Fibroblasts are generally believed to play an important role in pulmonary fibrosis10. Although drugs that block fibroblasts can slow the decline in lung function in patients with pulmonary fibrosis, they cannot reduce the mortality of patients. Therefore, there is still much work to do to identify the important events and molecular mechanisms in pulmonary fibrosis to provide targets for pulmonary fibrosis treatments. Our study found that AT2-like cells proliferated and secreted IgA into the lung ECM. These cells account for much more than other lung cells in the total number of cells, and the activation of these cells greatly changes the composition of lung ECM.
IgA is usually distributed on the mucosal surface of the digestive tract and respiratory tract and is secreted by B cells. In our pulmonary fibrosis model, IgA is deposited in large amounts in the lung ECM, which is the first report in the world thus far. In fact, pulmonary fibrosis disease is clinically associated with autoimmune diseases such sarcoidosis and systemic sclerosis32. Studies have shown that IgA in fibrotic lungs can activate fibroblasts, promote their transdifferentiation into myofibroblasts and secrete collagen. Our findings provide a basis for fine contact between IgA and fibroblasts.
Lung alveolar progenitor cells can self-renew, proliferate and replenish damaged alveolar epithelium33. When they proliferate abnormally, they cause alveolar structural disorders15,16. Three kinds of lung progenitor cells have been reported. In our study, AT2-like cell cluster 1 was more similar to Sftpc+ Scgb3a1+ lung progenitor cells, and they secreted IgA to the ECM. Although immunoglobulins are thought to be derived from B cells, studies have shown that in lung cancer, epithelial cells can also secrete immunoglobulin IgG34. This is the first discovery that epithelial cells produce immunoglobulins in noncancer diseases.
It has been suggested that idiopathic pulmonary fibrosis will be reclassified in the next 10 years35. With this in mind, the findings in this study inspire us to investigate other types of pulmonary fibrosis and classify them according to the presence or absence of IgA deposition. Targeting the ablation of overproliferating AT2-like cells or blocking the production of IgA may become a potential target for the treatment of pulmonary fibrosis.
Extended data
Ethics
All animal experiments were approved by the Laboratory Animal Care and Use Committee of Southeast University. All procedures were conducted in accordance with the Declaration of Helsinki.
Mice
Male C57BL/6 mice were purchased from Hangzhou Ziyuan Experimental Animal Co., Ltd. Three to five mice were raised in a cage at a stable temperature (22 °C ± 2 °C) and humidity (40% ± 10%) with a 12-h light–dark cycle. Mice were allowed free access to food and water at specified feeding times.
Animal model
Silica with a diameter of 5 µm (80% of particles) was purchased from Sigma® (S5631). It was incubated at 180 degrees for 16 h to inactivate endotoxin. We used NS to generate a working solution of 50 mg/ml silica suspension before use. Male C57BL/6J mice (6 weeks of age, 22±1 g weight) were anaesthetized with pentobarbital sodium (1%, 50 mg/kg in ddH2O) administered by intraperitoneal injection. After the fur on the neck was shaved, the skin was disinfected with 75% alcohol, and an incision (1 cm) was made on the skin along the midline. We bluntly separated the soft tissue in front of the neck to expose the trachea. For the mice in the SiO2-7d and SiO2-56d groups, we administered 100 µl of silica suspension (50 mg/ml) to the lungs via intratracheal instillation. For the mice in the NS-7d and NS-56d groups, we administered NS via the same method. The mouse skin was sutured and disinfected, and they were closely observed until they recovered. All mice were fasted with water for 6 hours before and after the operation. We observed the mice every 8 hours for the first three days after the operation and once a day afterwards.
CT scanning
Mice were anaesthetized with inhaled isoflurane (induced concentration 3–44%, maintained concentration 1–1.5%), and when their breathing was stable, they were placed on the mouse platform, and CT (Hiscan XM Micro CT, Suzhou Hiscan Information Technology Co., Ltd.) detection was performed. The X-ray tube settings were 60 kV and 133 µA, and images were acquired at 50 µm resolution. A 0.5° rotation step through a 360° angular range with 50 ms exposure per step was used. The images were reconstructed with Hiscan Reconstruct software (Version 3.0, Suzhou Hiscan Information Technology Co., Ltd.) and analysed with Hiscan Analyzer software (Version 3.0, Suzhou Hiscan Information Technology Co., Ltd.).
Pulmonary function test
Mice were anaesthetized by pentobarbital sodium injection as described above. Then, we inserted a tracheal catheter and fastened it to the trachea. After finishing the preparatory work, we tested mouse pulmonary function (Cchord, FEV75 (volume expired in the first 75 ms of fast expiration), IC (inspiratory capacity, volume inspired during slow inspiration), MMEF (mean mid expiratory flow, average flow between 25%-75%), FVC (forced vital capacity, volume expired during fast expiration) and TLC (total lung capacity, FRC+IC) with the Forced Manoeuvres System (EMMS, Hants, UK). Each mouse was tested three times, and the most reliable result (removal of unusually high or unusually low value) was used for analysis. Finally, mice were sacrificed to harvest lung tissue, and the bodies were prepared for cremation by the university.
Processing of mouse lung tissue for histology
Fresh lung tissues were immediately fixed in 4% paraformaldehyde for 24 h at 4°C. Then, lung tissues were transferred to a 20% sucrose gradient overnight for dehydration. Then, they were transferred to a 30% sucrose gradient overnight for further dehydration. After being fully dehydrated, these lung tissues were stored at -80°C before the next experiment.
H&E staining
Tissues were cut into 8 µm sections at -20°C by a cryostat (Leica, Germany). The lung tissue sections were stained according to the instructions provided with the H&E staining kit (Biyun Tian, China). Briefly, the tissue sections were washed 3 times in precooled phosphate-buffered saline (1×PBS) and then stained with haematoxylin for 5 minutes and then transferred into acid alcohol fast differentiation solution for 10 seconds. After that, the sections were soaked in tap water for 15 minutes. Next, the cells were stained with Esion for 5 minutes and washed in running water. Then, the sections were dehydrated with 75%, 95%, 100%, 100% alcohol and 100% xylene for 1 min each. Finally, neutral gum and glass slides were used to cover the tissue.
After the sections were dried in air, the images were observed and photographed at 10x and 20x resolution with a microscope (EVOS FL AUTO2, Thermo, America). Images at 10x resolution were captured using EVOS Automated Imaging System software.
Sirius red staining
All tissue sections were stained according to the instructions of the Sirius Star Chromosome Kit (Abcam, USA). Briefly, 8-µm-thick lung tissue sections were washed in PBS 3 times for 5 minutes and then stained with Sirius red for 1 hour, followed by brief rinsing with 1% acetic acid. Finally, the images were observed and photographed at 10x and 20x resolution with a microscope (EVOS FL AUTO2, Thermo, America). Images at 10x resolution were captured using EVOS Automated Imaging System software.
ECM processing
1. ECM collection
Fresh lung tissues were cut into 200-µm-thick pieces by cryostat sectioning. After rinsing with PBS, 15 ml of lysis buffer (1% SDS in ddH2O) was added, and the tissue mixture was incubated at room temperature for 1 h on a shaker. Then, the lung tissues were transferred to new lysis buffer and incubated for 1 h again. Next, lung tissues were transferred into another tube with new lysis buffer and incubated overnight at room temperature. On the second day, the sections were incubated with 1% Triton X-100 (diluted with ddH2O) at room temperature for 1 h; this incubation was repeated twice. Next, the 1% Triton X-100 solution was renewed and incubated overnight at room temperature. On the third day, the slices were washed with PBS and ddH2O in turn for 5 minutes at room temperature. Next, they were put in sodium chloride solution (1 M) for 1 h incubation and then washed with PBS and ddH2O in turn for 5 minutes at room temperature. After that, the slices were incubated in solution containing DNase (20 µg/ml) and MgCl2 (4.2 mM) at 37°C for 1 h. Finally, the sections were washed with ddH2O. Membrane proteins and nuclear proteins were extracted, and lung ECM was obtained.
2. Extraction of ECM proteins
Lung ECM was added to an appropriate amount of SDS lysis buffer (Biyun Tian, China) and incubated at 37°C for 1 h, followed by centrifugation at room temperature at 15000 rcf for 15 min. The supernatant was collected as the ECM protein solution. The ECM protein solution was stored in PBS at 4°C before use.
Proteomic analysis of the ECM (PXD028194)
1. Experimental procedures
Total protein was extracted from 6 lung ECM samples (3 for the NS-56d group, namely, con111, con116, con117; 3 for the SiO2-56d group, namely, M80, M101, M107), and a portion (10 µl) of the protein was used to measure the protein concentration, followed by SDS-PAGE separation. Another part was collected for trypsin digestion and labelled with TMT (Tandem Mass Tags) reagents, con111 with 126, con 116 with 127, con117 with 128; M80 with 129, M101 with 130, M107 with 131. Equal amounts of each labelled sample were mixed, and an appropriate quantity of protein was taken to perform chromatographic separation. Finally, the samples were analysed by LC-MS (liquid chromatography mass spectrometry).
2. Analysis of LC-MS/MS data
The LC-MS/MS raw data was processed using Proteome Discover 2.4 (Thermo, USA). According to the unique peptide ≥ 1, keep any group of samples with the expression value ≥ 50% of the protein. Then, the missing values were imputed with the mean of the protein expression in corresponding group. Next, the data was median normalized and log2-transformed to obtain credible proteins. Then we performed statistical and visual display of these proteins using R software (version 4.2) ggplot2 package (version 3.2.2), including principal component analysis (PCA), sample correlation analysis (Extended Data Fig. 2a), sample hierarchical cluster analysis (Extended Data Fig. 2b), visual display of data after standardization (Extended Data Fig. 2c) and density plot (Extended Data Fig. 2d).
Based on credible proteins, we performed Student’s t test to identify significant difference of proteins in NS-56d group and SiO2-56d group. Fold change (FC) is used to evaluate the expression level of a certain protein between samples. The p-value (P) calculated by the t-test shows the significance of the difference between samples. Difference screening conditions are FC≥2.0 and P≤0.05. The clustering heat map based on R software (version 4.2) pheatmap package (version 1.0.12) can be used for quality control of standardized experimental data and data display after enrichment of differential data. Generally, samples of the same group can appear in the same cluster through clustering.
For the identified proteins, extract the annotation information based on such as Uniprot databases. After obtaining the differentially expressed proteins (FC≥2, P≤0.05), the GO and KEGG functional enrichment analyses of up-regulated proteins were performed with R software (version 4.2) ggplot2 package (version 3.2.2).
Western blotting
The proteins were treated with loading buffer at 100°C for 5 min. Then, equal amounts of proteins (20 µg) of lung ECM were added to a 12% SDS-PAGE gel, and after separation, the proteins were transferred to a 0.25 µm PVDF membrane. Next, the PVDF membrane was placed in 5% skim milk at room temperature for one hour to block nonspecific binding sites. Then, the membranes were incubated in primary antibody (anti-pro/mature surfactant protein B, ab40876, Abcam, 1:5000) at 4°C overnight. After being washed 4 times with TBST at room temperature for 8 min, the PVDF membranes were incubated with HRP-conjugated goat anti-mouse or anti-rabbit secondary antibodies (according to what primary antibody origination was used) for 1 h at room temperature. For the detection of IgA, we used goat anti-mouse IgA-HRP (Southern Biotech, SBA-1040-05, 1:5000) as both the primary antibody and secondary antibody. After unbound secondary antibodies were fully washed away with TBST, the signal was detected with ECL chemiluminescence solution (Millipore, America), and photos were taken with a chemiluminescence detection system (Tanon Scientific & Technology Co., China). For lung ECM samples, β-actin was used as an internal reference protein. For MLE-12 cell line samples, GAPDH was used as an internal reference protein.
Spatial transcriptomics(GSE183683)
1. Sample collection
For the model group, mice with obvious fibrotic lesions on CT were thought to be a successful model. Lung tissues were trimmed near the hilum in the horizontal direction and then immediately frozen in OCT on dry ice. These samples were stored at -80°C before next step.
2. Spatial transcriptomics sequencing
2.1 Staining and imaging
Cryosections were cut at 10 μm thickness with a cryostat (Leica, Germany) and mounted onto GEX arrays. The arrays were placed on a Thermocycler Adaptor with the active surface facing up and then incubated for 1 min at 37°C, fixed for 30 min with methyl alcohol at -20°C, and stained with H&E. Bright field images of the whole slide were acquired on a Leica DMI8 whole-slide scanner at 10x resolution.
2.2 Permeabilization and reverse transcription
Spatial gene expression analysis was performed using the Visium Spatial Gene Expression Slide and Reagent Kit (10x Genomics, PN-1000184). For each well, a slide cassette was used to create leakproof wells for adding reagents. Then, 70 μl of permeabilization enzyme was added, and the samples were incubated at 37°C. For the NS-7d, SiO2-7d and NS-56d samples, the incubation time was 24 min. However, for the SiO2-56d group, because of severe lung fibrosis, a 30-min incubation time was used. Each well was washed with 100 μl of SSC, and 75 μl of RT master mix was added for cDNA synthesis (65°C, 15 minutes, hold at 4°C).
2.3 cDNA library preparation for sequencing
At the end of first-strand synthesis, RT Master Mix was removed from the wells. Then, 75 μl of 0.08 M KOH was added and incubated for 5 min at room temperature. Then, KOH was removed from the wells, which were washed with 100 µl of EB buffer. Then, 75 μl of Second Strand Mix was added to each well for second-strand synthesis. cDNA amplification (98°C 3 min; 98°C 15 s, 63°C 20 s, 72°C 1 min, 14 cycles; 72°C 1 min, hold at 4°C)
was performed on a S1000TM Touch Thermal Cycler (Bio-Rad). According to the manufacturer’s instructions, Visium spatial libraries were constructed using a Visium spatial library construction kit (10x Genomics, PN-1000184). The libraries were sequenced using an Illumina NovaSeq6000 sequencer with a sequencing depth of at least 100,000 reads per spot with a paired-end 150 bp (PE150) reading strategy (performed by CapitalBio Technology, Beijing).
3. Analysis of spatial transcriptomics data
The 10X Space Ranger software, which process, align and summarize unique molecular identifier (UMI) counts against mmu10 mouse reference genome for each spot, was used to generate feature-barcode matrix. Only spots overlaying tissue sections were retained for further analysis. Unsupervised clustering was based on graph based algorithm with 10 principal components. The t-distributed stochastic neighborhood embedding (t-SNE) was performed to visualize the spots in two-dimensional space. Spatial feature expression plots were generated by Loupe Browser 4.1.0.
Single-cell sequencing (GSE183682)
1. Sample collection
For the model group (mice instilled with SiO2 suspension), mice with obviously high-density shadows on CT were included. Lung samples for single-cell sequencing were collected from four mice, namely, the NS-7d, SiO2-7d, NS-56d, and SiO2-56d samples. Whole lungs of each mouse were removed within 2 min of euthanasia and quickly washed in precooled PBS 3 times.
2. scRNA-seq
2.1 Cell capture and cDNA synthesis
Whole lung tissue was cut into small pieces (approximately 1 mm) and dissociated into single cells using Lung Dissociation Kit (Miltenyi Biotech, 130-095-927, Germany). With the Single-Cell 5' Library and Gel Bead Kit (10x Genomics, 1000169) and Chromium Single-Cell G Chip Kit (10x Genomics, 1000120), a cell suspension (300–600 living cells per microlitre determined by CountStar) was loaded onto a Chromium single-cell controller (10x Genomics) to generate single-cell gel beads in emulsion (GEMs) according to the manufacturer’s protocol. In short, single cells were suspended in PBS containing 0.04% BSA. Approximately 20,000 cells were added to each channel, and the target cell recovery was estimated to be approximately 10,000 cells. Captured cells were lysed, and the released RNA was barcoded through reverse transcription in individual GEMs. Reverse transcription was performed on a S1000TM Touch Thermal Cycler (Bio-Rad) at 53°C for 45 min, followed by 85°C for 5 min and a hold at 4°C. cDNA was generated and then amplified, and quality was assessed using an Agilent 4200 system (performed by CapitalBio Technology, Beijing).
2.2 scRNA-seq library preparation
The scRNA-seq libraries were constructed using the Single-Cell 5' Library and Gel Bead Kit, Single Cell V(D)J Enrichment Kit, Human T Cell (1000005) and Single Cell V(D)J Enrichment Kit according to the manufacturers’ instructions. The libraries were sequenced using an Illumina NovaSeq6000 sequencer with a sequencing depth of at least 100,000 reads per cell with a paired-end 150 bp (PE150) reading strategy (performed by CapitalBio Technology, Beijing).
3. Data preprocessing
3.1 Analysis of scRNA-seq data
Cell barcode filtering, alignment of reads and UMI counting were performed with Cell Ranger 4.0.0 (https://www.10xgenomics.com/). The scRNA-seq data for four samples was combined though Cellranger aggr. Principal component analysis (PCA) is performed on the normalized data. Unsupervised clustering was performed by Cellranger reanalyze using graph based algorithm. The top 10 principal components was used for clustering and t-SNE projections. The differential expression of genes between clusters were computed by sSEq and edgeR based method. The GO and KEGG functional enrichment analyses of marker genes (FC≥2, P≤0.05) in cluster 1 and 6 were performed by R package Metascape.
3.2 Cell type annotation
Cell types were determined by clustering and marker gene expression. Cluster 1, 3 and 15 highly expressed genes Sftpc, Sftpb and Sftpa1, which were defined as AT2-like cell. Cluster 6 highly expressed genes Itgax, Csf1r and Ly86, and was inferred to be macrophage. Monocyte was made up two clusters (cluster 4 and 8), which highly expressed Ccr2 and Csf1r. Other clusters highly expressed markers specific for dentritic cell (Ear1, Plet1), clara cell (Sftpb, Sftpc, Scgb3a1), AT1 cell (Cldn3, Epcam), Ccl3- Ccl4- neutrophils (Cxcr2, Stfa2l1), Ccl3+ Ccl4+ neutrophils (Il1f9, Ccl3, Ccl4), T cell (Thy1, Cd8a), B cell (Cd79a, Cd79b, Cd19), endothelial cell (Cdh5, Pecam1, Clec14a), fibroblast (Col1a1, Col3a1, Col6a2), red blood cell (Hba-a1, Hba-a2)
- BCR and TCR sequence reconstruction
Using a Chromium Single-Cell V(D)J Enrichment kit, we reconstructed full-length TCR/BCR V(D)J segments from amplified cDNA in 5′ libraries via PCR amplification of sc-RNA seq following the manufacturer’s protocol (10x Genomics). Using the Cell Ranger (v.3.0.2) vdj pipeline coupled with the mouse reference genome mm10, we conducted demultiplexing, gene quantification and TCR/BCR clonotype assignment. Cells with at least one complete TCR α-chain (TRA) or TCR β-chain (TRB) were retained for TCR analysis, and only cells with at least one complete heavy chain (IGH) or one light chain (IGK or IGL) were retained for BCR analysis. Each unique TRA(s)-TRB(s) pair or IGH(s)-IGK/IGL(s) pair was defined as an effective clonotype for further clonal expansion analysis. Cells with the same clonotype were considered clonal cells. Based on barcode information, cells with a TCR or BCR clonotype were projected on UMAP plots.
RNAscope
1. Preparation of lung tissue section
Lung tissues harvested from mouse of NS-56d and SiO2-56d (mouse and modeling methods are the same as in scRNA sequencing) were immediately frozen in OCT on dry ice. These samples were stored at -80°C before next step.
2. RNAscope in situ hybridization
All RNAscope experiments are performed strictly in accordance with ACD's instructions. After being equilibrated at -20°C for 1h, the tissues were cut into 12 µm sections at -20°C by a cryostat (Leica, Germany) and mounted onto SuperFrost Plus slides (Fisher, Scientific, 12-550-15). The tissue sections were air-dried at -20°C for 20 minutes and then immediately transferred into 4% paraformaldehyde (precooled at 4°C ) for 15 minutes. Next, the tissue sections were dehydrated in 50% ethanol, 70% ethanol, 100% ethanol and 100% ethanol in order at room temperature, each for 5 minutes. The sections were air-dried for 5 minutes at room temperature and then the boundaries of sections were drawn on slides using an ImmEdge hydrophobic pen. After the hydrophobic boundaries were dried, tissue sections were incubated with hydrogen peroxide solution for 10 minutes at room temperature, followed by briefly washed twice in PBS at room temperature. Tissue sections then were incubated with protease IV for 15 minutes at room temperature followed by briefly washed twice in PBS at room temperature.
The tissue sections were placed in humidity box and incubated with a mixture of Sftpc probe (C3, 570) and Igha probe (C2, 520) ( 1:50 diluted with probe diluent) at 4°C for 2 hours, followed by washed twice in 1×washing buffer at room temperature for 2 minutes. Then tissue sections were incubated with AMP-1, AMP-2, AMP-3 reagents for 30, 30, 15 minutes respectively at 40°C. After each amplification, the sections were washed twice in 1×washing buffer at room temperature for 2 minutes. The sections were incubated with HRP-C2 reagent at 40°C for 15 minutes, followed by washed twice in 1×washing buffer at room temperature for 2 minutes. After that, the sections were incubated with Opal 520 (1:1000 diluted with TSA diluent) at 40°C for 30 minutes, followed by washed twice in 1×washing buffer at room temperature for 2 minutes. The sections were incubated with HRP-C3 reagent at 40°C for 15 minutes, followed by washed twice in 1×washing buffer at room temperature for 2 minutes. After that, the sections were incubated with Opal 570 (1:1000 diluted with TSA diluent) at 40°C for 15 minutes, followed by washed twice in 1×washing buffer at room temperature for 2 minutes.
Finally, the sections were incubated with DAPI reagent for 30s. After briefly washed in PBS, the sections were covered with Prolong Gold Antifade mounting medium (Thermo Fisher Scientific, P36930, USA). All the slides were fully air-dried and then stored in dark at 4°C.
3. Image capture
Images were captured using a confocal (Olympus FV1000, Japan). We observe the overall condition of the sample through a 10× objective lens. The detailed pictures were taken with a 60× objective oil lens. Images of different channels were taken separately and merged using Olympus Fluoview software.
Cell culture
MLE12 mouse lung epithelial cells (ScienCell) were cultured in DMEM (Gibco) supplemented with 10% FBS (Corning) and 1% penicillin-streptomycin (Gibco) in a 5% CO2 and 37°C incubator (Thermo).
Protein extraction
Cells were washed 3 times with precooled PBS and then lysed with RIPA lysis buffer (Beyotime Biotechnology, China) supplemented with a protease inhibitor cocktail (Beyotime Biotechnology, China) on ice for 1 h, followed by centrifugation at 12000 rpm for 15 min. Then, the protein concentration in the supernatant was quantified using a BCA assay (Beyotime Biotechnology, China), and the extracted protein was heated for 5 min at 100°C. The protein solution was stored at -80°C before use.
scRNA-seq and spatial RNA-seq integration
In order to obtain the composition of cell types in each spot, we used anchor-based integration method in Seurat v3.2, which could transfer the label of scRNA-seq reference to a spatial RNA-seq query. First, both the scRNA-seq and spatial RNA-seq data were normalized with the SCTransform function. Then, the cell type prediction probabilities score were calculated for each spot through FindTransferAnchors and TransferData functions. Finally, the spot is assigned to the cell type with the highest score.