A subscription to JoVE is required to view this content. Sign in or start your free trial.

In This Article

  • Summary
  • Abstract
  • Introduction
  • Protocol
  • Results
  • Discussion
  • Disclosures
  • Acknowledgements
  • Materials
  • References
  • Reprints and Permissions

Summary

The presented method describes the generation of a CRISPR-mediated gene knockout in the human embryonic stem cell (hESC) line H9, which stably expresses sgRNAs targeting the L2HGDH gene using a highly efficient lentiviral-mediated gene delivery system.

Abstract

The CRISPR-Cas9 system for genome editing has revolutionized gene function studies in mammalian cells, including stem cells. However, the practical application of this technique, particularly in pluripotent stem cells, presents certain challenges, such as being time- and labor-intensive and having low editing efficiency. Here, we describe the generation of a CRISPR-mediated gene knockout in a human embryonic stem cell (hESC) line stably expressing sgRNAs for the L2HGDH gene, using a highly efficient and stable lentiviral-mediated gene delivery system. The sgRNAs targeting exon 1 of the L2HGDH gene were chemically synthesized and cloned into the lentiCRISPR v2-puro vector, which combines the constitutive expression of sgRNAs with Cas9 in a highly efficient single-vector system to achieve higher lentiviral titers for hESC infection and stable selection using puromycin. Puromycin-selected cells were further expanded, and single-cell clones were obtained using the limited dilution method. The single clones were expanded, and several homozygous knockout clones for the L2HGDH gene were obtained, as confirmed by a 100% reduction in L2HGDH expression using Western blot analysis. Furthermore, using MSBSP-PCR, the CRISPR mutation site was mapped upstream of the PAM recognition sequence of Cas9 in the selected homozygous clones. Sanger sequencing was performed to analyze the exact insertions/deletions, and functional characterization of the clones was conducted. This method produced a significantly higher percentage of homozygous deletions compared to previously reported non-viral gene delivery methods. Although this report focuses on the L2HGDH gene, this robust and cost-effective approach can be used to create homozygous knockouts for other genes in pluripotent stem cells for gene function studies.

Introduction

Human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) are stem cells with the potential to differentiate into all cell types in the body. These cells serve as valuable tools for studying human development, as well as for understanding the underlying mechanisms of various diseases, thus offering tremendous promise for regenerative medicine, disease modeling, and drug discovery. Such studies involve investigating how specific genes contribute to the development, functioning, and regulation of organisms1,2.

Various techniques and approaches are employed to decipher gene function, including genetic manipulation, such as gene knockout or overexpression, and genome editing. Among these, CRISPR-Cas9 technology has emerged as the most efficient approach for gene knockout and gene editing studies1,2,3. The CRISPR-Cas9 system works by utilizing a single guide RNA (sgRNA) molecule specifically designed to identify and bind to a particular DNA sequence of interest. Acting as a molecular guide, the sgRNA directs the Cas9 enzyme to the precise location in the genome that requires modification. Once bound, Cas9 initiates a double-stranded break in the DNA at the designated site. Following the cleavage of DNA, the cell's inherent repair mechanisms are activated. These include two main repair pathways: non-homologous end joining (NHEJ) and homology-directed repair (HDR). NHEJ often results in insertions or deletions (indels) at the break site, leading to gene disruption or inactivation. Conversely, HDR enables the insertion of new DNA sequences at the break site, facilitating the introduction of targeted genetic alterations4.

Given the importance of gene deletions in pluripotent stem cells, several protocols have been published on CRISPR-Cas9-mediated gene knockouts in hESCs/iPSCs. However, many of these protocols face significant limitations, such as being extremely time-consuming, labor-intensive, and having low efficiency due to the use of non-viral gene delivery methods5. These challenges are even more pronounced in hESCs/iPSCs, as these cells are known to have lower editing efficiency compared to other cell types5. Some of these limitations can be addressed by increasing the efficiency of plasmid delivery containing Cas9 and sgRNAs. This can be successfully achieved using a lentiviral vector system, which can significantly improve gene editing outcomes. Lentivirus packaging protocols are well-established and straightforward, allowing easy adoption in laboratories, even by researchers with limited experience. Lentiviruses exhibit high infection efficiency across various cell types, including hESCs and iPSCs. Therefore, utilizing a lentiviral system for Cas9-sgRNA expression is ideal for routine gene editing experiments in hESCs/iPSCs for gene function studies.

Here, we provide a simple and straightforward method for highly efficient CRISPR-Cas9-based gene deletions in hESCs in a comparatively shorter time duration than conventional protocols (Figure 1). Although a lentiviral vector with constitutive expression of Cas9 and sgRNA has been used, it could easily be replaced with drug-inducible Cas9 expression for controllable Cas9 expression.

Protocol

The details of the gene sequences, reagents, and equipment used in this study are listed in the Table of Materials.

1. Single guide RNA (sgRNA) design, cloning, and lentiviral vector production

NOTE: Two different sgRNA sequences targeting the exon 1 of the L2HGDH gene, both adapted from Qiu et al.6with PAM sites of AGG and TGG for sequences 1 and 2, respectively, are used. Both sgRNAs were 20 bp in length, and ends were modified to add linker sequences for restriction enzyme Bsmb1 to be cloned in the target vector (LentiCRISPRv2) as described in the previous report6. Linker sequences were added during the designing of sgRNAs for cloning purposes.

  1. Anneal chemically synthesized sgRNAs6 and clone into Bsmb1 single cut lentiviral vector, LentiCRISPRv2 using optimized protocol as previously reported7.
  2. Proceed with lentiviral packaging and concentration using standardized and optimized protocol7.
  3. For lentivirus production, seed HEK293T cells (1 Γ— 105 cells/cm2) using DMEM, 10% FBS and 1x penicillin/streptomycin (P/S) and incubate overnight in tissue culture incubator at 37 Β°C in an atmosphere of 5% CO2 under humid conditions.
  4. When 90% confluent, co-transfect HEK293T cells with entry vector (empty backbone or sgRNA expressing plasmids) and packaging plasmids using low-cost cationic polymer PEI as described previously7.
  5. Collect the conditioned media containing Lentiviral supernatant (LVS) particles at 48 h and 72 h post-transfection and proceed with ultracentrifugation using sucrose cushion and spin the tubes at 1,25,000 x g for 2 h at 4 Β°C.
  6. Proceed with the LVS particle concentration to at least 200 times the original volume in PBS, aliquot, and store at -80Β Β°C until use.
  7. Determine the titer of lentiviral particles by using the qPCR Lentivirus Titer Kit following the manufacturer's instructions (see Table of Materials).

2. Lentiviral infections and single-cell clonal propagation

  1. For infections with LVS particles, seed hESC (H9) cell suspension having 1 Γ— 105 cells/0.5 mL of hESC culture media (Basal media + P/S + 10 Β΅M of Rock Inhibitor) on complete Matrigel (1:50) coated P24 well plates in 500 Β΅L of media and incubate overnight to allow the cells to attach. Seed extra wells that would not be infected with LVS but treated with puromycin to serve as non-infected control.
    NOTE: Cell counting can be performed using a manual hemocytometer or automated cell counter.
  2. The following day, infect the cells at Multiplicity of Infection (MOI) of 10 along with 8 Β΅g/mL of polybrene and incubate at 37Β Β°C for 8 h followed by media replacement with fresh hESC media + P/S without Rock Inhibitor and continue culturing until cells are 90% confluent.
  3. Start puromycin selection by supplementing the media with 0.8 Β΅g/mL concentration of puromycin when cells reach 90% confluency, which is usually 48-72 h post-infection, and continue the selection until all cells die in the non-infected cells (control group).
  4. After the selection is complete (usually 4-6 days), split stable cells (1:4) and expand for cryopreservation and further analysis.
  5. Perform single-cell selection and clonal expansion using cells expressing L2HGDH-sgRNA-16.
  6. For this purpose, prepare a cell suspension equivalent to 500 cells/10 mL of complete growth media and seed 100 Β΅L of this suspension in each well of a 96-well plate.
  7. Leave the cells undisturbed for 3 days and then observe.
  8. Mark the wells that yield single clones and change the media every other day until a sufficient size is achieved for the colonies (usually 2 weeks) to expand further, cryopreserve, and analyze.

3. gDNA extraction, MS-BSP PCR, and Sanger sequencing

  1. Isolate gDNA from cells using the genomic DNA isolation Kit following the manufacturer's instructions (see Table of Materials).
  2. Proceed with the mapping of mutation sites upstream of the PAM recognition sequences using Mutation Sites Based Specific Primers (MS-BSP) analysis8.
  3. For this, an unbiased right primer L2H-UMSBSP-R1 is designed to amplify the region outside the Exon1 to amplify any targets.
  4. Design a biased left primer L2H-BMSBSP-F1 with an identical sequence to sgRNA to amplify the target sequence close to PAM recognition sequences.
    NOTE: At very high stringent conditions of PCR, the product will be observed on the gel in unmutated clones. No product would be observed in CRISPR-knockout clones exhibiting mutations close to upstream of PAM recognition sequences.
  5. In order to map single base pair mutations, PCR amplify a 468 bp sequence spanning the whole exon 1 of L2HGDH and subject to Sanger sequencing followed by multiple sequence alignment analysis using clustalw8.

4. hESC-differentiation and embryoid body (EB) formation assay

  1. Proceed with the directed differentiation of control and different CRISPR clones of hESCs (H9) towards neuro-ectoderm fate following established protocols as previously described using dual smad inhibition method9,10,11.
  2. When at 90% confluency, treat the cells with LDN193189 (200 nM) and SB431542 (10 Β΅M) for an initial 24 h in 100% KSR media followed by the addition of XAV939 (2 Β΅M) for additional 2 days.
  3. After 3 days, reduce the percentage of KSR media (Knockout DMEM supplemented with 15% (v/v) KSR, 1% (v/v) L-glutamine, 1% (v/v) P/S, 1% (v/v) 10 mM MEM, and 0.1% (v/v) 2-mercaptoethanol (75%, 50%, 25%) by combining with N2 media (DMEM/F12 supplemented with 1x N2 supplement, 1x P/S,) to 100% N2 media over a period of 8 days.
  4. At the beginning of Day 12, fix the cells for immunostaining using PAX6 as a neuro-ectoderm marker.
  5. For mesoderm and endodermal fate determination studies, employ a small molecule CHIR99021-based approach described in previous publications12,13.
  6. For this, when the cells reach 70% confluency, treat with 3 Β΅M of CHIR99021 in Definitive Endoderm (DE) media for 24 h followed by fixation for immunostaining using Brachuary as mesoderm-specific marker.
  7. For the endodermal stage, culture the cells for an additional 24 h in DE media alone without the addition of CHIR99021 before fixing for immunostaining using FOXA2.
  8. For EB formation assay, seed the cells on low attachment cell surfaces without using Matrigel to culture cells under suspension conditions for 24 h before taking micrographs.

5. Western blot analysis

  1. Wash the cells twice using PBS and lyse using 1x RIPA buffer containing 1% SDS and 1x protease and phosphatase inhibitor cocktail.
  2. Clear the lysates by centrifugation at 16,000 x g for 10 min at 4 Β°C followed by collection of the supernatants.
  3. Quantify the total cell protein using the BCA protein assay kit following the manufacturer's instructions. Adjust the samples to 2 Β΅g/Β΅L using 4Γ— loading dye sample buffer.
  4. Denature the protein samples at 70 Β°C for 10 min, load equal amounts of each sample, and resolve using 4%-12% gradient SDS-PAGE gels7 followed by transfer to PVDF membrane at a constant voltage of 100 V for 1 h at 4 Β°C.
  5. Block the membranes using 5% non-fat milk and incubate in primary antibody dilutions at 4 Β°C overnight with rotation.
  6. Next, wash the membranes 5x using PBST buffer and incubate in HRP-conjugated appropriate secondary antibodies for 1 h at room temperature.
  7. Wash the membranes again with PBST 5x, incubate with chemiluminescent substrate, and develop using X-ray films.

6. Immunostaining

  1. Seed the cells on P4 well plates and incubate for at least 24 h before fixation to allow proper attachment of cells to surfaces.
  2. Wash the cells 3x with PBS to remove any dead cells as well as media components, followed by fixation using 4% PFA for 15 min at room temperature.
  3. Permeabilize the cells by using 0.3% triton X-100 followed by blocking for nonspecific binding by using 2% BSA in PBS for 1 h at room temperature.
  4. Incubate the samples with primary antibodies (OCT4, NANOG, SOX2, KI67, PAX6, Brachuary, FOXA2) diluted in 1% BSA overnight at 4 Β°C.
  5. Wash the cells 3x with PBS and incubate with appropriate secondary antibodies (Goat anti-mouse 488, Goat anti-rabbit 488, Goat anti-mouse 546, Goat anti-rabbit 546) diluted in 1% BSA for 1 h at room temperature.
  6. Finally, wash the samples with PBS 3x and counterstain them with DAPI and image using a fluorescence microscope.

Results

Cloning of L2HGDH sgRNAs in lentiCRISPRv2 puro
lentiCRISPRv2 puro vector was commercially obtained (see Table of Materials) and digested with BsmB1, which resulted in the release of a 1.8 Kb stuffer fragment. As shown in Figure 2A, a complete digestion of the vector was observed. For each construct, six clones were screened for the presence or absence of insert using reverse sgRNA sequence as a primer and a forward primer (U6-459F) from within the vect...

Discussion

This study has standardized a method that enables highly efficient and cost-effective gene deletions in hESCs through CRISPR-Cas9 technology. This method successfully achieved homozygous deletion of the L2HGDH gene in hESCs within 3-4 weeks, starting from hESC infection to single-cell clonal selection and propagation (Table 1). Although CRISPR-Cas9-mediated gene manipulations can be achieved by transient transfections in most cells, this becomes challenging in stem cells due to poor transfection efficien...

Disclosures

The authors declare that there is no conflict of interest.

Acknowledgements

This work was supported by research grants from United Arab Emirates University (UAEU) - grant #12M105, grant #12R167 (Zayed Center for Health Sciences), 21R105 (Zayed Bin Sultan Charitable and Humanitarian Foundation (ZCHF)), and ASPIRE, the technology program management pillar of Abu Dhabi's Advanced Technology Research Council (ATRC), via the ASPIRE Precision Medicine Research Institute Abu Dhabi (ASPIREPMRIAD) award grant number VRI-20-10.

Materials

NameCompanyCatalog NumberComments
2-MERCAPTOETHANOLΒ Invitrogen31350010
38.5 mL, Sterile + Certified Free Open-Top
Thinwall Ultra-Clear Tubes
Beckman CoulterC14292
AccutaseStem cell technologies7920
bFGF Recombinant humanInvitrogenPHG0261
Brachyury Rabbit mAbAbclonalA5078
BsmBI-v2NEBR0739S
chir99021Tocris4423/10
Corning Matrigel Basement Membrane Matrix, LDEV-freeCorning354234
CyclopamineStem cell technologies72074
DMEM mediaInvitrogen11995073
DMEM NUTRIENT MIX F12Β Invitrogen11320033
DPBS w/o: Ca and MgPAN BiotechP04-36500
Fetal bovie serumInvitrogen10270106
FoxA2/HNF3Ξ²Β CST8186
GAPDH (14C10) Rabbit mAb AntibodyCST2118S
Gentle Cell Dissociation ReagentStem cell technologies7174
HyClone Non Essential Amino Acids (NEAA) 100x SolutionGE healthcareSH30238.01
Ki-67 (D3B5) Rabbit mAbCST9129
KnockOut Serum ReplacementInvitrogen10828028
L GLUTAMINE, 100xInvitrogen2924190090
L2H-BMSBSP-F1MacrogenCGTGCGGGTTCGCGTCTGGG
L2HGDH Polyclonal antibodyProteintech15707-1-AP
L2HGDH-SgRNA1-FMacrogenCACCGCGTGCGG
GTTCGCGTCTGGG
L2HGDH-SgRNA1-RMacrogenAAACCCCAGACGC
GAACCCGCACGC
L2HGDH-SgRNA2-FMacrogenCACCGCCCGCGG
GCTTTTCGCCGG
L2HGDH-SgRNA2-RMacrogenAAACCCGGCGAA
AAGCCCGCGGGC
L2H-SeqF1MacrogenGCTAAAGAGCGC
GGGTCCTCGG
L2H-SeqR1MacrogenGTGGACGGGTTG
TTCAAAGCCAGAG
L2H-UMSBSP-R1MacrogenGTGGACGGGTTG
TTCAAAGCCAGAG
LentiCRISPRv2Addgene52961
mTesR1 complete mediaStem cell technologies85850
Nanog AntibodyCST3580
NEUROBASAL MEDIUM 1x CTSInvitrogenA1371201
Neuropan 2 Supplement 100xPAN BiotechP07-11050
Neuropan 27 Supplement 50xPAN BiotechP07-07200
Oct-4 AntibodyCST2750
Pax6 (D3A9V) XP Rabbit mAbCST60433
PENICILLIN STREPTOMYCIN SOLInvitrogen15140122
pMD2.GAddgene12259
Polybrene infection reagentSigmaTR1003- G
Polyethylenimine, branchedSigma408727
psPAX2.0Addgene12260
PuromycinInvitrogenA1113802
qPCR Lentivirus Titer KitAbmLV900
Rock inhibitor Y-27632
dihydrochlorideΒ 
Tocris1254
SB 431542Tocris1614/10
Sox2 AntibodyCST2748
SucroseSigma57-50-1
TRYPSIN .05% EDTAΒ Invitrogen25300062
U6-459FMacrogenGAGGGCCTATT
TCCCATGATTC
Wizard Genomic DNA Purification KitΒ PromegaA1120
XAV 939Tocris3748/10

References

  1. Jinek, M., et al. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science. 337 (6096), 816-821 (2012).
  2. Jiang, F., Doudna, J. A. CRISPR-Cas9 structures and mechanisms. Annu Rev Biophys. 46 (1), 505-529 (2017).
  3. Mali, P., et al. RNA-guided human genome engineering via Cas9. Science. 339 (6121), 823-826 (2013).
  4. Xue, C., Greene, E. C. DNA repair pathway choices in CRISPR-Cas9-mediated genome editing. Trends Genet. 37 (7), 639-656 (2021).
  5. Zhang, Z., et al. CRISPR/Cas9 genome-editing system in human stem cells: Current status and future prospects. Mol Ther Nucleic Acids. 9, 230-241 (2017).
  6. Qiu, Z., et al. MYC regulation of D2HGDH and L2HGDH influences the epigenome and epitranscriptome. Cell Chem Biol. 27 (5), 538-550.e537 (2020).
  7. Sheikh, M. A., Ansari, S. A. Lentiviral mediated delivery of shRNAs to hESCs and NPCs using low-cost cationic polymer polyethylenimine (PEI). J Vis Exp. (183), e63953 (2022).
  8. Guo, J., et al. A simple and cost-effective method for screening of CRISPR/Cas9-induced homozygous/biallelic mutants. Plant Methods. 14 (1), 40 (2018).
  9. Ardah, M. T., Parween, S., Varghese, D. S., Emerald, B. S., Ansari, S. A. Saturated fatty acid alters embryonic cortical neurogenesis through modulation of gene expression in neural stem cells. J Nutr Biochem. 62, 230-246 (2018).
  10. Parween, S., et al. Higher O-GlcNAc levels are associated with defects in progenitor proliferation and premature neuronal differentiation. Front Cell Neurosci. 11, 415 (2017).
  11. Parween, S., et al. Nutrient-sensitive protein O-GlcNAcylation modulates the transcriptome through epigenetic mechanisms during embryonic neurogenesis. Life Sci Alliance. 5 (8), e202201385 (2022).
  12. Varghese, D. S., et al. Developmental modeling of hepatogenesis using obese iPSCs-hepatocyte differentiation uncovers pathological features. Cell Death Dis. 13 (7), 670 (2022).
  13. Varghese, D. S., Alawathugoda, T. T., Ansari, S. A. Fine-tuning of hepatocyte differentiation from human embryonic stem cells: Growth factor. Stem Cells Int. 2019 (5), 5968236 (2019).
  14. Jacobs, E. Z., et al. CRISPR/Cas9-mediated genome editing in naΓ―ve human embryonic stem cells. Sci Rep. 7 (1), 16650 (2017).
  15. Cuevas-OcaΓ±a, S., et al. A cell-based optimized approach for rapid and efficient gene editing of human pluripotent stem cells. Int J Mol Sci. 24 (3), 12345 (2023).
  16. Veres, A., et al. Low incidence of off-target mutations in individual CRISPR-Cas9 and TALEN-targeted human stem cell clones detected by whole-genome sequencing. Cell Stem Cell. 15 (1), 27-30 (2014).
  17. Smith, C., Gore, A., Yan, W., Abalde-Atristain, L., Li, Z., He, C. Whole-genome sequencing analysis reveals high specificity of CRISPR/Cas9 and TALEN-based genome editing in human iPSCs. Cell Stem Cell. 15 (1), 12-13 (2014).
  18. Zhang, X. H., Tee, L. Y., Wang, X. G., Huang, Q. S., Yang, S. H. Off-target effects in CRISPR/Cas9-mediated genome engineering. Mol Ther Nucleic Acids. 4 (7), e264 (2015).

Reprints and Permissions

Request permission to reuse the text or figures of this JoVE article

Request Permission

Explore More Articles

CRISPR Cas9Gene DeletionGene KnockoutHuman Pluripotent Stem CellsHESCL2HGDH GeneLentiviral mediated DeliverySgRNAsPuromycin SelectionHomozygous ClonesWestern Blot AnalysisMSBSP PCRSanger SequencingGene Function Studies

This article has been published

Video Coming Soon

JoVE Logo

Privacy

Terms of Use

Policies

Research

Education

ABOUT JoVE

Copyright Β© 2025 MyJoVE Corporation. All rights reserved