Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
J Immunol ; 207(6): 1507-1512, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34400524

ABSTRACT

Resistance and tolerance are vital for survivability of the host-pathogen relationship. Virulence during Toxoplasma infection in mice is mediated by parasite kinase-dependent antagonism of IFN-γ-induced host resistance. Whether avirulence requires expression of parasite factors that induce host tolerance mechanisms or is a default status reflecting the absence of resistance-interfering factors is not known. In this study, we present evidence that avirulence in Toxoplasma requires parasite engagement of the scavenger receptor CD36. CD36 promotes macrophage tropism but is dispensable for the development of resistance mechanisms. Instead CD36 is critical for re-establishing tissue homeostasis and survival following the acute phase of infection. The CD36-binding capacity of T. gondii strains is negatively controlled by the virulence factor, ROP18. Thus, the absence of resistance-interfering virulence factors and the presence of tolerance-inducing avirulence factors are both required for long-term host-pathogen survival.


Subject(s)
CD36 Antigens/deficiency , CD36 Antigens/metabolism , Macrophages, Peritoneal/immunology , Macrophages, Peritoneal/parasitology , Toxoplasma/metabolism , Toxoplasma/pathogenicity , Toxoplasmosis, Animal/immunology , Animals , CD36 Antigens/genetics , CHO Cells , Cricetulus , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Immune Tolerance/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Serine-Threonine Kinases/metabolism , Protozoan Proteins/metabolism , RAW 264.7 Cells , Toxoplasmosis, Animal/metabolism , Toxoplasmosis, Animal/parasitology , Virulence/genetics , Virulence Factors/metabolism
2.
PLoS Pathog ; 16(5): e1008572, 2020 05.
Article in English | MEDLINE | ID: mdl-32413093

ABSTRACT

The apicomplexan Toxoplasma gondii induces strong protective immunity dependent upon recognition by Toll-like receptors (TLR)11 and 12 operating in conjunction with MyD88 in the murine host. However, TLR11 and 12 proteins are not present in humans, inspiring us to investigate MyD88-independent pathways of resistance. Using bicistronic IL-12-YFP reporter mice on MyD88+/+ and MyD88-/- genetic backgrounds, we show that CD11c+MHCII+F4/80- dendritic cells, F4/80+ macrophages, and Ly6G+ neutrophils were the dominant cellular sources of IL-12 in both wild type and MyD88 deficient mice after parasite challenge. Parasite dense granule protein GRA24 induces p38 MAPK activation and subsequent IL-12 production in host macrophages. We show that Toxoplasma triggers an early and late p38 MAPK phosphorylation response in MyD88+/+ and MyD88-/- bone marrow-derived macrophages. Using the uracil auxotrophic Type I T. gondii strain cps1-1, we demonstrate that the late response does not require active parasite proliferation, but strictly depends upon GRA24. By i. p. inoculation with cps1-1 and cps1-1:Δgra24, we identified unique subsets of chemokines and cytokines that were up and downregulated by GRA24. Finally, we demonstrate that cps1-1 triggers a strong host-protective GRA24-dependent Th1 response in the absence of MyD88. Our data identify GRA24 as a major mediator of p38 MAPK activation, IL-12 induction and protective immunity that operates independently of the TLR/MyD88 cascade.


Subject(s)
Interleukin-12/immunology , MAP Kinase Signaling System/immunology , Macrophages/immunology , Myeloid Differentiation Factor 88/immunology , Protozoan Proteins/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , p38 Mitogen-Activated Protein Kinases/immunology , Animals , Enzyme Activation/genetics , Enzyme Activation/immunology , Interleukin-12/genetics , MAP Kinase Signaling System/genetics , Macrophages/parasitology , Macrophages/pathology , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Protozoan Proteins/genetics , Toxoplasma/genetics , Toxoplasmosis/genetics , Toxoplasmosis/pathology , p38 Mitogen-Activated Protein Kinases/genetics
3.
Infect Immun ; 87(8)2019 08.
Article in English | MEDLINE | ID: mdl-31109947

ABSTRACT

Little is known about whether pathogen invasion of neural tissue is affected by immune-based mechanisms in endothelial cells. We examined the effects of endothelial cell CD40 on Toxoplasma gondii invasion of the retina and brain, organs seeded hematogenously. T. gondii circulates in the bloodstream within infected leukocytes (including monocytes and dendritic cells) and as extracellular tachyzoites. After T. gondii infection, mice that expressed CD40 restricted to endothelial cells exhibited diminished parasite loads and histopathology in the retina and brain. These mice also had lower parasite loads in the retina and brain after intravenous (i.v.) injection of infected monocytes or dendritic cells. The protective effect of endothelial cell CD40 was not explained by changes in cellular or humoral immunity, reduced transmigration of leukocytes into neural tissue, or reduced invasion by extracellular parasites. Circulating T. gondii-infected leukocytes (dendritic cells used as a model) led to infection of neural endothelial cells. The number of foci of infection in these cells were reduced if endothelial cells expressed CD40. Infected dendritic cells and macrophages expressed membrane-associated inducible Hsp70. Infected leukocytes triggered Hsp70-dependent autophagy in CD40+ endothelial cells and anti-T. gondii activity dependent on ULK1 and beclin 1. Reduced parasite load in the retina and brain not only required CD40 expression in endothelial cells but was also dependent on beclin 1 and the expression of inducible Hsp70 in dendritic cells. These studies suggest that during endothelial cell-leukocyte interaction, CD40 restricts T. gondii invasion of neural tissue through a mechanism that appears mediated by endothelial cell anti-parasitic activity stimulated by Hsp70.


Subject(s)
Brain/parasitology , CD40 Antigens/physiology , Endothelial Cells/immunology , Retina/parasitology , Toxoplasma/pathogenicity , Animals , Autophagy , Cell Movement , HSP70 Heat-Shock Proteins/physiology , Leukocytes/physiology , Mice , Mice, Inbred C57BL
4.
PLoS Genet ; 12(7): e1006189, 2016 07.
Article in English | MEDLINE | ID: mdl-27447180

ABSTRACT

Nonreplicating type I uracil auxotrophic mutants of Toxoplasma gondii possess a potent ability to activate therapeutic immunity to established solid tumors by reversing immune suppression in the tumor microenvironment. Here we engineered targeted deletions of parasite secreted effector proteins using a genetically tractable Δku80 vaccine strain to show that the secretion of specific rhoptry (ROP) and dense granule (GRA) proteins by uracil auxotrophic mutants of T. gondii in conjunction with host cell invasion activates antitumor immunity through host responses involving CD8α+ dendritic cells, the IL-12/interferon-gamma (IFN-γ) TH1 axis, as well as CD4+ and CD8+ T cells. Deletion of parasitophorous vacuole membrane (PVM) associated proteins ROP5, ROP17, ROP18, ROP35 or ROP38, intravacuolar network associated dense granule proteins GRA2 or GRA12, and GRA24 which traffics past the PVM to the host cell nucleus severely abrogated the antitumor response. In contrast, deletion of other secreted effector molecules such as GRA15, GRA16, or ROP16 that manipulate host cell signaling and transcriptional pathways, or deletion of PVM associated ROP21 or GRA3 molecules did not affect the antitumor activity. Association of ROP18 with the PVM was found to be essential for the development of the antitumor responses. Surprisingly, the ROP18 kinase activity required for resistance to IFN-γ activated host innate immunity related GTPases and virulence was not essential for the antitumor response. These data show that PVM functions of parasite secreted effector molecules, including ROP18, manipulate host cell responses through ROP18 kinase virulence independent mechanisms to activate potent antitumor responses. Our results demonstrate that PVM associated rhoptry effector proteins secreted prior to host cell invasion and dense granule effector proteins localized to the intravacuolar network and host nucleus that are secreted after host cell invasion coordinately control the development of host immune responses that provide effective antitumor immunity against established ovarian cancer.


Subject(s)
Cancer Vaccines/immunology , Immunity, Innate/genetics , Ovarian Neoplasms/immunology , Protein Serine-Threonine Kinases/immunology , Toxoplasma/immunology , Animals , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Cancer Vaccines/genetics , Dendritic Cells/immunology , Female , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/immunology , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Mice , Ovarian Neoplasms/prevention & control , Ovarian Neoplasms/therapy , Parasitic Diseases/immunology , Protein Serine-Threonine Kinases/genetics , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Signal Transduction , T-Lymphocytes/immunology , Toxoplasma/pathogenicity , Tumor Microenvironment/immunology , Uracil/metabolism , Virulence Factors/genetics , Virulence Factors/immunology
5.
Parasitol Res ; 118(6): 1899-1918, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30949853

ABSTRACT

After host cell invasion, Toxoplasma secretes a variety of dense granule proteins (GRA proteins) from its secretory dense granules, which are involved in the biogenesis of the parasitophorous vacuole (PV). TgGRA8I is predicted to contain proline-rich domains, which are structural features of some cytoskeleton-related proteins. In agreement with this observation, previous proteomic analyses revealed the presence of TgGRA8I in the Toxoplasma sub-pellicular cytoskeleton. In the present study, we show (1) by docking analyses that TgGRA8I may interact with both Toxoplasma ß-tubulin and actin; (2) by immunoelectron microscopy, proteomic, biochemical, and cellular approaches that TgGRA8I associates with sub-pellicular microtubules and actin at the parasite sub-pellicular cytoskeleton; (3) that type I parasites (RH strain) lacking the GRA8 gene (RHΔku80Δgra8) exhibit loss of conoid extrusion, diminished cell infection, and egress capabilities, and that these motility impairments were likely due to important alterations in their sub-pellicular cytoskeleton, in particular their sub-pellicular microtubules and meshwork. Parasites lacking the GRA4 gene (RHΔku80Δgra4) did not show modifications in the organization of the sub-pellicular cytoskeleton. Collectively, these results demonstrated that TgGRA8I is a dense granule protein that, besides its role in the formation of the PV, contributes to the organization of the parasite sub-pellicular cytoskeleton and motility. This is the first proline-rich protein described in the Toxoplasma cytoskeleton, which is a key organelle for both the parasite motility and the invasion process. Knowledge about the function of cytoskeleton components in Toxoplasma is fundamental to understand the motility process and the host cell invasion mechanism. Refining this knowledge should lead to the design of novel pharmacological strategies for the treatment against toxoplasmosis.


Subject(s)
Actins/metabolism , Antigens, Protozoan/metabolism , Cell Movement/genetics , Cytoskeleton/metabolism , Protozoan Proteins/metabolism , Toxoplasma/metabolism , Toxoplasma/pathogenicity , Tubulin/metabolism , Animals , Antigens, Protozoan/genetics , Biological Transport , Microscopy, Immunoelectron , Microtubules/metabolism , Molecular Docking Simulation , Proteomics , Protozoan Proteins/genetics , Secretory Vesicles/metabolism , Toxoplasma/genetics , Toxoplasmosis/parasitology , Toxoplasmosis/pathology , Vacuoles/parasitology
6.
Proc Natl Acad Sci U S A ; 111(17): 6437-42, 2014 Apr 29.
Article in English | MEDLINE | ID: mdl-24733931

ABSTRACT

Unlike most intracellular pathogens that gain access into host cells through endocytic pathways, Toxoplasma gondii initiates infection at the cell surface by active penetration through a moving junction and subsequent formation of a parasitophorous vacuole. Here, we describe a noncanonical pathway for T. gondii infection of macrophages, in which parasites are initially internalized through phagocytosis, and then actively invade from within a phagosomal compartment to form a parasitophorous vacuole. This phagosome to vacuole invasion (PTVI) pathway may represent an intermediary link between the endocytic and the penetrative routes for host cell entry by intracellular pathogens. The PTVI pathway is preferentially used by avirulent strains of T. gondii and confers an infectious advantage over virulent strains for macrophage tropism.


Subject(s)
Macrophages/parasitology , Phagosomes/parasitology , Toxoplasma/pathogenicity , Animals , Cell Line , Macrophages/pathology , Macrophages/ultrastructure , Mice , Mice, Inbred C57BL , Phagocytosis , Phagosomes/pathology , Phagosomes/ultrastructure , Toxoplasma/ultrastructure , Toxoplasmosis/parasitology , Toxoplasmosis/pathology , Tropism , Vacuoles/parasitology , Vacuoles/pathology , Vacuoles/ultrastructure
7.
Infect Immun ; 84(5): 1252-1254, 2016 05.
Article in English | MEDLINE | ID: mdl-26953326

ABSTRACT

Calcium ion signaling regulates central aspects of the biology controlling stage and life cycle transitions of apicomplexan parasites. In the current issue of Infection and Immunity, Long and coworkers (S. Long, Q. Wang, and L. D. Sibley, Infect Immun 84:1262-1273, 2016, http://dx.doi.org/10.1128/IAI.01173-15) describe a powerful genetic system enabling reliable serial genetic dissection of a large gene family encoding novel calcium-dependent protein kinases (CDPKs) that provides new insights into the roles of CDPKs during Toxoplasma gondii infection.


Subject(s)
Parasites , Toxoplasma/genetics , Animals , Life Cycle Stages
8.
Infect Immun ; 84(10): 2974-81, 2016 10.
Article in English | MEDLINE | ID: mdl-27481247

ABSTRACT

Dihydroorotate dehydrogenase (DHODH) mediates the fourth step of de novo pyrimidine biosynthesis and is a proven drug target for inducing immunosuppression in therapy of human disease as well as a rapidly emerging drug target for treatment of malaria. In Toxoplasma gondii, disruption of the first, fifth, or sixth step of de novo pyrimidine biosynthesis induced uracil auxotrophy. However, previous attempts to generate uracil auxotrophy by genetically deleting the mitochondrion-associated DHODH of T. gondii (TgDHODH) failed. To further address the essentiality of TgDHODH, mutant gene alleles deficient in TgDHODH activity were designed to ablate the enzyme activity. Replacement of the endogenous DHODH gene with catalytically deficient DHODH gene alleles induced uracil auxotrophy. Catalytically deficient TgDHODH localized to the mitochondria, and parasites retained mitochondrial membrane potential. These results show that TgDHODH is essential for the synthesis of pyrimidines and suggest that TgDHODH is required for a second essential function independent of its role in pyrimidine biosynthesis.


Subject(s)
Mitochondria/enzymology , Oxidoreductases Acting on CH-CH Group Donors/physiology , Pyrimidines/biosynthesis , Toxoplasma/enzymology , Toxoplasmosis/metabolism , Biosynthetic Pathways , Dihydroorotate Dehydrogenase , Fibroblasts/metabolism , Fibroblasts/parasitology , Gene Knockout Techniques , Humans , Oxidoreductases Acting on CH-CH Group Donors/genetics , Oxidoreductases Acting on CH-CH Group Donors/metabolism , Uracil/metabolism
9.
Infect Immun ; 83(5): 2148-55, 2015 May.
Article in English | MEDLINE | ID: mdl-25776745

ABSTRACT

Live attenuated vaccine strains, such as type I nonreplicating uracil auxotroph mutants, are highly effective in eliciting lifelong immunity to virulent acute infection by Toxoplasma gondii. However, it is currently unknown whether vaccine-elicited immunity can provide protection against acute infection and also prevent chronic infection. To address this problem, we developed nonreverting, nonreplicating, live attenuated uracil auxotroph vaccine strains in the type II Δku80 genetic background by targeting the deletion of the orotidine 5'-monophosphate decarboxylase (OMPDC) and uridine phosphorylase (UP) genes. Deletion of OMPDC induced a severe uracil auxotrophy with loss of replication, loss of virulence in mice, and loss of the ability to develop cysts and chronic infection. Vaccination of mice using type II Δku80 Δompdc mutants stimulated a fully protective CD8(+) T cell-dependent immunity that prevented acute infection by type I and type II strains of T. gondii, and this vaccination also severely reduced or prevented cyst formation after type II challenge infection. Nonreverting, nonreplicating, and non-cyst-forming Δompdc mutants provide new tools to examine protective immune responses elicited by vaccination with a live attenuated type II vaccine.


Subject(s)
Protozoan Vaccines/immunology , Toxoplasma/immunology , Toxoplasmosis/prevention & control , Animals , CD8-Positive T-Lymphocytes/immunology , Mice, Inbred C57BL , Mice, Knockout , Protozoan Vaccines/administration & dosage , Toxoplasmosis/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
10.
Infect Immun ; 83(10): 3865-80, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26195549

ABSTRACT

Toxoplasma gondii is an obligate intracellular protozoan parasite. This apicomplexan is the causative agent of toxoplasmosis, a leading cause of central nervous system disease in AIDS. It has long been known that T. gondii interferes with major histocompatibility complex class II (MHC-II) antigen presentation to attenuate CD4(+) T cell responses and establish persisting infections. Transcriptional downregulation of MHC-II genes by T. gondii was previously established, but the precise mechanisms inhibiting MHC-II function are currently unknown. Here, we show that, in addition to transcriptional regulation of MHC-II, the parasite modulates the expression of key components of the MHC-II antigen presentation pathway, namely, the MHC-II-associated invariant chain (Ii or CD74) and the peptide editor H2-DM, in professional antigen-presenting cells (pAPCs). Genetic deletion of CD74 restored the ability of infected dendritic cells to present a parasite antigen in the context of MHC-II in vitro. CD74 mRNA and protein levels were, surprisingly, elevated in infected cells, whereas MHC-II and H2-DM expression was inhibited. CD74 accumulated mainly in the endoplasmic reticulum (ER), and this phenotype required live parasites, but not active replication. Finally, we compared the impacts of genetic deletion of CD74 and H2-DM genes on parasite dissemination toward lymphoid organs in mice, as well as activation of CD4(+) T cells and interferon gamma (IFN-γ) levels during acute infection. Cyst burdens and survival during the chronic phase of infection were also evaluated in wild-type and knockout mice. These results highlight the fact that the infection is influenced by multiple levels of parasite manipulation of the MHC-II antigen presentation pathway.


Subject(s)
Antigens, Differentiation, B-Lymphocyte/immunology , Histocompatibility Antigens Class II/immunology , Toxoplasma/immunology , Toxoplasmosis/immunology , Animals , Antigen Presentation , Antigen-Presenting Cells/immunology , Antigens, Differentiation, B-Lymphocyte/genetics , CD4-Positive T-Lymphocytes , Dendritic Cells/immunology , Dendritic Cells/parasitology , Female , Histocompatibility Antigens Class II/genetics , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Macrophages/immunology , Macrophages/parasitology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Toxoplasma/genetics , Toxoplasma/physiology , Toxoplasmosis/genetics , Toxoplasmosis/parasitology
11.
PLoS Pathog ; 9(12): e1003823, 2013.
Article in English | MEDLINE | ID: mdl-24385904

ABSTRACT

Toxoplasma gondii infects up to one third of the world's population. A key to the success of T. gondii as a parasite is its ability to persist for the life of its host as bradyzoites within tissue cysts. The glycosylated cyst wall is the key structural feature that facilitates persistence and oral transmission of this parasite. Because most of the antibodies and reagents that recognize the cyst wall recognize carbohydrates, identification of the components of the cyst wall has been technically challenging. We have identified CST1 (TGME49_064660) as a 250 kDa SRS (SAG1 related sequence) domain protein with a large mucin-like domain. CST1 is responsible for the Dolichos biflorus Agglutinin (DBA) lectin binding characteristic of T. gondii cysts. Deletion of CST1 results in reduced cyst number and a fragile brain cyst phenotype characterized by a thinning and disruption of the underlying region of the cyst wall. These defects are reversed by complementation of CST1. Additional complementation experiments demonstrate that the CST1-mucin domain is necessary for the formation of a normal cyst wall structure, the ability of the cyst to resist mechanical stress, and binding of DBA to the cyst wall. RNA-seq transcriptome analysis demonstrated dysregulation of bradyzoite genes within the various cst1 mutants. These results indicate that CST1 functions as a key structural component that confers essential sturdiness to the T. gondii tissue cyst critical for persistence of bradyzoite forms.


Subject(s)
Cysts/genetics , Protozoan Proteins/physiology , Spores, Protozoan/genetics , Toxoplasma , Amino Acid Sequence , Antibodies, Monoclonal/metabolism , Cells, Cultured , Cysts/metabolism , Humans , Immune Evasion/genetics , Life Cycle Stages/genetics , Permeability , Spores, Protozoan/metabolism , Toxoplasma/genetics , Toxoplasma/growth & development , Toxoplasma/immunology , Toxoplasmosis/immunology , Toxoplasmosis/parasitology
12.
J Immunol ; 190(1): 469-78, 2013 Jan 01.
Article in English | MEDLINE | ID: mdl-23225891

ABSTRACT

Immune recognition of tumors can limit cancer development, but antitumor immune responses are often blocked by tumor-mediated immunosuppression. Because microbes or microbial constituents are powerful adjuvants to stimulate immune responses, we evaluated whether intratumoral administration of a highly immunogenic but attenuated parasite could induce rejection of an established poorly immunogenic tumor. We treated intradermal B16F10 murine melanoma by intratumoral injection of an attenuated strain of Toxoplasma gondii (cps) that cannot replicate in vivo and therefore is not infective. The cps treatment stimulated a strong CD8(+) T cell-mediated antitumor immune response in vivo that regressed established primary melanoma. The cps monotherapy rapidly modified the tumor microenvironment, halting tumor growth, and subsequently, as tumor-reactive T cells expanded, the tumors disappeared and rarely returned. The treatment required live cps that could invade cells and also required CD8(+) T cells and NK cells, but did not require CD4(+) T cells. Furthermore, we demonstrate that IL-12, IFN-γ, and the CXCR3-stimulating cytokines are required for full treatment efficacy. The treatment developed systemic antitumor immune activity as well as antitumor immune memory and therefore might have an impact against human metastatic disease. The approach is not specific for either B16F10 or melanoma. Direct intratumoral injection of cps has efficacy against an inducible genetic melanoma model and transplantable lung and ovarian tumors, demonstrating potential for broad clinical use. The combination of efficacy, systemic antitumor immune response, and complete attenuation with no observed host toxicity demonstrates the potential value of this novel cancer therapy.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Cancer Vaccines/administration & dosage , Melanoma, Experimental/immunology , Skin Neoplasms/immunology , Toxoplasma/immunology , Adjuvants, Immunologic/therapeutic use , Animals , Cancer Vaccines/immunology , Cell Line, Tumor , Injections, Intradermal , Melanoma, Experimental/prevention & control , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/prevention & control , Skin Neoplasms/prevention & control , Tumor Escape/immunology , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
14.
Vaccine ; 42(9): 2299-2309, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38429153

ABSTRACT

Toxoplasma gondii is a pervasive protozoan parasite that is responsible for significant zoonoses. A wide array of vaccines using different effector molecules of T. gondii have been studied worldwide to control toxoplasmosis. None of the existing vaccines are sufficiently effective to confer protective immunity. Among the different Toxoplasma-derived effector molecules, T. gondii dense granule protein 15 from the type II strain (GRA15 (II)) was recently characterized as an immunomodulatory molecule that induced host immunity via NF-κB. Therefore, we assessed the immunostimulatory and protective efficacy of recombinant GRA15 (II) (rGRA15) against T. gondii infection in a C57BL/6 mouse model. We observed that rGRA15 treatment increased the production of IL-12p40 from mouse peritoneal macrophages in vitro. Immunization of mice with rGRA15 induced the production of anti-TgGRA15-specific IgG, IgG1 and IgG2c antibodies. The rGRA15-sensitized spleen cells from mice inoculated with the same antigen strongly promoted spleen cell proliferation and IFN-γ production. Immunization with rGRA15 significantly enhanced the survival rate of mice and dramatically decreased parasite burden in mice challenged with the Pru (type II) strain. These results suggested that rGRA15 triggered humoral and cellular immune responses to control infection. However, all of the immunized mice died when challenged with the GRA15-deficient Pru strain or the RH (type I) strain. These results suggest that GRA15 (II)-dependent immunity plays a crucial role in protection against challenge infection with the type II strain of T. gondii. This study is the first report to show GRA15 (II) as a recombinant vaccine antigen against Toxoplasma infection.


Subject(s)
Protozoan Vaccines , Toxoplasma , Toxoplasmosis, Animal , Toxoplasmosis , Vaccines, DNA , Vaccines , Animals , Mice , Protozoan Proteins , Mice, Inbred C57BL , Toxoplasmosis/prevention & control , Recombinant Proteins/metabolism , Antibodies, Protozoan , Toxoplasmosis, Animal/prevention & control , Mice, Inbred BALB C
15.
Front Cell Infect Microbiol ; 14: 1359888, 2024.
Article in English | MEDLINE | ID: mdl-38828265

ABSTRACT

Toxoplasma, an important intracellular parasite of humans and animals, causes life-threatening toxoplasmosis in immunocompromised individuals. Although Toxoplasma secretory proteins during acute infection (tachyzoite, which divides rapidly and causes inflammation) have been extensively characterized, those involved in chronic infection (bradyzoite, which divides slowly and is surrounded by a cyst wall) remain uncertain. Regulation of the cyst wall is essential to the parasite life cycle, and polysaccharides, such as chitin, in the cyst wall are necessary to sustain latent infection. Toxoplasma secretory proteins during the bradyzoite stage may have important roles in regulating the cyst wall via polysaccharides. Here, we focused on characterizing the hypothetical T. gondii chitinase, chitinase-like protein 1 (TgCLP1). We found that the chitinase-like domain containing TgCLP1 is partially present in the bradyzoite microneme and confirmed, albeit partially, its previous identification in the tachyzoite microneme. Furthermore, although parasites lacking TgCLP1 could convert from tachyzoites to bradyzoites and make an intact cyst wall, they failed to convert from bradyzoites to tachyzoites, indicating that TgCLP1 is necessary for bradyzoite reactivation. Taken together, our findings deepen our understanding of the molecular basis of recrudescence and could contribute to the development of novel strategies for the control of toxoplasmosis.


Subject(s)
Chitinases , Protozoan Proteins , Toxoplasma , Toxoplasmosis , Animals , Humans , Mice , Chitinases/metabolism , Chitinases/genetics , Life Cycle Stages , Protozoan Proteins/metabolism , Protozoan Proteins/genetics , Toxoplasma/enzymology , Toxoplasma/genetics , Toxoplasma/growth & development , Toxoplasma/metabolism , Toxoplasmosis/parasitology
16.
iScience ; 27(1): 108477, 2024 Jan 19.
Article in English | MEDLINE | ID: mdl-38205261

ABSTRACT

Toxoplasma gondii causes morbidity, mortality, and disseminates widely via cat sexual stages. Here, we find T. gondii ornithine aminotransferase (OAT) is conserved across phyla. We solve TgO/GABA-AT structures with bound inactivators at 1.55 Å and identify an inactivator selective for TgO/GABA-AT over human OAT and GABA-AT. However, abrogating TgO/GABA-AT genetically does not diminish replication, virulence, cyst-formation, or eliminate cat's oocyst shedding. Increased sporozoite/merozoite TgO/GABA-AT expression led to our study of a mutagenized clone with oocyst formation blocked, arresting after forming male and female gametes, with "Rosetta stone"-like mutations in genes expressed in merozoites. Mutations are similar to those in organisms from plants to mammals, causing defects in conception and zygote formation, affecting merozoite capacitation, pH/ionicity/sodium-GABA concentrations, drawing attention to cyclic AMP/PKA, and genes enhancing energy or substrate formation in TgO/GABA-AT-related-pathways. These candidates potentially influence merozoite's capacity to make gametes that fuse to become zygotes, thereby contaminating environments and causing disease.

17.
PLoS Pathog ; 7(9): e1002236, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21931552

ABSTRACT

The ROP16 kinase of Toxoplasma gondii is injected into the host cell cytosol where it activates signal transducer and activator of transcription (STAT)-3 and STAT6. Here, we generated a ROP16 deletion mutant on a Type I parasite strain background, as well as a control complementation mutant with restored ROP16 expression. We investigated the biological role of the ROP16 molecule during T. gondii infection. Infection of mouse bone marrow-derived macrophages with rop16-deleted (ΔROP16) parasites resulted in increased amounts of IL-12p40 production relative to the ROP16-positive RH parental strain. High level IL-12p40 production in ΔROP16 infection was dependent on the host cell adaptor molecule MyD88, but surprisingly was independent of any previously recognized T. gondii triggered pathway linking to MyD88 (TLR2, TLR4, TLR9, TLR11, IL-1ß and IL-18). In addition, ROP16 was found to mediate the suppressive effects of Toxoplasma on LPS-induced cytokine synthesis in macrophages and on IFN-γ-induced nitric oxide production by astrocytes and microglial cells. Furthermore, ROP16 triggered synthesis of host cell arginase-1 in a STAT6-dependent manner. In fibroblasts and macrophages, failure to induce arginase-1 by ΔROP16 tachyzoites resulted in resistance to starvation conditions of limiting arginine, an essential amino acid for replication and virulence of this parasite. ΔROP16 tachyzoites that failed to induce host cell arginase-1 displayed increased replication and dissemination during in vivo infection. We conclude that encounter between Toxoplasma ROP16 and the host cell STAT signaling cascade has pleiotropic downstream effects that act in multiple and complex ways to direct the course of infection.


Subject(s)
Arginase/metabolism , Cytokines/immunology , Protein-Tyrosine Kinases/metabolism , Protozoan Proteins/metabolism , STAT3 Transcription Factor/metabolism , STAT6 Transcription Factor/metabolism , Toxoplasma/pathogenicity , Animals , Arginase/antagonists & inhibitors , Arginase/genetics , Cells, Cultured , Female , Gene Deletion , Gene Knockout Techniques , Interleukin-12 Subunit p40/immunology , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Macrophages/immunology , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/metabolism , Phosphorylation , Plasmids , Protein-Tyrosine Kinases/genetics , Protozoan Proteins/genetics , STAT3 Transcription Factor/genetics , STAT6 Transcription Factor/genetics , Signal Transduction , Toxoplasma/enzymology , Toxoplasma/genetics
18.
Front Immunol ; 14: 1272221, 2023.
Article in English | MEDLINE | ID: mdl-37868957

ABSTRACT

Toxoplasma gondii establishes chronic infection by forming tissue cysts, and this chronic infection is one of the most common parasitic infections in humans. Our recent studies revealed that whereas CD8+ T cells of genetically resistant BALB/c mice have the capability to remove the tissue cysts of the parasite through their perforin-mediated activities, small portions of the cysts are capable of persisting in the presence of the anti-cyst CD8+ T cells. It is currently unknown how those small portions of the cysts resist or escape the T-cell immunity and persist in the hosts. In the present study, we discovered that the cysts, which persisted in the presence of the perforin-mediated CD8+ T-cell immunity, have significantly greater mRNA levels for four dense granule proteins, GRA1, GRA2, GRA3, and GRA7, and one rhoptry protein, ROP35, than the total population of the cysts present in the absence of the T cells. In addition, increased levels of mRNA for GRA1, GRA3, and ROP35 in the cysts significantly correlated with their successful persistence through the condition in which greater degrees of reduction of the cyst burden occurred through anti-cyst CD8+ T cells. In addition, GRA3-deficient T. gondii displayed significantly enhanced elimination of the cysts by anti-cyst CD8+ T cells when compared to the wild-type parasite. These results indicate that GRA3 is a key molecule that mediates in the capability of T. gondii cysts to persist by resisting or evading the anti-cyst activity of CD8+ T cells during the later stage of infection.


Subject(s)
Parasites , Toxoplasma , Humans , Animals , Mice , CD8-Positive T-Lymphocytes , Protozoan Proteins/genetics , Perforin , Persistent Infection , RNA, Messenger
19.
Infect Immun ; 80(2): 476-82, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22104110

ABSTRACT

The intracellular protozoan Toxoplasma gondii is well known for its skill at invading and living within host cells. New discoveries are now also revealing the astounding ability of the parasite to inject effector proteins into the cytoplasm to seize control of the host cell. This review summarizes recent advances in our understanding of one such secretory protein called ROP16. This molecule is released from rhoptries into the host cell during invasion. The ROP16 molecule acts as a kinase, directly activating both signal transducer and activator of transcription 3 (STAT3) and STAT6 signaling pathways. In macrophages, an important and preferential target cell of parasite infection, the injection of ROP16 has multiple consequences, including downregulation of proinflammatory cytokine signaling and macrophage deviation to an alternatively activated phenotype.


Subject(s)
Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/physiology , Toxoplasma/physiology , Animals
20.
Eukaryot Cell ; 10(9): 1193-206, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21531875

ABSTRACT

Type II Toxoplasma gondii KU80 knockouts (Δku80) deficient in nonhomologous end joining were developed to delete the dominant pathway mediating random integration of targeting episomes. Gene targeting frequency in the type II Δku80 Δhxgprt strain measured at the orotate (OPRT) and the uracil (UPRT) phosphoribosyltransferase loci was highly efficient. To assess the potential of the type II Δku80 Δhxgprt strain to examine gene function affecting cyst biology and latent stages of infection, we targeted the deletion of four parasite antigen genes (GRA4, GRA6, ROP7, and tgd057) that encode characterized CD8(+) T cell epitopes that elicit corresponding antigen-specific CD8(+) T cell populations associated with control of infection. Cyst development in these type II mutant strains was not found to be strictly dependent on antigen-specific CD8(+) T cell host responses. In contrast, a significant biological role was revealed for the dense granule proteins GRA4 and GRA6 in cyst development since brain tissue cyst burdens were drastically reduced specifically in mutant strains with GRA4 and/or GRA6 deleted. Complementation of the Δgra4 and Δgra6 mutant strains using a functional allele of the deleted GRA coding region placed under the control of the endogenous UPRT locus was found to significantly restore brain cyst burdens. These results reveal that GRA proteins play a functional role in establishing cyst burdens and latent infection. Collectively, our results suggest that a type II Δku80 Δhxgprt genetic background enables a higher-throughput functional analysis of the parasite genome to reveal fundamental aspects of parasite biology controlling virulence, pathogenesis, and transmission.


Subject(s)
Antigens, Protozoan/genetics , Gene Deletion , Protozoan Proteins/genetics , Toxoplasma/genetics , Toxoplasma/metabolism , Toxoplasmosis, Animal/parasitology , Animals , Antigens, Protozoan/metabolism , CD8 Antigens/immunology , CD8 Antigens/metabolism , Communicable Diseases/microbiology , Gene Knockout Techniques , Gene Targeting , Mice , Mice, Inbred C57BL , Mice, Knockout , Protozoan Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL