Summary

Determination of Chemical Inhibitor Efficiency against Intracellular Toxoplasma Gondii Growth Using a Luciferase-Based Growth Assay

Published: April 29, 2020
doi:

Summary

Presented here is a protocol to evaluate the inhibition efficacy of chemical compounds against in vitro intracellular growth of Toxoplasma gondii using a luciferase-based growth assay. The technique is used to confirm inhibition specificity by genetic deletion of the corresponding target gene. The inhibition of LHVS against TgCPL protease is evaluated as an example.

Abstract

Toxoplasma gondii is a protozoan pathogen that widely affects the human population. The current antibiotics used for treating clinical toxoplasmosis are limited. In addition, they exhibit adverse side effects in certain groups of people. Therefore, discovery of novel therapeutics for clinical toxoplasmosis is imperative. The first step of novel antibiotic development is to identify chemical compounds showing high efficacy in inhibition of parasite growth using a high throughput screening strategy. As an obligate intracellular pathogen, Toxoplasma can only replicate within host cells, which prohibits the use of optical absorbance measurements as a quick indicator of growth. Presented here is a detailed protocol for a luciferase-based growth assay. As an example, this method is used to calculate the doubling time of wild-type Toxoplasma parasites and measure the efficacy of morpholinurea-leucyl-homophenyl-vinyl sulfone phenyl (LHVS, a cysteine protease-targeting compound) regarding inhibition of parasite intracellular growth. Also described, is a CRISPR-Cas9-based gene deletion protocol in Toxoplasma using 50 bp homologous regions for homology-dependent recombination (HDR). By quantifying the inhibition efficacies of LHVS in wild-type and TgCPL (Toxoplasma cathepsin L-like protease)-deficient parasites, it is shown that LHVS inhibits wild-type parasite growth more efficiently than Δcpl growth, suggesting that TgCPL is a target that LHVS binds to in Toxoplasma. The high sensitivity and easy operation of this luciferase-based growth assay make it suitable for monitoring Toxoplasma proliferation and evaluating drug efficacy in a high throughput manner.

Introduction

Toxoplasma gondii is a highly successful obligate intracellular parasite that infects approximately one-third of the human population. Its high transmission rate is predominantly due to its diverse routes of transmission, including consumption of undercooked meat, exposure to mammalian reservoirs, and congenital transmission during birth. T. gondii mainly causes opportunistic infections that can lead to severe morbidity and mortality in immunocompromised individuals1,2,3,4,5,6. The antibiotics currently used for treating acute toxoplasmosis are particularly inefficient in treating congenital and latent infections and cause severe reactions in some individuals3,7,8. Thus, an urgent need to identify novel therapeutics exists. Understanding the differences in subcellular processes within Toxoplasma and its host will help to identify potential drug targets. Therefore, efficient and convenient genome manipulation techniques are required to study the roles of individual genes within Toxoplasma. Additionally, Toxoplasma belongs to the phylum Apicomplexa, which includes several other significant human pathogens, such as Plasmodium spp. and Cryptosporidium spp. Hence, Toxoplasma can be used as a model organism to help study basic biology in other apicomplexan parasites.

To identify novel antibiotics against microbial pathogens, high throughput screening of a library of chemical compounds is initially performed to determine their efficacy in the repression of microbial growth. So far, several microplate-based growth assays have been developed for measuring intracellular growth of T. gondii (i.e., radioactive 3H-uracil incorporation-based quantification9, quantitative ELISA-based parasite detection using T. gondii-specific antibodies10,11, reporter protein-based measurement using β-galactosidase or YFP-expressing Toxoplasma strains12,13, and a recently developed high-content imaging assay14).

These individual strategies all have unique advantages; however, certain limitations also restrict their applications. For example, since Toxoplasma can only replicate within nucleated animal cells, autofluorescence and non-specific binding of anti-T. gondii antibodies to host cells cause interference in fluorescence-based measurements. Furthermore, usage of radioactive isotopes requires special safety compliance and potential safety issues. Some of these assays are more suitable for assessing growth at a single timepoint rather than continuous monitoring of growth.

Presented here is a luciferase-based protocol for the quantification of intracellular Toxoplasma growth. In a previous study, the NanoLuc luciferase gene was cloned under the Toxoplasma tubulin promoter, and this luciferase expression construct was transfected into wild-type (RHΔku80Δhxg strain) parasites to create an RHΔku80Δhxg::NLuc strain (referred to as RHΔku80::NLuc hereafter)15. This strain served as the parental strain for intracellular growth determination and gene deletion in this study. Using the RHΔku80::NLuc strain, parasite growth in human foreskin fibroblasts (HFFs) was monitored over a 96 h period post-infection to calculate parasite doubling time.

In addition, the inhibition efficacy of LHVS against parasite growth can be determined by plotting Toxoplasma growth rates against serial LHVS concentrations to identify the IC50 value. Previous literature has reported that TgCPL is a major target of LHVS in parasites and that treatment with LHVS decreases the development of acute and chronic Toxoplasma infections16,17,18,19. Additionally, RHΔku80::NLuc was used as the parental strain for genome modification to generate a TgCPL-deficient strain (RHΔku80Δcpl::NLuc), and the inhibition of LHVS was measured against this mutant. By observing an upshift of IC50 values for LHVS in the TgCPL-deficient parasites compared to the WT strain, it was validated that TgCPL is targeted by LHVS in vivo.

In this protocol, RHΔku80::NLuc is used as the parental strain, which lacks an efficient non-homologous end-joining pathway (NHEJ), thereby facilitating double crossover homology-dependent recombination (HDR)20,21. Additionally, 50 bp homologous regions are flanked at both ends of a drug resistance cassette by PCR. The PCR product serves as a repair template to remove the entire gene locus via HDR using CRISPR-Cas9-based genome editing tools. Such short homologous regions can be easily incorporated into primers, providing a convenient strategy for production of the repair template. This protocol can be modified to perform universal gene deletion and endogenous gene tagging.

For instance, in our most recent publication, three protease genes, TgCPL, TgCPB (Toxoplasma cathepsin B-like protease), and TgSUB1 (Toxoplasma subtilisin-like protease 1), were genetically ablated in TgCRT (Toxoplasma chloroquine-resistance transporter)-deficient parasites using this method15. Additionally, TgAMN (a putative aminopeptidase N [TgAMN, TGGT1_221310]) was endogenously tagged15. The Lourido lab also reported using short homologous regions in the range of 40-43 bp for the introduction of site-directed gene mutation and endogenous gene tagging in the Toxoplasma genome using a similar method22. These successful genome modifications suggest that a 40-50 bp homologous region is sufficient for efficient DNA recombination in the TgKU80-deficient strain, which greatly simplifies genome manipulation in Toxoplasma gondii.

Protocol

Toxoplasma gondii is categorized in Risk Group 2 and must be handled at a Biosafety Level 2 (BSL-2). The protocol has been reviewed and approved by the Institutional Biosafety Committee at Clemson University. 1. Luciferase-based Toxoplasma growth assay Seed human foreskin fibroblasts (HFFs) 1 week before parasite inoculation to ensure that host cells are fully confluent. Perform a mock assay in a transparent plate to ensure that parasites remain intracellular thro…

Representative Results

Figure 1 represents an example of a growth curve for the RHΔku80::NLuc strain and the derived calculation for its doubling time. Generally, the assay is performed in three technical replicates for each of the three biological replicates to account for variations of luciferase activity readings. In order to calculate the normalized fold change of parasite growth, each reading at 24-96 h post-infection was divided by the initial reading a…

Discussion

++This protocol describes a luciferase-based protocol to assess intracellular Toxoplasma growth and evaluate the inhibition efficacy of chemical compounds against parasite growth. Compared to the existing strategies available for measuring intracellular Toxoplasma growth, this method exhibits high sensitivity and specificity. While monitoring parasite growth, a mock assay in a clear 96 well microplate is recommended to confirm that the tested strain does not prematurely lyse host cells before the end of…

Disclosures

The authors have nothing to disclose.

Acknowledgements

The authors would like to thank Drs. Sibley and Carruthers for sharing pSAG1-Cas9-sgRNA-TgUPRT plasmid and anti-TgCPL and TgActin antibodies. This work was supported by the Clemson Startup fund (to Z.D.), Knights Templar Eye Foundation Pediatric Ophthalmology Career-Starter Research Grant (to Z.D.), a pilot grant of an NIH COBRE grant P20GM109094 (to Z.D.), and NIH R01AI143707 (to Z.D.). The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Materials

Agarose gel extraction kit New England BioLabs T1020L
BamHI New England BioLabs R0316S
Biotek Synergy H1 Hybrid Multi-Mode Microplate Reader BioTek Instuments
BTX Gemini Twin Waveform Electroporation System Harvard Apparatus
Chemically competent E. coli cells New England BioLabs C29871
CloneAmp HiFi PCR premix Takara Bio 639298
Coelenterazine h Prolume 301-10 hCTZ
EcoRV New England BioLabs R3195S
Phire Tissue Direct PCR Master Mix Thermo Scientific F170L
Plasmid miniprep kit Zymo Research D4054
Q5 Site-Directed Mutagenesis kit New England BioLabs E0554S
Software
Geneious software for sgRNA design (version: R11)
GraphPad Prism software (8th version)
SnapGene for molecular cloning (version: 4.2.11)

References

  1. Blader, I. J., Coleman, B. I., Chen, C. T., Gubbels, M. J. Lytic Cycle of Toxoplasma gondii: 15 Years Later. Annual Review of Microbiology. 69 (1), 1-23 (2014).
  2. Jones, J. L., Kruszon-Moran, D., Rivera, H., Price, C., Wilkins, P. P. Toxoplasma gondii Seroprevalence in the United States 2009-2010 and Comparison with the Past Two Decades. The American Journal of Tropical Medicine and Hygiene. 90 (6), (2014).
  3. Kieffer, F., Wallon, M. Congenital toxoplasmosis. Handbook of Clinical Neurology. 112, 1099-1101 (2013).
  4. Hoffmann, S., Batz, M. B., Morris, G. J. Annual cost of illness and quality-adjusted life year losses in the United States due to 14 foodborne pathogens. Journal of Food Protection. 75 (7), 1292-1302 (2012).
  5. Dubey, J. Toxoplasmosis. Journal of the American Veterinary Medical Association. 205 (11), 1593-1598 (1994).
  6. Lindsay, D., Dubey, J. Toxoplasma gondii: the changing paradigm of congenital toxoplasmosis. Parasitology. 138 (14), 1-3 (2011).
  7. Deng, Y., Wu, T., Zhai, S., Li, C. Recent progress on anti-Toxoplasma drugs discovery: Design, synthesis and screening. European Journal of Medicinal Chemistry. 183, 111711 (2019).
  8. Butler, N. J., Furtado, J. M., Winthrop, K. L., Smith, J. R. Ocular toxoplasmosis II: clinical features, pathology and management. Clinical & Experimental Ophthalmology. 41 (1), 95-108 (2013).
  9. Pfefferko, E., Pfefferko, L. C. Specific Labeling of Intracellular Toxoplasma gondii with Uracil. Journal of Eukaryotic Microbiology. 24 (3), 449-453 (1977).
  10. Merli, A., Canessa, A., Melioli, G. Enzyme immunoassay for evaluation of Toxoplasma gondii growth in tissue culture. Journal of Clinical Microbiology. 21 (1), 88-91 (1985).
  11. Derouin, F., Chastang, C. Enzyme immunoassay to assess effect of antimicrobial agents on Toxoplasma gondii in tissue culture. Antimicrobial Agents and Chemotherapy. 32 (3), 303-307 (1988).
  12. McFadden, D., Seeber, F., Boothroyd, J. Use of Toxoplasma gondii expressing beta-galactosidase for colorimetric assessment of drug activity in vitro. Antimicrobial Agents and Chemotherapy. 41 (9), 1849-1853 (1997).
  13. Gubbels, M. J., Li, C., Striepen, B. High-Throughput Growth Assay for Toxoplasma gondii Using Yellow Fluorescent Protein. Antimicrobial Agents and Chemotherapy. 47 (1), 309-316 (2003).
  14. Touquet, B., et al. High-content imaging assay to evaluate Toxoplasma gondii infection and proliferation: A multiparametric assay to screen new compounds. PLoS ONE. 13 (8), e0201678 (2018).
  15. Thornton, L. B., et al. An ortholog of Plasmodium falciparum chloroquine resistance transporter (PfCRT) plays a key role in maintaining the integrity of the endolysosomal system in Toxoplasma gondii to facilitate host invasion. PLOS Pathogens. 15 (6), e1007775 (2019).
  16. Larson, E. T., et al. Toxoplasma gondii cathepsin L is the primary target of the invasion-inhibitory compound morpholinurea-leucyl-homophenyl-vinyl sulfone phenyl. The Journal of Biological Chemistry. 284 (39), 26839-26850 (2009).
  17. Dou, Z., McGovern, O. L., Cristina, M., Carruthers, V. B. Toxoplasma gondii Ingests and Digests Host Cytosolic Proteins. mBio. 5 (4), e01188-14 (2014).
  18. Cristina, M., et al. Toxoplasma depends on lysosomal consumption of autophagosomes for persistent infection. Nature Microbiology. 2, 17096 (2017).
  19. Parussini, F., Coppens, I., Shah, P. P., Diamond, S. L., Carruthers, V. B. Cathepsin L occupies a vacuolar compartment and is a protein maturase within the endo/exocytic system of Toxoplasma gondii. Molecular Microbiology. 76 (6), 1340-1357 (2010).
  20. Huynh, M. H., Carruthers, V. B. Tagging of endogenous genes in a Toxoplasma gondii strain lacking Ku80. Eukaryotic cell. 8 (4), 530-539 (2009).
  21. Fox, B. A., Ristuccia, J. G., Gigley, J. P., Bzik, D. J. Efficient gene replacements in Toxoplasma gondii strains deficient for nonhomologous end joining. Eukaryotic Cell. 8 (4), 520-529 (2009).
  22. Sidik, S. M., Hackett, C. G., Tran, F., Westwood, N. J., Lourido, S. Efficient Genome Engineering of Toxoplasma gondii Using CRISPR/Cas9. PLoS ONE. 9 (6), e100450 (2014).
  23. Shen, B., Brown, K. M., Lee, T. D., Sibley, D. L. Efficient Gene Disruption in Diverse Strains of Toxoplasma gondii Using CRISPR/CAS9. mBio. 5 (3), e01114-14 (2014).
  24. Radke, J. R., et al. Defining the cell cycle for the tachyzoite stage of Toxoplasma gondii. Molecular and Biochemical Parasitology. 115 (2), 165-175 (2001).
  25. Ran, A. F., et al. Genome engineering using the CRISPR-Cas9 system. Nature Protocols. 8 (11), 2281-2308 (2013).
  26. Labun, K., Montague, T. G., Gagnon, J. A., Thyme, S. B., Valen, E. CHOPCHOP v2: a web tool for the next generation of CRISPR genome engineering. Nucleic Acids Research. 44 (W1), W272-W276 (2016).
  27. Heigwer, F., Kerr, G., Boutros, M. E-CRISP: fast CRISPR target site identification. Nature Methods. 11 (2), 2812 (2014).
  28. Peng, D., Tarleton, R. EuPaGDT: a web tool tailored to design CRISPR guide RNAs for eukaryotic pathogens. Microbial Genomics. 1 (4), e000033 (2015).
  29. Doench, J. G., et al. Rational design of highly active sgRNAs for CRISPR-Cas9-mediated gene inactivation. Nature Biotechnology. 32 (12), 1262-1267 (2014).
  30. Sidik, S. M., et al. A Genome-wide CRISPR Screen in Toxoplasma Identifies Essential Apicomplexan Genes. Cell. 166 (6), 1423-1435 (2016).
check_url/60985?article_type=t

Play Video

Cite This Article
Key, M., Bergmann, A., Micchelli, C., Thornton, L. B., Millard, S., Dou, Z. Determination of Chemical Inhibitor Efficiency against Intracellular Toxoplasma Gondii Growth Using a Luciferase-Based Growth Assay. J. Vis. Exp. (158), e60985, doi:10.3791/60985 (2020).

View Video