Journal of Biomedical Science (Mar 2024)

Engineered extracellular vesicles carrying let-7a-5p for alleviating inflammation in acute lung injury

  • Sin-Yu Chen,
  • Yi-Ling Chen,
  • Po-Chen Li,
  • Tai-Shan Cheng,
  • Yeh-Shiu Chu,
  • Yi-Shan Shen,
  • Hsin-Tung Chen,
  • Wei-Ni Tsai,
  • Chien-Ling Huang,
  • Martin Sieber,
  • Yuan-Chieh Yeh,
  • Hsiao-Sheng Liu,
  • Chi-Ling Chiang,
  • Chih-Hung Chang,
  • Andrew S. Lee,
  • Yen-Han Tseng,
  • Ly James Lee,
  • Hsiu-Jung Liao,
  • Hon-Kan Yip,
  • Chi-Ying F. Huang

DOI
https://doi.org/10.1186/s12929-024-01019-4
Journal volume & issue
Vol. 31, no. 1
pp. 1 – 18

Abstract

Read online

Abstract Background Acute lung injury (ALI) is a life-threatening respiratory condition characterized by severe inflammation and lung tissue damage, frequently causing rapid respiratory failure and long-term complications. The microRNA let-7a-5p is involved in the progression of lung injury, inflammation, and fibrosis by regulating immune cell activation and cytokine production. This study aims to use an innovative cellular electroporation platform to generate extracellular vesicles (EVs) carring let-7a-5p (EV-let-7a-5p) derived from transfected Wharton’s jelly-mesenchymal stem cells (WJ-MSCs) as a potential gene therapy for ALI. Methods A cellular nanoporation (CNP) method was used to induce the production and release of EV-let-7a-5p from WJ-MSCs transfected with the relevant plasmid DNA. EV-let-7a-5p in the conditioned medium were isolated using a tangential flow filtration (TFF) system. EV characterization followed the minimal consensus guidelines outlined by the International Society for Extracellular Vesicles. We conducted a thorough set of therapeutic assessments, including the antifibrotic effects using a transforming growth factor beta (TGF-β)-induced cell model, the modulation effects on macrophage polarization, and the influence of EV-let-7a-5p in a rat model of hyperoxia-induced ALI. Results The CNP platform significantly increased EV secretion from transfected WJ-MSCs, and the encapsulated let-7a-5p in engineered EVs was markedly higher than that in untreated WJ-MSCs. These EV-let-7a-5p did not influence cell proliferation and effectively mitigated the TGF-β-induced fibrotic phenotype by downregulating SMAD2/3 phosphorylation in LL29 cells. Furthermore, EV-let-7a-5p regulated M2-like macrophage activation in an inflammatory microenvironment and significantly induced interleukin (IL)-10 secretion, demonstrating their modulatory effect on inflammation. Administering EVs from untreated WJ-MSCs slightly improved lung function and increased let-7a-5p expression in plasma in the hyperoxia-induced ALI rat model. In comparison, EV-let-7a-5p significantly reduced macrophage infiltration and collagen deposition while increasing IL-10 expression, causing a substantial improvement in lung function. Conclusion This study reveals that the use of the CNP platform to stimulate and transfect WJ-MSCs could generate an abundance of let-7a-5p-enriched EVs, which underscores the therapeutic potential in countering inflammatory responses, fibrotic activation, and hyperoxia-induced lung injury. These results provide potential avenues for developing innovative therapeutic approaches for more effective interventions in ALI.

Keywords