Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.591
Filter
1.
Ann Parasitol ; 70(2): 101-111, 2024.
Article in English | MEDLINE | ID: mdl-39154197

ABSTRACT

Toxoplasma gondii and Toxocara spp. zoonotic infections may cause severe systemic and ocular illness in infected individuals. Cats play a significant role in environmental contamination and the transmission of parasites. The goal of the present study was to investigate the prevalence of Toxoplasma gondii (T. gondii) and Toxocara spp. infection among stray cats at Ahvaz Jundishapur University of Medical Sciences campus. The current descriptive study began with the collection of 170 fresh cat faecal samples from various sites in the Ahvaz Jundishapur University of Medical Sciences area. Sheather's sugar flotation method was applied to all specimens, and parasites were identified and examined microscopically. Next, a nested-PCR assay, sequencing, and real-time PCR with high-resolution melting curve (HRM) analysis were performed. In this study, out of 170 cat faecal samples microscopically evaluated, 8 (4.70%) and 37 (21.76%) were infected with T. gondii oocysts and Toxocara eggs, respectively. Using nested PCR, 8 out of 170 samples (4.70%) were found to be infected with T. gondii. HRM analysis showed that all isolates could be classified into three genetic lineages. Considerable prevalence, exceeding 50% for Toxocara and surpassing 25% for Toxoplasma in certain instances, along with genetic diversity, was observed in the present study. Hence, it is suggested that all individuals, including kindergarten children, students, employees, workers, and pregnant women who are in contact with their surroundings, take the necessary precautions.


Subject(s)
Cat Diseases , Feces , Toxocara , Toxoplasma , Animals , Cats , Toxoplasma/isolation & purification , Toxoplasma/genetics , Toxocara/isolation & purification , Toxocara/genetics , Feces/parasitology , Cat Diseases/parasitology , Cat Diseases/epidemiology , Universities , Toxocariasis/epidemiology , Toxocariasis/parasitology , Toxoplasmosis, Animal/epidemiology , Toxoplasmosis, Animal/parasitology , Toxoplasmosis/epidemiology , Toxoplasmosis/parasitology
2.
PLoS Biol ; 22(8): e3002745, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39137211

ABSTRACT

Rhoptries are specialized secretory organelles conserved across the Apicomplexa phylum, essential for host cell invasion and critical for subverting of host cellular and immune functions. They contain proteins and membranous materials injected directly into the host cells, participating in parasitophorous vacuole formation. Toxoplasma gondii tachyzoites harbor 8 to 12 rhoptries, 2 of which are docked to an apical vesicle (AV), a central element associated with a rhoptry secretory apparatus prior to injection into the host cell. This parasite is also equipped with 5 to 6 microtubule-associated vesicles, presumably serving as AV replenishment for iterative rhoptry discharge. Here, we characterized a rhoptry protein, rhoptry discharge factor 3 (RDF3), crucial for rhoptry discharge and invasion. RDF3 enters the secretory pathway, localizing near the AV and associated with the rhoptry bulb. Upon invasion, RDF3 dynamically delocalizes, suggesting a critical role at the time of rhoptry discharge. Cryo-electron tomography analysis of RDF3-depleted parasites reveals irregularity in microtubule-associated vesicles morphology, presumably impacting on their preparedness to function as an AV. Our findings suggest that RDF3 is priming the microtubule-associated vesicles for rhoptry discharge by a mechanism distinct from the rhoptry secretory apparatus contribution.


Subject(s)
Microtubules , Protozoan Proteins , Toxoplasma , Toxoplasma/metabolism , Toxoplasma/pathogenicity , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Microtubules/metabolism , Animals , Mice , Host-Parasite Interactions , Humans , Organelles/metabolism , Electron Microscope Tomography , Toxoplasmosis/parasitology , Toxoplasmosis/metabolism
3.
Gut Microbes ; 16(1): 2391535, 2024.
Article in English | MEDLINE | ID: mdl-39182245

ABSTRACT

BACKGROUND: Chronic infection with the neurotropic parasite Toxoplasma gondii (T. gondii) can cause anxiety and gut microbiota dysbiosis in hosts. However, the potential role of gut microbiota in anxiety induced by the parasite remains unclear. METHODS: C57BL/6J mice were infected with 10 cysts of T. gondii. Antibiotic depletion of gut microbiota and fecal microbiota transplantation experiments were utilized to investigate the causal relationship between gut microbiota and anxiety. Anxiety-like behaviors were examined by the elevated plus maze test and the open field test; blood, feces, colon and amygdala were collected to evaluate the profiles of serum endotoxin (Lipopolysaccharide, LPS) and serotonin (5-hydroxytryptamine, 5-HT), gut microbiota composition, metabolomics, global transcriptome and neuroinflammation in the amygdala. Furthermore, the effects of Diethyl butylmalonate (DBM, an inhibitor of mitochondrial succinate transporter, which causes the accumulation of endogenous succinate) on the disorders of the gut-brain axis were evaluated. RESULTS: Here, we found that T. gondii chronic infection induced anxiety-like behaviors and disturbed the composition of the gut microbiota in mice. In the amygdala, T. gondii infection triggered the microglial activation and neuroinflammation. In the colon, T. gondii infection caused the intestinal dyshomeostasis including elevated colonic inflammation, enhanced bacterial endotoxin translocation to blood and compromised intestinal barrier. In the serum, T. gondii infection increased the LPS levels and decreased the 5-HT levels. Interestingly, antibiotics ablation of gut microbiota alleviated the anxiety-like behaviors induced by T. gondii infection. More importantly, transplantation of the fecal microbiota from T. gondii-infected mice resulted in anxiety and the transcriptomic alteration in the amygdala of the antibiotic-pretreated mice. Notably, the decreased abundance of succinate-producing bacteria and the decreased production of succinate were observed in the feces of the T. gondii-infected mice. Moreover, DBM administration ameliorated the anxiety and gut barrier impairment induced by T. gondii infection. CONCLUSIONS: The present study uncovers a novel role of gut microbiota in mediating the anxiety-like behaviors induced by chronic T. gondii infection. Moreover, we show that DBM supplementation has a beneficial effect on anxiety. Overall, these findings provide new insights into the treatment of T. gondii-related mental disorders.


Subject(s)
Anxiety , Gastrointestinal Microbiome , Mice, Inbred C57BL , Toxoplasma , Animals , Mice , Anxiety/microbiology , Toxoplasma/physiology , Male , Fecal Microbiota Transplantation , Dysbiosis/microbiology , Amygdala/metabolism , Behavior, Animal , Toxoplasmosis/physiopathology , Toxoplasmosis/psychology , Toxoplasmosis/parasitology , Toxoplasmosis/microbiology , Chronic Disease , Brain-Gut Axis/physiology , Disease Models, Animal , Colon/microbiology , Colon/parasitology
4.
Turkiye Parazitol Derg ; 48(2): 128-132, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38958491

ABSTRACT

Toxoplasma gondii (T. gondii) is an obligate intracellular, zoonotic protozoan parasite of interest to physicians and veterinarians with its highly complex structure. It is known to infect about one-third of the world's population. Since it is a zoonotic disease, it is necessary to keep the animal population under control in order to prevent human exposure. Many studies have been conducted on the detection of T. gondii and it has been determined that there are three clonal groups consisting of types 1, 2, 3. Developments in molecular studies have led to changes in the taxonomy and new developments in parasitic diseases. It has helped in diagnosis, treatment, development of antiparasitic drugs and research on resistance. They also provided research on vaccine studies, genetic typing and phylogenetics of parasitic diseases. Conventional polymerase chain reaction (PCR), real-time PCR and genotyping studies conducted today increase our knowledge about T. gondii. Methods such as B1, SAG1, SAG2, GRA1, 529-bp repeat element, OWP genes and 18S rRNAs are mostly used in PCR, and methods such as MS, MLST, PCR-RFLP, RAPD-PCR and HRM are used in genotyping. Toxoplasmosis is a disease that is within the framework of the concept of one health and must attract attention, has not yet been eradicated in the world and needs joint studies for humans, animals and ecosystems to be eradicated. This can only be possible by establishing interdisciplinary groups, conducting surveys and training.


Subject(s)
Toxoplasma , Toxoplasmosis, Animal , Toxoplasmosis , Toxoplasma/genetics , Toxoplasma/classification , Animals , Humans , Toxoplasmosis/parasitology , Toxoplasmosis, Animal/parasitology , Toxoplasmosis, Animal/diagnosis , Zoonoses/parasitology , Genotype
5.
Zhongguo Xue Xi Chong Bing Fang Zhi Za Zhi ; 36(3): 304-309, 2024 Jun 07.
Article in Chinese | MEDLINE | ID: mdl-38952318

ABSTRACT

OBJECTIVE: To investigate the development and dynamic changes of cysts in the brain of mice following infection with different forms of Toxoplasma gondii, so as to provide insights into for toxoplasmosis prevention and control. METHODS: ICR mice at ages of 6 to 8 weeks, each weighing 20 to 25 g, were intraperitoneally injected with tachyzoites of the T. gondii PRU strain at a dose of 1 × 105 tachyzoites per mouse, orally administered with cysts at a dose of 20 oocysts per mouse or oocysts at a dose of 200 oocysts per mouse for modeling chronic T. gondii infection in mice, and the clinical symptoms and survival of mice were observed post-infection. Mice were orally infected with T. gondii cysts at doses of 10 (low-dose group), 20 (medium-dose group), 40 cysts per mouse (high-dose group), and the effect of different doses of T. gondii infections on the number of cysts was examined in the mouse brain. Mice were orally administered with T. gondii cysts at a dose of 20 cysts per mouse, and grouped according to gender (female and male) and time points of infections (20, 30, 60, 90, 120, 150, 180 days post-infection), and the effects of gender and time points of infections on the number of cysts was examined in the mouse brain. In addition, mice were divided into the tachyzoite group (Group T), the first-generation cyst group (Group C1), the second-generation cyst group (Group C2), the third-generation cyst (Group C3) and the fourth-generation cyst group (Group C4). Mice in the Group T were intraperitoneally injected with T. gondii tachyzoites at a dose of 1 × 105 tachyzoites per mouse, and the cysts were collected from the mouse brain tissues 30 days post-infection, while mice in the Group C1 were orally infected with the collected cysts at a dose of 30 cysts per mouse. Continuous passage was performed by oral administration with cysts produced by the previous generation in mice, and the effect of continuous passage on the number of cysts was examined in the mouse brain. RESULTS: Following infection with T. gondii tachyzoites, cysts and oocysts in mice, obvious clinical symptoms were observed on days 6 to 13 and mice frequently died on days 7 to 12. The survival rates of mice were 67.0%, 87.0% and 53.0%, and the mean numbers of cysts were (516.0 ± 257.2), (1 203.0 ± 502.0) and (581.0 ± 183.1) in the mouse brain (F = 11.94, P < 0.01) on day 30 post-infection with T. gondii tachyzoites, cysts and oocysts, respectively, and the numbers of cysts in the brain tissues were significantly lower in mice infected with T. gondii tachyzoites and oocysts than in those infected with cysts (all P values < 0.01). The survival rates of mice were 87.0%, 87.0% and 60.0%, and the mean numbers of cysts were (953.0 ± 355.5), (1 084.0 ± 474.3) and (1 113.0 ± 546.0) in the mouse brain in the low-, medium- and high-dose groups on day 30 post-infection, respectively (F = 0.42, P > 0.05). The survival rates of male and female mice were 73.0% and 80.0%, and the mean numbers of cysts were (946.4 ± 411.4) and (932.1 ± 322.4) in the brain tissues of male and female mice, respectively (F = 1.63, P > 0.05). Following continuous passage, the mean numbers of cysts were (516.0 ± 257.2), (1 203.0 ± 502.0), (896.8 ± 332.3), (782.5 ± 423.9) and (829.2 ± 306.0) in the brain tissues of mice in the T, C1, C2, C3 and C4 groups, respectively (F = 4.82, P < 0.01), and the number of cysts was higher in the mouse brain in Group 1 than in Group T (P < 0.01). Following oral administration of 20 T. gondii cysts in mice, cysts were found in the moues brain for the first time on day 20 post-infection, and the number of cysts gradually increased over time, peaked on days 30 and 90 post-infection and then gradually decreased; however, the cysts were still found in the mouse brain on day 180 post-infection. CONCLUSIONS: There is a higher possibility of developing chronic T. gondii infection in mice following infection with cysts than with oocysts or tachyzoites and the most severe chronic infection is seen following infection with cysts. The number of cysts does not correlate with the severity of chronic T. gondii infection, and the number of cysts peaks in the mouse brain on days 30 and 90 post-infection.


Subject(s)
Brain , Mice, Inbred ICR , Toxoplasma , Toxoplasmosis, Animal , Animals , Mice , Female , Male , Brain/parasitology , Chronic Disease , Toxoplasmosis, Animal/parasitology , Toxoplasma/physiology , Toxoplasmosis/parasitology , Disease Models, Animal
6.
Front Immunol ; 15: 1428232, 2024.
Article in English | MEDLINE | ID: mdl-39040112

ABSTRACT

In the decades since the discovery, Type I interferon (IFN-I) has been intensively studied for their antiviral activity. However, increasing evidences suggest that it may also play an important role in the infection of Toxoplasma gondii, a model organism for intracellular parasites. Recent studies demonstrated that the induction of IFN-I by the parasite depends on cell type, strain genotype, and mouse strain. IFN-I can inhibit the proliferation of T. gondii, but few studies showed that it is beneficial to the growth of the parasite. Meanwhile, T. gondii also can secrete proteins that impact the pathway of IFN-I production and downstream induced interferon-stimulated genes (ISGs) regulation, thereby escaping immune destruction by the host. This article reviews the major findings and progress in the production, function, and regulation of IFN-I during T. gondii infection, to thoroughly understand the innate immune mechanism of T. gondii infection, which provides a new target for subsequent intervention and treatment.


Subject(s)
Interferon Type I , Toxoplasma , Toxoplasmosis , Toxoplasma/immunology , Animals , Interferon Type I/immunology , Interferon Type I/metabolism , Humans , Toxoplasmosis/immunology , Toxoplasmosis/parasitology , Host-Parasite Interactions/immunology , Immunity, Innate , Signal Transduction , Gene Expression Regulation , Mice
7.
Immun Inflamm Dis ; 12(6): e1329, 2024 Jun.
Article in English | MEDLINE | ID: mdl-39031850

ABSTRACT

BACKGROUND: Toxoplasma gondii is an obligate intracellular protozoan parasite that can invade all mammalian cells. It is well established that natural killer (NK) cells have critical protective roles in innate immunity during infections by intracellular pathogens. In the current study, we conducted an in vitro experiment to evaluate NK cell differentiation and activation from human umbilical cord blood mononuclear cells (UCB-MNCs) after infection with T. gondii tachyzoites. METHODS: UCB-MNCs were infected by fresh tachyzoites of type I (RH) or type II (PTG) strains of T. gondii pre-expanded in mesenchymal stem cells for 2 weeks in a medium enriched with stem cell factor, Flt3, IL-2, and IL-15. Flow cytometry analysis and western blot analysis were performed to measure the CD57+, CD56+, and Granzyme A (GZMA). RESULTS: Data revealed that incubation of UCB-MNCs with NK cell differentiation medium increased the CD57+, CD56+, and GZMA. UCB-MNCs cocultured with PTG tachyzoites showed a significant reduction of CD56+ and GZMA, but nonsignificant changes, in the levels of CD56+ compared to the control UCB-MNCs (p > .05). Noteworthy, 2-week culture of UCB-MNCs with type I (RH) tachyzoites significantly suppressed CD57+, CD56+, and GZMA, showing reduction of NK cell differentiation from cord blood cells. CONCLUSION: Our findings suggest that virulent T. gondii tachyzoites with cytopathic effects inhibit NK cell activation and eliminate innate immune responses during infection, and consequently enable the parasite to continue its survival in the host body.


Subject(s)
Cell Differentiation , Fetal Blood , Killer Cells, Natural , Toxoplasma , Humans , Killer Cells, Natural/immunology , Fetal Blood/cytology , Fetal Blood/immunology , Fetal Blood/parasitology , Cell Differentiation/immunology , Toxoplasma/immunology , Cells, Cultured , Toxoplasmosis/immunology , Toxoplasmosis/parasitology , Immunity, Innate , Lymphocyte Activation/immunology , Leukocytes, Mononuclear/immunology
8.
Acta Trop ; 257: 107283, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38955322

ABSTRACT

Toxoplasmosis, a zoonotic parasitic disease caused by Toxoplasma gondii (T. gondii), is prevalent worldwide. The fact should be emphasized that a considerable proportion of individuals infected with T. gondii may remain asymptomatic; nevertheless, the condition can have severe implications for pregnant women or immunocompromised individuals. The current treatment of toxoplasmosis primarily relies on medication; however, traditional anti-toxoplasmosis drugs exhibit significant limitations in terms of efficacy, side effects, and drug resistance. The life cycles of T. gondii are characterized by distinct stages and its body morphology goes through dynamic alterations during the growth cycle that are intricately governed by a wide array of post-translational modifications (PTMs). Ubiquitin (Ub) signaling and ubiquitin-like (Ubl) signaling are two crucial post-translational modification pathways within cells, regulating protein function, localization, stability, or interactions by attaching Ub or ubiquitin-like proteins (Ubls) to target proteins. While these signaling mechanisms share some functional similarities, they have distinct regulatory mechanisms and effects. T. gondii possesses both Ub and Ubls and plays a significant role in regulating the parasite's life cycle and maintaining its morphology through PTMs of substrate proteins. Investigating the role and mechanism of protein ubiquitination in T. gondii will provide valuable insights for preventing and treating toxoplasmosis. This review explores the distinctive characteristics of Ub and Ubl signaling in T. gondii, with the aim of inspiring research ideas for the identification of safer and more effective drug targets against toxoplasmosis.


Subject(s)
Signal Transduction , Toxoplasma , Toxoplasmosis , Ubiquitin , Toxoplasma/metabolism , Toxoplasma/physiology , Toxoplasma/drug effects , Ubiquitin/metabolism , Humans , Toxoplasmosis/parasitology , Toxoplasmosis/drug therapy , Toxoplasmosis/metabolism , Animals , Protozoan Proteins/metabolism , Ubiquitination , Protein Processing, Post-Translational , Ubiquitins/metabolism , Life Cycle Stages
9.
Int J Mol Sci ; 25(14)2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39063076

ABSTRACT

Eukaryotic translation initiation factors (eIFs) are crucial for initiating protein translation and ensuring the correct assembly of mRNA-ribosomal subunit complexes. In this study, we investigated the effects of deleting six eIFs in the apicomplexan parasite Toxoplasma gondii using the CRISPR-Cas9 system. We determined the subcellular localization of these eIFs using C-terminal endogenous tagging and immunofluorescence analysis. Four eIFs (RH::315150-6HA, RH::286090-6HA, RH::249370-6HA, and RH::211410-6HA) were localized in the cytoplasm, while RH::224235-6HA was localized in the apicoplast. Additionally, RH::272640-6HA was found in both the basal complex and the cytoplasm of T. gondii. Functional characterization of the six RHΔeIFs strains was conducted using plaque assay, cell invasion assay, intracellular growth assay and egress assay in vitro, and virulence assay in mice. Disruption of five eIF genes (RHΔ315150, RHΔ272640, RHΔ249370, RHΔ211410, and RHΔ224235) did not affect the ability of the T. gondii RH strain to invade, replicate, form plaques and egress in vitro, or virulence in Kunming mice (p > 0.05). However, the RHΔ286090 strain showed slightly reduced invasion efficiency and virulence (p < 0.01) compared to the other five RHΔeIFs strains and the wild-type strain. The disruption of the TGGT1_286090 gene significantly impaired the ability of tachyzoites to differentiate into bradyzoites in both type I RH and type II Pru strains. These findings reveal that the eukaryotic translation initiation factor TGGT1_286090 is crucial for T. gondii bradyzoite differentiation and may serve as a potential target for drug development and an attenuated vaccine against T. gondii.


Subject(s)
CRISPR-Cas Systems , Eukaryotic Initiation Factors , Protozoan Proteins , Toxoplasma , Toxoplasma/genetics , Toxoplasma/pathogenicity , Toxoplasma/metabolism , Toxoplasma/growth & development , Animals , Mice , Protozoan Proteins/genetics , Protozoan Proteins/metabolism , Eukaryotic Initiation Factors/genetics , Eukaryotic Initiation Factors/metabolism , Virulence/genetics , Toxoplasmosis/parasitology , Toxoplasmosis/genetics , Humans
10.
Int J Parasitol Drugs Drug Resist ; 25: 100552, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38986389

ABSTRACT

Toxoplasma gondii (T. gondii) is a highly successful global parasite, infecting about one-third of the world's population and significantly affecting human life and the economy. However, current drugs for toxoplasmosis treatment have considerable side effects, and there is no specific drug to meet current needs. This study aims to evaluate the anti-T. gondii activity of broxaldine (BRO) in vitro and in vivo and explore its mechanism of action. Our results showed that compared to the control group, the invasion rate of tachyzoites in the 4 µg/mL BRO group was only 14.31%, and the proliferation rate of tachyzoites in host cells was only 1.23%. Furthermore, BRO disrupted the lytic cycle of T. gondii and reduced the size and number of cysts in vitro. A mouse model of acute toxoplasmosis reported a 41.5% survival rate after BRO treatment, with reduced parasite load in tissues and blood. The subcellular structure of T. gondii was observed, including disintegration of T. gondii, mitochondrial swelling, increased liposomes, and the presence of autophagic lysosomes. Further investigation revealed enhanced autophagy, increased neutral lipids, and decreased mitochondrial membrane potential in T. gondii treated with BRO. The results also showed a significant decrease in ATP levels. Overall, BRO demonstrates good anti-T. gondii activity in vitro and in vivo; therefore, it has the potential to be used as a lead compound for anti-T. gondii treatment.


Subject(s)
Toxoplasma , Toxoplasma/drug effects , Animals , Mice , Toxoplasmosis/drug therapy , Toxoplasmosis/parasitology , Female , Antiprotozoal Agents/pharmacology , Antiprotozoal Agents/therapeutic use , Disease Models, Animal , Toxoplasmosis, Animal/drug therapy , Toxoplasmosis, Animal/parasitology , Humans , Autophagy/drug effects , Parasite Load , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred BALB C
11.
Int J Mol Sci ; 25(13)2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39000057

ABSTRACT

Toxoplasma gondii, an important opportunistic pathogen, underscores the necessity of developing novel therapeutic drugs and identifying new drug targets. Our findings indicate that the half-maximal inhibitory concentrations (IC50) of KU60019 and CP466722 (abbreviated as KU and CP) against T. gondii are 0.522 µM and 0.702 µM, respectively, with selection indices (SI) of 68 and 10. Treatment with KU and CP affects the in vitro growth of T. gondii, inducing aberrant division in the daughter parasites. Transmission electron microscopy reveals that KU and CP prompt the anomalous division of T. gondii, accompanied by cellular enlargement, nuclear shrinkage, and an increased dense granule density, suggesting potential damage to parasite vesicle transport. Subsequent investigations unveil their ability to modulate the expression of certain secreted proteins and FAS II (type II fatty acid synthesis) in T. gondii, as well as including the dot-like aggregation of the autophagy-related protein ATG8 (autophagy-related protein 8), thereby expediting programmed death. Leveraging DARTS (drug affinity responsive target stability) in conjunction with 4D-Label-free quantitative proteomics technology, we identified seven target proteins binding to KU, implicated in pivotal biological processes such as the fatty acid metabolism, mitochondrial ATP transmission, microtubule formation, and Golgi proteins transport in T. gondii. Molecular docking predicts their good binding affinity. Furthermore, KU has a slight protective effect on mice infected with T. gondii. Elucidating the function of those target proteins and their mechanism of action with ATM kinase inhibitors may potentially enhance the treatment paradigm for toxoplasmosis.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , Protein Kinase Inhibitors , Toxoplasma , Toxoplasma/drug effects , Toxoplasma/enzymology , Animals , Mice , Ataxia Telangiectasia Mutated Proteins/metabolism , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Toxoplasmosis/drug therapy , Toxoplasmosis/parasitology , Humans , Protozoan Proteins/metabolism , Protozoan Proteins/antagonists & inhibitors , Female
12.
Biomed Environ Sci ; 37(6): 647-660, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38988115

ABSTRACT

Toxoplasma gondii( T. gondii or Tg), is an obligatory intracellular parasite with humans as its intermediate hosts. In recent years, significant correlations between T. gondii infection and schizophrenia have been reported, including the possible mediating mechanisms. Currently, mechanisms and hypotheses focus on central neurotransmitters, immunity, neuroinflammation, and epigenetics; however, the exact underlying mechanisms remain unclear. In this article, we review the studies related to T. gondii infection and schizophrenia, particularly the latest research progress. Research on dopamine (DA) and other neurotransmitters, the blood-brain barrier, inflammatory factors, disease heterogeneity, and other confounders is also discussed. In addition, we also summarized the results of some new epidemiological investigations.


Subject(s)
Schizophrenia , Toxoplasma , Toxoplasmosis , Schizophrenia/parasitology , Schizophrenia/etiology , Humans , Toxoplasmosis/complications , Toxoplasmosis/epidemiology , Toxoplasmosis/parasitology , Animals
13.
mSphere ; 9(7): e0036924, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38980070

ABSTRACT

Toxoplasma gondii, a widespread parasite, has the ability to infect nearly any nucleated cell in warm-blooded vertebrates. It is estimated that around 2 billion people globally have been infected by this pathogen. Although most healthy individuals can effectively control parasite replication, certain parasites may evade the immune response, establishing cysts in the brain that are refractory to the immune system and resistant to available drugs. For its chronic persistence in the brain, the parasite relies on host cells' nutrients, particularly amino acids and lipids. Therefore, understanding how latent parasites persist in the brain is crucial for identifying potential drug targets against chronic forms. While shielded within parasitophorous vacuoles (PVs) or cysts, Toxoplasma exploits the host endoplasmic reticulum (ER) metabolism to sustain its persistence in the brain, resulting in host neurological alterations. In this study, we demonstrate that T. gondii disrupts the host ER homeostasis, resulting in the accumulation of unfolded protein within the host ER. The host counters this stress by initiating an autophagic pathway known as ER-phagy, which breaks down unfolded proteins into amino acids, promoting their recycling. Our findings unveil the underlying mechanisms employed by T. gondii to exploit host ER and lysosomal pathways, enhancing nutrient levels during infection. These insights provide new strategies for the treatment of toxoplasmosis. IMPORTANCE: Intracellular parasites employ several mechanisms to manipulate the cellular environment, enabling them to persist in the host. Toxoplasma gondii, a single-celled parasite, possesses the ability to infect virtually any nucleated cell of warm-blooded vertebrates, including nearly 2 billion people worldwide. Unfortunately, existing treatments and immune responses are not entirely effective in eliminating the chronic persisting forms of the parasite. This study reveals that T. gondii induces the host's autophagic pathway to boost amino acid levels in infected cells. The depletion of amino acids, in turn, influences the persistence of the parasite's chronic forms. Significantly, our investigation establishes the crucial role of host endoplasmic reticulum (ER)-phagy in the parasite's persistence within the host during latent infection.


Subject(s)
Amino Acids , Autophagy , Endoplasmic Reticulum , Toxoplasma , Toxoplasma/physiology , Amino Acids/metabolism , Animals , Endoplasmic Reticulum/metabolism , Mice , Toxoplasmosis/parasitology , Toxoplasmosis/metabolism , Humans , Brain/parasitology , Host-Parasite Interactions
14.
Parasitol Res ; 123(7): 286, 2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39046555

ABSTRACT

Despite being the initial choice for treating toxoplasmosis, sulfadiazine and pyrimethamine have limited effectiveness in eliminating the infection and were linked to a variety of adverse effects. Therefore, the search for new effective therapeutic strategies against toxoplasmosis is still required. The current work is the first research to assess the efficacy of spiramycin-loaded maltodextrin nanoparticles (SPM-loaded MNPs) as a novel alternative drug therapy against toxoplasmosis in a murine model. Fifty laboratory-bred Swiss albino mice were divided into five groups: normal control group (GI, n = 10), positive control group (GII, n = 10), orally treated with spiramycin (SPM) alone (GIII, n = 10), intranasal treated with SPM-loaded MNPs (GIV, n = 10), and orally treated with SPM-loaded MNPs (GV, n = 10). Cysts of Toxoplasma gondii ME-49 strain were used to infect the mice. Tested drugs were administered 2 months after the infection. Drug efficacy was assessed by counting brain cysts, histopathological examination, and measures of serum CD19 by flow cytometer. The orally treated group with SPM-loaded MNPs (GV) showed a marked reduction of brain cyst count (88.7%), histopathological improvement changes, and an increasing mean level of CD19 (80.2%) with significant differences. SPM-loaded MNPs showed potent therapeutic effects against chronic toxoplasmosis. Further research should be conducted to assess it in the treatment of human toxoplasmosis, especially during pregnancy.


Subject(s)
Disease Models, Animal , Nanoparticles , Polysaccharides , Spiramycin , Toxoplasmosis, Animal , Animals , Spiramycin/therapeutic use , Spiramycin/administration & dosage , Mice , Polysaccharides/administration & dosage , Polysaccharides/therapeutic use , Polysaccharides/pharmacology , Nanoparticles/chemistry , Toxoplasmosis, Animal/drug therapy , Toxoplasma/drug effects , Female , Brain/parasitology , Brain/pathology , Antiprotozoal Agents/administration & dosage , Antiprotozoal Agents/therapeutic use , Toxoplasmosis/drug therapy , Toxoplasmosis/parasitology , Drug Carriers
15.
mBio ; 15(8): e0072724, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-38975793

ABSTRACT

Toxoplasma gondii, the causative agent of toxoplasmosis, is an obligate intracellular parasite that infects warm-blooded vertebrates across the world. In humans, seropositivity rates of T. gondii range from 10% to 90% across communities. Despite its prevalence, few studies address how T. gondii infection changes the metabolism of host cells. In this study, we investigate how T. gondii manipulates the host cell metabolic environment by monitoring the metabolic response over time using noninvasive autofluorescence lifetime imaging of single cells, metabolite analysis, extracellular flux analysis, and reactive oxygen species (ROS) production. Autofluorescence lifetime imaging indicates that infected host cells become more oxidized and have an increased proportion of bound NAD(P)H compared to uninfected controls. Over time, infected cells also show decreases in levels of intracellular glucose and lactate, increases in oxygen consumption, and variability in ROS production. We further examined changes associated with the pre-invasion "kiss and spit" process using autofluorescence lifetime imaging, which also showed a more oxidized host cell with an increased proportion of bound NAD(P)H over 48 hours compared to uninfected controls, suggesting that metabolic changes in host cells are induced by T. gondii kiss and spit even without invasion.IMPORTANCEThis study sheds light on previously unexplored changes in host cell metabolism induced by T. gondii infection using noninvasive, label-free autofluorescence imaging. In this study, we use optical metabolic imaging (OMI) to measure the optical redox ratio (ORR) in conjunction with fluorescence lifetime imaging microscopy (FLIM) to noninvasively monitor single host cell response to T. gondii infection over 48 hours. Collectively, our results affirm the value of using autofluorescence lifetime imaging to noninvasively monitor metabolic changes in host cells over the time course of a microbial infection. Understanding this metabolic relationship between the host cell and the parasite could uncover new treatment and prevention options for T. gondii infections worldwide.


Subject(s)
Optical Imaging , Reactive Oxygen Species , Toxoplasma , Toxoplasma/metabolism , Optical Imaging/methods , Humans , Reactive Oxygen Species/metabolism , Toxoplasmosis/metabolism , Toxoplasmosis/parasitology , Animals , NADP/metabolism , Oxidation-Reduction , Glucose/metabolism , Host-Parasite Interactions
16.
mBio ; 15(8): e0121124, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-38995035

ABSTRACT

Intracellular infection by a pathogen induces significant rewiring of host cell signaling and biological processes. Understanding how an intracellular pathogen such as Toxoplasma gondii modulates host cell metabolism with single-cell resolution has been challenged by the variability of infection within cultures and difficulties in separating host and parasite metabolic processes. A new study from Gallego-Lopez and colleagues (G. M. Gallego-López, E. C. Guzman, D. E. Desa, L. J. Knoll, M. C. Skala, mBio e00727-24, 2024, https://doi.org/10.1128/mbio.00727-24) applies a quantitative imaging approach to evaluate the host cell metabolism during intracellular infection with Toxoplasma. This study provides important insights into host metabolic responses to Toxoplasma infection and offers a valuable tool to dissect the mechanisms underlying parasite infection and pathophysiology.


Subject(s)
Toxoplasma , Toxoplasmosis , Toxoplasma/metabolism , Toxoplasma/genetics , Humans , Toxoplasmosis/parasitology , Toxoplasmosis/metabolism , Host-Parasite Interactions , Animals
17.
Cell Mol Life Sci ; 81(1): 294, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38977495

ABSTRACT

The obligate intracellular parasite Toxoplasma gondii causes life-threatening toxoplasmosis to immunocompromised individuals. The pathogenesis of Toxoplasma relies on its swift dissemination to the central nervous system through a 'Trojan Horse' mechanism using infected leukocytes as carriers. Previous work found TgWIP, a protein secreted from Toxoplasma, played a role in altering the actin cytoskeleton and promoting cell migration in infected dendritic cells (DCs). However, the mechanism behind these changes was unknown. Here, we report that TgWIP harbors two SH2-binding motifs that interact with tyrosine phosphatases Shp1 and Shp2, leading to phosphatase activation. DCs infected with Toxoplasma exhibited hypermigration, accompanying enhanced F-actin stress fibers and increased membrane protrusions such as filopodia and pseudopodia. By contrast, these phenotypes were abrogated in DCs infected with Toxoplasma expressing a mutant TgWIP lacking the SH2-binding motifs. We further demonstrated that the Rho-associated kinase (Rock) is involved in the induction of these phenotypes, in a TgWIP-Shp1/2 dependent manner. Collectively, the data uncover a molecular mechanism by which TgWIP modulates the migration dynamics of infected DCs in vitro.


Subject(s)
Cell Movement , Dendritic Cells , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Protein Tyrosine Phosphatase, Non-Receptor Type 6 , Protozoan Proteins , Toxoplasma , Toxoplasma/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 11/genetics , Dendritic Cells/metabolism , Dendritic Cells/parasitology , Animals , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Humans , Mice , rho-Associated Kinases/metabolism , Toxoplasmosis/metabolism , Toxoplasmosis/parasitology , Toxoplasmosis/pathology , Mice, Inbred C57BL
18.
Exp Parasitol ; 263-264: 108799, 2024.
Article in English | MEDLINE | ID: mdl-39025462

ABSTRACT

One of the many warm-blooded hosts that toxoplasmosis-causing intracellular protozoan parasite Toxoplasma gondii can infect is humans. Cytokines are crucial to stimulate an effective immune response against T. gondii. Interleukin-33 (IL-33) is a unique anti-inflammatory cytokine that suppresses the immune response. The levels of cytokine gene expression are regulated by genetics, and the genetic polymorphisms of these cytokines play a functional role in this process. Single nucleotide polymorphisms (SNPs) are prognostic indicators of illnesses. This study aimed to determine whether toxoplasmosis interacts with serum levels of IL-33 and its SNP in miscarriage women as well as whether serum levels and IL-33 gene expression are related in toxoplasmosis-positive miscarriage women. Two hundred blood samples from patients and controls were collected from AL-Alawiya Maternity Teaching Hospital and AL-Yarmouk Teaching Hospital in Baghdad, Iraq from 2021 to 2022 in order to evaluate the serum level of IL-33 using ELISA test. For the SNP of IL-33, the allelic high-resolution approach was utilized, and real time-PCR was performed to assess gene expression. The results showed that compared to healthy and pregnant women, recurrent miscarriage with toxoplasmosis and recurrent miscarriage women had lower IL-33 concentrations. Additionally, there were significant differences among healthy women, pregnant women, and women with repeated miscarriage who experienced toxoplasmosis. Furthermore, no differences between patients and controls were revealed by gene expression data. The results revealed that recurrent miscarriage, pregnancy, and healthy women all had a slightly higher amount of the IL-33 gene fold. Additionally, the SNP of IL-33 data demonstrated that there was no significant genetic relationship between patients and controls. Recurrent miscarriage women with toxoplasmosis have showed significant differences from pregnant women in the genotypes GG and AA as well as the alleles A and G. There were notable variations between recurrent miscarriage with and without toxoplasmosis in terms of the genotypes AA and AC. The genotypes GG, AA, and allele A in recurrent miscarriage women with toxoplasmosis and recurrent miscarriage women is a protective factor. Taking together, there was a statistically significant negative correlation between toxoplasmosis and IL-33 gene expression, which calls for more quantitative investigation in order to fully comprehend the interaction of mRNA and protein.


Subject(s)
Abortion, Habitual , Interleukin-33 , Polymorphism, Single Nucleotide , Toxoplasmosis , Humans , Female , Interleukin-33/blood , Interleukin-33/genetics , Abortion, Habitual/genetics , Abortion, Habitual/blood , Abortion, Habitual/parasitology , Pregnancy , Iraq , Adult , Toxoplasmosis/blood , Toxoplasmosis/complications , Toxoplasmosis/parasitology , Gene Expression , Case-Control Studies , Young Adult , Enzyme-Linked Immunosorbent Assay , Toxoplasma/immunology , Toxoplasma/genetics , Real-Time Polymerase Chain Reaction , Genotype , Pregnancy Complications, Parasitic/blood , Pregnancy Complications, Parasitic/parasitology , Pregnancy Complications, Parasitic/genetics
19.
Parasitol Int ; 102: 102922, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38997003

ABSTRACT

BACKGROUND: Toxoplasmosis is caused by the protozoan parasite Toxoplasma gondii, a food- and water-borne zoonotic protozoan parasite that is able to infect almost all warm-blooded vertebrates. It has a major effect on public health, particularly in underdeveloped nations. Immune-competent individuals typically exhibit no symptoms or experience a mild influenza-like sickness, while there is a possibility of severe manifestation and fatal or high-risk for life-threatening diseases in immunocompromised people like pregnant women and HIV/AIDS patients and lead to severe pathological effects on the fetus. METHOD: We conducted a systematic search of databases (PubMed, Google Scholar, Science Direct, EMBASE, and Scopus) using the PRISMA criteria. We used specific keywords such as Toxoplasma gondii, Toxoplasmosis, pregnant women, prevalence, HIV/AIDS, and worldwide studies published from 2018 to 2022. We use Stata (version 14) software to estimate the pooled prevalence and heterogeneity of toxoplasmosis in pregnant women and HIV-infected people using a random-effects model and the Cochran's Q-test, respectively. The Joanna Briggs Institute Critical Appraisal Instrument and Egger's regression asymmetry test were used to assess study quality and publication bias, respectively, while the single study omission analysis was used to test the robustness of a pooled estimate. RESULTS: We included and analyzed a total of 12,887 individuals in this review. The pooled prevalence of T. gondii in this review was 40% (95% CI = 0.31-0.50). The sub-group analysis revealed that the evaluation included 11,967 pregnant women. In pregnant women, the pooled sero-prevalence was 40% (95% CI = 0.31-0.50). In pregnant women and HIV/AIDS patients, 920 individuals were evaluated, and the pooled sero-prevalence was 41% (95% CI = 0.20-0.61). CONCLUSION: This review identified an overall sero-prevalence of Toxoplasma infection of 40% among pregnant women and HIV/AIDS. The expansion of prevention and control strategies, with a primary focus on enhancing educational initiatives, is necessary to avoid reactivation and stop the spread of infection, so investigative sero-prevalence is important work among pregnant women and HIV patients. In order to achieve a comprehensive explanation of the disease condition and reach this goal, we conducted a systematic review and meta-analysis in Worldwide for future use.


Subject(s)
HIV Infections , Toxoplasma , Toxoplasmosis , Humans , Female , Toxoplasmosis/epidemiology , Toxoplasmosis/parasitology , Pregnancy , Toxoplasma/immunology , HIV Infections/epidemiology , HIV Infections/complications , Prevalence , Pregnancy Complications, Infectious/epidemiology , Pregnancy Complications, Infectious/parasitology , Acquired Immunodeficiency Syndrome/complications , Acquired Immunodeficiency Syndrome/epidemiology , Global Health , Seroepidemiologic Studies
20.
J Cell Mol Med ; 28(14): e18542, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39046369

ABSTRACT

This study aims to investigate the relationship between toxoplasmosis and this pathway, which may be effective in the formation of epilepsy by acting through the HMGB1/RAGE/TLR4/NF-κB signalling pathway in patients with idiopathic epilepsy. In the study, four different experimental groups were formed by selecting Toxoplasma gondii IgG positive and negative patients with idiopathic epilepsy and healthy controls. Experimental groups were as follows: Group 1: Epilepsy+/Toxo- (E+, T-) (n = 10), Group 2: Epilepsy-/Toxo- (E-, T-) (n = 10), Group 3: Epilepsy-/Toxo+ (E-, T+) (n = 10), Group 4: Epilepsy+/Toxo+ (E+, T+) (n = 10). HMGB1, RAGE, TLR4, TLR1, TLR2, TLR3, IRAK1, IRAK2, IKBKB, IKBKG, BCL3, IL1ß, IL10, 1 L8 and TNFα mRNA expression levels in the HMGB/RAGE/TLR4/NF-κB signalling pathway were determined by quantitative simultaneous PCR (qRT-PCR) after collecting blood samples from all patients in the groups. Statistical analysis was performed by one-way ANOVA followed by LSD post-hoc tests, and p < 0.05 was considered to denote statistical significance. The gene expression levels of HMGB1, TLR4, IL10, IL1B, IL8, and TLR2 were significantly higher in the G1 group than in the other groups (p < 0.05). In the G3 group, RAGE and BCL3 gene expression levels were significantly higher than in the other groups (p < 0.05). In the G4 group, however, IRAK2, IKBKB, and IKBKG gene expression levels were significantly higher than in the other groups (p < 0.05). HMGB1, TLR4, IRAK2, IKBKB, IL10, IL1B, IL1B, and IL8 in this signalling pathway are highly expressed in epilepsy patients in G1 and seizures occur with the stimulation of excitatory mechanisms by acting through this pathway. The signalling pathway in epilepsy may be activated by HMGB1, TLR4, and TLR2, which are considered to increase the level of proinflammatory cytokines. In T. gondii, this pathway is activated by RAGE and BCL3.


Subject(s)
Epilepsy , HMGB1 Protein , NF-kappa B , Signal Transduction , Toll-Like Receptor 4 , Toxoplasmosis , Humans , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 4/genetics , HMGB1 Protein/metabolism , HMGB1 Protein/genetics , NF-kappa B/metabolism , NF-kappa B/genetics , Male , Female , Epilepsy/metabolism , Epilepsy/genetics , Epilepsy/parasitology , Adult , Toxoplasmosis/parasitology , Toxoplasmosis/metabolism , Toxoplasmosis/complications , Toxoplasmosis/blood , Toxoplasmosis/genetics , Receptor for Advanced Glycation End Products/metabolism , Receptor for Advanced Glycation End Products/genetics , Case-Control Studies , Young Adult , Middle Aged , Antigens, Neoplasm , Mitogen-Activated Protein Kinases
SELECTION OF CITATIONS
SEARCH DETAIL