Inflammation and Regeneration (Oct 2023)

CXCL10 deficiency limits macrophage infiltration, preserves lung matrix, and enables lung growth in bronchopulmonary dysplasia

  • Dharmesh V. Hirani,
  • Florian Thielen,
  • Siavash Mansouri,
  • Soula Danopoulos,
  • Christina Vohlen,
  • Pinar Haznedar-Karakaya,
  • Jasmine Mohr,
  • Rebecca Wilke,
  • Jaco Selle,
  • Thomas Grosch,
  • Ivana Mizik,
  • Margarete Odenthal,
  • Cristina M. Alvira,
  • Celien Kuiper-Makris,
  • Gloria S. Pryhuber,
  • Christian Pallasch,
  • S. van Koningsbruggen-Rietschel,
  • Denise Al-Alam,
  • Werner Seeger,
  • Rajkumar Savai,
  • Jörg Dötsch,
  • Miguel A. Alejandre Alcazar

DOI
https://doi.org/10.1186/s41232-023-00301-6
Journal volume & issue
Vol. 43, no. 1
pp. 1 – 20

Abstract

Read online

Abstract Preterm infants with oxygen supplementation are at high risk for bronchopulmonary dysplasia (BPD), a neonatal chronic lung disease. Inflammation with macrophage activation is central to the pathogenesis of BPD. CXCL10, a chemotactic and pro-inflammatory chemokine, is elevated in the lungs of infants evolving BPD and in hyperoxia-based BPD in mice. Here, we tested if CXCL10 deficiency preserves lung growth after neonatal hyperoxia by preventing macrophage activation. To this end, we exposed Cxcl10 knockout (Cxcl10 −/−) and wild-type mice to an experimental model of hyperoxia (85% O2)-induced neonatal lung injury and subsequent regeneration. In addition, cultured primary human macrophages and murine macrophages (J744A.1) were treated with CXCL10 and/or CXCR3 antagonist. Our transcriptomic analysis identified CXCL10 as a central hub in the inflammatory network of neonatal mouse lungs after hyperoxia. Quantitative histomorphometric analysis revealed that Cxcl10 −/− mice are in part protected from reduced alveolar. These findings were related to the preserved spatial distribution of elastic fibers, reduced collagen deposition, and protection from macrophage recruitment/infiltration to the lungs in Cxcl10 −/− mice during acute injury and regeneration. Complimentary, studies with cultured human and murine macrophages showed that hyperoxia induces Cxcl10 expression that in turn triggers M1-like activation and migration of macrophages through CXCR3. Finally, we demonstrated a temporal increase of macrophage-related CXCL10 in the lungs of infants with BPD. In conclusion, our data demonstrate macrophage-derived CXCL10 in experimental and clinical BPD that drives macrophage chemotaxis through CXCR3, causing pro-fibrotic lung remodeling and arrest of alveolarization. Thus, targeting the CXCL10-CXCR3 axis could offer a new therapeutic avenue for BPD.

Keywords