Targeted Gene Editing in Human Primary T Cells Using CRISPR/Cas9 Ribonucleoproteins

Document Type : Original Article (s)

10.22122/jims.v39i1.13586

Abstract

Background: Clustered regularly interspaced short palindromic repeats/ CRISPR-associated protein 9 (CRISPR/Cas9)-mediated gene knockout of primary T cell has several limitations for clinical applications. Direct delivery of recombinant Cas9 protein and synthetic gRNA, as a pre-assembled ribonucleoprotein (RNP) complex, has become a potent approach to introduce highly efficient gene editing in primary T cells. In this study, we employed Cas9 RNP-based delivery system for targeted T Cell receptor alpha constant (TRAC) and β2 microglobulin (B2M) genes knockout in human primary T cells.
Methods: Specific gRNAs were designed to target the first exons of TRAC and B2M genes. Cas9 protein and respective synthetic gRNAs were then mixed separately, and electroporated into human primary T cells isolated from peripheral blood mononuclear cells (PBMCs). The gene editing efficiency was quantified using tracking of indels by decomposition (TIDE) analysis and flow cytometry.
Findings: Three days after electroporation of primary T cells with the TRAC and B2M targeting RNP complexes, TIDE analysis revealed the knockout efficiency of 13-60 percent for the TRAC-targeting gRNAs and 21-53 percent for B2M-targeting gRNAs. Flow cytometry analysis confirmed ~76% and ~27% complete loss of expression for the most efficient gRNAs targeting TRAC (TRAC-gRNA3) and B2M (B2M-gRNA2), respectively.
Conclusion: Our results demonstrate that Cas9 RNP system can be efficiently delivered into primary T cells and result in targeted gene knockout. The protocol described here enables a streamlined and highly efficient solution for maximizing editing efficiency in primary T cells, and simplifies the gene editing process for next-generation immunotherapies.

Keywords


Jinek M, Chylinski K, Fonfara I, Hauer M, Doudna JA, Charpentier E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 2012; 337(6096): 816-21.
Wang W, Ye C, Liu J, Zhang D, Kimata JT, Zhou P. CCR5 gene disruption via lentiviral vectors expressing Cas9 and single guided RNA renders cells resistant to HIV-1 infection. PLoS One 2014; 9(12): e115987.
Li C, Guan X, Du T, Jin W, Wu B, Liu Y, et al. Inhibition of HIV-1 infection of primary CD4+ T-cells by gene editing of CCR5 using adenovirus-delivered CRISPR/Cas9. J Gen Virol 2015; 96(8): 2381-93.
Mandal PK, Ferreira LM, Collins R, Meissner TB, Boutwell CL, Friesen M, et al. Efficient ablation of genes in human hematopoietic stem and effector cells using CRISPR/Cas9. Cell Stem Cell 2014; 15(5): 643-52.
Su S, Hu B, Shao J, Shen B, Du J, Du Y, et al. CRISPR-Cas9 mediated efficient PD-1 disruption on human primary T cells from cancer patients. Sci Rep 2016; 6: 20070.
Gomes-Silva D, Srinivasan M, Sharma S, Lee CM, Wagner DL, Davis TH, et al. CD7-edited T cells expressing a CD7-specific CAR for the therapy of T-cell malignancies. Blood 2017; 130(3): 285-96.
Ren J, Zhang X, Liu X, Fang C, Jiang S, June CH, et al. A versatile system for rapid multiplex genome-edited CAR T cell generation. Oncotarget 2017; 8(10): 17002-11.
Seki A, Rutz S. Optimized RNP transfection for highly efficient CRISPR/Cas9-mediated gene knockout in primary T cells. J Exp Med 2018; 215(3): 985-97.
Liang X, Potter J, Kumar S, Zou Y, Quintanilla R, Sridharan M, et al. Rapid and highly efficient mammalian cell engineering via Cas9 protein transfection. J Biotechnol 2015; 208: 44-53.
Morgan NV, Goddard S, Cardno TS, McDonald D, Rahman F, Barge D, et al. Mutation in the TCRalpha subunit constant gene (TRAC) leads to a human immunodeficiency disorder characterized by a lack of TCRalphabeta+ T cells. J Clin Invest 2011; 121(2): 695-702.
Bjorkman PJ, Saper MA, Samraoui B, Bennett WS, Strominger JL, Wiley DC. Structure of the human class I histocompatibility antigen, HLA-A2. Nature 1987; 329(6139): 506-12.
Concordet JP, Haeussler M. CRISPOR: Intuitive guide selection for CRISPR/Cas9 genome editing experiments and screens. Nucleic Acids Res 2018; 46(W1): W242-W245.
Doench JG, Fusi N, Sullender M, Hegde M, Vaimberg EW, Donovan KF, et al. Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9. Nat Biotechnol 2016; 34(2): 184-91.
Hsu PD, Scott DA, Weinstein JA, Ran FA, Konermann S, Agarwala V, et al. DNA targeting specificity of RNA-guided Cas9 nucleases. Nat Biotechnol 2013; 31(9): 827-32.
Boyum A. Isolation of mononuclear cells and granulocytes from human blood. Isolation of monuclear cells by one centrifugation, and of granulocytes by combining centrifugation and sedimentation at 1 g. Scand J Clin Lab Invest Suppl 1968; 97: 77-89.
Trickett A, Kwan YL. T cell stimulation and expansion using anti-CD3/CD28 beads. J Immunol Methods 2003; 275(1-2): 251-5.
Brinkman EK, Chen T, Amendola M, van Steensel B. Easy quantitative assessment of genome editing by sequence trace decomposition. Nucleic Acids Res 2014; 42(22): e168.
Geisler C, Kuhlmann J, Rubin B. Assembly, intracellular processing, and expression at the cell surface of the human alpha beta T cell receptor/CD3 complex. Function of the CD3-zeta chain. J Immunol 1989; 143(12): 4069-77.
Serreze DV, Leiter EH, Christianson GJ, Greiner D, Roopenian DC. Major histocompatibility complex class I-deficient NOD-B2mnull mice are diabetes and insulitis resistant. Diabetes 1994; 43(3): 505-9.
Liu X, Zhang Y, Cheng C, Cheng AW, Zhang X, Li N, et al. CRISPR-Cas9-mediated multiplex gene editing in CAR-T cells. Cell Res 2017; 27(1): 154-7.
Torikai H, Reik A, Liu PQ, Zhou Y, Zhang L, Maiti S, et al. A foundation for universal T-cell based immunotherapy: T cells engineered to express a CD19-specific chimeric-antigen-receptor and eliminate expression of endogenous TCR. Blood 2012; 119(24): 5697-705.
Berdien B, Mock U, Atanackovic D, Fehse B. TALEN-mediated editing of endogenous T-cell receptors facilitates efficient reprogramming of T lymphocytes by lentiviral gene transfer. Gene Ther 2014; 21(6): 539-48.
Poirot L, Philip B, Schiffer-Mannioui C, Le CD, Chion-Sotinel I, Derniame S, et al. Multiplex genome-edited T-cell manufacturing platform for "off-the-shelf" adoptive T-cell immunotherapies. Cancer Res 2015; 75(18): 3853-64.
Valton J, Guyot V, Marechal A, Filhol JM, Juillerat A, Duclert A, et al. A Multidrug-resistant engineered CAR T cell for allogeneic combination immunotherapy. Mol Ther 2015; 23(9): 1507-18.
Ren J, Liu X, Fang C, Jiang S, June CH, Zhao Y. Multiplex genome editing to generate universal CAR T cells resistant to PD1 inhibition. Clin Cancer Res 2017; 23(9): 2255-66.
Chu VT, Weber T, Graf R, Sommermann T, Petsch K, Sack U, et al. Efficient generation of Rosa26 knock-in mice using CRISPR/Cas9 in C57BL/6 zygotes. BMC Biotechnol 2016; 16: 4.
Platt RJ, Chen S, Zhou Y, Yim MJ, Swiech L, Kempton HR, et al. CRISPR-Cas9 knockin mice for genome editing and cancer modeling. Cell 2014; 159(2): 440-55.
Hendel A, Bak RO, Clark JT, Kennedy AB, Ryan DE, Roy S, et al. Chemically modified guide RNAs enhance CRISPR-Cas genome editing in human primary cells. Nat Biotechnol 2015; 33(9): 985-9.
Rupp LJ, Schumann K, Roybal KT, Gate RE, Ye CJ, Lim WA, et al. CRISPR/Cas9-mediated PD-1 disruption enhances anti-tumor efficacy of human chimeric antigen receptor T cells. Sci Rep 2017; 7(1): 737.
Schumann K, Lin S, Boyer E, Simeonov DR, Subramaniam M, Gate RE, et al. Generation of knock-in primary human T cells using Cas9 ribonucleoproteins. Proc Natl Acad Sci USA 2015; 112(33): 10437-42.