Abstract
Parasitic protozoa are major threats to human health affecting millions of people around the world. Control of these infections by the host immune system relies on a myriad of immunological mechanisms that includes both humoral and cellular immunity. CD8+ T cells contribute to the control of these parasitic infections in both animals and humans. Here, we will focus on the CD8+ T cell response against a subset of these protozoa: Plasmodium, Toxoplasma gondii, Leishmania and Trypanosoma cruzi, with an emphasis on experimental rodent systems. It is evident a complex interaction occurs between CD8+ T cells and the invading protozoa. A detailed understanding of how CD8+ T cells mediate protection should provide the basis for the development of effective vaccines that prevent and control infections by these parasites.
Keywords: CD8+ T cell, immunity, Leishmania, malaria, Plasmodium, protozoa, Toxoplasma, Trypanosoma.
INTRODUCTION
The parasitic protozoa, Plasmodium spps., Toxoplasma gondii, Leishmania spps. and Trypanosoma cruzi are medically important pathogens around the world, causing significant disease in humans. Resistance and control of infections by these parasites relies on a competent immune system with multiple immunological mechanisms contributing. Plasmodium spps., Toxoplasma gondii, Leishmania spps. and Trypanosoma cruzi are intracellular parasites in both humans and mice. As a consequence of the intracellular infection these parasites are susceptible to immune mediated control by CD8+ T cells, which target intracellular pathogens. Indeed, there is strong evidence that CD8+ T cell responses are an important component of the host defense mechanism against these parasites. However, the degree of protection afforded by CD8+ T cells against these pathogens depends on the parasite, and may also differ among species of a given protozoa. Despite the induction of a robust immune response, these protozoa can delay or prevent immune clearance, thus, allowing the establishment of chronic infection in the host. Here we will review the contribution of CD8+ T cells during Plasmodium, T. gondii, Leishmania and T. cruzi infections. A more thorough understanding of CD8+ T cell responses against these intracellular parasites may be applicable to other intracellular parasites. Furthermore, there are no licensed vaccines that target these parasites through the induction of protective CD8+ T cells. Therefore, there is a need for continued research on understanding CD8+ T cell responses against these parasitic infections.
GENERATION OF PLASMODIUM-SPECIFIC CD8+ T CELL RESPONSES
Plasmodium infections in humans and rodents begin when an infected female Anopheles mosquito injects saliva containing Plasmodium sporozoites into the dermis during a blood meal. After deposition of sporozoites into the skin, the parasites enter host blood vessels where they travel to the liver and establish infection in hepatocytes. In the case of both humans and rodents, the initial liver stage of infection is relatively short lived [1, 2], resulting in little if any opportunity for the host to mount CD8+ T cell responses that are capable of eliminating infected hepatocytes during the initial infection. However, using the rodent model of malaria it has been shown that memory CD8+ T cells recognize parasite-infected hepatocytes upon re-exposure to the parasite, and are capable of preventing the parasite from progressing into the erythrocytic stage of infection [3, 4].
Following inoculation of sporozoites, priming of CD8+ T cells may occur at two different anatomical locations, skin draining lymph nodes (DLNs) and the liver [5-7]. It was long assumed that activation of sporozoite-specific CD8+ T cells occurred in the liver. This idea was challenged when Zavala and colleagues, demonstrated that lymph nodes draining the infection site play a fundamental role in priming liver stage-specific CD8+ T cells. The authors observed a marked decrease in the number of activated circumsporozoite protein (CSP)-specific CD8+ T cells in the liver of mice treated with FTY720, which blocks T cell egress from lymph nodes [8], prior to injection of sporozoites, or following the removal of the skin DLN at the site of sporozoite inoculation [7]. These results demonstrated the importance of lymph nodes in mounting CD8+ T cell responses against Plasmodium. Furthermore, CD8+ T cell priming in skin DLNs is sufficient for the induction of protective immunity against sporozoite challenge [6].
A small fraction of sporozoites at the site of inoculation have been shown to mature into infectious merozoites [9], however it has also been shown that these skin exo- erythrocytic infections are not capable of initiating blood stage infections [10]. Alternatively, sporozoites can leave the infection site by entering either the blood or lymphatic circulation [11, 12]. Approximately 15–20% of the inoculum ends up in the skin DLN [6, 12]. Those sporozoites that reach the DLN represent a critical portion of the inoculum that primes CD8+ T cells. This is supported by the observation that 48 hours after inoculation of sporozoites, by either bites of irradiated infected mosquitoes or via intradermal inoculation, CSP-specific CD8+ T cells producing IFN-γ were only detected in the skin DLN [7]. Although analysis of sporozoite-specific CD4+ T cells, which can contribute towards control of liver stage parasites [13, 14], has not been determined, it is likely they are also induced in the skin DLN.
Once sporozoites enter lymph nodes, dendritic cells (DCs) phagocytose the parasites, and then process and present parasite antigens via cross-presentation [5, 7, 15, 16]. CD11c+DCs play a key role in the activation of Plasmodium-specific CD8+ T cells [7, 16], as in vivo depletion of these cells abolished the induction of parasite-specific CD8+ T cells [16]. However, the specific DC population responsible for the induction of parasite-specific CD8+ T cells is not known. There are multiple subsets of DCs in the dermis (e.g. resident dermal CD103+ and CD11b+ DC subsets, Langer-hans cells or inflammatory monocyte-derived DCs) that are capable of cross-presenting viral antigens [17], and thus may be relevant in the activation of Plasmodium-specific CD8+ T cells. It is also possible that priming of parasite-specific CD8+ T cells requires a collaborative effort between skin migratory DCs and lymphoid-resident DCs [12, 18]. Thus, activation of Plasmodium-specific CD8+ T cells in skin DLNs might not rely on a single DC subset, but on the interaction of several DC populations.
As mentioned above, priming of Plasmodium-specific CD8+ T cells may also occur in the liver. Liver resident CD8α+CD11c+ DCs activate CD8+ T cells after immunization with irradiated sporozoites [5], and both liver sinusoidal endothelial cells and Kupffer cells are capable of processing and presenting antigens to naïve CD8+ T cells [19]. However, CD8+ T cells primed by liver antigen-presenting cells exhibit lower levels of activation (i.e., diminished expression of the activation markers CD44 and CD25) [19]. Finally, prolonged antigen presentation, following immunization with irradiated sporozoites, is also important in the optimal induction of sporozoite-specific CD8+ T cell responses [20]. Collectively, efficient generation of Plasmodium-specific effector CD8+ T cells, in rodent malaria, seems to be shaped by at least 3 factors (i) number of sporozoites inoculated into the host [21, 22], (ii) priming of CD8+ T cells in skin DLNs [7] and (iii) prolonged antigen presentation [20]. However, there are still many unknowns, including why natural infections and vaccines tested to date fail to induce protective liver stage-specific CD8+ T cell responses.
REGULATION OF PLASMODIUM-INDUCED CD8+ T CELL ACTIVATION
The precise mechanism by which naïve Plasmodium-specific CD8+ T cells become activated is not clear, but clues are emerging. For example, it has been shown that NK cells, probably via IL-12, are necessary for optimal priming as depletion of NK cells significantly reduced CD8+ T cell priming [23]. CD4+ T cells also participate in generating liver stage-specific CD8+ T cells. Zavala and colleagues showed that in the absence of CD4+ T cells, CD8+ T cell responses are impaired as a consequence of undergoing premature contraction [24, 25]. Following sporozoite infection CD4+ T cells were shown to secrete IL-4, a cytokine with strong in vivo and in vitro anti-apoptotic effects on activated and resting CD8+ T cells [26], which signals directly to parasite-specific CD8+ T cells to help maintain a memory CD8+ T cell population [24]. Of note, these studies were conducted in BALB/c mice, which favor production of IL-4 and consequently Th2 biased responses. Thus, it will be important to determine whether the contribution of IL-4 to expansion of sporozoite-specific CD8+ T cells is universal (e.g., is it also important in C57BL/6 mice which favor production of IFN-γ and Th1 biased responses?) or a consequence of using BALB/c mice. In contrast to these signals that favor robust liver stage-specific CD8+ T cells, there are also negative signals that function to dampen CD8+ T cell responses. For example, activated CD8+ T cells can negatively regulate the subsequent activation of additional naïve CD8+ T cells via competition for antigen on antigen-presenting cells [27, 28]. Furthermore, skin CD4+ regulatory T cells (Tregs) have been suggested to decrease expression of MHC class II and CD86 on skin DCs, which may impair activation of liver stage-specific CD8+ T cells [29]. Much of what we know about the precise mechanisms involved in the induction of liver stage-specific CD8+ T cells has been explored in the context of irradiated sporozoites. Thus, it is imperative we gain a better understanding of how liver stage-specific CD8+ T cells are generated following infection with live sporozoites via their natural route of infection and why Plasmodium-infected humans fail to induce protective CD8+ T cell responses in spite of repeated infections.
MECHANISMS OF CD8+ T CELL MEDIATED PROTECTION DURING THE LIVER STAGE
Once sporozoites enter the liver they glide along sinusoids where they ultimately invade and infect the liver parenchyma through an elegant process that has been reviewed elsewhere [30]. The liver stage of the Plasmodium life cycle is marked by an exponential expansion of parasite numbers and differentiation into merozoites that infect red blood cells when released from hepatocytes. It is estimated that one sporozoite can give rise to about 40,000 merozoites [31]. The liver stage of the life cycle is also relatively short, lasting about 2 days in mice [1], and about one-week in humans [2]. Thus, liver stage-specific CD8+ T cells must overcome substantial hurdles (i.e., the relatively few infected hepatocytes, the short duration of the liver stage, and the necessity to eliminate every infected hepatocyte) if they are to prevent the parasite from progressing from the asymptomatic liver stage to the symptomatic blood stage. Given the short duration of the liver stage, Plasmodium-specific CD8+ T cells primed during the initial exposure, which require one to two weeks for optimal expansion [32], likely contribute very little to liver stage immunity. However, liver stage-specific memory CD8+ T cells can play a critical role at controlling Plasmodium infected hepatocytes during a secondary infection [3, 33, 34].
Circulating memory CD8+ T cells can be broadly defined as either effector memory T cells (CD62Llo/CD27lo/IL-2lo) or central memory T cells (CD62Lhi/CD27hi/IL-2hi) [33]. Consistent with enhanced protection mediated by effector memory T cells following infection with Listeria monocytogenes and lymphocytic choriomeningitis virus [35, 36], effector memory T cells also provide increased protection against Plasmodium infected hepatocytes compared to central memory T cells [37-39]. Nevertheless, central memory CD8+ T cells correlate with sustained protection against malaria in mice [34], which is likely explained by the long-term stability of central memory CD8+ T cell numbers [40].
CD8+ T cells are endowed with multiple effector pathways, which include direct and indirect mechanisms, to eliminate target cells. In the case of liver stage-specific CD8+ T cells both are involved in controlling the parasite infection. Direct effector pathways used by Plasmodium liver stage-specific CD8+ T cells include the release of perforin and granzymes [27, 41], whereas indirect effector mechanisms include the production of IFN-γ and TNF [5, 42-44].
Among CD8+ T cell effector mechanisms, IFN-γ is important for controlling infected hepatocytes [5, 42-44]. The exact mechanism by which IFN-γ exerts its protective effect against Plasmodium is not fully known, but probably involves multiple mechanisms. IFN-γ causes increased expression of MHC class I, which enhances the recognition of antigens by memory CD8+ T cells [45]. Similarly, IFN-γ facilitates the conversion of the proteasome to the immune proteasome, which increases production of peptides that occupy MHC class I molecules [46, 47]. Another mechanism by which IFN-γ suppresses parasite development is through direct impairment of parasite differentiation in hepatocytes [48]. IFN-γ, from Plasmodium-specific CD8+ T cells, has also been suggested to increase expression of inducible nitric oxide synthetase, which results in increased production of nitric oxide that confers protection against Plasmodium [49], however the mechanism by which nitric oxide inhibits the development of liver stage parasites is not known.
Although production of IFN-γ may be the most critical mechanism by which CD8+ T cells eliminate infected hepatocytes, TNF also participates in Plasmodium control during the liver stage. For instance, in vitro administration of TNF prevents the development of human and rodent malaria pre-erythrocytic stages, but the mechanism of action of TNF against the parasite is unclear [4, 50]. However, an earlier study suggested TNF inhibits P. yoelii liver stages in vitro via synthesis of IL-6 [51], which shows anti-parasite activity potentially mediated by oxidative burst [52]. It was also shown that in vivo neutralization of TNF, via treatment with anti-TNF monoclonal antibodies, substantially reduced protection against either P. berghei or P. yoelii sporozoite challenge in a CD8+ T cell-dependent model [53].
There is also evidence to support a role for CD8+ T cells mediating pre-erythrocytic protection via direct cell contact. Following vaccination of mice with P. yoelii genetically attenuated parasites (GAPs) protection against subsequent P. yoelii sporozoite challenge is perforin-dependent [54]. The absence of perforin in memory CD8+ T cells also results in a 50% decrease in protection against P. yoelii sporozoite challenge in a prime-boost model that generates only P. yoelii CSP-specific CD8+ T cells [53]. However, the requirement for perforin in CD8+ T cell mediated protection in this model is species specific, as perforin-deficiency had no effect on protection against P. berghei sporozoite challenge [53]. Species-specific requirements for CD8+ T cell effector molecules was also noted in mice vaccinated with radiation-attenuated sporozoites (RAS) [13]. These data are also consistent with the observation that the numerical threshold of CSP-specific CD8+ T cells required for protection against sporozoite challenge is dependent on both Plasmodium species and the genetic background of the host [13, 39].
It is well established that Plasmodium-specific CD8+ T cells are able to prevent progression of Plasmodium infections from the pre-erythrocytic stage to the erythrocytic stage, however how such events occur needs to be elucidated. Cockburn and colleagues, provide useful clues using key developments in several technologies that allowed them to visualize Plasmodium-specific CD8+ T cells interacting with Plasmodium infected hepatocytes in mice in real time in vivo. Upon recognition of infected cells, Plasmodium-specific CD8+ T cells form large clusters around infected hepatocytes [55]. The formation of these cellular clusters may facilitate CD8+ T cells to eliminate parasites from the liver. The development of these technologies may contribute important findings as to how CD8+ T cells identify and eliminate infected hepatocytes.
APPROACHES TO GENERATE PROTECTIVE CD8+ T CELLS
Given the protective capacity of Plasmodium liver stage-specific CD8+ T cells in rodents substantial research has been directed towards developing CD8+ T cell based vaccines. Currently, there are four approaches to generate Plasmodium-specific CD8+ T cells, which can provide complete or partial protection against Plasmodium sporozoite challenge. These techniques include the use of RAS, GAP, wild-type sporozoites with chemoprophylaxis (CPS) and viral vectors (VV) that express Plasmodium antigens. Attenuation of Plasmodium by either radiation or targeted gene deletion results in viable sporozoites that infect hepatocytes and subsequently arrest within the liver without progressing into the blood stage. Consequently, the host is exposed to the full complement of sporozoite antigens. Likewise, vaccination with wild-type sporozoites with CPS not only exposes the host to all sporozoite antigens, but also to antigens expressed during the liver and blood stage. This allows the host to mount a diverse immune response including CD8+ T cells, CD4+ T cells, and antibodies. In contrast, VV induce an immune response to a small subset of parasite antigens. Of note, a diverse immune response including CD8+ and CD4+ T cells and antibodies directed against both pre-erythrocytic and erythrocytic antigens will likely be necessary in the development of an efficacious vaccine against Plasmodium.
Radiation-Attenuated Sporozoites
RAS have been used to induce sterilizing liver stage specific immunity not only in rodents [56] but also in humans [57]. RAS involve the application of radiation (gamma or X ray) to sporozoites, which leads to random DNA damage and impairs subsequent gene transcription [58]. Radiation-induced DNA damage does not alter the capacity of the parasite to infect hepatocytes, however the life cycle is arrested at early stages [59-61]. The level of protection by RAS is not influenced by the source of radiation, but the dose of radiation is critical [3, 59, 62]. Large doses of radiation may kill sporozoites and limit their ability to infect hepatocytes and induce protective immunity, while low radiation doses allow the parasite to complete the liver stage and progress into the blood stage. RAS induced protection is CD8+ T cell-dependent [13, 53] and correlates with effector memory CD8+ T cells [33]. Of note, use of RAS also induces sporozoite-specific antibodies that potentially contribute to protection, as well as CD4+ T cells that have been shown in some cases to provide protective immunity in mice [13, 63-65].
In spite of the technical hurdles associated with large-scale implementation of RAS vaccination, Hoffman and colleagues have made significant strides to make the RAS approach a practical vaccine procedure [66]. So far in fact, only the RAS approach [57] and a vaccine that uses immunogenic fragments of Plasmodium falciparum CSP known as the RTS,S vaccine [67, 68] have shown promise as human vaccines. RTS,S has reached large-scale phase III testing, however results have been disappointing and suggest the vaccine affords only limited protection (~16-40%) against severe disease while no protection against infection or mortality [69-71]. One hopes the RAS vaccination approach will prove to be more efficacious than RTS,S when fully tested.
Genetically Attenuated Parasites
Over the last decade advances in Plasmodium genetics have resulted in the generation of parasites lacking genes necessary for completion of the liver stage [61]. Subsequent infection with sporozoites from these GAPs are as immunogenic as RAS, with protective immunity dependent on CD8+ T cells [54]. However, like RAS vaccination it is possible CD4+ T cells and antibodies also contribute to GAP induced protective immunity.
The specific gene or combination of genes deleted determines the point at which the parasite arrests during liver stage development [72]. Currently, ten genes (P36p/P36, UIS3, UIS4, E1α, E3, SAP1/SLARP, FABI, FABB/F, FABZ, and PKG) have been deleted to manipulate the life cycle of the parasite [61]. Deletion of P36p results in normal sporozoite motility and hepatocyte invasion, but causes early arrest of the parasite in the liver due to impaired formation of the parasitophorus vacuole (PV) [73, 74]. When sporozoite and liver-stage asparagine-rich protein (SLARP) in P. berghei, or its P. yoelii ortholog sporozoite asparagine-rich protein 1 (SAP1) are deleted, sporozoites can still invade hepatocytes and form the PV, but they do not progress further in the liver stage [75, 76]. Deletion of UIS3 and UIS4 arrests the differentiation from trophozoite into schizonts [77, 78]. In contrast to P36p and SAP1, deletion of genes associated with the fatty acid metabolism (E1α, E3, FABI, FABB/F, FABZ, and PKG) exhibit normal development until the final differentiation and release of merozoites [79-81].
Vaccination with GAPs that arrest early in the liver stage can induce protective immunity, but GAPs that arrest later during the liver stage are more effective [82]. One explanation for this difference is the increased antigen repertoire the host is exposed to in late arresting GAPs compared to early arresting GAPs [61]. This was shown by Butler and colleagues, who demonstrated superior protective immunity in mice vaccinated with late arresting GAPs, compared to either early arresting GAPs or RAS, was the result of a larger and broader CD8+ T cell response in late arresting GAP vaccinated mice compared to early arresting GAP or RAS vaccinated mice [83]. This observation is consistent with prior reports that demonstrate the magnitude of Plasmodium specific CD8+ T cells is important in providing protection against sporozoite challenge [39, 84]. Furthermore, late arresting GAPs generate a host immune response that exhibits cross-stage specificity targeting both the liver and blood stages [83].
Wild Type Sporozoites with Chemoprophylaxis
The use of wild type sporozoites with CPS to stimulate the immune system is based on the administration of viable sporozoites in conjunction with anti-parasitic drugs to induce host immune responses while controlling the perpetuation of the parasite. So far several drugs have been used, which target the parasite at either the late liver or blood stages. Pyrimethamine, centanamycin and primaquine prevent nuclear division of liver schizont stages [85]. Azithromycin and clindamycin exert delayed action by directly inhibiting apicoplast maturation of liver schizont stages while chloroquine affects the blood stage of the parasite life cycle [86-88]. Consequently, chloroquine provides the latest arrest of the parasite life cycle of all the strategies mentioned above [86, 87], which increases the number of parasite antigens the host is exposed to. Furthermore, chloroquine mediated CPS and late arresting GAP immunization strategies may also afford enhanced protection as a consequence of increased parasite biomass.
Vaccination with viable sporozoites under CPS is very similar to either RAS or GAP vaccination. However, protective immunity in humans was induced following exposure to just 10 bites from infected mosquitoes [89] while protective immunity elicited by RAS in humans required the bites of >1000 infected mosquitoes [90, 91]. Whether this differential outcome is associated with exposure to blood stage antigens following sporozoite infection and CPS compared to RAS is not known, but these results clearly highlight the potency of this approach over RAS. Although vaccination of viable sporozoites under CPS induces CD4+ T cell and antibody responses, protection in humans correlates with liver stage-specific CD8+ T cells [42].
Subunit Vaccines
Viral vectors have been extensively evaluated as malaria vaccine candidates based on their ability to encode Plasmodium antigens and induce subsequent CD8+ T cell responses [92]. Examples of viral vector platforms for inducing Plasmodium-specific CD8+ T cell responses include replication-deficient adenoviruses (e.g., human, simian, and chimpanzee serotypes) and replication-deficient orthopoxviruses (e.g., modified vaccinia virus Ankara (MVA) and fowl pox 9 virus) [93, 94]. In addition, alphavirus, flavivirus, and morbillivirus may represent platforms to generate Plasmodium-specific CD8+ T cell responses [94]. In comparison with the other techniques used for induction of Plasmodium-specific CD8+ T cell responses, viral vectors overcome many manufacturing complications related with mosquito and/or sporozoite based formulations [95]. Another advantage of viral vectors is their ability to carry multiple transgenes and immune-stimulatory molecules, such as TLR agonists [96]. They also afford the ability to introduce cross-stage antigens to induce both liver and blood stage immune responses.
One of the main limitations of viral vectors is pre-existing immunity to the viral vector itself, which can dampen the host immune response to the transgene [97, 98]. Furthermore, as a consequence of the host immune system responding to the viral vector, it is essential that subsequent booster immunizations be done with different viral vectors engineered to carry the same Plasmodium transgene. Of the viral vectors evaluated as candidate malaria vaccines, poxviruses have been the most extensively studied in the clinic, however they have provided only modest protection against sporozoite challenge in humans [99, 100]. Consequently, chimpanzee adenoviruses have been targeted based on their ability to both prime robust CD8+ T cell responses and avoid pre-existing vector immunity [101].
ROLE OF CD8+ T CELLS DURING THE BLOOD STAGE OF PLASMODIUM
Experimental models have demonstrated CD8+ T cells are important in the immune response against liver stage parasites. In contrast, they contribute little to protective immunity during the blood stage [102, 103] and are potentially pathogenic [104-106]. The limited role of CD8+ T cells during the blood stage Plasmodium infection is explained by the lack of MHC class I on the surface of infected red blood cells [107]. Although blood stage-specific CD8+ T cells contribute little to protective immunity they are efficiently primed during infection in a process that involves cross-presentation mediated by CD8α+ DCs [108]. In contrast, it has been reported that IL-10 impairs the ability of DCs to fully prime CD8+ T cells during malaria, resulting in decreased proliferation and cytokine production [109]. Regardless of their induction, there is strong support for blood stage-specific CD8+ T cells contributing to Plasmodium-induced pathology during experimental cerebral malaria in mice [104, 110-113]. There appears to be at least two mechanisms by which this occurs. First, CD8+ T cells, through an unknown mechanism involving IFN-γ, contribute to parasite accumulation in the brain [114]. Second, following recognition of antigen, Plasmodium-specific CD8+ T cells release perforin and granzyme B, which leads to experimental cerebral malaria [112, 115]. Curiously, CD8+ T cells do not recognize parasite infected RBCs, thus it’s not clear what cells stimulate the CD8+ T cells to release perforin and granzyme B. One possibility is that vascular endothelial cells acquire antigen from infected RBCs during cytoadherance, which is then recognized by CD8+ T cells. Of note, it is unknown if CD8+ T cells contribute to cerebral malaria in humans.
CD8+ T CELL MEDIATED IMMUNITY AGAINST TOXOPLASMA GONDII
Toxoplasma gondii is the causative agent of toxoplasmosis, its an apicomplexan parasite that infects a wide range of vertebrates including humans [116, 117]. This parasite is transmitted between animals by ingestion of oocysts found in feline feces or tissue cysts in infected vertebrates [118]. Once in the intermediate host, the parasite undergoes asexual replication and disseminates throughout the body, including the brain, where it establishes intracellular infections and the formation of cysts [118]. Control of T. gondii requires the synergic interaction of multiple soluble (i.e. IL-12, IFN-γ) and cellular components (natural killer cells, DCs, macrophages, and CD4+ and CD8+ T cells) of the host immune system [119].
Depletion of CD8+ T cells, but not CD4+ T cells, using monoclonal antibodies accelerates the mortality of mice chronically infected with T. gondii [120-122], which provides support for the role of CD8+ T cells in controlling the parasite during the chronic phase of the infection. CD8+ T cell mediated control of T. gondii is dependent upon the production of IFN-γ [123, 124] and perforin [125-127]. One mechanism by which IFN-γ contributes to the control of T. gondii is by stimulating monocytes, macrophages and non-hematopoietic cells to produce nitric oxide [128, 129], however the precise mechanism by which these molecules afford protection is not known. Both IL-2 and CXCL10 also contribute to CD8+ T cell control of T. gondii. IL-2, which is produced by CD8+ T cells, increases IFN-γ production during the secondary response to T. gondii through an autocrine feedback loop [124], while CXCL10 was demonstrated to maintain effector CD8+ T cells in the brain and regulate migration speed towards T. gondii infected cells [130].
ROLE OF CD8+ T CELLS DURING LEISHMANIA INFECTION
Leishmaniasis is caused by various species of Leishmania (L. major, L. donovani, L. braziliensis, L. infantum, etc.), and is transmitted by 30 different sand fly species [131]. Clinical manifestations range from self-healing cutaneous lesions to deadly visceral disease. The contribution of CD8+ T cells in mediating protection against experimental cutaneous Leishmanias has been controversial. Early evidence suggested CD8+ T cells contributed to protective immunity [132]. However, subsequent studies identified CD4+ Th1 cells as the primary cells involved in controlling infection [133]. These contradictions were later resolved when it was shown that following low dose infection CD8+ T cell produced IFN-γ was necessary for the development of Th1-polarized CD4+ T cells, while after high dose infection CD8+ T cells were not required for the generation of a protective Th1 response [134]. In addition to their role in cutaneous Leishmaniasis, CD8+ T cells also provide protection against visceral Leishmaniasis. During visceral Leishmaniasis CD8+ T cells aid in the development of granulomas in the liver of infected mice [135], and the reduction of parasite burden in the spleen [136]. Moreover, CD8+ T cells have been shown to contribute to protective immunity during secondary Leishmania infections in mice [137]. The insights learned from experimental Leishmaniasis appear to hold up in humans, where CD8+ T cells also correlate with protective immunity (reviewed by Stagar and Rafati [138]).
ROLE OF CD8+ T CELLS DURING TRYPANOSOMA CRUZI INFECTION
T. cruzi is the causative parasite of Chagas disease, a zoonotic chronic inflammatory disease transmitted by haematophagous triatomine insects [139]. CD8+ T cells contribute to control of the parasite during acute and chronic stages of the disease [140-142]. A role for CD8+ T cells in the control of acute T. cruzi infection was shown through the use of β2-m-deficient mice, which lack MHC class I expression on the cell surface, as these mice succumb to acute infection [143]. Additionally, CD8+ T cells are required for control of chronic T. cruzi infection as depletion of CD8+ T cells during chronic infection resulted in exacerbation of inflammation within the heart, the site of chronic infection, and an increase in parasite burden [144]. Moreover, recent work has identified a number of CD8+ T cell epitopes within the T. cruzi genome, including an immune dominant epitope located in the trans-sialidase gene [145-148]. Identification of these epitopes may facilitate additional studies to evaluate the contribution of CD8+ T cells to protective immunity against T. cruzi and may also guide sub-unit based vaccines against this parasitic infection.
In spite of the induction of T. cruzi-specific CD8+ T cells there are several notable abnormalities associated with this response, which may contribute to impaired clearance of the parasite and progression to a chronic infection. For example, expansion of CD8+ T cells is delayed and remains relatively low in numbers during the first week of infection [148-150], which likely contributes to dissemination and an increase in the parasite burden throughout the host. Effector and memory CD8+ T cells accumulate at the site of infection [151], however effector functions of CD8+ T cells are attenuated and the CD8+ T cells eventually become exhausted [151-154]. Given the impact of T. cruzi on human health there is still much to be learned about the host immune response, including CD8+ T cells, to this parasite.
CONCLUSION
CD8+ T cells contribute to protective immunity against multiple intracellular parasitic infections. In recent years we have learned a great deal about how CD8+ T cells are primed, expand into effector and memory populations, and contribute to protective immunity against Plasmodium spps. and T. gondii. However, the contribution of CD8+ T cells in host immune responses to Leishmania and T. cruzi are not as well defined. A greater understanding of the requirements for CD8+ T cells to mediate protective immunity against these parasitic infections, especially in humans, is needed in order to develop effective vaccines against these pathogens.
ACKNOWLEDGEMENTS
This work was funded by the University of Tennessee. We would like to thank Dr. Barry Rouse for editing the manuscript.
CONFLICT OF INTEREST
The authors confirm that this article content has no conflicts of interest.
REFERENCES
- 1.Sturm A, Amino R, van de Sand C, et al. Manipulation of host hepatocytes by the malaria parasite for delivery into liver sinusoids. Science. 2006;313(5791):1287–90. doi: 10.1126/science.1129720. [DOI] [PubMed] [Google Scholar]
- 2.Vaughan AM, Mikolajczak SA, Wilson EM, et al. Complete Plasmodium falciparum liver-stage development in liver-chimeric mice. J Clin Invest. 2012;122(10):3618–28. doi: 10.1172/JCI62684. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Weiss WR, Sedegah M, Beaudoin RL, Miller LH, Good MF. CD8+ T cells (cytotoxic/suppressors) are required for protection in mice immunized with malaria sporozoites. Proc Natl Acad Sci USA. 1988;85(2):573–6. doi: 10.1073/pnas.85.2.573. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Schofield L, Villaquiran J, Ferreira A, Schellekens H, Nussenzweig R, Nussenzweig V. Gamma interferon, CD8+ T cells and antibodies required for immunity to malaria sporozoites. Nature. 1987;330(6149):664–6. doi: 10.1038/330664a0. [DOI] [PubMed] [Google Scholar]
- 5.Jobe O, Donofrio G, Sun G, Liepinsh D, Schwenk R, Krzych U. Immunization with radiation-attenuated Plasmodium berghei sporozoites induces liver cCD8alpha+DC that activate CD8+T cells against liver-stage malaria. PLoS One. 2009;4(4):e5075. doi: 10.1371/journal.pone.0005075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Obeid M, Franetich JF, Lorthiois A, et al. Skin-draining lymph node priming is sufficient to induce sterile immunity against pre-erythrocytic malaria. EMBO Mol Med. 2013;5(2):250–63. doi: 10.1002/emmm.201201677. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Chakravarty S, Cockburn IA, Kuk S, Overstreet MG, Sacci JB, Zavala F. CD8+ T lymphocytes protective against malaria liver stages are primed in skin-draining lymph nodes. Nat Med. 2007;13(9):1035–41. doi: 10.1038/nm1628. [DOI] [PubMed] [Google Scholar]
- 8.Luo ZJ, Tanaka T, Kimura F, Miyasaka M. Analysis of the mode of action of a novel immunosuppressant FTY720 in mice. Immunopharmacology. 1999;41(3):199–207. doi: 10.1016/s0162-3109(99)00004-1. [DOI] [PubMed] [Google Scholar]
- 9.Gueirard P, Tavares J, Thiberge S, et al. Development of the malaria parasite in the skin of the mammalian host. Proc Natl Acad Sci USA. 2010;107(43):18640–5. doi: 10.1073/pnas.1009346107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Voza T, Miller JL, Kappe SH, Sinnis P. Extrahepatic exoerythrocytic forms of rodent malaria parasites at the site of inoculation: clearance after immunization, susceptibility to primaquine, and contribution to blood-stage infection. Infect Immun. 2012;80(6):2158–64. doi: 10.1128/IAI.00246-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Yamauchi LM, Coppi A, Snounou G, Sinnis P. Plasmodium sporozoites trickle out of the injection site. Cell Microbiol. 2007;9(5):1215–22. doi: 10.1111/j.1462-5822.2006.00861.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Amino R, Thiberge S, Shorte S, Frischknecht F, Ménard R. Quantitative imaging of Plasmodium sporozoites in the mammalian host. C R Biol. 2006;329(11):858–62. doi: 10.1016/j.crvi.2006.04.003. [DOI] [PubMed] [Google Scholar]
- 13.Doolan DL, Hoffman SL. The complexity of protective immunity against liver-stage malaria. J Immunol. 2000;165(3):1453–62. doi: 10.4049/jimmunol.165.3.1453. [DOI] [PubMed] [Google Scholar]
- 14.Nardin EH, Oliveira GA, Calvo-Calle JM, et al. Synthetic malaria peptide vaccine elicits high levels of antibodies in vaccinees of defined HLA genotypes. J Infect Dis. 2000;182(5):1486–96. doi: 10.1086/315871. [DOI] [PubMed] [Google Scholar]
- 15.Plebanski M, Hannan CM, Behboudi S, et al. Direct processing and presentation of antigen from malaria sporozoites by professional antigen-presenting cells in the induction of CD8 T-cell responses. Immunol Cell Biol. 2005;83(3):307–12. doi: 10.1111/j.1440-1711.2005.01325.x. [DOI] [PubMed] [Google Scholar]
- 16.Jung S, Unutmaz D, Wong P, et al. In vivo depletion of CD11c+ dendritic cells abrogates priming of CD8+ T cells by exogenous cell-associated antigens. Immunity. 2002;17(2):211–20. doi: 10.1016/s1074-7613(02)00365-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Bedoui S, Whitney PG, Waithman J, et al. Cross-presentation of viral and self antigens by skin-derived CD103+ dendritic cells. Nat Immunol. 2009;10(5):488–95. doi: 10.1038/ni.1724. [DOI] [PubMed] [Google Scholar]
- 18.Allan RS, Waithman J, Bedoui S, et al. Migratory dendritic cells transfer antigen to a lymph node-resident dendritic cell population for efficient CTL priming. Immunity. 2006;25(1):153–62. doi: 10.1016/j.immuni.2006.04.017. [DOI] [PubMed] [Google Scholar]
- 19.Ebrahimkhani MR, Mohar I, Crispe IN. Cross-presentation of antigen by diverse subsets of murine liver cells. Hepatology. 2011;54(4):1379–87. doi: 10.1002/hep.24508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Cockburn IA, Chen YC, Overstreet MG, et al. Prolonged antigen presentation is required for optimal CD8+ T cell responses against malaria liver stage parasites. PLoS Pathog. 2010;6(5):e1000877. doi: 10.1371/journal.ppat.1000877. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hafalla JC, Sano G, Carvalho LH, Morrot A, Zavala F. Short-term antigen presentation and single clonal burst limit the magnitude of the CD8(+) T cell responses to malaria liver stages. Proc Natl Acad Sci USA. 2002;99(18):11819–24. doi: 10.1073/pnas.182189999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Morrot A, Zavala F. Effector and memory CD8+ T cells as seen in immunity to malaria. Immunol Rev. 2004;201:291–303. doi: 10.1111/j.0105-2896.2004.00175.x. [DOI] [PubMed] [Google Scholar]
- 23.Ryg-Cornejo V, Nie CQ, Bernard NJ, et al. NK cells and conventional dendritic cells engage in reciprocal activation for the induction of inflammatory responses during Plasmodium berghei ANKA infection. Immunobiology. 2013;218(2):263–71. doi: 10.1016/j.imbio.2012.05.018. [DOI] [PubMed] [Google Scholar]
- 24.Carvalho LH, Sano G, Hafalla JC, Morrot A, Curotto de Lafaille MA, Zavala F. IL-4-secreting CD4+ T cells are crucial to the development of CD8+ T-cell responses against malaria liver stages. Nat Med. 2002;8(2):166–70. doi: 10.1038/nm0202-166. [DOI] [PubMed] [Google Scholar]
- 25.Morrot A, Hafalla JC, Cockburn IA, Carvalho LH, Zavala F. IL-4 receptor expression on CD8+ T cells is required for the development of protective memory responses against liver stages of malaria parasites. J Exp Med. 2005;202(4):551–60. doi: 10.1084/jem.20042463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Acacia de Sa Pinheiro A, Morrot A, Chakravarty S, et al. IL-4 induces a wide-spectrum intracellular signaling cascade in CD8+ T cells. J Leukoc Biol. 2007;81(4):1102–10. doi: 10.1189/jlb.0906583. [DOI] [PubMed] [Google Scholar]
- 27.Morrot A, Zavala F. Regulation of the CD8+ T cell responses against Plasmodium liver stages in mice. Int J Parasitol. 2004;34(13-14):1529–34. doi: 10.1016/j.ijpara.2004.10.001. [DOI] [PubMed] [Google Scholar]
- 28.Hafalla JC, Morrot A, Sano G, Milon G, Lafaille JJ, Zavala F. Early self-regulatory mechanisms control the magnitude of CD8+ T cell responses against liver stages of murine malaria. J Immunol. 2003;171(2):964–70. doi: 10.4049/jimmunol.171.2.964. [DOI] [PubMed] [Google Scholar]
- 29.Silva HB, Caetano SS, Monteiro I, et al. Early skin immunological disturbance after Plasmodium-infected mosquito bites. Cell Immunol. 2012;277(1-2):22–32. doi: 10.1016/j.cellimm.2012.06.003. [DOI] [PubMed] [Google Scholar]
- 30.Rankin KE, Graewe S, Heussler VT, Stanway RR. Imaging liver-stage malaria parasites. Cell Microbiol. 2010;12(5):569–79. doi: 10.1111/j.1462-5822.2010.01454.x. [DOI] [PubMed] [Google Scholar]
- 31.Baer K, Klotz C, Kappe SH, Schnieder T, Frevert U. Release of hepatic Plasmodium yoelii merozoites into the pulmonary microvasculature. PLoS Pathog. 2007;3(11):e171. doi: 10.1371/journal.ppat.0030171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Miller JD, van der Most RG, Akondy RS, et al. Human effector and memory CD8+ T cell responses to smallpox and yellow fever vaccines. Immunity. 2008;28(5):710–22. doi: 10.1016/j.immuni.2008.02.020. [DOI] [PubMed] [Google Scholar]
- 33.Krzych U, Dalai S, Zarling S, Pichugin A. Memory CD8 T cells specific for plasmodia liver-stage antigens maintain protracted protection against malaria. Front Immunol. 2012;3:370. doi: 10.3389/fimmu.2012.00370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Zarling S, Berenzon D, Dalai S, Liepinsh D, Steers N, Krzych U. The survival of memory CD8 T cells that is mediated by IL-15 correlates with sustained protection against malaria. J Immunol. 2013;190(10):5128–41. doi: 10.4049/jimmunol.1203396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Nolz JC, Harty JT. Protective capacity of memory CD8+ T cells is dictated by antigen exposure history and nature of the infection. Immunity. 2011;34(5):781–93. doi: 10.1016/j.immuni.2011.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Wherry EJ, Teichgräber V, Becker TC, et al. Lineage relationship and protective immunity of memory CD8 T cell subsets. Nat Immunol. 2003;4(3):225–34. doi: 10.1038/ni889. [DOI] [PubMed] [Google Scholar]
- 37.Reyes-Sandoval A, Wyllie DH, Bauza K, et al. CD8+ T effector memory cells protect against liver-stage malaria. J Immunol. 2011;187(3):1347–57. doi: 10.4049/jimmunol.1100302. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Schmidt NW, Podyminogin RL, Butler NS, et al. Memory CD8 T cell responses exceeding a large but definable threshold provide long-term immunity to malaria. Proc Natl Acad Sci USA. 2008;105(37):14017–22. doi: 10.1073/pnas.0805452105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Schmidt NW, Butler NS, Harty JT. Plasmodium-host interactions directly influence the threshold of memory CD8 T cells required for protective immunity. J Immunol. 2011;186(10):5873–84. doi: 10.4049/jimmunol.1100194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Homann D, Teyton L, Oldstone MB. Differential regulation of antiviral T-cell immunity results in stable CD8+ but declining CD4+ T-cell memory. Nat Med. 2001;7(8):913–9. doi: 10.1038/90950. [DOI] [PubMed] [Google Scholar]
- 41.Renggli J, Hahne M, Matile H, Betschart B, Tschopp J, Corradin G. Elimination of P. berghei liver stages is independent of Fas (CD95/Apo-I) or perforin-mediated cytotoxicity. Parasite Immunol. 1997;19(3):145–8. doi: 10.1046/j.1365-3024.1997.d01-190.x. [DOI] [PubMed] [Google Scholar]
- 42.Nganou-Makamdop K, van Gemert GJ, Arens T, Hermsen CC, Sauerwein RW. Long term protection after immunization with P. berghei sporozoites correlates with sustained IFNγ responses of hepatic CD8+ memory T cells. PLoS One. 2012;7(5):e36508. doi: 10.1371/journal.pone.0036508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Jobe O, Lumsden J, Mueller AK, et al. Genetically attenuated Plasmodium berghei liver stages induce sterile protracted protection that is mediated by major histocompatibility complex Class I-dependent interferon-gamma-producing CD8+ T cells. J Infect Dis. 2007;196(4):599–607. doi: 10.1086/519743. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Clark IA, Hunt NH, Butcher GA, Cowden WB. Inhibition of murine malaria (Plasmodium chabaudi) in vivo by recombinant interferon-gamma or tumor necrosis factor, and its enhancement by butylated hydroxyanisole. J Immunol. 1987;139(10):3493–6. [PubMed] [Google Scholar]
- 45.Zhou F. Molecular mechanisms of IFN-gamma to up-regulate MHC class I antigen processing and presentation. Int Rev Immunol. 2009;28(3-4):239–60. doi: 10.1080/08830180902978120. [DOI] [PubMed] [Google Scholar]
- 46.Akiyama K, Yokota K, Kagawa S, et al. cDNA cloning and interferon gamma down-regulation of proteasomal subunits X and Y. Science. 1994;265(5176):1231–4. doi: 10.1126/science.8066462. [DOI] [PubMed] [Google Scholar]
- 47.Boes B, Hengel H, Ruppert T, Multhaup G, Koszinowski UH, Kloetzel PM. Interferon gamma stimulation modulates the proteolytic activity and cleavage site preference of 20S mouse proteasomes. J Exp Med. 1994;179(3):901–9. doi: 10.1084/jem.179.3.901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Mellouk S, Maheshwari RK, Rhodes-Feuillette A, et al. Inhibitory activity of interferons and interleukin 1 on the development of Plasmodium falciparum in human hepatocyte cultures. J Immunol. 1987;139(12):4192–5. [PubMed] [Google Scholar]
- 49.Seguin MC, Klotz FW, Schneider I, et al. Induction of nitric oxide synthase protects against malaria in mice exposed to irradiated Plasmodium berghei infected mosquitoes: involvement of interferon gamma and CD8+ T cells. J Exp Med. 1994;180(1):353–8. doi: 10.1084/jem.180.1.353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Depinay N, Franetich JF, Grüner AC, et al. Inhibitory effect of TNF-α on malaria pre-erythrocytic stage development: influence of host hepatocyte/parasite combinations. PLoS One. 2011;6(3):e17464. doi: 10.1371/journal.pone.0017464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Nussler A, Pied S, Goma J, Rénia L, Miltgen F, Grau GE, et al. TNF inhibits malaria hepatic stages in vitro via synthesis of IL-6. Int Immunol. 1991;3(4):317–21. doi: 10.1093/intimm/3.4.317. [DOI] [PubMed] [Google Scholar]
- 52.Pied S, Rénia L, Nüssler A, Miltgen F, Mazier D. Inhibitory activity of IL-6 on malaria hepatic stages. Parasite Immunol. 1991;13(2):211–7. doi: 10.1111/j.1365-3024.1991.tb00276.x. [DOI] [PubMed] [Google Scholar]
- 53.Butler NS, Schmidt NW, Harty JT. Differential effector pathways regulate memory CD8 T cell immunity against Plasmodium berghei versus P. yoelii sporozoites. J Immunol. 2010;184(5):2528–38. doi: 10.4049/jimmunol.0903529. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Trimnell A, Takagi A, Gupta M, Richie TL, Kappe SH, Wang R. Genetically attenuated parasite vaccines induce contact-dependent CD8+ T cell killing of Plasmodium yoelii liver stage-infected hepatocytes. J Immunol. 2009;183(9):5870–8. doi: 10.4049/jimmunol.0900302. [DOI] [PubMed] [Google Scholar]
- 55.Cockburn IA, Amino R, Kelemen RK, et al. In vivo imaging of CD8+ T cell-mediated elimination of malaria liver stages. Proc Natl Acad Sci USA. 2013;110(22):9090–5. doi: 10.1073/pnas.1303858110. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Nussenzweig RS, Vanderberg J, Most H, Orton C. Protective immunity produced by the injection of x-irradiated sporozoites of plasmodium berghei. Nature. 1967;216(5111):160–2. doi: 10.1038/216160a0. [DOI] [PubMed] [Google Scholar]
- 57.Clyde DF. Immunization of man against falciparum and vivax malaria by use of attenuated sporozoites. Am J Trop Med Hyg. 1975;24(3):397–401. doi: 10.4269/ajtmh.1975.24.397. [DOI] [PubMed] [Google Scholar]
- 58.Hoffman BU, Chattopadhyay R. Plasmodium falciparum: effect of radiation on levels of gene transcripts in sporozoites. Exp Parasitol. 2008;118(2):247–52. doi: 10.1016/j.exppara.2007.08.014. [DOI] [PubMed] [Google Scholar]
- 59.Suhrbier A, Winger LA, Castellano E, Sinden RE. Survival and antigenic profile of irradiated malarial sporozoites in infected liver cells. Infect Immun. 1990;58(9):2834–9. doi: 10.1128/iai.58.9.2834-2839.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Silvie O, Semblat JP, Franetich JF, Hannoun L, Eling W, Mazier D. Effects of irradiation on Plasmodium falciparum sporozoite hepatic development: implications for the design of pre-erythrocytic malaria vaccines. Parasite Immunol. 2002;24(4):221–3. doi: 10.1046/j.1365-3024.2002.00450.x. [DOI] [PubMed] [Google Scholar]
- 61.Nganou-Makamdop K, Sauerwein RW. Liver or blood-stage arrest during malaria sporozoite immunization: the later the better? Trends Parasitol. 2013;29(6):304–10. doi: 10.1016/j.pt.2013.03.008. [DOI] [PubMed] [Google Scholar]
- 62.Chattopadhyay R, Conteh S, Li M, James ER, Epstein JE, Hoffman SL. The Effects of radiation on the safety and protective efficacy of an attenuated Plasmodium yoelii sporozoite malaria vaccine. Vaccine. 2009;27(27):3675–80. doi: 10.1016/j.vaccine.2008.11.073. [DOI] [PubMed] [Google Scholar]
- 63.Nardin E, Zavala F, Nussenzweig V, Nussenzweig RS. Pre-erythrocytic malaria vaccine: mechanisms of protective immunity and human vaccine trials. Parassitologia. 1999;41(1-3):397–402. [PubMed] [Google Scholar]
- 64.Frevert U, Nardin E. Cellular effector mechanisms against Plasmodium liver stages. Cell Microbiol. 2008;10(10):1956–67. doi: 10.1111/j.1462-5822.2008.01211.x. [DOI] [PubMed] [Google Scholar]
- 65.Calvo-Calle JM, Oliveira GA, Nardin EH. Human CD4+ T cells induced by synthetic peptide malaria vaccine are comparable to cells elicited by attenuated Plasmodium falciparum sporozoites. J Immunol. 2005;175(11):7575–85. doi: 10.4049/jimmunol.175.11.7575. [DOI] [PubMed] [Google Scholar]
- 66.Epstein JE, Tewari K, Lyke KE, et al. Live attenuated malaria vaccine designed to protect through hepatic CD8+ T cell immunity. Science. 2011;334(6055):475–80. doi: 10.1126/science.1211548. [DOI] [PubMed] [Google Scholar]
- 67.Gordon DM, McGovern TW, Krzych U, et al. Safety, immunogenicity, and efficacy of a recombinantly produced Plasmodium falciparum circumsporozoite protein-hepatitis B surface antigen subunit vaccine. J Infect Dis. 1995;171(6):1576–85. doi: 10.1093/infdis/171.6.1576. [DOI] [PubMed] [Google Scholar]
- 68.Kester KE, McKinney DA, Tornieporth N, et al. Efficacy of recombinant circumsporozoite protein vaccine regimens against experimental Plasmodium falciparum malaria. J Infect Dis. 2001;183(4):640–7. doi: 10.1086/318534. [DOI] [PubMed] [Google Scholar]
- 69.Olotu A, Fegan G, Wambua J, et al. Four-year efficacy of RTS,S/AS01E and its interaction with malaria exposure. N Engl J Med. 2013;368(12):1111–20. doi: 10.1056/NEJMoa1207564. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Agnandji ST, Lell B, Fernandes JF, et al. A phase 3 trial of RTS,S/AS01 malaria vaccine in African infants. N Engl J Med. 2012;367(24):2284–95. doi: 10.1056/NEJMoa1208394. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Agnandji ST, Lell B, Soulanoudjingar SS, et al. First results of phase 3 trial of RTS,S/AS01 malaria vaccine in African children. N Engl J Med. 2011;365(20):1863–75. doi: 10.1056/NEJMoa1102287. [DOI] [PubMed] [Google Scholar]
- 72.Vaughan AM, Kappe SH. Vaccination using radiation- or genetically attenuated live sporozoites. Methods Mol Biol. 2013;923:549–66. doi: 10.1007/978-1-62703-026-7_38. [DOI] [PubMed] [Google Scholar]
- 73.van Dijk MR, Douradinha B, Franke-Fayard B, et al. Genetically attenuated, P36p-deficient malarial sporozoites induce protective immunity and apoptosis of infected liver cells. Proc Natl Acad Sci USA. 2005;102(34):12194–9. doi: 10.1073/pnas.0500925102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Labaied M, Harupa A, Dumpit RF, Coppens I, Mikolajczak SA, Kappe SH. Plasmodium yoelii sporozoites with simultaneous deletion of P52 and P36 are completely attenuated and confer sterile immunity against infection. Infect Immun. 2007;75(8):3758–68. doi: 10.1128/IAI.00225-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Aly AS, Lindner SE, MacKellar DC, Peng X, Kappe SH. SAP1 is a critical post-transcriptional regulator of infectivity in malaria parasite sporozoite stages. Mol Microbiol. 2011;79(4):929–39. doi: 10.1111/j.1365-2958.2010.07497.x. [DOI] [PubMed] [Google Scholar]
- 76.Aly AS, Mikolajczak SA, Rivera HS, et al. Targeted deletion of SAP1 abolishes the expression of infectivity factors necessary for successful malaria parasite liver infection. Mol Microbiol. 2008;69(1):152–63. doi: 10.1111/j.1365-2958.2008.06271.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Mueller AK, Camargo N, Kaiser K, et al. Plasmodium liver stage developmental arrest by depletion of a protein at the parasite-host interface. Proc Natl Acad Sci USA. 2005;102(8):3022–7. doi: 10.1073/pnas.0408442102. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Tarun AS, Dumpit RF, Camargo N, et al. Protracted sterile protection with Plasmodium yoelii pre-erythrocytic genetically attenuated parasite malaria vaccines is independent of significant liver-stage persistence and is mediated by CD8+ T cells. J Infect Dis. 2007;196(4):608–16. doi: 10.1086/519742. [DOI] [PubMed] [Google Scholar]
- 79.Yu M, Kumar TR, Nkrumah LJ, et al. The fatty acid biosynthesis enzyme FabI plays a key role in the development of liver-stage malarial parasites. Cell Host Microbe. 2008;4(6):567–78. doi: 10.1016/j.chom.2008.11.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Vaughan AM, O'Neill MT, Tarun AS, et al. Type II fatty acid synthesis is essential only for malaria parasite late liver stage development. Cell Microbiol. 2009;11(3):506–20. doi: 10.1111/j.1462-5822.2008.01270.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Falae A, Combe A, Amaladoss A, Carvalho T, Menard R, Bhanot P. Role of Plasmodium berghei cGMP-dependent protein kinase in late liver stage development. J Biol Chem. 2010;285(5):3282–8. doi: 10.1074/jbc.M109.070367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Friesen J, Matuschewski K. Comparative efficacy of pre-erythrocytic whole organism vaccine strategies against the malaria parasite. Vaccine. 2011;29(40):7002–8. doi: 10.1016/j.vaccine.2011.07.034. [DOI] [PubMed] [Google Scholar]
- 83.Butler NS, Schmidt NW, Vaughan AM, Aly AS, Kappe SH, Harty JT. Superior antimalarial immunity after vaccination with late liver stage-arresting genetically attenuated parasites. Cell Host Microbe. 2011;9(6):451–62. doi: 10.1016/j.chom.2011.05.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Schmidt NW, Harty JT. Cutting edge: attrition of Plasmodium-specific memory CD8 T cells results in decreased protection that is rescued by booster immunization. J Immunol. 2011;186(7):3836–40. doi: 10.4049/jimmunol.1003949. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Cunha-Rodrigues M, Prudêncio M, Mota MM, Haas W. Antimalarial drugs - host targets (re)visited. Biotechnol J. 2006;1(3):321–32. doi: 10.1002/biot.200500038. [DOI] [PubMed] [Google Scholar]
- 86.Belnoue E, Costa FT, Frankenberg T, et al. Protective T cell immunity against malaria liver stage after vaccination with live sporozoites under chloroquine treatment. J Immunol. 2004;172(4):2487–95. doi: 10.4049/jimmunol.172.4.2487. [DOI] [PubMed] [Google Scholar]
- 87.Schlitzer M. Antimalarial drugs - what is in use and what is in the pipeline. Arch Pharm (Weinheim) 2008;341(3):149–63. doi: 10.1002/ardp.200700184. [DOI] [PubMed] [Google Scholar]
- 88.Friesen J, Silvie O, Putrianti ED, Hafalla JC, Matuschewski K, Borrmann S. Natural immunization against malaria: causal prophylaxis with antibiotics. Sci Transl Med. 2010;2(40):40ra9. doi: 10.1126/scitranslmed.3001058. [DOI] [PubMed] [Google Scholar]
- 89.Roestenberg M, McCall M, Hopman J, et al. Protection against a malaria challenge by sporozoite inoculation. N Engl J Med. 2009;361(5):468–77. doi: 10.1056/NEJMoa0805832. [DOI] [PubMed] [Google Scholar]
- 90.Clyde DF, McCarthy VC, Miller RM, Hornick RB. Specificity of protection of man immunized against sporozoite-induced falciparum malaria. Am J Med Sci. 1973;266(6):398–403. doi: 10.1097/00000441-197312000-00001. [DOI] [PubMed] [Google Scholar]
- 91.Hoffman SL, Goh LM, Luke TC, et al. Protection of humans against malaria by immunization with radiation-attenuated Plasmodium falciparum sporozoites. J Infect Dis. 2002;185(8):1155–64. doi: 10.1086/339409. [DOI] [PubMed] [Google Scholar]
- 92.Schuldt NJ, Aldhamen YA, Godbehere-Roosa S, Seregin SS, Kousa YA, Amalfitano A. Immunogenicity when utilizing adenovirus serotype 4 and 5 vaccines expressing circumsporozoite protein in naïve and adenovirus (Ad5) immune mice. Malar J. 2012;11:209. doi: 10.1186/1475-2875-11-209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Anderson RJ, Hannan CM, Gilbert SC, et al. Enhanced CD8+ T cell immune responses and protection elicited against Plasmodium berghei malaria by prime boost immunization regimens using a novel attenuated fowlpox virus. J Immunol. 2004;172(5):3094–100. doi: 10.4049/jimmunol.172.5.3094. [DOI] [PubMed] [Google Scholar]
- 94.Limbach KJ, Richie TL. Viral vectors in malaria vaccine development. Parasite Immunol. 2009;31(9):501–19. doi: 10.1111/j.1365-3024.2009.01141.x. [DOI] [PubMed] [Google Scholar]
- 95.Schuldt NJ, Amalfitano A. Malaria vaccines: focus on adenovirus based vectors. Vaccine. 2012;30(35):5191–8. doi: 10.1016/j.vaccine.2012.05.048. [DOI] [PubMed] [Google Scholar]
- 96.Barlan AU, Griffin TM, McGuire KA, Wiethoff CM. Adenovirus membrane penetration activates the NLRP3 inflammasome. J Virol. 2011;85(1):146–55. doi: 10.1128/JVI.01265-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Roberts DM, Nanda A, Havenga MJ, et al. Hexon-chimaeric adenovirus serotype 5 vectors circumvent pre-existing anti-vector immunity. Nature. 2006;441(7090):239–43. doi: 10.1038/nature04721. [DOI] [PubMed] [Google Scholar]
- 98.Saxena M, Van TT, Baird FJ, Coloe PJ, Smooker PM. Pre-existing immunity against vaccine vectors--friend or foe? Microbiology. 2013;159(Pt 1):1–11. doi: 10.1099/mic.0.049601-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Walther M, Dunachie S, Keating S, et al. Safety, immunogenicity and efficacy of a pre-erythrocytic malaria candidate vaccine, ICC-1132 formulated in Seppic ISA 720. Vaccine. 2005;23(7):857–64. doi: 10.1016/j.vaccine.2004.08.020. [DOI] [PubMed] [Google Scholar]
- 100.Ockenhouse CF, Sun PF, Lanar DE, et al. Phase I/IIa safety, immunogenicity, and efficacy trial of NYVAC-Pf7, a pox-vectored, multiantigen, multistage vaccine candidate for Plasmodium falciparum malaria. J Infect Dis. 1998;177(6):1664–73. doi: 10.1086/515331. [DOI] [PubMed] [Google Scholar]
- 101.Shiratsuchi T, Rai U, Krause A, Worgall S, Tsuji M. Replacing adenoviral vector HVR1 with a malaria B cell epitope improves immunogenicity and circumvents preexisting immunity to adenovirus in mice. J Clin Invest. 2010;120(10):3688–701. doi: 10.1172/JCI39812. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Kumar S, Miller LH. Cellular mechanisms in immunity to blood stage infection. Immunol Lett. 1990;25(1-3):109–14. doi: 10.1016/0165-2478(90)90100-5. [DOI] [PubMed] [Google Scholar]
- 103.Vinetz JM, Kumar S, Good MF, Fowlkes BJ, Berzofsky JA, Miller LH. Adoptive transfer of CD8+ T cells from immune animals does not transfer immunity to blood stage Plasmodium yoelii malaria. J Immunol. 1990;144(3):1069–74. [PubMed] [Google Scholar]
- 104.Belnoue E, Kayibanda M, Vigario AM, et al. On the pathogenic role of brain-sequestered alphabeta CD8+ T cells in experimental cerebral malaria. J Immunol. 2002;169(11):6369–75. doi: 10.4049/jimmunol.169.11.6369. [DOI] [PubMed] [Google Scholar]
- 105.Lau LS, Fernandez Ruiz D, Davey GM, et al. Blood-stage Plasmodium berghei infection generates a potent, specific CD8+ T-cell response despite residence largely in cells lacking MHC I processing machinery. J Infect Dis. 2011;204(12):1989–96. doi: 10.1093/infdis/jir656. [DOI] [PubMed] [Google Scholar]
- 106.Howe L, Castro IC, Schoener ER, Hunter S, Barraclough RK, Alley MR. Malaria parasites (Plasmodium spp.) infecting introduced, native and endemic New Zealand birds. Parasitol Res. 2012;110(2):913–23. doi: 10.1007/s00436-011-2577-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Miyakoda M, Kimura D, Honma K, Kimura K, Yuda M, Yui K. Development of memory CD8+ T cells and their recall responses during blood-stage infection with Plasmodium berghei ANKA. J Immunol. 2012;189(9):4396–404. doi: 10.4049/jimmunol.1200781. [DOI] [PubMed] [Google Scholar]
- 108.Lundie RJ, de Koning-Ward TF, Davey GM, et al. Blood-stage Plasmodium infection induces CD8+ T lymphocytes to parasite-expressed antigens, largely regulated by CD8alpha+ dendritic cells. Proc Natl Acad Sci USA. 2008;105(38):14509–14. doi: 10.1073/pnas.0806727105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Pouniotis DS, Proudfoot O, Bogdanoska V, et al. Selectively impaired CD8+ but not CD4+ T cell cycle arrest during priming as a consequence of dendritic cell interaction with plasmodium-infected red cells. J Immunol. 2005;175(6):3525–33. doi: 10.4049/jimmunol.175.6.3525. [DOI] [PubMed] [Google Scholar]
- 110.Hermsen C, van de Wiel T, Mommers E, Sauerwein R, Eling W. Depletion of CD4+ or CD8+ T-cells prevents Plasmodium berghei induced cerebral malaria in end-stage disease. Parasitology. 1997;114( Pt 1):7–12. doi: 10.1017/s0031182096008293. [DOI] [PubMed] [Google Scholar]
- 111.deWalick S, Amante FH, McSweeney KA, et al. Cutting edge: conventional dendritic cells are the critical APC required for the induction of experimental cerebral malaria. J Immunol. 2007;178(10):6033–7. doi: 10.4049/jimmunol.178.10.6033. [DOI] [PubMed] [Google Scholar]
- 112.Nitcheu J, Bonduelle O, Combadiere C, et al. Perforin-dependent brain-infiltrating cytotoxic CD8+ T lymphocytes mediate experimental cerebral malaria pathogenesis. J Immunol. 2003;170(4):2221–8. doi: 10.4049/jimmunol.170.4.2221. [DOI] [PubMed] [Google Scholar]
- 113.Randall LM, Amante FH, McSweeney KA, et al. Common strategies to prevent and modulate experimental cerebral malaria in mouse strains with different susceptibilities. Infect Immun. 2008;76(7):3312–20. doi: 10.1128/IAI.01475-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Claser C, Malleret B, Gun SY, et al. CD8+ T cells and IFN-γ mediate the time-dependent accumulation of infected red blood cells in deep organs during experimental cerebral malaria. PLoS One. 2011;6(4):e18720. doi: 10.1371/journal.pone.0018720. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Haque A, Best SE, Unosson K, et al. Granzyme B expression by CD8+ T cells is required for the development of experimental cerebral malaria. J Immunol. 2011;186(11):6148–56. doi: 10.4049/jimmunol.1003955. [DOI] [PubMed] [Google Scholar]
- 116.Jacobs L. Toxoplasma and toxoplasmosis. Adv Parasitol. 1967;5:1–45. doi: 10.1016/s0065-308x(08)60374-7. [DOI] [PubMed] [Google Scholar]
- 117.Esch KJ, Petersen CA. Transmission and epidemiology of zoonotic protozoal diseases of companion animals. Clin Microbiol Rev. 2013;26(1):58–85. doi: 10.1128/CMR.00067-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Dubey JP. Advances in the life cycle of Toxoplasma gondii. Int J Parasitol. 1998;28(7):1019–24. doi: 10.1016/s0020-7519(98)00023-x. [DOI] [PubMed] [Google Scholar]
- 119.Dupont CD, Christian DA, Hunter CA. Immune response and immunopathology during toxoplasmosis. Semin Immunopathol. 2012;34(6):793–813. doi: 10.1007/s00281-012-0339-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Goldszmid RS, Coppens I, Lev A, Caspar P, Mellman I, Sher A. Host ER-parasitophorous vacuole interaction provides a route of entry for antigen cross-presentation in Toxoplasma gondii-infected dendritic cells. J Exp Med. 2009;206(2):399–410. doi: 10.1084/jem.20082108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Gazzinelli RT, Hakim FT, Hieny S, Shearer GM, Sher A. Synergistic role of CD4+ and CD8+ T lymphocytes in IFN-gamma production and protective immunity induced by an attenuated Toxoplasma gondii vaccine. J Immunol. 1991;146(1):286–92. [PubMed] [Google Scholar]
- 122.Gazzinelli R, Xu Y, Hieny S, Cheever A, Sher A. Simultaneous depletion of CD4+ and CD8+ T lymphocytes is required to reactivate chronic infection with Toxoplasma gondii. J Immunol. 1992;149(1):175–80. [PubMed] [Google Scholar]
- 123.Scharton-Kersten T, Caspar P, Sher A, Denkers EY. Toxoplasma gondii: evidence for interleukin-12-dependent and-independent pathways of interferon-gamma production induced by an attenuated parasite strain. Exp Parasitol. 1996;84(2):102–14. doi: 10.1006/expr.1996.0096. [DOI] [PubMed] [Google Scholar]
- 124.Sa Q, Woodward J, Suzuki Y. IL-2 produced by CD8+ immune T cells can augment their IFN-γ production independently from their proliferation in the secondary response to an intracellular pathogen. J Immunol. 2013;190(5):2199–207. doi: 10.4049/jimmunol.1202256. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Hakim FT, Gazzinelli RT, Denkers E, Hieny S, Shearer GM, Sher A. CD8+ T cells from mice vaccinated against Toxoplasma gondii are cytotoxic for parasite-infected or antigen-pulsed host cells. J Immunol. 1991;147(7):2310–6. [PubMed] [Google Scholar]
- 126.Subauste CS, Koniaris AH, Remington JS. Murine CD8+ cytotoxic T lymphocytes lyse Toxoplasma gondii-infected cells. J Immunol. 1991;147(11):3955–9. [PubMed] [Google Scholar]
- 127.Denkers EY, Yap G, Scharton-Kersten T, et al. Perforin-mediated cytolysis plays a limited role in host resistance to Toxoplasma gondii. J Immunol. 1997;159(4):1903–8. [PubMed] [Google Scholar]
- 128.Adams LB, Hibbs JB, Taintor RR, Krahenbuhl JL. Microbiostatic effect of murine-activated macrophages for Toxoplasma gondii. Role for synthesis of inorganic nitrogen oxides from L-arginine. J Immunol. 1990;144(7):2725–9. [PubMed] [Google Scholar]
- 129.Takács AC, Swierzy IJ, Lüder CG. Interferon-γ restricts Toxoplasma gondii development in murine skeletal muscle cells via nitric oxide production and immunity-related GTPases. PLoS One. 2012;7(9):e45440. doi: 10.1371/journal.pone.0045440. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Harris TH, Banigan EJ, Christian DA, et al. Generalized Lévy walks and the role of chemokines in migration of effector CD8+ T cells. Nature. 2012;486(7404):545–8. doi: 10.1038/nature11098. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Ashford RW. The leishmaniases as emerging and reemerging zoonoses. Int J Parasitol. 2000;30(12-13):1269–81. doi: 10.1016/s0020-7519(00)00136-3. [DOI] [PubMed] [Google Scholar]
- 132.Bates PA. Transmission of Leishmania metacyclic promastigotes by phlebotomine sand flies. Int J Parasitol. 2007;37(10):1097–106. doi: 10.1016/j.ijpara.2007.04.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Alexander CE, Kaye PM, Engwerda CR. CD95 is required for the early control of parasite burden in the liver of Leishmania donovani-infected mice. Eur J Immunol. 2001;31(4):1199–210. doi: 10.1002/1521-4141(200104)31:4<1199::aid-immu1199>3.0.co;2-6. [DOI] [PubMed] [Google Scholar]
- 134.Badovinac VP, Haring JS, Harty JT. Initial T cell receptor transgenic cell precursor frequency dictates critical aspects of the CD8(+) T cell response to infection. Immunity. 2007;26(6):827–41. doi: 10.1016/j.immuni.2007.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Barral-Netto M, Barral A, Brodskyn C, Carvalho EM, Reed SG. Cytotoxicity in human mucosal and cutaneous leishmaniasis. Parasite Immunol. 1995;17(1):21–8. doi: 10.1111/j.1365-3024.1995.tb00962.x. [DOI] [PubMed] [Google Scholar]
- 136.Basu R, Roy S, Walden P. HLA class I-restricted T cell epitopes of the kinetoplastid membrane protein-11 presented by Leishmania donovani-infected human macrophages. J Infect Dis. 2007;195(9):1373–80. doi: 10.1086/513439. [DOI] [PubMed] [Google Scholar]
- 137.Belkaid Y, Von Stebut E, Mendez S, et al. CD8+ T cells are required for primary immunity in C57BL/6 mice following lowdose, intradermal challenge with Leishmania major. J Immunol. 2002;168(8):3992–4000. doi: 10.4049/jimmunol.168.8.3992. [DOI] [PubMed] [Google Scholar]
- 138.Stäger S, Rafati S. CD8(+) T cells in leishmania infections: friends or foes? Front Immunol. 2012;3:5. doi: 10.3389/fimmu.2012.00005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Tanowitz HB, Kirchhoff LV, Simon D, Morris SA, Weiss LM, Wittner M. Chagas' disease. Clin Microbiol Rev. 1992;5(4):400–19. doi: 10.1128/cmr.5.4.400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Bustamante JM, Bixby LM, Tarleton RL. Drug-induced cure drives conversion to a stable and protective CD8+ T central memory response in chronic Chagas disease. Nat Med. 2008;14(5):542–50. doi: 10.1038/nm1744. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Bixby LM, Tarleton RL. Stable CD8+ T cell memory during persistent Trypanosoma cruzi infection. J Immunol. 2008;181(4):2644–50. doi: 10.4049/jimmunol.181.4.2644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Padilla AM, Simpson LJ, Tarleton RL. Insufficient TLR activation contributes to the slow development of CD8+ T cell responses in Trypanosoma cruzi infection. J Immunol. 2009;183(2):1245–52. doi: 10.4049/jimmunol.0901178. [DOI] [PubMed] [Google Scholar]
- 143.Tarleton RL, Koller BH, Latour A, Postan M. Susceptibility of beta 2-microglobulin-deficient mice to Trypanosoma cruzi infection. Nature. 1992;356(6367):338–40. doi: 10.1038/356338a0. [DOI] [PubMed] [Google Scholar]
- 144.Tarleton RL, Sun J, Zhang L, Postan M. Depletion of T-cell subpopulations results in exacerbation of myocarditis and parasitism in experimental Chagas' disease. Infect Immun. 1994;62(5):1820–9. doi: 10.1128/iai.62.5.1820-1829.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Wizel B, Nunes M, Tarleton RL. Identification of Trypanosoma cruzi trans-sialidase family members as targets of protective CD8+ TC1 responses. J Immunol. 1997;159(12):6120–30. [PubMed] [Google Scholar]
- 146.Wrightsman RA, Luhrs KA, Fouts D, Manning JE. Paraflagellar rod protein-specific CD8+ cytotoxic T lymphocytes target Trypanosoma cruzi-infected host cells. Parasite Immunol. 2002;24(8):401–12. doi: 10.1046/j.1365-3024.2002.00479.x. [DOI] [PubMed] [Google Scholar]
- 147.Fralish BH, Tarleton RL. Genetic immunization with LYT1 or a pool of trans-sialidase genes protects mice from lethal Trypanosoma cruzi infection. Vaccine. 2003;21(21-22):3070–80. doi: 10.1016/s0264-410x(03)00121-x. [DOI] [PubMed] [Google Scholar]
- 148.Martin DL, Weatherly DB, Laucella SA, et al. CD8+ T-Cell responses to Trypanosoma cruzi are highly focused on strain-variant trans-sialidase epitopes. PLoS Pathog. 2006;2(8):e77. doi: 10.1371/journal.ppat.0020077. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Tzelepis F, de Alencar BC, Penido ML, Gazzinelli RT, Persechini PM, Rodrigues MM. Distinct kinetics of effector CD8+ cytotoxic T cells after infection with Trypanosoma cruzi in naive or vaccinated mice. Infect Immun. 2006;74(4):2477–81. doi: 10.1128/IAI.74.4.2477-2481.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Tzelepis F, Persechini PM, Rodrigues MM. Modulation of CD4(+) T cell-dependent specific cytotoxic CD8(+) T cells differentiation and proliferation by the timing of increase in the pathogen load. PLoS One. 2007;2(4):e393. doi: 10.1371/journal.pone.0000393. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Leavey JK, Tarleton RL. Cutting edge: dysfunctional CD8+ T cells reside in nonlymphoid tissues during chronic Trypanosoma cruzi infection. J Immunol. 2003;170(5):2264–8. doi: 10.4049/jimmunol.170.5.2264. [DOI] [PubMed] [Google Scholar]
- 152.Laucella SA, Postan M, Martin D, et al. Frequency of interferon- gamma -producing T cells specific for Trypanosoma cruzi inversely correlates with disease severity in chronic human Chagas disease. J Infect Dis. 2004;189(5):909–18. doi: 10.1086/381682. [DOI] [PubMed] [Google Scholar]
- 153.Albareda MC, Olivera GC, Laucella SA, et al. Chronic human infection with Trypanosoma cruzi drives CD4+ T cells to immune senescence. J Immunol. 2009;183(6):4103–8. doi: 10.4049/jimmunol.0900852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Albareda MC, Laucella SA, Alvarez MG, et al. Trypanosoma cruzi modulates the profile of memory CD8+ T cells in chronic Chagas' disease patients. Int Immunol. 2006;18(3):465–71. doi: 10.1093/intimm/dxh387. [DOI] [PubMed] [Google Scholar]