4.6 Article

Airway Macrophages Encompass Transcriptionally and Functionally Distinct Subsets Altered by Smoking

出版社

AMER THORACIC SOC
DOI: 10.1165/rcmb.2021-0563OC

关键词

lung; airway macrophages; smoking; single-cell and bulk RNA-seq; COPD

资金

  1. Fonds de la Recherche scientifique (FRS)-FNRS
  2. FRFS-Walloon Excellence in Life Sciences and Biotechnology (FRFS-WELBIO) [20263584, 35049229]
  3. Acteria Foundation
  4. European Research Council
  5. FRFS-WELBIO
  6. Excellence of Science grant

向作者/读者索取更多资源

This study explores the heterogeneity of human alveolar macrophages (AMs) isolated from different groups, finding distinct subpopulations of AMs in BAL fluid based on autofluorescence levels. Single-cell RNA sequencing analyses reveal transcriptionally distinct clusters of classical and monocyte-derived AM enriched in smokers with and without COPD. Signs of gene signatures related to detoxification, oxidative stress, and proinflammatory responses are observed in smoking-associated clusters, indicating potential implications for COPD initiation or progression.
Alveolar macrophages (AMs) are functionally important innate cells involved in lung homeostasis and immunity and whose diversity in health and disease is a subject of intense investigations. Yet, it remains unclear to what extent conditions like smoking or chronic obstructive pulmonary disease (COPD) trigger changes in the AM compartment. Here, we aimed to explore heterogeneity of human AMs isolated from healthy nonsmokers, smokers without COPD, and smokers with COPD by analyzing BAL fluid cells by flow cytometry and bulk and single-cell RNA sequencing. We found that subpopulations of BAL fluid CD2061 macrophages could be distinguished based on their degree of autofluorescence in each subject analyzed. CD2061 autofluorescenthigh AMs were identified as classical, self-proliferative AM, whereas autofluorescentlow AMs were expressing both monocyte and classical AM-related genes, supportive of a monocytic origin. Of note, monocyte-derived autofluorescentlow AMs exhibited a functionally distinct immunoregulatory profile, including the ability to secrete the immunosuppressive cytokine IL-10. Interestingly, single-cell RNA-sequencing analyses showed that transcriptionally distinct clusters of classical and monocyte-derived AM were uniquely enriched in smokers with and without COPD as compared with healthy nonsmokers. Of note, such smoking-associated clusters exhibited gene signatures enriched in detoxification, oxidative stress, and proinflammatory responses. Our study independently confirms previous reports supporting that monocyte-derived macrophages coexist with classical AM in the airways of healthy subjects and patients with COPD and identifies smoking-associated changes in the AM compartment that may favor COPD initiation or progression.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.6
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据