Intracellular Membranes/ultrastructure , Mice , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Membranes/ultrastructure , Mitochondrial Proteins/metabolism , Protein Binding , Stress, Physiological , Toxoplasma/growth & development , Toxoplasma/ultrastructure , Toxoplasmosis/parasitology , Vacuoles/physiology , Vacuoles/ultrastructure
8.
Nat Commun ; 13(1): 345, 2022 01 17.
Article En | MEDLINE | ID: mdl-35039477

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.


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
9.
Nat Commun ; 13(1): 459, 2022 01 24.
Article En | MEDLINE | ID: mdl-35075105

Toxoplasma gondii commonly infects humans and while most infections are controlled by the immune response, currently approved drugs are not capable of clearing chronic infection in humans. Hence, approximately one third of the world's human population is at risk of reactivation, potentially leading to severe sequelae. To identify new candidates for treating chronic infection, we investigated a series of compounds derived from diversity-oriented synthesis. Bicyclic azetidines are potent low nanomolar inhibitors of phenylalanine tRNA synthetase (PheRS) in T. gondii, with excellent selectivity. Biochemical and genetic studies validate PheRS as the primary target of bicyclic azetidines in T. gondii, providing a structural basis for rational design of improved analogs. Favorable pharmacokinetic properties of a lead compound provide excellent protection from acute infection and partial protection from chronic infection in an immunocompromised mouse model of toxoplasmosis. Collectively, PheRS inhibitors of the bicyclic azetidine series offer promise for treatment of chronic toxoplasmosis.


Antiprotozoal Agents/administration & dosage , Azetidines/administration & dosage , Enzyme Inhibitors/administration & dosage , Phenylalanine-tRNA Ligase/antagonists & inhibitors , Protozoan Proteins/antagonists & inhibitors , Toxoplasma/drug effects , Toxoplasma/enzymology , Toxoplasmosis/drug therapy , Animals , Antiprotozoal Agents/chemistry , Azetidines/chemistry , Enzyme Inhibitors/chemistry , Female , Humans , Kinetics , Male , Mice , Mice, Inbred CBA , Phenylalanine-tRNA Ligase/chemistry , Phenylalanine-tRNA Ligase/metabolism , Protozoan Proteins/chemistry , Protozoan Proteins/metabolism , Toxoplasma/genetics , Toxoplasma/growth & development , Toxoplasmosis/parasitology
10.
PLoS Pathog ; 17(11): e1010096, 2021 11.
Article En | MEDLINE | ID: mdl-34793583

Iron-sulfur (Fe-S) clusters are one of the most ancient and ubiquitous prosthetic groups, and they are required by a variety of proteins involved in important metabolic processes. Apicomplexan parasites have inherited different plastidic and mitochondrial Fe-S clusters biosynthesis pathways through endosymbiosis. We have investigated the relative contributions of these pathways to the fitness of Toxoplasma gondii, an apicomplexan parasite causing disease in humans, by generating specific mutants. Phenotypic analysis and quantitative proteomics allowed us to highlight notable differences in these mutants. Both Fe-S cluster synthesis pathways are necessary for optimal parasite growth in vitro, but their disruption leads to markedly different fates: impairment of the plastidic pathway leads to a loss of the organelle and to parasite death, while disruption of the mitochondrial pathway trigger differentiation into a stress resistance stage. This highlights that otherwise similar biochemical pathways hosted by different sub-cellular compartments can have very different contributions to the biology of the parasites, which is something to consider when exploring novel strategies for therapeutic intervention.


Iron-Sulfur Proteins/metabolism , Mitochondria/parasitology , Plastids/parasitology , Protozoan Proteins/metabolism , Symbiosis , Toxoplasma/growth & development , Toxoplasmosis/parasitology , Humans , Iron-Sulfur Proteins/genetics , Mitochondria/metabolism , Plastids/metabolism , Proteome/analysis , Proteome/metabolism , Protozoan Proteins/genetics , Toxoplasma/metabolism , Toxoplasmosis/genetics , Toxoplasmosis/metabolism
11.
Nat Commun ; 12(1): 5802, 2021 10 04.
Article En | MEDLINE | ID: mdl-34608145

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.


Calcium Channels/metabolism , Protozoan Proteins/metabolism , Toxoplasma/growth & development , Toxoplasma/metabolism , Amino Acid Sequence , Apicoplasts/metabolism , Calcium/metabolism , Calcium Channels/chemistry , Calcium Channels/genetics , Calcium Signaling , DNA/metabolism , Endoplasmic Reticulum/metabolism , Humans , Mutation , Organelle Biogenesis , Phylogeny , Protozoan Proteins/chemistry , Protozoan Proteins/genetics
12.
Bioorg Med Chem ; 50: 116467, 2021 11 15.
Article En | MEDLINE | ID: mdl-34666274

Toxoplasma gondiiis an apicomplexan parasite, the causative agent of toxoplasmosis, a common disease in the world. Toxoplasmosis could be severe, especially in immunocompromised patients. The current therapy is limited, where pyrimethamine and sulfadiazine are the best choices despite being associated with side effects and ineffective against the bradyzoites, the parasitic form present during the chronic phase of the infection. Thus, new therapies against both tachyzoites and bradyzoites from T. gondii are urgent. Herein, we present the anti-T. gondii effect of 1,10-phenanthroline and its N-phenyl-1,10-phenanthroline-2-amine derivatives. The chemical modification of 1,10-phenanthroline tonew derivatives improved the anti-T. gondiiactivity 3.4 fold. The most active derivative presented ED50in the nanomolar range, the smallest value found was for Ph8, 0.1 µM for 96 h of treatment. The host cell viability was maintained after the treatment with the compounds, which were found to be highly selective presenting large selectivity indexes. Treatment with derivatives for 96 h was able to eliminate the T. gondii infection irreversibly. The ultrastructural alterations caused after the treatment with the most effective derivative (Ph8) included signs of cell death, specifically revealed by the Tunel assay for detection of DNA fragmentation. The Phen derivatives were also able to control the growth of the in vitro-derived bradyzoite forms of T. gondii EGS strain, causing its lysis and death. These findings promote the 1,10-phenanthroline derivatives as potential lead compounds for the development of a treatment for acute and chronic phases of toxoplasmosis.


Antiprotozoal Agents/pharmacology , Toxoplasma/drug effects , Antiprotozoal Agents/chemical synthesis , Antiprotozoal Agents/chemistry , Dose-Response Relationship, Drug , Molecular Structure , Parasitic Sensitivity Tests , Structure-Activity Relationship , Toxoplasma/growth & development
13.
J Enzyme Inhib Med Chem ; 36(1): 1145-1164, 2021 Dec.
Article En | MEDLINE | ID: mdl-34074198

We report herein anti-proliferation effects of 4-arylthiosemicarbazides, with a cyclopentane substitution at N1 position, on highly virulent RH strain of Toxoplasma gondii. Among them, the highest in vitro anti-Toxoplasma activity was found with the meta-iodo derivative. Further experiments demonstrated inhibitory effects of thiosemicarbazides on tyrosinase (Tyr) activity, and good correlation was found between percentage of Tyr inhibition and IC50Tg. To confirm the concept that thiosemicarbazides are able to disrupt tyrosine metabolism in Toxoplasma tachyzoites, the most potent Tyr inhibitors were tested for their efficacy of T. gondii growth inhibition. All of them significantly reduced the number of tachyzoites in the parasitophorous vacuoles (PVs) compared to untreated cells, as well as inhibited tachyzoites growth by impeding cell division. Collectively, these results indicate that compounds with the thiosemicarbazide scaffold are able to disrupt tyrosine metabolism in Toxoplasma tachyzoites by deregulation of their crucial enzyme tyrosine hydroxylase (TyrH).


Antiprotozoal Agents/pharmacology , Enzyme Inhibitors/pharmacology , Monophenol Monooxygenase/antagonists & inhibitors , Semicarbazides/pharmacology , Toxoplasma/drug effects , Antiprotozoal Agents/chemical synthesis , Antiprotozoal Agents/chemistry , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Molecular Structure , Monophenol Monooxygenase/metabolism , Parasitic Sensitivity Tests , Semicarbazides/chemical synthesis , Semicarbazides/chemistry , Structure-Activity Relationship , Toxoplasma/growth & development
14.
Sci Rep ; 11(1): 12709, 2021 06 16.
Article En | MEDLINE | ID: mdl-34135407

Congenital toxoplasmosis is represented by the transplacental passage of Toxoplasma gondii from the mother to the fetus. Our studies demonstrated that T. gondii developed mechanisms to evade of the host immune response, such as cyclooxygenase (COX)-2 and prostaglandin E2 (PGE2) induction, and these mediators can be produced/stored in lipid droplets (LDs). The aim of this study was to evaluate the role of COX-2 and LDs during T. gondii infection in human trophoblast cells and villous explants. Our data demonstrated that COX-2 inhibitors decreased T. gondii replication in trophoblast cells and villous. In BeWo cells, the COX-2 inhibitors induced an increase of pro-inflammatory cytokines (IL-6 and MIF), and a decrease in anti-inflammatory cytokines (IL-4 and IL-10). In HTR-8/SVneo cells, the COX-2 inhibitors induced an increase of IL-6 and nitrite and decreased IL-4 and TGF-ß1. In villous explants, the COX-2 inhibitors increased MIF and decreased TNF-α and IL-10. Furthermore, T. gondii induced an increase in LDs in BeWo and HTR-8/SVneo, but COX-2 inhibitors reduced LDs in both cells type. We highlighted that COX-2 is a key factor to T. gondii proliferation in human trophoblast cells, since its inhibition induced a pro-inflammatory response capable of controlling parasitism and leading to a decrease in the availability of LDs, which are essentials for parasite growth.


Chorionic Villi/parasitology , Cyclooxygenase 2/metabolism , Lipid Droplets/metabolism , Toxoplasma/growth & development , Trophoblasts/parasitology , Cell Line , Cell Survival/drug effects , Chorionic Villi/immunology , Chorionic Villi/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Extracellular Matrix Proteins/metabolism , Host-Parasite Interactions , Humans , Interleukins/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Nitrites/metabolism , Toxoplasma/immunology , Transforming Growth Factor beta/metabolism , Trophoblasts/immunology , Trophoblasts/metabolism
15.
Food Microbiol ; 99: 103816, 2021 Oct.
Article En | MEDLINE | ID: mdl-34119101

Protozoan contamination in produce is of growing importance due to their capacity to cause illnesses in consumers of fresh leafy greens. Viability assays are essential to accurately estimate health risk caused by viable parasites that contaminate food. We evaluated the efficacy of reverse transcription quantitative PCR (RT-qPCR), propidium monoazide coupled with (q)PCR, and viability staining using propidium iodide through systematic laboratory spiking experiments for selective detection of viable Cryptosporidium parvum, Giardia enterica, and Toxoplasma gondii. In the presence of only viable protozoa, the RT-qPCR assays could accurately detect two to nine (oo)cysts/g spinach (in 10 g processed). When different proportions of viable and inactivated parasite were spiked, mRNA concentrations correlated with increasing proportions of viable (oo)cysts, although low levels of false-positive mRNA signals were detectable in the presence of high amounts of inactivated protozoa. Our study demonstrated that among the methods tested, RT-qPCR performed more effectively to discriminate viable from inactivated C. parvum, G. enterica and T. gondii on spinach. This application of viability methods on leafy greens can be adopted by the produce industry and regulatory agencies charged with protection of human public health to screen leafy greens for the presence of viable protozoan pathogen contamination.


Cryptosporidium parvum/isolation & purification , Food Parasitology/methods , Giardia/isolation & purification , Spinacia oleracea/parasitology , Toxoplasma/isolation & purification , Animals , Azides/chemistry , Cryptosporidium parvum/chemistry , Cryptosporidium parvum/genetics , Cryptosporidium parvum/growth & development , Food Contamination/analysis , Giardia/chemistry , Giardia/genetics , Giardia/growth & development , Oocysts/chemistry , Oocysts/growth & development , Oocysts/isolation & purification , Plant Leaves/parasitology , Propidium/analogs & derivatives , Propidium/chemistry , Real-Time Polymerase Chain Reaction , Staining and Labeling , Toxoplasma/chemistry , Toxoplasma/genetics , Toxoplasma/growth & development
16.
Cell Chem Biol ; 28(10): 1501-1513.e5, 2021 10 21.
Article En | MEDLINE | ID: mdl-34043961

The intracellular protozoan parasite Toxoplasma gondii must scavenge cholesterol and other lipids from the host to facilitate intracellular growth and replication. Enzymes responsible for neutral lipid synthesis have been identified but there is no evidence for enzymes that catalyze lipolysis of cholesterol esters and esterified lipids. Here, we characterize several T. gondii serine hydrolases with esterase and thioesterase activities that were previously thought to be depalmitoylating enzymes. We find they do not cleave palmitoyl thiol esters but rather hydrolyze short-chain lipid esters. Deletion of one of the hydrolases results in alterations in levels of multiple lipids species. We also identify small-molecule inhibitors of these hydrolases and show that treatment of parasites results in phenotypic defects reminiscent of parasites exposed to excess cholesterol or oleic acid. Together, these data characterize enzymes necessary for processing lipids critical for infection and highlight the potential for targeting parasite hydrolases for therapeutic applications.


Lipid Metabolism/physiology , Protozoan Proteins/metabolism , Serine Endopeptidases/metabolism , Toxoplasma/enzymology , Amino Acid Sequence , Catalytic Domain , Hydrolysis , Kinetics , Phylogeny , Protozoan Proteins/classification , Protozoan Proteins/genetics , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Sequence Alignment , Serine Endopeptidases/classification , Serine Endopeptidases/genetics , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism , Substrate Specificity , Toxoplasma/growth & development , Toxoplasma/physiology
17.
Food Microbiol ; 98: 103792, 2021 Sep.
Article En | MEDLINE | ID: mdl-33875219

To investigate the presence of Cyclospora cayetanensis, Toxoplasma gondii and Echinococcus spp. in fresh produce sold in Italy, 324 locally produced 'ready-to-eat' (RTE) mixed-salad packages belonging to three brands and 324 berries packages (blueberries and blackberries imported from Peru and Mexico, respectively, and raspberries grown in Italy) were purchased at retail. Nine individual packages from each of the six types of fresh produce were collected monthly for one year, and with the same produce pooled, this resulted in a total of 72 pools for the whole year. Using microscopy (FLOTAC), a Cyclospora-like oocyst was detected in a blueberry sample and a taeniid egg was detected in a RTE-salad sample. Molecular tools confirmed these to be C. cayetanensis and Echinococcus multilocularis, respectively. Toxoplasma gondii was not detected in any of the samples. This study shows for the first time in Europe that imported berries on the Italian market may be contaminated with C. cayetanensis and RTE salads grown in Italy with E. multilocularis. The results indicate a new epidemiological scenario and highlight that current management of fresh produce, locally produced or imported, does not ensure products are free from parasite contamination.


Cyclospora/growth & development , Echinococcus multilocularis/growth & development , Fast Foods/parasitology , Food Contamination/analysis , Fruit/parasitology , Animals , Blueberry Plants/parasitology , Cyclospora/genetics , Cyclospora/isolation & purification , Echinococcus multilocularis/genetics , Echinococcus multilocularis/isolation & purification , Italy , Mexico , Oocysts/genetics , Oocysts/isolation & purification , Rubus/parasitology , Toxoplasma/genetics , Toxoplasma/growth & development , Toxoplasma/isolation & purification
18.
Sci Rep ; 11(1): 6787, 2021 03 24.
Article En | MEDLINE | ID: mdl-33762657

Toxoplasma gondii and Plasmodium falciparum parasites both extrude L-lactate, a byproduct of glycolysis. The P. falciparum Formate Nitrite Transporter, PfFNT, mediates L-lactate transport across the plasma membrane of P. falciparum parasites and has been validated as a drug target. The T. gondii genome encodes three FNTs that have been shown to transport L-lactate, and which are proposed to be the targets of several inhibitors of T. gondii proliferation. Here, we show that each of the TgFNTs localize to the T. gondii plasma membrane and are capable of transporting L-lactate across it, with TgFNT1 making the primary contribution to L-lactate transport during the disease-causing lytic cycle of the parasite. We use the Xenopus oocyte expression system to provide direct measurements of L-lactate transport via TgFNT1. We undertake a genetic analysis of the importance of the tgfnt genes for parasite proliferation, and demonstrate that all three tgfnt genes can be disrupted individually and together without affecting the lytic cycle under in vitro culture conditions. Together, our experiments identify the major lactate transporter in the disease causing stage of T. gondii, and reveal that this transporter is not required for parasite proliferation, indicating that TgFNTs are unlikely to be targets for anti-Toxoplasma drugs.


Monocarboxylic Acid Transporters/metabolism , Protozoan Proteins/metabolism , Toxoplasma/metabolism , Animals , Cell Membrane/metabolism , Lactic Acid/metabolism , Monocarboxylic Acid Transporters/genetics , Oocytes/metabolism , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protozoan Proteins/genetics , Toxoplasma/growth & development , Xenopus/growth & development
19.
mBio ; 12(2)2021 03 02.
Article En | MEDLINE | ID: mdl-33653884

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.


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
20.
Nat Rev Microbiol ; 19(7): 467-480, 2021 07.
Article En | MEDLINE | ID: mdl-33627834

Toxoplasma gondii is a parasite that infects a wide range of animals and causes zoonotic infections in humans. Although it normally only results in mild illness in healthy individuals, toxoplasmosis is a common opportunistic infection with high mortality in individuals who are immunocompromised, most commonly due to reactivation of infection in the central nervous system. In the acute phase of infection, interferon-dependent immune responses control rapid parasite expansion and mitigate acute disease symptoms. However, after dissemination the parasite differentiates into semi-dormant cysts that form within muscle cells and neurons, where they persist for life in the infected host. Control of infection in the central nervous system, a compartment of immune privilege, relies on modified immune responses that aim to balance infection control while limiting potential damage due to inflammation. In response to the activation of interferon-mediated pathways, the parasite deploys an array of effector proteins to escape immune clearance and ensure latent survival. Although these pathways are best studied in the laboratory mouse, emerging evidence points to unique mechanisms of control in human toxoplasmosis. In this Review, we explore some of these recent findings that extend our understanding for proliferation, establishment and control of toxoplasmosis in humans.


Toxoplasma/physiology , Toxoplasmosis/parasitology , Animals , Central Nervous System Infections/immunology , Central Nervous System Infections/parasitology , Central Nervous System Infections/pathology , Chronic Disease , Humans , Interferons/immunology , Toxoplasma/growth & development , Toxoplasma/pathogenicity , Toxoplasmosis/drug therapy , Toxoplasmosis/immunology , Virulence
...