Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Sci Rep ; 14(1): 10433, 2024 05 07.
Article in English | MEDLINE | ID: mdl-38714696

ABSTRACT

Toxoplasma gondii (T. gondii) is a protozoan parasite that infects approximately one-third of the global human population, often leading to chronic infection. While acute T. gondii infection can cause neural damage in the central nervous system and result in toxoplasmic encephalitis, the consequences of T. gondii chronic infection (TCI) are generally asymptomatic. However, emerging evidence suggests that TCI may be linked to behavioral changes or mental disorders in hosts. Astrocyte polarization, particularly the A1 subtype associated with neuronal apoptosis, has been identified in various neurodegenerative diseases. Nevertheless, the role of astrocyte polarization in TCI still needs to be better understood. This study aimed to establish a mouse model of chronic TCI and examine the transcription and expression levels of glial fibrillary acidic protein (GFAP), C3, C1q, IL-1α, and TNF-α in the brain tissues of the mice. Quantitative real-time PCR (qRT-PCR), enzyme-linked immunosorbent assay, and Western blotting were employed to assess these levels. Additionally, the expression level of the A1 astrocyte-specific marker C3 was evaluated using indirect fluorescent assay (IFA). In mice with TCI, the transcriptional and expression levels of the inflammatory factors C1q, IL-1α, and TNF-α followed an up-down-up pattern, although they remained elevated compared to the control group. These findings suggest a potential association between astrocyte polarization towards the A1 subtype and synchronized changes in these three inflammatory mediators. Furthermore, immunofluorescence assay (IFA) revealed a significant increase in the A1 astrocytes (GFAP+C3+) proportion in TCI mice. This study provides evidence that TCI can induce astrocyte polarization, a biological process that may be influenced by changes in the levels of three inflammatory factors: C1q, IL-1α, and TNF-α. Additionally, the release of neurotoxic substances by A1 astrocytes may be associated with the development of TCI.


Subject(s)
Astrocytes , Brain , Toxoplasma , Animals , Astrocytes/metabolism , Astrocytes/parasitology , Astrocytes/pathology , Mice , Toxoplasma/pathogenicity , Toxoplasma/physiology , Brain/parasitology , Brain/metabolism , Brain/pathology , Disease Models, Animal , Female , Chronic Disease , Cell Polarity , Glial Fibrillary Acidic Protein/metabolism , Glial Fibrillary Acidic Protein/genetics , Toxoplasmosis/metabolism , Toxoplasmosis/parasitology , Toxoplasmosis/pathology , Tumor Necrosis Factor-alpha/metabolism , Toxoplasmosis, Cerebral/parasitology , Toxoplasmosis, Cerebral/pathology , Toxoplasmosis, Cerebral/metabolism
2.
PLoS Pathog ; 18(9): e1010637, 2022 09.
Article in English | MEDLINE | ID: mdl-36067217

ABSTRACT

Toxoplasma gondii is a ubiquitous intracellular protozoan parasite that establishes a life-long chronic infection largely restricted to the central nervous system (CNS). Constant immune pressure, notably IFN-γ-STAT1 signaling, is required for preventing fatal pathology during T. gondii infection. Here, we report that abrogation of STAT1 signaling in microglia, the resident immune cells of the CNS, is sufficient to induce a loss of parasite control in the CNS and susceptibility to toxoplasmic encephalitis during the early stages of chronic infection. Using a microglia-specific genetic labeling and targeting system that discriminates microglia from blood-derived myeloid cells that infiltrate the brain during infection, we find that, contrary to previous in vitro reports, microglia do not express inducible nitric-oxide synthase (iNOS) during T. gondii infection in vivo. Instead, transcriptomic analyses of microglia reveal that STAT1 regulates both (i) a transcriptional shift from homeostatic to "disease-associated microglia" (DAM) phenotype conserved across several neuroinflammatory models, including T. gondii infection, and (ii) the expression of anti-parasitic cytosolic molecules that are required for eliminating T. gondii in a cell-intrinsic manner. Further, genetic deletion of Stat1 from microglia during T. gondii challenge leads to fatal pathology despite largely equivalent or enhanced immune effector functions displayed by brain-infiltrating immune populations. Finally, we show that microglial STAT1-deficiency results in the overrepresentation of the highly replicative, lytic tachyzoite form of T. gondii, relative to its quiescent, semi-dormant bradyzoite form typical of chronic CNS infection. Our data suggest an overall protective role of CNS-resident microglia against T. gondii infection, illuminating (i) general mechanisms of CNS-specific immunity to infection (ii) and a clear role for IFN-STAT1 signaling in regulating a microglial activation phenotype observed across diverse neuroinflammatory disease states.


Subject(s)
Encephalitis , STAT1 Transcription Factor , Toxoplasma , Toxoplasmosis, Cerebral , Animals , Brain/pathology , Encephalitis/metabolism , Encephalitis/pathology , Mice , Microglia/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Toxoplasma/metabolism , Toxoplasmosis, Cerebral/metabolism
3.
Molecules ; 27(17)2022 Aug 30.
Article in English | MEDLINE | ID: mdl-36080339

ABSTRACT

BACKGROUND: Toxoplasma gondii, an intracellular protozoan parasite, exists in the host brain as cysts, which can result in Toxoplasmic Encephalitis (TE) and neurological diseases. However, few studies have been conducted on TE, particularly on how to prevent it. Previous proteomics studies have showed that the expression of C3 in rat brains was up-regulated after T. gondii infection. METHODS: In this study, we used T. gondii to infect mice and bEnd 3 cells to confirm the relation between T. gondii and the expression of C3. BEnd3 cells membrane proteins which directly interacted with C3a were screened by pull down. Finally, animal behavior experiments were conducted to compare the differences in the inhibitory ability of TE by four chemotherapeutic compounds (SB290157, CVF, NSC23766, and Anxa1). RESULTS: All chemotherapeutic compounds in this study can inhibit TE and cognitive behavior in the host. However, Anxa 1 is the most suitable material to inhibit mice TE. CONCLUSION: T. gondii infection promotes TE by promoting host C3 production. Anxa1 was selected as the most appropriate material to prevent TE among four chemotherapeutic compounds closely related to C3.


Subject(s)
Toxoplasma , Toxoplasmosis, Cerebral , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Mice , Proteomics , Toxoplasmosis, Cerebral/drug therapy , Toxoplasmosis, Cerebral/metabolism , Toxoplasmosis, Cerebral/parasitology
4.
J Neuroinflammation ; 19(1): 17, 2022 Jan 13.
Article in English | MEDLINE | ID: mdl-35027063

ABSTRACT

BACKGROUND: Toxoplasma gondii (T. gondii) is a highly successful parasite being able to cross all biological barriers of the body, finally reaching the central nervous system (CNS). Previous studies have highlighted the critical involvement of the blood-brain barrier (BBB) during T. gondii invasion and development of subsequent neuroinflammation. Still, the potential contribution of the choroid plexus (CP), the main structure forming the blood-cerebrospinal fluid (CSF) barrier (BCSFB) have not been addressed. METHODS: To investigate T. gondii invasion at the onset of neuroinflammation, the CP and brain microvessels (BMV) were isolated and analyzed for parasite burden. Additionally, immuno-stained brain sections and three-dimensional whole mount preparations were evaluated for parasite localization and morphological alterations. Activation of choroidal and brain endothelial cells were characterized by flow cytometry. To evaluate the impact of early immune responses on CP and BMV, expression levels of inflammatory mediators, tight junctions (TJ) and matrix metalloproteinases (MMPs) were quantified. Additionally, FITC-dextran was applied to determine infection-related changes in BCSFB permeability. Finally, the response of primary CP epithelial cells to T. gondii parasites was tested in vitro. RESULTS: Here we revealed that endothelial cells in the CP are initially infected by T. gondii, and become activated prior to BBB endothelial cells indicated by MHCII upregulation. Additionally, CP elicited early local immune response with upregulation of IFN-γ, TNF, IL-6, host-defence factors as well as swift expression of CXCL9 chemokine, when compared to the BMV. Consequently, we uncovered distinct TJ disturbances of claudins, associated with upregulation of MMP-8 and MMP-13 expression in infected CP in vivo, which was confirmed by in vitro infection of primary CP epithelial cells. Notably, we detected early barrier damage and functional loss by increased BCSFB permeability to FITC-dextran in vivo, which was extended over the infection course. CONCLUSIONS: Altogether, our data reveal a close interaction between T. gondii infection at the CP and the impairment of the BCSFB function indicating that infection-related neuroinflammation is initiated in the CP.


Subject(s)
Choroid Plexus , Toxoplasmosis, Cerebral , Blood-Brain Barrier/metabolism , Choroid Plexus/metabolism , Endothelial Cells , Humans , Immunity , Toxoplasmosis, Cerebral/metabolism
5.
Front Immunol ; 12: 681242, 2021.
Article in English | MEDLINE | ID: mdl-34367142

ABSTRACT

Toxoplasma gondii is an obligate intracellular parasite capable of establishing persistent infection within the host brain and inducing severe neuropathology. Peptides are important native molecules responsible for a wide range of biological functions within the central nervous system. However, peptidome profiling in host brain during T. gondii infection has never been investigated. Using a label-free peptidomics approach (LC-MS/MS), we identified a total of 2,735 endogenous peptides from acutely infected, chronically infected and control brain samples following T. gondii infection. Quantitative analysis revealed 478 and 344 significantly differentially expressed peptides (DEPs) in the acute and chronic infection stages, respectively. Functional analysis of DEPs by Gene Ontology suggested these DEPs mainly originated from cell part and took part in cellular process. We also identified three novel neuropeptides derived from the precursor protein cholecystokinin. These results demonstrated the usefulness of quantitative peptidomics in determining bioactive peptides and elucidating their functions in the regulation of behavior modification during T. gondii infection.


Subject(s)
Brain/metabolism , Brain/parasitology , Neuropeptides/metabolism , Proteomics , Toxoplasma , Toxoplasmosis, Cerebral/metabolism , Toxoplasmosis, Cerebral/parasitology , Animals , Brain/pathology , Chromatography, Liquid , Computational Biology/methods , Female , Host-Parasite Interactions , Immunohistochemistry , Mice , Proteomics/methods , Tandem Mass Spectrometry , Toxoplasmosis, Animal , Toxoplasmosis, Cerebral/pathology
6.
Front Immunol ; 12: 619465, 2021.
Article in English | MEDLINE | ID: mdl-33968021

ABSTRACT

Cell survival and function critically relies on the fine-tuned balance of protein synthesis and degradation. In the steady state, the standard proteasome is sufficient to maintain this proteostasis. However, upon inflammation, the sharp increase in protein production requires additional mechanisms to limit protein-associated cellular stress. Under inflammatory conditions and the release of interferons, the immunoproteasome (IP) is induced to support protein processing and recycling. In antigen-presenting cells constitutively expressing IPs, inflammation-related mechanisms contribute to the formation of MHC class I/II-peptide complexes, which are required for the induction of T cell responses. The control of Toxoplasma gondii infection relies on Interferon-γ (IFNγ)-related T cell responses. Whether and how the IP affects the course of anti-parasitic T cell responses along the infection as well as inflammation of the central nervous system is still unknown. To answer this question we used triple knockout (TKO) mice lacking the 3 catalytic subunits of the immunoproteasome (ß1i/LMP2, ß2i/MECL-1 and ß5i/LMP7). Here we show that the numbers of dendritic cells, monocytes and CD8+ T cells were reduced in Toxoplasma gondii-infected TKO mice. Furthermore, impaired IFNγ, TNF and iNOS production was accompanied by dysregulated chemokine expression and altered immune cell recruitment to the brain. T cell differentiation was altered, apoptosis rates of microglia and monocytes were elevated and STAT3 downstream signaling was diminished. Consequently, anti-parasitic immune responses were impaired in TKO mice leading to elevated T. gondii burden and prolonged neuroinflammation. In summary we provide evidence for a critical role of the IP subunits ß1i/LMP2, ß2i/MECL-1 and ß5i/LMP7 for the control of cerebral Toxoplasma gondii infection and subsequent neuroinflammation.


Subject(s)
Cysteine Endopeptidases/metabolism , Immunomodulation , Proteasome Endopeptidase Complex/metabolism , Toxoplasmosis, Cerebral/etiology , Toxoplasmosis, Cerebral/metabolism , Animals , Apoptosis , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Mice , Signal Transduction , Toxoplasma
7.
Sci Rep ; 11(1): 4549, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33633185

ABSTRACT

Central nervous system (CNS) injury and infection can result in profound tissue remodeling in the brain, the mechanism and purpose of which is poorly understood. Infection with the protozoan parasite Toxoplasma gondii causes chronic infection and inflammation in the brain parenchyma. Control of parasite replication requires the continuous presence of IFNγ-producing T cells to keep T. gondii in its slowly replicating cyst form. During infection, a network of extracellular matrix fibers, revealed using multiphoton microscopy, forms in the brain. The origin and composition of these structures are unknown but the fibers have been observed to act as a substrate for migrating T cells. In this study, we show a critical regulator of extracellular matrix (ECM) remodeling, Secreted Protein, Acidic, Rich in Cysteine (SPARC), is upregulated in the brain during the early phases of infection in the frontal cortex. In the absence of SPARC, a reduced and disordered fibrous network, increased parasite burden, and reduced antigen-specific T cell entry into the brain points to a role for SPARC in T cell recruitment to and migration within the brain. We also report SPARC can directly bind to CCR7 ligands CCL19 and CCL21 but not CXCL10, and enhance migration toward a chemokine gradient. Measurement of T cell behavior points to tissue remodeling being important for access of immune cells to the brain and facilitating cellular locomotion. Together, these data identify SPARC as an important regulatory component of immune cell trafficking and access to the inflamed CNS.


Subject(s)
Extracellular Matrix/metabolism , Osteonectin/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Toxoplasma/physiology , Toxoplasmosis, Cerebral/etiology , Toxoplasmosis, Cerebral/metabolism , Animals , Antigens, Protozoan/immunology , Biomarkers , Brain/blood supply , Brain/immunology , Brain/metabolism , Brain/parasitology , Cell Movement/immunology , Chemokine CCL21/metabolism , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Gene Expression Regulation , Host-Parasite Interactions/immunology , Mice , Mice, Knockout , Neurons/metabolism , Osteonectin/genetics , Protein Binding , Receptors, CCR7
8.
J Ethnopharmacol ; 267: 113525, 2021 Mar 01.
Article in English | MEDLINE | ID: mdl-33129946

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Hypericum perforatum L. has been widely used as a natural antidepressant. However, it is unknown whether it is effective in treating infection-induced neuropsychiatric disorders. AIM OF THE STUDY: In order to evaluate the effectiveness of H. perforatum against infection with neurotropic parasite Toxoplasma gondii, which has been linked to neuropsychiatric disorders, this study investigated the anti-Toxoplasma activity using in vitro models. MATERIALS AND METHODS: Dried alcoholic extracts were prepared from three Hypericum species: H. perforatum, H. erectum, and H. ascyron. H. perforatum extract was further separated by solvent-partitioning. Hyperforin and hypericin levels in the extracts and fractions were analyzed by high resolution LC-MS. Anti-Toxoplasma activities were tested in vitro, using cell lines (Vero and Raw264), murine primary mixed glia, and primary neuron-glia. Toxoplasma proliferation and stage conversion were analyzed by qPCR. Infection-induced damages to the host cells were analyzed by Sulforhodamine B cytotoxicity assay (Vero) and immunofluorescent microscopy (neurons). Infection-induced inflammatory responses in glial cells were analysed by qPCR and immunofluorescent microscopy. RESULTS: Hyperforin was identified only in H. perforatum among the three tested species, whereas hypericin was present in H. perforatum and H. erectum. H. perforatum extract and hyperforin-enriched fraction, as well as hyperforin, exhibited significant anti-Toxoplasma property as well as inhibitory activity against infection-induced inflammatory responses in glial cells. In addition, H. perforatum-derived hyperforin-enriched fraction restored neuro-supportive environment in mixed neuron-glia culture. CONCLUSIONS: H. perforatum and its major constituent hyperforin are promising anti-Toxoplasma agents that could potentially protect neurons and glial cells against infection-induced damages. Further study is warranted to establish in vivo efficacy.


Subject(s)
Coccidiostats/pharmacology , Hypericum , Neuroglia/drug effects , Neuroprotective Agents/pharmacology , Phloroglucinol/analogs & derivatives , Plant Extracts/pharmacology , Terpenes/pharmacology , Toxoplasma/drug effects , Toxoplasmosis, Cerebral/drug therapy , Animals , Chlorocebus aethiops , Coccidiostats/isolation & purification , Cytokines , Hypericum/chemistry , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Neuroglia/metabolism , Neuroglia/parasitology , Neuroglia/pathology , Neuroprotective Agents/isolation & purification , Phloroglucinol/isolation & purification , Phloroglucinol/pharmacology , Plant Extracts/isolation & purification , RAW 264.7 Cells , Terpenes/isolation & purification , Toxoplasma/growth & development , Toxoplasmosis, Cerebral/metabolism , Toxoplasmosis, Cerebral/parasitology , Toxoplasmosis, Cerebral/pathology , Vero Cells
9.
PLoS Pathog ; 16(10): e1009027, 2020 10.
Article in English | MEDLINE | ID: mdl-33108405

ABSTRACT

It is of great interest to understand how invading pathogens are sensed within the brain, a tissue with unique challenges to mounting an immune response. The eukaryotic parasite Toxoplasma gondii colonizes the brain of its hosts, and initiates robust immune cell recruitment, but little is known about pattern recognition of T. gondii within brain tissue. The host damage signal IL-33 is one protein that has been implicated in control of chronic T. gondii infection, but, like many other pattern recognition pathways, IL-33 can signal peripherally, and the specific impact of IL-33 signaling within the brain is unclear. Here, we show that IL-33 is expressed by oligodendrocytes and astrocytes during T. gondii infection, is released locally into the cerebrospinal fluid of T. gondii-infected animals, and is required for control of infection. IL-33 signaling promotes chemokine expression within brain tissue and is required for the recruitment and/or maintenance of blood-derived anti-parasitic immune cells, including proliferating, IFN-γ-expressing T cells and iNOS-expressing monocytes. Importantly, we find that the beneficial effects of IL-33 during chronic infection are not a result of signaling on infiltrating immune cells, but rather on radio-resistant responders, and specifically, astrocytes. Mice with IL-33 receptor-deficient astrocytes fail to mount an adequate adaptive immune response in the CNS to control parasite burden-demonstrating, genetically, that astrocytes can directly respond to IL-33 in vivo. Together, these results indicate a brain-specific mechanism by which IL-33 is released locally, and sensed locally, to engage the peripheral immune system in controlling a pathogen.


Subject(s)
Astrocytes/immunology , Interleukin-33/immunology , Toxoplasmosis, Cerebral/immunology , Adult , Animals , Astrocytes/metabolism , Astrocytes/physiology , Brain/metabolism , Female , Humans , Immunity , Interferon-gamma/immunology , Interleukin-33/metabolism , Male , Mice , Mice, Inbred C57BL , Monocytes/immunology , Signal Transduction , Toxoplasma/metabolism , Toxoplasma/parasitology , Toxoplasmosis/metabolism , Toxoplasmosis, Cerebral/metabolism
10.
Brain Behav Immun ; 89: 480-490, 2020 10.
Article in English | MEDLINE | ID: mdl-32717399

ABSTRACT

The incidence of infectious diseases affecting the central nervous system (CNS) has been increasing over the last several years. Among the reasons for the expansion of these diseases and the appearance of new neuropathogens are globalization, global warming, and the increased proximity between humans and wild animals due to human activities such as deforestation. Neurotropism affecting normal brain function is shared by organisms such as viruses, bacteria, fungi, and parasites. Neuroinfections caused by these agents activate immune responses, inducing neuroinflammation, excitotoxicity, and neurodegeneration. Purinergic signaling is an evolutionarily conserved signaling pathway associated with these neuropathologies. During neuroinfections, host cells release ATP as an extracellular danger signal with pro-inflammatory activities. ATP is metabolized to its derivatives by ectonucleotidases such as CD39 and CD73; ATP and its metabolites modulate neuronal and immune mechanisms through P1 and P2 purinergic receptors that are involved in pathophysiological mechanisms of neuroinfections. In this review we discuss the beneficial or deleterious effects of various components of the purinergic signaling pathway in infectious diseases that affect the CNS, including human immunodeficiency virus (HIV-1) infection, herpes simplex virus type 1 (HSV-1) infection, bacterial meningitis, sepsis, cryptococcosis, toxoplasmosis, and malaria. We also provide a description of this signaling pathway in emerging viral infections with neurological implications such as Zika and SARS-CoV-2.


Subject(s)
Central Nervous System Infections/metabolism , Receptors, Purinergic P1/metabolism , Receptors, Purinergic P2X/metabolism , Receptors, Purinergic P2Y/metabolism , AIDS Dementia Complex/metabolism , Betacoronavirus , COVID-19 , Coronavirus Infections/metabolism , Encephalitis, Herpes Simplex/metabolism , Humans , Malaria/metabolism , Meningitis, Bacterial/metabolism , Meningitis, Cryptococcal/metabolism , Pandemics , Pneumonia, Viral/metabolism , SARS-CoV-2 , Sepsis/metabolism , Signal Transduction , Toxoplasmosis, Cerebral/metabolism , Zika Virus Infection/metabolism
11.
Microvasc Res ; 131: 104024, 2020 09.
Article in English | MEDLINE | ID: mdl-32502488

ABSTRACT

Congenital toxoplasmosis is a parasitic disease that occurs due vertical transmission of the protozoan Toxoplasma gondii (T. gondii) during pregnancy. The parasite crosses the placental barrier and reaches the developing brain, infecting progenitor, glial, neuronal and vascular cell types. Although the role of Radial glia (RG) neural stem cells in the development of the brain vasculature has been recently investigated, the impact of T. gondii infection in these events is not yet understood. Herein, we studied the role of T. gondii infection on RG cell function and its interaction with endothelial cells. By infecting isolated RG cultures with T. gondii tachyzoites, we observed a cytotoxic effect with reduced numbers of RG populations together with decrease neuronal and oligodendrocyte progenitor populations. Conditioned medium (CM) from RG control cultures increased ZO-1 protein levels and organization on endothelial bEnd.3 cells membranes, which was impaired by CM from infected RG, accompanied by decreased trans-endothelial electrical resistance (TEER). ELISA assays revealed reduced levels of anti-inflammatory cytokine TGF-ß1 in CM from T. gondii-infected RG cells. Treatment with recombinant TGF-ß1 concomitantly with CM from infected RG cultures led to restoration of ZO-1 staining in bEnd.3 cells. Congenital infection in Swiss Webster mice led to abnormalities in the cortical microvasculature in comparison to uninfected embryos. Our results suggest that infection of RG cells by T. gondii negatively modulates cytokine secretion, which might contribute to endothelial loss of barrier properties, thus leading to impairment of neurovascular interaction establishment.


Subject(s)
Cell Differentiation , Cerebral Cortex/blood supply , Endothelial Cells/parasitology , Ependymoglial Cells/parasitology , Microvessels/parasitology , Neurovascular Coupling , Toxoplasma/pathogenicity , Toxoplasmosis, Cerebral/parasitology , Toxoplasmosis, Congenital/parasitology , Animals , Cell Line , Disease Models, Animal , Electric Impedance , Endothelial Cells/metabolism , Endothelial Cells/pathology , Ependymoglial Cells/metabolism , Ependymoglial Cells/pathology , Mice, Inbred C57BL , Microvessels/metabolism , Microvessels/pathology , Tight Junctions/metabolism , Tight Junctions/parasitology , Tight Junctions/pathology , Toxoplasmosis, Cerebral/metabolism , Toxoplasmosis, Cerebral/pathology , Toxoplasmosis, Congenital/metabolism , Toxoplasmosis, Congenital/pathology , Transforming Growth Factor beta1/metabolism , Zonula Occludens-1 Protein/metabolism
12.
PLoS One ; 15(5): e0232552, 2020.
Article in English | MEDLINE | ID: mdl-32374791

ABSTRACT

Toxoplasma gondii is an obligate intracellular parasite which is capable of establishing life-long chronic infection in any mammalian host. During the intracellular life cycle, the parasite secretes an array of proteins into the parasitophorous vacuole (PV) where it resides. Specialized organelles called the dense granules secrete GRA proteins that are known to participate in nutrient acquisition, immune evasion, and host cell-cycle manipulation. Although many GRAs have been discovered which are expressed during the acute infection mediated by tachyzoites, little is known about those that participate in the chronic infection mediated by the bradyzoite form of the parasite. In this study, we sought to uncover novel bradyzoite-upregulated GRA proteins using proximity biotinylation, which we previously used to examine the secreted proteome of the tachyzoites. Using a fusion of the bradyzoite upregulated protein MAG1 to BirA* as bait and a strain with improved switch efficiency, we identified a number of novel GRA proteins which are expressed in bradyzoites. After using the CRISPR/Cas9 system to characterize these proteins by gene knockout, we focused on one of these GRAs (GRA55) and found it was important for the establishment or maintenance of cysts in the mouse brain. These findings highlight new components of the GRA proteome of the tissue-cyst life stage of T. gondii and identify potential targets that are important for maintenance of parasite persistence in vivo.


Subject(s)
Protozoan Proteins/metabolism , Toxoplasma/physiology , Animals , Biotinylation , Brain/metabolism , Brain/parasitology , CRISPR-Cas Systems , Female , Gene Knockout Techniques , Genes, Protozoan , Humans , Life Cycle Stages , Mice , Mice, Inbred C57BL , Proteome/metabolism , Protozoan Proteins/genetics , Toxoplasma/genetics , Toxoplasma/growth & development , Toxoplasmosis, Animal/metabolism , Toxoplasmosis, Animal/parasitology , Toxoplasmosis, Cerebral/metabolism , Toxoplasmosis, Cerebral/parasitology , Vacuoles/metabolism , Virulence
13.
Front Immunol ; 11: 603924, 2020.
Article in English | MEDLINE | ID: mdl-33613523

ABSTRACT

Toxoplasma gondii is a neurotropic protozoan parasite, which is linked to neurological manifestations in immunocompromised individuals as well as severe neurodevelopmental sequelae in congenital toxoplasmosis. While the complement system is the first line of host defense that plays a significant role in the prevention of parasite dissemination, Toxoplasma artfully evades complement-mediated clearance via recruiting complement regulatory proteins to their surface. On the other hand, the details of Toxoplasma and the complement system interaction in the brain parenchyma remain elusive. In this study, infection-induced changes in the mRNA levels of complement components were analyzed by quantitative PCR using a murine Toxoplasma infection model in vivo and primary glial cells in vitro. In addition to the core components C3 and C1q, anaphylatoxin C3a and C5a receptors (C3aR and C5aR1), as well as alternative complement pathway components properdin (CFP) and factor B (CFB), were significantly upregulated 2 weeks after inoculation. Two months post-infection, CFB, C3, C3aR, and C5aR1 expression remained higher than in controls, while CFP upregulation was transient. Furthermore, Toxoplasma infection induced significant increase in CFP, CFB, C3, and C5aR1 in mixed glial culture, which was abrogated when microglial activation was inhibited by pre-treatment with minocycline. This study sheds new light on the roles for the complement system in the brain parenchyma during Toxoplasma infection, which may lead to the development of novel therapeutic approaches to Toxoplasma infection-induced neurological disorders.


Subject(s)
Brain/parasitology , Complement Factor B/metabolism , Complement Pathway, Alternative , Microglia/parasitology , Receptor, Anaphylatoxin C5a/metabolism , Toxoplasma/pathogenicity , Toxoplasmosis, Animal/parasitology , Toxoplasmosis, Cerebral/parasitology , Animals , Brain/immunology , Brain/metabolism , Cells, Cultured , Complement Factor B/genetics , Disease Models, Animal , Host-Parasite Interactions , Male , Mice, Inbred C57BL , Microglia/immunology , Microglia/metabolism , Receptor, Anaphylatoxin C5a/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Time Factors , Toxoplasma/immunology , Toxoplasmosis, Animal/genetics , Toxoplasmosis, Animal/immunology , Toxoplasmosis, Animal/metabolism , Toxoplasmosis, Cerebral/genetics , Toxoplasmosis, Cerebral/immunology , Toxoplasmosis, Cerebral/metabolism , Up-Regulation
14.
Parasitol Res ; 118(9): 2591-2600, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31350619

ABSTRACT

Infective larvae of Toxocara canis and T. cati, the common roundworms of dogs and cats, may invade the central nervous system of paratenic hosts, including humans, causing neurotoxocarosis (NT). Previous studies on NT in the model organism "mouse" have indicated distinct differences between T. canis and T. cati regarding larval migration patterns as well as the severity of clinical symptoms and behavioural alterations. The objective of the present study was to provide an extensive characterization of the underlying histopathological alterations, comparing T. canis- and T. cati-induced changes in different brain areas over the course of murine infection. Four histological sections of five brains each of T. canis- and T. cati-infected as well as uninfected C57Bl/6 mice were investigated 7, 14, 28, 42, 70 and 98 days post infection (dpi), while brains of T. cati-infected and control mice were also available 120 and 150 dpi. In addition to haematoxylin-eosin and luxol fast blue-cresyl violet staining, immunohistochemistry was employed to study microglia/macrophage cell morphology and to detect accumulation of ß-amyloid precursor protein (ß-APP) as an indicator of axonal damage. Haemorrhages, eosinophilic vasculitis and activated microglia/macrophages were detected in both infection groups starting 7 dpi, followed by eosinophilic meningitis in cerebra as from 14 dpi. Overall, little differences in the proportion of animals affected by these alterations were found between the two infection groups. In contrast, the proportion of animals displaying ß-APP accumulation was significantly higher in the T. canis than T. cati group as from 28 dpi regarding the cerebrum as well as at 98 dpi regarding the cerebellum. In T. canis-infected mice, myelinophagic microglia/macrophages ("gitter cells") appeared as from 14 dpi, whereas these were first observed at 70 dpi in T. cati-infected animals. The proportion of animals displaying demyelination and/or gitter cells in the cerebrum was significantly higher in the T. canis than T. cati group as from 28 dpi, and at 28 and 42 dpi regarding the cerebellum. Earlier and more severe neurodegeneration during T. canis- than T. cati-induced NT, especially in the cerebrum, may explain the differences in behavioural alterations observed in previous studies. In addition to differences in larval migration preferences, immunological processes may contribute to these patterns, which warrant further investigation.


Subject(s)
Toxocara canis/physiology , Toxocariasis/parasitology , Toxoplasmosis, Cerebral/parasitology , Amyloid beta-Peptides/metabolism , Animals , Brain/metabolism , Brain/parasitology , Brain/pathology , Disease Models, Animal , Female , Humans , Larva/physiology , Male , Mice , Mice, Inbred C57BL , Toxocara canis/immunology , Toxocariasis/metabolism , Toxocariasis/pathology , Toxoplasmosis, Cerebral/metabolism , Toxoplasmosis, Cerebral/pathology
15.
Am J Pathol ; 189(4): 730-738, 2019 04.
Article in English | MEDLINE | ID: mdl-30653952

ABSTRACT

Toxoplasmosis is a neglected disease that affects millions of individuals worldwide. Toxoplasma gondii infection is an asymptomatic disease, with lethal cases occurring mostly in HIV patients and organ transplant recipients. Nevertheless, atypical strains of T. gondii in endemic locations cause severe pathology in healthy individuals. Toxoplasmosis has no cure but it can be controlled by the proinflammatory immune response. The purinergic receptor P2X7 (P2X7) is involved in many inflammatory events and has been associated with genes that confer resistance against toxoplasmosis in humans. In vitro studies have reported parasite death after P2X7-receptor activation in various cell types. To understand the contribution of P2X7 during cerebral toxoplasmosis, wild-type and P2rx7 knockout mice were infected orally with T. gondii and their pathologic profiles were analyzed. We found that all P2rx7-/- mice died 8 weeks after infection with an increased number of cysts and fewer inflammatory infiltrates in their brains. The cytokines interleukin-1ß, interleukin-12, tumor necrosis factor-α, and reactive oxygen species were absent or reduced in P2rx7-/- mice. Taken together, these data suggest that the P2X7 receptor promotes inflammatory infiltrates, proinflammatory cytokines, and reactive oxygen species production in the brain, and that P2X7 signaling mediates major events that confer resistance to cerebral toxoplasmosis.


Subject(s)
Brain/pathology , Disease Susceptibility , Inflammation/etiology , Receptors, Purinergic P2X7/physiology , Toxoplasma/pathogenicity , Toxoplasmosis, Cerebral/etiology , Animals , Brain/metabolism , Brain/microbiology , Cytokines/metabolism , Female , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Toxoplasmosis, Cerebral/metabolism , Toxoplasmosis, Cerebral/pathology
16.
Front Immunol ; 9: 1648, 2018.
Article in English | MEDLINE | ID: mdl-30108583

ABSTRACT

Toxoplasmic encephalitis (TE), an opportunistic infection, is a severe health problem in immunocompromised patients. Previous studies have revealed that C57BL/6 mice are susceptible and BALB/c mice are resistant to TE. To investigate the mechanisms involved in the immunopathogenesis of TE in susceptible C57BL/6 and resistant BALB/c mice, both strains of mice were perorally infected with the Prugniuad (Pru) strain of Toxoplasma gondii. Our results showed that compared with BALB/c mice, C57BL/6 mice infected with T. gondii Pru strain had more severe brain histopathological damage, and higher mRNA expression levels of tachyzoite-specific surface antigen 1, bradyzoite-specific antigen 1, interferon gamma (IFNγ), interleukin (IL)-10, arginase1 (Arg1) (M2 marker), galectin (Gal)-3, Gal-9, T. gondii microneme protein 1 (TgMIC1), TgMIC4, and TgMIC6 during the course of infection by using quantitative real-time reverse transcription-polymerase chain reaction. Further analysis displayed that BALB/c mice showed higher numbers of microglial cells and higher levels of IL-1ß, inducible nitric oxide synthase (iNOS) (M1 marker), and chitinase-3-like protein 3 (Ym1) (M2 marker) in the early infective stage [at day 14 or 35 post infection (p.i.)] compared with C57BL/6 mice, whereas C57BL/6 mice showed higher numbers of microglial cells and higher levels of IL-10, iNOS (M1 marker), and Ym1 (M2 marker) at days 35, 50, or 70 p.i. compared with BALB/c mice. Correlation analysis showed that significant positive correlations existed between Gal-3 and IL-4/IL-10/iNOS/Ym1 and between Gal-9 and IL-4/Ym1 in C57BL/6 mice; between Gal-3 and IFNγ/Arg1 and between Gal-9 and IFNγ/Arg1 in BALB/c mice. Together, our data demonstrated that different Gal-3 and Gal-9 expressions as well as different positive correlations were found between Gal-3 and T helper 1 (Th1)/Th2/M1/M2 cytokines or between Gal-9 and Th1/Th2/M2 cytokines in the brains of T. gondii Pru strain-infected C57BL/6 and BALB/c mice.


Subject(s)
Brain/metabolism , Galectin 3/metabolism , Galectins/metabolism , Infectious Encephalitis/metabolism , Microglia/metabolism , Toxoplasma , Toxoplasmosis, Cerebral/metabolism , Animals , Biomarkers/metabolism , Brain/immunology , Cytokines/metabolism , Genetic Predisposition to Disease , Humans , Infectious Encephalitis/genetics , Infectious Encephalitis/immunology , Infectious Encephalitis/physiopathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microglia/immunology , Species Specificity , Toxoplasmosis, Cerebral/genetics , Toxoplasmosis, Cerebral/immunology , Toxoplasmosis, Cerebral/physiopathology
17.
Behav Brain Res ; 347: 193-200, 2018 07 16.
Article in English | MEDLINE | ID: mdl-29555339

ABSTRACT

Infection with the protozoan parasite, Toxoplasma gondii (T. gondii), has been associated with the increased risk for several psychiatric disorders. The exact mechanisms of a hypothesized contribution of T. gondii infection are poorly understood. The T. gondii genome contains two aromatic amino acid hydroxylase genes (AAH1 and AAH2) that encode proteins that can produce L-DOPA. One popular hypothesis posits that these encoded enzymes might influence dopamine (DA) production and hence DA synaptic transmission, leading to neurobehavioral abnormalities in the infected host. Prior studies have shown that deletion of these genes does not alter DA levels in the brain or exploratory activity in infected mice. However, possible effects of AAH gene deficiency on infection-induced brain and behavior alterations that are directly linked to DA synaptic transmission have not been evaluated. We found that chronic T. gondii infection of BALB/c mice leads to blunted response to amphetamine or cocaine and decreased expression of Dopamine Transporter (DAT) and Vesicular Monoamine Transporter 2 (VMAT2). Deletion of AAH2 had no effects on these changes in infected mice. Both wild type and Δaah2 strains produced comparable levels of neuroinflammation. Our findings demonstrate that AAH2 is not required for T. gondii infection-produced DA-dependent neurobehavioral abnormalities.


Subject(s)
Brain/metabolism , Protozoan Proteins/metabolism , Toxoplasma/metabolism , Toxoplasmosis, Animal/metabolism , Toxoplasmosis, Cerebral/metabolism , Amphetamine/pharmacology , Animals , Animals, Genetically Modified , Astrocytes/drug effects , Astrocytes/metabolism , Astrocytes/parasitology , Astrocytes/pathology , Brain/drug effects , Brain/parasitology , Brain/pathology , Central Nervous System Stimulants/pharmacology , Chronic Disease , Cocaine/pharmacology , Dopamine Plasma Membrane Transport Proteins/metabolism , Male , Mice, Inbred BALB C , Microglia/drug effects , Microglia/metabolism , Microglia/parasitology , Microglia/pathology , Motor Activity/drug effects , Motor Activity/physiology , Prepulse Inhibition/drug effects , Prepulse Inhibition/physiology , Protozoan Proteins/genetics , Reflex, Startle/drug effects , Reflex, Startle/physiology , Toxoplasma/genetics , Vesicular Monoamine Transport Proteins/metabolism
18.
Clin Nucl Med ; 42(12): e506-e508, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29076908

ABSTRACT

We report the case of a 40-year-old woman with a progressive right-sided hemiparesis. Standard MRI revealed a contrast-enhancing brain lesion within the left basal ganglia. Ffluoroethyl-L-tyrosine (F-FET) PET showed a distinct tracer uptake (lesion-to-brain ratio [LBR]: LBRmax = 2.03, LBRmean = 1.68) with a significant larger metabolic lesion volume than contrast-enhancement in MRI, indicating cerebral glioma. Surprisingly, histopathologic analysis demonstrated central nervous system toxoplasmosis with pronounced inflammatory reaction (reactive astrogliosis, microglia activation, macrophage, and T-lymphocyte infiltration), which was associated with strong LAT1/LAT2/CD98 expression. In conclusion, inflammatory brain lesions, such as cerebral toxoplasmosis, represent a potential pitfall of F-FET PET mimicking a brain tumor.


Subject(s)
Acquired Immunodeficiency Syndrome/complications , Adaptor Proteins, Signal Transducing/metabolism , Gene Expression Regulation , Large Neutral Amino Acid-Transporter 1/metabolism , Positron-Emission Tomography , Toxoplasmosis, Cerebral/diagnostic imaging , Tyrosine/analogs & derivatives , Adult , Biological Transport , Brain Neoplasms/diagnostic imaging , Diagnosis, Differential , Female , Humans , Inflammation/pathology , Toxoplasmosis, Cerebral/complications , Toxoplasmosis, Cerebral/metabolism , Tyrosine/metabolism
19.
J Proteomics ; 160: 74-83, 2017 05 08.
Article in English | MEDLINE | ID: mdl-28323244

ABSTRACT

The aim of our study was to detect differentially regulated proteins and specific signaling pathways in Mongolian gerbil brains during chronic Toxoplasma gondii (T.gondii) PRU strain infection. We use a iTRAQ-based strategy to detecte 4935 proteins, out of which 110 proteins were differentially expressed (>/=2.0-fold, p value <0.05) when the brain of gerbils infected with T.gondii was compared to control brain tissues. We confirmed the authenticity and the accuracy of iTRAQ results through quantitative real-time PCR and western blot (WB), which was consistent with mass spectrometry analysis. Pathway analysis and GO (Gene Ontology) annotations indicated the deregulation of several pathways related to immune response, metabolism and neurological processes, like neuronal growth and neurotransmitter transport. Through the iTRAQ-based strategy, we obtained a comparative proteome profile of brain tissues from Mongolian gerbils with chronic infection of T.gondii. Several differentially expressed proteins involved in neurological pathways, like Parvalbumin, Drebrin or Synaptotagmin, can be further investigated to enhance our understanding of central nervous system (CNS) injury caused by T.gondii. BIOLOGICAL SIGNIFICANCE: T.gondii can infect almost all nucleated cells with a preference for the CNS, which can induce Toxoplasma encephalitis (TE). However, the pathogenesis and mechanisms between the parasite and host associated with TE are largely unexplored. Around 30% of the world population is considered to have latent infection with T.gondii and >90% patients died of TE, while the proportion of secondary paralysis is also high. Patients of TE may have highly varied neurological symptoms with both focal and diffuse neurological lesions, while mental symptoms and behavior disorders are frequently accompanied, like the Alzheimer's disease (AD). We present a comparative proteomics analysis to explore the differences of protein expression caused by chronic T.gondii infection. The results of this analysis can be helpful for identifying key proteins involved in the pathogenesis of TE. In addition, the study can contribute to a better understanding of molecular mechanisms underlying the host-parasite relationship in chronic infection of T.gondii and facilitate further development of new therapies for TE.


Subject(s)
Brain/parasitology , Proteomics/methods , Toxoplasma/chemistry , Animals , Blotting, Western , Brain/metabolism , Gene Expression Regulation , Gene Ontology , Gerbillinae , Host-Parasite Interactions , Mass Spectrometry , Real-Time Polymerase Chain Reaction , Toxoplasmosis, Cerebral/metabolism
20.
PLoS Pathog ; 12(6): e1005643, 2016 06.
Article in English | MEDLINE | ID: mdl-27281462

ABSTRACT

The immune privileged nature of the CNS can make it vulnerable to chronic and latent infections. Little is known about the effects of lifelong brain infections, and thus inflammation, on the neurological health of the host. Toxoplasma gondii is a parasite that can infect any mammalian nucleated cell with average worldwide seroprevalence rates of 30%. Infection by Toxoplasma is characterized by the lifelong presence of parasitic cysts within neurons in the brain, requiring a competent immune system to prevent parasite reactivation and encephalitis. In the immunocompetent individual, Toxoplasma infection is largely asymptomatic, however many recent studies suggest a strong correlation with certain neurodegenerative and psychiatric disorders. Here, we demonstrate a significant reduction in the primary astrocytic glutamate transporter, GLT-1, following infection with Toxoplasma. Using microdialysis of the murine frontal cortex over the course of infection, a significant increase in extracellular concentrations of glutamate is observed. Consistent with glutamate dysregulation, analysis of neurons reveal changes in morphology including a reduction in dendritic spines, VGlut1 and NeuN immunoreactivity. Furthermore, behavioral testing and EEG recordings point to significant changes in neuronal output. Finally, these changes in neuronal connectivity are dependent on infection-induced downregulation of GLT-1 as treatment with the ß-lactam antibiotic ceftriaxone, rescues extracellular glutamate concentrations, neuronal pathology and function. Altogether, these data demonstrate that following an infection with T. gondii, the delicate regulation of glutamate by astrocytes is disrupted and accounts for a range of deficits observed in chronic infection.


Subject(s)
Astrocytes/metabolism , Brain/microbiology , Excitatory Amino Acid Transporter 2/metabolism , Glutamic Acid/metabolism , Homeostasis , Neurons/metabolism , Toxoplasmosis, Cerebral/metabolism , Animals , Blotting, Western , Brain/metabolism , Central Nervous System/metabolism , Central Nervous System/microbiology , Disease Models, Animal , Electroencephalography , Female , Homeostasis/physiology , Immunohistochemistry , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Microdialysis , Microscopy, Electron, Transmission , Real-Time Polymerase Chain Reaction , Toxoplasma
SELECTION OF CITATIONS
SEARCH DETAIL
...