788
Views
0
CrossRef citations to date
0
Altmetric
Review

Immunopathogenesis of ocular toxoplasmosis and implications for treatment

ORCID Icon, ORCID Icon & ORCID Icon
Pages 7-26 | Received 31 Jul 2023, Accepted 31 Oct 2023, Published online: 29 Nov 2023

ABSTRACT

Introduction

Ocular toxoplasmosis appears after primary infection or during the reactivation of chronic infection by the protozoa Toxoplasma gondii. The risk of ocular involvement and the heterogeneity of clinical manifestations, their complications, and the probability of recurrences are linked to polymorphisms in immune response-related genes, cytokine networks, lymphocyte subpopulation, and parasite virulence factors. Appropriate clinical management and evidence-based advisory recommendations for patients require a clear understanding of the immunopathological mechanisms of this parasitic disease.

Areas covered

Narrative review of the scientific literature in human ocular toxoplasmosis related to parasitological and immunological characteristics, genetic polymorphisms linked to ocular involvement, and the clinical correlations of the cytokinome in aqueous humor and experiments with peripheral blood mononuclear cells.

Expert Opinion/Commentary

The greater severity in people infected by South American strains is partly explained by parasite protein kinases interfering with the effector immune functions of interferon-gamma, resulting in lower antiparasitic activity and more significant inflammation. Future therapies should point to the increase in IFN-γ production (for example, by stimulating CD4+ memory T cells subset). Thus, immune-based interventions could be promising in inducing an appropriate response for treating and preventing ocular damage and recurrences. Drugs targeting tissue cysts responsible for reactivations are a current priority.

Plain Language Summary

Ocular toxoplasmosis is a persistent eye condition affecting patients’ visual health and quality of life. This disease may manifest after a primary infection or during the reactivation of a latent infection by the protozoan Toxoplasma gondii. In our opinion, based on the principle of precaution, all recent primary infections, symptomatic or asymptomatic, caused by Toxoplasma, should be treated, as routine screening of the susceptibility genetic factors remains unavailable. Clinicians often encounter inquiries about the source of the infection, the factors contributing to ocular involvement (which, in the majority of the population, approximately 90%, remains asymptomatic), the likelihood of recurrent episodes, and the potential expansion of ocular damage. Current scientific knowledge indicates that genetic determinants governing specific immune responses, particularly the ability to produce protective cytokines while restraining inflammatory responses, may contribute to understanding the development and characteristics of ocular toxoplasmosis in humans. The role of genetic polymorphisms has been substantiated by the analysis of cytokine profiles in aqueous humor and experimental investigations using human peripheral blood mononuclear cells (PBMCs). In South America, where virulent strains of Toxoplasma prevail, the disease can manifest itself more severely. Numerous parasite protein kinases function as virulence factors, impeding the effector immune functions of interferon-gamma, decreasing antiparasitic activity, and exacerbating inflammation. The interaction between infection by virulent strains and genetic host susceptibility factors intervene in the magnitude of retinochoroidal damage.

In light of these insights, developing new therapies becomes imperative for managing and preventing recurrent ocular toxoplasmosis. Additionally, pursuing drugs capable of eradicating tissue cysts responsible for recurrences and reactivations is a current research priority.

1. Introduction: epidemiology and risk factors for ocular toxoplasmosis

Ocular toxoplasmosis results from retinal infection by the protozoan parasite Toxoplasma gondii [Citation1]]. This successful ubiquitous apicomplexan protozoan can infect various vertebrate hosts, considered the most frequent zoonotic disease worldwide [Citation2–4]. The infection is cosmopolitan but is more prevalent in tropical and subtropical regions, where environmental conditions favor the parasite’s survival, especially the humidity and a high rate of precipitations [Citation5–8]. An umbrella review concluded that in the human general population, the global seroprevalence reach 42%, varying largely according to geographical regions [Citation2]. It is generally estimated that one-third of the human population has had the infection [Citation9]. Although this high prevalence of the infection in the human population, the development of symptoms such as ocular clinical manifestations are relatively low, estimated to range from 1% to 18% of all Toxoplasma infections [Citation1,Citation10]. In recent years, some reports indicate a decline in the prevalence in the general population in Europe and North America [Citation11] but not in South America [Citation12]. Parallelly, the incidence of ocular toxoplasmosis was reported to decline in some places in North America [Citation13] but increased in South America over the years [Citation14]. It should be noted that climate change would impact the trends in the risk of infection for many regions of the world, and an increase in toxoplasmosis frequency can be expected for the years to come [Citation15–17].

Toxoplasmosis in humans is a food and waterborne infection [Citation18–21]. The attributable fraction from each factor changes from region to region; thus, infections due to uncooked meat are more common in countries with elevated incomes and low-income countries due to water conditions [Citation22]. Waterborne origin has been linked with outbreaks of ocular toxoplasmosis [Citation23–26]. Congenital transmission from mother to fetus contributes to a lower fraction of ocular toxoplasmosis, with postnatal infection the most critical origin of ocular disease due to toxoplasmosis [Citation7,Citation27].

2. Parasitological, clinical, and pathological characteristics of human ocular toxoplasmosis

Toxoplasma can infect almost any cell type, unlike other protozoa that cause human diseases such as Plasmodium or Leishmania, invading specific cell lineages [Citation28]. However, humans’ most common clinical damage is observed in the eye and brain [Citation29]. During the acute phase of the disease, Toxoplasma multiplies actively within cells in the tachyzoite stage with rapid replication [Citation30]. After the host develops an immune response parasite turns to a stage with slower proliferation, the bradyzoite, residing within a cyst tissue [Citation30,Citation31]. This infection characteristic is crucial to understanding clinical consequences, such as reactivation and subsequent recurrence of ocular lesions [Citation32].

The most prominent clinical characteristic of toxoplasmosis as a human disease for immunocompetent individuals is neurotropism, with the eye and brain as primary targets for damage [Citation33,Citation34]. Our classical view that chronic infection is asymptomatic and without significant consequences for most infected people has recently been challenged [Citation35–37]. Voluminous and cumulative evidence indicates that chronic infection can lead to mental disease [Citation37,Citation38]. Although the clinical and pathological characteristics of immunodeficient individuals with toxoplasmosis differ, the brain and eye remain the main sites of disease manifestations [Citation39].

One clinical key feature of ocular toxoplasmosis is that it is unilateral for most (~70%) of the immunocompetent hosts in primary and during recurrent episodes in the posterior eye segment [Citation40]. The infection can cause visual impairment and ocular complications, including retinal scars, retinal detachment, cataracts, glaucoma, and vision loss [Citation39,Citation41–43]. The severity, recurrences, and location of the lesions within the eye can vary, leading to a wide range of clinical manifestations [Citation39,Citation42,Citation43].

One crucial question is how many infected people have the parasite in the eye and how many result in retinochoroiditis. Eye fundoscopy screening in the general population can indicate how many seropositive people have retinochoroidal scars in the population [Citation44,Citation45]. This kind of screening has the advantage of identifying people unaware of the presence of retinal scars because they are peripheral or because of a lack of access to visual health care [Citation7]. The reports of eye fund screening for chorioretinal scars [Citation46] showed a prevalence ranging between 0.6% in Maryland, United States [Citation47], and 17.7% in Brazil [Citation44]. However, fund eye screening has the problem that up to 30% of retinal lesions seen as typical of toxoplasmosis can originate from other microorganisms [Citation48]. Another source of information comes from the reports of ocular involvement during outbreaks [Citation27,Citation49] as occurred in Canada [Citation47], Brazil [Citation49] and India [Citation23]. The rates of ocular involvement in these outbreaks ranged between 2.7% to 19% [Citation1]. The most critical data to date comes from Brazil through a longitudinal follow-up, where the risk of new chorioretinal lesions after diagnosis was 6.4 per 100 persons/year [Citation27].

Another work was made in postmortem specimens from bank eyes from two cities in Brazil [Citation46]. The eyes were collected consecutively from deceased persons, and the overall prevalence of retinal scars in 270 eyes was 10% [Citation46]. After that, PCR analysis for Toxoplasma was performed in 57 eyes, and different prevalences were found according to the city: in Joinville, in southern Brazil, it was 87% (13/15), and in São Paulo, 7% (3/42). The eyes from Joinville tested positive for parasite DNA had a 1:1 correlation with seroprevalence, which may indicate that no apparent barrier to parasite entry into the eye existed in this geographic region [Citation46]. Even more interesting was that the 16 eyes that were positive by PCR did not have any chorioretinal scar, indicating that bearing parasite infections in their eyes do not force the development of ocular lesion, like the finding that only 30%–50% of AIDS Toxoplasma seropositive patients experience recrudescent ocular or brain disease [Citation50].

The data from the studies based on eye screening, outbreaks, and eye banks can give us a picture of what can occur after a primary Toxoplasma infection and, at the same time, can answer some crucial questions. The parasite, after infection, can arrive in the eye and remain as tissue cysts without triggering retinal damage for most people. This can be explained by an efficacious local immune response mediated by innate () and adaptative () immune response [Citation32,Citation51]. Although some people presumably can eliminate the parasite and become seronegative for antibodies and even revert to a negative T cell response [Citation52] this is not a general situation, as can be deducted from the high rate of reactivation and the association of an increased risk of ocular toxoplasmosis reactivation with age [Citation53]. Two additional facts indicate that persistent cyst tissue in the eye is responsible for reactivation: the lack of acute markers for most acute episodes (~80%) of ocular toxoplasmosis [Citation5,Citation54–56] and the presence of parasite DNA in eye bank specimens [Citation46]. Our estimation of the mean length of specific IgM after primary infection is two years in the Colombian population [Citation57] therefore, we can estimate that, at least in Colombian patients, when active retinochoroiditis manifests, this occurs after infection was acquired more than two years before.

Figure 1. Scheme of the innate and adaptive immune response occurred during human ocular toxoplasmosis. Created with BioRender.com. a. local innate immune response components in the human eye responsible for the control of the Toxoplasma gondii retinal infection. b. Local adaptative immune response in the human eye responsible for controlling the Toxoplasma gondii retinal infection. Abbreviations: TLR: toll-like receptors, NK: natural Killer cells; RPE: retinal pigmentary epithelium; IDO: indoleamine Oxygenase; GBP: guanylate binding proteins; PMN: polymorph mononuclear cells; MՓ: macrophages.

Figure 1. Scheme of the innate and adaptive immune response occurred during human ocular toxoplasmosis. Created with BioRender.com. a. local innate immune response components in the human eye responsible for the control of the Toxoplasma gondii retinal infection. b. Local adaptative immune response in the human eye responsible for controlling the Toxoplasma gondii retinal infection. Abbreviations: TLR: toll-like receptors, NK: natural Killer cells; RPE: retinal pigmentary epithelium; IDO: indoleamine Oxygenase; GBP: guanylate binding proteins; PMN: polymorph mononuclear cells; MՓ: macrophages.

When infection leads to retinal damage, the pathological description can inform what cells are affected by the main characteristics of the inflammatory reaction [Citation28]. Helenor Wilder accomplished the first description of the pathological changes induced by toxoplasmosis in 1952 in a case series of 53 enucleated eyes with granulomatous lesions where central necrosis and the presence of tachyzoites of T. gondii were consistently found in the necrotic areas [Citation58]. The lesions comprised focal necrotizing retinitis, characterized by inflammation and tissue damage with mixed inflammatory reactions, mainly lymphocytes and monocytes [Citation58]. In pathological material, a well-demarcated area of coagulative necrosis is observed with adjacent choroiditis, vasculitis, hemorrhage, and vitritis [Citation40,Citation57]. Viable tachyzoites and tissue cysts may be found in superficial layers of the infected retina, along with an intense mononuclear inflammatory cell reaction seen in the involved retina and adjacent choroid and vitreous [Citation40].

One interesting noninvasive imaging technique providing in vivo information is optical coherence tomography (OCT), an interferometric, noninvasive optical tomographic imaging technique offering micrometer penetration [Citation59]. The results of this technique have demonstrated the involvement of retinal layers during acute episodes of ocular toxoplasmosis [Citation60,Citation61]. Typical OCT findings in toxoplasmic chorioretinitis are the retinal pigment epithelium proliferation and hyperpigmentation, an increased backscatter from the choroid consistent with pigmentary atrophy, and the preferential involvement and thinned of superficial layers of the retina, which is consistent with the preference of toxoplasmosis for neural tissue [Citation59]. In one study from Brazil, in all patients at the active lesion site, the inner retinal layers were abnormally hyper-reflective with thickening of the posterior hyaloid as well as of the subjacent choroid, and changes at the retinal pigmentary epithelium in two-thirds of the patients [Citation62]. At the same time, only one-fifth had subretinal fluid [Citation62]. The recent introduction of optical coherence tomography angiography demonstrated that retinal vasculitis is another common finding of ocular toxoplasmosis, in addition to typical focal necrotizing retinitis [Citation60,Citation63].

Additional data come from ex vivo cups of the human eye [Citation64] This model provides essential information that indicates tachyzoites of T. gondii cross the vascular endothelium to access the human retina by at least three routes: inside leukocytes that migrate to the retina, as a transmigrating tachyzoite, and after infecting endothelial cells [Citation64]. The retinal Müller glial cells are the preferred initial host cells; then, by invading retinal pigment epithelial cells, the secretion of growth factors is altered and induces the proliferation of adjacent uninfected epithelial cells [Citation64]. Indeed, this model gave a very realistic dynamic view of the eye invasion process.

3. The role and mechanisms of parasite strains diversity and virulence in ocular toxoplasmosis severity

The clinical forms of ocular toxoplasmosis in immunocompetent people infected in South America are more severe than in other continents [Citation65–68]. This has been linked to infection with virulent strains [Citation68,Citation69]. Notoriously, Toxoplasma has a biphasic distribution of the parasite population [Citation70]. The strains in the northern hemisphere have a clonal distribution and are less polymorphic; in contrast, the parasite strains from South America are more diverse genetically and have virulent alleles more frequently than strains from other continents [Citation71–73]. This can be explained because the most probable apparition of Toxoplasma species was in South America, specifically, the Colombian Atrato region, as deduced from the phylogeographic analysis [Citation74]. In South America, parasite dissemination occurred through slow feline dispersion in a wild environment, keeping remarkable genetic polymorphism and conserving virulent alleles. In contrast, the species was transmitted faster through domestic cats associated with human populations in Africa, Asia, and Europe privileging attenuated clonal species [Citation74–76]. The virulent alleles are related to genes coding for protein kinases derived from the organelle rhoptry of the parasite [Citation77]. Some groups of rhoptry-derived protein kinases fundamentally affect the host immune response. For instance, ROP16 coopts the intracellular signaling of STAT proteins necessary for the IFN-mediated response [Citation78–80] and ROP18 inhibits the host proteins that lyse the parasite vacuole [Citation81].

The first demonstration of the pertinence of Toxoplasma virulent factors in humans described a higher inflammatory response in patients harboring strains with the ROP18 virulent allele [Citation82]. While any strain can induce ocular damage in human toxoplasmosis, more severe clinical forms are related to infections by strains from serotype I [Citation76]. This serotype possesses the virulent versions of many rhoptry proteins, being the ROP5, the critical locus for virulence [Citation83].

4. Cytokine networks in ocular toxoplasmosis

An essential element in the immune response is how cells communicate with each other to activate or regulate inflammatory processes through networks of cytokines [Citation84]. These networks have been studied in patients with ocular toxoplasmosis, allowing us to know which cytokines are involved in the inflammatory and reactivation processes of ocular toxoplasmosis [Citation85]. The eye has a specific and independent intraocular immune response, making it an immunologically privileged organ [Citation86]. For this reason, exploring and comparing the systemic and intraocular cytokine network response are fascinating topics in ocular toxoplasmosis [Citation87].

The role of cytokines in toxoplasmic uveitis is complex because the predominance of a particular set of cytokines varies according to the time, the intensity of the antigenic stimuli, and the simultaneous presence of cytokines with antagonistic effects [Citation88]. It should be remembered that the immune response looks for an equilibrium between the efficacious inhibitions of the parasite’s replication or its destruction and the control of an excessive detrimental inflammatory response [Citation89,Citation90]. Therefore, mapping the cytokine levels in the eye (cytokinome) can enhance our understanding of the disease’s mechanisms and help develop targeted treatments [Citation85]. Examining cytokine levels might also help predict relapses and monitor disease activity in patients with uveitis [Citation84,Citation91]. For this reason, some studies have sought to generate a methodology to determine the degree and patterns of cytokine secretion in uveitis, considering its potential clinical utility and the knowledge it can provide about the immunopathogenesis of this condition [Citation69,Citation84,Citation92].

In this sense, some studies have measured several intraocular cytokines/chemokines in patients diagnosed with OT in the aqueous humor [Citation66,Citation93]. Correlations were evaluated between cytokine/chemokine levels, type of inflammatory response (Th1, Th2, Th17, Treg), and clinical characteristics [Citation68,Citation94–96]. A predominant Th2 response was associated with more severe clinical features [Citation68,Citation85]. The presence of VEGF and IL-5 was related to a higher number of recurrences [Citation85]. Growth factors (VEGF, FGF, PDGF-β) were related to more lesions [Citation85]. Patients infected by type-I/III strains had a particular intraocular cytokine pattern related to more severe clinical characteristics [Citation85]. Additional studies of the aqueous humor cytokinome showed slight differences between primary acquired ocular toxoplasmosis and recurrent cases [Citation93].

In addition, it has been described that severe ocular lesions occur from a combination of host gene susceptibility and exposure to more aggressive strains [Citation69]. The parasite and its host’s relationship could affect clinical presentation, treatment, and prognosis [Citation97]. In this context, host cytokines and promoter sequences polymorphisms have been studied and observed more frequently in individuals with ocular lesions than controls in the same geographical areas [Citation98–102].

Considering the characteristics in different geographical regions, comparing the cytokine profile in the eye between French and Colombian patients revealed that Colombian patients exhibited an intraocular polarized Th2 cytokine response [Citation85]. Similarly, Colombian patients with ocular toxoplasmosis displayed a peripheral Th2-skewed response [Citation103]. The peripheral immune response also showed a significant increase in IL-6, IL-10, and TGF-β mRNA levels in the patients with ocular toxoplasmosis compared to the chronically infected individual without ocular lesions and negative controls [Citation104]. TNF-α and IL-12 mRNA levels were up-regulated in patients with ocular lesions but did not reach statistical significance. Furthermore, IL-27 and IFN-γ mRNA levels were higher in patients with ocular toxoplasmosis than in negative controls, and these differences were statistically significant [Citation104].

South American patients experience more severe clinical symptoms than European patients, including higher levels of inflammation, more lesions, and larger lesion sizes [Citation68]. This could be attributed to the South American strains’ ability to weaken the protective effect of IFN-γ by manipulating the immune response through a Th2 cytokine profile and upregulation of IL-17 [Citation105]. IL-17, produced by Th17 and Müller cells, negatively impacts IFN-γ production, decreasing the protective immune response against Toxoplasma gondii [Citation105].

The production of IFN-γ by human peripheral blood mononuclear cells (PBMC) in response to intracellular pathogens like T. gondii is influenced by both genetic factors, such as polymorphisms in the IFN-γ gene and genes that inhibit IFN-γ synthesis like IL-10 [Citation69,Citation106,Citation107]. Other factors, including DNA methylation, posttranscriptional mechanisms like miRNA modification of IFN-γ mRNA, the type of infection (congenital or acquired), and the presence of ocular or cerebral lesions, also affect IFN-γ production [Citation108,Citation109]. Patients with ocular hypertension associated with ocular toxoplasmosis who produce high levels of intraocular IFN-γ during reactivation of toxoplasmic retinochoroiditis can be identified as high releasers of IFN-γ by PBMCs in cytokine release assays [Citation106]. These assays can also help to identify patients with acquired ocular toxoplasmosis who exhibit low release of IFN-γ [Citation106]. This patient group has a higher risk of experiencing extended disease progression and severe complications. Including local strains in the cytokine release assay when using T. gondii lysates is recommended [Citation106].

Moreover, the release of IFN-γ by Natural Killer cells (NK cells) plays a crucial role in innate immunity during T. gondii infection. Interleukin 12 (IL-12) is a critical cytokine that promotes the generation of IFN-γ by NK cells. Various studies have demonstrated the influence of cytokines on NK cell activation, with IL-2 being an important stimulatory factor [Citation110].

Focusing on the interferon role, an in vitro model examined the immune interaction among retinal cells, specifically concentrating on the involvement of type I and III interferons in the barrier function [Citation111]. It was shown that IFN-γ influenced the parasite proliferation, and the regulation of the barrier function of the outer blood-retinal barrier (oBRB) is associated with type I and III interferons producing IFN-λ1 in a manner independent of STAT1 [Citation111]. These findings may help identify potential immunological targets related to these interferons and provide possible treatment options for this ocular disease [Citation111].

In conclusion, the information provided by studies focused on evaluating the network and kinetics of intraocular cytokines () in humans has contributed the most to understanding what happens in real-life scenarios within the eyes of individuals infected with Toxoplasma gondii. This information has been precious in studying and comparing ocular immune responses of cytokines among patients infected with parasite strains in various geographic areas, considering that the local response in an immunologically privileged organ like the eye provides the most specific information about what occurs at the local level. Investigating the cytokinome within the eye can enhance our comprehension of the mechanisms underlying the disease and aid in developing precise and advanced targeted therapies. This information could also potentially facilitate the prediction of relapses and enable the monitoring of disease activity in patients with ocular toxoplasmosis.

5. Immunogenetics studies

The fact that most people who acquire toxoplasmosis do not develop ocular disease is a solid argument for considering individual susceptibility [Citation112,Citation113]. Certain genetic predisposition variations have been associated with an increased likelihood of developing ocular involvement. summarizes the polymorphisms by cytokine that showed significant association with ocular toxoplasmosis. Many polymorphisms related to recurrences, like IL-1α −889 C/T [Citation101], IL-10 -1082A/G [Citation69,Citation102] and IFN-γ +874 T/A [Citation101,Citation121], should be considered indicative biomarkers for prophylaxis against recurrences [Citation133,Citation134]. The findings of theses immunogenetic studies confirm the importance of gene regulation in the immune response and the development of toxoplasmic retinochoroiditis [Citation112,Citation113]. However, due to the intricate nature of the immune response to parasites, it is improbable that genetic variation at a single location can fully explain the differences in immune responses and clinical manifestations among hosts [Citation112,Citation113]. Understanding disease susceptibility and genetic factors is essential for the functional assessment of disease-related gene variations [Citation112,Citation113]. Most studies analyze the putative association between the frequency of the gene polymorphisms and the presence of the disease. Still, very few go until the demonstration of how the specific polymorphisms affect the synthesis or activity of the cytokine [Citation112,Citation113].

Table 1. Genetic polymorphisms associated to ocular toxoplasmosis.

Important and notoriously, in the + 874 site of the promotor for IFN γ (SNP rs24305619), the T/A or A/A polymorphisms are present in 95% of Colombian cases with ocular toxoplasmosis. In contrast, in people without ocular toxoplasmosis, 80% have the TT or TA alleles [Citation98]. In Santa Rita de Cassia, in Brazil, 91% of patients had T/A or A/A polymorphisms but with a similar frequency of theses alleles in community seropositive controls; however, no IgM was performed to establish that non-recent infections were present, and no follow-up was reported to show apparition of ocular lesions in this high prevalent region [Citation101]. The + 874 site of the IFN γ promotor is a NFkB binding site [Citation135], and in health volunteers, the presence of the T allele was associated with increased IDO activity [Citation136]. The significance of these polymorphisms was proved by T. gondii stimulation in ex vivo experiments, showing that the leukocytes from people with ocular toxoplasmosis possessing the T/A allele significantly produced less IFN γ than those with the T/T allele [Citation137]. The polymorphisms in this genetic locus are associated with susceptibility to severe infections by many microorganisms [Citation135,Citation138,Citation139].

6. Lymphocyte subpopulations

The studies about lymphocyte subpopulations (CD4+ memory, effector) are the start point to understanding how cytokines synthesis is modulated. Still, most of the studies in ocular toxoplasmosis remain in the initial characterization of the systemic CD4+ and CD8+ T cell immune responses and the cytokines they produce [Citation140–142]. In addition, other leukocytes are essential in this landscape, including Natural Killer (NK) cells and neutrophils [Citation110,Citation143,Citation144]. All these leukocyte subsets can produce interferon-gamma (IFN-γ), providing a significant response against the parasite and mediating host resistance [Citation145]. Consistently, human eyes removed from patients with ocular toxoplasmosis showed a heterogeneous leucocytic infiltrate [Citation32,Citation146,Citation147].

Also, in the eye environment, it is essential to consider the retinal pigment epithelium (PE), which modulates the activation status of multiple lymphocyte subsets and reduces intraocular inflammation [Citation148]. Therefore, regarding CD8+ T cells, a key lymphocyte population controlling the infection against T. gondii, a recent study explored the interaction between human CD8+ T cells and retinal PE in the setting of T. gondii tachyzoite infection [Citation145]. The authors exposed confluent human retinal PE monolayers to GT-1 strain tachyzoites and subsequently co-cultured the cell monolayers with CD8+ T cells obtained from human peripheral blood [Citation64]. They found a reduced production of IL-2 (an indicator of T cell activation) in T cells and observed differential expression of the immunomodulatory marker (programmed cell death-ligand 1, PD-L1 transcript), which increased in retinal PE cells infected with the parasite [Citation64]. Since PD-L1 inhibits T cell activation, its elevated levels in retinal PE cells were expected to inhibit IL-2 production by CD8+ T cells [Citation64]. This finding could support what was previously suggested about how retinal PE cells can convert CD4+ T and CD8+ T cells into T regulatory cells, along with expressing TGFβ and through the expression of ligands for programmed cell death pathways, such as FAS ligand (FasL) and PD-L1 [Citation64]. We recently confirmed these results by analyzing the expression of CD8+ T – cell exhaustion immunomodulatory markers in peripheral blood mononuclear cells (PBMCs) from patients with ocular toxoplasmosis, and we found that the gene expression of PD-1 (programmed cell death 1 protein) was higher in ocular toxoplasmosis compared to asymptomatic or uninfected individuals [Citation149]. Additionally, membrane expression of PD1 was observed in the central memory CD8+ T lymphocytes subset, and most patients with ocular toxoplasmosis showed a total exhaustion phenotype [Citation149]. Also, when PBMCs were stimulated ex vivo with total antigen from T. gondii, it was found an inverse correlation between the exhaustion markers and clinical characteristics (lesion size, recurrence index, and number of lesions), suggesting that PD-1 expression could have a beneficial role during immediate response to a new challenge and therefore by controlling an excessive inflammatory response [Citation149].

Similar research, using PBMCs from patients with toxoplasmic retinochoroiditis and after stimulation with T. gondii antigen, found that CD4+ and CD8+ T cells were the main sources of IL-10 cytokine, but also for IFN-γ and TNF-α, other lymphocyte populations were a relevant source of inflammatory cytokines [Citation150]. Interestingly, a negative correlation was observed between ocular lesion size and IL-10 expression by CD4+ lymphocytes. Therefore, this study showed that T cells are the main lymphocyte populations expressing IL-10 and its protective role for excessive damage in patients with toxoplasmic retinochoroiditis [Citation150]. Besides the IL-10 response from CD4+ T lymphocytes, it has been detected the IL-17A production by resident retinal cells rather than infiltrating T cells in the eye [Citation151]. The observed distribution and early secretion of IL-17A in the eye led to suspect that resident cells, such as glial cells and astrocytes, may be responsible for this early production of IL-17A [Citation151]. Local IL-17A production by resident cells plays a central role in the pathology of ocular toxoplasmosis [Citation151,Citation152]. The balance between Th17 and Th1 responses, by regulating especially IFN-γ levels, is crucial for the outcome of infection [Citation151,Citation152].

Additional key cytokines participating in this cellular response involve IL-23, which induces the proliferation of IL-17-producing cells, and IL-27, counterplayer to IL-17 May 2001regulate Th1-cell-mediated responses in ocular toxoplasmosis [Citation51]. The importance of these cytokines in experimental models of uveitis and encephalitis has been reported. CD4+CD25+ regulatory T-cells may control the local inflammatory response and protect the host against collateral inflammatory tissue damage. The responses of these cells to ocular infection by T. gondii may be suitably tailored to cope with either an acquired or a congenital origin [Citation51]. On the other side, the beneficial or pathogenic roles of the cytokines produced by Th1 and Th17 cells and the protective and homeostatic roles of IL-10, TGF-β, and IL-27 in modulating the hypersensitivity responses induced by T. gondii are generally recognized [Citation153]. The IFN-γ and IL-17 cytokines produced by Th1 and Th17 cell responses were analyzed in patients with active ocular toxoplasmosis or presumed ocular toxoplasmosis. The authors found an increased IL-17A production in both groups of individuals, where the main cellular source of IL-17A was CD4(+) CD45RO (+) T-bet (-) IFN-γ (-) T-helper 17 cells [Citation151]. Furthermore, they found that the NOD2 gene influences the production of IL-17A by CD4+ T lymphocytes and might contribute to the development of ocular toxoplasmosis [Citation154].

Regarding other cell populations involved in the immune response inside the eye, it has been found the involvement of neutrophils in retinal inflammation in the setting of ocular toxoplasmosis [Citation155]. Neutrophils generated more reactive oxygen species when co-cultured with infected versus uninfected retinal pigment epithelial cells (ARPE-19 cells) [Citation155]. Infected ARPE-19 cells also induced neutrophils to produce inflammatory cytokines, like TNF-α and IL-1β, but this effect was not replicated in primary cells for TNF-α and proved to be donor-dependent for IL-1β [Citation155]. These results suggested infiltrating neutrophils contribute to retinal damage in ocular toxoplasmosis [Citation64].

Finally, the NK cells have also been involved in the presentation and diagnosis of ocular toxoplasmosis; a recent study found these cells as part of the putative biomarkers for early diagnosis and prognosis of congenital ocular toxoplasmosis [Citation156]. The analysis of in vitro T. gondii-specific IL5+CD4+ T-cells and IFN-γ+NK-cells displayed a high accuracy for early prognosis of the ocular lesion in infants with congenital toxoplasmosis attaining a global diagnostic accuracy of 0,8 and 0,9, respectively [Citation156]. The authors concluded about the relevance of employing the elements of the cell-mediated immune response as biomarkers with the potential to endorse early diagnosis and prognosis of congenital ocular toxoplasmosis to contribute to precise clinical management and effective therapeutic intervention [Citation156].

In , we summarize the evidence of T lymphocyte subpopulations; the most striking characteristic of ocular toxoplasmosis patients is that during the active phase of infection, it would be necessary to stimulate CD8+ effector subset cells expressing CD244 to limit the damage caused by inflammation [Citation149]. Contrarily, during the chronic phase, the CD8+ central memory subset expressing CD160+ should be limited [Citation149].

Table 2. CD8+ T lymphocyte subpopulations and exhaustion phenotype in patients with ocular toxoplasmosis.

7. Trigger signals for reactivation

Recurrences of lesions are present in patients with ocular toxoplasmosis in 56% in South America, in 46% in Europe and 39% in North America [Citation161]. How a quiescent parasite tissue cyst in the retina becomes active is a central question (and an anguish factor for patients and their families). Some clear conditions that trigger recurrences (or worsening clinical picture) are the use of subconjunctival steroids [Citation162], early initiation of steroids without antiparasitic drug [Citation163,Citation164], pregnancy [Citation165] age [Citation53] and ocular trauma [Citation166]. These triggers are related to an immunosuppressive status like immunosenescence [Citation167] or alteration in the blood-retinal barrier [Citation66], pointing to the critical maintenance of a protective local immune response [Citation64]. We found that drinking boiled water, a protective factor against reinfection, was associated with a lower rate of recurrences [Citation168]. Secondary infection with a different strain can lead to a superinfection status where CD8 T cell IFN-γ response is impaired [Citation169]. The risk of recurrence is higher immediately after an active episode with a clustering pattern [Citation66,Citation170,Citation171].[Citation165,Citation170]

In our opinion, based on the physiopathological factors involved in this disease, ocular toxoplasmosis should always be treated with a combination of steroids and antiparasitic drugs, even for small lesions, because at the moment it is not possible to predict who or when an individual with ocular lesions will have recurrences [Citation66,Citation162]. The steroids reduce the time to clearance of the vitreous and enhance lesion size regression, shortening the time for visual recovery [Citation172]. In turn, antiparasites will inhibit parasite replication, limiting additional tissue damage and probably potentializing the host immune response by limiting the production of parasite virulence factors [Citation173,Citation174]. The lack of evidence of efficacity of antiparasites in ocular toxoplasmosis is because not appropriate outcome have been defined to evaluate the benefit of therapy, thus antiparasite treatment should be given with the aim of reducing the risk of recurrences after treatment [Citation171,Citation172,Citation175].

8. Implications for current and next-generation treatments

The understanding of the role of different cells of the immune system to control invasion and permanence of the parasite within the cells of the retina has illuminated the search for new therapeutic strategies improving the efficacity of the antibiotics traditionally used to treat the active disease or reduce recurrences [Citation32,Citation176,Citation177]. A potential therapeutic target is studying and understanding inflammatory pathways and cytokine networks [Citation64,Citation177]. Blocking inflammatory molecules may help arrest excessive inflammation and subsequent retinal damage [Citation94,Citation105]. However, much remains to be discovered in this intricate mechanism that becomes even more complicated if we consider the characteristics of the eye as an immunologically privileged organ [Citation148,Citation178]. The crucial cytokine defending the host retinal cells against T. gondii is IFN γ [Citation145]. But, the potential activation of other IFNs like the type I (α and β) or type III (λ) pathways are also under study [Citation111]. However, some in vitro results show that inhibition of type I IFN can be beneficial [Citation179]. The apparent contrary effect of cytokines in different in vitro and in vivo models of infection can be explained by considering that the positive or detrimental effect of one given cytokine depends on the stage of the infection and the fine-tuning of their levels [Citation153,Citation180]. Additionally, while targeted cytokine inhibitors can mitigate tissue destruction mediated by higher cytokine production, cytokine-targeted therapeutics’ effectiveness is difficult to predict due to disease heterogeneity [Citation181]. To establish the IFN type, the stage of the infection will be beneficial, and the clinical criteria to be applied is critical for future immune therapies [Citation181].

The IL-17 axis, as a potent inflammatory agent in ocular toxoplasmosis, leading to severe inflammation and subsequent tissue damage, is, in theory, an attractive target for inhibitory antibodies [Citation95,Citation105,Citation152]. Currently, several available therapies, particularly monoclonal antibodies like ixekizumab, brodalumab, and secukinumab, that target the IL17 pathway, are being evaluated or used to treat autoimmune diseases such as psoriasis [Citation182,Citation183], ankylosing spondyloarthritis [Citation184] and ustekinumab targeting the IL-23/IL-17 axis used in psoriatic arthritis, Crohn’s disease, and uveitis [Citation185]. However, the potential use of these medications in controlling intraocular inflammation secondary to T. gondii has not been studied. The possible side effects of these therapies in this parasitic infection are unknown, especially in patients infected with virulent strains that downregulate the IL17 secretion and yet present severe tissue damage [Citation68].

Targeting other molecules like IL6, a pleiotropic cytokine involved in the inflammatory process, including intraocular inflammation, using monoclonal antibodies such as tocilizumab could offer alternative therapeutic options [Citation186–188].

The role of therapies targeting different molecules, such as IL-5 and growth factors (VEGF, FGF, PDGF-β), that have been related to a higher number of recurrences and more lesions must be elucidated [Citation85]. Anti-IL-5 is used to treat eosinophilic asthma and anti-VEGF as antiangiogenic therapy [Citation189]. Nevertheless, the role of these therapies in ocular toxoplasmosis has not been explored.

While these medications may alleviate inflammation, their effectiveness as antiparasitic agents is not their main activity. Therefore, proper antiparasitic drugs should be included in all these potential treatment regimens. Nonetheless, immune-based treatments could be promising in inducing an appropriate response to prevent tissue damage, reduce recurrences, and hopefully, in the future, limit parasite growth [Citation32].

9. Expert opinion

Altogether, the data from eye fund screening and from outbreaks with ocular involvement in different continents and geographical areas (such as India, Canada, United States, Colombia, or Brazil) point to a risk of ocular involvement that can be manifested months after primary infection in around one-tenth of infected people. Some genetic polymorphism exists linked to the susceptibility to develop an ocular episode, especially the + 874 nucleotide of the promotor for IFN γ (SNP rs24305619). Still, it has not been evaluated for use in routine practice. Additional polymorphisms should be explored as biomarkers for recurrence (IL-1α −889 C/T; and IL-10 -1082A/G) and their use as criteria to establish antiparasite prophylaxis. More robust evidence is needed regarding the necessity of treatment of primary infection to reduce the development of ocular toxoplasmosis and a separate risk of later development of mental disease. However, on the precaution principle, in our opinion, we recommend treating acute acquired toxoplasmosis, yet clinical trials are necessary to validate this recommendation for all geographical areas. Although our understanding of the underlying causes of ocular toxoplasmosis has improved, many unanswered questions remain. The complete mapping of the immune response has yet to be entirely recognized.

The ideal in the future is to find an antibiotic that will penetrate cysts containing bradyzoites in the intraocular tissues, which has not been possible until now. Treatments with molecules that block inflammatory pathways can be adjuvant treatments, but the essence of curative treatment is the elimination of tissue cysts. Multiple tests have been proposed by in vitro and animal models, but in real life in patients infected with Toxoplasma gondii, it has not been possible to eradicate the parasites encysted in the retina that remain latent for the host’s entire life.

Article highlights

  • Ocular toxoplasmosis develops in around one-tenth of people after a primary infection.

  • Susceptibility to developing ocular toxoplasmosis and its severity result from the interplay between host genetic factors and parasite virulence.

  • The most critical genetic markers related to susceptibility to ocular toxoplasmosis are polymorphisms in the IFN γ promoter region.

  • Future treatments with molecules that block inflammatory pathways could be adjuvant treatments, but the essence of curative treatment is the elimination of tissue cysts.

  • Medications targeting tissue cysts responsible for recurrences are a current priority.

Declaration of interests

The authors have no relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript. This includes employment, consultancies, honoraria, stock ownership or options, expert testimony, grants or patents received or pending, or royalties.

Reviewer disclosures

Peer reviewers on this manuscript have no relevant financial or other relationships to disclose.

Geolocation information

H83Q+HW Armenia, Quindío

Acknowledgments

To all our patients, who are also part of the research team and altruistically participated and contributed with their information, clinical samples, and support without awaiting an immediate benefit but always hoping for a future benefit for them and others. To Dr. William Rojas Carabali for help in Figures elaboration.

Additional information

Funding

This paper was not funded.

References

  • Gomez-Marin JE, de-la-Torre A Ocular disease due to Toxoplasma gondii. Toxoplasma gondii [Internet]. Elsevier; 2020 [cited 2023 Jan 24]. p. 229–291. Available from: https://linkinghub.elsevier.com/retrieve/pii/B9780128150412000050
  • Rahmanian V, Rahmanian K, Jahromi A, et al. Seroprevalence of toxoplasma gondii infection: an umbrella review of updated systematic reviews and meta-analyses. J Family Med Prim Care [Internet]. 2020 [cited 2023 Feb 13];9:3848. 8. doi: 10.4103/jfmpc.jfmpc_753_20
  • Dubey JP. Toxoplasmosis – a waterborne zoonosis. Vet Parasitol Internet. 2004;126(1–2):57–72. doi: 10.1016/j.vetpar.2004.09.005
  • Tenter AM, Heckeroth AR, Weiss LM. Toxoplasma gondii: from animals to humans.Int J Parasitol Internet. 2000 [cited 2015 Aug 5];30(12–13):1217–1258. doi: 10.1016/S0020-7519(00)00124-7
  • Nsiangani Lusambo N, Kaimbo Wa Kaimbo D, Mumba Ngoyi D, et al. Clinical and serological characteristics of ocular toxoplasmosis in the democratic Republic of Congo. 2022. Internet [cited 2023 Jul 17]; doi: 10.1080/0927394820222140297
  • Lusambo NN, Kaimbo Wa Kaimbo D, Mumba Ngoyi D. Risk factors for ocular toxoplasmosis among uveitis patients in Kinshasa, DR Congo.BMJ Open Ophthalmol Internet. 2023 [cited 2023 Jul 17];8(1):1198. doi: 10.1136/bmjophth-2022-001198
  • Gómez-Marín JE, Muñoz-Ortiz J, Mejía-Oquendo M, et al. High frequency of ocular toxoplasmosis in Quindío, Colombia and risk factors related to the infection. Heliyon Internet. 2021 [cited 2021 Apr 8];7(4):e06659. doi: 10.1016/j.heliyon.2021.e06659
  • Ferreira AIC, De Mattos CCB, Frederico FB, et al. Risk factors for ocular toxoplasmosis in Brazil. Epidemiol Infect [Internet]. 2014 [cited 2020 Feb 28];142(1):142–148. doi: 10.1017/S0950268813000526
  • Robert-Gangneux F, Dardé M-L. Epidemiology of and diagnostic strategies for toxoplasmosis. Clin Microbiol Rev Internet. 2012;25(2):264–296. doi: 10.1128/CMR.05013-11
  • Petersen E, Kijlstra A, Stanford M. Epidemiology of ocular toxoplasmosis. Ocul Immunol Inflamm. 2012;20(2):68–75. InternetAvailable from: http://www.ncbi.nlm.nih.gov/pubmed/22409558%5Cnhttp://informahealthcare.com/doi/abs/10.3109/09273948.2012.661115
  • Milne GC, Webster JP, Walker M. Is the incidence of congenital toxoplasmosis declining? Trends Parasitol Internet. 2023 [cited 2023 Jan 25];39(1):26–37. doi: 10.1016/j.pt.2022.10.003
  • Mejia-Oquendo M, Marulanda-Ibarra E, Gomez-Marin JE. Evaluation of the impact of the first evidence-based guidelines for congenital toxoplasmosis in Armenia (Quindío) Colombia: an observational retrospective analysis. Lancet Reg Health - Am Internet. 2021 [cited 2023 Feb 24];1:100010. doi: 10.1016/j.lana.2021.100010
  • Hou JH, Patel SS, Farooq AV, et al. Decline in ocular toxoplasmosis over 40 years at a tertiary referral practice in the United States. Ocul Immunol InflammInternet. 2018 [cited 2023 Jul 17];26:577–583. https://click.endnote.com/viewer?doi=10.1080%2F09273948.2016.1246665&token=WzEwNzI1NCwiMTAuMTA4MC8wOTI3Mzk0OC4yMDE2LjEyNDY2NjUiXQ.XM8ZuJkfFpbxDBf3-0YhGhz-aK8
  • Cifuentes-González C, Zapata-Bravo E, Sierra-Cote MC, et al. Colombian ocular infectious Epidemiology study (COIES): ocular toxoplasmosis incidence and sociodemographic Characterization, 2015-2019. Inter J Infect Dis. 2022;117:349–355. doi: 10.1016/j.ijid.2022.02.028
  • Semenza JC, Rocklöv J, Ebi KL. Climate change and cascading risks from infectious disease. Infect Dis Ther. 2022;11(4):1371–1390. doi: 10.1007/s40121-022-00647-3
  • Pozio E. How globalization and climate change could affect foodborne parasites. Exp Parasitol Internet. 2020 [cited 2022 Jul 13];208:107807. doi: 10.1016/j.exppara.2019.107807
  • Gomez-Marin JE, Pinto Y, Lora F, et al. Recipe ingredients for re emergent protozoa: climatic change, rain, zoonosis, mountain and food. Infectio Internet. 2022 [cited 2023 Jan 25];26:381–383. doi: 10.22354/24223794.1080
  • FAO; WHO. Multicriteria-based ranking for risk management of food-born parasites [Internet]. First. WHO; FAO, Editor. Microbiological Risk Assessment Series No. 23. Rome. Rome: FAO/WHO; 2014 [cited 2015 Oct 2]. Available from: http://www.fao.org/3/a-i3649e.pdf
  • Jones JL, Dubey JP. Foodborne toxoplasmosis.Clin Infect Dis Internet. 2012 [cited 2015 Sep 18];55(6):845–851. doi: 10.1093/cid/cis508
  • Foodborne Disease Burden Epidemiology Reference Group 2007-2015. WHO estimates of the global burden of foodborne diseases: foodborne diseases burden epidemiology reference group 2007-2015 [Internet]. 2015 [cited 2022 Jul 8]. Available from: https://www.who.int/publications/i/item/9789241565165
  • Shapiro K, Bahia-Oliveira L, Dixon B, et al. Environmental transmission of Toxoplasma gondii: oocysts in water, soil and food. Food Waterborne Parasitol Internet. 2019;15:e00049. doi: 10.1016/j.fawpar.2019.e00049
  • Nessim J, Luna-Ramirez JC, Moreno-Gómez GA, et al. Estimations of the number people with mental diseases associated with toxoplasmosis and identification of risk factors by continent. Psychiatry Res Internet. 2023 [cited 2023 Mar 9];115130:115130. doi: 10.1016/j.psychres.2023.115130
  • Palanisamy M, Madhavan B, Balasundaram M, et al. Outbreak of ocular toxoplasmosis in Coimbatore, India. Indian J Ophthalmol Internet. 2006;54(2):129. doi: 10.4103/0301-4738.25839
  • Bowie WR, King AS, Werker DH, et al. Outbreak of toxoplasmosis associated with municipal drinking water. The BC toxoplasma investigation team. Lancet Internet. 1997;350(9072):173–177. doi: 10.1016/S0140-6736(96)11105-3
  • Dubey JP. Outbreaks of clinical toxoplasmosis in humans: five decades of personal experience, perspectives and lessons learned. Parasites Vectors Internet. 2021 [cited 2023 Jan 25];14(1): doi: 10.1186/s13071-021-04769-4
  • Vaudaux JD, Muccioli C, James ER, et al. Identification of an atypical strain of Toxoplasma gondii as the cause of a waterborne outbreak of toxoplasmosis in Santa Isabel do Ivai, Brazil. J Infect Dis. 2010;202(8):1226–1233.
  • Arantes TEFF, Silveira C, Holland GN, et al. Ocular involvement following Postnatally acquired Toxoplasma gondii infection in Southern Brazil: a 28-year experience. Am J Ophthalmol Internet. 2015 [cited 2015 Jul 1];159(6):1002–1012.e2. doi: 10.1016/j.ajo.2015.02.015
  • Frenkel JK. Pathophysiology of toxoplasmosis. Parasitol Today Internet. 1988;4(10):273–278. doi: 10.1016/0169-4758(88)90018-X
  • Sharma P, Chitnis CE. Key molecular events during host cell invasion by apicomplexan pathogens. Curr Opin Microbiol. 2013;16(4):432–437. doi: 10.1016/j.mib.2013.07.004
  • Blader IJ, Coleman BI, Chen C-T, et al. Lytic Cycle of Toxoplasma gondii : 15 Years Later. Annu Rev Microbiol [Internet]. 2015 [cited 2020 Oct 14];69:463–485. 1 doi: 10.1146/annurev-micro-091014-104100
  • Skariah S, McIntyre MK, Mordue DG. Toxoplasma gondii: determinants of tachyzoite to bradyzoite conversion.Parasitol Res Internet. 2010 [cited 2023 Jul 23];107(2):253–260. doi: 10.1007/s00436-010-1899-6
  • Greigert V, Bittich-Fahmi F, Pfaff AW. Pathophysiology of ocular toxoplasmosis: facts and open questions. Taylan Ozkan A, editor. PLoS Negl Trop Dis [Internet] Available from. 2020 [cited 2021 Sep 1];14(12):e0008905. doi: 10.1371/journal.pntd.0008905
  • Daré LO, Bruand P-E, Gérard D, et al. Associations of mental disorders and neurotropic parasitic diseases: a meta-analysis in developing and emerging countries. BMC Public Health [Internet]. 2019 [cited 2020 Mar 29];19(1):1645. doi: 10.1186/s12889-019-7933-4
  • Petersen E, Ajzenberg D, Mandelbrot L, et al. Protozoan Diseases: Toxoplasmosis. International Encyclopedia Of Public Health [Internet]. Amsterdan: Elsevier; 2017 [cited 2019 May 26]. p. 114–132. Available from: https://www.sciencedirect.com/science/article/pii/B9780128036785003611
  • Xiao J. Toxoplasma-induced behavioral changes: an aspecific consequence of neuroinflammation. Trends Parasitol Internet. 2020 [cited 2021 Jun 5];36(4):317–318. doi: 10.1016/j.pt.2020.01.005
  • Flegr J. Effects of Toxoplasma on human behavior. Schizophr Bull Internet. 2007;33(3):757–760. doi: 10.1093/schbul/sbl074
  • Milne G, Webster JP, Walker M. Toxoplasma gondii: an underestimated threat? Trends Parasitol Internet. 2020 [cited 2021 Jun 5];36(12):959–969. doi: 10.1016/j.pt.2020.08.005
  • Flegr J. How and why Toxoplasma makes us crazy. Trends Parasitol. 2013;29(4):156–163. Internet: https://linkinghub.elsevier.com/retrieve/pii/S1471492213000202
  • Delair E, Latkany P, Noble AG, et al. Clinical manifestations of ocular toxoplasmosis. Ocul Immunol Inflamm Internet. 2011;19(2):91–102. doi: 10.3109/09273948.2011.564068
  • Butler NJ, Furtado JM, Winthrop KL, et al. Ocular toxoplasmosis II: clinical features, pathology and management. Clin Exp Ophthalmol. 2013;41(1):95–108.
  • Huang PK, Jianping C, Vasconcelos-Santos DV, et al. Ocular toxoplasmosis in tropical areas: analysis and outcome of 190 patients from a multicenter collaborative study.Ocul Immunol Inflamm [Internet]. 2017 [cited 2023 Jul 24];26(8):1289–1296. doi: 10.1080/0927394820171367407
  • Bosch-Driessen LEHH, Berendschot TTJMJM, Ongkosuwito JV, et al. Ocular toxoplasmosis: clinical features and prognosis of 154 patients. Ophthalmol. 2002;109(5):869–878.
  • de-la-Torre A, López-Castillo CA, Gómez-Marín JE. Incidence and clinical characteristics in a Colombian cohort of ocular toxoplasmosis. Eye Internet. 2009 [cited 2015 Jan 22];23(5):1090–1093. doi: 10.1038/eye.2008.219
  • Glasner PD, Silveira C, Kruszon-Moran D, et al. An unusually high prevalence of ocular toxoplasmosis in Southern Brazil. Am J Ophthalmol [Internet]. 1992 [cited 2023 Jul 23];114(2):136–144. doi: 10.1016/S0002-9394(14)73976-5
  • de-la-Torre A, González G, Díaz-Ramirez J, et al. Screening by Ophthalmoscopy for Toxoplasma Retinochoroiditis in Colombia. Am J Ophthalmol Internet. 2007 [cited 2015 Jan 22];143(2):354–356. doi: 10.1016/j.ajo.2006.09.048
  • Commodaro AG, Chiasson M, Sundar N, et al. Elevated toxoplasma gondii infection rates for retinas from eye banks, Southern Brazil. Emerg Infect Dis [Internet]. 2016 [cited 2023 Jul 23];22:691–693. 4. doi: 10.3201/eid2204.141819
  • Burnett AJ, Shortt SG, Isaac-Renton J, et al. Multiple cases of acquired toxoplasmosis retinitis presenting in an outbreak. Ophthalmol. 1998;105(6):1032–1037.
  • de-la-Torre A, Valdés-Camacho J, de Mesa CL, et al. Coinfections and differential diagnosis in immunocompetent patients with uveitis of infectious origin. BMC Infect Dis Internet. 2019;19(1):91. https://bmcinfectdis.biomedcentral.com/articles/10.1186/s12879-018-3613-8
  • Silveira C, Muccioli C, Holland GN, et al. Ocular involvement following an epidemic of Toxoplasma gondii infection in Santa Isabel do ivaí, Brazil. Am J Ophthalmol Internet. 2015 [cited 2015 Jul 18];159(6):1013–1021.e3. doi: 10.1016/j.ajo.2015.02.017
  • de-la-Torre A, Gómez-Marín J. Disease of the Year 2019: ocular toxoplasmosis in HIV-infected patients. Ocul Immunol Inflamm. 2020;28(7):1031–1039. Internet: https://www.tandfonline.com/doi/full/10.1080/09273948.2020.1735450
  • Garweg JG, Candolfi E. Immunopathology in ocular toxoplasmosis: facts and clues. Mem Inst Oswaldo Cruz Internet. 2009;104(2):211–220. doi: 10.1590/S0074-02762009000200014
  • Rougier S, Montoya JG, Peyron F. Lifelong persistence of Toxoplasma cysts: a questionable dogma? Trends Parasitol Internet. 2016 [cited 2016 Dec 25]; Available from;33(2):93–101. doi: 10.1016/j.pt.2016.10.007
  • Holland GN. Ocular toxoplasmosis: the influence of patient age. Mem Inst Oswaldo Cruz Internet. 2009;104(2):351–357. doi: 10.1590/S0074-02762009000200031
  • Papadia M, Aldigeri R, Herbort CP. The role of serology in active ocular toxoplasmosis.Int Ophthalmol Internet. 2011 [cited 2023 Jul 26];31(6):461–465. doi: 10.1007/s10792-011-9507-z
  • Gómez-Marín JE, Montoya-de-Londoño MT, Castaño-Osorio JC, et al. Frequency of specific anti-Toxoplasma gondii IgM, IgA and IgE in Colombian patients with acute and chronic ocular toxoplasmosis. Mem Inst Oswaldo Cruz [Internet]. 2000 [cited 2015 Jan 22];95(1):89–94. doi: 10.1590/S0074-02762000000100014
  • Ongkosuwito JV, Bosch-Driessen EH, Kijlstra A, et al. Serologic evaluation of patients with primary and recurrent ocular toxoplasmosis for evidence of recent infection. Am J Ophthalmol Internet. 1999;128(4):407–412. doi: 10.1016/S0002-9394(99)00266-4
  • Bonfioli AA, Orefice F. Toxoplasmosis. Semin Ophthalmol. 2005;20(3):129–141. doi: 10.1080/08820530500231961
  • Wilder HC. TOXOPLASMA CHORIORETINITIS in ADULTS. AMA Arch Ophthalmol.Arch Ophtalmol Internet. 1952 [cited 2023 Jul 23];48(2):127–136. doi: 10.1001/archopht.1952.00920010132001
  • Gallagher MJ, Yilmaz T, Cervantes-Castañeda RA, et al. The characteristic features of optical coherence tomography in posterior uveitis. Br J Ophthalmol [Internet]. 2007 [cited 2023 Jul 24];91(12):1680–1685. doi: 10.1136/bjo.2007.124099
  • Sofia O, Wahyudi INSA, Susianti H, et al. Optical coherence tomography angiography findings in ocular toxoplasmosis with Multiple recurrences. Int Med Case Rep J Internet. 2023 [cited 2023 Feb 5];16:35–43. doi: 10.2147/IMCRJ.S395600
  • Oréfice JL, Costa RA, Campos W, et al. Third-generation optical coherence tomography findings in punctate retinal toxoplasmosis. Am J Ophthalmol. 2006;142(3):503–505.e2.
  • Oréfice JL, Costa RA, Scott IU, et al. Spectral optical coherence tomography findings in patients with ocular toxoplasmosis and active satellite lesions (MINAS report 1). Acta Ophthalmol [Internet]. 2013 [cited 2023 Jul 24];91(1):e41–e47. doi: 10.1111/j.1755-3768.2012.02531.x
  • Diniz B, Regatieri, Andrade R, et al. Evaluation of spectral domain and time domain optical coherence tomography findings in toxoplasmic retinochoroiditis. Clin Ophthalmol Internet. 2011;5:645. http://www.dovepress.com/evaluation-of-spectral-domain-and-time-domain-optical-coherence-tomogr-peer-reviewed-article-OPTH
  • Smith JR, Ashander LM, Arruda SL, et al. Pathogenesis of ocular toxoplasmosis. Prog Retin Eye Res [Internet]. 2021 [cited 2023 Jul 26];81:100882. doi: 10.1016/j.preteyeres.2020.100882
  • Carme B, Bissuel F, Ajzenberg D, et al. Severe acquired toxoplasmosis in immunocompetent adult patients in French Guiana. J Clin Microbiol Internet. 2002;40(11):4037–4044. doi: 10.1128/JCM.40.11.4037-4044.2002
  • Matet A, Paris L, Fardeau C, et al. Clinical and biological factors associated with recurrences of severe toxoplasmic retinochoroiditis confirmed by aqueous humor analysis. Am J Ophthalmol. 2019;199:82–93. doi: 10.1016/j.ajo.2018.11.013
  • Cortés DA, Aguilar MC, Ríos HA, et al. Severe acute multi-systemic failure with bilateral ocular toxoplasmosis in immunocompetent patients from urban settings in Colombia: case reports. Am J Ophthalmol Case Rep Internet. 2020 [cited 2022 Jul 18];18:100661. doi: 10.1016/j.ajoc.2020.100661
  • de-la-Torre A, Sauer A, Pfaff AW, et al. Severe South American ocular toxoplasmosis is associated with decreased ifn-γ/Il-17a and increased Il-6/Il-13 intraocular levels. Jardim A, editor. PLoS Negl Trop Dis [Internet].2013 [cited 2015 Jan 22];7(11):e2541. https://dx.plos.org/10.1371/journal.pntd.0002541
  • Mantilla-Muriel LE, Hernández-de-Los-Ríos A, Rincón M, et al. Serotyping, host genes and cytokines response in human ocular toxoplasmosis. Microb Pathog Internet. 2020 [[cited 2020 Sep 9]];148:104465. doi: 10.1016/j.micpath.2020.104465
  • Su C, Khan A, Zhou P, et al. Globally diverse Toxoplasma gondii isolates comprise six major clades originating from a small number of distinct ancestral lineages. Proc Natl Acad Sci U S A Internet. 2012;109(15):5844–5849. doi: 10.1073/pnas.1203190109
  • Khan A, Ajzenberg D, Mercier A, et al. Geographic separation of domestic and wild strains of Toxoplasma gondii in French Guiana Correlates with a monomorphic version of Chromosome1a. PLoS Negl Trop Dis. 2014;8(9):e3182.
  • Shwab EK, Jiang T, Pena HFJ, et al. The ROP18 and ROP5 gene allele types are highly predictive of virulence in mice across globally distributed strains of Toxoplasma gondii. Int J Parasitol [Internet]. 2016 [cited 2018 Dec 26];46(2):141–146. doi: 10.1016/j.ijpara.2015.10.005
  • Behnke MS, Khan A, Lauron EJ, et al. Rhoptry proteins ROP5 and ROP18 are Major murine virulence factors in genetically divergent South American strains of Toxoplasma gondii. Malik HS, editor. PLoS Genet [Internet] Available from. 2015 [cited 2018 Dec 26];11(8):e1005434. doi: 10.1371/journal.pgen.1005434
  • Bertranpetit E, Jombart T, Paradis E, et al. Phylogeography of Toxoplasma gondii points to a South American origin. Infect Genet Evol Internet. 2017;48:150–155. doi: 10.1016/j.meegid.2016.12.020
  • Gazzinelli RT, Mendonça-Neto R, Lilue J, et al. Innate resistance against Toxoplasma gondii: an evolutionary tale of mice, cats, and men. Cell Host Microbe Cell Press. 2014;15(2):132–138.
  • Lehmann T, Marcet PL, Graham DH, et al. Globalization and the population structure of Toxoplasma gondii. Proc Natl Acad Sci U S A Internet. 2006;103(30):11423–11428. doi: 10.1073/pnas.0601438103
  • Wang J-L, Li T-T, Elsheikha HM, et al. Functional Characterization of Rhoptry Kinome in the virulent Toxoplasma gondii RH strain. Front Microbiol Internet. 2017;8:1–8. doi: 10.3389/fmicb.2017.00084
  • Chen L, Christian DA, Kochanowsky JA, et al. The Toxoplasma gondii virulence factor ROP16 acts in cis and trans, and suppresses T cell responses. J Exp Med. 2020 [[cited 2020 Oct 14]];217(3): Internet. doi: 10.1084/jem.20181757
  • Butcher BA, Fox BA, Rommereim LM, et al. Toxoplasma gondii rhoptry kinase ROP16 activates STAT3 and STAT6 resulting in cytokine inhibition and arginase-1-dependent growth control. Hunter CA, editor. PLoS Pathog [Internet] Available from. 2011 [cited 2020 Oct 14];7(9):e1002236. doi: 10.1371/journal.ppat.1002236
  • Saeij JPJ, Coller S, Boyle JP, et al. Toxoplasma co-opts host gene expression by injection of a polymorphic kinase homologue. Nature. 2007;445(7125):324–327.
  • Fentress SJ, Steinfeldt T, Howard JC, et al. The arginine-rich N-terminal domain of ROP18 is necessary for vacuole targeting and virulence of Toxoplasma gondii. Cell Microbiol. 2012;14(12):1921–1933.
  • Sánchez V, De-la-Torre A, Gómez-Marín JE. Characterization of ROP18 alleles in human toxoplasmosis.Parasitol Int Internet. 2014 [cited 2015 Jan 17];63(2):463–469. doi: 10.1016/j.parint.2013.10.012
  • Behnke MS, Khan A, Wootton JC, et al. Virulence differences in Toxoplasma mediated by amplification of a family of polymorphic pseudokinases. Proc Natl Acad Sci U S A [Internet]. 2011 [cited 2023 Jul 26];108(23):9631–9636. doi: 10.1073/pnas.1015338108
  • Bayram Delibaş S, Turgay N, Gürüz AY The role of cytokines in the immunopathogenesis of toxoplasmosis. Turkiye Klinikleri J Med Sci. 2009;29:1217–1221.
  • de-la-Torre A, Pfaff AW, Grigg ME, et al. Ocular cytokinome is linked to clinical characteristics in ocular toxoplasmosis. Cytokine Internet. 2014;68(1):23–31. http://www.ncbi.nlm.nih.gov/pubmed/24787053
  • Cone RE, Pais R. Anterior Chamber-associated immune Deviation (ACAID): an acute response to ocular insult protects from Future immune-mediated damage? Internet. 2009 [cited 2023 Jul 25];1;OED.S2858. https://click.endnote.com/viewer?doi=10.4137%2Foed.s2858&token=WzEwNzI1NCwiMTAuNDEzNy9vZWQuczI4NTgiXQ.BMb28RbIx80X-cQ7TJdeDSmVCUE
  • Acosta Davila JA, Hernandez De Los Rios A, Acosta JAD, et al. An overview of peripheral blood mononuclear cells as a model for immunological Research of Toxoplasma gondii and other apicomplexan parasites. Front Cell Infect Microbiol Internet. 2019 [cited 2019 May 15];9:24. doi: 10.3389/fcimb.2019.00024
  • Costantini S, Castello G, Colonna G. Human cytokinome: a new challenge for systems biology.Bioinformation Internet. 2010 [cited 2016 Nov 14];5(4):166–167. doi: 10.6026/97320630005166
  • Frickel EM, Hunter CA. Lessons from toxoplasma: Host responses that mediate parasite control and the microbial effectors that subvert them. J Exp Med Internet. 2021 [cited 2022 Jan 23];218(11): doi: 10.1084/jem.20201314
  • Fisch D, Clough B, Frickel E-M. Human immunity to Toxoplasma gondii. Coers J, editor. PLoS Pathog [Internet]. 2019;15(12):e1008097. Available from: https://dx.plos.org/10.1371/journal.ppat.1008097
  • Ooi KGJ, Galatowicz G, Calder VL, et al. Cytokines and Chemokines in uveitis – is there a correlation with clinical phenotype? Clinical Medicine & Research Internet. 2006 [cited 2023 Jul 25];4(4):294–309. doi: 10.3121/cmr.4.4.294
  • Maia MM, Meira-Strejevitch CS, Pereira-Chioccola VL, et al. Evaluation of gene expression levels for cytokines in ocular toxoplasmosis. Parasite Immunol Internet. 2017;39(10):e12462. doi: 10.1111/pim.12462
  • Thieme C, Schlickeiser S, Metzner S, et al. Immune Mediator profile in aqueous humor differs in patients with primary acquired ocular toxoplasmosis and recurrent acute ocular toxoplasmosis. Mediators Inflamm Internet. 2019 [[cited 2023 Jul 25]];2019:1–12. doi: 10.1155/2019/9356728
  • Sauer A, Rochet E, Lahmar I, et al. The local immune response to intraocular Toxoplasma re-challenge: less pathology and better parasite control through Treg/Th1/Th2 induction. Int J Parasitol Internet. 2013;43(9):721–728. doi: 10.1016/j.ijpara.2013.04.004
  • Sauer A, Villard O, Creuzot-Garcher C, et al. Intraocular levels of interleukin 17A (IL-17A) and IL-10 as respective determinant markers of toxoplasmosis and viral uveitis. Papasian CJ, editor. Clin Vaccin Immunol [Internet]. 2015 [cited 2017 Mar 5];22(1):72–78. doi: 10.1128/CVI.00423-14
  • Sauer A, Pfaff AW, Villard O, et al. Interleukin 17A as an effective target for anti-inflammatory and antiparasitic treatment of toxoplasmic uveitis. J Infect Dis. 2012;206(8):1319–1329.
  • Kalogeropoulos D, Kalogeropoulos C, Sakkas H, et al. Pathophysiological aspects of ocular toxoplasmosis: host-parasite interactions. Ocul Immunol Inflamm [Internet]. 2022 [cited 2023 Jul 26];30(3):560–569. doi: 10.1080/09273948.2021.1922706
  • Naranjo-Galvis CA, de-la-Torre A, Mantilla-Muriel LE, et al. Genetic polymorphisms in cytokine genes in Colombian patients with ocular toxoplasmosis. Adams JH, editor. Infect Immun Internet. 2018;86(4): doi: 10.1128/IAI.00597-17
  • Cordeiro CA, Moreira PR, Costa GC, et al. Interleukin-1 gene polymorphisms and toxoplasmic retinochoroiditis. Mol VisInternet. 2008 [cited 2017 May 11];14:1845–1849. http://www.ncbi.nlm.nih.gov/pubmed/18941541
  • de Neves ES, Curi ALL, de Albuquerque MC, et al. Genetic polymorphism for IFNγ +874T/A in patients with acute toxoplasmosis. Rev Soc Bras Med Trop Internet. 2012 [cited 2017 May 11];45(6):757–760. doi: 10.1590/S0037-86822012000600020
  • de Albuquerque MC, Do Aleixo ALQ C, Benchimol EI, et al. The IFN-³+874T/A gene polymorphism is associated with retinochoroiditis toxoplasmosis susceptibility. Mem Inst Oswaldo Cruz Internet. 2009 [cited 2017 May 11];104(3):451–455. doi: 10.1590/S0074-02762009000300009
  • Cordeiro CA, Moreira PR, Andrade MS, et al. Interleukin-10 gene polymorphism (−1082G/A) is associated with Toxoplasmic Retinochoroiditis. Invest Ophthalmol Vis Sci Internet. 2008 [cited 2017 May 11];49(5):1979–1982. doi: 10.1167/iovs.07-1393
  • Torres-Morales E, Taborda L, Cardona N, et al. Th1 and Th2 immune response to P30 and ROP18 peptides in human toxoplasmosis. Med Microbiol Immunol [Internet]. 2014 [cited 2015 Jan 22];203(5):315–322. doi: 10.1007/s00430-014-0339-0
  • Maia MM, Meira-Strejevitch CS, Pereira-Chioccola VL, et al. Evaluation of gene expression levels for cytokines in ocular toxoplasmosis. Parasite Immunol Internet. 2017 [cited 2023 Jul 26];39(10):e12462. doi: 10.1111/pim.12462
  • Pfaff AW, de-la-Torre A, Rochet E, et al. New clinical and experimental insights into old world and neotropical ocular toxoplasmosis. Int J Parasitol [Internet]. 2014 [cited 2015 Jan 22];44(2):99–107. doi: 10.1016/j.ijpara.2013.09.007
  • Rudzinski M, Pardini L, Bernstein M, et al. Interferon-γ and il-10 release assay for patients with ocular toxoplasmosis. Am J Trop Med Hyg [Internet]. 2020 [cited 2023 Jul 26];103(6):2239–2243. doi: 10.4269/ajtmh.20-0124
  • Barbosa BF, Lopes-Maria JB, Gomes AO, et al. IL10, TGF Beta1, and IFN gamma modulate intracellular signaling pathways and cytokine production to control Toxoplasma gondii infection in BeWo trophoblast Cells1. Biol Reprod. 2015;92(3). doi: 10.1095/biolreprod.114.124115
  • Cai Y, Shen J. Modulation of host immune responses to Toxoplasma gondii by microRnas. Parasite Immunol Internet. 2017 [cited 2023 Jul 26];39(2):e12417. doi: 10.1111/pim.12417
  • Meira CS, Pereira-Chioccola VL, Vidal JE, et al. Cerebral and ocular toxoplasmosis related with IFN-γ, TNF-α, and IL-10 levels. Front Microbiol. 2014;5:105138. doi: 10.3389/fmicb.2014.00492
  • Mahmoudzadeh S, Nozad Charoudeh H, Marques CS, et al. The role of IL-12 in stimulating NK cells against Toxoplasma gondii infection: a mini-review. Parasitol Res. 2021;120(7):2303–2309.
  • Geiller B, Greigert V, Hillenbrand CA, et al. Type I and III interferons shape the retinal cytokine network and barrier function in an in vitro model of ocular toxoplasmosis. Front Immunol. 2023;14:1148037. doi: 10.3389/fimmu.2023.1148037
  • Roberts CW, Prasad S, Khaliq F, et al. Adaptive immunity and genetics of the host immune response. Toxoplasma Gondii [Internet]. Second Edi. Elsevier; 2014. p. 819–994. Available from: https://linkinghub.elsevier.com/retrieve/pii/B9780123964816000258
  • Jamieson SE, Cordell H, Petersen E, et al. Host genetic and epigenetic factors in toxoplasmosis. Mem Inst Oswaldo Cruz. 2009;104(2):162–169.
  • Wujcicka W, Gaj Z, Wilczyński J, et al. Contribution of IL6 −174 G>C and IL1B +3954 C>T polymorphisms to congenital infection with Toxoplasma gondii. Eur J Clin Microbiol Infect Dis. 2015;34(11):2287–2294.
  • Cordeiro CA, MMoreira PR, Costa GC, et al. Interleukin-1 gene polymorphisms and toxoplasmic retinochoroiditis. Mol VisInternet. 2008 [cited 2023 Jul 29];14:1845–1849. https://www.ncbi.nlm.nih.gov/pmc/articles/PMC2568892/pdf/mv-v14-1845.pdf
  • Araujo WMR, Ayo CM, Previato M, et al. Role of interleukin 1β and interleukin 10 variants on ocular toxoplasmosis in Brazilian individuals. Front Ophthalmol Internet. 2023;3. https://www.frontiersin.org/articles/10.3389/fopht.2023.1183167/full
  • Cordeiro CA, Moreira PR, Bessa TF, et al. Interleukin-6 gene polymorphism (−174 G/C) is associated with toxoplasmic retinochoroiditis. Acta Ophthalmol. 2013;91:e311–e314. doi: 10.1111/aos.12046
  • Abu EK, Boampong JN, Kyei S, et al. Associations between polymorphisms within interferon gamma and Tumor necrosis factor genes and Toxoplasma Retinochoroiditis in Ghanaian patients. Ocul Immunol Inflamm. 2017;25(5):683–689.
  • Cordeiro CA, Moreira PR, Costa GC, et al. TNF- gene polymorphism (-308G/A) and toxoplasmic retinochoroiditis. Br J Ophthalmol. 2008;92(7):986–988.
  • de Albuquerque MC, Do Aleixo AC, Benchimol EI, et al. The IFN-³+874T/A gene polymorphism is associated with retinochoroiditis toxoplasmosis susceptibility. Mem Inst Oswaldo Cruz. 2009;104(3):451–455.
  • Do Aleixo AC, Vasconcelos C, de Oliveira R, et al. Toxoplasmic retinochoroiditis: the influence of age, number of retinochoroidal lesions and genetic polymorphism for IFN-γ +874 T/A as risk factors for recurrence in a survival analysis. PLoS One. 2019;14(2):e0211627.
  • de S NE, Curi ALL, de AM, et al. Genetic polymorphism for IFNγ +874T/A in patients with acute toxoplasmosis. Rev Soc Bras Med Trop. 2012;45(6):757–760.
  • Peixe RG, Boechat MSB, Rangel ALP, et al. Single nucleotide polymorphisms in the interferon gamma gene are associated with distinct types of retinochoroidal scar lesions presumably caused by Toxoplasma gondii infection. Mem Inst Oswaldo Cruz. 2014;109(1):99–107.
  • de Faria Junior GM, Ayo CM, de Oliveira AP, et al. CCR5 chemokine receptor gene polymorphisms in ocular toxoplasmosis. Acta Trop. 2018;178:276–280. doi: 10.1016/j.actatropica.2017.12.012
  • Jamieson SE, Peixoto-Rangel AL, Hargrave AC, et al. Evidence for associations between the purinergic receptor P2X7 (P2RX7) and toxoplasmosis. Genes Immun. 2010;11(5):374–383.
  • Witola WH, Liu SR, Montpetit A, et al. ALOX12 in human toxoplasmosis. Infect Immun. 2014;82(7):2670–2679.
  • Peixoto-Rangel AL, Miller EN, Castellucci L, et al. Candidate gene analysis of ocular toxoplasmosis in Brazil: evidence for a role for toll-like receptor 9 (TLR9). Mem Inst Oswaldo Cruz. 2009;104(8):1187–1190.
  • Dutra MS, Bela SR, Peixoto-Rangel AL, et al. Association of a NOD2 gene polymorphism and T-Helper 17 cells with presumed ocular toxoplasmosis. J Infect Dis. 2013;207(1):152–163.
  • Witola WH, Mui E, Hargrave A, et al. NALP1 influences susceptibility to human congenital toxoplasmosis, proinflammatory cytokine response, and fate of Toxoplasma gondii -infected monocytic cells. Infect Immun. 2011;79(2):756–766.
  • Ayo CM, da Camargo AS, Frederico FB, et al. MHC class I Chain-related gene a polymorphisms and linkage disequilibrium with HLA-B and HLA-C alleles in ocular toxoplasmosis. PLoS One. 2015;10(12):e0144534.
  • Ayo CM, Frederico FB, Siqueira RC, et al. Ocular toxoplasmosis: susceptibility in respect to the genes encoding the KIR receptors and their HLA class I ligands. Sci Rep. 2016;6(1):36632.
  • de Perce-da-Silva DS, Joaquim TE, Do Aleixo AC, et al. Influence of killer immunoglobulin-like receptors genes on the recurrence rate of ocular toxoplasmosis in Brazil. Mem Inst Oswaldo Cruz Internet. 2023;118 [[cited 2023 Jul 29]]. http://www.scielo.br/scielo.php?script=sci_arttext&pid=S0074-02762023000101103&tlng=en
  • Felix JPF, Lira RPC, Zacchia RS, et al. Trimethoprim-sulfamethoxazole versus placebo to reduce the risk of recurrences of Toxoplasma gondii retinochoroiditis: randomized controlled clinical trial. Am J Ophthalmol Internet. 2014;157(4):762–766.e1. doi: 10.1016/j.ajo.2013.12.022
  • Fernandes Felix JP, Cavalcanti Lira RP, Cosimo AB, et al. Trimethoprim-sulfamethoxazole versus placebo in reducing the risk of toxoplasmic retinochoroiditis recurrences: a three-year follow-up. Am J Ophthalmol Internet. 2016 [[cited 2019 Apr 26]];170:176–182. doi: 10.1016/j.ajo.2016.08.003
  • Amim LHLV, Pacheco AG, Fonseca-Costa J, et al. Role of IFN-γ +874 T/A single nucleotide polymorphism in the tuberculosis outcome among Brazilians subjects. Mol Biol Rep [Internet]. 2008 [cited 2023 Jul 28];35(4):563–566. doi:10.1007/s11033-007-9123-1
  • Sica A, Dorman L, Viggiano V, et al. Interaction of NF-κB and NFAT with the interferon-γ promoter. J Biol Chem. 1997;272(48):30412–30420.
  • Hernández-de-Los-Ríos A, Murillo-Leon M, Mantilla-Muriel LELE, et al. Influence of two major Toxoplasma gondii virulence factors (ROP16 and ROP18) on the immune response of peripheral blood mononuclear cells to human toxoplasmosis infection. Front Cell Infect Microbiol Internet. 2019;9:413. doi: 10.3389/fcimb.2019.00413
  • El-Bendary M, Neamatallah M, Elalfy H, et al. Association of interferon gamma gene polymorphism and susceptibility to hepatitis C virus infection in Egyptian patients: A multicenter, family‐based study. JGH Open [Internet]. 2017 [cited 2023 Jul 28];1:140 4 10.1002/jgh3.12024
  • Barrett S, Collins M, Kenny C, et al. Polymorphisms in tumour necrosis factor-α, transforming growth factor-β, interleukin-10, interleukin-6, interferon-γ, and outcome of hepatitis C virus infection. J Med Virol. 2003;71(2):212–218.
  • Silva JLA, Rezende-Oliveira K, da Silva MV, et al. IL-17-expressing CD4+ and CD8+ T lymphocytes in human toxoplasmosis. Mediators InflammInternet. 2014 [cited 2017 Mar 5];2014:573825. http://www.hindawi.com/journals/mi/2014/573825/
  • Fatoohi F, Cozon GJN, Wallon M, et al. Systemic T cell response to Toxoplasma gondii antigen in patients with ocular toxoplasmosis. Jpn J Ophthalmol Internet. 2006;50(2):103–110. doi: 10.1007/s10384-005-0295-8
  • Feron EJ, Klaren VN, Wierenga EA, et al. Characterization of Toxoplasma gondii-specific T cells recovered from vitreous fluid of patients with ocular toxoplasmosis. Invest Ophthalmol Vis SciInternet. 2001 [cited 2018 Oct 26];42:3228–3232. https://iovs.arvojournals.org/article.aspx?articleid=2123203
  • Guan H, Moretto M, Bzik DJ, et al. NK cells enhance dendritic cell response against parasite antigens via NKG2D pathway. J Immunol. 2007;179(1):590–596.
  • Hunter CA. How are NK cell responses regulated during infection? Exp Parasitol. 1996;84(3):444–448. doi: 10.1006/expr.1996.0133
  • Sturge CR, Yarovinsky F, Andrews-Polymenis HL. Complex immune cell interplay in the gamma interferon response during Toxoplasma gondii infection.Infect Immun Internet. 2014 [cited 2023 Jul 26];82(8):3090–3097. doi: 10.1128/IAI.01722-14
  • Frenkel JK, Jacobs L. Ocular toxoplasmosis; pathogenesis, diagnosis and treatment. AMA Arch Ophthalmol. AMA Archives Of Ophthalmol Internet. 1958 [cited 2018 Jul 5];59(2):260–279. doi: 10.1001/archopht.1958.00940030128013
  • Wilder HC. Toxoplasma chorioretinitis in adults: a preliminary study of forty-one Cases diagnosed by microscopic examination.AMA Arch Ophthalmol [Internet]. 1952 [[cited 2018 Jul 4]];47(4):425. doi: 10.1001/archopht.1952.01700030435003
  • Mochizuki M, Sugita S, Kamoi K. Immunological homeostasis of the eye. 2012 [cited 2023 Jul 26]. doi: 10.1016/j.preteyeres.2012.10.002
  • García-López LL, Vargas-Montes M, Osorio-Méndez JF, et al. CD8+ T–cell exhaustion phenotype in human asymptomatic and ocular toxoplasmosis.Ocul Immunol Inflamm Internet. 2023 [cited 2023 Jul 11].1–10. doi: 10.1080/0927394820232217906
  • Cordeiro CA, Vieira ELM, Castro VM, et al. T cell immunoregulation in active ocular toxoplasmosis. Immunol Lett Internet. 2017 [[cited 2023 Jul 26]];184:84–91. doi: 10.1016/j.imlet.2017.02.009
  • Sauer A, Pfaff AW, Villard O, et al. Interleukin 17A as an effective target for anti-inflammatory and antiparasitic treatment of toxoplasmic uveitis. J Infect Dis Internet. 2012 [[cited 2023 Jul 26]];206:1319–1329. doi:10.1093/infdis/jis486
  • Kelly MN, Kolls JK, Happel K, et al. Interleukin-17/interleukin-17 receptor-mediated signaling is important for generation of an optimal polymorphonuclear response against Toxoplasma gondii infection. Infect Immun. 2005;73(1):617–621.
  • Gaddi PJ, Yap GS. Cytokine regulation of immunopathology in toxoplasmosis.Immunol Cell Biol Internet. 2007 [cited 2023 Jul 26];85(2):155–159. doi: 10.1038/sj.icb.7100038
  • Dutra MS, Bela SR, Peixoto-Rangel AL, et al. Association of a NOD2 gene polymorphism and Th17 lymphocytes with presumed ocular toxoplasmosis. J Infect Dis Internet. 2012;207(1):152–163. http://www.ncbi.nlm.nih.gov/pubmed/23100559
  • Ashander LM, Lie S, Ma Y, et al. Neutrophil activities in human ocular toxoplasmosis: an in vitro study with human cells. Invest Ophthalmol Vis Sci [Internet]. 2019 [cited 2023 Jul 26];60(14):4652–4660. doi: 10.1167/iovs.19-28306
  • de Araújo TE, dos Santos LI, Gomes AO, et al. Putative biomarkers for early diagnosis and prognosis of congenital ocular toxoplasmosis. Sci Rep Internet. 2020 [cited 2023 Jul 26];10(11):1–14. doi: 10.1038/s41598-020-73265-z
  • Seder RA, Ahmed R. Similarities and differences in CD4+ and CD8+ effector and memory T cell generation.Nat Immunol Internet. 2003 [cited 2023 Jul 28];4(9):835–842. doi: 10.1038/ni969
  • Wherry EJ. T cell exhaustion. Nat Immunol. 2011;12(6):492–499. doi: 10.1038/ni.2035
  • Trautmann L, Janbazian L, Chomont N, et al. Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nature Med 12:10Internet. 2006 [cited 2021 Aug 31];12(10):1198–1202. doi: 10.1038/nm1482
  • de Oliveira BC, da Silva AA, de Andrade Cavalcante MK, et al. Central and effector memory human CD4+ and CD8+ T cells during Cutaneous leishmaniasis and after in vitro stimulation with leishmania (viannia) braziliensis epitopes. Vaccines (Basel) Internet. 2023 [[cited 2023 Jul 11]];11(1): doi: 10.3390/vaccines11010158
  • Cifuentes-González C, Rojas-Carabali W, Pérez ÁO, et al. Risk factors for recurrences and visual impairment in patients with ocular toxoplasmosis: a systematic review and meta-analysis. PLoS One [Internet]. 2023 [cited 2023 Jul 28];18. 4 e0283845. doi: 10.1371/journal.pone.0283845
  • De-la-Torre A, Rios-Cadavid ACC, Cardozo-García CMM, et al. Frequency and factors associated with recurrences of ocular toxoplasmosis in a referral centre in Colombia. Br J Ophthalmol Internet. 2009;93(8):1001–1004. doi: 10.1136/bjo.2008.155861
  • Lin H-Y, Lee W-J. The role of corticosteroids in treating acute ocular toxoplasmosis in an immunocompetent patient: a case report. Front Med Internet. 2022;9. doi: 10.3389/fmed.2022.843050
  • Oray M, Ozdal PC, Cebeci Z, et al. Fulminant ocular toxoplasmosis: the hazards of corticosteroid monotherapy. Ocul Immunol Inflamm. 2016;24(6):637–646.
  • Garweg JG, Scherrer J, Wallon M, et al. Reactivation of ocular toxoplasmosis during pregnancy. BJOG [Internet]. 2005 [cited 2023 Jul 28];112(2):241–242. doi: 10.1111/j.1471-0528.2004.00302.x
  • Inchauspe S, Palacio A, Arriazu G, et al. Association between ocular Trauma and activation of ocular toxoplasmosis. Ocul Immunol Inflamm Internet. 2023 [cited 2023 Jul 28]; 1–5. https://click.endnote.com/viewer?doi=10.1080%2F09273948.2023.2203215&token=WzEwNzI1NCwiMTAuMTA4MC8wOTI3Mzk0OC4yMDIzLjIyMDMyMTUiXQ.q0vjbL8ahzWFPviXMj69q1rIV6Q
  • Lee KA, Flores RR, Jang IH, et al. Immune Senescence, Immunosenescence and Aging. Frontiers In Aging Internet. 2022 [cited 2023 Jul 28]; 3. doi: 10.3389/fragi.2022.900028
  • Velasco-Velásquez S, Celis-Giraldo D, Botero Hincapié A, et al. Clinical, socio-economic and environmental factors related with recurrences in ocular toxoplasmosis in Quindío, Colombia. Ophthalmic Epidemiol Internet. 2020;28(3):258–264. https://www.tandfonline.com/doi/full/10.1080/09286586.2020.1839509
  • Jensen KDC, Camejo A, Melo MB, et al. Toxoplasma gondii superinfection and virulence during secondary infection correlate with the exact ROP5/ROP18 allelic combination. Weiss LM, editor. MBio [Internet]. 2015;6(2): doi: 10.1128/mBio.02280-14
  • Holland GN, Crespi CM, ten Dam-van Loon N, et al. Analysis of recurrence patterns associated with Toxoplasmic Retinochoroiditis. Am J Ophthalmol Internet. 2008;145(6):1007–1013.e1. doi: 10.1016/j.ajo.2008.01.023
  • Reich M, Ruppenstein M, Becker MD, et al. Time patterns of recurrences and factors predisposing for a higher risk of recurrence of ocular toxoplasmosis. Retina [Internet]. 2015 [cited 2023 Sep 24];35(4):809–819. doi: 10.1097/IAE.0000000000000361
  • Garweg JG, Pleyer U. Treatment strategy in human ocular toxoplasmosis: why antibiotics have failed. J Clin Med Internet. 2021 [[cited 2023 Sep 24]];10(5):1–20. doi: 10.3390/jcm10051090
  • Fentress SJ, Behnke MS, Dunay IR, et al. Phosphorylation of immunity-related GTPases by a Toxoplasma gondii-secreted kinase promotes macrophage survival and virulence. Cell Host Microbe [Internet]. 2010 [cited 2020 Oct 14];8(6):484–495. doi: 10.1016/j.chom.2010.11.005
  • BLADER IJ, SAEIJ JP. Communication between Toxoplasma gondii and its host: impact on parasite growth, development, immune evasion, and virulence. APMIS Internet. 2009;117(5–6):458–476. doi: 10.1111/j.1600-0463.2009.02453.x
  • Reich M, Becker MD, Mackensen F. Influence of drug therapy on the risk of recurrence of ocular toxoplasmosis. Br J Ophthalmol bjophthalmol-2015. 2015;100(2):195–199. doi: 10.1136/bjophthalmol-2015-306650
  • Gómez Marín JE. Possibilities for immunomodulation in congenital toxoplasmosis. J Infect Dis Internet. 2016 [cited 2016 Sep 13];214(4):656.1–656. doi: 10.1093/infdis/jiw210
  • Gómez Marín JE, El Bissati K. Editorial: Innovative Therapeutic and Immunomodulatory Strategies for Protozoan Infections. Front Cell Infect Microbiol Internet. 2019;9. doi: 10.3389/fcimb.2019.00293
  • Streilein JW. Ocular immune privilege: the eye takes a dim but practical view of immunity and inflammation.Journal Of Leukocyte Biology Internet. 2003 [cited 2018 Oct 26];74(2):179–185. doi: 10.1189/jlb.1102574
  • Hu Z, Wu D, Lu J, et al. Inflammasome activation dampens type I IFN signaling to strengthen anti-Toxoplasma immunity. MBio. 2022 [[cited 2023 Jul 27]];13(6): Internet. doi: 10.1128/mbio.02361-22
  • Bretscher PA. On the mechanism determining the Th1/Th2 phenotype of an immune response, and its pertinence to strategies for the prevention, and treatment, of certain infectious diseases.Scand J Immunol Internet. 2014 [cited 2023 May 27];79(6):361–376. doi: 10.1111/sji.12175
  • McBride D, Kerr M, Dorn N, et al. Triggers, timescales, and treatments for cytokine-mediated tissue damage. EMJ Innovations Internet. 2020 [[cited 2023 Jul 27]]. https://click.endnote.com/viewer?doi=10.33590%2Femjinnov%2F20-00203&token=WzEwNzI1NCwiMTAuMzM1OTAvZW1qaW5ub3YvMjAtMDAyMDMiXQ.zIek2f43CMQ0WpRUMnWDoDoGHGc
  • Mease PJ, Van Der Heijde D, Ritchlin CT, et al. Ixekizumab, an interleukin-17A specific monoclonal antibody, for the treatment of biologic-naive patients with active psoriatic arthritis: results from the 24-week randomised, double-blind, placebocontrolled and active (adalimumab)-controlled period of the phase III trial SPIRIT-P1. Ann Rheum DisInternet. 2017 [cited 2023 Jul 27];76:79–87. https://click.endnote.com/viewer?doi=10.1136%2Fannrheumdis-2016-209709&token=WzEwNzI1NCwiMTAuMTEzNi9hbm5yaGV1bWRpcy0yMDE2LTIwOTcwOSJd.KrsXr2mzaaCvvXXLShfGu79Cchw
  • Lebwohl M, Strober B, Menter A, et al. Phase 3 Studies Comparing Brodalumab with Ustekinumab in Psoriasis. N Engl J Med [Internet]. 2015 [cited 2023 Jul 27];373:1318–1328. 14 10.1056/NEJMoa1503824
  • Baeten D, Sieper J, Braun J, et al. Secukinumab, an Interleukin-17A Inhibitor, in Ankylosing Spondylitis. N Engl J Med [Internet]. 2015 [cited 2023 Jul 27];373(26):2534–2548. doi: 10.1056/NEJMoa1505066
  • Pepple KL, Lin P. Targeting interleukin-23 in the treatment of noninfectious uveitis.Ophthalmol Internet. 2018 [cited 2023 Jul 27];125(12):1977. doi: 10.1016/j.ophtha.2018.05.014
  • Atienza-Mateo B, Calvo-Río V, Beltrán E, et al. Anti-interleukin 6 receptor tocilizumab in refractory uveitis associated with Behçet’s disease: multicentre retrospective study. Rheumatology [Internet]. 2018 [cited 2023 Jul 27];57:856–864. doi: 10.1093/rheumatology/kex480. 5
  • Syambani Ulhaq Z, Vita Soraya G, Retno Wulandari L. The role of IL-6-174 G/C polymorphism and intraocular IL-6 levels in the pathogenesis of ocular diseases: a systematic review and meta-analysis. Sci Rep. 2020 [cited 2023 Jul 27];10(1). doi: 10.1038/s41598-020-74203-9
  • Yang JY, Goldberg D, Sobrin L. Interleukin-6 and Macular Edema: a review of Outcomes with inhibition. Int J Mol Sci 2023, Vol 24, Page 4676Internet. 2023 [cited 2023 Jul 27];24(5):4676. doi: 10.3390/ijms24054676
  • Ciulla TA, Hussain RM, Taraborelli D, et al. Longer-Term Anti-VEGF Therapy Outcomes in Neovascular Age-Related Macular Degeneration, Diabetic Macular Edema, and Vein Occlusion-Related Macular Edema: Clinical Outcomes in 130 247 Eyes. Ophthalmol Retina. 2022;6(9):796–806.