Abstract

CRISPR-Cas9 technologies have enabled genome engineering in an unprecedented array of species, accelerating biological studies in both model and nonmodel systems. However, Cas9 can be inherently toxic, which has limited its use in some organisms. We previously described the serendipitous discovery of a single guide RNA (sgRNA) that helped overcome Cas9 toxicity in the apicomplexan parasite Toxoplasma gondii, enabling the first genome-wide loss-of-function screens in any apicomplexan. Even in the presence of the buffering sgRNA, low-level Cas9 toxicity persists and results in frequent loss of Cas9 expression, which can affect the outcome of these screens. Similar Cas9-mediated toxicity has also been described in other organisms. We therefore sought to define the requirements for stable Cas9 expression, comparing different expression constructs and characterizing the role of the buffering sgRNA to understand the basis of Cas9 toxicity. We find that viral 2A peptides can substantially improve the selection and stability of Cas9 expression. We also demonstrate that the sgRNA has two functions: primarily facilitating integration of the Cas9-expression construct following initial genome targeting and secondarily improving long-term parasite fitness by alleviating Cas9 toxicity. We define a set of guidelines for the expression of Cas9 with improved stability and selection stringency, which are directly applicable to a variety of genetic approaches in diverse organisms. Our work also emphasizes the need for further characterizing the effects of Cas9 expression.IMPORTANCE Toxoplasma gondii is an intracellular parasite that causes life-threatening disease in immunocompromised patients and affects the developing fetus when contracted during pregnancy. Closely related species cause malaria and severe diarrhea, thereby constituting leading causes for childhood mortality. Despite their importance to global health, this family of parasites has remained enigmatic. Given its remarkable experimental tractability, T. gondii has emerged as a model also for the study of related parasites. Genetic approaches are important tools for studying the biology of organisms, including T. gondii As such, the recent developments of CRISPR-Cas9-based techniques for genome editing have vastly expanded our ability to study the biology of numerous species. In some organisms, however, CRISPR-Cas9 has been difficult to implement due to its inherent toxicity. Our research characterizes the basis of the observed toxicity, using T. gondii as a model, allowing us to develop approaches to aid the use of CRISPR-Cas9 in diverse species.

Highlights

  • CRISPR-Cas9 technologies have enabled genome engineering in an unprecedented array of species, accelerating biological studies in both model and nonmodel systems

  • Our initial studies suggested that Cas9 expression in the absence of an single guide RNA (sgRNA) is toxic for T. gondii

  • Characterizing the role of the coexpressed sgRNA, we found that its primary function is to enhance construct integration by targeting Cas9 to create a DNA double-strand breaks (DSBs)

Read more

Summary

Introduction

CRISPR-Cas technologies have enabled genome engineering in an unprecedented array of species, accelerating biological studies in both model and nonmodel systems. Related species cause malaria and severe diarrhea, thereby constituting leading causes for childhood mortality Despite their importance to global health, this family of parasites has remained enigmatic. Our research characterizes the basis of the observed toxicity, using T. gondii as a model, allowing us to develop approaches to aid the use of CRISPR-Cas in diverse species. While CRISPR-Cas technologies have enabled genome editing in an unprecedented array of species, their use is often limited by the cytotoxic effects of heterologous Cas expression. Motivated by the pioneering work in mammalian cell culture systems [7,8,9], we sought to use the CRISPR-Cas system to conduct genome-wide loss-offunction (LOF) screens in order to measure the fitness contribution of the many still-uncharacterized T. gondii genes. Recent studies that investigated the DNA-binding kinetics of dCas demonstrated that the protein can bind PAM sequences nonspecifically— especially in the absence of an sgRNA—which may explain the deleterious effects of both Cas and dCas9 [20,21,22]

Objectives
Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call