Abstract

BackgroundReporter methods to quantitatively measure the efficiency and specificity of genome editing tools are important for the development of novel editing techniques and successful applications of available ones. However, the existing methods have major limitations in sensitivity, accuracy, and/or readiness for in vivo applications. Here, we aim to develop a straight-forward method by using nucleotide insertion/deletion resulted from genome editing. In this system, a target sequence with frame-shifting length is inserted after the start codon of a cerulean fluorescence protein (CFP) to inactivate its fluorescence. As such, only a new insertion/deletion event in the target sequence will reactivate the fluorescence. This reporter is therefore termed as “Insertion/deletion-activated frame-shift fluorescence protein”. To increase its traceability, an internal ribosome entry site and a red fluorescence protein mCherryFP are placed downstream of the reporter. The percentage of CFP-positive cells can be quantified by fluorescence measuring devices such as flow cytometer as the readout for genome editing frequency.ResultsTo test the background noise level, sensitivity, and quantitative capacity of this new reporter, we applied this approach to examine the efficiency of genome editing of CRISPR/Cas9 on two different targeting sequences and in three different cell lines, in the presence or absence of guide-RNAs with or without efficiency-compromising mutations. We found that the insertion/deletion-activated frame-shift fluorescence protein has very low background signal, can detect low-efficiency genome editing events driven by mutated guideRNAs, and can quantitatively distinguish genome editing by normal or mutated guideRNA. To further test whether the positive editing event detected by this reporter indeed correspond to genuine insertion/deletion on the genome, we enriched the CFP-positive cells to examine their fluorescence under confocal microscope and to analyze the DNA sequence of the reporter in the genome by Sanger sequencing. We found that the positive events captured by this reporter indeed correlates with genuine DNA insertion/deletion in the expected genome location.ConclusionThe insertion/deletion-activated frame-shift fluorescence protein reporter has very low background, high sensitivity, and is quantitative in nature. It will be able to facilitate the development of new genome editing tools as well as the application of existing tools.

Highlights

  • Reporter methods to quantitatively measure the efficiency and specificity of genome editing tools are important for the development of novel editing techniques and successful applications of available ones

  • Assuming completely random length in spontaneous in/del, there is a 33.3% chance the coding sequence for cerulean fluorescence protein (CFP) will become in-frame for the start codon, leading to reactivation of CFP fluorescence. This reporter is termed as frame-shift CFP (FsCFP)

  • We found that the FsCFP-mCherryFP clearly detected substantial positive events with the vascular endothelial growth factor (VEGF)-targeting guide RNA (gRNA), and a much reduced level with the mutant gRNA (Fig. 2b)

Read more

Summary

Introduction

Reporter methods to quantitatively measure the efficiency and specificity of genome editing tools are important for the development of novel editing techniques and successful applications of available ones. Through the efforts of many researchers in last two decades, a variety of genome editing tools have been introduced, which include meganucleases [2], zinc finger nucleases (ZFNs) [3], transcription activator-like effector-based nucleases (TALEN) [4], the clustered regularly interspaced short palindromic repeats (CRISPR) and CRISPR-associated genes (Cas) [5, 6]. To further propel the progress of genome editing for cell modification and eventual therapeutic application, extensive efforts are currently being invested for developing new genome editing tools to achieve higher efficiency, better precision, and wider application conditions. Such tasks will require a convenient method to quantitatively evaluate the efficiency and specificity of genome editing.

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