Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
PLoS Pathog ; 9(12): e1003779, 2013.
Article in English | MEDLINE | ID: mdl-24367253

ABSTRACT

Most isolates of Toxoplasma from Europe and North America fall into one of three genetically distinct clonal lineages, the type I, II and III lineages. However, in South America these strains are rarely isolated and instead a great variety of other strains are found. T. gondii strains differ widely in a number of phenotypes in mice, such as virulence, persistence, oral infectivity, migratory capacity, induction of cytokine expression and modulation of host gene expression. The outcome of toxoplasmosis in patients is also variable and we hypothesize that, besides host and environmental factors, the genotype of the parasite strain plays a major role. The molecular basis for these differences in pathogenesis, especially in strains other than the clonal lineages, remains largely unexplored. Macrophages play an essential role in the early immune response against T. gondii and are also the cell type preferentially infected in vivo. To determine if non-canonical Toxoplasma strains have unique interactions with the host cell, we infected murine macrophages with 29 different Toxoplasma strains, representing global diversity, and used RNA-sequencing to determine host and parasite transcriptomes. We identified large differences between strains in the expression level of known parasite effectors and large chromosomal structural variation in some strains. We also identified novel strain-specifically regulated host pathways, including the regulation of the type I interferon response by some atypical strains. IFNß production by infected cells was associated with parasite killing, independent of interferon gamma activation, and dependent on endosomal Toll-like receptors in macrophages and the cytoplasmic receptor retinoic acid-inducible gene 1 (RIG-I) in fibroblasts.


Subject(s)
Host-Parasite Interactions/genetics , Macrophages/metabolism , Macrophages/parasitology , Toxoplasma/pathogenicity , Animals , Cells, Cultured , Female , Gene Expression Profiling , Gene Expression Regulation , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Multigene Family , Signal Transduction/genetics
2.
J Biol Chem ; 288(48): 34968-80, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-24129568

ABSTRACT

At least a third of the human population is infected with the intracellular parasite Toxoplasma gondii, which contributes significantly to the disease burden in immunocompromised and neutropenic hosts and causes serious congenital complications when vertically transmitted to the fetus. Genetic analyses have identified the Toxoplasma ROP18 Ser/Thr protein kinase as a major factor mediating acute virulence in mice. ROP18 is secreted into the host cell during the invasion process, and its catalytic activity is required for the acute virulence phenotype. However, its precise molecular function and regulation are not fully understood. We have determined the crystal structure of the ROP18 kinase domain, which is inconsistent with a previously proposed autoinhibitory mechanism of regulation. Furthermore, a sucrose molecule bound to our structure identifies an additional ligand-binding pocket outside of the active site cleft. Mutational analysis confirms an important role for this pocket in virulence.


Subject(s)
Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/genetics , Toxoplasma/pathogenicity , Toxoplasmosis/genetics , Animals , Binding Sites , Crystallography, X-Ray , DNA Mutational Analysis , Humans , Ligands , Mice , Protein Binding , Protein Conformation , Protein Structure, Tertiary/genetics , Protozoan Proteins , Toxoplasma/genetics , Toxoplasmosis/microbiology
3.
Infect Immun ; 81(6): 2156-67, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23545295

ABSTRACT

Toxoplasma gondii transmission between intermediate hosts is dependent on the ingestion of walled cysts formed during the chronic phase of infection. Immediately following consumption, the parasite must ensure survival of the host by preventing adverse inflammatory responses and/or by limiting its own replication. Since the Toxoplasma secreted effectors rhoptry 16 kinase (ROP16) and dense granule 15 (GRA15) activate the JAK-STAT3/6 and NF-κB signaling pathways, respectively, we explored whether a particular combination of these effectors impacted intestinal inflammation and parasite survival in vivo. Here we report that expression of the STAT-activating version of ROP16 in the type II strain (strain II+ROP16I) promotes host resistance to oral infection only in the context of endogenous GRA15 expression. Protection was characterized by a lower intestinal parasite burden and dampened inflammation. Host resistance to the II+ROP16I strain occurred independently of STAT6 and the T cell coinhibitory receptors B7-DC and B7-H1, two receptors that are upregulated by ROP16. In addition, coexpression of ROP16 and GRA15 enhanced parasite susceptibility within tumor necrosis factor alpha/gamma interferon-stimulated macrophages in a STAT3/6-independent manner. Transcriptional profiling of infected STAT3- and STAT6-deficient macrophages and parasitized Peyer's patches from mice orally challenged with strain II+ROP16I suggested that ROP16 activated STAT5 to modulate host gene expression. Consistent with this supposition, the ROP16 kinase induced the sustained phosphorylation and nuclear localization of STAT5 in Toxoplasma-infected cells. In summary, only the combined expression of both GRA15 and ROP16 promoted host resistance to acute oral infection, and Toxoplasma may possibly target the STAT5 signaling pathway to generate protective immunity in the gut.


Subject(s)
Antigens, Protozoan/metabolism , Inflammation/pathology , Intestines/pathology , Protein-Tyrosine Kinases/metabolism , Protozoan Proteins/metabolism , Toxoplasma/enzymology , Toxoplasmosis, Animal/parasitology , Animals , Antigens, Protozoan/genetics , Cytokines/genetics , Cytokines/metabolism , Female , Gene Expression Regulation/physiology , Mice , Mice, Knockout , Peyer's Patches/parasitology , Protein-Tyrosine Kinases/genetics , Protozoan Proteins/genetics , Signal Transduction , Toxoplasmosis, Animal/pathology
4.
BMC Genomics ; 14: 467, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23837824

ABSTRACT

BACKGROUND: Toxoplasma gondii has a largely clonal population in North America and Europe, with types I, II and III clonal lineages accounting for the majority of strains isolated from patients. RH, a particular type I strain, is most frequently used to characterize Toxoplasma biology. However, compared to other type I strains, RH has unique characteristics such as faster growth, increased extracellular survival rate and inability to form orally infectious cysts. Thus, to identify candidate genes that could account for these parasite phenotypic differences, we determined genetic differences and differential parasite gene expression between RH and another type I strain, GT1. Moreover, as differences in host cell modulation could affect Toxoplasma replication in the host, we determined differentially modulated host processes among the type I strains through host transcriptional profiling. RESULTS: Through whole genome sequencing, we identified 1,394 single nucleotide polymorphisms (SNPs) and insertions/deletions (indels) between RH and GT1. These SNPs/indels together with parasite gene expression differences between RH and GT1 were used to identify candidate genes that could account for type I phenotypic differences. A polymorphism in dense granule protein, GRA2, determined RH and GT1 differences in the evasion of the interferon gamma response. In addition, host transcriptional profiling identified that genes regulated by NF-ĸB, such as interleukin (IL)-12p40, were differentially modulated by the different type I strains. We subsequently showed that this difference in NF-ĸB activation was due to polymorphisms in GRA15. Furthermore, we observed that RH, but not other type I strains, recruited phosphorylated IĸBα (a component of the NF-ĸB complex) to the parasitophorous vacuole membrane and this recruitment of p- IĸBα was partially dependent on GRA2. CONCLUSIONS: We identified candidate parasite genes that could be responsible for phenotypic variation among the type I strains through comparative genomics and transcriptomics. We also identified differentially modulated host pathways among the type I strains, and these can serve as a guideline for future studies in examining the phenotypic differences among type I strains.


Subject(s)
Phenotype , Toxoplasma/genetics , Toxoplasma/physiology , Animals , Fibroblasts/parasitology , Gene Expression Regulation , Genes, Protozoan/genetics , HEK293 Cells , Humans , Interleukin-12 Subunit p40/metabolism , Intracellular Membranes/metabolism , Intracellular Membranes/parasitology , Macrophages/metabolism , Macrophages/parasitology , Mice , NF-kappa B/metabolism , Polymorphism, Single Nucleotide , Protein Transport , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Species Specificity , Toxoplasma/metabolism , Vacuoles/metabolism
5.
Cell Host Microbe ; 26(4): 478-492.e8, 2019 10 09.
Article in English | MEDLINE | ID: mdl-31600500

ABSTRACT

Toxoplasma can reach distant organs, especially the brain, leading to a lifelong chronic phase. However, genes involved in related in vivo processes are currently unknown. Here, we use focused CRISPR libraries to identify Toxoplasma genes that affect in vivo fitness. We focus on TgWIP, whose deletion affects Toxoplasma dissemination to distant organs. We show that TgWIP is secreted into the host cell upon invasion and interacts with the host WAVE regulatory complex and SHP2 phosphatase, both of which regulate actin dynamics. TgWIP affects the morphology of dendritic cells and mediates the dissolution of podosomes, which dendritic cells use to adhere to extracellular matrix. TgWIP enhances the motility and transmigration of parasitized dendritic cells, likely explaining its effect on in vivo fitness. Our results provide a framework for systemic identification of Toxoplasma genes with in vivo effects at the site of infection or on dissemination to distant organs, including the brain.


Subject(s)
Cell Movement/physiology , Cytoskeletal Proteins/metabolism , Dendritic Cells/physiology , Protozoan Proteins/genetics , Toxoplasma/genetics , Animals , Cell Line , Clustered Regularly Interspaced Short Palindromic Repeats/genetics , Humans , Mice , Mice, Inbred C57BL , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Toxoplasma/pathogenicity , Virulence Factors/genetics , Wiskott-Aldrich Syndrome Protein Family/metabolism
6.
mBio ; 6(2): e02280, 2015 Feb 24.
Article in English | MEDLINE | ID: mdl-25714710

ABSTRACT

UNLABELLED: The intracellular parasite Toxoplasma gondii infects a wide variety of vertebrate species globally. Infection in most hosts causes a lifelong chronic infection and generates immunological memory responses that protect the host against new infections. In regions where the organism is endemic, multiple exposures to T. gondii likely occur with great frequency, yet little is known about the interaction between a chronically infected host and the parasite strains from these areas. A widely used model to explore secondary infection entails challenge of chronically infected or vaccinated mice with the highly virulent type I RH strain. Here, we show that although vaccinated or chronically infected C57BL/6 mice are protected against the type I RH strain, they are not protected against challenge with most strains prevalent in South America or another type I strain, GT1. Genetic and genomic analyses implicated the parasite-secreted rhoptry effectors ROP5 and ROP18, which antagonize the host's gamma interferon-induced immunity-regulated GTPases (IRGs), as primary requirements for virulence during secondary infection. ROP5 and ROP18 promoted parasite superinfection in the brains of challenged survivors. We hypothesize that superinfection may be an important mechanism to generate T. gondii strain diversity, simply because two parasite strains would be present in a single meal consumed by the feline definitive host. Superinfection may drive the genetic diversity of Toxoplasma strains in South America, where most isolates are IRG resistant, compared to North America, where most strains are IRG susceptible and are derived from a few clonal lineages. In summary, ROP5 and ROP18 promote Toxoplasma virulence during reinfection. IMPORTANCE: Toxoplasma gondii is a widespread parasite of warm-blooded animals and currently infects one-third of the human population. A long-standing assumption in the field is that prior exposure to this parasite protects the host from subsequent reexposure, due to the generation of protective immunological memory. However, this assumption is based on clinical data and mouse models that analyze infections with strains common to Europe infections with strains common to Europe and North America. In contrast, we found that the majority of strains sampled from around the world, in particular those from South America, were able to kill or reinfect the brains of hosts previously exposed to T. gondii. The T. gondii virulence factors ROP5 and ROP18, which inhibit key host effectors that mediate parasite killing, were required for these phenotypes. We speculate that these results underpin clinical observations that pregnant women previously exposed to Toxoplasma can develop congenital infection upon reexposure to South American strains.


Subject(s)
Alleles , Coinfection , Protozoan Proteins/genetics , Superinfection , Toxoplasma/genetics , Toxoplasma/pathogenicity , Toxoplasmosis, Animal/parasitology , Animals , Mice, Inbred C57BL , North America , South America , Virulence
7.
Int J Parasitol ; 44(2): 147-60, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24070999

ABSTRACT

The rhoptries are key secretory organelles from apicomplexan parasites that contain proteins involved in invasion and modulation of the host cell. Some rhoptry proteins are restricted to the posterior bulb (ROPs) and others to the anterior neck (RONs). As many rhoptry proteins have been shown to be key players in Toxoplasma invasion and virulence, it is important to identify, understand and characterise the biological function of the components of the rhoptries. In this report, we identified putative novel rhoptry genes by identifying Toxoplasma genes with similar cyclical expression profiles as known rhoptry protein encoding genes. Using this approach we identified two new rhoptry bulb (ROP47 and ROP48) and one new rhoptry neck protein (RON12). ROP47 is secreted and traffics to the host cell nucleus, RON12 was not detected at the moving junction during invasion. Deletion of ROP47 or ROP48 in a type II strain did not show major influence in in vitro growth or virulence in mice.


Subject(s)
Protozoan Proteins/metabolism , Toxoplasma/metabolism , Toxoplasmosis, Animal/parasitology , Animals , Female , Gene Expression Regulation , Mice , Mice, Inbred C57BL , Protozoan Proteins/genetics , Toxoplasma/chemistry , Toxoplasma/genetics
8.
J Exp Med ; 208(1): 195-212, 2011 Jan 17.
Article in English | MEDLINE | ID: mdl-21199955

ABSTRACT

NF-κB is an integral component of the immune response to Toxoplasma gondii. Although evidence exists that T. gondii can directly modulate the NF-κB pathway, the parasite-derived effectors involved are unknown. We determined that type II strains of T. gondii activate more NF-κB than type I or type III strains, and using forward genetics we found that this difference is a result of the polymorphic protein GRA15, a novel dense granule protein which T. gondii secretes into the host cell upon invasion. A GRA15-deficient type II strain has a severe defect in both NF-κB nuclear translocation and NF-κB-mediated transcription. Furthermore, human cells expressing type II GRA15 also activate NF-κB, demonstrating that GRA15 alone is sufficient for NF-κB activation. Along with the rhoptry protein ROP16, GRA15 is responsible for a large part of the strain differences in the induction of IL-12 secretion by infected mouse macrophages. In vivo bioluminescent imaging showed that a GRA15-deficient type II strain grows faster compared with wild-type, most likely through its reduced induction of IFN-γ. These results show for the first time that a dense granule protein can modulate host signaling pathways, and dense granule proteins can therefore join rhoptry proteins in T. gondii's host cell-modifying arsenal.


Subject(s)
Protozoan Proteins/metabolism , Signal Transduction , Toxoplasma/metabolism , Transcription Factor RelA/metabolism , Animals , Cells, Cultured , Gene Expression Regulation , Genome, Protozoan , Humans , Interleukin-12/biosynthesis , Mice , Mice, Knockout , Protein Transport , Toxoplasma/genetics , Transcription Factor RelA/deficiency , Transcription, Genetic
9.
PLoS One ; 6(9): e24434, 2011.
Article in English | MEDLINE | ID: mdl-21931713

ABSTRACT

IFN-γ is a major cytokine that mediates resistance against the intracellular parasite Toxoplasma gondii. The p65 guanylate-binding proteins (GBPs) are strongly induced by IFN-γ. We studied the behavior of murine GBP1 (mGBP1) upon infection with T. gondii in vitro and confirmed that IFN-γ-dependent re-localization of mGBP1 to the parasitophorous vacuole (PV) correlates with the virulence type of the parasite. We identified three parasitic factors, ROP16, ROP18, and GRA15 that determine strain-specific accumulation of mGBP1 on the PV. These highly polymorphic proteins are held responsible for a large part of the strain-specific differences in virulence. Therefore, our data suggest that virulence of T. gondii in animals may rely in part on recognition by GBPs. However, phagosomes or vacuoles containing Trypanosoma cruzi did not recruit mGBP1. Co-immunoprecipitation revealed mGBP2, mGBP4, and mGBP5 as binding partners of mGBP1. Indeed, mGBP2 and mGBP5 co-localize with mGBP1 in T. gondii-infected cells. T. gondii thus elicits a cell-autonomous immune response in mice with GBPs involved. Three parasitic virulence factors and unknown IFN-γ-dependent host factors regulate this complex process. Depending on the virulence of the strains involved, numerous GBPs are brought to the PV as part of a large, multimeric structure to combat T. gondii.


Subject(s)
GTP-Binding Proteins/physiology , Gene Expression Regulation , Toxoplasma/metabolism , Vacuoles/parasitology , Animals , Antigens, Protozoan/metabolism , Fibroblasts/cytology , GTP-Binding Proteins/metabolism , Guanosine Triphosphate/metabolism , Humans , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Phagosomes/metabolism , Polymorphism, Genetic , Protein Binding , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Protozoan Proteins/metabolism , Vacuoles/metabolism
10.
Planta ; 227(5): 991-1000, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18183417

ABSTRACT

Proper cell morphogenesis is dependent on the establishment and expression of cellular polarity. In the fucoid zygote, cell shape is critical for establishing the developmental pattern of the adult, and is achieved by guiding insertion of new membrane and wall to the rhizoid tip. Selection and growth of the appropriate tip site are accompanied by formation of dynamic actin arrays associated with the actin-nucleating Arp2/3 complex. In eukaryotes, a major pathway for activation of the Arp2/3 complex is via the Rho family GTPase, Rac1, which stimulates the Scar/WAVE complex. To determine whether Rac1 controls actin nucleation in Silvetia compressa (J. Agardh) E. Serrao, T. O. Cho, S. M. Boo et Brawley, we tested the effects of the Rac1-specific inhibitory compound, NSC23766, on actin dependent processes and on actin arrays. We found that NSC23766 disrupted polar secretion of adhesive, polarization of endomembranes, and tip-focused growth in the rhizoid. Similarly, NSC23766 altered actin and Arp2 localization in the growing rhizoid. In contrast, NSC23766 had no effect on selection of the growth site or on cytokinesis. These data suggest that Rac1 participates in nucleation of specific actin arrays in the developing zygote.


Subject(s)
Aminoquinolines/pharmacology , Phaeophyceae/drug effects , Pyrimidines/pharmacology , rac1 GTP-Binding Protein/antagonists & inhibitors , Actins/metabolism , Cell Membrane/drug effects , Cell Membrane/metabolism , Phaeophyceae/growth & development , Phaeophyceae/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL