Parasites and cell cultures
N. caninum tachyzoites were cultured and passaged in Human Foreskin Fibroblast (HFF) and African Green Monkey kidney cells (vero) lines. The cells and parasites were maintained in a humidified incubator at 37°C with a 5% CO2 concentration. The standard growth medium used was Dulbecco’s Modified Eagle Medium (DMEM), supplemented with 10% fetal bovine serum (FBS, Gibco, USA), 25 mM glucose and 4 mM glutamine.
Generation of transgenic strains
The primer sequences are available in Table S1.
For the generation of NcGRA41-3xHA parasites, sgRNA was designed from the ncgra41 stop codon downstream, approximately 200-300bp sequence, using the ToxoDB database. The sgRNA sequence was incorporated to U6 downstream and AMP upstream primers. Subsequently, the Cas9 skeleton, U6 promotor and Amp fragments were amplified from the previously constructed pNcCRISPR/Cas9 plasmid in our lab. These fragments were then ligated by seamless cloning to form a new pNcCRISPR/Cas9 plasmid, named pNcCRISPR/Cas9::sgNcGRA41. The 5′ and 3′ flanks of the ncgra41 stop codon, front 1000bp, and sgRNA behind 1000bp sequences were amplified from the DNA of Nc1 parasites and inserted into the backbone of the pLIC-GRA41-DHFR-3×HA plasmid using seamless cloning. The 5′ flank-DHFR-3×HA-3′ flank fragment from the newly constructed pLIC-GRA41-DHFR-3×HA plasmid and pNcCRISPR/Cas9::sgNcGRA41 plasmid were co-transfected into Nc1 parasites and screened with pyrimethamine. Clones were verified through western blot (WB) and immunofluorescence assay (IFA).
To construct the NcGRA41 deficient strain (Δncgra41), 5′ and 3′ flank fragment of NcGRA41 gene before and behind 1000bp sequence were amplified from the genomic DNA of Nc1 parasites. DHFR and pTCR backbone were amplified from the previously generated pTCR-DHFR plasmid. The fragments were linked and sequenced to form a new pTCR-DHFR-NcGRA41 plasmid. The 5′ flank-DHFR-3′ flank fragment was then amplified from this plasmid. The pCRISPR/CAS9::sgΔncgra41 was constructed using a similar method as described above, with the sgRNA selected from the NcGRA41 gene. The linearized 5′ flank-DHFR-3′ flank fragment and pNcCRISPR/Cas9::sgΔncgra41 plasmid were co-transfected into Nc1 parasites and screened with pyrimethamine. Clones were verified through PCR.
For the generation of NcGRA41 complemented parasites (comΔncgra41), sgRNA was selected from the NcUPRT locus, and the pCRISPR/CAS9::sgNcUPRT was constructed accordingly, with sgRNA selected from the NcUPRT locus. The NcUPRT gene, before and behind 1000bp sequence, and the NcGRA41 gene were amplified from the DNA of Nc1 parasites. These fragments were then inserted into the pNcUPRT::DHFR backbone vector by homologous recombination to construct the pNcUPRT::DHFR-NcGRA41 plasmid. Finally, the pNcCRISPR/Cas9::sgNcUPRT plasmid and linearized fragment were co-transfected into Δncgra41 strain, and the complementary parasites were selected using fluorodeoxyribose (FUDR) and confirmed through PCR and IFA.
Western blot
Freshly isolated parasites were harvested and purified through a 5 µm filter, followed by centrifugation at 1400×g for 8 min, and washing in PBS. Purified tachyzoites were lysed in radio immunoprecipitation assay lysis buffer (RIPA) containing a cocktail of protease inhibitors. The lysates were then separated on 12.5% (w/v) SDS-PAGE gels and transferred onto polyvinylidene fluoride (PVDF) membranes (Millipore, MA, USA). The membranes were blocked with 5% (w/v) powdered milk (BD Difco, USA) and probed with a 1:500 dilution of mouse anti-HA antibody (Sigma-Aldrich) for 1 h. Mouse anti-NcActin antibody (National Animal Protozoa Laboratory, China Agricultural University) was used as a loading control for N. caninum F-actin subunit beta. After washing with PBST (PBS with 1% Tween-20), the membranes were incubated with an HRP-conjugated goat anti-mouse IgG (Sigma, USA) at 1:5 000 dilution for 1 h. Following three washes with PBST, visualization was performed using Plus ECL (CoWin Biotech Co., Ltd, China).
Immunofluorescence assay
Tachyzoites (1×104) were seeded onto HFFs previously prepared on glass coverslips in 12-well plates and incubated at 37℃ with 5% CO2 for 30 h. and then fixed for 30 in 4% formaldehyde for 30 min. Subsequently, cells were permeabilized with 0.25% Triton X-100 for 15 min, followed by blocking with 3% bovine serum albumin (BSA) for 1 h. The cells were incubated with a mouse anti-HA monoclonal antibody (1:500, Sigma-Aldrich), rabbit anti-NcGRA1 polyclonal antibody (1:200), and rabbit anti-NcSRS2 polyclonal antibody (1:500) at 37 ℃ for 1 h. Following incubation, parasitess were treated with FITC-conjugated goat-anti mouse IgG (H + L) and Cy3-conjugated goat-anti rabbit IgG (H + L) (Sigma, USA). Hoechst33258 (1:100, Sigma, USA) was used for nuclear DNA staining. Images were captured with a Leica confocal microscope system (Leica, TCS SP52, Germany). Antibodies against NcGRA1 and NcSRS2 were provided by the National Animal Protozoa Laboratory, China Agricultural University. To analyze NcGRA41 localization in extracellular parasites, filtered parasites were allowed to adhere to coverslips precoated with poly-lysine at 4℃ overnight. The IFA process and image collection followed the procedures described above.
Plaque assay
Plaque assay was performed using HFFs cultured in 12-well cell culture plates. The HFFs monolayer, reaching 80–90% confluency, was infected with 200 freshly isolated parasites per well and then incubated in a 37°C incubator with 5% CO2 for 7 days. After incubation, cells were washed twice with PBS, fixed with 4% paraformaldehyde, and stained with 2% crystal violet solution for 2–4 h. Plaque areas were measured as described previously, and data were compiled from three independent experiments [22].
Invasion assay
Invasion assay was performed by inoculating freshly isolated parasites (1×106) on HFFs in12-well cell culture plates. After invasion for 1 hour at 37°C, extracellular parasites were removed by washing with PBS. Following 30 h of incubation, infected cells were fixed with 4% paraformaldehyde. Parasites were then stained with a rabbit anti-NcSRS2 polyclonal antibody (1:500), and host cell nucleus were stained with Hoechst33258 (1:100, Sigma, USA) following the IFA protocol. For each experiment, a minimum of 100 parasites were counted from 10 fields per treatment. Invasion efficiency was calculated as the ratio of the numbers of vacuoles to the numbers of host nuclei. Results represent the mean ± standard deviation of three independent biological replicate experiments.
Proliferation assay
A total of 1×106 freshly tachyzoites were inoculated into HFF cells cultured in 12-well cell culture plates. After 1 hour invasion, the cells were washed with PBS three times, and fresh medium was added for continuously cultivation for 30 h. Infected HFFs were fixed with 4% paraformaldehyde and permeabilized with 0.25% Triton X-100. The parasites were labeled with a rabbit anti-NcSRS2 polyclonal antibody (1:500), and the nucleus was stained with Hoechst33258. At least 100 vacuoles were quantified per strain per experiment, and the number of tachyzoites contained in each vacuole was counted separately (e.g., 2, 4, 8, 16 tachyzoites). Three independent experiments were conducted.
Egress assay
HFFs monolayers on 12-well coverslip were infected with 1×105 parasites for 1 hour at 37°C. The medium containing uninvaded parasites was removed, and the cells were washed with PBS. Fresh 2% FBS DHEM medium was added and the cells were allowed to grow for 30–33 h until large vacuoles were visualized, but parasites were not released, as observed under an inverted microscope. For induction of egress, host cells were incubated for 3–5 min with PBS containing 1 µM A23187 or 5% absolute ethyl alcohol. This process was carried out in the dark. Following this, cells were fixed with 4% paraformaldehyde, and the subsequent IFA assay was conducted, connecting with invasion protocol. A minimum of 100 parasites were counted from randomly selected fields. The efficiency of egress was calculated as the ratio of vacuoles with released parasites to the total counted number of vacuoles. Three independent experiments were performed.
Gliding assay
Glass coverslips were coated overnight at 4°C with 20% FBS dissolved in PBS. Freshly lysed and filtered tachyzoites were resuspended in Ringer buffer (composition: NaCl 9.1 g, KCl 0.23 g, CaCl2·2H2O 0.29 g, MgCl2 ·6H2O 0.2 g, NaH2PO4·2H2O 0.468 g, HEPEs 2.4 g, Glucose 1.8 g, dissolved in 900 mL distilled water). Subsequently, 250 µL of this suspension was added to the coated slides and incubated for 4–5 min on ice. Ringer buffer containing 2 µM A23187 was then added, followed by incubation for 20 min at 37℃. The slides were fixed with 4% paraformaldehyde. Trails left by gliding parasites were visualized by staining with rabbit anti-NcSRS2 antibody (1:500). A total of 100 parasites were enumerated per strain/treatment, and the numbers of parasites trails was recoded to calculate the proportion of gliding tachyzoites. Trail lengths were measured using ImageJ. Three independent experiments were performed.
Parasite virulence assay
To assess the virulence of the transgenic strain compared to the Nc1 strain, groups of 5 female BALB/c mice aged between 7–8 weeks were intraperitoneally (i. p.) infected with 8×106 tachyzoites post-adaptation. Mice were monitored daily over a 60-day period for clinical symptoms and survival time post-infection. Data were collected from three independent experiments. Throughout the entire experiment, mice were provided with an ample food and water, along with appropriate environmental conditions to ensure their well-being.
Quantitative real-time PCR
A total of 1×107 tachyzoites collected at 46–48 h post-invasion were used for total RNA extraction following the manufacturer’s instructions with TRIZOL® reagent (Invitrogen). The concentration of RNA was measured using a NanoDrop 2000c spectrophotometer (Thermo Scientific). Equal amounts of total RNA were then synthesized into cDNA utilizing a First-Strand cDNA Synthesis kit (Invitrogen). ABgene Absolute qPCR SYBR Green Master Mix (ABgene, Surrey, UK) with ROX dye was used for all PCR protocols. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) reactions were conducted in triplicate in 96-well plates on an ABI Prism®7900HT real-time PCR machine. The thermal cycling conditions consisted of one cycle at 50°C for 2 min for enzyme activation, followed by one cycle at 95°C for 15 min for enzyme activation. Subsequently, forty cycles were run at 95°C for 15 sec for denaturation and 60°C for 1 min for the anneal/extension step. The threshold cycle number (Ct) of the target gene was calculated, with NcActin serving as the reference gene. Delta–delta Ct values of the genes were presented as relative fold induction.
Microneme secretion assay
The microneme secretion assay was conducted following a previously established protocol [23]. In brief, 1×108 purified tachyzoites from both Nc1 and Δncgra41 strains were suspended in 1 mL DMEM containing 1 µM A23187. The mixture was incubated at 37°C in a water bath for 30 min. Subsequently, the mixture was centrifuged at 1300×g for 15 min at 4°C to separate the supernatant and precipitation. The precipitation was then combined with 98 µL RIPA and 2 µL Cocktail, followed by a 30 min incubation on ice. The supernatants and lysate were processed using SDS-PAGE followed by a western blot protocol to detect the secretion and expression of NcMICs. The primary antibodies used included mouse anti-NcActin monoclonal antibody (1:4 000), mouse anti-NcMIC1 polyclonal antibody (1:300), mouse anti-NcMIC4 polyclonal antibody (1:300), and mouse anti-NcMIC8 polyclonal antibody (1:1 000). The secondary antibody used was an HRP labeled goat anti-mouse lgG monoclonal antibody (1:5 000).
Statistical analysis
The statistical analysis was conducted using GraphPad Prism 5. Log-rank (Mantel-Cox) Test was used for data analysis. The results were presented as mean ± SD, and values with P < 0.05 and P < 0.01 were considered statistically significant.