loading page

CRISPR-Cas13d for CHO Cell Engineering and Antibody Production
  • +6
  • Mei-Wei Lin,
  • Chih-Che Shen ,
  • Yen-Ju Lin,
  • Min-Yuan Chou,
  • Nam Ngoc Pham,
  • Yi-Hao Chang,
  • Chin-Wei Chang,
  • Jih Ru Hwu,
  • Yu-Chen Hu
Mei-Wei Lin
National Tsing Hua University

Corresponding Author:[email protected]

Author Profile
Chih-Che Shen
National Tsing Hua University
Author Profile
Yen-Ju Lin
Industrial Technology Research Institute
Author Profile
Min-Yuan Chou
Industrial Technology Research Institute
Author Profile
Nam Ngoc Pham
National Tsing Hua University
Author Profile
Yi-Hao Chang
National Tsing Hua University
Author Profile
Chin-Wei Chang
National Tsing Hua University
Author Profile
Jih Ru Hwu
National Tsing Hua University
Author Profile
Yu-Chen Hu
National Tsing Hua University
Author Profile

Abstract

Chinese hamster ovary (CHO) cell is the predominant cell factory to produce biopharmaceuticals such as immunoglobulin G (IgG), but in CHO cells lactate accumulation and cell death compromise IgG production while fucosylation mitigates the antibody-dependent cellular cytotoxicity (ADCC) of IgG. To tackle these problems, we harnessed a burgeoning gene silencing system, CRISPR-Cas13d, to effectively suppress the endogenous genes governing lactate production (LDHA), fucosylation (GFT) and cell death (DDIT3), hence repressing lactate accumulation and core fucosylation. We further exploited the Sleeping Beauty system to integrate the CRISPR-Cas13d module co-targeting these 3 genes and generated a CHO cell platform for IgG production. The new platform exhibited simultaneous knockdown of LDHA, GFT and DDIT3, accumulated less lactate, had prolonged longevity, produced more IgG with less fucosylation and stronger ADCC efficacy. These data collectively warrant the potentials of CRISPR-Cas13d for CHO cell engineering and improving antibody production with regard to quantity and quality.