key: cord-0851354-rrknvhk6 authors: Zhao, Zixian; Zhao, Yu; Zhou, Yueqing; Wang, Xiaofan; Zhang, Ting; Zuo, Wei title: Single-cell analysis reveals the function of lung progenitor cells in COVID-19 patients date: 2020-07-13 journal: bioRxiv DOI: 10.1101/2020.07.13.200188 sha: e7417bbb88023c950d0bd58eb2ccf3a7e91235cf doc_id: 851354 cord_uid: rrknvhk6 The high mortality of severe 2019 novel coronavirus disease (COVID-19) cases is mainly caused by acute respiratory distress syndrome (ARDS), which is characterized by increased permeability of the alveolar epithelial barriers, pulmonary edema and consequently inflammatory tissue damage. Some but not all patients showed full functional recovery after the devastating lung damage, and so far there is little knowledge about the lung repair process1. Here by analyzing the bronchoalveolar lavage fluid (BALF) of COVID-19 patients through single cell RNA-sequencing (scRNA-Seq), we found that in severe (or critical) cases, there is remarkable expansion of TM4SF1+ and KRT5+ lung progenitor cells. The two distinct populations of progenitor cells could play crucial roles in alveolar cell regeneration and epithelial barrier re-establishment, respectively. In order to understand the function of KRT5+ progenitors in vivo, we transplanted a single KRT5+ cell-derived cell population into damaged mouse lung. Time-course single-cell transcriptomic analysis showed that the transplanted KRT5+ progenitors could long-term engrafted into host lung and differentiate into HOPX+ OCLN+ alveolar barrier cell which restored the epithelial barrier and efficiently prevented inflammatory cell infiltration. Similar barrier cells were also identified in some COVID-19 patients with massive leukocyte infiltration. Altogether this work uncovered the mechanism that how various lung progenitor cells work in concert to prevent and replenish alveoli loss post severe SARS-CoV-2 infection. worldwide. Pathological studies of COVID-19 postmortem lungs have shown that the effect of mild virus infection is limited in upper airway and had little influence on the lung tissue integrity. However, severe virus infection leads to diffuse alveolar damage (DAD) characterized apoptosis, desquamation of alveolar epithelial cells, and infiltration of inflammatory cells into alveolar cavity, which could eventually lead to hypoxemia, pulmonary tissue fibrosis and death of patients. Hyperplasia of type II alveolar cells (ATII) was also noted in most cases, which could suggest an undergoing regenerative process mediated by ATII lung progenitor cells [2] [3] [4] . In order to fully elucidate the epithelial damage and repair mechanism, we analyzed the single cell transcriptomic profile of lung BALF to quantify the major events post infection and focused on structural epithelial cells. BALF is a useful technique for sampling the human lung, providing landscape information of the whole lower respiratory tract. The current study was based on public scRNA-Seq datasets on BALF cells from three patients with moderate COVID-19 (M1 -M3), six patients with severe/critical infection (S1-S6) and four healthy controls (HC1-HC4) 5, 6 . Firstly, we performed unsupervised clustering analysis on the whole dataset to separate EPCAM+/TPPP3+/KRT18+ epithelial cells from other cells types (mostly immune cells) in the BALF (Extended Data Fig.1a,b) . Reclustering analysis identified 12 epithelial cell clusters, among them 4 were identified to be co-expressing immune markers which could be epithelial cells engulfed by leukocytes (Extended Data Fig. 1c,d) . The other 8 distinct epithelial cell clusters composed of Club/goblet cells (Cluster 0. SCGB1A1+/MUC5AC+), various types of ciliated cells (Cluster 1-5. FOXJ1+), alveolar cells (Cluster 6. HOPX+/SPC+). Most interestingly, a cluster of lung progenitor cells (Cluster 7. TM4SF1+/KRT5+/SOX9+) were identified, which will be analyzed in details as below ( Fig. 1a and Extended Data Fig. 2 ). When we compare the HC group with the other two infected groups, we found significant higher proportion of alveolar cell clusters (Cluster 6) in the BALF of patients with severe infection (Fig. 1b-d) . Of note, the HOPX+/AGER+ type I alveolar cells (ATI) and SPC+/LAMP3+ ATII were almost undetectable in the BALFs of healthy control persons due to the tissue integrity of their lungs. In contrast, in the severe COVID-19 patients, both ATI and ATII cell markers were detected in the lavage fluid, probably due to the tissue collapse and desquamation of alveolar cells (Fig. 1e ). This phenomenon was not obvious in moderate COVID-19 patients, which was also consistent with previous pathological observation 7 . Therefore, the number of alveolar cells (or the alveolar marker gene expression level) in BALFs could be clinically used to measure the structural integrity of lung, which could serve as an index of disease severity for COVID-19 patients. In the BALFs of patients with severe infection, we also found significant higher proportions of progenitor cell clusters (Cluster 7) (Fig. 1b-d) . Multiple stem/progenitor cell populations have been reported to play critical roles in damage repair after various types of acute lung injury 8 . Among them, a rare population of Wnt-responsive ATII is regarded as the major facultative progenitors 9,10 , which can be specifically marked by TM4SF1 expression in human lung 11 . In current study, we found that in the patients of severe group, the number of TM4SF1+ cells increased remarkably, which implicated the rapid activation of such progenitor cells by tissue damage (Fig. 2a,b) . Consistently, we observed co-expression of TM4SF1 and mature alveolar cell markers, AGER (also known as RAGE) and SPC, in a substantial proportion of cells in patients of severe group (Fig. 2c) . These results suggested that the TM4SF1+ progenitor cells had the potential to differentiate into mature alveolar cells and regenerate the damaged alveoli of COVID-19 patients. KRT5+ cells are also reported to have lung stem/progenitor characteristics. Such cells are originated from various primitive progenitors in proximal or distal airways and could expand/migrate to inflamed damaged lung parenchymal region to form "KRT5 pods" once activated by various types of tissue injury including influenza virus infection [12] [13] [14] [15] [16] [17] . Recent studies showed that the expanded KRT5 cells could give rise to new pulmonary epithelium, which was now believed to have important epithelial barrier function to protect the lung tissue from further damage 11, 17, 18 . Specific ablation of the newly expanded KRT5+ cells resulted in persistent hypoxemia, confirming the contribution of these cells in recovery of lung function 15 . In current study, we found that in the patients of severe group, the number of KRT5+ progenitor cells increased remarkably, in together with the elevated expression of another related progenitor marker gene SOX9 19, 20 (Fig. 2d and Extended Data Fig. 2c ). Then we examined the function of genes whose expression level were significantly up-regulated in KRT5+ cells by Gene Ontology analysis. The results showed that KRT5+ cells were responsive to the low oxygen condition of severe COVID-19 patients, and actively participated in the development and generation of respiratory epithelial system. More importantly, such cells highly expressed multiple tissue integrity genes including Claudin1, Claudin4, TJP1, Stratifin, AQP3 and Scnn1A, which were associated with epithelial barrier establishment, tight junction assembly, maintenance of fluid homeostasis and prevention of leukocyte mediated cytotoxicity (Fig. 2e) . These functions of KRT5+ cells were rather important for a damaged lung as reconstitution of tight alveolar barrier between atmosphere and fluid-filled tissue is required for recovery of normal gas exchange, while persistent disruption of the alveolar barrier could result in catastrophic consequences including alveolar flooding, "cytokine storm" attack from the circulating leukocytes and subsequent fibrotic scarring 21 . In order to elucidate the process that how KRT5+ progenitor cells restored mature alveolar barrier in injured lung, we isolated the mouse KRT5+ progenitor cells (previously also named distal airway stem cells) 15 for transplantation assay as described in Fig. 3a . Briefly, the cell population was trypsinized into single-cell suspension and a single cell-derived pedigree clone was propagated, followed by GFP labeling by lentiviral infection for further analysis. Immunostaining showed that all cultured GFP+ KRT5+ progenitor cells expressed KRT5 and P63 marker genes (Fig. 3b) . Then we transplanted the progenitors and the actively cycling Ki67+ KRT5+ progenitors. We also identified two KRT5-differentiated ABCs which both highly expressed early alveolar marker HOPX and paracellular adhesion marker Claudin7. One of the two ABC clusters highly expressing tight junction marker Occludin (OCLN) was supposed to be more mature than the other one ( Fig. 3f,g) . Gene ontology analysis demonstrated that the P63+ KRT5+ progenitors highly expressed multiple genes whose function was related to cell stemness maintenance, chemotaxis and inhibition of cell apoptosis. In contrast, the mature ABC highly expressed multiple genes (including HOPX, SPD, OCLN, CDH1, Claudin7, Claudin4, AQP3, etc.) whose function was related to lung morphogenesis, tight junction assembly and water homeostasis (Extended Data Fig. 3b ,c). To further dissect the lineage relationship between different clusters of engrafted cells, we performed the Monocle pseudo-time analysis based on the scRNA-Seq data. The result indicated that the P63+ KRT5+ progenitors could differentiate into P63-KRT5+ progenitors and immature ABCs, which eventually give rise to OCLN+ mature ABCs (Fig. 4a,b ). In consistent with the pseudo-time analytical data, we noticed that comparing to the 30-day engrafted cells, the 90-day engrafted cells had relatively more mature ABCs, less immature ABCs and less P63-KRT5+ progenitors (Fig. 4c ). Altogether such data revealed that the tight alveolar barrier would be gradually established by KRT5+ progenitor cell differentiation. Next we asked which molecular signaling pathways were involved in the establishment of alveolar barrier. Previous studies indicated that Notch signaling is critical for activation of P63+ KRT5+ progenitors in lung, but persistent Notch signaling prevents further differentiation of cells 16 . Consistently, here we found that the expression of multiple Notch pathway component genes was up-regulated in P63+ KRT5+ progenitors but gradually down-regulated when the cells were differentiating to mature ABC (Fig. 4d ). In addition, sense of low oxygen level through hypoxia pathway is known to be critical for the expansion of KRT5+ progenitors 22 . Here we found that the hypoxia pathway component gene expressions were relatively low in P63+ KRT5+ progenitors but were gradually up-regulated when the cells were differentiating to ABC (Fig. 4d) . (Fig. 4f) . So why the 4 patients have much more mature ABC than others? Interestingly, we found that generally the individuals' mature ABC cell numbers were positively correlated with their FCN1+ macrophage cell numbers in BALF (P=0.002), and the patient S2, S4, S5 and S6 had much more FCN1+ macrophages in BALF than most of the other individuals (Fig. 4g) . FCN1+ macrophages were reported to be highly proinflammatory and responsible for the tissue damage in COVID-19 patients 5 . Therefore, it seems that the establishment of new alveolar barrier was closely associated with severity of tissue damage and inflammation at individual level. Abnormal pulmonary function in COVID-19 patients at time of hospital discharge. The European respiratory journal 55 Pathological study of the 2019 novel coronavirus disease (COVID-19) through postmortem core biopsies. Modern pathology : an official journal of the United States and Canadian Academy of Pathology the lungs: report of 10 cases by postmortem needle autopsy Pathological findings of COVID-19 associated with acute respiratory distress syndrome. The Lancet Single-cell landscape of bronchoalveolar immune cells in patients with COVID-19 Proliferating SPP1/MERTK-expressing macrophages in idiopathic pulmonary fibrosis. The European respiratory journal 54 Pulmonary Pathology of Early-Phase Journal of thoracic oncology : official publication of the International Association for the Study of Lung Cancer The Cellular and Physiological Basis for Lung Repair and Regeneration: Past, Present, and Future Type 2 alveolar cells are stem cells in adult lung Single-cell Wnt signaling niches maintain stemness of alveolar type 2 cells Regeneration of the lung alveolus by an evolutionarily conserved epithelial progenitor Spatial-Temporal Lineage Restrictions of Embryonic p63(+) Progenitors Establish Distinct Stem Cell Pools in Adult Airways Rare SOX2(+) Airway Progenitor Cells Generate KRT5(+) Cells that Repopulate Damaged Alveolar Parenchyma following Influenza Virus Infection Distal airway stem cells yield alveoli in vitro and during lung regeneration following H1N1 influenza infection p63+Krt5+ distal airway stem cells are essential for lung regeneration Lineage-negative progenitors mobilize to regenerate lung epithelium after major injury Distinct Airway Epithelial Stem Cells Hide among Club Cells but Mobilize to Promote Alveolar Regeneration Persistent Pathology in Influenza-Infected Mouse Lungs Regeneration of functional alveoli by adult human SOX9 + airway basal cell transplantation Isolation and 3D expansion of multipotent Sox9(+) mouse lung progenitors Breakdown of Epithelial Barrier Integrity and Overdrive Activation of Alveolar Epithelial Cells in the Pathogenesis of Acute Respiratory Distress Syndrome and Lung Fibrosis Local lung hypoxia determines epithelial fate decisions during alveolar regeneration MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data clusterProfiler: an R package for comparing biological themes among gene clusters The Molecular Signatures Database (MSigDB) hallmark gene set collection The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells Construction of a human cell landscape at single-cell level Acknowledgements W. Zuo We salute to medical workers who sacrificed their lives in fight against the COVID-19 pandemic.