Your browser doesn't support javascript.
loading
Increased airway epithelial cell-derived exosomes activate macrophage-mediated allergic inflammation via CD100 shedding.
Yu, Yi; Zhou, Yao; Di, Caixia; Zhao, Caiqi; Chen, Jie; Su, Wen; Wu, Qun; Wu, Min; Su, Xiao; Xia, Zhenwei.
Afiliação
  • Yu Y; Department of Pediatrics, Ruijin Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai, China.
  • Zhou Y; Department of Pediatrics, Ruijin Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai, China.
  • Di C; Department of Pediatrics, Ruijin Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai, China.
  • Zhao C; Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.
  • Chen J; Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.
  • Su W; Department of Pediatrics, Ruijin Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai, China.
  • Wu Q; Department of Pediatrics, Ruijin Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai, China.
  • Wu M; School of Medicine and Health Sciences, Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND, USA.
  • Su X; Unit of Respiratory Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.
  • Xia Z; Department of Pediatrics, Ruijin Hospital, Shanghai Jiao-tong University School of Medicine, Shanghai, China.
J Cell Mol Med ; 25(18): 8850-8862, 2021 09.
Article em En | MEDLINE | ID: mdl-34414666
ABSTRACT
Airway epithelial cells (AECs) participate in allergic airway inflammation by producing mediators in response to allergen stimulation. Whether ovalbumin (OVA) challenge promotes exosome release from AECs (OVA-challenged AEC-derived exosomes (OAEs)), thereby affecting airway inflammation, as well as the underlying mechanisms, is unknown. Our study showed that AECs released an increased number of exosomes after OVA challenge, and the expression of Plexin B2 (PLXNB2; a natural CD100 ligand) was increased by a massive 85.7-fold in OAEs than in PBS-treated AEC-derived exosomes (PAEs). CD100+ F4/80+ macrophages engulfed OAEs to trigger the transcription of pro-inflammatory chemokines and cytokines. Plxnb2 transcripts increased in asthmatic lungs, and similarly, PLXNB2 protein was highly enriched in exosomes purified from asthmatic bronchoalveolar lavage (BAL) fluid. Furthermore, aspiration of PLXNB2 or OAEs increased the recruitment of lung neutrophils, monocytes, eosinophils and dendritic cells in OVA-challenged mice. Mechanistically, OAE aspiration enhanced the cleavage of CD100 by MMP14, which manifested as an increase in the soluble CD100 (sCD100) level in BAL fluid and lung homogenates. Knockdown of Mmp14 in macrophages prevented the cleavage of CD100 and reduced Ccl2, Ccl5 and Cxcl2 transcription. These data indicate that PLXNB2-containing OAEs aggravate airway asthmatic inflammation via cleavage of CD100 by MMP14, suggesting potential therapeutic targets of OAE-mediated asthma exacerbations.
Assuntos
Palavras-chave

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Asma / Antígenos CD / Semaforinas / Exossomos / Inflamação Limite: Animals / Female / Humans Idioma: En Ano de publicação: 2021 Tipo de documento: Article

Texto completo: 1 Coleções: 01-internacional Base de dados: MEDLINE Assunto principal: Asma / Antígenos CD / Semaforinas / Exossomos / Inflamação Limite: Animals / Female / Humans Idioma: En Ano de publicação: 2021 Tipo de documento: Article