Abstract

Simple SummaryCRISPR/Cas9 system as a potential gene editing platform has been widely applied in biological engineering and disease therapies. To achieve precise gene targeting, active CRISPR/Cas9 components must be efficiently transported to targeted cells. As a simple and effective strategy, Cold Atmospheric Plasma (CAP) treatment has been demonstrated for the transmembrane delivery of various exogenous materials. In comparison with carrier-dependent delivery methods, this carrier-free platform provides a promising alternative to circumvent the obstacles of biosafety and complicated preparation processes. In this work, a CAP-based CRISPR/Cas9 carrier-free delivery platform has been established and corresponding mechanism related to efficient transportation has been explored. Briefly, the efficient production of bioactive species in culture media after CAP treatment alters cell membrane potential and permeability, which facilitates cytosolic delivery of active CRISPR/Cas9 components via passive diffusion and ATP-dependent endocytosis pathways, resulting in efficient genome editing and gene silencing. This carrier-free strategy using CAP-based transportation may also be extended to other active biomolecules in drug delivery and gene therapy.A carrier-free CRISPR/Cas9 ribonucleoprotein delivery strategy for genome editing mediated by a cold atmospheric plasma (CAP) is described. The CAP is promising in many biomedical applications due to efficient production of bioactive ionized species. The MCF-7 cancer cells after CAP exposure exhibit increased extracellular reactive oxygen and nitrogen species (RONS) and altered membrane potential and permeability. Hence, transmembrane transport of Ca2+ into the cells increases and accelerates ATP hydrolysis, resulting in enhanced ATP-dependent endocytosis. Afterwards, the increased Ca2+ and ATP contents promote the release of cargo into cytoplasm due to the enhanced endosomal escape. The increased membrane permeability also facilitates passive diffusion of foreign species across the membrane into the cytosol. After CAP exposure, the MCF-7 cells incubated with Cas9 ribonucleoprotein (Cas9-sgRNA complex, Cas9sg) with a size of about 15 nm show 88.9% uptake efficiency and 65.9% nuclear import efficiency via passive diffusion and ATP-dependent endocytosis pathways. The efficient transportation of active Cas9sg after the CAP treatment leads to 21.7% and 30.2% indel efficiencies in HEK293T and MCF-7 cells, respectively. This CAP-mediated transportation process provides a simple and robust alternative for the delivery of active CRISPR/Cas9 ribonucleoprotein. Additionally, the technique can be extended to other macro-biomolecules and nanomaterials to cater to different biomedical applications.

Highlights

  • Cas9 (CRISPR-associate protein 9) is an RNA-guided endonuclease protein associated with the CRISPR system, which is involved in bacteria adaptive immunity against invading nucleic acids [1]

  • A grounded electrode was wrapped around the nozzle of the outer dielectric tube with a cooper sheet

  • Biology 2021, 10, x FOR PEER REVIEW show lower fluorescence signals from Cas9sg-488 and less changes in fluorescence signals9 of 18 after being pre-treated with inhibitors (Supporting Information, Figures S11 and S12) as compared with cells treated with cold atmospheric plasma (CAP), suggesting that the endocytosis of Cas9sg-488 is very weak in cells without CAP exposures. These results suggest that endocytosis is the and nuclear import of Cas9sg-488 are both extremely low, confirming that CAP can major cellular uptake pathway

Read more

Summary

Introduction

Cas (CRISPR-associate protein 9) is an RNA-guided endonuclease protein associated with the CRISPR (clustered regularly interspaced short palindromic repeats) system, which is involved in bacteria adaptive immunity against invading nucleic acids [1]. Various approaches of CRISPR/Cas have been developed to deliver DNA fragments encoding Cas ribonucleoprotein (Cas9-sgRNA complex, Cas9sg) via microinjection [8,9], electroporation [10,11], or viral vectors [12,13]. DNA fragments and associated overexpression of functional Cas9sg may increase the risk of insertional mutagenesis [14] as well as off-target effects [15]. To overcome these obstacles, cytosolic delivery of Cas9sg is a promising alternative with therapeutic potentials [16,17,18,19,20,21,22]. New strategies of carrier-free delivery of Cas ribonucleoprotein with excellent bioactivity are highly desirable

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