Elsevier

Methods in Enzymology

Volume 546, 2014, Pages 215-250
Methods in Enzymology

Chapter Eleven - The iCRISPR Platform for Rapid Genome Editing in Human Pluripotent Stem Cells

https://doi.org/10.1016/B978-0-12-801185-0.00011-8Get rights and content

Abstract

Human pluripotent stem cells (hPSCs) have the potential to generate all adult cell types, including rare or inaccessible human cell populations, thus providing a unique platform for disease studies. To realize this promise, it is essential to develop methods for efficient genetic manipulations in hPSCs. Established using TALEN (transcription activator-like effector nuclease) and CRISPR (clustered regularly interspaced short palindromic repeats)/Cas (CRISPR-associated) systems, the iCRISPR platform supports a variety of genome-engineering approaches with high efficiencies. Here, we first describe the establishment of the iCRISPR platform through TALEN-mediated targeting of inducible Cas9 expression cassettes into the AAVS1 locus. Next, we provide a series of technical procedures for using iCRISPR to achieve one-step knockout of one or multiple gene(s), “scarless” introduction of precise nucleotide alterations, as well as inducible knockout during hPSC differentiation. We present an optimized workflow, as well as guidelines for the selection of CRISPR targeting sequences and the design of single-stranded DNA (ssDNA) homology-directed DNA repair templates for the introduction of specific nucleotide alterations. We have successfully used these protocols in four different hPSC lines, including human embryonic stem cells and induced pluripotent stem cells. Once the iCRISPR platform is established, clonal lines with desired genetic modifications can be established in as little as 1 month. The methods described here enable a wide range of genome-engineering applications in hPSCs, thus providing a valuable resource for the creation of diverse hPSC-based disease models with superior speed and ease.

Introduction

Functional analysis of sequence variants affecting diverse human traits, including disease susceptibility, is a key to understanding human biology and disease mechanisms. With their unlimited self-renewal capacity and the potential to generate all adult cell types, human pluripotent stem cells (hPSCs), including human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs), offer an ideal platform for biological and disease studies (Zhu & Huangfu, 2013). To meet this goal, it is mandatory to develop efficient methods for genome engineering in hPSCs.

The development of programmable site-specific nucleases has significantly facilitated targeted-genome editing in a wide range of organisms and cultured cell types (Joung and Sander, 2013, Ran et al., 2013, Urnov et al., 2010). These customized nucleases induce DNA double-strand breaks (DSBs) at desired genomic loci, triggering the endogenous DNA repair machinery through two competing pathways: error-prone nonhomologous end-joining (NHEJ), leading to insertion/deletion mutations (Indels), or homology-directed repair (HDR), which can be co-opted to introduce precise nucleotide alterations using a homologous DNA template (Jasin, 1996, Rouet et al., 1994). Among various customized nuclease systems developed so far, the transcription activator-like effector (TALE) nuclease (TALEN) and the clustered, regularly interspaced, short palindromic repeat (CRISPR) technologies have emerged as powerful and versatile tools for genome editing in hPSCs.

The DNA target specificity of TALENs is guided by the TALE DNA-binding domain. Originally discovered in the plant pathogenic bacteria Xanthomonas, TALEs can bind to the promoter of various genes of the plant host, hijacking the transcriptional machinery to promote bacterial infection (Rossier et al., 1999, Szurek et al., 2001). The DNA-binding domain of TALE is composed of ∼ 34 amino acids repeats (TALE repeats) arranged in tandem. Each repeat contains two variable adjacent amino acids called the “repeat variable diresidue,” which determine the single base-recognition specificity. Thus, each TALE repeat independently specifies one target base (Boch et al., 2009, Moscou and Bogdanove, 2009). To introduce DSBs, TALENs are designed as pairs, recognizing the genomic sequences flanking the target site. Each TALEN consists of a programmable, sequence-specific TALE DNA-binding domain fused to the cleavage domain of the bacterial endonuclease FokI. The binding of a TALEN pair to DNA allows FokI dimerization and DNA cleavage (Cermak et al., 2011, Miller et al., 2011).

Recently, the CRISPR technology has been developed for genome engineering in mammalian systems (Cho et al., 2013, Cong et al., 2013, Jinek et al., 2013, Mali, Yang, et al., 2013, Wang et al., 2013). The CRISPR/Cas system is derived from Streptococcus pyogenes where it functions as part of an immune system to provide acquired resistance against invading viruses (van der Oost, Westra, Jackson, & Wiedenheft, 2014). CRISPR/Cas-mediated genome engineering requires two components: the constant RNA-guided DNA endonuclease Cas9 protein required for DNA cleavage and a variable CRISPR RNA (crRNA) and trans-activating crRNA (tracrRNA) duplex that specifies DNA target recognition (Jinek et al., 2012). Most applications now replace the crRNA/tracrRNA duplex with a single chimeric guide RNA (sgRNA), which works more efficiently than the original duplex design (Hsu et al., 2013, Jinek et al., 2012).

sgRNA directs Cas9 to its target genomic locus by recognizing a 20 nucleotide (nt) sequence (protospacer) followed by an NGG motif (protospacer-associated motif or PAM, where N can be A, T, G, or C), and DNA cleavage occurs 3 bp upstream of the PAM sequence. In our experience with hPSCs, the CRISPR/Cas system tends to outperform TALENs, which has also been observed by others (Ding et al., 2013). Compared to TALENs, the CRISPR/Cas system is easier to engineer and simplifies multiplexing. However, there have also been concerns regarding its off-target effects (Cho et al., 2014, Fu et al., 2013, Hsu et al., 2013, Mali, Aach, et al., 2013, Pattanayak et al., 2013), which will be discussed further in Section 6.

A number of studies have now used CRISPR/Cas to establish modified hPSC lines with variable efficiencies. Several studies use HDR-mediated editing to target a selectable marker into the locus of interest, which allows enrichment of correctly targeted cells after selection (An et al., 2014, Hou et al., 2013, Ye et al., 2014). Although efficient, the construction of the targeting construct could be time consuming, and it is often desirable to remove the selectable marker to allow more precise modeling of the disease conditions. Alternatively, the CRISPR/Cas system also supports efficient NHEJ or HDR-mediated genome editing without the need for drug selection (Ding et al., 2013, Gonzalez et al., 2014, Horii et al., 2013, Wang et al., 2014).

To further improve the efficiency, and to also achieve multiplexable and inducible genome editing in hPSCs, we have developed a genome-engineering platform called iCRISPR (Gonzalez et al., 2014). Through TALEN-mediated gene targeting, hPSC lines are engineered for doxycycline-inducible expression of Cas9 (referred to as iCas9 hPSCs). Upon doxycycline treatment, these lines can then be transfected with (a) a single or multiple sgRNA(s) to generate biallelic knockout hPSC lines for individual or multiple genes; (b) a sgRNA together with a HDR template to generate knockin alleles; and (c) a sgRNA at specific stages of hPSC differentiation to achieve inducible gene knockout.

Below we describe an optimized protocol for the establishment of the iCRISPR platform through TALEN-mediated targeting of inducible Cas9 expression cassettes into the AAVS1 locus of hPSCs (Fig. 11.1). We have successfully used this protocol on four different hPSC lines and obtained similar results: ~ 50% of the lines are correctly targeted with no additional random integrations. Next, we provide detailed protocols for using iCRISPR to achieve one-step knockout of one or multiple gene(s), “scarless” introduction of precise nucleotide alterations, as well as inducible knockout during hPSC differentiation. We also provide guidelines for the selection of CRISPR targeting sequence, and for the design of single-stranded DNA (ssDNA) HDR templates for introduction of specific nucleotide modifications. Based on our successful experience using both hESCs (including HUES8, HUES9, and MEL-1) and hiPSCs, we believe the methods described here are generally applicable to most hPSC lines with minor adjustment.

iCRISPR supports a wide range of genome-engineering applications, and once established, our optimized workflow enables the generation of clonal lines with desired genetic modifications in as little as 1 month. Genome editing in hPSCs may finally become a routine laboratory procedure instead of a difficult and time-consuming task.

Section snippets

Generation of iCas9 hPSCs

Compared with transient, plasmid-mediated expression, targeted integration, and inducible expression of Cas9 from a “safe harbor” locus provides a precise and reliable approach to express the invariable component of the CRISPR/Cas system. In this configuration, Cas9-expressing hPSCs can be easily transfected with sgRNAs due to their small size (~ 100 nucleotides), leading to reproducible and highly efficient genome editing in target loci.

We generate iCas9 hPSCs by TALEN-mediated gene targeting

sgRNA design

NHEJ-mediated repair of DNA DSBs leads to random Indel mutations, which is useful for generating loss-of-function mutations or knockouts. Thus, the design of the gene targeting strategy aims at generating premature stop codons through the creation of frameshift Indel mutations, and the target sequence need to be strategically chosen to maximize the possibility of disrupting the function of the corresponding protein. It is important to identify all possible splice variants of the gene of

Generation of Precise Nucleotide Alterations Using iCRISPR

Without a DNA repair template, NHEJ repair of DSBs introduces random Indels. However, in the presence of a DNA repair template, HDR of DSBs could lead to precise nucleotide alterations of the hPSC genome, which is important for either creating disease-specific variants in wild-type cells or correcting disease-associated mutations in patient cells. Compared with double-stranded DNAs, synthetic ssDNAs (~ 80–200 nt) are easy to produce and can be used as DNA repair template without selection (Chen,

Inducible Gene Knockout in hPSCs Using iCRISPR

Inducible gene knockout during differentiation of hPSCs into specific cell types is of great importance for studying genes with pleiotropic effects. With the iCRISPR platform, inducible gene knockout could be achieved by inducible Cas9 expression and temporally regulated delivery of sgRNA due to the low toxicity of lipid-mediated sgRNA transfection (Fig. 11.6A). Alternatively, inducible gene knockout could also be achieved through generation of iCr lines including a constitutive sgRNA

Conclusions and Future Directions

Below we discuss expected results, common issues and considerations, and potential additional use and extension of the iCRISPR platform.

Acknowledgments

We thank Feng Zhang and Rudolf Jaenisch for providing vectors through Addgene, and Dirk Hockemeyer for the Neo-M2rtTA donor. This study was funded in part by NIH (R01DK096239), NYSTEM (C029156), and March of Dimes Foundation (Basil O'Connor Starter Scholar Research Award Grant 5-FY12-82). F.G. and Z.Z. were supported by the New York State Stem Cell Science fellowship from the Center for Stem Cell Biology of the Sloan-Kettering Institute

References (51)

  • M.C. An et al.

    Polyglutamine disease modeling: Epitope based screen for homologous recombination using CRISPR/Cas9 system

    PLoS Currents

    (2014)
  • S. Bae et al.

    Microhomology-based choice of Cas9 nuclease target sites

    Nature Methods

    (2014)
  • J. Boch et al.

    Breaking the code of DNA binding specificity of TAL-type III effectors

    Science

    (2009)
  • T. Cermak et al.

    Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting

    Nucleic Acids Research

    (2011)
  • G. Chen et al.

    Chemically defined conditions for human iPSC derivation and culture

    Nature Methods

    (2011)
  • F. Chen et al.

    High-frequency genome editing using ssDNA oligonucleotides with zinc-finger nucleases

    Nature Methods

    (2011)
  • S.W. Cho et al.

    Targeted genome engineering in human cells with the Cas9 RNA-guided endonuclease

    Nature Biotechnology

    (2013)
  • S.W. Cho et al.

    Analysis of off-target effects of CRISPR/Cas-derived RNA-guided endonucleases and nickases

    Genome Research

    (2014)
  • L. Cong et al.

    Multiplex genome engineering using CRISPR/Cas systems

    Science

    (2013)
  • M. Costa et al.

    A method for genetic modification of human embryonic stem cells using electroporation

    Nature Protocols

    (2007)
  • R.P. Davis et al.

    A protocol for removal of antibiotic resistance cassettes from human embryonic stem cells genetically modified by homologous recombination or transgenesis

    Nature Protocols

    (2008)
  • R.C. DeKelver et al.

    Functional genomics, proteomics, and regulatory DNA analysis in isogenic settings using zinc finger nuclease-driven transgenesis into a safe harbor locus in the human genome

    Genome Research

    (2010)
  • Y. Fu et al.

    High-frequency off-target mutagenesis induced by CRISPR-Cas nucleases in human cells

    Nature Biotechnology

    (2013)
  • D. Hockemeyer et al.

    Genetic engineering of human pluripotent cells using TALE nucleases

    Nature Biotechnology

    (2011)
  • T. Horii et al.

    Generation of an ICF syndrome model by efficient genome editing of human induced pluripotent stem cells using the CRISPR system

    International Journal of Molecular Sciences

    (2013)
  • Cited by (56)

    • Modeling Monogenic Diabetes using Human ESCs Reveals Developmental and Metabolic Deficiencies Caused by Mutations in HNF1A

      2019, Cell Stem Cell
      Citation Excerpt :

      The use of human pluripotent stem cells and in vitro differentiation protocols that mimic in vivo pancreatic development are well suited to interrogate monogenic diseases of the pancreas. A number of relevant pancreatic phenotypes due to mutations in GATA4, GATA6, PDX1, and RFX6 have been described (Tiyaboonchai et al., 2017; Shi et al., 2017; Zhu et al., 2014; Zeng et al., 2016). This study uses genetically modified embryonic stem cells (ESCs) for human in vitro disease modeling to understand the role of HNF1A in pancreatic development and β-cell function.

    View all citing articles on Scopus
    1

    These authors contributed equally to this work.

    View full text