Single-Step, High-Efficiency CRISPR-Cas9 Genome Editing in Primary Human Disease-Derived Fibroblasts.

Authors:
Martufi M, Good RB, Rapiteanu R, Schmidt T, Patili E, Tvermosegaard K, New M, Nanthakumar CB, Betts J, Blanchard AD, Maratou K.
In:
Source: CRISPR J
Publication Date: (2019)
Issue: 2: 31-40
Research Area:
Gene Expression
Respiratory Research
Cells used in publication:
Fibroblast, lung, human normal (NHLF)
Species: human
Tissue Origin: lung
CD4+, human
Species: human
Tissue Origin: blood
Platform:
Nucleofector® I/II/2b
4D-Nucleofector® X-Unit
Experiment

250,000 cells (IPF) in 20 µL were used per electroporation. Cells were washed once in phosphate-buffered saline (PBS), and pelleted by centrifugation at 90g, 10min, at room temperature. After PBS removal, cells were re-suspended in 15.5 µL of P3 solution from a P3 Primary Cell 4D-Nucleofector X Kit (Lonza) and mixed together with 4.5 µL of RNP complex to reach a total volume of 20 µL. Electroporation was performed in 16-well Nucleocuvette strips using
program CM-138. For a double KO generation using the 4D Nucleofector system, RNPs targeting SMAD2 exon 6 and SMAD3 exon 6 were complexed in vitro as  described above. A total of 250,000 cells were washed once in PBS, and pelleted by centrifugation at 90g, 10 min, at room temperature. After PBS removal, cells were re-suspended in 11µL of P3 solution and mixed together with 9 µL of RNPs to reach a final volume of 20 µL. Electroporation was performed in 16-well Nucleocuvette Strips using program CM-138.

Expanded CD4+ T cells (n = 1,000,000) derived from peripheral blood were re-suspended in 20 µL of P3 primary Cell4D-Nucleofector X Kit Buffer (Lonza) and were then
mixed with 15 lL of RNP complex for a total of 35 µL. Electroporation was carried out using a 4D Nucleofector system in 16-well Nucleocuvette Strips with the EH-115 program.

Abstract

Genome editing is a tool that has many applications, including the validation of potential drug targets. However, performing genome editing in low-passage primary human cells with the greatest physiological relevance is notoriously difficult. High editing efficiency is desired because it enables gene knockouts (KO) to be generated in bulk cellular populations and circumvents the problem of having to generate clonal cell isolates. Here, we describe a single-step workflow enabling >90% KO generation in primary human lung fibroblasts via CRISPR ribonucleoprotein delivery in the absence of antibiotic selection or clonal expansion. As proof of concept, we edited two SMAD family members and demonstrated that in response to transforming growth factor beta, SMAD3, but not SMAD2, is critical for deposition of type I collagen in the fibrotic response. The optimization of this workflow can be readily transferred to other primary cell types.