1. Academic Validation
  2. Engineering Extracellular Vesicles Enriched with Palmitoylated ACE2 as COVID-19 Therapy

Engineering Extracellular Vesicles Enriched with Palmitoylated ACE2 as COVID-19 Therapy

  • Adv Mater. 2021 Dec;33(49):e2103471. doi: 10.1002/adma.202103471.
Feng Xie 1 2 Peng Su 3 Ting Pan 4 Xiaoxue Zhou 3 Heyu Li 3 Huizhe Huang 5 Aijun Wang 6 Fangwei Wang 3 Jun Huang 3 Haiyan Yan 1 Linghui Zeng 1 Long Zhang 3 Fangfang Zhou 2
Affiliations

Affiliations

  • 1 School of Medicine, Zhejiang University City College, Hangzhou, 310015, China.
  • 2 Institutes of Biology and Medical Science, Soochow University, Suzhou, 215123, China.
  • 3 MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, 310058, China.
  • 4 Center for Infection and Immunity Studies, School of Medicine, Sun Yat-sen University, Shenzhen, 518107, China.
  • 5 Faculty of Basic Medical Sciences, Chongqing Medical University, Medical College Road 1, Chongqing, 400016, China.
  • 6 Department of Surgery, School of Medicine, UC Davis, Davis, CA, 95817, USA.
Abstract

Angiotensin converting Enzyme 2 (ACE2) is a key receptor present on cell surfaces that directly interacts with the viral spike (S) protein of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). It is proposed that inhibiting this interaction can be promising in treating COVID-19. Here, the presence of ACE2 in extracellular vesicles (EVs) is reported and the EV-ACE2 levels are determined by protein palmitoylation. The Cys141 and Cys498 residues on ACE2 are S-palmitoylated by zinc finger DHHC-Type Palmitoyltransferase 3 (ZDHHC3) and de-palmitoylated by acyl protein thioesterase 1 (LYPLA1), which is critical for the membrane-targeting of ACE2 and their EV secretion. Importantly, by fusing the S-palmitoylation-dependent plasma membrane (PM) targeting sequence with ACE2, EVs enriched with ACE2 on their surface (referred to as PM-ACE2-EVs) are engineered. It is shown that PM-ACE2-EVs can bind to the SARS-CoV-2 S-RBD with high affinity and block its interaction with cell surface ACE2 in vitro. PM-ACE2-EVs show neutralization potency against pseudotyped and authentic SARS-CoV-2 in human ACE2 (hACE2) transgenic mice, efficiently block viral load of authentic SARS-CoV-2, and thus protect host against SARS-CoV-2-induced lung inflammation. The study provides an efficient engineering protocol for constructing a promising, novel biomaterial for application in prophylactic and therapeutic treatments against COVID-19.

Keywords

ACE2; SARS-CoV-2; extracellular vesicles; palmitoylation.

Figures
Products
Inhibitors & Agonists
Other Products