Improved FLP Recombinase, FLPe, Efficiently Removes Marker Gene from Transgene Locus Developed by Cre–lox Mediated Site-Specific Gene Integration in Rice

2011 ◽  
Vol 49 (1) ◽  
pp. 82-89 ◽  
Author(s):  
M. Aydın Akbudak ◽  
Vibha Srivastava
2012 ◽  
Vol 97 (9) ◽  
pp. 4039-4048 ◽  
Author(s):  
Tetsurou Muroi ◽  
Takaaki Kokuzawa ◽  
Yoshihiko Kihara ◽  
Ryuichi Kobayashi ◽  
Nobutaka Hirano ◽  
...  

2010 ◽  
Vol 45 (1) ◽  
pp. 15-23 ◽  
Author(s):  
M. Aydın Akbudak ◽  
Anjali B. More ◽  
Soumen Nandy ◽  
Vibha Srivastava

2020 ◽  
Vol 19 ◽  
pp. S28-S29
Author(s):  
Z.P. Zhou ◽  
Z.R. Chen ◽  
H. Cao ◽  
R. Duan ◽  
J. Li ◽  
...  

2004 ◽  
Vol 2 (2) ◽  
pp. 169-179 ◽  
Author(s):  
Vibha Srivastava ◽  
Magnolia Ariza-Nieto ◽  
Andrea J. Wilson

Cancers ◽  
2020 ◽  
Vol 12 (6) ◽  
pp. 1704
Author(s):  
Zelda Odé ◽  
Jose Condori ◽  
Nicolas Peterson ◽  
Sheng Zhou ◽  
Giedre Krenciute

T cells engineered with chimeric antigen receptors (CARs) show great promise in the treatment of some cancers. Modifying T cells to express CARs generally relies on T-cell transduction using viral vectors carrying a transgene, resulting in semi-random DNA integration within the T-cell genome. While this approach has proven successful and is used in generating the Food and Drug Administration (FDA, USA) approved B-lymphocyte antigen CD19-specific CAR T cells, it is possible the transgene could integrate into a locus that would lead to malignant transformation of the engineered T cells. In addition, manufacturing viral vectors is time-consuming and expensive. One way to overcome these challenges is site-specific gene integration, which can be achieved through clustered regularly interspaced short palindromic repeat (CRISPR) mediated editing and non-viral DNA, which serves as a template for homology-directed repair (HDR). This non-viral gene editing approach provides a rapid, highly specific, and inexpensive way to engineer T cells. Here, we describe an optimized protocol for the site-specific knock-in of a large transgene in primary human T cells using non-viral double stranded DNA as a repair template. As proof-of-principle, we targeted the T-cell receptor alpha constant (TRAC) locus for insertion of a large transgene containing green fluorescence protein (GFP) and interleukin-15 (IL-15). To optimize the knock-in conditions we tested template DNA concentration, homology arm length, cell number, and knock-in efficiency over time. We then applied these established guidelines to target the TRAC or interleukin-13 (IL-13) locus for the knock-in of synthetic molecules, such as a CAR, bispecific T-cell engager (BiTE), and other transgenes. While integration efficiency depends on the targeted gene locus and selected transgene, this optimized protocol reliably generates the desired insertion at rates upwards of 20%. Thus, it should serve as a good starting point for investigators who are interested in knocking in transgenes into specific loci.


2006 ◽  
Vol 4 (2) ◽  
pp. 209-218 ◽  
Author(s):  
Rekha Chawla ◽  
Magnolia Ariza-Nieto ◽  
Andrea J. Wilson ◽  
Sarah K. Moore ◽  
Vibha Srivastava

Sign in / Sign up

Export Citation Format

Share Document