Your browser doesn't support javascript.
loading
Modeling COVID-19 with Human Pluripotent Stem Cell-Derived Cells Reveals Synergistic Effects of Anti-inflammatory Macrophages with ACE2 Inhibition Against SARS-CoV-2.
Duan, Fuyu; Guo, Liyan; Yang, Liuliu; Han, Yuling; Thakur, Abhimanyu; Nilsson-Payant, Benjamin E; Wang, Pengfei; Zhang, Zhao; Yan Ma, Chui; Zhou, Xiaoya; Han, Teng; Zhang, Tuo; Wang, Xing; Xu, Dong; Duan, Xiaohua; Xiang, Jenny; Tse, Hung-Fat; Liao, Can; Luo, Weiren; Huang, Fang-Ping; Chen, Ya-Wen; Evans, Todd; Schwartz, Robert E; tenOever, Benjamin; Ho, David D; Chen, Shuibing; Na, Jie; Lian, Qizhou; Chen, Huanhuan Joyce.
Affiliation
  • Duan F; School of Medicine, Tsinghua University.
  • Guo L; Prenatal Diagnostic Centre and Cord Blood Bank, Guangzhou Women and Children's Medical Center, Guangzhou Medical University.
  • Yang L; Department of Surgery, Weill Cornell Medicine.
  • Han Y; Department of Surgery, Weill Cornell Medicine.
  • Thakur A; The Pritzker School of Molecular Engineering, the University of Chicago.
  • Nilsson-Payant BE; Department of Microbiology, Icahn School of Medicine at Mount Sinai.
  • Wang P; Aaron Diamond AIDS Research Center, Columbia University Irving Medical Center.
  • Zhang Z; Department of Medicine, Li Ka Shing Faculty of Medicine; the University of Hong Kong.
  • Yan Ma C; Department of Medicine, Li Ka Shing Faculty of Medicine; the University of Hong Kong.
  • Zhou X; Prenatal Diagnostic Centre and Cord Blood Bank, Guangzhou Women and Children's Medical Cent.
  • Han T; Sandra and Edward Meyer Cancer Center, Department of Medicine, Weill Cornell Medicine.
  • Zhang T; Genomic Resource Core Facility, Weill Cornell Medicine.
  • Wang X; Genomic Resource Core Facility, Weill Cornell Medicine.
  • Xu D; Genomic Resource Core Facility, Weill Cornell Medicine.
  • Duan X; Department of Surgery, Weill Cornell Medicine.
  • Xiang J; Genomic Resource Core Facility, Weill Cornell Medicine.
  • Tse HF; Department of Medicine, Li Ka Shing Faculty of Medicine; the University of Hong Kong.
  • Liao C; Prenatal Diagnostic Centre and Cord Blood Bank, Guangzhou Women and Children's Medical Center, Guangzhou Medical University.
  • Luo W; Department of Pathology, The Second Affiliated Hospital of Southern University of Science and Technology.
  • Huang FP; Institute for Advanced Study (IAS), Shenzhen University.
  • Chen YW; Department of Medicine, Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine.
  • Evans T; Department of Surgery, Weill Cornell Medicine.
  • Schwartz RE; Division of Gastroenterology and Hepatology, Department of Medicine, Weill Cornell Medicine.
  • tenOever B; Department of Microbiology, Icahn School of Medicine at Mount Sinai.
  • Ho DD; Department of Microbiology, Icahn School of Medicine at Mount Sinai.
  • Chen S; Department of Surgery, Weill Cornell Medicine.
  • Na J; School of Medicine, Tsinghua University.
  • Lian Q; Department of Medicine, Li Ka Shing Faculty of Medicine; the University of Hong Kong.
  • Chen HJ; The Pritzker School of Molecular Engineering, the University of Chicago.
Res Sq ; 2020 Sep 15.
Article in En | MEDLINE | ID: mdl-32839764
ABSTRACT
Dysfunctional immune responses contribute critically to the progression of Coronavirus Disease-2019 (COVID-19) from mild to severe stages including fatality, with pro-inflammatory macrophages as one of the main mediators of lung hyper-inflammation. Therefore, there is an urgent need to better understand the interactions among SARS-CoV-2 permissive cells, macrophage, and the SARS-CoV-2 virus, thereby offering important insights into new therapeutic strategies. Here, we used directed differentiation of human pluripotent stem cells (hPSCs) to establish a lung and macrophage co-culture system and model the host-pathogen interaction and immune response caused by SARS-CoV-2 infection. Among the hPSC-derived lung cells, alveolar type II and ciliated cells are the major cell populations expressing the viral receptor ACE2 and co-effector TMPRSS2, and both were highly permissive to viral infection. We found that alternatively polarized macrophages (M2) and classically polarized macrophages (M1) had similar inhibitory effects on SARS-CoV-2 infection. However, only M1 macrophages significantly up-regulated inflammatory factors including IL-6 and IL-18, inhibiting growth and enhancing apoptosis of lung cells. Inhibiting viral entry into target cells using an ACE2 blocking antibody enhanced the activity of M2 macrophages, resulting in nearly complete clearance of virus and protection of lung cells. These results suggest a potential therapeutic strategy, in that by blocking viral entrance to target cells while boosting anti-inflammatory action of macrophages at an early stage of infection, M2 macrophages can eliminate SARS-CoV-2, while sparing lung cells and suppressing the dysfunctional hyper-inflammatory response mediated by M1 macrophages.
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Type of study: Prognostic_studies Language: En Journal: Res Sq Year: 2020 Document type: Article

Full text: 1 Collection: 01-internacional Database: MEDLINE Type of study: Prognostic_studies Language: En Journal: Res Sq Year: 2020 Document type: Article