79
Views
20
CrossRef citations to date
0
Altmetric
Review

Molecular markers of susceptibility to ocular toxoplasmosis, host and guest behaving badly

, , , , , , & show all
Pages 837-848 | Published online: 05 Dec 2008

Abstract

Infection with Toxoplasma gondii results in retinochoroiditis in 6% to 20% of immunocompetent individuals. The outcome of infection is the result of a set of interactions involving host genetic background, environmental, and social factors, and the genetic background of the parasite, all of which can be further modified by additional infections or even reinfection. Genes that encode several components of the immune system exhibit polymorphisms in their regulatory and coding regions that affect level and type of expression in response to stimuli, directing the immune response into different pathways. These variant alleles have been associated with susceptibility to immune-mediated diseases and with severity of pathology. We have investigated polymorphisms in several of these genes, identified as candidates for progression to retinochoroiditis caused by toxoplasmosis, namely chemokine (C-C motif) receptor 5 (CCR5), toll-like receptor-2 (TLR2), and TLR4. Furthermore, because interleukin-12 (IL-12) has been shown to be fundamental both in mice and in man to control a protective response against T. gondii, molecules that have a key function in IL-12 production will be emphasized in this review, in addition to discussing the importance of the genetic background of the parasite in the establishment of ocular disease.

Toxoplasma gondii, an obligate intracellular protozoan parasite, infects close to a billion people worldwide (CitationTenter et al 2000) and is an important agent of animal and human disease worldwide (CitationDubey and Beattie 1988; CitationWong and Remington 1993; CitationPetersen and Dubey 2001).

Toxoplasmosis in humans and other mammals is acquired by oral ingestion of either tissue cysts in raw or undercooked meat from chronically infected intermediate hosts or by ingestion of oocysts shed by cats by way of fecal contamination of food or water (CitationFrenkel 1988, Citation1990). Once ingested, the cyst wall is digested within the lumen of the small intestine. The acute phase of infection is characterized by wide-spread dissemination of rapidly dividing tachyzoites that invade virtually all cell types.

During the inflammatory process, soluble mediators and cellular components work together in a systematic manner in the attempt to contain and to eliminate the agents causing physical distress. The nature and portal of entry of the foreign substance and, to some degree, the nature and circumstances of a particular individual influence the way in which the inflammatory process is initiated.

Pathogens can initiate inflammation by a number of distinct and characteristic mechanisms, including activation of the plasma protease systems by interaction with degradation products of the pathogens and by secretion of toxins that can activate the inflammatory response directly. Injured cells themselves can release degradation products that initiate one or more of the plasma protease cascades and augment expression of proinflammatory cytokines that promote the inflammatory process. The physiological changes in inflammation are crucial to maintain the health and integrity of an organism, however, the inflammatory process can result in massive tissue destruction when poorly controlled.

During invasion of T. gondii, the host cell is essentially passive and no change is detected in membrane ruffling, actin cytoskeleton, or phosphorylation of host cell proteins (CitationFurtado et al 1992; CitationManger et al 1998; CitationOrtega-Barria and Boothroyd 1999; CitationJacquet et al 2001). The invasion is an active parasite-mediated process, initiated by contact between the apex of T. gondii and the host cell surface, involving many host receptors (proteoglycans like heparin and heparin-sulphate, β-integrins) and parasite ligands such as surface antigens (SAG), surface antigen-related sequences (SRS), microneme proteins (MIC), and laminin (CitationYap and Sher 1999).

Intestinal epithelial cells probably are the first cells infected by T. gondii. Chemokines released by these cells play a critical role in the initiation and modulation of immune response to various pathogens (CitationPerez de Lema et al 2001). They are responsible for the chemoattraction of polymorphonuclear neutrophils (PMNs), dendritic cells (DC), macrophages (MØ), and lymphocytes (CitationMackay 2001). T. gondii infects and can be disseminated by these cells to other organs throughout the host, especially to, muscle and central nervous system.

The disease in animal models

The T. gondii mouse infection model, because of its simplicity and robust responses it generates, has proven a powerful tool for studying host-microbe interactions. Because of that, most of the data published on immune responses and genes implicated in susceptibility derive from studies employing inbred mouse strains. Infection of mice leads to lifelong persistence of the parasite. Outcome is variable, depending on the interaction of many factors like inoculum size (CitationLiesenfeld 1999), virulence of the organism (CitationSu et al 2002), life cycle stage of the parasite (CitationJohnson 1984; CitationMcLeod et al 1989a; CitationSuzuki et al 1989a, Citation1993), genetic background (CitationWilliams et al 1978; CitationMcLeod et al 1989b; CitationSuzuki et al 1996; CitationLu et al 2005), gender (CitationRoberts et al 1995), and immunological status. All these factors seem to affect the course of infection in human beings and in animal models of toxoplasmosis.

Murine susceptibility to T. gondii or resistance to mortality following acute oral infection is under multigenic control by the host (CitationWilliams et al 1978; CitationMcLeod et al 1989a; CitationJohnson et al 2002). In C57BL/6 mice, genes both within and outside of the major histocompatibility complex (MHC) are involved in impaired intracerebral immune response (CitationBrown and McLeod 1990; CitationDeckert-Schluter et al 1994).

The ability of mice to survive T. gondii infection is dependent on strong T cell-mediated immunity. Both CD4+ T helper (Th) 1 cells and CD8+ cytolytic T lymphocytes are vital in providing protective immunity and long-term survival during chronic infection, as determined by in vivo depletion studies and adoptive transfer of CD4+ and CD8+ T cell lines and clones (CitationSuzuki et al 1988; CitationGazzinelli et al 1991, Citation1992; CitationParker et al 1991; CitationKasper et al 1992; CitationBuzoni-Gatel et al 1997). If the host becomes immunosuppressed, chronic infection can be reactivated, leading to toxoplasmic encephalitis, which is often fatal if not treated (CitationSuzuki 2002).

The protective competence of these cell types is due to their ability to produce interferon (IFN)-γ, a pro-inflammatory cytokine that has become well known as the major mediator of resistance to T. gondii (CitationSuzuki et al 1988). Mice that do not express IFN-γ (IFN-γ knockout mice) are unable to survive the acute phase of infection (CitationScharton-Kersten et al 1996). Antibody-mediated depletion of this cytokine during chronic infection demonstrates that continued IFN-γ production is necessary for long-term survival (CitationGazzinelli et al 1992). However, pathologic consequences due to overproduction of cytokines and excessive activation of the immune system are observed in IL-10 knockout (IL-10 KO) mice infected with T. gondii (CitationGazzinelli et al 1996) or wild type mice infected with highly virulent strains (CitationMordue et al 2001).

The activation of murine macrophages (MØ) by IFN-γ in the presence of additional signals, such as lipopolysaccharide (LPS) or tumor necrosis factor-α (TNF-α), is necessary to trigger the cytotoxic activity of MØ against T. gondii (CitationSibley et al 1993). IFN-γ exerts its antimicrobial activity through induction of specific effector molecules, including reactive nitrogen intermediates that interfere with key metabolic enzymes (CitationKhan et al 1997; CitationScharton-Kersten et al 1997; CitationRoberts et al 2000), indoleamine 2,3-dioxygenase (IDO) that induces tryptophan degradation thereby interfering with viability of the parasite (CitationMacKenzie et al 1999; CitationFujigaki et al 2002; CitationSilva et al 2002), and a family of GTP-binding proteins. IFN-γ-inducible GTP-binding proteins IGTP and LRP-47 are required for the mice to survive acute T. gondii infection, while nitric oxide-dependent protection may be more important during chronic infection (CitationTaylor et al 2000; CitationCollazo et al 2001).

The intraocular inflammatory response to the parasite is also mediated primarily by the CD4+ T- and CD8+ T-cell responses. CD8+ T cells recognize and destroy parasite-infected cells in an MHC class I restricted manner. However, as part of the local immune privilege, cells in the eye express low levels of MHC class I while infected mice usually show increased expression (CitationLyons et al 2001). This would, in turn, permit the precise targeting of T. gondii-infected cells by CD8+ cytolytic T lymphocytes and unlike other killing mechanisms that rely on soluble mediators, would limit collateral damage to uninfected cells. Indeed, depletion of the CD8+ cell subset resulted in increased lesion formation and cyst burden in an acquired disease model of infection (CitationGazzinelli et al 1994a), and a severe necrosis accompanied by higher ocular parasite burdens, in CD8-deficient mice immunized and challenged by a intraocular inoculum (CitationLu et al 2004). B cells may also contribute by limiting tachyzoite proliferation in the eyes (CitationLu et al 2004).

Programmed cell death is an important protective strategy used in immune-privileged organs. Fas is expressed on a variety of immune and nonimmune cells, whilst FasL is evident on T cells as well as in immune-privileged tissues. Ligation of these two surface molecules results in apoptosis of the Fas-expressing cell. In this way, the eye can prevent the damage caused by infiltrating lymphocytes and activated T cells can induce apoptosis in infected cells. T. gondii induces increased Fas and FasL expression on the cells of mouse retina. When intracamerally inoculated with tachyzoites of one virulent strain, Fas- and FasL-defective mice exhibit greater inflammatory scores and a more severe histopathology than similarly infected wild-type mice, added to a reduced ability to limit parasite proliferation early in infection (CitationHu et al 1999, Citation2001). However, when intraperitoneally inoculated with a less virulent strain, the same mice showed no difference in the degree of ocular inflammation and apoptosis. The higher level of IFN-γ and NO apparent early in these mice must have provided an alternative means to control parasitemia in the absence of Fas or FasL expression (CitationShen et al 2001).

In the immunosuppressed environment of the eye with high levels of TGF-β antigen-presenting cells can produce IL-10 when activated, which would stimulate the expansion of a regulatory T cell subset able to counteract the Th1 type pro-inflammatory responses and thus maintain the immune privilege. Nevertheless, the ocular pathogenesis in C57BL/6 mice is more severe than that of BALB/c and CBA/J mice and the serum levels of IFN-γ and TNF-α in C57BL/6 mice are significantly higher than those in BALB/c and CBA/J mice following ocular infection with a virulent strain of T. gondii (CitationLu et al 2005). There are higher levels of IFN-γ mRNA expression in the retinas of C57BL/6 mice than in those of BALB/c mice infected with T. gondii (CitationNorose et al 2003). Furthermore, a decline in IFN-γ production in CD4-KO mice protects mice from mortality due to an exacerbated immune response (CitationCasciotti et al 2002). IFN-γ has been shown to regulate the T. gondii load and inter-conversion between the bradyzoite and tachyzoite stages of T. gondii in the murine eye (CitationNorose et al 2003). Therefore, IFN-γ mediates immunopathology and contributes to early death following T. gondii infection (CitationLiesenfeld et al 1996).

Toxoplasmosis

Some recent studies support the idea that T. gondii is a master manipulator of host immune responses. The parasite simultaneously triggers the secretion of protective cytokines (IFN-γ and IL-12) and paradoxically suppresses the same type of response (CitationDenkers et al 2004; CitationGaddi and Yap 2007). This dual capacity of the parasite could actually be beneficial and allow the establishment of a stable host–parasite interaction. Failure to successfully synchronize these responses has negative consequences for both. Antimicrobial protection is ensured by the coordinated action of the innate and adaptative immune systems. Molecular communication between the host innate immune system and the intracellular protozoan T. gondii is fast emerging as a dramatic example of these key principles in action.

In the immunocompetent host the acute infection is usually asymptomatic or causes a influenza-like syndrome, and rarely leads to severe symptoms such as interstitial pneumonia, pericardial effusions, myositis, myocarditis, and neurological disorders (retinochoroiditis, Guillain-Barré syndrome, mental confusion) (CitationBossi and Bricaire 2004). Encephalitis is most commonly seen in immunocompromised individuals whereas uveitis is the most common lesion appearing in immunocompetent individuals (CitationHill and Dubey 2002). The infection incites a quasiefficient immune response, which controls parasitemia but allows tissue cysts to form in the brain, eyes, and muscles, leading to lifelong persistence of the parasite and variable medical outcome.

Dissemination of T. gondii in immunocompromised hosts, such as AIDS patients with very low CD4 counts, patients under immunosuppression to prevent or treat transplant rejection, and fetuses, is common, and often represents a reactivation of an earlier infection, rather than a newly acquired one. In these individuals the parasite can, besides encephalitis and retinochoroiditis, cause carditis, pneumonia, and meningitis, amongst other manifestations (CitationHill and Dubey 2002; CitationMele et al 2002; CitationMontoya and Liesenfeld 2004).

Ocular lesions may result from congenital or after birth-acquired infections. The lesion is often necrotic, destroying the architecture of the neural retina and sometimes involving the choroid (retinochoroiditis). Typical findings of toxoplasmic retinochoroiditis include white focal lesions with an intense overlaying and vitreal inflammatory reaction. Toxoplasmic retinochoroiditis is the most common lesion caused by infection with this protozoan and it may occur either immediately or long after the initial infection (CitationSilveira et al 1988; CitationCouvreur and Thulliez 1996; CitationMontoya and Remington 1996) or reactivation (CitationBosch-Driessen et al 2002).

Recurrent lesions are usually identified at the borders of the retinochoroidal scars, are typically found in clusters, and have been attributed to the rupture of tissue cysts within these old lesions or eventually coming from other tissues due to activation of memory immune cells. It is thought that the satellite lesions occurring both in the brain and the eye are caused by rupture of local cysts (CitationHolland 2003, Citation2004), but the origin of active infections in remote tissues has been credited to either clinically unapparent tissue cysts or to re-infection (CitationHill and Dubey 2002). In this last case, it is conceivable that the immune response by memory cells might be triggered by circulating parasites or even by other infections, through the activation of IFN-γ and other inflammatory mediators.

Many questions about the disease still remain that confound ophthalmologists (CitationVallochi et al 2002; CitationHolland 2003, Citation2004). The response to antibiotic therapy alone or in combination with corticosteroids varies widely among patients (CitationStanford et al 2003). Clinical presentation also varies, with some patients presenting only one episode of mild inflammation whereas others have multiple recurrences of severe uveitis leading to loss of eyesight. The immune response against retinal antigens does not explain the extensive damage to the eye in patients with ocular toxoplasmosis. Furthermore, we have presented data suggesting that a controlled autoimmune response may develop after infection with T. gondii in some patients and, in addition, may help to prevent the parasite from spreading through the retina thus associated with a milder disease (CitationVallochi et al 2005a).

Familial ocular toxoplasmosis is found in southern Brazil, where the incidence of ocular toxoplasmosis is higher than in most regions of the globe, and accounts for 70% to 90% of all uveitis cases seen in rural areas. A household survey in the endemic area showed 17,7% (186/1,042) of the population had toxoplasmic scars in the retina (CitationSilveira et al 1988; CitationGlasner et al 1992), a finding that cannot be explained by congenital infection alone (CitationHolland 2003, Citation2004). The same population was reassessed 7 and 11 years later and showed that seroconversion was more frequent in individuals under 17 years of age or above 50 years of age, with a risk of 10% of ocular involvement (CitationSilveira et al 2001; CitationSilveira 2002). Finally, acquired infection causing late necrotizing retinochoroiditis is seen not only in Brazil (CitationNussenblatt and Belfort 1994), but also in Europe (CitationBrezin and Cisneros 1999) and in the USA (CitationMontoya and Remington 1996).

Analysis of systemic cellular response to T. gondii antigen in 136 subjects with a diagnosis of ocular toxoplasmosis, no ocular disease or no infection by T. gondii suggested that resistance to the development of ocular toxoplasmosis has been associated with the ability to mount a lasting T-helper (Th) 1-type response (IL-2 and IFN-γ production) to the parasite antigens. On the other hand, susceptibility has also been associated with an inflammatory response, mostly mediated by cytokines (IL-1 and TNF-α) possibly produced by activated macrophages (MØ), which may themselves take part in the pathological features of the disease (CitationYamamoto et al 2000).

A recent study performed in France is in opposition to our study. Analyzing the induction of CD25 expression, no significant difference was observed in immune response among 16 individuals with a diagnosis of congenital, acquired, or undetermined ocular toxoplasmosis or between patients with active or inactive ocular lesions. Furthermore, higher levels of IFN-γ were detected in stimulated blood cultures from infected patients than in those from controls, with no difference between patients with asymptomatic or ocular toxoplasmosis (CitationFatoohi et al 2006).

TNF-α also plays an important role in murine resistance to acute and chronic toxoplasmosis (CitationChang et al 1990; CitationJohnson 1992; CitationLangermans et al 1992). However, high levels of this cytokine produced during any lethal infection can potentially contribute to harmful cerebral and hepatic effects (CitationBlack et al 1989; CitationBeaman et al 1992; CitationMarshall et al 1999). Furthermore, TNF-α may aid the intracerebral dissemination of T. gondii in mice (CitationGrau et al 1992) and may be increased in toxoplasmic retinochoroidal in humans (CitationYamamoto et al 2000).

We have shown that individuals with ocular toxoplasmosis from congenital infection have a diminished immune response towards T. gondii antigens when compared with individuals with ocular disease due to acquired infection (CitationYamamoto et al 2000). Interestingly, unlike mice, the diminished response is not caused by a superantigen (CitationVallochi et al 2001). This means that, despite the similarity at macroscopic and histopathologic level, the lesions seen on ocular toxoplasmosis may be generated by different mechanisms, therefore explaining the wide range of responses to available therapy.

Toxoplasma genotype and pathology

Variation of clinical presentation and severity of disease in susceptible individuals has been attributed to several factors, including host genetic heterogeneity and parasite genotype (CitationBoothroyd and Grigg 2002). We have typed T. gondii strains in Brazil (CitationVallochi et al 2005b) and our findings reveal that the highly clonal population structure of T. gondii seen in North America and Europe does not predominate in South America (CitationKhan et al 2006). Identification of the direct players in host–pathogen interactions during the initial infection and chronic phases of the disease is perhaps the best hope for better clinical treatment in this region.

Population studies analyzing more than 50 different genetic markers have identified a limited number of T. gondii genotypes in nature, despite the presence of a sexual phase in the life cycle, a wide geographical distribution and a broad range of intermediate hosts. Phylogenetic analysis detects 2 main groups in T. gondii population. From a practical point of view, 3 main lineages (type I, type II, and type III) that despite sharing about 98% genetic identity differ in virulence and epidemiological pattern of occurrence (CitationDarde 2004; CitationSwitaj et al 2005), are usually considered. For most groups, allelic polymorphism is low (2 to 4 alleles) (CitationDarde et al 1992; CitationHowe and Sibley 1995; CitationAjzenberg et al 2002), indicating that they descend recently from two closely-related parents that underwent a small number of genetic recombination events. Less than 1% of the previously studied strains contain unique genotypes and high divergence of DNA sequence, and have therefore been considered ‘exotic’ or ‘atypical’ strains (CitationSu et al 2003). However, it was demonstrated that sexual recombination combining polymorphisms in two of these distinct and competing clonal lines, can be a powerful force driving the natural evolution of virulence in this highly successful pathogen (CitationGrigg et al 2001).

The apparently low genetic diversity in T. gondii may have been underestimated because most parasite strains in previous studies were collected from human patients and domestic animals in North America and Europe. The choice largely employed by researchers is using the SAG2 locus for rapid typing since it is capable of distinguishing all three alleles from a single locus (CitationHowe et al 1997). This method is also widely used in other genes where the population structure has not been studied so extensively, therefore genetic diversity is under-represented. Several studies have examined the distribution of genotypes in chickens from countries such as Egypt, Argentina, India, and Brazil (CitationDubey et al 2002, Citation2003a, Citation2003b, Citation2003c; CitationSreekumar et al 2003; CitationLehmann et al 2004), but the genetic studies were limited by the use of only one marker (SAG2) (except [CitationSreekumar et al 2003; CitationLehmann et al 2004]), and because chickens in farms are indicators of strain prevalence in a domestic or peridomestic environment. Strains isolated from humans and animals (rabbit, mouse, goat, chickens, and dogs) in Minas Gerais, Brazil were analyzed by polymerase chain reaction-restriction fragment length polymorphism (PCR–RFLP) at eight independent loci and all strains carried recombinant genotypes, with typical types I, II, or III in almost all loci assessed (Citationde Melo Ferreira et al 2006).

We also have identified type I T. gondii strains by PCR-based typing at the SAG2 locus in retinochoroidal specimens from Brazil (CitationVallochi et al 2005b). When analyzed by multilocus PCR-RFLP, our recent findings reveal that the highly clonal population structure of T. gondii seen in North America and Europe does not predominate in South America (CitationKhan et al 2006). Consistent with this observation, T. gondii strains isolated from French Guiana patients are also highly diversified and not characteristic of a clonal organism as seen in North America strains (CitationAjzenberg et al 2004).

In the Brazilian strains, genotyping showed no cases of mixed infection (two alleles at a given locus). Unfortunately, the true extent of sequence divergence is not captured by multilocus RFLP and microsatellite analysis tends to overestimate genetic divergence. The UPRT-1 intron sequence was compared, and all except one strain from Brazil proved to have multiple additional polymorphisms not seen in the clonal lineages. This analysis suggested the presence of more than one predominant haplotype in Brazil along with less common unique genotypes (CitationKhan et al 2006).

It has been suggested previously that such divergent strains are more ancient in origin than the recently derived clonal lineages and that T. gondii expansion is linked to enhanced oral transmission between intermediate hosts, a trait not shared by other closely related parasites (CitationSu et al 2003). Despite of high serologic prevalence and level of recurrent ocular disease in Erechim, Brazil (CitationGlasner et al 1992), it is premature to say that sexual reproduction plays a central role in the population structure of T. gondii, since there were no cases of mixed infection. Given that the samples are from two small outbreaks and one of them due to food borne. It is likewise early to describe the epidemiology of toxoplasmosis in Brazil, since it is a country of continential size, boasting tropical and subtropical areas.

Recent studies in mice have identified some genes responsible for the strain-specific differences in virulence (CitationTaylor et al 2006; CitationSaeij et al 2007), where type I strains are more virulent than types II and III. As virulence is a multigenic trait, crossing the less virulent type II and type III strains generates some F1 progeny in which the virulence is greatly enhanced. The use of QTL mapping in such progeny identified a total of five virulence loci, including one on chromosome VIIa, which was found to correspond to ROP18, and one on chromosome VIIb, which corresponded to a gene encoding another rhoptry kinase, ROP16 (CitationSaeij et al 2007). ROP18 encodes the ROP18 protein, which is a functional serine kinase and is secreted into host cells (CitationTaylor et al 2006). ROP16 is a key modulator of the host response and the three clonal lineages differ in their level of activation of the STAT signaling pathway. As this is the pathway to the crucial cytokine interleukin 12, ROP16 is involved in this effect (CitationSaeij et al 2007). It seems that these eukaryotic pathogens can secrete protein kinases into host cells to subvert host-cell signaling pathways and that this explains many of the differences in virulence among the three dominant clonal lineages.

Our findings reveal that the genetic makeup of T. gondii is more complex than previously recognized and are in agreement that unique or divergent genotypes may contribute to different clinical outcomes of toxoplasmosis in different localities. Further strain comparisons based on a wider set of sequence based markers will be necessary to define the global population structure of T. gondii and to resolve the relationships between major strain types seen in different geographic regions.

Host genotype and the immune response to T. gondii

A wide array of studies has further demonstrated differences in genotype and allele frequencies of cytokine gene polymorphisms depending on ethnicity and race (CitationGolovleva et al 1997; CitationCox et al 2001; CitationUboldi de Capei et al 2003). Cytokines and chemokines and Toll-like receptors play a key role in the regulation of the type and magnitude of immune response, and the polymorphic nature of these genes may confer further flexibility to the immune response (CitationSchroder and Schumann 2005). We have analyzed some of the genes involved in the response against T. gondii infection, whose functions are detailed in the following pages.

Toll-like receptors

Toll-like receptors (TLRs) have been recently defined as important transmembrane proteins that can confer a certain degree of specificity to the cells of the innate immune compartment. TLRs, also referred to as pattern recognition receptors (PRRs), have been implicated in recognition of every known category of pathogen that causes human disease and whose signaling through the common adaptor molecule myeloid differentiation primary-response protein 88 (MyD88) is essential in proinflammatory cytokine responses to many microbial pathogens.

TLRs can recognize traces of microbial components and orchestrate an early defense, largely dependent on the activation of nuclear factor-kappaB (NF-κB), which will lead to the production of proinflammatory cytokines and triggering of microbiostatic/microbicidal effector mechanisms (CitationIwasaki and Medzhitov 2004). While major advances have been made in the assignment of individual TLRs to defined roles in bacterial infections (CitationTakeda et al 2003), such identification has only recently begun to emerge in protozoan parasites (CitationGazzinelli et al 2004).

Mice that are deficient in the TLR adaptor protein MyD88 were found to have a pronounced decrease in IL-12 production by peritoneal macrophages, neutrophils, and splenic DC in response to soluble tachyzoite antigen (STAg) stimulation in vivo and in vitro being highly susceptible to the acute infection with low doses of the infective stage of T. gondii (CitationScanga et al 2002). At this lower parasite dose, neither TLR2 nor TLR4 were important for mouse resistance to infection with T. gondii. In addition, DC from TLR2- or TLR4-KO mice developed normal IL-12 responses to STAg (CitationScanga et al 2002) just as PMNs from TLR2-KO animals did (CitationDel Rio et al 2004). TLR2-KO mice produce sufficient IL-12 to mediate partial protection that is nonetheless not enough to provide complete resistance to high dose infection, as compared to the wild type or TLR4-KO mice (CitationMun et al 2003). Hence, TLR2 might be involved in the early activation of the innate immune system during infection with T. gondii to be partially involved in the parasite recognition and MyD88 activation during early stages of infection with T. gondii. It seems that the defect seen in TLR2-KO mice is due to inefficient activation of microbicidal functions, with diminished nitric-oxide production by macrophages (CitationMun et al 2003).

Alternatively, MyD88-dependent neutrophil production of CCL2 (monocyte chemoattractant protein 1), but not IL-12, is dependent upon TLR2 signaling and defective CCL2 release could confer partial susceptibility to infection (CitationDel Rio et al 2004). Of note, neutrophil IL-12 production is not dependent upon parasite cyclophilin-18 (C-18), a T. gondii protein shown to trigger DC IL-12 through CCR5 ligation (CitationAliberti et al 2003). Thus, T. gondii possesses multiple molecules triggering distinct MyD88-dependent signaling cascades, and as these pathways are independently regulated, they may lead to distinct profiles of cytokine production (CitationDel Rio et al 2004).

MyD88 signaling occurs also through IL-1R or IL-18R, but ICE-KO mice (lacking functional IL-1 and IL-18) exhibit no increased susceptibility to infection suggesting that signaling through IL-1R and IL-18R is not essential for responses against T. gondii infection. Importantly, TLR1-, TLR2-, TLR4-, TLR6-, and TLR9-KO mice exhibited similar resistance to infection compared with wild-type mice suggesting that these receptors are not independently essential for control of T. gondii infection (CitationHitziger et al 2005).

A recent study from our laboratory indicates that there is no association between polymorphisms in TLR2 (Arg677Trp and Arg763Gln) and TLR4 (Asp299Gly) and ocular toxoplasmosis in Brazilian patients (unpublished observations), a finding in keeping with the observations in the murine model.

On the other hand, of the whole family of TLRs, two distinct TLR have been implicated in the recognition of protein ligands. TLR5 has been shown to be triggered by bacterial flagellin while TLR11 signaling is stimulated by protease sensitive molecules in uropathogenic bacteria (CitationHayashi et al 2001; CitationZhang et al 2004). TLR11 was implicated in resistance to T. gondii when an additional ligand in the parasite, T. gondii profilin (PTG), that apparently triggers high DC IL-12 production by a MyD88 dependent but CCR5 independent pathway, was discovered (CitationYarovinsky et al 2005). Interestingly, while DC IL-12 production appeared to be almost totally impaired in TLR11-KO mice infected with T. gondii, these animals, unlike either MyD88 or IL-12-KO mice (CitationReis e Sousa et al 1997) retained partial resistance to challenge and survived the acute phase of the infection, most likely due to a residual IFN-γ response (CitationYarovinsky et al 2005). It is important to note however, that human TLR11 is nonfunctional because of the presence of a stop codon in the gene (CitationZhang et al 2004). At present it is not clear whether TLR11 recognition of T. gondii is of importance in limiting infection in other mammalian species or whether humans utilize alternative pattern recognition receptors in the innate response to T. gondii.

Interleukin-12

Immunity to T. gondii involves acute inflammatory responses and antigen-specific adaptive immunity (CitationDenkers and Gazzinelli 1998; CitationCai et al 2000). Central to host resistance is the generation of IFN-γ by innate-type natural killer (NK) cells and adaptive CD4 and CD8 T lymphocytes (CitationSuzuki et al 1988, Citation1989b). IL-12 has been identified as critical for driving prompt IFN-γ production and proper differentiation of Th1 lymphocytes during the immune response to T. gondii (CitationGazzinelli et al 1994b; CitationYap et al 2000).

IL-12 is produced by PMNs, DC, B cells and other antigen-presenting cells (APCs). PMNs store cytokines in pre-formed pools namely IL-12, IL-6, MIP-2 in mouse neutrophils in the absence of infection (CitationTerebuh et al 1992; CitationBliss et al 2000; CitationMatzer et al 2001), and IL-4 in PMN from healthy human donors (CitationBrandt et al 2000). T. gondii induces secretion of IL-12 directly and rapidly by both human and mouse neutrophils, DCs as well as MØ (CitationGazzinelli et al 1993; CitationReis e Sousa et al 1997; CitationBliss et al 1999a, Citation1999b; CitationDenkers et al 2003; CitationRobben et al 2004). Release of IL-12 does not require IFN-γ priming since responses are intact in gene-targeted mice lacking this cytokine. On the other hand, recombinant TNF-α alone stimulates CCL2 and CCL4 production by PMN, and therefore, the ability of T. gondii to trigger TNF-α may provide an important positive feedback loop for release of neutrophil CCL2 and CCL4, important chemokines which recruit macrophages and other cells to infected tissue. T. gondii also induces up regulation of RANTES (CCL5), MIP-3α (CCL20) and macrophage chemoattractant protein-1 (MCP-1; CCL2) gene transcription by PMN (CitationBennouna et al 2003), which recruited to the site of infection by chemokines such as IL-8 and MCP-1, are themselves equipped to immediately release cytokines and chemokines that contribute further to cell recruitment and activation.

IL-12 is composed of two subunits, IL-12 p40 and IL-12 p35. In the absence of the IL-12 p40 gene or the IL-12-receptor-associated signal transducers Tyk2 and STAT-4, IFN-γ production is severely impaired resulting in enhanced susceptibility to acute T. gondii infection (CitationYap and Sher 1999; CitationCai et al 2000; CitationShaw et al 2003). In contrast, the IL-12-related cytokine, IL-23, which is composed of the shared IL-12 p40 subunit and a unique p19 subunit is not required or essential for mouse immunity to T. gondii infection (CitationLieberman et al 2004). Thus, IL-12 p40 production by DC and via a MyD88-dependent recognition mechanism is critical and perhaps sufficient for initiating IFN-γ-mediated immunity to T. gondii (CitationReis e Sousa et al 1997; CitationScanga et al 2002). Related cytokines such as IL-18 and IL-23 may instead play a pathogenic role (CitationMordue et al 2001; CitationGaddi and Yap 2007).

Studies of differential production of IL-12 in BALB/c and DBA/2 mice suggest that strain-specific variation not encoded by MHC genes affects IL-12 production with a skewing of antigen-specific recall responses to either a Th1 or a Th2 direction (CitationGieni et al 1996). In humans, deletions of IL-12 p40 or IL-12 receptors lead to serious impairment of immunity against intracellular bacteria (CitationAltare et al 1998; Citationde Jong et al 1998). A complete genomic sequence analysis of the IL-12 gene encoding its p40 subunit (IL-12B) identified several intronic polymorphisms, a single-nucleotide polymorphism (SNP) at position +16974 (+16974 A/C) in the 3′-untranslated region (UTR) of IL-12B (CitationHall et al 2000; CitationHuang et al 2000) and a promoter polymorphism (IL-12Bpro) (CitationMorahan et al 2002).

Association between genotype (+16974 A/C) and IL-12p40 production by stimulated PBMC was observed and depend on the stimuli used. There is a significantly decreased IL-12 p40 secretion for the following order of genotypes: AA > CA > CC, after stimulation of PBMC with C3-binding glycoprotein (C3bgp) in contrast to lipopolysaccharide, phytohaemagglutinin and pokeweed mitogen (CitationStanilova and Miteva 2005). Furthermore, polymorphisms in the IL12B 3′UTR gene have been show to influence the secretion of IL-12 and susceptibility to Type 1 diabetes (CitationWindsor et al 2004), and the promoter polymorphism may influence the outcome of malaria infection in at least one African population (CitationMorahan et al 2002).

CCR5

Although low-level IFN-γ production occurs in the absence of IL-12 signaling, optimal induction is strictly IL-12-dependent (CitationCai et al 2000). Therefore, much effort has been dedicated to disclose the mechanism(s) by which T. gondii tachyzoites elicit the production of IL-12 during the initial stages of infection.

The best defined mechanism by which T. gondii induces the production of IL-12 is through the parasite-derived cyclophilin-18, which signals murine DCs to produce IL-12 via the chemokine receptor CCR5 (CitationAliberti et al 2000; CitationAliberti et al 2003). The biological relevance of the unusual requirement for a chemokine receptor to participate in microbial recognition by DCs is supported by the observation that IL-12 production is decreased during acute infections of CCR5-KO animals (CitationAliberti et al 2000), although defects in cell migration could also contribute to this susceptible phenotype. In theory, cell-surface CCR5 on DCs could deliver signals derived from stimulation with autologous chemokines that have been induced following microbial stimulation or could be directly triggered by pathogen derived ligands.

In humans, a mutant allele of the CCR5 gene, bearing a 32-bp deletion, results in reduced CCR5 cell surface expression (CitationBlanpain et al 2000). However, we couldn’t observe association between the allele CCR5Δ32 and ocular toxoplasmosis in Brazilian patients (unpublished observations). Incidentally, we and others have found this variant to be quite rare in Brazilian population samples (unpublished results).

The IL-12 levels observed after stimulation of murine DCs with C-18 are much lower than those seen after stimulation with whole-parasite lysate or with a pool of tachyzoite-secreted proteins, indicating that pathways other than those initiated by CCR5 and C-18 might also be important for IL-12 production by DCs (CitationAliberti et al 2003).

Conclusion

While the data summarized in this review demonstrate that the binomial IL-12 and IFN-γ production is critical to resistance to T. gondii infection in mice, and that an intricate mechanism involving simultaneous activation of different signaling pathways could be required for induction and control of IL-12 during acute infection with T. gondii, much remains to be learned about the mechanisms involved and their relevance to parasite and host interactions during natural infection in humans. We had discussed the importance of the genetic background of both the parasite and the host in the establishment of ocular disease and the major questions concerning the specific receptors and parasite ligands required for innate immunity response triggering and how these interactions result in resistance to T. gondii.

Acknowledgments

Supported by Fundação de Amparo a Pesquisa do Estado de São Paulo (FAPESP), Conselho Nacional de Desenvolvimento Científi co e Tecnológico (CNPq), and the Pan-American Association of Ophthalmology (PAAO).

References

  • AjzenbergDBanulsALSuC2004Genetic diversity, clonality and sexuality in Toxoplasma gondiiInt J Parasitol3411859615380690
  • AjzenbergDCogneNParisL2002Genotype of 86 Toxoplasma gondii isolates associated with human congenital toxoplasmosis, and correlation with clinical findingsJ Infect Dis186684912195356
  • AlibertiJReis e SousaCSchitoM2000CCR5 provides a signal for microbial induced production of IL-12 by CD8 alpha+ dendritic cellsNat Immunol183710881180
  • AlibertiJValenzuelaJGCarruthersVB2003Molecular mimicry of a CCR5 binding-domain in the microbial activation of dendritic cellsNat Immunol44859012665855
  • AltareFDurandyALammasD1998Impairment of mycobacterial immunity in human interleukin-12 receptor deficiencyScience280143259603732
  • BeamanMHWongSYRemingtonJS1992Cytokines, Toxoplasma and intracellular parasitismImmunol Rev127971171506009
  • BennounaSBlissSKCurielTJ2003Cross-talk in the innate immune system: neutrophils instruct recruitment and activation of dendritic cells during microbial infectionJ Immunol1716052814634118
  • BlackCMIsraelskiDMSuzukiY1989Effect of recombinant tumour necrosis factor on acute infection in mice with Toxoplasma gondii or Trypanosoma cruziImmunology6857042514141
  • BlanpainCLeeBTackoenM2000Multiple nonfunctional alleles of CCR5 are frequent in various human populationsBlood9616384510961858
  • BlissSKButcherBADenkersEY2000Rapid recruitment of neutrophils containing prestored IL-12 during microbial infectionJ Immunol16545152111035091
  • BlissSKMarshallAJZhangY1999aHuman polymorphonuclear leukocytes produce IL-12, TNF-alpha, and the chemokines macrophage-inflammatory protein-1 alpha and -1 beta in response to Toxoplasma gondii antigensJ Immunol16273697510358188
  • BlissSKZhangYDenkersEY1999bMurine neutrophil stimulation by Toxoplasma gondii antigen drives high level production of IFN-gamma-independent IL-12J Immunol1632081810438947
  • BoothroydJCGriggME2002Population biology of Toxoplasma gondii and its relevance to human infection: do different strains cause different disease?Curr Opin Microbiol54384212160866
  • Bosch-DriessenLEBerendschotTTOngkosuwitoJV2002Ocular toxoplasmosis: clinical features and prognosis of 154 patientsOphthalmology1098697811986090
  • BossiPBricaireF2004Severe acute disseminated toxoplasmosisLancet36457915313352
  • BrandtEWoerlyGYounesAB2000IL-4 production by human polymorphonuclear neutrophilsJ Leukoc Biol681253010914499
  • BrezinAPCisnerosB1999Congenital and acquired infection in ocular toxoplasmosis [ARVO abstract]Invest Ophthalmol Vis Sci40S383
  • BrownCRMcLeodR1990Class I MHC genes and CD8+ T cells determine cyst number in Toxoplasma gondii infectionJ Immunol1453438412121825
  • Buzoni-GatelDLepageACDimier-PoissonIH1997Adoptive transfer of gut intraepithelial lymphocytes protects against murine infection with Toxoplasma gondiiJ Immunol158588399190941
  • CaiGRadzanowskiTVillegasEN2000Identification of STAT4-dependent and independent mechanisms of resistance to Toxoplasma gondiiJ Immunol16526192710946290
  • CasciottiLElyKHWilliamsME2002CD8(+)-T-cell immunity against Toxoplasma gondii can be induced but not maintained in mice lacking conventional CD4(+) T cellsInfect Immun704344311796568
  • ChangHRGrauGEPechereJC1990Role of TNF and IL-1 in infections with Toxoplasma gondiiImmunology693372107144
  • CollazoCMYapGSSempowskiGD2001Inactivation of LRG-47 and IRG-47 reveals a family of interferon gamma-inducible genes with essential, pathogen-specific roles in resistance to infectionJ Exp Med194181811457893
  • CouvreurJThulliezP1996[Acquired toxoplasmosis of ocular or neurologic site: 49 cases]Presse Med25438428685192
  • CoxEDHoffmannSCDiMercurioBS2001Cytokine polymorphic analyses indicate ethnic differences in the allelic distribution of interleukin-2 and interleukin-6Transplantation72720611544437
  • DardeML2004Genetic analysis of the diversity in Toxoplasma gondiiAnn Ist Super Sanita40576315269453
  • DardeMLBouteilleBPestre-AlexandreM1992Isoenzyme analysis of 35 Toxoplasma gondii isolates and the biological and epidemiological implicationsJ Parasitol78786941403418
  • de JongRAltareFHaagenIA1998Severe mycobacterial and Salmonella infections in interleukin-12 receptor-deficient patientsScience280143589603733
  • de Melo FerreiraAVitorRWGazzinelliRT2006Genetic analysis of natural recombinant Brazilian Toxoplasmagondii strains by multilocus PCR-RFLPInfect Genet Evol6223116376837
  • Deckert-SchluterMSchluterDSchmidtD1994Toxoplasma encephalitis in congenic B10 and BALB mice: impact of genetic factors on the immune responseInfect Immun6222188262631
  • Del RioLButcherBABennounaS2004Toxoplasma gondii triggers myeloid differentiation factor 88-dependent IL-12 and chemokine ligand 2 (monocyte chemoattractant protein 1) responses using distinct parasite molecules and host receptorsJ Immunol17269546015153515
  • DenkersEYButcherBADel RioL2004Manipulation of mitogen-activated protein kinase/nuclear factor-kappaB-signaling cascades during intracellular Toxoplasma gondii infectionImmunol Rev20119120515361242
  • DenkersEYDel RioLBennounaS2003Neutrophil production of IL-12 and other cytokines during microbial infectionChem Immunol Allergy839511412947981
  • DenkersEYGazzinelliRT1998Regulation and function of T-cell-mediated immunity during Toxoplasma gondii infectionClin Microbiol Rev11569889767056
  • DubeyJPBeattieCP1988Toxoplasmosis of animals and manBoca Raton, FLCRC Press
  • DubeyJPGrahamDHBlackstonCR2002Biological and genetic characterisation of Toxoplasma gondii isolates from chickens (Gallus domesticus) from Sao Paulo, Brazil: unexpected findingsInt J Parasitol329910511796127
  • DubeyJPGrahamDHda SilvaDS2003aToxoplasma gondii isolates of free-ranging chickens from Rio de Janeiro, Brazil: mouse mortality, genotype, and oocyst shedding by catsJ Parasitol89851314533703
  • DubeyJPGrahamDHDahlE2003bIsolation and molecular characterization of Toxoplasma gondii from chickens and ducks from EgyptVet Parasitol114899512781471
  • DubeyJPVenturiniMCVenturiniL2003cIsolation and genotyping of Toxoplasma gondii from free-ranging chickens from ArgentinaJ Parasitol891063414627159
  • FatoohiFCozonGJWallonM2006Systemic T cell response to Toxoplasma gondii antigen in patients with ocular toxoplasmosisJpn J Ophthalmol501031016604384
  • FrenkelJK1988Pathophysiology of toxoplasmosisParasitol Today4273815463000
  • FrenkelJK1990Toxoplasmosis in human beingsJ Am Vet Med Assoc19624082404922
  • FujigakiSSaitoKTakemuraM2002L-tryptophan-L-kynurenine pathway metabolism accelerated by Toxoplasma gondii infection is abolished in gamma interferon-gene-deficient mice: cross-regulation between inducible nitric oxide synthase and indoleamine-2,3-dioxygenaseInfect Immun707798611796611
  • FurtadoGCCaoYJoinerKA1992Laminin on Toxoplasma gondii mediates parasite binding to the beta 1 integrin receptor alpha 6 beta 1 on human foreskin fibroblasts and Chinese hamster ovary cellsInfect Immun604925311399003
  • GaddiPJYapGS2007Cytokine regulation of immunopathology in toxoplasmosisImmunol Cell Biol85155917228318
  • GazzinelliRXuYHienyS1992Simultaneous depletion of CD4+ and CD8+ T lymphocytes is required to reactivate chronic infection with Toxoplasma gondiiJ Immunol149175801351500
  • GazzinelliRTBrezinALiQ1994aToxoplasma gondii: acquired ocular toxoplasmosis in the murine model, protective role of TNF-alpha and IFN-gammaExp Parasitol78217298119376
  • GazzinelliRTHakimFTHienyS1991Synergistic role of CD4+ and CD8+ T lymphocytes in IFN-gamma production and protective immunity induced by an attenuated Toxoplasma gondii vaccineJ Immunol146286921670604
  • GazzinelliRTHienySWynnTA1993Interleukin 12 is required for the T-lymphocyte-independent induction of interferon gamma by an intracellular parasite and induces resistance in T-cell-deficient hostsProc Natl Acad Sci U S A906115198100999
  • GazzinelliRTRopertCCamposMA2004Role of the Toll/interleukin-1 receptor signaling pathway in host resistance and pathogenesis during infection with protozoan parasitesImmunol Rev20192515361229
  • GazzinelliRTWysockaMHayashiS1994bParasite-induced IL-12 stimulates early IFN-gamma synthesis and resistance during acute infection with Toxoplasma gondiiJ Immunol1532533437915739
  • GazzinelliRTWysockaMHienyS1996In 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-alphaJ Immunol1577988058752931
  • GieniRSFangYTrinchieriG1996Differential production of IL-12 in BALB/c and DBA/2 mice controls IL-4 versus IFN-gamma synthesis in primed CD4 lymphocytesInt Immunol81511208921430
  • GlasnerPDSilveiraCKruszon-MoranD1992An unusually high prevalence of ocular toxoplasmosis in southern BrazilAm J Ophthalmol114136441642287
  • GolovlevaISahaNBeckmanL1997Ethnic differences in interferon-alpha allele frequenciesHum Hered4718589239504
  • GrauGETacchini-CoolerFPiguetPF1992Is TNF beneficial or deleterious in toxoplasmic encephalitisParasitol Today83224 discussion 35–615463526
  • GriggMEBonnefoySHehlAB2001Success and virulence in Toxoplasma as the result of sexual recombination between two distinct ancestriesScience294161511588262
  • HallMAMcGlinnECoakleyG2000Genetic polymorphism of IL-12 p40 gene in immune-mediated diseaseGenes Immun12192411196715
  • HayashiFSmithKDOzinskyA2001The innate immune response to bacterial flagellin is mediated by Toll-like receptor 5Nature410109910311323673
  • HillDDubeyJP2002Toxoplasma gondii: transmission, diagnosis and preventionClin Microbiol Infect86344012390281
  • HitzigerNDellacasaIAlbigerB2005Dissemination of Toxoplasma gondii to immunoprivileged organs and role of Toll/interleukin-1 receptor signalling for host resistance assessed by in vivo bioluminescence imagingCell Microbiol78374815888086
  • HollandGN2003Ocular toxoplasmosis: a global reassessment. Part I: epidemiology and course of diseaseAm J Ophthalmol1369738814644206
  • HollandGN2004Ocular toxoplasmosis: a global reassessment. Part II: disease manifestations and managementAm J Ophthalmol13711714700638
  • HoweDKHonoreSDerouinF1997Determination of genotypes of Toxoplasma gondii strains isolated from patients with toxoplasmosisJ Clin Microbiol351411149163454
  • HoweDKSibleyLD1995Toxoplasma gondii comprises three clonal lineages: correlation of parasite genotype with human diseaseJ Infect Dis172156167594717
  • HuMSSchwartzmanJDYeamanGR1999Fas-FasL interaction involved in pathogenesis of ocular toxoplasmosis in miceInfect Immun67928359916110
  • HuSSchwartzmanJDKasperLH2001Apoptosis within mouse eye induced by Toxoplasma gondiiChin Med J (Engl)114640411780444
  • HuangDCancillaMRMorahanG2000Complete primary structure, chromosomal localisation, and definition of polymorphisms of the gene encoding the human interleukin-12 p40 subunitGenes Immun15152011197695
  • IwasakiAMedzhitovR2004Toll-like receptor control of the adaptive immune responsesNat Immunol59879515454922
  • JacquetACoulonLDe NeveJ2001The surface antigen SAG3 mediates the attachment of Toxoplasma gondii to cell-surface proteoglycansMol Biochem Parasitol116354411463464
  • JohnsonAM1984Strain-dependent, route of challenge-dependent, murine susceptibility to toxoplasmosisZ Parasitenkd7030396741220
  • JohnsonJSuzukiYMackD2002Genetic analysis of influences on survival following Toxoplasma gondii infectionInt J Parasitol321798511812495
  • JohnsonLL1992A protective role for endogenous tumor necrosis factor in Toxoplasma gondii infectionInfect Immun601979831563790
  • KasperLHKhanIAElyKH1992Antigen-specific (p30) mouse CD8+ T cells are cytotoxic against Toxoplasma gondii-infected peritoneal macrophagesJ Immunol148149381538132
  • KhanAJordanCMuccioliC2006Genetic divergence of Toxoplasma gondii strains associated with ocular toxoplasmosis, BrazilEmerg Infect Dis12942916707050
  • KhanIASchwartzmanJDMatsuuraT1997A dichotomous role for nitric oxide during acute Toxoplasma gondii infection in miceProc Natl Acad Sci U S A9413955609391134
  • LambertHHitzigerNDellacasaI2006Induction of dendritic cell migration upon Toxoplasma gondii infection potentiates parasite disseminationCell Microbiol816112316984416
  • LangermansJAVan der HulstMENibberingPH1992IFN-gamma-induced L-arginine-dependent toxoplasmastatic activity in murine peritoneal macrophages is mediated by endogenous tumor necrosis factor-alphaJ Immunol148568741729374
  • LehmannTGrahamDHDahlER2004Variation in the structure of Toxoplasma gondii and the roles of selfing, drift, and epistatic selection in maintaining linkage disequilibriaInfect Genet Evol40714
  • LiebermanLACardilloFOwyangAM2004IL-23 provides a limited mechanism of resistance to acute toxoplasmosis in the absence of IL-12J Immunol17318879315265921
  • LiesenfeldO1999Immune responses to Toxoplasma gondii in the gutImmunobiology2012293910631572
  • LiesenfeldOKosekJRemingtonJS1996Association of CD4+ T cell-dependent, interferon-gamma-mediated necrosis of the small intestine with genetic susceptibility of mice to peroral infection with Toxoplasma gondiiJ Exp Med1845976078760813
  • LuFHuangSHuMS2005Experimental ocular toxoplasmosis in genetically susceptible and resistant miceInfect Immun735160516041033
  • LuFHuangSKasperLH2004CD4+ T cells in the pathogenesis of murine ocular toxoplasmosisInfect Immun7249667215321988
  • LyonsREAnthonyJPFergusonDJ2001Immunological studies of chronic ocular toxoplasmosis: up-regulation of major histocompatibility complex class I and transforming growth factor beta and a protective role for interleukin-6Infect Immun6925899511254623
  • MackayCR2001Chemokines: immunology’s high impact factorsNat Immunol29510111175800
  • MacKenzieCRGonzalezRGKniepE1999Cytokine mediated regulation of interferon-gamma-induced IDO activationAdv Exp Med Biol467533910721097
  • MangerIDHehlABBoothroydJC1998The surface of Toxoplasma tachyzoites is dominated by a family of glycosylphosphatidylinositol-anchored antigens related to SAG1Infect Immun662237449573113
  • MarshallAJBrunetLRvan GesselY1999Toxoplasma gondii and Schistosoma mansoni synergize to promote hepatocyte dysfunction associated with high levels of plasma TNF-alpha and early death in C57BL/6 miceJ Immunol16320899710438948
  • MatzerSPBaumannTLukacsNW2001Constitutive expression of macrophage-inflammatory protein 2 (MIP-2) mRNA in bone marrow gives rise to peripheral neutrophils with preformed MIP-2 proteinJ Immunol16746354311591793
  • McLeodREisenhauerPMackD1989aImmune responses associated with early survival after peroral infection with Toxoplasma gondiiJ Immunol1423247552496163
  • McLeodRSkameneEBrownCR1989bGenetic regulation of early survival and cyst number after peroral Toxoplasma gondii infection of A × B/B × A recombinant inbred and B10 congenic miceJ Immunol143303142809214
  • MeleAPatersonPJPrenticeHG2002Toxoplasmosis in bone marrow transplantation: a report of two cases and systematic review of the literatureBone Marrow Transplant29691812180115
  • MontoyaJGLiesenfeldO2004ToxoplasmosisLancet36319657615194258
  • MontoyaJGRemingtonJS1996Toxoplasmic chorioretinitis in the setting of acute acquired toxoplasmosisClin Infect Dis23277828842263
  • MorahanGBoutlisCSHuangD2002A promoter polymorphism in the gene encoding interleukin-12 p40 (IL12B) is associated with mortality from cerebral malaria and with reduced nitric oxide productionGenes Immun34141812424623
  • MordueDGMonroyFLa ReginaM2001Acute toxoplasmosis leads to lethal overproduction of Th1 cytokinesJ Immunol16745748411591786
  • MunHSAosaiFNoroseK2003TLR2 as an essential molecule for protective immunity against Toxoplasma gondii infectionInt Immunol151081712917260
  • NoroseKMunHSAosaiF2003IFN-gamma-regulated Toxoplasma gondii distribution and load in the murine eyeInvest Ophthalmol Vis Sci4443758114507882
  • NussenblattRBBelfortRJr1994Ocular toxoplasmosis. An old disease revisitedJAMA27130478295291
  • Ortega-BarriaEBoothroydJC1999A Toxoplasma lectin-like activity specific for sulfated polysaccharides is involved in host cell infectionJ Biol Chem2741267769880495
  • ParkerSJRobertsCWAlexanderJ1991CD8+ T cells are the major lymphocyte subpopulation involved in the protective immune response to Toxoplasma gondii in miceClin Exp Immunol84207121902762
  • Perez de LemaGMaierHNietoE2001Chemokine expression precedes inflammatory cell infiltration and chemokine receptor and cytokine expression during the initiation of murine lupus nephritisJ Am Soc Nephrol1213698211423566
  • PetersenEDubeyJPJoynsonDHMWreghittTG2001Biology of toxoplasmosisToxoplasmosis: a comprehensive clinical guideCambridge, UKCambridge University Press142
  • Reis e SousaCHienySScharton-KerstenT1997In vivo microbial stimulation induces rapid CD40 ligand-independent production of interleukin 12 by dendritic cells and their redistribution to T cell areasJ Exp Med1861819299382881
  • RobbenPMMordueDGTruscottSM2004Production of IL-12 by macrophages infected with Toxoplasma gondii depends on the parasite genotypeJ Immunol17236869415004172
  • RobertsCWCruickshankSMAlexanderJ1995Sex-determined resistance to Toxoplasma gondii is associated with temporal differences in cytokine productionInfect Immun632549557790068
  • RobertsFRobertsCWFergusonDJ2000Inhibition of nitric oxide production exacerbates chronic ocular toxoplasmosisParasite Immunol221510607284
  • SaeijJPCollerSBoyleJP2007Toxoplasma co-opts host gene expression by injection of a polymorphic kinase homologueNature445324717183270
  • ScangaCAAlibertiJJankovicD2002Cutting edge: MyD88 is required for resistance to Toxoplasma gondii infection and regulates parasite-induced IL-12 production by dendritic cellsJ Immunol1685997600112055206
  • Scharton-KerstenTMWynnTADenkersEY1996In the absence of endogenous IFN-gamma, mice develop unimpaired IL-12 responses to Toxoplasma gondii while failing to control acute infectionJ Immunol1574045548892638
  • Scharton-KerstenTMYapGMagramJ1997Inducible nitric oxide is essential for host control of persistent but not acute infection with the intracellular pathogen Toxoplasma gondiiJ Exp Med1851261739104813
  • SchroderNWSchumannRR2005Single nucleotide polymorphisms of Toll-like receptors and susceptibility to infectious diseaseLancet Infect Dis51566415766650
  • ShawMHBoyartchukVWongS2003A natural mutation in the Tyk2 pseudokinase domain underlies altered susceptibility of B10. Q/J mice to infection and autoimmunityProc Natl Acad Sci U S A10011594914500783
  • ShenDFMattesonDMTuaillonN2001Involvement of apoptosis and interferon-gamma in murine toxoplasmosisInvest Ophthalmol Vis Sci422031611481268
  • SibleyLDAdamsLBKrahenbuhlJL1993Macrophage interactions in toxoplasmosisRes Immunol14438408451516
  • SilvaNMRodriguesCVSantoroMM2002Expression of indoleamine 2,3-dioxygenase, tryptophan degradation, and kynurenine formation during in vivo infection with Toxoplasma gondii: induction by endogenous gamma interferon and requirement of interferon regulatory factor 1Infect Immun708596811796621
  • SilveiraC2002Toxoplasmose: Dúvidas e controvérsiasErechim, RSEdiFAPES
  • SilveiraCBelfortRJrBurnierMJr1988Acquired toxoplasmic infection as the cause of toxoplasmic retinochoroiditis in familiesAm J Ophthalmol10636243262308
  • SilveiraCBelfortRJrMuccioliC2001A follow-up study of Toxoplasma gondii infection in southern BrazilAm J Ophthalmol131351411239868
  • SreekumarCGrahamDHDahlE2003Genotyping of Toxoplasma gondii isolates from chickens from IndiaVet Parasitol1181879414729166
  • StanfordMRSeeSEJonesLV2003Antibiotics for toxoplasmic retinochoroiditis: an evidence-based systematic reviewOphthalmology11092631 quiz 31–212750091
  • StanilovaSMitevaL2005Taq-I polymorphism in 3′UTR of the IL-12B and association with IL-12p40 production from human PBMCGenes Immun6364615858599
  • SuCEvansDColeRH2003Recent expansion of Toxoplasma through enhanced oral transmissionScience2994141612532022
  • SuCHoweDKDubeyJP2002Identification of quantitative trait loci controlling acute virulence in Toxoplasma gondiiProc Natl Acad Sci U S A9910753812149482
  • SuzukiY2002Immunopathogenesis of cerebral toxoplasmosisJ Infect Dis186Suppl 2S2344012424703
  • SuzukiYConleyFKRemingtonJS1989aDifferences in virulence and development of encephalitis during chronic infection vary with the strain of Toxoplasma gondiiJ Infect Dis15979042926171
  • SuzukiYConleyFKRemingtonJS1989bImportance of endogenous IFN-gamma for prevention of toxoplasmic encephalitis in miceJ Immunol1432045502506275
  • SuzukiYOrellanaMASchreiberRD1988Interferon-gamma: the major mediator of resistance against Toxoplasma gondiiScience240516183128869
  • SuzukiYOrellanaMAWongSY1993Susceptibility to chronic infection with Toxoplasma gondii does not correlate with susceptibility to acute infection in miceInfect Immun61228488500870
  • SuzukiYWongSYGrumetFC1996Evidence for genetic regulation of susceptibility to toxoplasmic encephalitis in AIDS patientsJ Infect Dis17326588537674
  • SwitajKMasterASkrzypczakM2005Recent trends in molecular diagnostics for Toxoplasma gondii infectionsClin Microbiol Infect11170615715713
  • TakedaKKaishoTAkiraS2003Toll-like receptorsAnnu Rev Immunol213357612524386
  • TaylorGACollazoCMYapGS2000Pathogen-specific loss of host resistance in mice lacking the IFN-gamma-inducible gene IGTPProc Natl Acad Sci U S A97751510639151
  • TaylorSBarraganASuC2006A secreted serine-threonine kinase determines virulence in the eukaryotic pathogen Toxoplasma gondiiScience31417768017170305
  • TenterAMHeckerothARWeissLM2000Toxoplasma gondii: from animals to humansInt J Parasitol3012175811113252
  • TerebuhPDOtternessIGStrieterRM1992Biologic and immunohistochemical analysis of interleukin-6 expression in vivo. Constitutive and induced expression in murine polymorphonuclear and mononuclear phagocytesAm J Pathol140649571372159
  • Uboldi de CapeiMUDamettoEFasanoME2003Genotyping for cytokine polymorphisms: allele frequencies in the Italian populationEur J Immunogenet3051012558814
  • VallochiALda Silva RiosLNakamuraMV2005aThe involvement of autoimmunity against retinal antigens in determining disease severity in toxoplasmosisJ Autoimmun24253215725573
  • VallochiALMuccioliCMartinsMC2005bThe genotype of Toxoplasma gondii strains causing ocular toxoplasmosis in humans in BrazilAm J Ophthalmol139350115734002
  • VallochiALNakamuraMVSchlesingerD2002Ocular toxoplasmosis: more than just what meets the eyeScand J Immunol55324811967112
  • VallochiALYamamotoJHSchlesingerD2001Lack of evidence for superantigen activity of Toxoplasma gondii towards human T cellsBraz J Med Biol Res3410233111471041
  • WilliamsDMGrumetFCRemingtonJS1978Genetic control of murine resistance to Toxoplasma gondiiInfect Immun1941620631881
  • WindsorLMorahanGHuangD2004Alleles of the IL12B 3′UTR associate with late onset of type 1 diabetesHum Immunol651432615603869
  • WongSYRemingtonJS1993Biology of Toxoplasma gondiiAIDS72993168471191
  • YamamotoJHVallochiALSilveiraC2000Discrimination between patients with acquired toxoplasmosis and congenital toxoplasmosis on the basis of the immune response to parasite antigensJ Infect Dis18120182210837184
  • YapGPesinMSherA2000Cutting edge: IL-12 is required for the maintenance of IFN-gamma production in T cells mediating chronic resistance to the intracellular pathogen, Toxoplasma gondiiJ Immunol1656283110878333
  • YapGSSherA1999Cell-mediated immunity to Toxoplasma gondii: initiation, regulation and effector functionImmunobiology201240710631573
  • YarovinskyFZhangDAndersenJF2005TLR11 activation of dendritic cells by a protozoan profilin-like proteinScience3081626915860593
  • ZhangDZhangGHaydenMS2004A toll-like receptor that prevents infection by uropathogenic bacteriaScience3031522615001781