Efficient gene knockout in primary human and murine myeloid cells by non-viral delivery of CRISPR-Cas9.

Authors:
Freund EC, Lock JY, Oh J, Maculins T, Delamarre L, Bohlen CJ, Haley B, Murthy A.
In:
Source: J Exp Med
Publication Date: (2020)
Issue: 217(7): e20191692
Research Area:
Cancer Research/Cell Biology
Immunotherapy / Hematology
Cells used in publication:
Dendritic cell (NHDC), human
Species: human
Tissue Origin: blood
Macrophage, human
Species: human
Tissue Origin: blood
Macrophage, mouse - C57BL/6
Species: mouse
Tissue Origin: bone marrow
Dendritic cell, mouse, immat. - C57BL/6
Species: mouse
Tissue Origin: blood
Platform:
4D-Nucleofector® 96-well Systems
4D-Nucleofector® X-Unit
Experiment

Murine BMDM Itgam/CD11b CRISPR-Cas9 optimization screen Day 5: BMDMs were resuspended in nucleofection solutions forprimary cells (Primary Cell Optimization 96-well Nucleofector Kit, Lonza, V4SP-9096) at a density of 5 × 105 cells per reaction in 20 µl nucleofection solution and mixed with Cas9-RNP containing B2M targeting gRNAs. This mixture was nucleofected using the Lonza 4D Nucleofector (4D-Nucleofector Core Unit: Lonza, AAF-1002B; 4D-Nucleofector X Unit: AAF-1002X). Immediately after nucleofection, ~180 µl of prewarmed BMDM culture medium was added to each well, and cells were harvested by gently washing the well. Each reaction was transferred to a single well in a 6-well TC-treated plate containing 2 ml of prewarmed BMDM culture medium.

Murine monocyte culture and eGFP CRISPR-Cas9 optimization screen: BM was harvested from tibias and femurs of eGFP-transgenic mice. Red blood cells were lysed with ACK lysis buffer. Monocytes were isolated using a negative selection kit (Miltenyi). Cells were washed once with 1× PBS and resuspended in the nucleofection solutions for primary cells (Primary Cell Optimization 96-well Nucleofector Kit, Lonza) at a density of 2 × 105 cells per well. Cells were nucleofected as above and immediately transferred to 6-well TC-treated plates containing prewarmed BMDM culture medium.

CRISPR-Cas9-mediated KO in murine BMDCs: BMcells were prepared, and red blood cells were lysed with ACK lysis buffer. BM cells were washed twice with 1× PBS and electroporated in the appropriate primary nucleofection solution (Primary Cell Optimization 4D-Nucleofector X Kit, P3 Primary Cell 4D-Nucleofector X Kit) using the Lonza 4DNucleofector (4DNucleofector Core Unit: Lonza, AAF-1002B; 4D-Nucleofector X Unit: AAF-1002X) as described above. Specifically, 2 × 106 BM cells per reaction were resuspended in 20 µl of primary nucleofection solution and mixed with Cas9-RNP containing targeting or NTC gRNAs.

Optimized conditons are then also transferred to the human monocytes and macropahages /DCs. please see table in reference. 

Abstract

Myeloid cells play critical and diverse roles in mammalian physiology, including tissue development and repair, innate defense against pathogens, and generation of adaptive immunity. As cells that show prolonged recruitment to sites of injury or pathology, myeloid cells represent therapeutic targets for a broad range of diseases. However, few approaches have been developed for gene editing of these cell types, likely owing to their sensitivity to foreign genetic material or virus-based manipulation. Here we describe optimized strategies for gene disruption in primary myeloid cells of human and murine origin. Using nucleofection-based delivery of Cas9-ribonuclear proteins (RNPs), we achieved near population-level genetic knockout of single and multiple targets in a range of cell types without selection or enrichment. Importantly, we show that cellular fitness and response to immunological stimuli is not significantly impacted by the gene editing process. This provides a significant advance in the study of myeloid cell biology, thus enabling pathway discovery and drug target validation across species in the field of innate immunity.