Single-cell transcriptomic analysis of human lung provides insights into the pathobiology of pulmonary fibrosis

PA Reyfman, JM Walter, N Joshi… - American journal of …, 2019 - atsjournals.org
PA Reyfman, JM Walter, N Joshi, KR Anekalla, AC McQuattie-Pimentel, S Chiu…
American journal of respiratory and critical care medicine, 2019atsjournals.org
Rationale: The contributions of diverse cell populations in the human lung to pulmonary
fibrosis pathogenesis are poorly understood. Single-cell RNA sequencing can reveal
changes within individual cell populations during pulmonary fibrosis that are important for
disease pathogenesis. Objectives: To determine whether single-cell RNA sequencing can
reveal disease-related heterogeneity within alveolar macrophages, epithelial cells, or other
cell types in lung tissue from subjects with pulmonary fibrosis compared with control …
Rationale: The contributions of diverse cell populations in the human lung to pulmonary fibrosis pathogenesis are poorly understood. Single-cell RNA sequencing can reveal changes within individual cell populations during pulmonary fibrosis that are important for disease pathogenesis.
Objectives: To determine whether single-cell RNA sequencing can reveal disease-related heterogeneity within alveolar macrophages, epithelial cells, or other cell types in lung tissue from subjects with pulmonary fibrosis compared with control subjects.
Methods: We performed single-cell RNA sequencing on lung tissue obtained from eight transplant donors and eight recipients with pulmonary fibrosis and on one bronchoscopic cryobiospy sample from a patient with idiopathic pulmonary fibrosis. We validated these data using in situ RNA hybridization, immunohistochemistry, and bulk RNA-sequencing on flow-sorted cells from 22 additional subjects.
Measurements and Main Results: We identified a distinct, novel population of profibrotic alveolar macrophages exclusively in patients with fibrosis. Within epithelial cells, the expression of genes involved in Wnt secretion and response was restricted to nonoverlapping cells. We identified rare cell populations including airway stem cells and senescent cells emerging during pulmonary fibrosis. We developed a web-based tool to explore these data.
Conclusions: We generated a single-cell atlas of pulmonary fibrosis. Using this atlas, we demonstrated heterogeneity within alveolar macrophages and epithelial cells from subjects with pulmonary fibrosis. These results support the feasibility of discovery-based approaches using next-generation sequencing technologies to identify signaling pathways for targeting in the development of personalized therapies for patients with pulmonary fibrosis.
ATS Journals