The enteric pathogen Cryptosporidium parvum exports proteins into the cytosol of the infected host cell

  1. Jennifer E Dumaine
  2. Adam Sateriale
  3. Alexis R Gibson
  4. Amita G Reddy
  5. Jodi A Gullicksrud
  6. Emma N Hunter
  7. Joseph T Clark
  8. Boris Striepen  Is a corresponding author
  1. Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, United States
  2. Franklin College of Arts and Science, University of Georgia, United States
6 figures, 2 tables and 3 additional files

Figures

Figure 1 with 3 supplements
MEDLE2 is exported to the host cell cytoplasm.

(A) Schematic overview of the chromosomal location for polymorphic gene families in the C. parvum genome. (B) Map of the MEDLE2 locus targeted in C. parvum for insertion of a 3× hemagglutinin (HA) epitope tag, a nanoluciferase reporter gene (Nluc), and neomycin phosphotransferase selection marker (Neo). (C) PCR mapping of the MEDLE2 locus using genomic DNA from wild type (WT) and transgenic (MEDLE2-HA) sporozoites, corresponding primer pairs are shown in (B), and thymidine kinase (TK) gene used as a control. Note the presence of two bands in the 5′–3′ amplification, indicating the presence of a transgene (3081 bp) and persistence of an unmodified copy (1174 bp), suggesting multiple copies of MEDLE2 in the C. parvum genome; also see Figure 1—figure supplement 2. (D, E) HCT-8 cultures were infected with WYLE4-HA (D) or MEDLE2-HA (E) transgenic parasites and fixed after 24 hr for immunofluorescence assay (IFA). Red, antibody to HA; green, Vicia villosa lectin stain, VVL (Gut and Nelson, 1999); blue, Hoechst DNA dye. Additional genes targeted and the localizations of their products are summarized in Table 1 and Figure 1—figure supplements 1 and 3.

Figure 1—figure supplement 1
Additional secretory proteins tested in this study.

(A–C) Schematic depicting the generation of in-locus gene fusion transgenic parasite strains for WYLE4 (A) and SKSR7 (B). In each case, the C terminus of the gene was targeted for integration of a repair construct containing a hemagglutinin (HA) epitope tag, a nanoluciferase reporter gene (Nluc), and a neomycin phosphotransferase (Neo) selectable marker. Individual primer pairs used to map integration in wild type (WT) and transgenic strains are shown. (C) SKSR7-HA does not localize to the host cell; rather, the protein (red) exhibits expression in the parasite (green).

Figure 1—figure supplement 2
Knockout of MEDLE2 reveals multiple copies of the gene in the genome.

(A) Schematic representation for the strategy used to generate a MEDLE2 KO line, in which the entire locus of MEDLE2 is replaced with a nanoluciferase reporter gene (Nluc) and the neomycin phosphotransferase (Neo) selection marker fused to a 2A peptide, and tdTomato, such that the parasites express a red fluorescent protein in their cytoplasm. The solid black arrow indicates the position of the Cas9-induced double-stranded break in the middle of the gene. Note that this is a different guide from the one used for C-terminal tagging. (B) PCR mapping modification of the MEDLE2 locus using genomic DNA from wild type (WT) and transgenic (MEDLE2 KO) sporozoites using the primer pairs shown in (A) and the thymidine kinase (TK) gene as a control. Note the persistence of a WT band (1392 bp) in the 5′–3′ amplification product, despite the presence of the transgene (3524 bp). (C) HCT-8 cultures were fixed 24 hr after being infected with MEDLE2 KO transgenic parasites. MEDLE2 KO parasites exhibit red fluorescence in their cytoplasm as expected (red, tdTomato, parasite cytoplasm; green, parasites VVL; blue, Hoechst). (D) The full gene PCR products from WT (1392 bp) and MEDLE2 KO parasites (3524 bp) were used for restriction digest with ScaI. A single ScaI restriction site is found in the C terminus of WT MEDLE2; however, integration of the repair cassette disrupts this site. ScaI digested WT PCR product results in two digest products: 331 bp and 1061 bp. Undigested MEDLE2 KO full gene product has the expected 3524 bp fragment, as well as a persisting 1392 bp WT band. ScaI digested MEDLE2 KO shows the 3525 bp repair cassette resistant to ScaI digest, as well as the 331 bp and 1061 bp fragments produced from digest of the unmodified MEDLE2 locus. As a result, there are multiple copies of MEDLE2 in the genome and we have only targeted one for knockout.

Figure 1—figure supplement 3
Other members of the MEDLE gene family are exported to the host cell.

(A) Schematic representation depicting the generation of a MEDLE6-HA transgenic parasite line, in which the endogenous locus of MEDLE6 (cgd6_5490) is a hemagglutinin (HA) epitope tagged at the C terminus. Proper integration at the desired locus was confirmed using PCR mapping with gDNA isolated from MEDLE6 transgenic parasites (M6) and a wild type control (WT). (B) MEDLE6-HA parasites were used to infect HCT-8 cells for an immunofluorescence assay. Cells were fixed every 12 hr and stained for immunofluorescence assay (IFA). Shown as a representative image, at 24 hr post infection, MEDLE6 (red) localizes in/around the parasite (green), as well as slightly in the host cell. Host cell expression is more apparent in multiply infected cells. (C) The MEDLE1 (cgd5_4580) locus was targeted for integration of an HA epitope tag at the C terminus. (D) MEDLE1-HA parasites were used to infect HCT-8 cells for a time-course infection, and IFA was performed on cells fixed every 12 hr. Shown as a representative image, at 12 hr post infection, MEDLE1 (red) localizes in/around the parasite (green), as well as at very low levels in the host cell. (E) Schematic representation for the strategy used to engineer a MEDLE1 overexpression line. The MEDLE2 promoter was used to drive expression of an ectopic copy of MEDLE1-HA expressed in the TK locus. Proper integration was assessed using PCR mapping with gDNA isolated from Medle2MEDLE1-HA (M2- M1 HA) and WT control (WT) parasites. (F) M2-M1 HA parasites were used to infect HCT-8 cells for IFA. At 24 hr post infection, MEDLE1-HA (red) can be seen in/around the parasite (green), as well as in the host cell when expression is driven by the MEDLE2 promoter.

Figure 2 with 1 supplement
Infected cells express MEDLE2-HA across the parasite life cycle.

(A) 200,000 MEDLE2-HA-tdNeon transgenic parasites were used to infect HCT-8 cells and fixed at intervals across a 72 hr time period. Data shown are representative images from triplicate coverslips processed for immunofluorescence assay (IFA). Red, hemagglutinin (HA)-tagged protein; green, parasites (mNeon); blue, Hoechst. (B, C) Quantification of MEDLE2-expressing cells (red) versus intracellular parasites (blue) for 3695 host cells evaluated across a 72 hr time course. 20 fields of view quantified using ImageJ to identify host cells and parasites (B). The percentage of cell exhibiting MEDLE2-HA and mNeon staining is constant across the time course with a cumulative 94 ± 1.83% (mean ± SD) (C). (D) HCT-8 cultures infected with MEDLE2-HA parasites were fixed for IFA at 48 hr when sexual life stages were present. Cells were stained with stage-specific antibodies for female (COWP1) and male (α- tubulin), demonstrating MEDLE2 is exported across the parasite life cycle. Red, HA-tagged protein; green, parasites (stage-specific antibody); blue, Hoechst. (E) IFA of cryosections from the small intestine of Ifng-/- mice infected with MEDLE2-HA-tdNeon C. parvum (images representative of samples from three mice). Red, HA-tagged protein; green, parasites (tdNeon); blue, Hoechst; gray, Phalloidin (actin).

Figure 2—source data 1

Numerical data used for the quantification of HA positive host cells and the intracellular parasites.

https://cdn.elifesciences.org/articles/70451/elife-70451-fig2-data1-v2.xlsx
Figure 2—figure supplement 1
Construction of a MEDLE2-HA cytoplasmic tdNeon reporter parasite.

(A) Schematic representation of the strategy to derive reporter parasite line in which MEDLE2 is 3× hemagglutinin (HA) epitope tagged and the parasite cytoplasm expresses a tandem mNeon green tag (tdNeon). The solid black arrow indicates the position of the Cas9-induced double-stranded break at the C terminus of the gene, which is the same guide used in Figure 1B to generate the MEDLE2-HA transgenic parasites. (B) PCR mapping modification of the MEDLE2 locus using genomic DNA from wild type (WT) and transgenic (MEDLE2-HA-tdNeon) sporozoites using the primer pairs shown in (A) and the thymidine kinase (TK) gene as a control. Note the presence of both a 1174 bp WT gene and a 4557 bp transgene. (C) HCT-8 cultures were infected with and MEDLE2-HA-tdNeon transgenic parasites and fixed at 24 hr. Red, HA-tagged protein; green, parasites (tdNeon); blue, Hoechst.

MEDLE2 is expressed by trophozoites and passes through the secretory pathway.

(A) Schematic representation of hypothetical patterns of MEDLE2 export in C. parvum. (B) Immunofluorescence assay (IFA) of wild type (WT) and MEDLE2-HA sporozoites fixed on poly-L-lysine-treated coverslips. We note that MEDLE2-HA is not observed in sporozoites. Red, hemagglutinin (HA)-tagged protein; green, sporozoite antigen Cp23; blue, Hoechst. (C) HCT-8 cells infected with MEDLE2-HA parasites were fixed in 30 min increments and processed for IFA. Data shown are representative images from a time-course bridging 3 hr (no observed MEDLE2-HA) and 6 hr (MEDLE2-HA abundant in host cell). White arrowheads denote parasite nuclei. Red, HA-tagged protein; blue, Hoechst. (D) MEDLE2-HA parasites were excysted and used to infect HCT-8 and after 3 hr media were supplemented with brefeldin A (BFA) (10 µg/mL). 10 hr post infection, cells were fixed and processed for IFA. Red, HA-tagged protein; green, parasites (VVL); blue, Hoechst (D). (E) The impact of BFA treatment on MEDLE2-HA export was quantified showing a significant reduction in MEDLE2 export when comparing BFA-treated (red) and untreated cells (blue) (n = 191 untreated, n = 98 treated; mean ± SEM; p<0.0001; unpaired t test with Welch’s correction).

Figure 3—source data 1

Numerical data used for the quantification of HA positive host cells in the absence and presence of Brefeldin A (BFA).

https://cdn.elifesciences.org/articles/70451/elife-70451-fig3-data1-v2.xlsx
Figure 4 with 3 supplements
Ordered reporters disrupt MEDLE2 export.

(A) Schematic map of the MEDLE2 locus targeted for insertion of three different reporter genes (mScarlet, beta-lactamase, or Cre recombinase), nanoluciferase (Nluc), and the selection marker (Neo). The guide RNA and flanking sequences used here were the same as those employed to generate MEDLE2-HA transgenic parasites (see Figure 2—figure supplement 1, Figure 4—figure supplement 1 for more detail). (B) MEDLE2-mScarlet parasites were used to infect HCT-8 cells and fixed for immunofluorescence assay (IFA) across a time course. Data shown are from 10 hr post infection, which is representative of the MEDLE2 localization observed at all time points. Red, Medle2-mScarlet; green, parasites (VVL); blue, Hoechst. (C) HCT-8 cells were infected with MEDLE2-BLA C. parvum for 24 hr before incubation with the CCF4-AM beta-lactamase substrate and visualization by live microscopy. This experiment was repeated three times. Red, parasites (tdTomato); green, uncleaved CC4F-AM; blue, cleaved CCF4-AM; gray, DIC. We attribute lack of CCF4-AM cleavage to failure of MEDLE2-BLA to export (Figure 4—figure supplement 1). (D) MEDLE2-Cre parasites were used to infect loxGFP/RFP color switch HCT-8 cells ((Figure 4—figure supplement 2) for schematic representation). After 48 hr, cells were subjected to flow cytometry. Live, single cells were gated based upon forward and side scatter, and green fluorescence (GFP) and red fluorescence (RFP) were measured to detect Cre recombinase activity. Despite robust infection, MEDLE2-Cre-infected cultures did not express RFP (Figure 4—figure supplement 2) compared to the positive control that was transiently transfected to express Cre recombinase.

Figure 4—figure supplement 1
MEDLE2-BLA is not exported.

(A) Schematic representation of the strategy used to generate a MEDLE2-mScarlet parasite line, in which the MEDLE2 is C-terminally tagged with the fluorescent protein mScarlet, as well as engineered to express a nanoluciferase reporter gene (Nluc) and the neomycin phosphotransferase (Neo) selection marker. The solid black guide hit sequence is the same as used in Figure 1B to generate the MEDLE2-HA transgenic parasites. Integration PCR using MEDLE2-mScarlet and WT gDNA confirms proper integration. (B) The C terminus of MEDLE2 was targeted for insertion of a construct encoding beta-lactamase (BLA), a nanoluciferase reporter gene (Nluc) and the neomycin phosphotransferase (Neo) selection marker fused to a 2A peptide and a tdTomato reporter gene, using the same tagging strategy previously utilized for this locus. Integration PCR mapping the MEDLE2 locus using genomic DNA from wild type (WT) and transgenic (MEDLE2-BLA) sporozoites with the corresponding primer pairs shown in (A) and the thymidine kinase (TK) gene as a control shows the locus was successfully modified. (C) MEDLE2-BLA transgenic parasites were used to infect HCT-8 cells and fixed at 24 hr for immunofluorescence assay (IFA). Red, parasites (tdTomato); green, beta-lactamase (beta-lactamase antibody); blue, Hoechst.

Figure 4—figure supplement 2
MEDLE2-Cre parasites infect loxGFP/RFP color switch cells.

(A) MEDLE2-Cre-expressing parasites were generated by targeting the MEDLE2 locus for insertion of a construct encoding Cre recombinase (Cre), a nanoluciferase reporter gene (Nluc), and the neomycin phosphotransferase (Neo) selection marker. The solid black arrow indicates the guide hit sequence, which is the same guide used in Figure 1B to generate the MEDLE2-HA transgenic parasites. (B) Integration PCR mapping the MEDLE2 locus using genomic DNA from wild type (WT) and transgenic (MEDLE2-Cre) sporozoites using the primer pairs shown in (A) and the thymidine kinase (TK) gene as a control. (C) Schematic of the LoxGFP/RFP color switch HCT-8. Introduction of Cre into these cells induces a color switch from green to red by recombinase-directed removal of the GFP coding sequence along with a stop codon preventing translation of RFP. (D) 1 × 106 transgenic MEDLE2-Cre oocyts were used to infect LoxGFP/RFP color switch HCT-8 cells for 48 hr before preparation of the sample for flow cytometry. 400,000 cells were removed from the sample preparation for nanoluciferase assay to determine infection in the culture. Mean nanoluciferase (relative luminescence) ± SEM is shown for three replicates (p<0.0001; one-way ANOVA with Dunnett’s multiple comparison test). Uninfected cells were used as a negative control, and Cre recombinase transfected cells were used as a positive control.

Figure 4—figure supplement 3
MEDLE2 is an intrinsically disordered protein.

MEDLE2 contains multiple single amino acid repeat regions and lacks a well-defined tertiary structure. The intrinsic disorder of MEDLE2 was assessed using IUPred2A (https://iupred2a.elte.hu/) and the resulting disordered plot is shown as greater than the threshold value of 0.5, represented by the horizontal black line.

Figure 5 with 3 supplements
MEDLE2 contains a host-targeting motif that is processed during export.

(A) Map showing the strategy used to engineer an ectopic copy of MEDLE2-HA in the thymidine kinase (TK) locus. Expression of an ectopic copy of MEDLE2-HA was driven by the MEDLE2 promoter. All point mutations were confirmed by Sanger sequencing (Figure 5—figure supplement 1). (B) Schematic representation of the MEDLE2 mutants generated using the strategy outlined in (A). The signal peptide (SP) is represented by dark blue, and low-complexity regions are shown in light blue. Candidate motifs targeted for mutagenesis are indicated with black triangles, and mutagenized amino acids are shown in red for two representative mutants. (C, D) Mutant parasites were used to infect HCT-8 cells and fixed for immunofluorescence assay (IFA) after 24 hr. For mutants shown in (C), the entire candidate motif was replaced with a matching number of alanine residues (e.g., KDVSLI/6A → AAAAAA). For mutants shown in (D), each individual amino acid in the KDVSLI sequence was changed to alanine. Red, hemagglutinin (HA)-tagged protein; green, parasites (VVL); blue, Hoechst. We note that SP and leucine 35 within the KDVSLI sequence are required for MEDLE2 export. (E, F) 5 × 106 transgenic oocysts were used to infect HCT-8 cells for 48 hr before preparation of whole-cell lysates. Proteins were separated by for SDS-PAGE and analyzed by western blot. The resulting blots for infections with whole motif mutants (E) and individual amino acid point mutants (F) are shown. Red, neomycin; green, HA. Note that when mutants are expressed in mammalian cells and not C. parvum the resulting proteins do not show any size differences (Figure 5—figure supplement 2).

Figure 5—figure supplement 1
Sanger sequencing confirming the generation of MEDLE2 mutants.

100,000 sporozoites were used from each mutant strain for genomic DNA extraction. The resulting gDNA was used for PCR mapping of the thymidine kinase (TK) locus to verify the desired mutagenesis. 5′ TK–Nluc PCR products were used for TopoTA cloning, and the resulting colonies were grown and Sanger sequenced. Three colonies were sequenced for each strain using the M13 forward, M13 reverse, and an internal MEDLE2-specific primer to confirm targeted mutagenesis. The Benchling alignment of the Sanger sequencing result (transgene) to the reference sequence is shown. The black box highlights the mutation engineered in each strain.

Figure 5—figure supplement 2
MEDLE2 mutants are of the same size as wild type (WT) MEDLE2 when expressed in HEK293T cells.

Plasmids encoding human codon-optimized MEDLE2 with the N-terminal signal peptide removed (aa 2–20) (WT) or encoding the desired point mutations (denoted residues were replaced with alanines) were transfected into HEK233T cells. After 24 hr, whole-cell lysates were prepared, and proteins separated by SDS-PAGE for western blot analysis. MEDLE2 mutants were indistinguishable in size from WT MEDLE2. Red, neomycin; green, hemagglutinin (HA).

Figure 5—figure supplement 3
Uncropped images of panels shown in Figure 5D including infected and uninfected cells.

Transgenic C. parvum parasites with point mutations engineered in an ectopic copy of MEDLE2 expressed in the thymidine kinase (TK) locus were used to infect HCT-8 cells for 24 hr before fixation for immunofluorescence assay. L35A mutation results in accumulation of MEDLE2-HA (red) with the parasite (green).

Figure 6 with 2 supplements
MEDLE2-expressing cells exhibit upregulation of genes involved in the unfolded protein response.

(A) HEK293T cells were transfected with plasmids encoding MEDLE2-GFP or GFP alone. After 24 hr, cells were fixed and processed for immunofluorescence assay (IFA). GFP is shown in green, Hoechst in blue. (B) 24 hr post transection, HEK293T cells were trypsinized and double sorted for live, GFP+ singlets directly into RNA lysis buffer and subjected to RNA sequencing. (C) Heat map depicting the differential gene expression between MEDLE2-GFP (top panel) and GFP control expressing cells (bottom panel). Upregulated gene expression is shown in red (row Z score > 0), while blue shows genes that are downregulated in expression (row Z score < 0). Expressing cells compared to GFP control cells. 413 transcripts showed upregulation in MEDLE2-GFP-expressing cells (right) and 487 genes had lower transcript abundance (left). The horizontal dashed line indicates p-value = 0.05. Gene set enrichment analysis (GSEA) performed on the 900 differentially expressed genes from the MEDLE2 transfection dataset identifies core enrichment of 20 genes that belong to ER stress response signaling pathways, which are indicated on the volcano plot in red. The most upregulated genes are identified by their gene ID. (E) The 234 genes with the greatest differential expression (p<0.01, log fold change absolute value > 1.5) were used to define a MEDLE2 gene set from the MEDLE2-GFP transfection dataset. This signature was used to perform GSEA using data from single-cell RNA sequencing on C. parvum-infected organoid-derived cultures, which showed enrichment of 51 genes with 22 genes in the core enrichment for the MEDLE2 response set highlighted in solid red. We note that we did not detect the MEDLE2 response signature in datasets from other enteric infections including rotavirus (Figure 6—figure supplement 1). (F) Ileal sections were removed from C. parvum-infected Ifng-/- mice and uninfected controls (each n = 3), and expression levels for the four differentially upregulated genes in the MEDLE2 response set (NUPR1, CHAC1, DDIT3, and TRIB3) were measured by qPCR. (G) HCT-8 cultures were pretreated for 2 hr with inhibitors (GSK2606414 and KIRA6) of ER stress signaling pathways prior to infection with 10,000 MEDLE2-HA parasites. After 24 hr, cells were lysed and nanoluciferase assay was performed as a measure of parasite growth. Inhibition of the IRE1 signaling pathway with KIRA6 significantly reduced parasite growth (one-way ANOVA, Dunnett’s multiple comparisons test p=0.0303; a representative experiment is shown [n = 6]). This experiment was repeated three times. (H) Ddit3-/- and C57BL/6J mice were treated with anti-mouse-IFN gamma antibody 1 day prior to infection with 10,000 MEDLE2-HA-tdNeon oocysts, and again at day 2 of infection. Fecal luminescence was determined by nanoluciferase activity to calculate the area under the curve for the duration of the infection. Ddit3-/- mice exhibited a 56% reduction in infection (1,416,227 ± 44,850; total peak area ± standard error) compared to control mice (3,189,123 ± 69,887; unpaired t test, p<0.001; n = 4 mice per group). One representative experiment is shown, which was repeated two more times, with a 54% reduction and no change in infection being observed.

Figure 6—figure supplement 1
MEDLE2 response signature is absent from intestinal epithelial cells infected with human rotavirus.

Genes with the greatest differential expression (p<0.01, log fold change absolute value > 1.5) were used to define a MEDLE2 gene set from the MEDLE2-GFP transfected cells. RNA sequencing from small intestinal enteroid cultures infected with human rotavirus shows an absence of the MEDLE2 signature created from the MEDLE2 transfection dataset, evidenced by the negative enrichment score resulting from gene set enrichment analysis (GSEA).

Figure 6—figure supplement 2
Transfection of HEK293T cells with T. gondii GRA16-GFP also results in ER stress.

HEK293T cells were transfected with plasmids encoding GFP-only, MEDLE2-GFP, and Tg. GRA16-GFP, RNA was extracted, and qPCR was performed to characterize the impact on ER stress response genes (CHAC1, DDIT3, NUPR1, and TRIB3). Introduction of disordered proteins, such as MEDLE2 (A) and GRA16 (B), both result in transcriptional changes in ER stress response genes.

Tables

Table 1
Members of multigene families for which localization of protein product was initially attempted in this study.
Gene familyGene IDResult
MEDLEcgd5_4590Exported to host cell
FLGNcgd4_4470Transgenic unsuccessful
GGCcgd5_3570Transgenic unsuccessful
SKSRcgd8_30Not in host cell
SKSRcgd8_40Not in host cell
WYLEcgd8_3560Not in host cell
Key resources table
Reagent type (species) or resourceDesignationSource or referenceIdentifiersAdditional information
Gene (Cryptosporidium parvum)MEDLE2Li et al., 2017cgd5_4590
Gene (C. parvum)MEDLE1Fei et al., 2018cgd5_4580
Gene (C. parvum)MEDLE6This papercgd6_5490Named according to the spatial localization in the genome similarly to MEDLE2 and MEDLE1
Gene (C. parvum)WYLE4This papercgd8_3560Named for the gene family and according to the spatial localization in the genome
Gene (C. parvum)SKSR7This paperCgd8_30Named for the gene family and according to the spatial localization in the genome
Strain, strain background (C. parvum)C. parvum oocysts, IOWAII strain (WT)Bunchgrass
Strain, strain background (Mus musculus)Ifng-/-, C57BL/6JThe Jackson LaboratoryJax 002287;
RRID:IMSR_JAX:002287
Strain, strain background (M. musculus)Ddit3tm2.1DronThe Jackson LaboratoryJax 005530;
RRID:IMSR_JAX:005530
Strain, strain background (M. musculus)C57BL/6JThe Jackson LaboratoryJax 000664;
RRID:IMSR_JAX:000664
Genetic reagent (C. parvum)MEDLE2-HAThis papercgd5_4590 modifiedStable transgenic parasite line expressing HA
Genetic reagent (C. parvum)WYLE4-HAThis papercgd8_3570 modifiedStable transgenic parasite line expressing HA
Genetic reagent (C. parvum)SKSR7-HAThis papercgd8_30 modifiedStable transgenic parasite line expressing HA
Genetic reagent (C. parvum)MEDLE2 KOThis paperCgd5_4590Stable transgenic parasite line with one copy of MEDLE2 knocked out
Genetic reagent (C. parvum)MEDLE1-HAThis papercgd5_4580 modifiedStable transgenic parasite line expressing HA
Genetic reagent (C. parvum)MEDLE6-HAThis papercgd6_5490 modifiedStable transgenic parasite line expressing HA
Genetic reagent (C. parvum)Medle2 MEDLE1-HAThis papercgd5_4440 modifiedStable transgenic parasite line expressing HA
Genetic reagent (C. parvum)MEDLE2-HA-tdNeonThis papercgd5_4590 modifiedStable transgenic parasite line expressing HA and tdNeon
Genetic reagent (C. parvum)MEDLE2-mScarletThis papercgd5_4590 modifiedStable transgenic parasite line expressing mScarlet
Genetic reagent (C. parvum)MEDLE2-Bla-2A-tdTomatoThis papercgd5_4590 modifiedStable transgenic parasite line expressing BLA and tdTomato
Genetic reagent (C. parvum)MEDLE2-CreThis papercgd5_4590 modifiedStable transgenic parasite line expressing Cre recombinase
Genetic reagent (C. parvum)Ectopic MEDLE2-HAThis papercgd5_4440 modifiedStable transgenic parasite line expressing extra copy of MEDLE2
Genetic reagent (C. parvum)ΔSPThis papercgd5_4440 modifiedStable transgenic parasite line expressing extra copy of MEDLE2 (aa 21–209)
Genetic reagent (C. parvum)KDVSLI/6AThis papercgd5_4440 modifiedStable transgenic parasite line expressing extra copy of MEDLE2 with KDVSLI (aa 31–36) mutated to six alanines
Genetic reagent (C. parvum)KPVLKN/6AThis papercgd5_4440 modifiedStable transgenic parasite line expressing extra copy of MEDLE2 with KPVLKN (aa 73–78) mutated to six alanines
Genetic reagent (C. parvum)KNVNLS/6AThis papercgd5_4440 modifiedStable transgenic parasite line expressing extra copy of MEDLE2 with KDVSLI (aa 77–82) mutated to six alanines
Genetic reagent (C. parvum)RGLLRGLSG/9AThis papercgd5_4440 modifiedStable transgenic parasite line expressing extra copy of MEDLE2 with KDVSLI (aa 191–199) mutated to six alanines
Genetic reagent (C. parvum)K31AThis papercgd5_4440 modifiedStable transgenic parasite line expressing extra copy of MEDLE2 with K31 mutated to alanine
Genetic reagent (C. parvum)D32AThis papercgd5_4440 modifiedStable transgenic parasite line expressing extra copy of MEDLE2 with D32 mutated to alanine
Genetic reagent (C. parvum)V33AThis papercgd5_4440 modifiedStable transgenic parasite line expressing extra copy of MEDLE2 with V33 mutated to alanine
Genetic reagent (C. parvum)S34AThis papercgd5_4440 modifiedStable transgenic parasite line expressing extra copy of MEDLE2 with S34 mutated to alanine
Genetic reagent (C. parvum)L35AThis papercgd5_4440 modifiedStable transgenic parasite line expressing extra copy of MEDLE2 with L35 mutated to alanine
Genetic reagent (C. parvum)I36AThis papercgd5_4440 modifiedStable transgenic parasite line expressing extra copy of MEDLE2 with I36 mutated to alanine
Cell line (human)HCT-8ATCCCCL-224;
RRID:CVCL_2478
Cell line (human)HEK293TATCCCRL-3216;
RRID:CVCL_0063
Cell line (Escherichia coli)GC5Genesee
Scientific
42-653Electrocompetent cells
Cell line (E. coli)One Shot
Topo10,
InvitrogenC404003Electrocompetent cells
Transfected construct (human)loxP GFP/RFP color switch lentivirusGenTarget IncCat#: LVP460-NeoTransfected construct (human)
Biological sample (M. musculus)Isolated sections of ileumIfng-/- miceJax 002287;
RRID:IMSR_ JAX:002287
6-week-old male mice
AntibodyAnti-HA (rat monoclonal)MilliporeSigmaCat# 11867431001; RRID:AB_390919IF (1:500), WB (1:500), IHC (1:500)
AntibodyAnti-Cp23 (mouse monoclonal)LS BioCat# LS-C137378; RRID:AB_10947007IF (1:100)
AntibodyAnti-alpha tubulin (mouse monoclonal)Developmental Studies Hybridoma BankCat#12G10; RRID:AB_1157911IF (1:1000)
AntibodyAnti-COWP1 (rat monoclonal)This paper, produced by GenScriptThis paperIF (1:100)
AntibodyAnti-neomycin phosphotransferase II (rabbit polyclonal)MilliporeSigmaMillipore Cat# 06-747; RRID:AB_310234WB (1:1000)
AntibodyAnti-mouse IFN gammaBio X CellClone: XMG1.2;
Cat# BE0055; RRID:AB_1107694
In vivo 100 µg
AntibodyGoat anti-rat polyclonal Alexa Fluor 594Thermo Fisher ScientificCat# A-21213; RRID:AB_2535799IFA (1:500)
AntibodyGoat anti-mouse polyclonal Alexa Fluor 488Thermo Fisher ScientificCat# A-11001; RRID:AB_2534069IFA (1:500)
Strain, strain backgroundAlexa Fluor 647 PhalloidinThermo Fisher ScientificA22287;
RRID:AB_2620155
IFA (1:1000)
AntibodyIRDye 800CW goat anti-rat IgGLI-COR926-32219;
RRID:AB_1850025
WB (1:10,000)
AntibodyIRDye 680RD goat anti-rabbit IgGLI-COR926-68071;
RRID:AB_2721181
WB (1:10,000)
Recombinant DNA reagentCas9 cgd5_4590 (plasmid)This paperGuide targeting C terminus of MEDLE2
Recombinant DNA reagentCas9 cgd8_3560 (plasmid)This paperGuide targeting C terminus of WYLE4
Recombinant DNA reagentCas9 cgd8_30 (plasmid)This paperGuide targeting C terminus of SKSR7
Recombinant DNA reagentCas9 cgd5_4580 (plasmid)This paperGuide targeting C terminus of MEDLE1
Recombinant DNA reagentCas9 Cgd6_5490 (plasmid)This paperGuide targeting C terminus of MEDLE6
Recombinant DNA reagentCas9 Tk guide int (plasmid)Tandel et al., 2019Guide targeting internal cgd5_4440
Recombinant DNA reagentCas9 MEDLE2
KO (plasmid)
This paperGuide targeting internal MEDLE2
Recombinant DNA reagentLic HA
(plasmid)
This paperCrypto expression vector for HA tagging
Recombinant DNA reagentLic tdTomato
KO (plasmid)
This paperCrypto expression vector for replacing gene KO with tdTomato
Recombinant DNA reagentLic medle2-MEDLE1 HA (plasmid)This paperCrypto expression vector with medle2 promoter driving MEDLE1-HA expression
Recombinant DNA reagentLic HA-2A-
TdNeon
(plasmid)
This paperCrypto expression vector for HA tagging and cytoplasmic tdNeon
Recombinant DNA reagentLic mScarlet (plasmid)This paperCrypto expression vector for mScarlet tagging
Recombinant DNA reagentLic Bla-2A-TdTomato
(plasmid)
This paperCrypto expression vector for BLA tagging and cytoplasmic tdTomato
Recombinant DNA reagentLic Cre
(plasmid)
This paperCrypto expression vector for Cre tagging
Recombinant DNA reagentLic Extra
MEDLE2-HA (plasmid)
This paperCrypto expression vector for extra copy of MEDLE2-HA (MEDLE2 promoter)
Recombinant DNA reagentLic ΔSP MEDLE2-HA (plasmid)This paperCrypto expression vector for extra copy of MEDLE2-HA (aa 21–209)
Recombinant DNA reagentLic KDVSLI/6A-HA (plasmid)This paperCrypto expression vector for extra copy of MEDLE2-HA with KDVSLI (aa 31–36) mutated to six alanines
Recombinant DNA reagentLic KPVLKN/6A-HA (plasmid)This paperCrypto expression vector for extra copy of MEDLE2-HA with KPVLKN (aa 73–78) mutated to six alanines
Recombinant DNA reagentLic KNVNLS/6A-HA (plasmid)This paperCrypto expression vector for extra copy of MEDLE2-HA with KNVNLS (aa 77–82) mutated to six alanines
Recombinant DNA reagentLic RGLLRGLSG/9A-HA (plasmid)This paperCrypto expression vector for extra copy of MEDLE2-HA with RGLLRGLS (aa 191–199) mutated to six alanines
Recombinant DNA reagentLic K31A-HA (plasmid)This paperCrypto expression vector for extra copy of MEDLE2-HA with K31 mutated to alanine
Recombinant DNA reagentLic D32A-HA (plasmid)This paperCrypto expression vector for extra copy of MEDLE2-HA with D32 mutated to alanine
Recombinant DNA reagentLic V33A-HA (plasmid)This paperCrypto expression vector for extra copy of MEDLE2-HA with V33 mutated to alanine
Recombinant DNA reagentLic S34A-HA (plasmid)This paperCrypto expression vector for extra copy of MEDLE2-HA with S34 mutated to alanine
Recombinant DNA reagentLic L35A-HA (plasmid)This paperCrypto expression vector for extra copy of MEDLE2-HA with L35 mutated to alanine
Recombinant DNA reagentLic I36A-HA (plasmid)This paperCrypto expression vector for extra copy of MEDLE2-HA with I36 mutated to alanine
Recombinant DNA reagentmEGFP-Lifeact-7 (plasmid)Addgene# 54610Used as a mammalian expression vector to clone codon optimized MEDLE2 into
Recombinant DNA reagentGFP-onlyThis paperRemoved Lifeact domain from Addgene plasmid #54610 for a GFP-only control plasmid
Recombinant DNA reagentRecod MEDLE2-GFP (plasmid)This paperHuman codon optimized MEDLE2 (aa 21–209) with a GFP tag
Recombinant DNA reagentGRA16-GFPThis paperT. gondii GRA16 (aa 24–505) with GFP tag.
Recombinant DNA reagentRecod MEDLE2-HA (plasmid)This paperHuman codon optimized MEDLE2 (aa 21–209) with a HA tag.
Recombinant DNA reagentRecod KDVSLI/6A-HA (plasmid)This paperHuman codon optimized MEDLE2 (aa 21–209) with a HA tag and KDVSLI (aa 31–36) mutated to six alanines
Recombinant DNA reagentRecod KPVLKN/6A-HA (plasmid)This paperHuman codon optimized MEDLE2 (aa 21–209) with a HA tag and KPVLKN (aa 73–78) mutated to six alanines
Recombinant DNA reagentRecod KNVNLS/6A-HA (plasmid)This paperHuman codon optimized MEDLE2 (aa 21–209) with a HA tag and KNVNLS (aa 77–82) mutated to six alanines
Recombinant DNA reagentRecod RGLLRGLSG/9A-HA (plasmid)This paperHuman codon optimized MEDLE2 (aa 21–209) with a HA tag and RGLLRGLS (aa 191–199) mutated to six alanines
Sequence-based
reagent
Recodonized MEDLE2Integrated DNA TechnologiesMEDLE2 (aa 21–209) codon optimized for human expressionSee Supplementary file 1 for sequence
Sequence-based
reagent
PCR primersThis paperPlease see Supplementary file 1
Commercial assay or kitDNeasy Blood & Tissue KitQIAGENCat# 69504
Commercial assay or kitZymoPureII Plasmid Maxiprep KitZymo ResearchCat# 11-555B
Commercial assay or kitNano-Glo Luciferase Assay SystemPromegaCat# N1130
Commercial assay or kitSF Cell Line 4D X Kit LLonzaCat# V4XC-2024
Commercial assay or kitLiveBLAzer FRET-B/G Loading KitThermo FisherCat# K1095
Commercial assay or kitZeroBlunt TopoTA KitInvitrogenCat# 450245
Commercial assay or kitRNeasy MicrokitQIAGENCat# 74004
Commercial assay or kitSMART cDNA synthesis kitTakaraCat# 635040
Commercial assay or kitNextera XT DNA Library Prep KitIlluminaCat# FC-131-1096
Commercial assay or kitRNeasy MiniKitQIAGENCat# 74104
Commercial assay or kitQIAshredderQIAGENCat# 79656
Commercial assay or kitSuperScript First Strand Synthesis kitThermo FisherCat#18091050
Commercial assay or kitLipofectamine 3000Thermo FisherCat# L3000015
Chemical compound, drugParomomycinGeminiCat# 400-155PUsed 16 g/L water
Chemical compound, drugBrefeldin A (BFA)BioLegendCat# 420601Used 10 µg/mL
Chemical compound, drugThapsigarginMedChemExpressHY-13433Used 1 µM
Chemical compound, drugGSK2606414MedChemExpressHY-18072Used 30 nm
Chemical compound, drugKIRA6MedChemExpressHY-19708Used 500 nm
Software, algorithmPrism 8GraphPadRRID:SCR_002798
Software, algorithmImageJFijiRRID:SCR_003070
Software, algorithmFlowJo v10, LLCTreeStarRRID:SCR_008520
Software, algorithmKallisto v0.44.0BioConductor (Bray et al., 2016)Pachter Lab
Software, algorithmLimma-VoomBioConductor (Law et al., 2014; Ritchie et al., 2015)
Software, algorithmBioconductor tximportBioConductor (Robinson et al., 2010)DOI: 10.18129/B9.bioc.tximport
Software, algorithmMolecular Signatures Database (MSigDB)UC San Diego and Broad Institute (Mootha et al., 2003; Subramanian et al., 2005)https://www.gsea-msigdb.org/gsea/msigdb
Software, algorithmCryptoDBVEuPathDBcryptodb.org
OtherFluorescin Vicia villosa lectin stainVector LabsCat# FL-1231-2IF (1:1000)
OtherDAPI stainInvitrogenCat# D1306Flow cytometry (1 µg/mL)
OtherHoechst 33342Thermo FisherCat# H3570IF (1:10,000)
OtherAlexa Fluor 647 PhalloidinThermo FisherCat# A22287IF (1:1000)

Additional files

Transparent reporting form
https://cdn.elifesciences.org/articles/70451/elife-70451-transrepform1-v2.docx
Supplementary file 1

Primer sequences used for this study.

https://cdn.elifesciences.org/articles/70451/elife-70451-supp1-v2.xlsx
Source code 1

Supplemental code detailing the R packages used for analysis of the MEDLE2 transfection RNAsequencing dataset.

https://cdn.elifesciences.org/articles/70451/elife-70451-supp2-v2.zip

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Jennifer E Dumaine
  2. Adam Sateriale
  3. Alexis R Gibson
  4. Amita G Reddy
  5. Jodi A Gullicksrud
  6. Emma N Hunter
  7. Joseph T Clark
  8. Boris Striepen
(2021)
The enteric pathogen Cryptosporidium parvum exports proteins into the cytosol of the infected host cell
eLife 10:e70451.
https://doi.org/10.7554/eLife.70451