KEY RESOURCES TABLE
REAGENT or RESOURCE
|
SOURCE
|
IDENTIFIER
|
Antibodies
|
|
|
Mouse monoclonal anti-CD68
|
eBioscience
|
#14-0688-82
|
Rabbit polyclonal anti-CD80
|
BOSTER
|
#A00196-1
|
CD80-PE, human
|
miltenyi Biotec
|
#130-117-683
|
CD206-PE, human
|
miltenyi Biotec
|
#130-095-220
|
Rabbit monoclonal anti-CD163
|
Abcam
|
#ab182422
|
Rabbit polyclonal anti-IL6
|
Affinity
|
#DF6087
|
Goat polyclonal anti-ACE2
|
R&D system
|
#AF933
|
PE-conjugated CD43
|
eBioscience
|
#eBio84-3C1
|
APC-conjugated CD34
|
BD Biosciences
|
clone 581
|
PE-conjugated CD68
|
Biolegend
|
clone Y1/82A
|
APC-conjugated CD11b
|
Biolegend
|
clone ICRF44
|
FITC-conjugated CD14
|
Biolegend
|
clone HCD14
|
Anti-NKX2.1 Antibody
|
Seven Hills Bioreagents
|
#WRAB-1231
|
Anti-FOXA2 Antibody
|
Santa Cruz
|
#sc-6554
|
Anti-SOX2 Antibody
|
Santa Cruz
|
#sc-17320
|
Anti-SP-B Antibody
|
Seven Hills Bioreagents
|
#WRAB-48604
|
Anti-Pro-SP-C Antibody
|
Seven Hills Bioreagents
|
#WRAB-9337
|
Anti-FOXJ1 Antibody
|
Sigma-Aldrich
|
#HPA005714-1
|
Firefly luciferase Monoclonal Antibody (CS 17)
|
Thermo Fisher Scientific
|
#35-6700
|
Recombinant Anti-Firefly Luciferase antibody
|
Abcam
|
#ab185924
|
Mouse Anti-SARS-CoV-Spike antibody
|
Provided by Dr. Tom Moran
|
2B3E5
|
Donkey anti-Mouse IgG (H+L) Highly Cross-Adsorbed Secondary Antibody, Alexa Fluor 488
|
Thermo Fisher Scientific
|
#A-21202
|
Alexa Fluor 488 AffiniPure Donkey Anti-Guinea Pig IgG (H+L)
|
Jackson Immunoresearch Labs
|
#706-545-148
|
Donkey anti-Mouse IgG (H+L) Highly Cross-Adsorbed Secondary Antibody, Alexa Fluor 594
|
Thermo Fisher Scientific
|
#A-21203
|
Donkey anti-Rabbit IgG (H+L) Secondary Antibody, Alexa Fluor 594 conjugate
|
Thermo Fisher Scientific
|
#A-21207
|
Donkey anti-Rabbit IgG (H+L) Secondary Antibody, Alexa Fluor 647 conjugate
|
Thermo Fisher Scientific
|
#A-31573
|
Donkey anti-Mouse IgG (H+L) Secondary Antibody, Alexa Fluor 647
|
Thermo Fisher Scientific
|
#A-31571
|
Donkey anti-Goat IgG (H+L) Cross-Adsorbed Secondary Antibody, Alexa Fluor 647
|
Thermo Fisher Scientific
|
#A-21447
|
Donkey anti-Chicken IgG (H+L) Cross-Adsorbed Secondary Antibody, Alexa Fluor 488
|
Jackson Immunoresearch Labs
|
#703-545-155
|
Donkey anti-Sheep IgG (H+L) Cross-Adsorbed Secondary Antibody, Alexa Fluor 647
|
Thermo Fisher Scientific
|
#A-21448
|
Chemicals, Peptides, and Recombinant Proteins
|
|
|
Activin A
|
R&D Systems
|
#338-AC-500/CF
|
Y-27632
|
MedchemExpress
|
#HY-10583
|
Recombinant Human BMP-4 Protein
|
R & D Systems
|
#314-BP
|
Recombinant Human bFGF
|
R&D Systems
|
#233-FB-500
|
Dorsomorphin dihydrochloride
|
R&D Systems
|
#3093/50
|
SB431542
|
R&D Systems
|
#1614/50
|
IWP2
|
R&D Systems
|
#3533/50
|
CHIR99021
|
Cayman Chemical
|
#13122
|
Recombinant Human FGF-10 Protein
|
R&D Systems
|
#345-FG-250
|
Recombinant Human KGF/FGF-7 Protein
|
R&D Systems
|
#251-KG-01M
|
Retinoic acid
|
Sigma-Aldrich
|
#R2625
|
Dexamethasone
|
Sigma-Aldrich
|
#D4902
|
8-Bromo-cAMP
|
Sigma-Aldrich
|
#B5386
|
IBMX
|
Sigma-Aldrich
|
#I5879
|
Recombinant Human VEGF Protein
|
R&D Systems
|
#293-VE-500/CF
|
Recombinant Human IL-3 Protein
|
R&D Systems
|
#203-IL-050/CF
|
Recombinant Human M-CSF Protein
|
R&D Systems
|
#216-MC-025
|
Recombinant Human IL4 Protein
|
R&D Systems
|
#204-IL-050
|
IFNγ
|
R&D Systems
|
#285-IF-100
|
LPS
|
Sigma-Aldrich
|
#L4391-1MG
|
DAPI
|
Santa Cruz
|
#sc-3598
|
Hoechst 33342
|
Sigma-Aldrich
|
# B2261-100mg
|
Wright-Giemsa Stain
|
Sigma-Aldrich
|
#WG16-500ML
|
Culture Medium
|
|
|
F12
|
Gibco Thermo Fisher
|
#31765035
|
β-mercaptoethanol
|
Sigma Aldrich
|
#M3148
|
Penicillin-Streptomycin (5,000 U/mL)
|
Gibco Thermo Fisher
|
#15070063
|
MEM Non-Essential Amino Acids Solution (100X)
|
Gibco Thermo Fisher
|
#11140050
|
IMDM
|
Gibco Thermo Fisher
|
#21056023
|
GlutaMAX Supplement
|
Thermo Fisher Scientific
|
#35050079
|
Accutase
|
Stemcell Technologies
|
#07920
|
Matrigel
|
Corning
|
#354234
|
Fibronectin (FN)
|
Thermo Fisher Scientific
|
#356008
|
N2 supplement
|
Thermo Fisher Scientific
|
#17502-048
|
B27
|
Thermo Fisher Scientific
|
#12587-010
|
DMEM/F12
|
Thermo Fisher Scientific
|
#10565-018
|
Knockout serum replacement(KOSR)
|
Thermo Fisher Scientific
|
#10828-028
|
FBS
|
Gibco Thermo Fisher
|
#10099141C
|
Monothioglycerol
|
Sigma Aldrich
|
#M6145
|
Ascorbic acid
|
Sigma Aldrich
|
#A4403
|
Bovine serum albumin(BSA)
|
Sigma Aldrich
|
#A9418
|
Experimental Models: Cell Lines
|
Seven Hills Bioreagents
|
#WRAB-1231
|
hESC line H1
|
Harvard University
|
#0014
|
hESC line-RUES2
|
The Rockefeller University
|
#0013
|
HEK293T
|
ATCC
|
#CRL-11268
|
Vero E6
|
ATCC
|
#CRL-1586
|
THP-1
|
ATCC
|
#TIB-202
|
U937
|
ATCC
|
#CRL-1593.2
|
Mouse embryonic fibroblasts
|
Global Stem
|
#GSC-6001G
|
Software and Algorithms
|
Cell Ranger
|
10X Genomics
|
https://support.10xgenomics.com/single-cell-gene-expression/software/overview/welcome
|
Scran
|
Lun ATL, McCarthy DJ, Marioni JC (2016). “A step-by-step workflow for low-level analysis of single-cell RNA-seq data with Bioconductor.” F1000Res., 5, 2122. doi: 10.12688/f1000research.9501.2.
|
https://bioconductor.org/packages/release/bioc/html/scran.html
|
Rstudio
|
Rstudio
|
https://rstudio.com
|
Seurat R package v3.1.4
|
1
|
https://satijalab.org/seurat/
|
DAVID6.8
|
LHRI
|
https://david.ncifcrf.gov/home.jsp
|
Adobe illustrator CC2017
|
Adobe
|
https://www.adobe.com/product/photoshop.html
|
Graphpad Prism 8.0
|
Graphpad software
|
https://www.graphpad.com
|
LEGENDplex v8.0
|
Biolegend
|
https://www.biolegend.com/en-us/legendplex
|
FlowJo v x.0.7
|
BD Biosciences
|
https://www.flowjo.com/
|
ToppCell Atlas
|
Toppgene
|
https://toppgene.cchmc.org/
|
METHOD DETAILS
Patient’s lung tissues
The paraffin-embedded lung tissues were acquired from the department of pathology in the 3rd hospital of Shenzhen. They recently reported the pathological changes of lungs from a 66-year-old male died in critical COVID-19 infection2. The patient developed respiratory failure and septic shock during the treatment and was done with transplant. Informed consent was obtained from the patient and family. The diagnosis of COVID-19 pneumonia was based on the "Coronavirus Pneumonia Prevention and Control Plan" (7th edition) newly issued by the National Health Commission, China 3. Nasopharyngeal swabs were collected and COVID-19 was detected by real-time polymerase chain reaction. Infection was defined as at least two positive test results. Surgical informed consent was obtained and the study was approved by IRB in the third People's hospital of Shenzhen.
hPSC lung differentiation
Protocols for maintenance of hPSCs and generation of lung cells were slightly modified from previous studies 4,5. The hESC line-RUES2 or H1 was cultured on irradiated mouse embryonic fibroblasts (Global Stem, cat. no. GSC-6001G) at a density of 20,000- 25,000 cells/cm2 in a medium of DMEM/F12, 20% knockout serum replacement (Life Technologies), 0.1 mM β-mercaptoethanol (Sigma Aldrich) and 20 ng/ml bFGF (R&D Systems), and medium was changed daily. hESC cultures were maintained in an undifferentiated state at 37 °C in a 5% CO2/air environment until stem cells reached about 90% confluence.
hESC differentiation into endoderm was performed in serum-free differentiation (SFD) medium of IMDM/F12 (3:1) (Life Technologies) supplemented with N2 (Life Technologies), B27 (Life Technologies), 50 μg/ml ascorbic acid, 2 mM Glutamax, 0.4 μM monothioglycerol, 0.05% BSA at 37 °C in a 5% CO2/5% O2/95% N2 environment. hESCs were treated with Accutase and plated onto low attachment 6-well plates (Corning Incorporated, Tewksbury MA), resuspended in endoderm induction medium containing 10 μM Y-27632, 0.5 ng/ml human BMP-4, 2.5 ng/ml human bFGF, 100 ng/ml human Activin A, for 72-76 hours dependent on the formation rates of endoderm cells. On day 3, the endoderm bodies were dissociated into single cells using 0.05% Trypsin/0.02% EDTA and plated onto fibronectin-coated, 24-well tissue culture plates (~100,000–150,000 cells/well). For induction of anterior foregut endoderm (AFE), the endoderm cells were cultured in SFD medium supplemented with 1.5 μM Dorsomorphin dihydrochloride (R&D Systems) and 10 μM SB431542 (R&D Systems) for 48 h, and then switched to 24 h of 10 μM SB431542 and 1 μM IWP2 (R&D Systems) treatment. For induction of early stage lung progenitor cells (day 6–15), the resulting anterior foregut endoderm was treated with 3 μM CHIR99021, 10 ng/ml human FGF10, 10 ng/ml human FGF-7, 10 ng/ml human BMP-4 and 50-60nM all-trans retinoic acid (ATRA), in SFD medium for 8–10 d. The day 10-15 cultures were maintained in a 5% CO2/air environment. On days 15 and 16, the lung progenitor cells were replated after one minute trypsinization onto fibronectin-coated plates, in the presence of SFD containing 3 μM CHIR99021, 10 ng/ml human FGF10, 10 ng/ml human FGF7, in a 5% CO2/air environment. For differentiation of mature lung cells (day 25 to 55), cultures were re-plated after brief trypsinization onto 3.3% Matrigel-coated 24-well plates in SFD media containing maturation components containing 3 μM CHIR99021, 10 ng/ml human FGF-10; 10 ng/ml human FGF-7, and DCI (50 nM Dexamethasone, 0.1 mM 8-bromo-cAMP (Sigma Aldrich ) and 0.1 mM IBMX (3,7-dihydro-1-methyl-3-(2-methylpropyl)-1H-purine-2,6-dione) (Sigma Aldrich)). The protocol details are summarized in Figure S1A.
All embryonic stem cell studies were approved by the Institutional Review Board (IRB) at the University of Chicago, or by the Tri-Institutional ESCRO committee (Weill Cornell Medicine, Memorial Sloan Kettering Cancer Center, and Rockefeller University).
hPSC macrophage differentiation
We derived macrophages from hESC line H1 or RUES2 and adapted based on previously reported protocols6-8. For macrophage differentiation, at day -2, hESCs were digested into single-cell suspension by 1 mg/ml Accutase (Stemcell Technologies) and plated onto Matrigel-coated culture dishes at a density of 2× 104 cells/cm2 in mTeSR1 medium with 5uM Y27632 (MedchemExpress). After 24 h, Y27632 was withdrawn from the medium and cells were cultured for another 24 h. At day 0, cells were firstly induced by macrophage differentiation basal medium (SFD-M) which is RPMI 1640 medium supplemented with 2% B27 (Thermo Fisher Scientific), 1% L-GlutaMAX-I and 50 μg/ml ascorbic acid (Sigma Aldrich) and 10 ng/ml BMP4 (R&D Systems) for 24 h. Afterward, the medium was changed to SFD-M medium containing 10 ng/ml BMP4 and 2 μM GSK3 inhibitor CHIR99021 (Cayman Chemical) for another 48 h. At day 3, cells were replated onto Matrigel-coated dishes at a density of 4 × 104 cells/ cm2 in SFD-M medium with 50 ng/ml VEGF (R&D Systems) and 10 ng ng/ml FGF2 (R&D Systems) for 48 h. At day 5, the medium was replaced with basal medium with 50 ng/ml VEGF, 10 ng ng/ml FGF2 and 10uM TGFβ inhibitor SB431542 (R&D Systems) for another 72 h. At day 8-10, floating cells were collected and medium was changed and supplemented with 50ng/ml M-CSF and 10ng/ml IL3 (R&D Systems) for another 3-5 days. From day 11-13 onward, the medium was changed to SFD-M medium with 50 ng/ml M-CSF for 3 days. All differentiation steps were cultured under normoxic conditions at 37 ℃, 5% CO2. The protocol details are summarized in Figure S4A.
All embryonic stem cell studies were approved by the Institutional Review Board (IRB) at the University of Chicago, or by the Tri-Institutional ESCRO committee (Weill Cornell Medicine, Memorial Sloan Kettering Cancer Center, and Rockefeller University).
hPSC monocyte polarization
hPSC-derived CD14+ cells were plated on tissue culture plates at a density of 2x104 cells/cm2 in SFD-M medium supplemented with 50 ng/mL M-CSF. After 2 days of culture, monocytes differentiated into M0 macrophages and polarized to M1 or M2 macrophages. For macrophages polarization, 100ng/mL LPS (Sigma-Aldrich) and 10ng/mL IFNγ (R&D Systems) were added for M1 induction, or 20 ng/m IL-4 (R&D Systems) was added for M2 induction in SFD-M medium supplemented with 50 ng/mL M-CSF, respectively. These cells were cultured for another three days before examination for expression of the M1 or M2 makers.
Giemsa Staining
Differentiating day 11-13 monocytes/macrophages were fixed on slides using Cytospin, followed by staining using Wright-Giemsa Stain (Sigma-Aldrich) according to the manufacturer’s instructions.
Immunohistochemical staining
Histological study of lung tissues was performed on paraffin-embedded sections as previously described 9. For immunohistochemical staining, paraffin-embedded sections were deparaffinized and incubated with primary antibodies at 4°C overnight and secondary antibodies at room temperature for 1h. Primary antibodies and secondary antibodies are described in the supplementary Table. Nuclei were counterstained by Hoechst 33342 (Sigma). positive cells in lungs were randomly counted from different visions of slides by confocal microscopy. 12 views in each lung section were counted and averaged cell numbers per 0.042 mm were used to define the distributions of positive cells in the lung tissues as described 10. Living cells in culture were directly fixed in 4% paraformaldehyde for 25 min, followed with 15 min permeabilization in 1% triton X-100. For immunofluorescence, cells or tissue sections were immunostained with antibodies and counterstained with 4,6-diamidino-2-phenylindole (DAPI). Adjacent sections stained with H and E were used for comparison. The antibodies used for immunostaining or western blot experiments are listed in the key resource table.
Fluorescent activated cell sorting (FACS)
For FACS analysis, cells were resuspended in a FACS buffer (PBS with 0.1 % BSA and 2.5 mM EDTA). The cell suspension was then stained with PE-conjugated CD43 (Biolegend, clone MEM-59), APC-conjugated CD34 (BD, clone 581) to detect hematopoietic stem/progenitor cells (HSPC). PE-conjugated CD68 (Biolegend, clone Y1/82A), APC-conjugated CD11b (Biolegend, clone ICRF44), FITC-conjugated CD14 (Biolegend, clone HCD14) were used to detect monocyte/macrophages. Basically, cells were incubated with antibodies for 30 minutes at 4°C, followed with washed and suspended in 0.1% BSA/PBS buffer. PE and APC filters were then used to detect cells double positive for CD43 and CD34 or CD68 and CD11b by signal intensity gating, FITC and APC were used to detect cells double positive for CD14 and CD11b. Negative controls stained with control IgG instead of primary antibodies were always performed with sample measurements. Flowcytometry machine of BD FACSAria II and software of Flowjo were mainly used to collect and analyze the flowcytometry data.
Cytokine analysis
Cytokines including hIL-1β, IFN-α2, hIFN-γ, hTNF-α, hMCP-1, hIL-6, hIL-8, hIL-10, hIL-12p70, IL-17A, hIL-18, hIL-23, hIL-33 were detected according to the instruction of LEGENDplexTM kit (Biolegend, cat. no. 740808). In brief, 25ul supernatant was taken from the co-culture medium and mixed with 25 µl of premixed beads and 25 µl of detection antibodies. The mixtures were placed on a shaker at 400 r.p.m. for 2 h at RT. Then 25 µl of SA-PE was added to each tube and placed on a shaker at 500 r.p.m. for 30 min. The data were obtained by flow cytometry (FACSAria II, BD) and were analyzed with LEGENDplex v.8.0 (Biolegend).
SARS-CoV-2-Pseudo-Entry Viruses
Recombinant Indiana VSV (rVSV) expressing SARS-CoV-2 spikes was generated as previously described 11-13. HEK293T cells were grown to 80% confluency before transfection with pCMV3-SARS-CoV2-spike (kindly provided by Dr. Peihui Wang, Shandong University, China) using FuGENE 6 (Promega). Cells were cultured overnight at 37°C with 5% CO2. The next day, the media was removed and VSV-G pseudotyped ΔG-luciferase (G*ΔG-luciferase, Kerafast) was used to infect the cells in DMEM at an MOI of 3 for 1 hr before washing the cells with 1X DPBS three times. DMEM supplemented with 2% FBS and 100 I.U. /mL penicillin and 100 μg/mL streptomycin was added to the infected cells and they were cultured overnight as described above. The next day, the supernatant was harvested and clarified by centrifugation at 300xg for 10 min before aliquoting and storing at −80°C.
SARS-CoV-2 infections
SARS-CoV-2, isolate USA-WA1/2020 (NR-52281) was deposited by the Center for Disease Control and Prevention and obtained through BEI Resources, NIAID, NIH. SARS-CoV-2 was propagated in Vero E6 cells in DMEM supplemented with 2% FBS, 4.5 g/L D-glucose, 4 mM L-glutamine, 10 mM Non-Essential Amino Acids, 1 mM Sodium Pyruvate and 10 mM HEPES as described previously (insert citation for https://doi.org/10.1016/j.cell.2020.04.026).
hESC-derived lung and macrophage co-cultures in 96-well plates were infected with SARS-CoV-2 for 24 h at an MOI of 0.01 in the medium containing SFD:SFD-M=1:1. For immunofluorescence staining, cells were washed three times in PBS and fixed for 24 h in 5% formaldehyde for 24 h for immunofluorescent staining, prior to safe removal from the BSL-3 facility.
All work involving live SARS-CoV-2 was performed in the CDC/USDA-approved BSL-3 facility of the Global Health and Emerging Pathogens Institute at the Icahn School of Medicine at Mount Sinai in accordance with institutional biosafety requirements
SARS-CoV-2 entry virus infections.
To assay pseudo-typed virus infection, cells were seeded in 96 well plates. Pseudo-typed virus was added for MOI=0.01. At 2 hpi, the infection medium was replaced with fresh medium. At 24 hpi, cells were harvested for luciferase assay or immunohistochemistry analysis. For liver and lung organoids, organoids were seeded in 24-well plates, pseudo-typed virus was added for MOI=0.01 and centrifuged the plate at 1200g, 1 hour. At 24 hpi, organoids were fixed for immunohistochemistry or harvested for luciferase assay following the Luciferase Assay System protocol (E1501, Promega)
Single cell sequencing of hPSC-derived lung cells
Single-cell capture, reverse transcription, cell lysis, and library preparation was performed using the Single Cell 3′ version 3 kit and chip according to the manufacturer’s protocol (10x Genomics, USA). Single-cell suspensions were generated by dissociating the cultured RUES2 cells with 0.05% Trypsin/0.02% EDTA for 10-15 min, followed with passing through 40µM strainer. The single cell suspension was achieved through sorting the dissociated cells in flow cytometry singlets. Cell count was adjusted to 1000-2000 cells per ul to target an estimated capture of 8000 cells. Six input wells were used. Sequencing was performed on NovaSeq6000 with setting 28 for read 1 and 91 for read 2. The sequencing data were primarily analyzed by CellRanger pipeline v3.0.2 (10x Genomics, USA). In particular, raw fastq data were generated by CellRanger mkfastq; A custom reference genome was built by integrating the virus and luciferase sequences into the 10x pre-built human reference (GRCh38 v3.0.0) using CellRanger mkref. Alignment of the raw reads to the custom reference genome, removing duplicated transcripts using the unique molecular identifiers (UMIs) and assignment to single cells was performed using CellRanger count.
Briefly, we used cells Seurat 3.1.4 R package for data analysis and visualization 1. The Seurat object is required at least 200 and at most 6000 unique molecular identifiers (UMIs), genes detected (UMI count > 0) in less than two cells were removed. In addition, cells were excluded if more than 10% of sequences mapped to mitochondrial genes. In total, 5,080 cells from the sample passed these filters for quality.
Following the package suggestions, we used a linear model to mitigate the variation stemming from the number of detected unique molecules per cell. The differentially expressed genes were found by ‘‘vst’’ method and the top 3,000 differentially expressed genes were selected for PCA analysis. We used an elbow plot to determine the number of PCs. 20 PCs were used for each group of cells. Clustering resolution was set at 0.2. For co-culture analysis, Macrophages and lung cells were re-clustered and re-analyzed, respectively. Macrophages were integrated using the first 20 dimensions of PCs and clustering resolution was set at 0.1. UMAP plots, heatmaps, violin plots and dotplots were generated by the Seurat toolkit FeaturePlot, DoHeatmap, VlnPlot amd DotPlot functions, respectively. Cell types were determined using a combination of marker genes identified from the literature and the web-based tool Topp CellAtlas (https://toppgene.cchmc.org/)
QUANTIFICATION AND STATSTICAL ANALYSIS
Sample sizes for all figures and tables were estimated based on our previous studies4,5,14,15. For each set of experiments, samples were prepared for all experimental arms at the same time. All statistical tests are 2-sided. No adjustments were made for multiple comparisons. The relevant investigators (FD, LG and LY) were blinded to experimental allocations among different experimental arms for all experiments. N=3 independent biological replicates were used for all experiments unless otherwise indicated. n.s. indicates a non-significant difference. For all parametric statistical analyses, data were determined to be normally distributed by the D'Agostino-Pearson test. For all parametric and non-parametric tests, variances were similar between groups being compared. For comparison between experimental and control groups at a specific time point or tissue site, 2-sided Student t- tests, one-way or two-way ANOVA test, chi-squared tests and two sided Kolmogorov–Smirnov test were used. All cells (RUES2, H1, HEK293T, THP-1, U937) were purchased from ATCC or WiCell in the past 2 years and were negative for mycoplasma. The hESC lines were regularly checked for chromosome abnormalities and maintained with normal chromosome numbers.