Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 19(3): e0300764, 2024.
Article in English | MEDLINE | ID: mdl-38551902

ABSTRACT

Toxoplasma gondii is an intracellular parasite that establishes a long-term infection in the brain of many warm-blooded hosts, including humans and rodents. Like all obligate intracellular microbes, Toxoplasma uses many effector proteins to manipulate the host cell to ensure parasite survival. While some of these effector proteins are universal to all Toxoplasma strains, some are polymorphic between Toxoplasma strains. One such polymorphic effector is GRA15. The gra15 allele carried by type II strains activates host NF-κB signaling, leading to the release of cytokines such as IL-12, TNF, and IL-1ß from immune cells infected with type II parasites. Prior work also suggested that GRA15 promotes early host control of parasites in vivo, but the effect of GRA15 on parasite persistence in the brain and the peripheral immune response has not been well defined. For this reason, we sought to address this gap by generating a new IIΔgra15 strain and comparing outcomes at 3 weeks post infection between WT and IIΔgra15 infected mice. We found that the brain parasite burden and the number of macrophages/microglia and T cells in the brain did not differ between WT and IIΔgra15 infected mice. In addition, while IIΔgra15 infected mice had a lower number and frequency of splenic M1-like macrophages and frequency of PD-1+ CTLA-4+ CD4+ T cells and NK cells compared to WT infected mice, the IFN-γ+ CD4 and CD8 T cell populations were equivalent. In summary, our results suggest that in vivo GRA15 may have a subtle effect on the peripheral immune response, but this effect is not strong enough to alter brain parasite burden or parenchymal immune cell number at 3 weeks post infection.


Subject(s)
Toxoplasma , Humans , Animals , Mice , Protozoan Proteins/metabolism , Signal Transduction , Cytokines/metabolism , NF-kappa B/metabolism
2.
PLoS Pathog ; 19(4): e1011347, 2023 04.
Article in English | MEDLINE | ID: mdl-37068104

ABSTRACT

Toxoplasma gondii establishes a long-lived latent infection in the central nervous system (CNS) of its hosts. Reactivation in immunocompromised individuals can lead to life threatening disease. Latent infection is driven by the ability of the parasite to convert from the acute-stage tachyzoite to the latent-stage bradyzoite which resides in long-lived intracellular cysts. While much work has focused on the parasitic factors that drive cyst development, the host factors that influence encystment are not well defined. Here we show that a polymorphic secreted parasite kinase (ROP16), that phosphorylates host cell proteins, mediates efficient encystment of T. gondii in a stress-induced model of encystment and primary neuronal cell cultures (PNCs) in a strain-specific manner. Using short-hairpin RNA (shRNA) knockdowns in human foreskin fibroblasts (HFFs) and PNCs from transgenic mice, we determined that ROP16's cyst enhancing abilities are mediated, in part, by phosphorylation-and therefore activation-of the host cell transcription factor STAT6. To test the role of STAT6 in vivo, we infected wild-type (WT) and STAT6KO mice, finding that, compared to WT mice, STAT6KO mice have a decrease in CNS cyst burden but not overall parasite burden or dissemination to the CNS. Finally, we found a similar ROP16-dependent encystment defect in human pluripotent stem cell-derived neurons. Together, these findings identify a host cell factor (STAT6) that T. gondii manipulates in a strain-specific manner to generate a favorable encystment environment.


Subject(s)
Toxoplasma , Mice , Animals , Humans , Toxoplasma/physiology , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Phosphorylation , Central Nervous System/metabolism , Gene Expression Regulation , STAT6 Transcription Factor/metabolism
3.
Nat Commun ; 13(1): 4605, 2022 08 08.
Article in English | MEDLINE | ID: mdl-35941154

ABSTRACT

Dogma holds that Toxoplasma gondii persists in neurons because neurons cannot clear intracellular parasites, even with IFN-γ stimulation. As several recent studies questioned this idea, here we use primary murine neuronal cultures from wild type and transgenic mice in combination with IFN-γ stimulation and parental and transgenic parasites to reassess IFN-γ dependent neuronal clearance of intracellular parasites. We find that neurons respond to IFN-γ and that a subset of neurons clear intracellular parasites via immunity regulated GTPases. Whole neuron reconstructions from mice infected with parasites that trigger neuron GFP expression only after full invasion reveal that ~50% of these T. gondii-invaded neurons no longer harbor parasites. Finally, IFN-γ stimulated human pluripotent stem cell derived neurons show an ~50% decrease in parasite infection rate when compared to unstimulated cultures. This work highlights the capability of human and murine neurons to mount cytokine-dependent anti-T. gondii defense mechanisms in vitro and in vivo.


Subject(s)
Parasites , Toxoplasma , Animals , GTP Phosphohydrolases/metabolism , Humans , Interferon-gamma/metabolism , Mice , Neurons/metabolism , Parasites/metabolism , Toxoplasma/metabolism
4.
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
5.
Cell ; 180(2): 216-218, 2020 01 23.
Article in English | MEDLINE | ID: mdl-31978340

ABSTRACT

Microbes that cause persistent infections (e.g., herpes viruses) do so by switching from fast-growing lytic states to slow-growing latent states. Waldman et al. have identified a single transcription factor that governs the switch between the lytic and latent forms of Toxoplasma gondii, a parasite that causes a persistent brain infection.


Subject(s)
Toxoplasma , Brain , Cell Differentiation , Gene Expression Regulation , Transcription Factors
6.
J Exp Med ; 217(3)2020 03 02.
Article in English | MEDLINE | ID: mdl-31961916

ABSTRACT

The ability of Toxoplasma gondii to inject the rhoptry kinase ROP16 into host cells results in the activation of the transcription factors STAT3 and STAT6, but it is unclear how these events impact infection. Here, parasites that inject Cre-recombinase with rhoptry proteins were used to distinguish infected macrophages from those only injected with parasite proteins. Transcriptional profiling revealed that injection of rhoptry proteins alone was sufficient to induce an M2 phenotype that is dependent on STAT3 and STAT6, but only infected cells displayed reduced expression of genes associated with antimicrobial activity and protective immunity. In vivo, the absence of STAT3 or STAT6 improved parasite control, while the loss of ROP16 resulted in a marked reduction in parasite numbers and heightened parasite-specific T cell responses. Thus, ROP16 is a virulence factor that can act in cis and trans to promote M2 programs and which limits the magnitude of parasite-specific T cell responses.


Subject(s)
Protein-Tyrosine Kinases/immunology , Protozoan Proteins/immunology , T-Lymphocytes/immunology , Toxoplasma/immunology , Virulence Factors/immunology , Animals , Macrophages/immunology , Mice , Mice, Inbred C57BL , STAT3 Transcription Factor/immunology , STAT6 Transcription Factor/immunology
7.
PLoS Pathog ; 15(10): e1007856, 2019 10.
Article in English | MEDLINE | ID: mdl-31648279

ABSTRACT

Toxoplasma gondii is an intracellular parasite that persistently infects the CNS and that has genetically distinct strains which provoke different acute immune responses. How differences in the acute immune response affect the CNS immune response is unknown. To address this question, we used two persistent Toxoplasma strains (type II and type III) and examined the CNS immune response at 21 days post infection (dpi). Contrary to acute infection studies, type III-infected mice had higher numbers of total CNS T cells and macrophages/microglia but fewer alternatively activated macrophages (M2s) and regulatory T cells (Tregs) than type II-infected mice. By profiling splenocytes at 5, 10, and 21 dpi, we determined that at 5 dpi type III-infected mice had more M2s while type II-infected mice had more pro-inflammatory macrophages and that these responses flipped over time. To test how these early differences influence the CNS immune response, we engineered the type III strain to lack ROP16 (IIIΔrop16), the polymorphic effector protein that drives the early type III-associated M2 response. IIIΔrop16-infected mice showed a type II-like neuroinflammatory response with fewer infiltrating T cells and macrophages/microglia and more M2s and an unexpectedly low CNS parasite burden. At 5 dpi, IIIΔrop16-infected mice showed a mixed inflammatory response with more pro-inflammatory macrophages, M2s, T effector cells, and Tregs, and decreased rates of infection of peritoneal exudative cells (PECs). These data suggested that type III parasites need the early ROP16-associated M2 response to avoid clearance, possibly by the Immunity-Related GTPases (IRGs), which are IFN-γ- dependent proteins essential for murine defenses against Toxoplasma. To test this possibility, we infected IRG-deficient mice and found that IIIΔrop16 parasites now maintained parental levels of PECs infection. Collectively, these studies suggest that, for the type III strain, rop16III plays a key role in parasite persistence and influences the subacute CNS immune response.


Subject(s)
Central Nervous System/immunology , Macrophages/immunology , Protein-Tyrosine Kinases/immunology , Protozoan Proteins/immunology , T-Lymphocytes/immunology , Toxoplasma/immunology , Toxoplasmosis, Animal/immunology , Animals , Central Nervous System/parasitology , GTP Phosphohydrolases/genetics , Mice , Mice, Knockout , Microglia/metabolism , Protein-Tyrosine Kinases/genetics , Protozoan Proteins/genetics , Toxoplasma/classification , Toxoplasma/genetics
8.
Curr Biol ; 28(14): R770-R771, 2018 07 23.
Article in English | MEDLINE | ID: mdl-30040931

ABSTRACT

Putting cat lovers on notice: Toxoplasma gondii is here to control your brain…or is it? Kochanowsky and Koshy shine the spotlight on this notorious pathogen.


Subject(s)
Life History Traits , Toxoplasma/cytology , Toxoplasma/physiology , Toxoplasmosis, Animal/parasitology , Toxoplasmosis/parasitology , Animals , Host-Parasite Interactions , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...