15 research outputs found

    Determinants of GBP Recruitment to Toxoplasma gondii Vacuoles and the Parasitic Factors That Control It

    Get PDF
    IFN-γ is a major cytokine that mediates resistance against the intracellular parasite Toxoplasma gondii. The p65 guanylate-binding proteins (GBPs) are strongly induced by IFN-γ. We studied the behavior of murine GBP1 (mGBP1) upon infection with T. gondii in vitro and confirmed that IFN-γ-dependent re-localization of mGBP1 to the parasitophorous vacuole (PV) correlates with the virulence type of the parasite. We identified three parasitic factors, ROP16, ROP18, and GRA15 that determine strain-specific accumulation of mGBP1 on the PV. These highly polymorphic proteins are held responsible for a large part of the strain-specific differences in virulence. Therefore, our data suggest that virulence of T. gondii in animals may rely in part on recognition by GBPs. However, phagosomes or vacuoles containing Trypanosoma cruzi did not recruit mGBP1. Co-immunoprecipitation revealed mGBP2, mGBP4, and mGBP5 as binding partners of mGBP1. Indeed, mGBP2 and mGBP5 co-localize with mGBP1 in T. gondii-infected cells. T. gondii thus elicits a cell-autonomous immune response in mice with GBPs involved. Three parasitic virulence factors and unknown IFN-γ-dependent host factors regulate this complex process. Depending on the virulence of the strains involved, numerous GBPs are brought to the PV as part of a large, multimeric structure to combat T. gondii.National Institutes of Health (U.S.)Massachusetts Life Sciences Center (New Investigator Award)National Institute of General Medical Sciences (U.S.) (Pre-Doctoral Grant in the Biological Sciences (5-T32-GM007287-33))Studienstiftung des deutschen VolkesCancer Research Institute (New York, N.Y.)Cleo and Paul Schimmel FoundationBayer HealthcareHuman Frontier Science Program (Strasbourg, France

    Preferential Brain Homing following Intranasal Administration of Trypanosoma cruzi▿

    No full text
    The Chagas’ disease parasite Trypanosoma cruzi commonly infects humans through skin abrasions or mucosa from reduviid bug excreta. Yet most studies on animal models start with subcutaneous or intraperitoneal injections, a distant approximation of the skin abrasion route. We show here that atraumatic placement of T. cruzi in the mouse nasal cavity produced low parasitemia, high survival rates, and preferential brain invasion compared to the case with subcutaneously injected parasites. Brain invasion was particularly prominent in the basal ganglia, peaked at a time when parasitemia was no longer detectable, and elicited a relatively large number of inflammatory foci. Yet, based on motor behavioral parameters and staining with Fluoro-Jade C, a dye that specifically recognizes apoptotic and necrotic neurons, brain invasion did not cause neurodegenerative events, in contrast to the neurodegeneration in the enteric nervous system. The results indicate that placement of T. cruzi on the mucosa in the mouse nasal cavity establishes a systemic infection with a robust yet harmless infection of the brain, seemingly analogous to disease progression in humans. The model may facilitate studies designed to understand mechanisms underlying T. cruzi infection of the central nervous system

    Individual islet respirometry reveals functional diversity within the islet population of mice and human donors

    No full text
    Objective: Islets from the same pancreas show remarkable variability in glucose sensitivity. While mitochondrial respiration is essential for glucose-stimulated insulin secretion, little is known regarding heterogeneity in mitochondrial function at the individual islet level. This is due in part to a lack of high-throughput and non-invasive methods for detecting single islet function. Methods: We have developed a novel non-invasive, high-throughput methodology capable of assessing mitochondrial respiration in large-sized individual islets using the XF96 analyzer (Agilent Technologies). Results: By increasing measurement sensitivity, we have reduced the minimal size of mouse and human islets needed to assess mitochondrial respiration to single large islets of >35,000 μm2 area (∼210 μm diameter). In addition, we have measured heterogeneous glucose-stimulated mitochondrial respiration among individual human and mouse islets from the same pancreas, allowing population analyses of islet mitochondrial function for the first time. Conclusions: We have developed a novel methodology capable of analyzing mitochondrial function in large-sized individual islets. By highlighting islet functional heterogeneity, we hope this methodology can significantly advance islet research. Keywords: Islets, Mitochondria, Respirometry, Glucos

    Dynamic host response signatures in <i>T</i>. <i>cruzi</i>-infected human fibroblasts.

    No full text
    <p>Expression patterns for selected genes in the most strongly modulated pathways in <i>T</i>. <i>cruzi</i> infected HFF. Genes in the following categories are highlighted. <b>(C) Mitotic Cell cycle:</b> <i>AURKA</i> (ENSG00000087586); <i>CDC6</i> (ENSG00000094804); <i>CDC20</i> (ENSG00000117399); <i>CDC25A</i> (ENSG00000164045); <i>CDK1</i> (ENSG00000170312); <i>CCNA2</i> (ENSG00000145386); <i>NEK2</i> (ENSG00000117650); <i>ORC1L</i> (ENSG00000085840); <i>PLK1</i> ENSG00000137807); KIF23 (ENSG00000137807). <b>(B) Cytokines/Chemokines:</b> <i>IL-8</i> (ENSG00000169429); <i>IL-6</i> (ENSG00000136244); <i>CCL8</i> (ENSG00000108700); <i>CCL2</i> (ENSG00000108691); <i>CXCL10</i> (ENSG00000169245). <b>(C) Type I Interferon:</b> <i>OAS</i> (ENSG00000089127); <i>IFNB</i> (ENSG00000171855); <i>ISG44</i> (ENSG00000137959); <i>GBP1</i> (ENSG00000117228); <i>ISG15</i> (ENSG00000187608). <b>(D) Mevalonate/Sterol biosynthesis:</b> <i>HMGCR</i> (ENSG00000113161); <i>HMGCS1</i> (ENSG00000112972); <i>DHCR7</i> (ENSG00000172893); <i>FDPS</i> (ENSG00000160752); <i>FDFT1</i> (ENSG00000079459); <i>HSD17B7</i> (ENSG00000132196); <i>LSS</i> (ENSG00000160285); <i>MVD</i> (ENSG00000167508); <i>SQLE</i> (ENSG00000104549); <i>MSMO1</i> (ENSG00000052802); <i>SC5D</i> (ENSG00000109929). All values are reported as log2 fold-change of the difference in expression of infected and matched uninfected controls at each time point as listed in <a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1005511#ppat.1005511.s017" target="_blank">S7 Table</a>.</p

    Simultaneous interrogation of parasite and host transcriptomes.

    No full text
    <p><b>(A) Intracellular <i>T</i>. <i>cruzi</i> life cycle and sample collection scheme.</b> Extracellular <i>T</i>. <i>cruzi</i> trypomastigotes actively penetrate mammalian cells where they receive cues to differentiate into amastigote forms that replicate in the host cell cytoplasm for 3–5 days, beginning at ~22 hpi with a doubling time of ~12 hr. Amastigote division ceases on day 4 or 5 post-infection and parasites differentiate back into trypomastigotes that rupture the host cell to initiate a new cellular infection cycle. For RNA-Seq analysis, total RNA was isolated from axenic <i>T</i>. <i>cruzi</i> epimastigotes (insect vector stage), extracellular trypomastigotes and from amastigote-containing human fibroblast monolayers at 6 time points spanning 4–72 hpi. Pie charts indicate the proportion of mapped sequence reads assigned to the parasite (pink) or human (grey). (<b>B) Distribution of global gene expression levels in a representative subset of <i>T</i>. <i>cruzi</i> and human samples.</b> Box plots showing comparisons of the distribution of per-gene counts (log2 counts per million with an offset of 1) normalized for sequencing library size. The ends of the whiskers represent the lowest datum still within 1.5 interquartile range (IQR) of the lower quartile, and the highest datum still within 1.5 IQR of the upper quartile. Genes with extremely high or low expression levels are shown as open circles above and below the whiskers, respectively. <b>(C) Principal component analysis plots of global transcriptome profiles.</b> Principal component analysis (PCA) plot for RNA-Seq data with <i>T</i>. <i>cruzi</i> and human samples plotted separately. The two first principal components (PC1 and PC2) are plotted with the proportion of variance explained by each component next to the axes labels. Each sample is represented by a dot and the color label corresponding to the sample group, such as the number of hours (hr) post-infection.</p

    Validation of predicted metabolic features of <i>T</i>. <i>cruzi</i> developmental stages.

    No full text
    <p><b>(A)</b> Heatmap of expression values of annotated <i>T</i>. <i>cruzi</i> genes predicted to function in intermediary metabolism with a focus on glycolysis, TCA cycle and Ox-PHOS. <b>(B)</b> Calculated basal respiratory capacity <b>(C)</b> ATP-linked respiration and <b>(D)</b> spare-respiratory capacity of extracellular trypomastigotes and isolated intracellular amastigotes (60 hpi) as pmol of oxygen consumed per min (oxygen consumption rate; OCR) normalized to <i>T</i>. <i>cruzi</i> DNA (ng) per well. <b>(E)</b> ATP content measured in isolated trypomastigotes and amastigotes in KHB buffer without a consumable carbon source at time points indicated. <b>(F)</b> OCR response to glutamine (10 mM) and ELQ271 (10 μM) in isolated <i>T</i>. <i>cruzi</i> trypomastigotes and amastigotes. <b>(G)</b> Dose-dependent inhibition of <i>T</i>. <i>cruzi</i> amastigote growth in HFF following addition of ELQ271 at 18 hpi and relative infection measured at 72 hpi. Host cell viability and growth (inset) is unaffected by the compound over the course of the assay. Graphs shown are representative of 3 independent experiments.</p

    Temporal expression of metabolic pathway genes in mammalian-infective stages of <i>T</i>. <i>cruzi</i>.

    No full text
    <p>Relative mRNA expression of selected genes in intracellular <i>T</i>. <i>cruzi</i> amastigote stages (4–72 hpi) compared to extracellular trypomastigotes (<b>T</b>). Genes in the following metabolic pathways are highlighted: (<b>A) Mevalonate pathway</b>: mevalonate kinase (TcCLB.436521.9), mevalonate diphosphate decarboxylase (TcCLB.507993.330), squalene monooxygenase (TcCLB.509589.20), farnesyl pyrophosphate synthase (TcCLB.508323.9), 3-hydroxy-3-methylglutaryl-CoA reductase (TcCLB.509167.20). <b>(B) Fatty Acid Synthesis:</b> fatty acid elongase 1 (ELO1) (TcCLB.506661.30), fatty acid elongase 2 (ELO2) (TcCLB.506661.20), fatty acid elongase 3 (ELO3) (TcCLB.506661.10). <b>(C) Glycolysis:</b> glyceraldehyde-3-phosphate dehydrogenase (TcCLB.506943.60); hexokinase (TcCLB.508951.20), triosephosphate isomerase (TcCLB.508647.200), phosphoglycerate kinase (TcCLB.511419.40), aldolase (TcCLB.504163.40). (<b>D) Tricarboxylic Acid / Oxidative Phosphorylation (TCA/Ox-PHOS):</b> cytochrome b5 (TcCLB.506773.44), ATPase subunit 9 (TcCLB.503579.70), cytochrome c oxidase subunit V (TcCLB.510565.30), succinate dehydrogenase 11 (TcCLB.504035.84), succinate dehydrogenase 6 (TcCLB.507091.30), cytochrome c oxidase subunit VII (TcCLB.509233.150), cytochrome c oxidase subunit VI (TcCLB.511145.10), cytochrome c oxidase subunit IV (TcCLB.506529.360). (<b>E) Fatty Acid Oxidation:</b> 3-ketoacyl-CoA thiolase (TcCLB.510507.20), enoyl-CoA hydratase/isomerase (TcCLB.511529.170); enoyl-CoA hydratase, mitochondrial (TcCLB.508185.10) enoyl-CoA hydratase/isomerase (TcCLB.510997.40). <b>(F) Glutamate Dehydrogenases (DH):</b> NADP+-GlutDH (TcCLB.507875.20), NAD+ GlutDH (TcCLB.509445.39). All values are reported as log2 fold-change of difference between expression at in the trypomastigote stage and intracellular stages as reported in <a href="http://www.plospathogens.org/article/info:doi/10.1371/journal.ppat.1005511#ppat.1005511.s015" target="_blank">S5 Table</a>.</p
    corecore