Next Article in Journal
Harnessing the Loop: The Perspective of Circular RNA in Modern Therapeutics
Previous Article in Journal
Advancing Reversed-Phase Chromatography Analytics of Influenza Vaccines Using Machine Learning Approaches on a Diverse Range of Antigens and Formulations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Toxoplasma gondii at the Host Interface: Immune Modulation and Translational Strategies for Infection Control

by
Billy J. Erazo Flores
and
Laura J. Knoll
*
Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI 53706, USA
*
Author to whom correspondence should be addressed.
Vaccines 2025, 13(8), 819; https://doi.org/10.3390/vaccines13080819 (registering DOI)
Submission received: 30 May 2025 / Revised: 18 July 2025 / Accepted: 30 July 2025 / Published: 31 July 2025
(This article belongs to the Special Issue Intracellular Parasites: Immunology, Resistance, and Therapeutics)

Abstract

Toxoplasma gondii is an intracellular protozoan found worldwide that is capable of infecting nearly all warm-blooded animals, including humans. Its parasitic success lies in its capacity to create chronic infections while avoiding immune detection, altering host immune responses, and disrupting programmed cell death pathways. This review examines the complex relationship between T. gondii and host immunity, focusing on how the parasite influences innate and adaptive immune responses to survive in immune-privileged tissues. We present recent findings on the immune modulation specific to various parasite strains, the immunopathology caused by imbalanced inflammation, and how the parasite undermines host cell death mechanisms such as apoptosis, necroptosis, and pyroptosis. These immune evasion tactics enable prolonged intracellular survival and pose significant challenges for treatment and vaccine development. We also review advancements in therapeutic strategies, including host-directed approaches, nanoparticle drug delivery, and CRISPR-based technologies, along with progress in vaccine development from subunit and DNA vaccines to live-attenuated candidates. This review emphasizes the importance of T. gondii as a model for chronic infections and points out potential avenues for developing innovative therapies and vaccines aimed at toxoplasmosis and similar intracellular pathogens.

1. Introduction

Toxoplasma gondii is an obligate intracellular protozoan parasite and the causative agent of toxoplasmosis, a potentially deadly disease [1]. It is estimated that approximately 30% of the global human population is infected with this parasite [2]. In most healthy individuals, T. gondii infection is either asymptomatic or presents with mild flu-like symptoms. The host immune system can typically control the infection damage if the strain encountered is not highly virulent and the host is immunocompetent. In cases of congenital infection or immunocompromised individuals, toxoplasmosis can cause severe complications, including fatal encephalitis, ocular damage, and stillbirth [3,4,5]. The early symptomatic phase of the infection, known as the acute stage, is characterized by the rapid replication of the parasite tachyzoite form. During this stage, tachyzoites disseminate throughout the host body. In reaction to the host immune response and other factors that remain incompletely understood, tachyzoites transition into the slow-growing encysted stage of the parasite, called bradyzoites [6]. This transition marks the onset of the chronic infection. In vitro, the conversion of tachyzoites to bradyzoites has been observed under high pH conditions, exposure to sodium arsenite, and temperature changes [7,8]. However, the precise mechanisms regulating this conversion in vivo are not yet fully characterized. Although T. gondii is capable of infecting nearly all nucleated cells [9], bradyzoite cysts are primarily found in mature skeletal muscle and brain neurons, potentially remaining in this area indefinitely and undetected [10]. With a weakened immune system, reactivation of the bradyzoite cysts to tachyzoites can occur [6] leading to severe pathology, such as encephalitis. T. gondii infection has been connected to various neurological effects in hosts, especially during the chronic stage. In both animal studies and humans, long-lasting bradyzoite cysts in neural tissue are associated with neuroinflammation [11], changes in neurotransmitter regulation [12,13], and behavioral alterations [12,14]. These findings raise concerns about the parasite’s possible influence on host cognition and neuropsychiatric disorders.
The host immune response against T. gondii is a complex, multi-layered defense system involving both innate and adaptive immunity. During the early stages of infection, the innate immune system plays a critical role in detecting and responding to the parasite. While it is critical for the host to develop a robust but balanced innate immune response, T. gondii has evolved sophisticated strategies to manipulate host defense pathways, allowing it to persist within host cells. This delicate interplay between host defense and parasite survival determines the outcome of the infection.
This review aims to provide a comprehensive overview of the host immune responses to T. gondii infection, emphasizing the delicate balance between protective immunity and immune evasion strategies employed by the parasite. It will explore the host’s innate and adaptive immune mechanisms, including cytokine signaling, cellular immune responses, and the activation of various immune pathways. Finally, the review will discuss the implications of these interactions for host–pathogen dynamics and potential therapeutic strategies.

Systematic Literature Search Methodology

To ensure a thorough coverage of relevant studies for this review, a systematic search was conducted across multiple scientific databases. Searches were performed in PubMed, Web of Science, and Google Scholar, covering articles published up to 2024. Keywords incorporated various combinations such as “Toxoplasma gondii,” “immune response,” “programmed cell death,” “host–pathogen interaction,” “immune evasion,” “vaccine development,” “host-directed therapy,” and “parasite persistence.” Boolean operators (AND/OR) were used to refine and include relevant intersections of topics.
The inclusion criteria encompassed original research articles, reviews, and preclinical or clinical studies focusing on T. gondii biology, host immune responses (both innate and adaptive), immune evasion mechanisms, programmed cell death, and therapeutic or vaccine strategies. Preference was given to peer-reviewed publications in English and available as full texts. Studies unrelated to T. gondii, lacking sufficient mechanistic detail, or non-peer-reviewed content were excluded. Additional relevant papers were found through manual screening of reference lists from key articles. In total, 150 articles were thoroughly reviewed, with 121 included in the final analysis. The selected literature was critically assessed for relevance, methodological rigor, and its contribution to current understanding. The articles included are cited, with an average per article of 236.52, and an H-index of 73.
To resolve conflicting results across various experimental models, including murine and human cell lines, and in vivo systems, a comparative evidence synthesis approach was employed. This method integrates data considering experimental context, strain differences, and immune responses. The emphasis was on ensuring mechanistic consistency and reproducibility across models to identify broadly supported conclusions, while also acknowledging model-specific limitations.

2. Life Cycle of T. gondii

T. gondii has a complex life cycle that includes both asexual and sexual stages. The asexual cycle occurs in all warm-blooded animals, including humans, where the parasite exists in two main forms: tachyzoites and bradyzoites [15,16]. In contrast, the sexual cycle is restricted to the feline intestine, the only environment in which T. gondii undergoes gametogenesis and oocyst formation. It was recently discovered that the sexual development of T. gondii is triggered by high levels of linoleic acid [17]. Linoleic acid accumulates due to the feline-specific lack of intestinal delta-6-desaturase activity, the rate limiting step in the conversion of linoleic acid to arachidonic acid. It was also recently observed that inhibition of T. gondii HDAC3 (TgHDAC3) hyperacetylates T. gondii histones and induces the expression of sexual stage genes [18]. The T. gondii transcription factors AP2XII-1 and AP2XI-2 bind DNA as heterodimers at merozoite promoters and recruit MORC and HDAC3, thereby limiting chromatin accessibility and transcription of early sexual stage genes [19]. The fact that inhibition of TgHDAC3 initiates the T. gondii sexual cycle suggests the presence of HDAC inhibitor (s) unique to the feline gut. Felines primarily become infected by ingesting tissue cysts containing bradyzoites from prey animals that have acquired toxoplasmosis. Cats can be infected with oocysts, but the prepatent period is longer compared to tissue cysts [20]. Within the feline intestinal epithelium, the parasite differentiates into male and female gametes, which fuse to form oocysts that are subsequently shed in the feces [21,22]. Oocysts undergo sporulation in the environment to produce infectious sporozoites, which can infect new intermediate hosts and thereby complete the life cycle [23].

2.1. Routes of Infection

The primary routes of human infection include ingesting contaminated food or water, zoonotic transmission, and congenital transmission [23]. The most common route is the oral ingestion of tissue cysts or oocysts [24]. Humans can become infected by consuming undercooked or raw meat that contains bradyzoite cysts. In addition, ingesting water, fruits, or vegetables contaminated with sporulated oocysts shed by infected cats represents a significant transmission pathway and may lead to more severe disease than infections derived from tissue cysts [25].
Once ingested, the cyst wall is dissolved by proteolytic enzymes found in the stomach and small intestine [7]. Tachyzoites are released in the gastrointestinal tract to infect cells and travel through the bloodstream to reach tissues such as the brain, eyes, and muscles. Congenital transmission occurs when a pregnant woman acquires a primary infection during pregnancy. Tachyzoites can cross the placental barrier, leading to congenital toxoplasmosis, which may cause severe complications such as neurological impairment, ocular lesions, or stillbirth [26]. The risk of transmission and the severity of the disease largely depend on the gestational age at the time of infection. Maternal infections during the first trimester have reduced fetal transmission rates, but are associated with more severe outcomes than those occurring later in pregnancy, with high levels of fetal transmission [27]. Direct contact with infected cat feces is a less common but important transmission route. Cats, as the definitive hosts, shed oocysts in their feces after ingesting infected prey. Humans may inadvertently ingest oocysts through improper handling of cat litter or contact with food grown in contaminated soil, particularly uncooked fruits and vegetables. Understanding these transmission routes is critical for effective prevention, especially among high-risk populations such as pregnant women and immunocompromised individuals. Preventive measures, including thoroughly cooking meat, washing fruits and vegetables, and practicing proper hygiene when handling cat litter or soil, can significantly reduce the risk of infection.

2.2. Acute and Chronic Infection

T. gondii infection progresses through two main stages: acute and chronic. In humans and mice, acute infection is characterized by the rapid proliferation and spread of the tachyzoite form. After oral ingestion of tissue cysts or oocysts, bradyzoites and sporozoites differentiate into tachyzoites that disseminate systemically through the bloodstream by the circulation of tachyzoite-infected leukocytes in the blood [28,29]. This stage often triggers a strong innate immune response, characterized by elevated levels of IL-12 and IFN-γ, which are critical for limiting parasite replication [30].
After the parasite has spread throughout the body, reaching areas such as the brain and muscle tissue, immune pressure drives the transition from tachyzoites to bradyzoites [6,31]. This slow-replicating form resides within long-lived tissue cysts, marking the onset of chronic infection. In humans, chronic toxoplasmosis is usually asymptomatic but can reactivate in immunosuppressed individuals, leading to severe conditions like encephalitis [4,32]. The ability of T. gondii to establish a lifelong chronic infection reflects its evolved strategies to evade immune clearance and highlights the challenges in therapeutic eradication.

3. Host Immune Response to T. gondii

T. gondii has evolved a complex life cycle and sophisticated strategies that enable it to thrive within host cells, evade immune detection, and manipulate host cellular processes for prolonged survival. A defining characteristic of T. gondii is its ability to infect nearly all nucleated cells and replicate rapidly within them [33]. Once inside a host cell, the parasite reprograms host functions to create a protective intracellular niche, exploit host resources, and evade immune clearance [34,35,36,37]. It can also disrupt critical host defense mechanisms, including programmed cell death (PCD) [38,39,40]. Understanding how the host immune system responds to T. gondii is crucial, as it determines the outcome of the infection and provides insight into potential therapeutic targets for limiting parasite replication and preventing disease progression.

3.1. Innate Immune Response

Survival during toxoplasmosis relies on a robust and balanced innate immune response. As the first line of defense, immune cells in the intestinal epithelium, including innate lymphoid cells, are among the initial responders to the parasite during oral infection [41,42,43]. These cells detect the presence of the parasite and initiate early immune responses to prevent its spread. Detection is facilitated by pattern recognition receptors, such as Toll-like receptors (TLRs), which identify pathogen-associated molecular patterns in the parasite [43,44,45].
Mice possess 13 TLRs, while humans express 10 [46]. In mice, TLR11 and TLR12 identify T. gondii profilin [47,48], triggering a MyD88-dependent signaling cascade [48,49] and the subsequent production of IL-12 by CD8α+ and CD8α dendritic cells [50,51,52,53]. IL-12 then stimulates natural killer cells and T cells to produce IFN-γ [54,55], a critical cytokine for controlling the parasite. In humans, TLR11 and TLR12 are non-functional [56]. Instead, detection is likely mediated through other TLRs, such as TLR3, TLR7, TLR8, and TLR9, which also signal through MyD88 to induce IL-12 [43,56].
MyD88, IL-12, and IFN-γ are critical for resistance to T. gondii; mice lacking any of these components quickly succumb to infection [52,57,58]. IFN-γ plays a central role in activating antimicrobial pathways, including the induction of immunity-related GTPases (IRGs) and guanylate-binding proteins (GBPs) [59,60], which disrupt the parasitophorous vacuole and compromise parasite survival [61,62]. Furthermore, nitric oxide (NO) produced by activated macrophages aids in parasite killing, although excessive NO production can lead to immunopathology [43,63,64].

3.2. Adaptive Immune Response

The adaptive immune response to T. gondii is initiated and shaped by early proinflammatory cytokines, notably IL-12 [65]. This response encompasses both B and T lymphocytes. Th1-polarized CD4+ T cells play a key role in the secretion of IFN-γ [66], which enhances macrophage activation [67,68], promotes antigen presentation [30], and stimulates the creation of reactive oxygen and nitrogen species to eradicate intracellular parasites [69,70]. Similarly, CD8+ T cells release IFN-γ [71] and eliminate T. gondii-infected cells via perforin and granzymes [72,73]. This cytotoxic function is crucial for eliminating the parasite when it is already present intracellularly. B cells assist the adaptive response by generating antigen-specific antibodies. During the early stage of acute infection, IgM antibodies are produced, whereas IgG antibodies are created later to provide long-term protection. These antibodies neutralize extracellular tachyzoites, hinder additional invasion of host cells, and are used as markers of exposure [1].

4. Immunopathology: Balancing Inflammation and Regulation

In both mice and humans, T. gondii elicits a robust proinflammatory Th1-polarized immune response that is pivotal for controlling the infection [65,74]. In immunocompetent individuals, this response usually eliminates most parasites, with any remaining ones confined to the tissue cysts, resulting in an asymptomatic or mild infection. However, failing to mount an effective proinflammatory response leads to uncontrolled parasite replication and severe disease manifestations such as toxoplasmic encephalitis [75].
While inflammation is essential to restrain parasite replication, an excessively vigorous immune response can cause collateral tissue damage [76,77]. Common sites of immunopathology include the brain and the eye, where intense inflammation can harm host tissues. For instance, oral high-dose infection of mice with T. gondii can result in the overproduction of inflammatory cytokines, leading to severe damage in the small intestine and necrotizing ileitis [78]. Similarly, immune-driven inflammation can damage retinal tissues in the eye, contributing to vision loss due to ocular toxoplasmosis [79]. These examples highlight the need for a balanced immune response that effectively eliminates the parasite while minimizing tissue injury.
To counteract immunopathology, the host employs anti-inflammatory mechanisms. The cytokines IL-10 and TGF-β and regulatory T cells (Tregs) are key players in this regulation. Tregs secrete IL-10 and TGF-β to suppress inflammatory mediators such as IFN-γ and IL-12 to reduce the activity of overactive effector T cells [80,81,82]. IL-10 plays a particularly crucial role in regulating Th1 responses; mouse models lacking IL-10 exhibit elevated levels of IFN-γ and IL-12, leading to lethal inflammation and necrotic liver lesions, even when parasite burdens are controlled [83]. Increased IL-10 or Treg activity can reduce inflammation, as seen in acute ocular toxoplasmosis models, where IL-10 regulates inflammation [84]. TGF-β, produced by Tregs and resident tissue cells, also helps suppress local inflammation and protects sensitive tissues such as neurons and retinal cells [85,86].
T. gondii can exploit these anti-inflammatory pathways to enhance its persistence. Parasite molecules, including microneme proteins MIC1 and MIC4, have been shown to induce IL-10 production in host cells [87], promoting a permissive environment that limits immune activation. By boosting IL-10 expression and expanding Treg responses, the parasite tempers host immunity to avoid elimination. This immunomodulatory strategy enables T. gondii to establish long-term chronic infections, particularly in immune-privileged tissues like the brain and eye, often with limited clinical symptoms. Therefore, the dynamic balance between inflammation and regulation is central to the pathogenesis of toxoplasmosis: a properly calibrated immune response controls the infection while minimizing damage, whereas an imbalance in either direction can lead to uncontrolled disease or tissue injury.

5. Strain-Specific Immune Responses

The host immune response to T. gondii is significantly affected by the genetic background of the parasite. The three primary clonal lineages—Type I, Type II, and Type III—demonstrate distinct profiles of virulence, immune modulation abilities, and disease outcomes in experimental models and natural infections [88,89]. Type I strains are highly virulent in mice and are characterized by rapid replication and a low lethal dose [90]. These strains suppress host immunity by secreting virulence factors such as ROP18 and ROP5, which antagonize IFN-γ induced IRGs [90,91]. As a result, the host’s innate and adaptive immune responses are quickly overwhelmed, often resulting in acute mortality [92]. Type II strains are less virulent but more immunogenic. They elicit robust IL-12 and IFN-γ responses, which are essential for controlling the parasite. These strains are commonly used in immunological studies due to their ability to establish chronic infections marked by tissue cysts in the brain and muscles. However, the strong inflammatory responses they provoke can contribute to immunopathology, as seen in ocular and cerebral toxoplasmosis models. In mice, type III strains are considered avirulent and induce a comparatively weak immune response. They stimulate lower levels of proinflammatory cytokines and minimal immune activation.
Beyond the canonical three clonal lineages, atypical and recombinant strains are common in South America [93], and exhibit a broad spectrum of virulence and immunomodulatory profiles. Some of these strains show high replication capacity and effective immune evasion mechanisms, presenting a greater risk of severe disease even in immunocompetent hosts. Ultimately, the outcome of infection, whether asymptomatic persistence or severe disease, is influenced by the complex interaction between the parasite genotype and the host’s immune competence. Understanding strain-specific immune responses is crucial for improving vaccine development, therapeutic interventions, and accurate risk assessment across different populations.

6. Programmed Cell Death in Host Defense

PCD plays a critical role in the host’s defense against intracellular pathogens such as T. gondii. By eliminating infected cells, PCD removes the intracellular niche, deprives the pathogen of access to host resources, and limits further spread. However, the type of cell death pathway activated, whether apoptosis, necroptosis, or another form, depends on various factors, including the pathogen’s characteristics, infectious burden, tissue environment, and the host’s immune status. Notably, even the same pathogen may trigger different forms of PCD based on the strain type and stage of infection, adding further complexity to the host’s response. This immune decision becomes even more challenging because apicomplexan parasites like T. gondii have developed sophisticated strategies to interfere with or subvert host PCD pathways for their benefit. Consequently, while PCD is essential for pathogen control, it also poses potential risks, including damage to surrounding host tissues due to excessive inflammation and immune cell recruitment [94].
Apoptosis is a non-inflammatory form of PCD that is characterized by the controlled fragmentation of cellular contents into membrane-bound apoptotic bodies, which are then phagocytosed without inducing inflammation. This containment is particularly beneficial in immune-privileged areas, such as the brain, where limiting immune-mediated damage is crucial. In the context of toxoplasmosis, apoptosis may aid in reducing neuroinflammation and protecting against severe pathology [95], especially in immunocompromised individuals. T. gondii can inhibit or delay apoptotic pathways, enabling continued intracellular replication. In contrast, necroptosis is a proinflammatory form of PCD that involves membrane rupture and the release of intracellular contents, which act as damage-associated molecular patterns, leading to the recruitment of immune cells and an amplification of inflammatory responses. This process can enhance pathogen clearance by exposing the parasite to immune effectors in the extracellular environment. However, if not tightly regulated, necroptosis can cause excessive tissue damage and contribute to immunopathology.
Thus, the host faces a strategic dilemma: inducing apoptosis may limit tissue damage but allow for parasite survival, while triggering necroptosis may enhance immune clearance at the expense of harming host tissues. The choice between these pathways reflects the ongoing evolutionary arms race between host defenses and parasite survival strategies and highlights the complexity of host–pathogen interactions.

7. Subversion of Host Cell Death and Immune Pathways by T. gondii

T. gondii has evolved various strategies to manipulate host cell death pathways and immune responses, thereby promoting its survival, replication, and long-term persistence. Apoptosis is significantly inhibited in T. gondii-infected cells. The parasite interferes with the intrinsic (mitochondrial) and extrinsic (death receptor) apoptotic pathways through various mechanisms, including the prevention of cytochrome c release, suppression of caspase activation, modulation of mitochondrial integrity, apoptosome assembly, and signaling intermediates [37]. Infected cells become resistant to normally lethal stimuli such as Fas ligand [96,97], growth factor withdrawal [98], and cytotoxic granzyme B [99] as well as the activation of pro-survival pathways like NF-κB [100]. By preventing premature host cell death, T. gondii preserves its intracellular niche and maintains an optimal environment for replication.
The inhibition of caspase-8 and death receptor signaling also impairs one arm of necroptosis. Nevertheless, under certain conditions, such as strong TNF-α or IFN-γ signaling, the activation of the RIPK3 pathway can still drive necroptosis in the absence of apoptosis. To avoid this, T. gondii appears to restrict the upstream signals necessary for necroptotic activation. The parasite also actively limits pyroptosis, a rapid, inflammasome-dependent form of cell death. In hosts with functional inflammasome sensors (e.g., NLRP1, NLRP3), macrophages may detect T. gondii and initiate pyroptosis [101]. However, in cases of regulating pyroptosis, T. gondii can potentially maintain the integrity of its parasitophorous vacuole, protecting its components from detection in the cytosol and thereby preventing inflammasome activation. By modulating apoptosis, necroptosis, and pyroptosis, T. gondii ensures the survival of its host cell until the parasite is ready to egress and infect new cells.
Beyond interfering with host cell death, T. gondii modulates immune signaling and antigen presentation to prevent immune clearance [102,103,104]. To counter the Th-1 type immune response needed for host control of the parasite, T. gondii deploys a range of secreted effectors. For instance, the dense granule protein GRA15 (expressed in Type II strains) activates NF-κB in host cells, enhancing proinflammatory cytokine production, including IL-12 [105]. In contrast, more virulent Type I strains expressing ROP18 can potentially suppress NF-κB signaling, thereby reducing IL-12 output and hindering early immune activation [106]. Also, the secreted effector TgIST traffics to the host nucleus, disrupting STAT1-dependent gene expression, a central aspect of IFN-γ signaling [107]. T. gondii infection also downregulates MHC class II expression on antigen-presenting cells, primarily by blocking STAT1 nuclear translocation and transcriptional activity [107]. This suppression of antigen presentation aids the parasite in evading detection by CD4+ T cells. Simultaneously, the parasite promotes host-derived immunosuppressive molecules, such as IL-10 and SOCS proteins [108], which further dampen Th1 responses and prevent complete immune activation.
T. gondii also directly interferes with intracellular immune effectors. In IFN-γ stimulated cells, particularly in mice, GTPases such as IRGs and GBPs target the parasitophorous vacuole, leading to its rupture and the destruction of the parasite. To counteract this, virulent strains of T. gondii express a set of rhoptry proteins, including ROP18, ROP17, ROP5, and GRA7, that phosphorylate and inactivate IRGs, preventing their oligomerization and targeting of the vacuole [109]. This action effectively turns off a major innate defense mechanism. A similar strategy applies to GBPs, whose recruitment to the vacuole is also hindered by parasite virulence factors. Although GBPs are IFN-inducible in mice and humans, their precise interactions with T. gondii effectors are still being unraveled. By blocking the recruitment and activation of IRGs and GBPs, T. gondii avoids vacuole lysis, exposure of its antigens, and subsequent inflammasome activation. This strategy allows the parasite to remain hidden and protected within its host cell.
Through the coordinated action of rhoptry and dense granule effectors like ROP18, GRA15, and TgIST, T. gondii manipulates both cell-intrinsic and immune signaling pathways. These mechanisms enable the parasite to evade immune clearance, replicate undetected, and establish chronic infections that may persist for the host’s lifetime. This highly evolved subversion of host defenses highlights T. gondii success as a widespread and enduring intracellular pathogen.

8. Therapeutic and Vaccine Strategies

Current treatments for toxoplasmosis primarily depend on antifolate drug combinations targeting the actively replicating tachyzoite stage of the parasite. The most common regimen includes pyrimethamine and sulfadiazine, supplemented with folinic acid to reduce bone marrow toxicity [110]. Alternatives like trimethoprim-sulfamethoxazole or combinations of pyrimethamine with clindamycin, azithromycin, or atovaquone are also used [110]. While these therapies can effectively manage acute infections, they have significant limitations. Chief among them are drug toxicity and the inability to eliminate the latent bradyzoite cysts that persist in tissues, particularly in the brain and muscles. Consequently, infections can reactivate if the immune system becomes compromised. Although rare, reports of T. gondii strains resistant to pyrimethamine, sulfonamides, and macrolides raise concerns about potential treatment failures [111]. These issues underscore the urgent need for new therapies that are safer and capable of targeting the chronic stages of infection.
To address these challenges, researchers are exploring various innovative therapeutic strategies. Host-directed therapies aim to enhance the immune system’s capacity to control infection by modulating cytokines or epigenetic regulators. Some repurposed drugs, such as the antidepressant sertraline, have demonstrated promising anti-toxoplasmosis effects through host-mediated pathways [112]. Sertraline treatment inhibited T. gondii proliferation and neuroinflammation by downregulating TNFR1/NF-κB signaling, thereby reducing parasite-induced depression-like behaviors [112]. Nanoparticle-based drug delivery systems are also under development to enhance drug uptake by infected cells and improve infiltration into hard-to-reach areas such as the brain [113,114,115]. Combining antiparasitic agents with copper-based nanoparticles in mouse models has improved cyst clearance [116]. Another cutting-edge approach involves CRISPR/Cas9 genome editing, which is being employed to identify essential parasite genes and create genetically attenuated strains that cannot form tissue cysts. These strains not only present targets for drug development but may also serve as candidates for live-attenuated vaccines. Emerging immunotherapeutic strategies include the adoptive transfer of T. gondii specific T cells and the use of immune checkpoint inhibitors like PD-1/PD-L1 blockade, which can reinvigorate exhausted T cells and diminish brain cyst burden in chronically infected mice [117]. Although these approaches show promising results in experimental settings, a systematic assessment of their success rates, model-specific factors, and translational challenges is crucial to evaluate their clinical feasibility and prioritize future development.
Vaccine development for human toxoplasmosis remains in the preclinical stage. Currently, no licensed vaccines are available for humans, primarily due to the parasite’s complex life cycle and the requirement for robust cell-mediated immunity, along with significant translational obstacles in moving from animal models to human application. Nevertheless, various experimental approaches are being investigated. Subunit and DNA vaccines encoding key antigens have demonstrated partial protection in animal models, especially when paired with potent adjuvants. Nanoparticle-based and mRNA vaccine platforms have shown promise by enhancing antigen presentation and immune activation. However, none of these approaches has yet achieved the sterilizing, long-term immunity necessary to fully prevent infection and cyst formation. More progress has been made in veterinary medicine. A live-attenuated vaccine (Toxovax®), based on the S48 strain of T. gondii, is successfully used in sheep to prevent abortion and reduce tissue cyst burden [118]. Nevertheless, this vaccine’s short shelf life, high production cost, and potential risk of reversion to virulence have limited its use to only a few countries. Similar efforts are underway in cats, the definitive hosts of the parasite. Experimental cat vaccines, including genetically modified strains lacking the HAP2 gene necessary for sexual development, have been found to prevent oocyst shedding, therefore interrupting environmental transmission [119]. Other attenuated strains, like T-263, also show promise in reducing oocyst production [120]. The T-263 strain has conferred sterilizing immunity in kittens vaccinated with T-263; they shed no oocysts when challenged, whereas 84% of unvaccinated kittens shed oocysts [121]. Although no commercial cat vaccine is currently available, partly due to cost and logistical hurdles in vaccinating cat populations, these advances in animal vaccines are vital to reducing zoonotic transmission through improved veterinary control.
In summary, while current therapies for toxoplasmosis can be lifesaving, they are limited by toxicity, restricted efficacy against tissue cysts, and the threat of resistance. Emerging therapies, including host-modulating drugs, nanocarriers, gene editing, and immunotherapies, offer promising new directions for more effective treatment. Vaccine development remains an ongoing challenge but has shown encouraging results in animal models and veterinary applications. Continued translational research bridging clinical, immunological, and veterinary advances will be essential for developing safe, broadly effective tools to combat this globally prevalent parasite.

9. Conclusions

Toxoplasmosis poses a considerable health risk to humans, wildlife, pets, and livestock. This risk arises from its ability to persist within cells, evade and modulate innate and adaptive immune responses, and establish long-term niches in immune-privileged areas like the brain. As this review highlights, T. gondii not only reveals the delicate balance between protective immunity and immunopathology but also exposes exploitable vulnerabilities in host–parasite interaction.
Enhancing our understanding of how T. gondii manipulates PCD, subverts antigen presentation, and alters cytokine networks has profound implications for the design of host-directed therapies. Developing safe and effective vaccines, especially those capable of blocking chronic infection or transmission, remains a priority. Innovative approaches, including CRISPR-attenuated strains, nanoparticle-based platforms, and mRNA vaccines, offer new hope in tackling these challenges.
Future research should focus on underexplored aspects such as the biology of bradyzoite cysts, strain-specific immunomodulation, and immune responses in tissue-resident niches. Leveraging omics technologies, in vivo models, and systems immunology will be essential to bridge these knowledge gaps. Ultimately, T. gondii provides a powerful model for understanding intracellular persistence, immune modulation, and the co-evolution of host–pathogen dynamics. Insights gained from this parasite can inform therapeutic and vaccine innovation for toxoplasmosis and a range of chronic infectious diseases, advancing our broader understanding of host immunity and translational immunology.

Author Contributions

Writing—original draft preparation, B.J.E.F.; Writing—review and editing, L.J.K. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the National Institutes of Health National Institute of Allergy and Infectious Diseases R01AI144016-01 (L.K.), graduate training in cellular and molecular pathogenesis of human diseases T32 fellowship (1T32GM135119-01) (B.E.F.), the parasitology and vector biology training program T32 fellowship (5T32AI007414-27) (B.E.F.), the Robert H. and Carol L. Deibel distinguished graduate fellowship in food safety research (B.E.F.). Any opinions, findings, conclusions, or recommendations expressed in this material are those of the authors and do not necessarily reflect the views of the funding agencies.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Liu, Q.; Wang, Z.-D.; Huang, S.-Y.; Zhu, X.-Q. Diagnosis of Toxoplasmosis and Typing of Toxoplasma gondii. Parasites Vectors 2015, 8, 292. [Google Scholar] [CrossRef] [PubMed]
  2. Dubey, J.P. Outbreaks of Clinical Toxoplasmosis in Humans: Five Decades of Personal Experience, Perspectives and Lessons Learned. Parasites Vectors 2021, 14, 263. [Google Scholar] [CrossRef]
  3. McAuley, J.B. Congenital Toxoplasmosis. J. Pediatr. Infect. Dis. Soc. 2014, 3, S30–S35. [Google Scholar] [CrossRef]
  4. Wang, Z.-D.; Liu, H.-H.; Ma, Z.-X.; Ma, H.-Y.; Li, Z.-Y.; Yang, Z.-B.; Zhu, X.-Q.; Xu, B.; Wei, F.; Liu, Q. Toxoplasma gondii Infection in Immunocompromised Patients: A Systematic Review and Meta-Analysis. Front. Microbiol. 2017, 8, 389. [Google Scholar] [CrossRef]
  5. Park, Y.-H.; Nam, H.-W. Clinical Features and Treatment of Ocular Toxoplasmosis. Korean J. Parasitol. 2013, 51, 393–399. [Google Scholar] [CrossRef]
  6. Skariah, S.; McIntyre, M.K.; Mordue, D.G. Toxoplasma gondii: Determinants of Tachyzoite to Bradyzoite Conversion. Parasitol. Res. 2010, 107, 253–260. [Google Scholar] [CrossRef] [PubMed]
  7. Dubey, J.P.; Lindsay, D.S.; Speer, C.A. Structures of Toxoplasma gondii Tachyzoites, Bradyzoites, and Sporozoites and Biology and Development of Tissue Cysts. Clin. Microbiol. Rev. 1998, 11, 267–299. [Google Scholar] [CrossRef]
  8. Weiss, L.M.; Ma, Y.F.; Takvorian, P.M.; Tanowitz, H.B.; Wittner, M. Bradyzoite Development in Toxoplasma gondii and the Hsp70 Stress Response. Infect. Immun. 1998, 66, 3295–3302. [Google Scholar] [CrossRef] [PubMed]
  9. Melo, M.B.; Jensen, K.D.C.; Saeij, J.P.J. Toxoplasma gondii Effectors Are Master Regulators of the Inflammatory Response. Trends Parasitol. 2011, 27, 487–495. [Google Scholar] [CrossRef] [PubMed]
  10. Cerutti, A.; Blanchard, N.; Besteiro, S. The Bradyzoite: A Key Developmental Stage for the Persistence and Pathogenesis of Toxoplasmosis. Pathogens 2020, 9, 234. [Google Scholar] [CrossRef]
  11. Mahmoudvand, H.; Ziaali, N.; Ghazvini, H.; Shojaee, S.; Keshavarz, H.; Esmaeilpour, K.; Sheibani, V. Toxoplasma gondii Infection Promotes Neuroinflammation Through Cytokine Networks and Induced Hyperalgesia in BALB/c Mice. Inflammation 2016, 39, 405–412. [Google Scholar] [CrossRef]
  12. Prandovszky, E.; Gaskell, E.; Martin, H.; Dubey, J.P.; Webster, J.P.; McConkey, G.A. The Neurotropic Parasite Toxoplasma gondii Increases Dopamine Metabolism. PLoS ONE 2011, 6, e23866. [Google Scholar] [CrossRef]
  13. Tedford, E.; McConkey, G. Neurophysiological Changes Induced by Chronic Toxoplasma gondii Infection. Pathogens 2017, 6, 19. [Google Scholar] [CrossRef] [PubMed]
  14. Castaño Barrios, L.; Da Silva Pinheiro, A.P.; Gibaldi, D.; Silva, A.A.; Machado Rodrigues e Silva, P.; Roffê, E.; da Costa Santiago, H.; Tostes Gazzinelli, R.; Mineo, J.R.; Silva, N.M.; et al. Behavioral Alterations in Long-Term Toxoplasma gondii Infection of C57BL/6 Mice Are Associated with Neuroinflammation and Disruption of the Blood Brain Barrier. PLoS ONE 2021, 16, e0258199. [Google Scholar] [CrossRef]
  15. Cleary, M.D.; Singh, U.; Blader, I.J.; Brewer, J.L.; Boothroyd, J.C. Toxoplasma gondii Asexual Development: Identification of Developmentally Regulated Genes and Distinct Patterns of Gene Expression. Eukaryot. Cell 2002, 1, 329–340. [Google Scholar] [CrossRef] [PubMed]
  16. Boothroyd, J.C.; Grigg, M.E. Population Biology of Toxoplasma gondii and Its Relevance to Human Infection: Do Different Strains Cause Different Disease? Curr. Opin. Microbiol. 2002, 5, 438–442. [Google Scholar] [CrossRef] [PubMed]
  17. Genova, B.M.D.; Wilson, S.K.; Dubey, J.P.; Knoll, L.J. Intestinal Delta-6-Desaturase Activity Determines Host Range for Toxoplasma Sexual Reproduction. PLoS Biol. 2019, 17, e3000364. [Google Scholar] [CrossRef]
  18. Farhat, D.C.; Swale, C.; Dard, C.; Cannella, D.; Ortet, P.; Barakat, M.; Sindikubwabo, F.; Belmudes, L.; De Bock, P.-J.; Couté, Y.; et al. A MORC-Driven Transcriptional Switch Controls Toxoplasma Developmental Trajectories and Sexual Commitment. Nat. Microbiol. 2020, 5, 570–583. [Google Scholar] [CrossRef]
  19. Antunes, A.V.; Shahinas, M.; Swale, C.; Farhat, D.C.; Ramakrishnan, C.; Bruley, C.; Cannella, D.; Robert, M.G.; Corrao, C.; Couté, Y.; et al. In Vitro Production of Cat-Restricted Toxoplasma Pre-Sexual Stages. Nature 2024, 625, 366–376. [Google Scholar] [CrossRef]
  20. Dubey, J.P.; Frenkel, J.K. Feline Toxoplasmosis from Acutely Infected Mice and the Development of Toxoplasma Cysts. J. Protozool. 1976, 23, 537–546. [Google Scholar] [CrossRef]
  21. Martorelli Di Genova, B.; Knoll, L.J. Comparisons of the Sexual Cycles for the Coccidian Parasites Eimeria and Toxoplasma. Front. Cell Infect. Microbiol. 2020, 10, 604897. [Google Scholar] [CrossRef]
  22. Sanchez, S.G.; Besteiro, S. The Pathogenicity and Virulence of Toxoplasma gondii. Virulence 2021, 12, 3095–3114. [Google Scholar] [CrossRef] [PubMed]
  23. Shapiro, K.; Bahia-Oliveira, L.; Dixon, B.; Dumètre, A.; de Wit, L.A.; VanWormer, E.; Villena, I. Environmental Transmission of Toxoplasma gondii: Oocysts in Water, Soil and Food. Food Waterborne Parasitol. 2019, 15, e00049. [Google Scholar] [CrossRef]
  24. Dubey, J.P.; Jones, J.L. Toxoplasma gondii Infection in Humans and Animals in the United States. Int. J. Parasitol. 2008, 38, 1257–1278. [Google Scholar] [CrossRef]
  25. Hill, D.; Dubey, J.P. Toxoplasma gondii: Transmission, Diagnosis and Prevention. Clin. Microbiol. Infect. 2002, 8, 634–640. [Google Scholar] [CrossRef]
  26. Robbins, J.R.; Zeldovich, V.B.; Poukchanski, A.; Boothroyd, J.C.; Bakardjiev, A.I. Tissue Barriers of the Human Placenta to Infection with Toxoplasma gondii. Infect. Immun. 2012, 80, 418–428. [Google Scholar] [CrossRef]
  27. Hampton, M.M. Congenital Toxoplasmosis: A Review. Neonatal Netw. 2015, 34, 274–278. [Google Scholar] [CrossRef]
  28. Gregg, B.; Taylor, B.C.; John, B.; Tait-Wojno, E.D.; Girgis, N.M.; Miller, N.; Wagage, S.; Roos, D.S.; Hunter, C.A. Replication and Distribution of Toxoplasma gondii in the Small Intestine after Oral Infection with Tissue Cysts. Infect. Immun. 2013, 81, 1635–1643. [Google Scholar] [CrossRef] [PubMed]
  29. Unno, A.; Suzuki, K.; Xuan, X.; Nishikawa, Y.; Kitoh, K.; Takashima, Y. Dissemination of Extracellular and Intracellular Toxoplasma gondii Tachyzoites in the Blood Flow. Parasitol. Int. 2008, 57, 515–518. [Google Scholar] [CrossRef] [PubMed]
  30. Denkers, E.Y. From Cells to Signaling Cascades: Manipulation of Innate Immunity by Toxoplasma gondii. FEMS Immunol. Med. Microbiol. 2003, 39, 193–203. [Google Scholar] [CrossRef] [PubMed]
  31. Pittman, K.J.; Aliota, M.T.; Knoll, L.J. Dual Transcriptional Profiling of Mice and Toxoplasma gondii during Acute and Chronic Infection. BMC Genom. 2014, 15, 806. [Google Scholar] [CrossRef] [PubMed]
  32. Derouin, F.; Pelloux, H. Prevention of Toxoplasmosis in Transplant Patients. Clin. Microbiol. Infect. 2008, 14, 1089–1101. [Google Scholar] [CrossRef] [PubMed]
  33. Sibley, D.L.; Charron, A.; Håkansson, S.; Mordue, D. Invasion and Intracellular Survival by Toxoplasma. In Madame Curie Bioscience Database [Internet]; Landes Bioscience: Austin, TX, USA, 2013. [Google Scholar]
  34. Hu, X.; Binns, D.; Reese, M.L. The Coccidian Parasites Toxoplasma and Neospora Dysregulate Mammalian Lipid Droplet Biogenesis. J. Biol. Chem. 2017, 292, 11009–11020. [Google Scholar] [CrossRef]
  35. Kemp, L.E.; Yamamoto, M.; Soldati-Favre, D. Subversion of Host Cellular Functions by the Apicomplexan Parasites. FEMS Microbiol. Rev. 2013, 37, 607–631. [Google Scholar] [CrossRef] [PubMed]
  36. Hakimi, M.-A.; Bougdour, A. Toxoplasma’s Ways of Manipulating the Host Transcriptome via Secreted Effectors. Curr. Opin. Microbiol. 2015, 26, 24–31. [Google Scholar] [CrossRef] [PubMed]
  37. Lima, T.S.; Lodoen, M.B. Mechanisms of Human Innate Immune Evasion by Toxoplasma gondii. Front. Cell Infect. Microbiol. 2019, 9, 103. [Google Scholar] [CrossRef] [PubMed]
  38. Mammari, N.; Halabi, M.A.; Yaacoub, S.; Chlala, H.; Dardé, M.-L.; Courtioux, B. Toxoplasma gondii Modulates the Host Cell Responses: An Overview of Apoptosis Pathways. Biomed. Res. Int. 2019, 2019, 6152489. [Google Scholar] [CrossRef] [PubMed]
  39. Ahmadpour, E.; Babaie, F.; Kazemi, T.; Mehrani Moghaddam, S.; Moghimi, A.; Hosseinzadeh, R.; Nissapatorn, V.; Pagheh, A.S. Overview of Apoptosis, Autophagy, and Inflammatory Processes in Toxoplasma gondii Infected Cells. Pathogens 2023, 12, 253. [Google Scholar] [CrossRef]
  40. Besteiro, S. Toxoplasma Control of Host Apoptosis: The Art of Not Biting Too Hard the Hand That Feeds You. Microb. Cell 2015, 2, 178–181. [Google Scholar] [CrossRef]
  41. Dunay, I.R.; Diefenbach, A. Group 1 Innate Lymphoid Cells in Toxoplasma gondii Infection. Parasite Immunol. 2018, 40, e12516. [Google Scholar] [CrossRef]
  42. Park, E.; Patel, S.; Wang, Q.; Andhey, P.; Zaitsev, K.; Porter, S.; Hershey, M.; Bern, M.; Plougastel-Douglas, B.; Collins, P.; et al. Toxoplasma gondii Infection Drives Conversion of NK Cells into ILC1-like Cells. eLife 2019, 8, e47605. [Google Scholar] [CrossRef] [PubMed]
  43. Sasai, M.; Yamamoto, M. Innate, Adaptive, and Cell-Autonomous Immunity against Toxoplasma gondii Infection. Exp. Mol. Med. 2019, 51, 1–10. [Google Scholar] [CrossRef] [PubMed]
  44. Kawai, T.; Akira, S. The Role of Pattern-Recognition Receptors in Innate Immunity: Update on Toll-like Receptors. Nat. Immunol. 2010, 11, 373–384. [Google Scholar] [CrossRef] [PubMed]
  45. Sher, A.; Tosh, K.; Jankovic, D. Innate Recognition of Toxoplasma gondii in Humans Involves a Mechanism Distinct from That Utilized by Rodents. Cell Mol. Immunol. 2017, 14, 36–42. [Google Scholar] [CrossRef]
  46. Kawai, T.; Akira, S. TLR Signaling. Cell Death Differ. 2006, 13, 816–825. [Google Scholar] [CrossRef]
  47. Yarovinsky, F.; Zhang, D.; Andersen, J.F.; Bannenberg, G.L.; Serhan, C.N.; Hayden, M.S.; Hieny, S.; Sutterwala, F.S.; Flavell, R.A.; Ghosh, S.; et al. TLR11 Activation of Dendritic Cells by a Protozoan Profilin-like Protein. Science 2005, 308, 1626–1629. [Google Scholar] [CrossRef]
  48. Koblansky, A.A.; Jankovic, D.; Oh, H.; Hieny, S.; Sungnak, W.; Mathur, R.; Hayden, M.S.; Akira, S.; Sher, A.; Ghosh, S. Recognition of Profilin by Toll-like Receptor 12 Is Critical for Host Resistance to Toxoplasma gondii. Immunity 2013, 38, 119–130. [Google Scholar] [CrossRef]
  49. Raetz, M.; Kibardin, A.; Sturge, C.R.; Pifer, R.; Li, H.; Burstein, E.; Ozato, K.; Larin, S.; Yarovinsky, F. Cooperation of TLR12 and TLR11 in the IRF8-Dependent IL-12 Response to Toxoplasma gondii Profilin. J. Immunol. 2013, 191, 4818–4827. [Google Scholar] [CrossRef]
  50. Mashayekhi, M.; Sandau, M.M.; Dunay, I.R.; Frickel, E.M.; Khan, A.; Goldszmid, R.S.; Sher, A.; Ploegh, H.L.; Murphy, T.L.; Sibley, L.D.; et al. CD8α+ Dendritic Cells Are the Critical Source of Interleukin-12 That Controls Acute Infection by Toxoplasma gondii Tachyzoites. Immunity 2011, 35, 249–259. [Google Scholar] [CrossRef]
  51. Sanecka, A.; and Frickel, E.-M. Use and Abuse of Dendritic Cells by Toxoplasma gondii. Virulence 2012, 3, 678–689. [Google Scholar] [CrossRef]
  52. Scanga, C.A.; Aliberti, J.; Jankovic, D.; Tilloy, F.; Bennouna, S.; Denkers, E.Y.; Medzhitov, R.; Sher, A. Cutting Edge: MyD88 Is Required for Resistance to Toxoplasma gondii Infection and Regulates Parasite-Induced IL-12 Production by Dendritic Cells. J. Immunol. 2002, 168, 5997–6001. [Google Scholar] [CrossRef]
  53. LaRosa, D.F.; Stumhofer, J.S.; Gelman, A.E.; Rahman, A.H.; Taylor, D.K.; Hunter, C.A.; Turka, L.A. T Cell Expression of MyD88 Is Required for Resistance to Toxoplasma gondii. Proc. Natl. Acad. Sci. USA 2008, 105, 3855–3860. [Google Scholar] [CrossRef]
  54. Mahmoudzadeh, S.; Nozad Charoudeh, H.; Marques, C.S.; Bahadory, S.; Ahmadpour, E. The Role of IL-12 in Stimulating NK Cells against Toxoplasma gondii Infection: A Mini-Review. Parasitol. Res. 2021, 120, 2303–2309. [Google Scholar] [CrossRef] [PubMed]
  55. Cai, G.; Kastelein, R.; Hunter, C.A. Interleukin-18 (IL-18) Enhances Innate IL-12-Mediated Resistance to Toxoplasma gondii. Infect. Immun. 2000, 68, 6932–6938. [Google Scholar] [CrossRef]
  56. Andrade, W.A.; Souza, M.d.C.; Ramos-Martinez, E.; Nagpal, K.; Dutra, M.S.; Melo, M.B.; Bartholomeu, D.C.; Ghosh, S.; Golenbock, D.T.; Gazzinelli, R.T. Combined Action of Nucleic Acid-Sensing Toll-like Receptors and TLR11/TLR12 Heterodimers Imparts Resistance to Toxoplasma gondii in Mice. Cell Host Microbe 2013, 13, 42–53. [Google Scholar] [CrossRef]
  57. Kim, L.; Butcher, B.A.; Lee, C.W.; Uematsu, S.; Akira, S.; Denkers, E.Y. Toxoplasma gondii Genotype Determines MyD88-Dependent Signaling in Infected Macrophages. J. Immunol. 2006, 177, 2584–2591. [Google Scholar] [CrossRef]
  58. Scharton-Kersten, T.M.; Wynn, T.A.; Denkers, E.Y.; Bala, S.; Grunvald, E.; Hieny, S.; Gazzinelli, R.T.; Sher, A. In the Absence of Endogenous IFN-Gamma, Mice Develop Unimpaired IL-12 Responses to Toxoplasma gondii While Failing to Control Acute Infection. J. Immunol. 1996, 157, 4045–4054. [Google Scholar] [CrossRef] [PubMed]
  59. Wallet, P.; Benaoudia, S.; Mosnier, A.; Lagrange, B.; Martin, A.; Lindgren, H.; Golovliov, I.; Michal, F.; Basso, P.; Djebali, S.; et al. IFN-γ Extends the Immune Functions of Guanylate Binding Proteins to Inflammasome-Independent Antibacterial Activities during Francisella Novicida Infection. PLoS Pathog. 2017, 13, e1006630. [Google Scholar] [CrossRef]
  60. Tretina, K.; Park, E.-S.; Maminska, A.; MacMicking, J.D. Interferon-Induced Guanylate-Binding Proteins: Guardians of Host Defense in Health and Disease. J. Exp. Med. 2019, 216, 482–500. [Google Scholar] [CrossRef] [PubMed]
  61. Zhao, Y.O.; Khaminets, A.; Hunn, J.P.; Howard, J.C. Disruption of the Toxoplasma gondii Parasitophorous Vacuole by IFNgamma-Inducible Immunity-Related GTPases (IRG Proteins) Triggers Necrotic Cell Death. PLoS Pathog. 2009, 5, e1000288. [Google Scholar] [CrossRef]
  62. Praefcke, G.J.K. Regulation of Innate Immune Functions by Guanylate-Binding Proteins. Int. J. Med. Microbiol. 2018, 308, 237–245. [Google Scholar] [CrossRef] [PubMed]
  63. Khan, I.A.; Matsuura, T.; Fonseka, S.; Kasper, L.H. Production of Nitric Oxide (NO) Is Not Essential for Protection against Acute Toxoplasma gondii Infection in IRF-1-/- Mice. J. Immunol. 1996, 156, 636–643. [Google Scholar] [CrossRef]
  64. Dincel, G.C.; Atmaca, H.T. Nitric Oxide Production Increases during Toxoplasma gondii Encephalitis in Mice. Exp. Parasitol. 2015, 156, 104–112. [Google Scholar] [CrossRef]
  65. Denkers, E.Y.; Gazzinelli, R.T. Regulation and Function of T-Cell-Mediated Immunity during Toxoplasma gondii Infection. Clin. Microbiol. Rev. 1998, 11, 569–588. [Google Scholar] [CrossRef] [PubMed]
  66. Suzuki, Y.; Sa, Q.; Gehman, M.; Ochiai, E. Interferon-Gamma- and Perforin-Mediated Immune Responses for Resistance in the Brain against Toxoplasma gondii. Expert. Rev. Mol. Med. 2011, 13, e31. [Google Scholar] [CrossRef] [PubMed]
  67. Ferreira, P.T.M.; Oliveira-Scussel, A.C.M.; Sousa, R.A.P.; Gomes, B.Q.; Félix, J.E.; Silva, R.J.; Millian, I.B.; Assunção, T.S.F.; Teixeira, S.C.; Gomes, M.d.L.M.; et al. Macrophage Migration Inhibitory Factor Contributes to Drive Phenotypic and Functional Macrophages Activation in Response to Toxoplasma gondii Infection. Immunobiology 2023, 228, 152357. [Google Scholar] [CrossRef]
  68. Suzuki, Y.; Orellana, M.A.; Schreiber, R.D.; Remington, J.S. Interferon-γ: The Major Mediator of Resistance Against Toxoplasma gondii. Science 1988, 240, 516–518. [Google Scholar] [CrossRef] [PubMed]
  69. Scharton-Kersten, T.M.; Yap, G.; Magram, J.; Sher, A. Inducible Nitric Oxide Is Essential for Host Control of Persistent but Not Acute Infection with the Intracellular Pathogen Toxoplasma gondii. J. Exp. Med. 1997, 185, 1261–1274. [Google Scholar] [CrossRef]
  70. Halonen, S.K.; Weiss, L.M. Investigation into the Mechanism of Gamma Interferon-Mediated Inhibition of Toxoplasma gondii in Murine Astrocytes. Infect. Immun. 2000, 68, 3426–3430. [Google Scholar] [CrossRef]
  71. Khan, I.A.; Green, W.R.; Kasper, L.H.; Green, K.A.; Schwartzman, J.D. Immune CD8+ T Cells Prevent Reactivation of Toxoplasma gondii Infection in the Immunocompromised Host. Infect. Immun. 1999, 67, 5869–5876. [Google Scholar] [CrossRef] [PubMed]
  72. Krishnamurthy, S.; Konstantinou, E.K.; Young, L.H.; Gold, D.A.; Saeij, J.P.J. The Human Immune Response to Toxoplasma: Autophagy versus Cell Death. PLoS Pathog. 2017, 13, e1006176. [Google Scholar] [CrossRef]
  73. Suzuki, Y.; Wang, X.; Jortner, B.S.; Payne, L.; Ni, Y.; Michie, S.A.; Xu, B.; Kudo, T.; Perkins, S. Removal of Toxoplasma gondii Cysts from the Brain by Perforin-Mediated Activity of CD8+ T Cells. Am. J. Pathol. 2010, 176, 1607–1613. [Google Scholar] [CrossRef] [PubMed]
  74. Cohen, S.B.; Denkers, E.Y. Border Maneuvers: Deployment of Mucosal Immune Defenses against Toxoplasma gondii. Mucosal Immunol. 2014, 7, 744–752. [Google Scholar] [CrossRef] [PubMed]
  75. Israelski, D.M.; Remington, J.S. Toxoplasmic Encephalitis in Patients with AIDS. Infect. Dis. Clin. N. Am. 1988, 2, 429–446. [Google Scholar] [CrossRef]
  76. Belkaid, Y.; Sun, C.M.; Bouladoux, N. Parasites and Immunoregulatory T Cells. Curr. Opin. Immunol. 2006, 18, 406–412. [Google Scholar] [CrossRef]
  77. Mahamed, D.A.; Toussaint, L.E.; Bynoe, M.S. CD73-Generated Adenosine Is Critical for Immune Regulation during Toxoplasma gondii Infection. Infect. Immun. 2015, 83, 721–729. [Google Scholar] [CrossRef]
  78. Heimesaat, M.M.; Escher, U.; Grunau, A.; Fiebiger, U.; Bereswill, S. Peroral Low-Dose Toxoplasma gondii Infection of Human Microbiota-Associated Mice—A Subacute Ileitis Model to Unravel Pathogen–Host Interactions. Eur. J. Microbiol. Immunol. 2018, 8, 53–61. [Google Scholar] [CrossRef]
  79. Toxoplasmosis—American Association for Pediatric Ophthalmology and Strabismus. Available online: https://www.aapos.org/glossary/toxoplasmosis (accessed on 21 May 2025).
  80. Smigiel, K.S.; Srivastava, S.; Stolley, J.M.; Campbell, D.J. Regulatory T Cell Homeostasis: Steady-State Maintenance and Modulation during Inflammation. Immunol. Rev. 2014, 259, 40–59. [Google Scholar] [CrossRef] [PubMed]
  81. Lei, H.; Schmidt-Bleek, K.; Dienelt, A.; Reinke, P.; Volk, H.-D. Regulatory T Cell-Mediated Anti-Inflammatory Effects Promote Successful Tissue Repair in Both Indirect and Direct Manners. Front. Pharmacol. 2015, 6, 184. [Google Scholar] [CrossRef]
  82. Wang, J.; Zhao, X.; Wan, Y.Y. Intricacies of TGF-β Signaling in Treg and Th17 Cell Biology. Cell Mol. Immunol. 2023, 20, 1002–1022. [Google Scholar] [CrossRef] [PubMed]
  83. Gazzinelli, R.T.; Wysocka, M.; Hieny, S.; Scharton-Kersten, T.; Cheever, A.; Kühn, R.; Müller, W.; Trinchieri, G.; Sher, A. In the Absence of Endogenous IL-10, Mice Acutely Infected with Toxoplasma gondii Succumb to a Lethal Immune Response Dependent on CD4+ T Cells and Accompanied by Overproduction of IL-12, IFN-Gamma and TNF-Alpha. J. Immunol. 1996, 157, 798–805. [Google Scholar] [CrossRef] [PubMed]
  84. Lu, F.; Huang, S.; Kasper, L.H. Interleukin-10 and Pathogenesis of Murine Ocular Toxoplasmosis. Infect. Immun. 2003, 71, 7159–7163. [Google Scholar] [CrossRef]
  85. Sanjabi, S.; Zenewicz, L.A.; Kamanaka, M.; Flavell, R.A. Anti- and Pro-Inflammatory Roles of TGF-β, IL-10, and IL-22 In Immunity and Autoimmunity. Curr. Opin. Pharmacol. 2009, 9, 447–453. [Google Scholar] [CrossRef]
  86. Deng, Z.; Fan, T.; Xiao, C.; Tian, H.; Zheng, Y.; Li, C.; He, J. TGF-β Signaling in Health, Disease and Therapeutics. Signal Transduct. Target. Ther. 2024, 9, 1–40. [Google Scholar] [CrossRef]
  87. Ricci-Azevedo, R.; Mendonça-Natividade, F.C.; Santana, A.C.; Alcoforado Diniz, J.; Roque-Barreira, M.C. Microneme Proteins 1 and 4 From Toxoplasma gondii Induce IL-10 Production by Macrophages Through TLR4 Endocytosis. Front. Immunol. 2021, 12, 655371. [Google Scholar] [CrossRef]
  88. Saeij, J.P.J.; Boyle, J.P.; Boothroyd, J.C. Differences among the Three Major Strains of Toxoplasma gondii and Their Specific Interactions with the Infected Host. Trends Parasitol. 2005, 21, 476–481. [Google Scholar] [CrossRef] [PubMed]
  89. Howe, D.K.; Sibley, L.D. Toxoplasma gondii Comprises Three Clonal Lineages: Correlation of Parasite Genotype with Human Disease. J. Infect. Dis. 1995, 172, 1561–1566. [Google Scholar] [CrossRef] [PubMed]
  90. Jensen, K.D.C.; Camejo, A.; Melo, M.B.; Cordeiro, C.; Julien, L.; Grotenbreg, G.M.; Frickel, E.-M.; Ploegh, H.L.; Young, L.; Saeij, J.P.J. Toxoplasma gondii Superinfection and Virulence during Secondary Infection Correlate with the Exact ROP5/ROP18 Allelic Combination. mBio 2015, 6, e02280-14. [Google Scholar] [CrossRef]
  91. Niedelman, W.; Gold, D.A.; Rosowski, E.E.; Sprokholt, J.K.; Lim, D.; Arenas, A.F.; Melo, M.B.; Spooner, E.; Yaffe, M.B.; Saeij, J.P.J. The Rhoptry Proteins ROP18 and ROP5 Mediate Toxoplasma gondii Evasion of the Murine, But Not the Human, Interferon-Gamma Response. PLOS Pathog. 2012, 8, e1002784. [Google Scholar] [CrossRef]
  92. Etheridge, R.D.; Alaganan, A.; Tang, K.; Lou, H.J.; Turk, B.E.; Sibley, L.D. The Toxoplasma Pseudokinase ROP5 Forms Complexes with ROP18 and ROP17 Kinases That Synergize to Control Acute Virulence in Mice. Cell Host Microbe 2014, 15, 537–550. [Google Scholar] [CrossRef]
  93. Galal, L.; Hamidović, A.; Dardé, M.L.; Mercier, M. Diversity of Toxoplasma gondii Strains at the Global Level and Its Determinants. Food Waterborne Parasitol. 2019, 15, e00052. [Google Scholar] [CrossRef]
  94. Ye, K.; Chen, Z.; Xu, Y. The Double-Edged Functions of Necroptosis. Cell Death Dis. 2023, 14, 163. [Google Scholar] [CrossRef]
  95. Labbé, K.; Saleh, M. Cell Death in the Host Response to Infection. Cell Death Differ. 2008, 15, 1339–1349. [Google Scholar] [CrossRef]
  96. Vutova, P.; Wirth, M.; Hippe, D.; Gross, U.; Schulze-Osthoff, K.; Schmitz, I.; Lüder, C.G.K. Toxoplasma gondii Inhibits Fas/CD95-Triggered Cell Death by Inducing Aberrant Processing and Degradation of Caspase 8. Cell. Microbiol. 2007, 9, 1556–1570. [Google Scholar] [CrossRef]
  97. Hippe, D.; Lytovchenko, O.; Schmitz, I.; Lüder, C.G.K. Fas/CD95-Mediated Apoptosis of Type II Cells Is Blocked by Toxoplasma gondii Primarily via Interference with the Mitochondrial Amplification Loop. Infect. Immun. 2008, 76, 2905–2912. [Google Scholar] [CrossRef]
  98. Nash, P.B.; Purner, M.B.; Leon, R.P.; Clarke, P.; Duke, R.C.; Curiel, T.J. Toxoplasma gondii-Infected Cells Are Resistant to Multiple Inducers of Apoptosis1. J. Immunol. 1998, 160, 1824–1830. [Google Scholar] [CrossRef] [PubMed]
  99. Yamada, T.; Tomita, T.; Weiss, L.M.; Orlofsky, A. Toxoplasma gondii Inhibits Granzyme B-Mediated Apoptosis by the Inhibition of Granzyme B Function in Host Cells. Int. J. Parasitol. 2011, 41, 595–607. [Google Scholar] [CrossRef] [PubMed]
  100. Shapira, S.; Speirs, K.; Gerstein, A.; Caamano, J.; Hunter, C.A. Suppression of NF-κB Activation by Infection with Toxoplasma gondii. J. Infect. Dis. 2002, 185, S66–S72. [Google Scholar] [CrossRef] [PubMed]
  101. Ewald, S.E.; Chavarria-Smith, J.; Boothroyd, J.C. NLRP1 Is an Inflammasome Sensor for Toxoplasma gondii. Infect. Immun. 2014, 82, 460–468. [Google Scholar] [CrossRef]
  102. McKee, A.S.; Dzierszinski, F.; Boes, M.; Roos, D.S.; Pearce, E.J. Functional Inactivation of Immature Dendritic Cells by the Intracellular Parasite Toxoplasma gondii. J. Immunol. 2004, 173, 2632–2640. [Google Scholar] [CrossRef]
  103. Lüder, C.G.K.; Lang, T.; Beuerle, B.; Gross, U. Down-Regulation of MHC Class II Molecules and Inability to up-Regulate Class I Molecules in Murine Macrophages after Infection with Toxoplasma gondii. Clin. Exp. Immunol. 1998, 112, 308–316. [Google Scholar] [CrossRef]
  104. Lüder, C.G.K.; Walter, W.; Beuerle, B.; Maeurer, M.J.; Gross, U. Toxoplasma gondii Down-Regulates MHC Class II Gene Expression and Antigen Presentation by Murine Macrophages via Interference with Nuclear Translocation of STAT1α. Eur. J. Immunol. 2001, 31, 1475–1484. [Google Scholar] [CrossRef]
  105. Rosowski, E.E.; Lu, D.; Julien, L.; Rodda, L.; Gaiser, R.A.; Jensen, K.D.C.; Saeij, J.P.J. Strain-Specific Activation of the NF-κB Pathway by GRA15, a Novel Toxoplasma gondii Dense Granule Protein. J. Exp. Med. 2011, 208, 195–212. [Google Scholar] [CrossRef]
  106. Du, J.; An, R.; Chen, L.; Shen, Y.; Chen, Y.; Cheng, L.; Jiang, Z.; Zhang, A.; Yu, L.; Chu, D.; et al. Toxoplasma gondii Virulence Factor ROP18 Inhibits the Host NF-κB Pathway by Promoting P65 Degradation*. J. Biol. Chem. 2014, 289, 12578–12592. [Google Scholar] [CrossRef] [PubMed]
  107. Gay, G.; Braun, L.; Brenier-Pinchart, M.-P.; Vollaire, J.; Josserand, V.; Bertini, R.-L.; Varesano, A.; Touquet, B.; De Bock, P.-J.; Coute, Y.; et al. Toxoplasma gondii TgIST Co-Opts Host Chromatin Repressors Dampening STAT1-Dependent Gene Regulation and IFN-γ–Mediated Host Defenses. J. Exp. Med. 2016, 213, 1779–1798. [Google Scholar] [CrossRef]
  108. Zimmermann, S.; Murray, P.J.; Heeg, K.; Dalpke, A.H. Induction of Suppressor of Cytokine Signaling-1 by Toxoplasma gondii Contributes to Immune Evasion in Macrophages by Blocking IFN-γ Signaling1. J. Immunol. 2006, 176, 1840–1847. [Google Scholar] [CrossRef]
  109. Alaganan, A.; Fentress, S.J.; Tang, K.; Wang, Q.; Sibley, L.D. Toxoplasma GRA7 Effector Increases Turnover of Immunity-Related GTPases and Contributes to Acute Virulence in the Mouse. Proc. Natl. Acad. Sci. USA 2014, 111, 1126–1131. [Google Scholar] [CrossRef] [PubMed]
  110. Dunay, I.R.; Gajurel, K.; Dhakal, R.; Liesenfeld, O.; Montoya, J.G. Treatment of Toxoplasmosis: Historical Perspective, Animal Models, and Current Clinical Practice. Clin. Microbiol. Rev. 2018, 31, e00057-17. [Google Scholar] [CrossRef] [PubMed]
  111. Montazeri, M.; Mehrzadi, S.; Sharif, M.; Sarvi, S.; Tanzifi, A.; Aghayan, S.A.; Daryani, A. Drug Resistance in Toxoplasma gondii. Front. Microbiol. 2018, 9, 2587. [Google Scholar] [CrossRef]
  112. Lan, H.-W.; Lu, Y.-N.; Zhao, X.-D.; Jin, G.-N.; Lu, J.-M.; Jin, C.-H.; Ma, J.; Jin, X.; Xu, X.; Piao, L.-X. New Role of Sertraline against Toxoplasma gondii-Induced Depression-like Behaviours in Mice. Parasite Immunol. 2021, 43, e12893. [Google Scholar] [CrossRef]
  113. Brito, C.; Lourenço, C.; Magalhães, J.; Reis, S.; Borges, M. Nanoparticles as a Delivery System of Antigens for the Development of an Effective Vaccine against Toxoplasma gondii. Vaccines 2023, 11, 733. [Google Scholar] [CrossRef]
  114. Abdelhamid Elgendy, W.M.; Haggag, Y.A.; El-Nouby, K.A.; El-Kowrany, S.I.; El Marhoumy, S.M. Evaluation of the Effect of Guanabenz-Loaded Nanoparticles on Chronic Toxoplasmosis in Mice. Exp. Parasitol. 2023, 246, 108460. [Google Scholar] [CrossRef] [PubMed]
  115. Costa, I.N.; Ribeiro, M.; Silva Franco, P.; da Silva, R.J.; de Araújo, T.E.; Milián, I.C.B.; Luz, L.C.; Guirelli, P.M.; Nakazato, G.; Mineo, J.R.; et al. Biogenic Silver Nanoparticles Can Control Toxoplasma gondii Infection in Both Human Trophoblast Cells and Villous Explants. Front. Microbiol. 2021, 11, 623947. [Google Scholar] [CrossRef]
  116. Albalawi, A.E.; Alanazi, A.D.; Alyousif, M.S.; Sepahvand, A.; Ebrahimi, K.; Niazi, M.; Mahmoudvand, H. The High Potency of Green Synthesized Copper Nanoparticles to Prevent the Toxoplasma gondii Infection in Mice. Acta Parasitol. 2021, 66, 1472–1479. [Google Scholar] [CrossRef]
  117. Xiao, J.; Li, Y.; Yolken, R.H.; Viscidi, R.P. PD-1 Immune Checkpoint Blockade Promotes Brain Leukocyte Infiltration and Diminishes Cyst Burden in a Mouse Model of Toxoplasma Infection. J. Neuroimmunol. 2018, 319, 55–62. [Google Scholar] [CrossRef]
  118. Buxton, D. Toxoplasmosis: The First Commercial Vaccine. Parasitol. Today 1993, 9, 335–337. [Google Scholar] [CrossRef] [PubMed]
  119. Ramakrishnan, C.; Maier, S.; Walker, R.A.; Rehrauer, H.; Joekel, D.E.; Winiger, R.R.; Basso, W.U.; Grigg, M.E.; Hehl, A.B.; Deplazes, P.; et al. An Experimental Genetically Attenuated Live Vaccine to Prevent Transmission of Toxoplasma gondii by Cats. Sci. Rep. 2019, 9, 1474. [Google Scholar] [CrossRef]
  120. Dubey, J.P. Schizogony and Gametogony of Oocyst-Deficient T-263 Strain of Toxoplasma gondii. Vet. Parasitol. 2017, 245, 160–162. [Google Scholar] [CrossRef]
  121. Frenkel, J.K.; Pfefferkorn, E.R.; Smith, D.D.; Fishback, J.L. Prospective Vaccine Prepared from a New Mutant of Toxoplasma gondii for Use in Cats. Am. J. Vet. Res. 1991, 52, 759–763. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Erazo Flores, B.J.; Knoll, L.J. Toxoplasma gondii at the Host Interface: Immune Modulation and Translational Strategies for Infection Control. Vaccines 2025, 13, 819. https://doi.org/10.3390/vaccines13080819

AMA Style

Erazo Flores BJ, Knoll LJ. Toxoplasma gondii at the Host Interface: Immune Modulation and Translational Strategies for Infection Control. Vaccines. 2025; 13(8):819. https://doi.org/10.3390/vaccines13080819

Chicago/Turabian Style

Erazo Flores, Billy J., and Laura J. Knoll. 2025. "Toxoplasma gondii at the Host Interface: Immune Modulation and Translational Strategies for Infection Control" Vaccines 13, no. 8: 819. https://doi.org/10.3390/vaccines13080819

APA Style

Erazo Flores, B. J., & Knoll, L. J. (2025). Toxoplasma gondii at the Host Interface: Immune Modulation and Translational Strategies for Infection Control. Vaccines, 13(8), 819. https://doi.org/10.3390/vaccines13080819

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop