Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Infect Immun ; 90(2): e0063821, 2022 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-34928716

RESUMO

Animals with a chronic infection of the parasite Toxoplasma gondii are protected against lethal secondary infection with other pathogens. Our group previously determined that soluble T. gondii antigens (STAg) can mimic this protection and be used as a treatment against several lethal pathogens. Because treatments are limited for the parasite Cryptosporidium parvum, we tested STAg as a C. parvum therapeutic. We determined that STAg treatment reduced C. parvum Iowa II oocyst shedding in gamma interferon knockout (IFN-γ-KO) mice. Murine intestinal sections were then sequenced to define the IFN-γ-independent transcriptomic response to C. parvum infection. Gene Ontology and transcript abundance comparisons showed host immune response and metabolism changes. Transcripts for type I interferon-responsive genes were more abundant in C. parvum-infected mice treated with STAg. Comparisons between phosphate-buffered saline (PBS) and STAg treatments showed no significant differences in C. parvum gene expression. C. parvum transcript abundance was highest in the ileum and mucin-like glycoproteins and the GDP-fucose transporter were among the most abundant. These results will assist the field in determining both host- and parasite-directed future therapeutic targets.


Assuntos
Criptosporidiose , Cryptosporidium parvum , Cryptosporidium , Animais , Cryptosporidium/genética , Imunidade , Interferon gama , Camundongos , Camundongos Endogâmicos C57BL , Transcriptoma
2.
PLoS Pathog ; 16(4): e1008432, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32255806

RESUMO

The obligate intracellular parasite Toxoplasma gondii is auxotrophic for several key metabolites and must scavenge these from the host. It is unclear how T. gondii manipulates host metabolism to support its overall growth rate and non-essential metabolites. To investigate this question, we measured changes in the joint host-parasite metabolome over a time course of infection. Host and parasite transcriptomes were simultaneously generated to determine potential changes in expression of metabolic enzymes. T. gondii infection changed metabolite abundance in multiple metabolic pathways, including the tricarboxylic acid cycle, the pentose phosphate pathway, glycolysis, amino acid synthesis, and nucleotide metabolism. Our analysis indicated that changes in some pathways, such as the tricarboxylic acid cycle, were mirrored by changes in parasite transcription, while changes in others, like the pentose phosphate pathway, were paired with changes in both the host and parasite transcriptomes. Further experiments led to the discovery of a T. gondii enzyme, sedoheptulose bisphosphatase, which funnels carbon from glycolysis into the pentose phosphate pathway through an energetically driven dephosphorylation reaction. This additional route for ribose synthesis appears to resolve the conflict between the T. gondii tricarboxylic acid cycle and pentose phosphate pathway, which are both NADP+ dependent. Sedoheptulose bisphosphatase represents a novel step in T. gondii central carbon metabolism that allows T. gondii to energetically-drive ribose synthesis without using NADP+.


Assuntos
Toxoplasma/metabolismo , Toxoplasmose/metabolismo , Toxoplasmose/parasitologia , Aminoácidos/biossíntese , Ciclo do Ácido Cítrico , Glicólise , Interações Hospedeiro-Parasita , Humanos , Metaboloma , Metabolômica , NADP/metabolismo , Via de Pentose Fosfato , Ribose/biossíntese , Toxoplasma/genética
3.
PLoS Pathog ; 16(7): e1008650, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32628723

RESUMO

Toxoplasma gondii is an obligate intracellular parasite that can invade any nucleated cell of any warm-blooded animal. In a previous screen to identify virulence determinants, disruption of gene TgME49_305140 generated a T. gondii mutant that could not establish a chronic infection in mice. The protein product of TgME49_305140, here named TgPL3, is a 277 kDa protein with a patatin-like phospholipase (PLP) domain and a microtubule binding domain. Antibodies generated against TgPL3 show that it is localized to the apical cap. Using a rapid selection FACS-based CRISPR/Cas-9 method, a TgPL3 deletion strain (ΔTgPL3) was generated. ΔTgPL3 parasites have defects in host cell invasion, which may be caused by reduced rhoptry secretion. We generated complementation clones with either wild type TgPL3 or an active site mutation in the PLP domain by converting the catalytic serine to an alanine, ΔTgPL3::TgPL3S1409A (S1409A). Complementation of ΔTgPL3 with wild type TgPL3 restored all phenotypes, while S1409A did not, suggesting that phospholipase activity is necessary for these phenotypes. ΔTgPL3 and S1409A parasites are also virtually avirulent in vivo but induce a robust antibody response. Vaccination with ΔTgPL3 and S1409A parasites protected mice against subsequent challenge with a lethal dose of Type I T. gondii parasites, making ΔTgPL3 a compelling vaccine candidate. These results demonstrate that TgPL3 has a role in rhoptry secretion, host cell invasion and survival of T. gondii during acute mouse infection.


Assuntos
Proteínas de Protozoários/metabolismo , Toxoplasma/patogenicidade , Toxoplasmose/metabolismo , Fatores de Virulência/metabolismo , Animais , Camundongos , Fosfolipases/genética , Fosfolipases/metabolismo , Proteínas de Protozoários/genética , Toxoplasma/genética , Toxoplasmose/enzimologia , Virulência
4.
PLoS Biol ; 17(8): e3000364, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31430281

RESUMO

Many eukaryotic microbes have complex life cycles that include both sexual and asexual phases with strict species specificity. Whereas the asexual cycle of the protistan parasite Toxoplasma gondii can occur in any warm-blooded mammal, the sexual cycle is restricted to the feline intestine. The molecular determinants that identify cats as the definitive host for T. gondii are unknown. Here, we defined the mechanism of species specificity for T. gondii sexual development and break the species barrier to allow the sexual cycle to occur in mice. We determined that T. gondii sexual development occurs when cultured feline intestinal epithelial cells are supplemented with linoleic acid. Felines are the only mammals that lack delta-6-desaturase activity in their intestines, which is required for linoleic acid metabolism, resulting in systemic excess of linoleic acid. We found that inhibition of murine delta-6-desaturase and supplementation of their diet with linoleic acid allowed T. gondii sexual development in mice. This mechanism of species specificity is the first defined for a parasite sexual cycle. This work highlights how host diet and metabolism shape coevolution with microbes. The key to unlocking the species boundaries for other eukaryotic microbes may also rely on the lipid composition of their environments as we see increasing evidence for the importance of host lipid metabolism during parasitic lifecycles. Pregnant women are advised against handling cat litter, as maternal infection with T. gondii can be transmitted to the fetus with potentially lethal outcomes. Knowing the molecular components that create a conducive environment for T. gondii sexual reproduction will allow for development of therapeutics that prevent shedding of T. gondii parasites. Finally, given the current reliance on companion animals to study T. gondii sexual development, this work will allow the T. gondii field to use of alternative models in future studies.


Assuntos
Linoleoil-CoA Desaturase/metabolismo , Toxoplasma/enzimologia , Animais , Gatos , Especificidade de Hospedeiro , Interações Hospedeiro-Parasita , Intestinos/parasitologia , Estágios do Ciclo de Vida/fisiologia , Ácido Linoleico/farmacologia , Camundongos , Camundongos Endogâmicos C57BL , Parasitos/metabolismo , Desenvolvimento Sexual/fisiologia , Especificidade da Espécie , Toxoplasma/crescimento & desenvolvimento , Toxoplasma/patogenicidade
5.
Infect Immun ; 89(12): e0050821, 2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34543124

RESUMO

Pancreatic ductal adenocarcinoma is the fourth leading cause of cancer-related death in the United States, with few effective treatments available and only 10% of those diagnosed surviving 5 years. Although immunotherapeutics is a growing field of study in cancer biology, there has been little progress in its use for the treatment of pancreatic cancer. Pancreatic cancer is considered a nonimmunogenic tumor because the tumor microenvironment does not easily allow for the immune system, even when stimulated, to attack the cancer. Infection with the protozoan parasite Toxoplasma gondii has been shown to enhance the immune response to clear cancer tumors. A subset of T. gondii proteins called soluble Toxoplasma antigen (STAg) contains an immunodominant protein called profilin. Both STAg and profilin have been shown to stimulate an immune response that reduces viral, bacterial, and parasitic burdens. Here, we use STAg and profilin to treat pancreatic cancer in a KPC mouse-derived allograft murine model. These mice exhibit pancreatic cancer with both Kras and P53 mutations as subcutaneous tumors. Pancreatic cancer tumors in C57BL/6J mice with a wild-type background showed a significant response to treatment with either profilin or STAg, exhibiting a decrease in tumor volume accompanied by an influx of CD4+ and CD8+ T cells into the tumors. Both IFN-γ-/- mice and Batf3-/- mice, which lack conventional dendritic cells, failed to show significant decreases in tumor volumes when treated. These results indicate that gamma interferon (IFN-γ) and dendritic cells may play critical roles in the immune response necessary to treat pancreatic cancer.


Assuntos
Antineoplásicos/farmacologia , Produtos Biológicos/farmacologia , Proteínas de Protozoários/farmacologia , Toxoplasma , Aloenxertos , Animais , Antígenos de Protozoários/imunologia , Antígenos de Protozoários/farmacologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Camundongos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Proteínas de Protozoários/imunologia , Toxoplasma/química , Toxoplasma/metabolismo
6.
Infect Immun ; 89(5)2021 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-33526566

RESUMO

Toxoplasma gondii infection activates pattern recognition receptor (PRR) pathways that drive innate inflammatory responses to control infection. Necroptosis is a proinflammatory cell death pathway apart from the innate immune response that has evolved to control pathogenic infection. In this study, we further defined the role of Z-DNA binding protein 1 (ZBP1) as a PRR and assessed its contribution to necroptosis as a host protection mechanism to T. gondii infection. We found that ZBP1 does not induce proinflammatory necroptosis cell death, and ZBP1 null mice have reduced survival after oral T. gondii infection. In contrast, mice deleted in receptor-interacting serine/threonine-protein kinase 3 (RIPK3-/-), a central mediator of necroptosis, have significantly improved survival after oral T. gondii infection without a reduction in parasite burden. The physiological consequences of RIPK3 activity did not show any differences in intestine villus immunopathology, but RIPK3-/- mice showed higher immune cell infiltration and edema in the lamina propria. The contribution of necroptosis to host survival was clarified with mixed-lineage kinase domain-like pseudokinase null (MLKL-/-) mice. We found MLKL-/- mice succumbed to oral T. gondii infection the same as wild-type mice, indicating necroptosis-independent RIPK3 activity impacts host survival. These results provide new insights on the impacts of proinflammatory cell death pathways as a mechanism of host defense to oral T. gondii infection.


Assuntos
Resistência à Doença , Interações Hospedeiro-Parasita , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Transdução de Sinais , Toxoplasmose/metabolismo , Toxoplasmose/parasitologia , Animais , Modelos Animais de Doenças , Resistência à Doença/genética , Resistência à Doença/imunologia , Interações Hospedeiro-Parasita/imunologia , Camundongos , Camundongos Knockout , Necroptose , Proteínas de Ligação a RNA , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Toxoplasmose/genética , Toxoplasmose/imunologia
7.
J Immunol ; 200(11): 3729-3738, 2018 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-29678951

RESUMO

Nonsteroidal anti-inflammatory drugs (NSAIDs) inhibit cyclooxygenase (COX) activity and are commonly used for pain relief and fever reduction. NSAIDs are used following childhood vaccinations and cancer immunotherapies; however, how NSAIDs influence the development of immunity following these therapies is unknown. We hypothesized that NSAIDs would modulate the development of an immune response to Listeria monocytogenes-based immunotherapy. Treatment of mice with the nonspecific COX inhibitor indomethacin impaired the generation of cell-mediated immunity. This phenotype was due to inhibition of the inducible COX-2 enzyme, as treatment with the COX-2-selective inhibitor celecoxib similarly inhibited the development of immunity. In contrast, loss of COX-1 activity improved immunity to L. monocytogenes Impairments in immunity were independent of bacterial burden, dendritic cell costimulation, or innate immune cell infiltrate. Instead, we observed that PGE2 production following L. monocytogenes is critical for the formation of an Ag-specific CD8+ T cell response. Use of the alternative analgesic acetaminophen did not impair immunity. Taken together, our results suggest that COX-2 is necessary for optimal CD8+ T cell responses to L. monocytogenes, whereas COX-1 is detrimental. Use of pharmacotherapies that spare COX-2 activity and the production of PGE2 like acetaminophen will be critical for the generation of optimal antitumor responses using L. monocytogenes.


Assuntos
Ciclo-Oxigenase 1/imunologia , Ciclo-Oxigenase 2/imunologia , Imunidade/imunologia , Listeria monocytogenes/imunologia , Proteínas de Membrana/imunologia , Acetaminofen/farmacologia , Animais , Anti-Inflamatórios não Esteroides/imunologia , Anti-Inflamatórios não Esteroides/farmacologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Inibidores de Ciclo-Oxigenase 2/farmacologia , Dinoprostona/imunologia , Feminino , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Listeriose/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
8.
Infect Immun ; 87(5)2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30858341

RESUMO

The long-term host effects caused by the protozoan parasite Toxoplasma gondii are poorly understood. High-throughput RNA sequencing analysis previously determined that the host response in the brain was greater and more complex at 28 days than at 10 days postinfection. Here, we analyzed the host transcriptional profile of age- and sex-matched mice during very early (21 days), early (28 days), mid (3 months), and late (6 months) chronic infection. We found that a majority of the host genes which increase in abundance at day 21 postinfection are still increased at 6 months postinfection for both male and female mice. While most of the differentially expressed genes were similar between sexes, females had far fewer genes that were significantly less abundant, which may have led to the slightly increased cyst burden in males. Transcripts for C-X-C motif chemokine ligand 13 and a C-C motif chemokine receptor 2 (CCR2) were significantly higher in females than in males during infection. As T. gondii chronic infection and profilin (PRF) confer resistance to Listeria monocytogenes infection in a CCR2-dependent manner, the differences in CCR2 expression led us to retest the protection of PRF in both sexes. Male mice were nearly as effective as female mice at reducing the bacterial burden either with a chronic infection or when treated with PRF. These data show that most of the host genes differentially expressed in response to T. gondii infection are similar between males and females. While differences in transcript abundance exist between the sexes, the infection phenotypes tested here did not show significant differences.


Assuntos
Interações Hospedeiro-Parasita/genética , Interações Hospedeiro-Parasita/imunologia , Toxoplasma/genética , Toxoplasma/imunologia , Toxoplasmose/genética , Toxoplasmose/imunologia , Animais , Doença Crônica , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos CBA , Fatores Sexuais , Fatores de Tempo
9.
BMC Genomics ; 20(1): 859, 2019 Nov 14.
Artigo em Inglês | MEDLINE | ID: mdl-31726967

RESUMO

BACKGROUND: The protozoan pathogen Toxoplasma gondii has the unique ability to develop a chronic infection in the brain of its host by transitioning from the fast growing tachyzoite morphology to latent bradyzoite morphology. A hallmark of the bradyzoite is the development of neuronal cysts that are resilient against host immune response and current therapeutics. The bradyzoite parasites within the cyst have a carbohydrate and protein-rich wall and a slow-replication cycle, allowing them to remain hidden from the host. The intracellular, encysted lifestyle of T. gondii has made them recalcitrant to molecular analysis in vivo. RESULTS: Here, we detail the results from transcriptional and proteomic analyses of bradyzoite-enriched fractions isolated from mouse brains infected with T. gondii over a time course of 21 to 150 days. The enrichment procedure afforded consistent identification of over 2000 parasitic peptides from the mixed-organism sample, representing 366 T. gondii proteins at 28, 90, and 120 day timepoints. Deep sequencing of transcripts expressed during these three timepoints revealed that a subpopulation of genes that are transcriptionally expressed at a high level. Approximately one-third of these transcripts are more enriched during bradyzoite conditions compared to tachyzoites and approximately half are expressed at similar levels during each phase. The T. gondii transcript which increased the most over the course of chronic infection, sporoAMA1, shows stage specific isoform expression of the gene. CONCLUSIONS: We have expanded the transcriptional profile of in vivo bradyzoites to 120 days post-infection and provided the first in vivo proteomic profile of T. gondii bradyzoites. The RNA sequencing depth of in vivo bradyzoite T. gondii was over 250-fold greater than previous reports and allowed us to identify low level transcripts and a novel bradyzoite-specific isoform of sporoAMA1.


Assuntos
Proteoma , Toxoplasma/genética , Toxoplasma/metabolismo , Toxoplasmose/parasitologia , Transcriptoma , Animais , Biologia Computacional/métodos , Perfilação da Expressão Gênica/métodos , Regulação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Estágios do Ciclo de Vida/genética , Camundongos , Proteômica/métodos , Toxoplasma/crescimento & desenvolvimento , Fluxo de Trabalho
10.
Mol Microbiol ; 107(1): 34-46, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29090840

RESUMO

Emerging lipidomic technologies have enabled researchers to dissect the complex roles of phospholipases in lipid metabolism, cellular signaling and immune regulation. Host phospholipase products are involved in stimulating and resolving the inflammatory response to pathogens. While many pathogen-derived phospholipases also manipulate the immune response, they have recently been shown to be involved in lipid remodeling and scavenging during replication. Animal and plant hosts as well as many pathogens contain a family of patatin-like phospholipases, which have been shown to have phospholipase A2 activity. Proteins containing patatin-like phospholipase domains have been identified in protozoan parasites within the Apicomplexa phylum. These parasites are the causative agents of some of the most widespread human diseases. Malaria, caused by Plasmodium spp., kills nearly half a million people worldwide each year. Toxoplasma and Cryptosporidium infect millions of people each year with lethal consequences in immunocompromised populations. Parasite-derived patatin-like phospholipases are likely effective drug targets and progress in the tools available to the Apicomplexan field will allow for a closer look at the interplay of lipid metabolism and immune regulation during host infection.


Assuntos
Metabolismo dos Lipídeos/fisiologia , Fosfolipases/metabolismo , Fosfolipases/fisiologia , Sequência de Aminoácidos , Animais , Antígenos de Plaquetas Humanas/imunologia , Antígenos de Plaquetas Humanas/metabolismo , Apicomplexa/imunologia , Apicomplexa/metabolismo , Ácidos Graxos/metabolismo , Humanos , Inflamação/metabolismo , Lipase/metabolismo , Lipídeos , Parasitos/metabolismo , Parasitos/parasitologia , Fosfolipases/imunologia
13.
Mol Microbiol ; 102(6): 1086-1098, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27671212

RESUMO

The Toxoplasma gondii cyst stage is resistant to drug therapy. To identify potential targets for new therapeutics, we screened insertional mutants of T. gondii for a reduced ability to form cysts in the brains of mice. In one of these mutants, named 38C3, the mutagenesis plasmid inserted into the mRNA of a protein that is highly conserved in microbes but is not present in humans. The mutation in 38C3 causes reduced brain cyst production during chronic infection, but does not affect acute virulence, so the disrupted gene and protein are called T. gondii Brain Colonization Protein 1 (TgBCP1). TgBCP1 has three potential in frame start codons that produce 51, 33 or 25 kDa proteins. In rapidly replicating tachyzoites, translation initiates at the third methionine, producing the 25 kDa form that is conserved in many bacteria and protozoans. Brain cysts exclusively express the 51 kDa form of TgBCP1, which is secreted from the parasites and localizes to the cyst wall. Only expression of the long form of TgBCP1 restored cyst formation in the 38C3 mutant. TgBCP1 is essential for cyst formation and is the first example of a developmental regulation in translation initiation site preference for a T. gondii protein.


Assuntos
Toxoplasma/metabolismo , Animais , Encéfalo/parasitologia , Genes de Protozoários , Teste de Complementação Genética , Camundongos , Camundongos Endogâmicos C57BL , Mutagênese Insercional , Iniciação Traducional da Cadeia Peptídica/fisiologia , Biossíntese de Proteínas , Proteínas de Protozoários/metabolismo , RNA Mensageiro/metabolismo , Toxoplasma/parasitologia , Toxoplasmose Animal/parasitologia , Virulência
15.
Infect Immun ; 84(10): 3063-70, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27481249

RESUMO

Intrinsic to Toxoplasma gondii infection is the parasite-induced modulation of the host immune response, which ensures establishment of a chronic lifelong infection. This manipulation of the host immune response allows T. gondii to not only dampen the ability of the host to eliminate the parasite but also trigger parasite differentiation to the slow-growing, encysted bradyzoite form. We previously used RNA sequencing (RNA-seq) to profile the transcriptomes of mice and T. gondii during acute and chronic stages of infection. One of the most abundant host transcripts during acute and chronic infection was Z-DNA binding protein 1 (ZBP1). In this study, we determined that ZBP1 functions to control T. gondii growth. In activated macrophages isolated from ZBP1 deletion (ZBP1(-/-)) mice, T. gondii has an increased rate of replication and a decreased rate of degradation. We also identified a novel function for ZBP1 as a regulator of nitric oxide (NO) production in activated macrophages, even in the absence of T. gondii infection. Upon stimulation, T. gondii-infected ZBP1(-/-) macrophages display increased proinflammatory cytokines compared to wild-type macrophages under the same conditions. These in vitro phenotypes were recapitulated in vivo, with ZBP1(-/-) mice having increased susceptibility to oral challenge, higher cyst burdens during chronic infection, and elevated inflammatory cytokine responses. Taken together, these results highlight a role for ZBP1 in assisting host control of T. gondii infection.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Interações Hospedeiro-Parasita/fisiologia , Toxoplasma/patogenicidade , Toxoplasmose Animal/microbiologia , Doença Aguda , Animais , Doença Crônica , Citocinas/metabolismo , DNA Forma Z , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Macrófagos/metabolismo , Macrófagos/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Óxido Nítrico/metabolismo , Análise de Sequência de RNA , Toxoplasma/crescimento & desenvolvimento , Toxoplasmose Animal/imunologia
16.
PLoS Pathog ; 10(6): e1004203, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24945711

RESUMO

Ly6C+ inflammatory monocytes are essential to host defense against Toxoplasma gondii, Listeria monocytogenes and other infections. During T. gondii infection impaired inflammatory monocyte emigration results in severe inflammation and failure to control parasite replication. However, the T. gondii factors that elicit these monocytes are unknown. Early studies from the Remington laboratory showed that mice with a chronic T. gondii infection survive lethal co-infections with unrelated pathogens, including L. monocytogenes, but a mechanistic analysis was not performed. Here we report that this enhanced survival against L. monocytogenes is due to early reduction of bacterial burdens and elicitation of Ly6C+ inflammatory monocytes. We demonstrate that a single TLR11/TLR12 ligand profilin (TgPRF) was sufficient to reduce bacterial burdens similar to T. gondii chronic infection. Stimulation with TgPRF was also sufficient to enhance animal survival when administered either pre- or post-Listeria infection. The ability of TgPRF to reduce L. monocytogenes burdens was dependent on TLR11 and required IFN-γ but was not dependent on IL-12 signaling. TgPRF induced rapid production of MCP-1 and resulted in trafficking of Ly6Chi CCR2+ inflammatory monocytes and Ly6G+ neutrophils into the blood and spleen. Stimulation with TgPRF reduced L. monocytogenes burdens in mice depleted with the Ly6G specific MAb 1A8, but not in Ly6C/Ly6G specific RB6-8C5 depleted or CCR2-/- mice, indicating that only inflammatory monocytes are required for TgPRF-induced reduction in bacterial burdens. These results demonstrate that stimulation of TLR11 by TgPRF is a mechanism to promote the emigration of Ly6Chi CCR2+ monocytes, and that TgPRF recruited inflammatory monocytes can provide an immunological benefit against an unrelated pathogen.


Assuntos
Listeria monocytogenes/imunologia , Listeriose/imunologia , Monócitos/imunologia , Neutrófilos/imunologia , Toxoplasma/imunologia , Animais , Antígenos Ly/imunologia , Quimiocina CCL2/biossíntese , Coinfecção/imunologia , Coinfecção/microbiologia , Coinfecção/parasitologia , Inflamação/imunologia , Interferon gama/biossíntese , Interleucina-12/biossíntese , Listeriose/microbiologia , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Camundongos Knockout , Profilinas/genética , Receptores CCR2/imunologia , Receptores de Interleucina-12/imunologia , Proteínas Recombinantes , Transdução de Sinais/imunologia , Receptores Toll-Like/imunologia , Toxoplasmose Animal/imunologia , Toxoplasmose Animal/parasitologia , Fator de Necrose Tumoral alfa/biossíntese
18.
Traffic ; 14(5): 519-31, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23356236

RESUMO

Apicomplexan parasites, such as Toxoplasma gondii and Plasmodium, secrete proteins for attachment, invasion and modulation of their host cells. The host targeting (HT), also known as the Plasmodium export element (PEXEL), directs Plasmodium proteins into erythrocytes to remodel the host cell and establish infection. Bioinformatic analysis of Toxoplasma revealed a HT/PEXEL-like motif at the N-terminus of several hypothetical unknown and dense granule proteins. Hemagglutinin-tagged versions of these uncharacterized proteins show co-localization with dense granule proteins found on the parasitophorous vacuole membrane (PVM). In contrast to Plasmodium, these Toxoplasma HT/PEXEL containing proteins are not exported into the host cell. Site directed mutagenesis of the Toxoplasma HT/PEXEL motif, RxLxD/E, shows that the arginine and leucine residues are permissible for protein cleavage. Mutations within the HT/PEXEL motif that prevent protein cleavage still allow for targeting to the PV but the proteins have a reduced association with the PVM. Addition of a Myc tag before and after the cleavage site shows that processed HT/PEXEL protein has increased PVM association. These findings suggest that while Toxoplasma and Plasmodium share similar HT/PEXEL motifs, Toxoplasma HT/PEXEL containing proteins interact with but do not cross the PVM.


Assuntos
Antígenos de Protozoários , Proteínas de Protozoários/química , Toxoplasma/metabolismo , Algoritmos , Motivos de Aminoácidos , Animais , Biologia Computacional , Detergentes/farmacologia , Fibroblastos/parasitologia , Hemaglutininas/química , Humanos , Microscopia de Fluorescência , Mutagênese Sítio-Dirigida , Octoxinol , Plasmídeos/metabolismo , Polietilenoglicóis/farmacologia , Ligação Proteica , Isoformas de Proteínas/química , Estrutura Terciária de Proteína , Transporte Proteico , Proteínas Proto-Oncogênicas c-myc/química , Vacúolos/metabolismo
20.
Infect Immun ; 82(3): 1343-53, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24396042

RESUMO

A chronic infection with the parasite Toxoplasma gondii has previously been shown to protect mice against subsequent viral, bacterial, or protozoal infections. Here we have shown that a chronic T. gondii infection can prevent Plasmodium berghei ANKA-induced experimental cerebral malaria (ECM) in C57BL/6 mice. Treatment with soluble T. gondii antigens (STAg) reduced parasite sequestration and T cell infiltration in the brains of P. berghei-infected mice. Administration of STAg also preserved blood-brain barrier function, reduced ECM symptoms, and significantly decreased mortality. STAg treatment 24 h post-P. berghei infection led to a rapid increase in serum levels of interleukin 12 (IL-12) and gamma interferon (IFN-γ). By 5 days after P. berghei infection, STAg-treated mice had reduced IFN-γ levels compared to those of mock-treated mice, suggesting that reductions in IFN-γ at the time of ECM onset protected against lethality. Using IL-10- and IL-12ßR-deficient mice, we found that STAg-induced protection from ECM is IL-10 independent but IL-12 dependent. Treatment of P. berghei-infected mice with recombinant IL-12 significantly decreased parasitemia and mortality. These data suggest that IL-12, either induced by STAg or injected as a recombinant protein, mediates protection from ECM-associated pathology potentially through early induction of IFN-γ and reduction in parasitemia. These results highlight the importance of early IL-12 induction in protection against ECM.


Assuntos
Interleucina-12/imunologia , Malária Cerebral/imunologia , Plasmodium berghei/imunologia , Toxoplasma/imunologia , Regulação para Cima/imunologia , Animais , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/microbiologia , Encéfalo/imunologia , Encéfalo/microbiologia , Células CHO , Cricetulus , Interferon gama/imunologia , Interleucina-10/imunologia , Malária Cerebral/microbiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Parasitemia/imunologia , Parasitemia/microbiologia , Receptores de Interleucina-11/imunologia , Linfócitos T/imunologia , Linfócitos T/microbiologia , Toxoplasmose/imunologia , Toxoplasmose/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA