Detailseite
Projekt Druckansicht

Die Rolle der Lungen Mesenchymalen Stromazellen in der Pathogenese der Bronchopulmonalen Dysplasie (BPD).

Antragstellerin Dr. Ivana Mizik, Ph.D.
Fachliche Zuordnung Pneumologie,Thoraxchirurgie
Zellbiologie
Förderung Förderung von 2019 bis 2021
Projektkennung Deutsche Forschungsgemeinschaft (DFG) - Projektnummer 417891009
 
Erstellungsjahr 2021

Zusammenfassung der Projektergebnisse

Defects of late lung development manifests in premature infants as chronic lung disease bronchopulmonary dysplasia (BPD). Previous studies demonstrated therapeutic properties of bone marrow and umbilical cord-derived mesenchymal stromal cells (MSCs) in animal models of BPD, however our knowledge of lung resident (L)-MSCs remains very limited. In this study we aimed to characterize L-MSCs in the developing lung, identify changes induced in L-MSCs in experimental BPD, and explore their therapeutic potential. We showed that L-MSCs express common MSC markers, differentiate and form colonies in culture. No significant differences were observed between the cells from healthy or BPD animals. Next, we assessed the therapeutic potential of L-MSCs in three different mouse models of BPD. Surprisingly, treatment with L-MSCs had no significant effect on lung architecture or function in developing BPD mice. In addition, L-MSCs derived from BPD animals did not induce lung injury when administered to normally developing mice. To better understand whether storage and culture may critically alter L-MSCs properties we decided to perform an extensive, state-of-the-art single-cell transcriptomic analysis of L-MSCs in both, in situ and in vitro settings. We utilized a single-cell RNA sequencing (scRNA- seq) approach, which allows for assessment of complex cellular dynamics during biological processes, such as development. First, we profiled over 66,000 cells from 36 mice at three timepoints during normal or impaired lung development secondary to hyperoxia-induced BPD. We observed dynamic populations of cells, including rare cell types and putative progenitors. Hyperoxia altered the composition of all cellular compartments, particularly alveolar epithelium, stromal fibroblasts, capillary endothelium, and macrophage populations. Pathway and cellular communication analysis further suggested inflammatory signaling as the main driver of hyperoxia-induced changes. Building up on this dataset, we further characterized the transcriptome of cultured L-MSCs. We described gene expression changes induced in L-MSCs by developmental age, exposure to hyperoxia, and culture. Finally, we determined the localization of L-MSCs in the developing lung and proposed novel, culture-stable markers for their identification. Results of these studies provide a new, important perspective on the nature of L-MSCs in the developing lung, their role in BPD pathogenesis, and the role cells preservation and handling play in L-MSCs transcriptional stability.

Projektbezogene Publikationen (Auswahl)

  • A lung tropic AAV vector improves survival in a mouse model of surfactant B deficiency. Nat Commun 2020;11(1):3929
    Kang MH, van Lieshout LP, Xu L, Domm JM, Vadivel A, Renesme L, Mühlfeld C, Hurskainen M, Mižíková I, Pei Y, van Vloten JP, Thomas SP, Milazzo C, Cyr-Depauw C, Whitsett JA, Nogee LM, Wootton SK, Thébaud B
    (Siehe online unter https://doi.org/10.1038/s41467-020-17577-8)
  • Looking at the developing lung in single-cell resolution. American Journal of Physiology-Lung Cellular and Molecular Physiology 2021;320:L680–L687
    Mižíková I, Thébaud B
    (Siehe online unter https://doi.org/10.1152/ajplung.00385.2020)
  • Single cell transcriptomic analysis of murine lung development on hyperoxia-induced damage. Nat Commun 2021;12:1565
    Hurskainen M, Mižíková I, Cook DP, Andersson N, Cyr-Deppauw C, Lesage F, Helle E, Renesme L, Jankov R, Heikinheimo M, Vanderhyden B, Thébaud B
    (Siehe online unter https://doi.org/10.1038/s41467-021-21865-2)
 
 

Zusatzinformationen

Textvergrößerung und Kontrastanpassung