File Download
  Links for fulltext
     (May Require Subscription)
Supplementary

Article: Modeling COVID-19 with Human Pluripotent Stem Cell-Derived Cells Reveals Synergistic Effects of Anti-inflammatory Macrophages with ACE2 Inhibition Against SARS-CoV-2

TitleModeling COVID-19 with Human Pluripotent Stem Cell-Derived Cells Reveals Synergistic Effects of Anti-inflammatory Macrophages with ACE2 Inhibition Against SARS-CoV-2
Authors
KeywordsCOVID-19
SARS-CoV-2
Human pluripotent stem cell
Macrophages
disease modeling
Issue Date2020
PublisherResearch Square Company. The Journal's web site is located at https://www.researchsquare.com/
Citation
Research Square, 2020, p. rs.3.rs-62758 How to Cite?
AbstractDysfunctional 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.
DescriptionVersion 2
Persistent Identifierhttp://hdl.handle.net/10722/307686
PubMed Central ID

 

DC FieldValueLanguage
dc.contributor.authorDuan, F-
dc.contributor.authorGuo, L-
dc.contributor.authorYang, L-
dc.contributor.authorHan, Y-
dc.contributor.authorThakur, A-
dc.contributor.authorNilsson-Payant, BE-
dc.contributor.authorWang, P-
dc.contributor.authorZhang, Z-
dc.contributor.authorMa, CY-
dc.contributor.authorZhou, X-
dc.contributor.authorHan, T-
dc.contributor.authorZhang, T-
dc.contributor.authorWang, X-
dc.contributor.authorXu, D-
dc.contributor.authorDuan, X-
dc.contributor.authorXiang, J-
dc.contributor.authorTse, HF-
dc.contributor.authorLiao, C-
dc.contributor.authorLuo, W-
dc.contributor.authorHuang, FP-
dc.contributor.authorChen, YW-
dc.contributor.authorEvans, T-
dc.contributor.authorSchwartz, RE-
dc.contributor.authortenOever, B-
dc.contributor.authorHo, DD-
dc.contributor.authorChen, S-
dc.contributor.authorLian, Q-
dc.contributor.authorChen, HJ-
dc.date.accessioned2021-11-12T13:36:20Z-
dc.date.available2021-11-12T13:36:20Z-
dc.date.issued2020-
dc.identifier.citationResearch Square, 2020, p. rs.3.rs-62758-
dc.identifier.urihttp://hdl.handle.net/10722/307686-
dc.descriptionVersion 2-
dc.description.abstractDysfunctional 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.-
dc.languageeng-
dc.publisherResearch Square Company. The Journal's web site is located at https://www.researchsquare.com/-
dc.relation.ispartofResearch Square-
dc.rightsThis work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.-
dc.subjectCOVID-19-
dc.subjectSARS-CoV-2-
dc.subjectHuman pluripotent stem cell-
dc.subjectMacrophages-
dc.subjectdisease modeling-
dc.titleModeling COVID-19 with Human Pluripotent Stem Cell-Derived Cells Reveals Synergistic Effects of Anti-inflammatory Macrophages with ACE2 Inhibition Against SARS-CoV-2-
dc.typeArticle-
dc.identifier.emailTse, HF: hftse@hkucc.hku.hk-
dc.identifier.emailLian, Q: qzlian@hku.hk-
dc.identifier.authorityTse, HF=rp00428-
dc.identifier.authorityLian, Q=rp00267-
dc.description.naturepreprint-
dc.identifier.doi10.21203/rs.3.rs-62758/v2-
dc.identifier.pmid32839764-
dc.identifier.pmcidPMC7444287-
dc.identifier.hkuros330248-
dc.identifier.volumeAug 20;rs.3-
dc.identifier.spagers.3.rs-
dc.identifier.epage62758-
dc.publisher.placeUnited States-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats