11 research outputs found

    SRF is Important for EGR1 and EGR2 Induction in Parasite-Infected Cells.

    No full text
    <p>A). SRF mRNA abundance was measured at the indicated time points after transfection with SRF or negative control siRNAs. B). HeLa cells were transfected with SRF or negative control siRNA and infected 48 h later. RNA was collected 4 and 18 h later, converted to cDNA, and EGR1 or EGR2 transcript levels measured by real time PCR. Shown are averages and standard deviations of three independent experiments performed in triplicate.</p

    <i>Toxoplasma</i> Activation of SRF is Dependent on Rhoptry Secretion.

    No full text
    <p>A.) SRE-CD8 COS cells were mock treated, infected with RH GFP, or treated with 1 µM cytochalasin D. Cells were collected 18 h later and analyzed by flow cytometry after staining with anti-CD8 antibody. Shown are representative results from three independent experiments. B.) SRE-luc-transfected MEFs were mock- infected or infected with untreated parasites (−4BPB) or parasites pretreated with 4-BPB (Toxo+4BPB). In addition, SRE-luc-transfected cells were pretreated with 4-BPB and then infected with untreated parasites (Host+4BPB). Luciferase activity was measured 18 h later. Shown are the averages and standard deviations of three independent experiments performed in triplicate.</p

    <i>Toxoplasma</i> Activates SRF.

    No full text
    <p>A). SRE-luc transfected MEFs were infected with increasing numbers of parasites for the indicated times and luciferase activity measured. Heat-killed parasites were added at a MOI of 10∶1 (parasites∶host cells). B). SRE-luc transfected MEFs were infected with the indicated parasites at a MOI of 10. Lysates were collected at the indicated times and luciferase activity measured. C). Cells were mock-infected or infected with equal numbers of RH and GT1 (Types I), Pru (Type II), and CTG (Type III) strains of <i>Toxoplasma</i> for indicated times before RNA was harvested. Real-time PCR was used to measure EGR2 transcript abundance. D). pEGR4x-Luc transfected cells were infected with each parasite strain and then luciferase activity was measured 16 h later. The dotted lines in all of the plots represent a 2-fold increase, which is considered the minimum increase level to be considered significant. Shown are averages and standard deviations of three independent experiments performed in triplicate.</p

    <i>Toxoplasma</i> Does Not Signal Through MAL.

    No full text
    <p>A.) SRE-luc transfected MEFs were incubated with TcdB-LF and PA for 6 hours. Cells were washed and either treated with 15% FBS for 6 h or infected for 16 h at which time luciferase activity was measured. B.) MEFs transfected with either SRE-luc (white bars) or pSM22α-luc (black bars) were parasite-infected or treated with 1 µM cytochalasin D. Luciferase was activity measured 16 h later. Shown are the averages and standard deviations of three independent experiments performed in triplicate.</p

    Activation of SRF by <i>Toxoplasma</i> is MAPK Dependent.

    No full text
    <p>A.) SRE-Luc transfected cells were mock-treated or treated for 30′ with 15 µM p38 MAPK inhibitor SB203580 (SB), 25 µM JNK inhibitor SP600125 (SP), 10 µM ERK inhibitor U0126 (U), or a mix of all three. The cells were then infected and luciferase activity measured 8 or 18 hpi. *,p<0.05 Student's <i>t</i> test, **,p<0.001 Student's <i>t</i> test. B.) SRE-luc-MEFs were co-transfected with either a control pCDNA3.1 empty vector (empty) or dominant negative p38 MAPK (DN p38). The cells were then mock- or parasite-infected and luciferase activity measured 18 h later. C.) SRE-Luc transfected cells were pretreated with LF and PA for 6 hours. Cells were washed and then either treated with EGF (50 ng/ml) for 6 hours or <i>Toxoplasma</i>-infected for 16 hours. Luciferase activity was measured at each time point. Shown are the averages and standard deviations of three independent experiments performed in triplicate.</p

    Generation of SBR mutants.

    No full text
    <p>A. Relative plaque formation in HFFs was determined for each parasite strain in the presence of increasing concentrations of SB505124. B–D. Parasite replication was measured by infecting HFFs on glass coverslips in the presence or absence of 3 µM SB505124 and then fixing the cells 24 hours later. Parasites and nuclei were detected with anti-SAG1 antibody and DAPI, respectively. B. Representative images. C. For each replicate, 100 vacuoles were monitored for parasites per vacuole and nuclei per parasite. Vacuoles were designated as being irregular if they contained an irregular number of parasites/vacuole (non 2<sup>n</sup>). Shown are averaged percentages and standard deviations of 2 independent experiments with two replicates each. D. Averaged percentages and standard deviations of irregular vacuoles (show in C) by nuclei per parasite.</p

    Forward Genetic Screening Identifies a Small Molecule That Blocks <i>Toxoplasma gondii</i> Growth by Inhibiting Both Host- and Parasite-Encoded Kinases

    No full text
    <div><p>The simultaneous targeting of host and pathogen processes represents an untapped approach for the treatment of intracellular infections. Hypoxia-inducible factor-1 (HIF-1) is a host cell transcription factor that is activated by and required for the growth of the intracellular protozoan parasite <i>Toxoplasma gondii</i> at physiological oxygen levels. Parasite activation of HIF-1 is blocked by inhibiting the family of closely related Activin-Like Kinase (ALK) host cell receptors ALK4, ALK5, and ALK7, which was determined in part by use of an ALK4,5,7 inhibitor named SB505124. Besides inhibiting HIF-1 activation, SB505124 also potently blocks parasite replication under normoxic conditions. To determine whether SB505124 inhibition of parasite growth was exclusively due to inhibition of ALK4,5,7 or because the drug inhibited a second kinase, SB505124-resistant parasites were isolated by chemical mutagenesis. Whole-genome sequencing of these mutants revealed mutations in the <i>Toxoplasma</i> MAP kinase, TgMAPK1. Allelic replacement of mutant TgMAPK1 alleles into wild-type parasites was sufficient to confer SB505124 resistance. SB505124 independently impacts TgMAPK1 and ALK4,5,7 signaling since drug resistant parasites could not activate HIF-1 in the presence of SB505124 or grow in HIF-1 deficient cells. In addition, TgMAPK1 kinase activity is inhibited by SB505124. Finally, mice treated with SB505124 had significantly lower tissue burdens following <i>Toxoplasma</i> infection. These data therefore identify SB505124 as a novel small molecule inhibitor that acts by inhibiting two distinct targets, host HIF-1 and TgMAPK1.</p></div

    SB505124 reduces parasite growth in <i>Toxoplasma</i>-infected mice.

    No full text
    <p>RH-GFP infected mice were intraperitoneally injected daily with 10 mg/kg SB505124 or DMSO alone. After 5 days post-infection, mice were sacrificed and flow cytometric analysis was performed on peritoneal exudate cells (3–4 mice per treatment group per experiment, 2 independent experiments). A. FACS plots (upper) and histograms (lower) showing percentages of infected (GFP<sup>+</sup>) cells of two representative mice per treatment group. B. Mean percentages of infected cells between treatment groups with standard deviations. C. Relative MFI of infected (GFP<sup>+</sup>) cells with standard deviations. D. ELISA determination of serum IFNγ levels of mock- and drug-treated, intraperitoneally infected mice 5 days post-infection. Shown are average and standard deviations.</p

    TgMAPK1 is an SBR gene.

    No full text
    <p>A. Venn diagram of whole genome sequencing data of codon-changing SNVs identified in each mutant. B. Amino acid positions of TgMAPK1<sup>SBR</sup> mutations. C. TgMAPK1<sup>SBR</sup> allelic replacement strategy. Primers 1 and 2 were used to amplify 944 bp fragments of genomic DNA containing the SBR allele and cloned into pCR2.1. Primers 3 and 4 were used to amplify 1055 bp fragments of genomic DNA to confirm allelic replacement by Sanger sequencing. D. RHΔku80 parasites were transfected with linearized TgMAPK1<sup>WT</sup> or TgMAPK1<sup>SBR</sup> replacement constructs and grown in 3 µM SB505124-treated HFFs. Shown are representative images depicting the ability of RHΔKu80:TgMAPK1<sup>SBR1</sup> to grow and form plaques after 5 days of growth in the presence of 3 µM SB505124.</p

    SB505124 impacts HIF1 and TgMAPK1 through distinct pathways.

    No full text
    <p>A. HIF-1 luciferase reporter activity in mock- or SB505124-treated MEFs was measured after 18 h of infection with RHΔ or SBR1-3. Shown are averaged measurements and standard deviations from at least 3 independent experiments performed in triplicate. B. HIF-1αWT and HIF-1α-/- MEFs were grown in 24 well plates and infected with RHΔ or SBR1-3. The plates were grown for 66 h at 3% O<sub>2</sub> and then 5 µCi <sup>3</sup>H-Uracil was added to each well to assess parasite growth. Shown are averaged data and standard deviations from 3 independent experiments performed in duplicate. C. HFFs grown on glass coverslips were infected with RH GFP at an MOI of 10 for 6 h in the presence or absence of 5 µM SB505124. Representative images are shown.</p
    corecore