↓ Skip to main content

Use of cilomilast-loaded phosphatiosomes to suppress neutrophilic inflammation for attenuating acute lung injury: the effect of nanovesicular surface charge

Overview of attention for article published in Journal of Nanobiotechnology, March 2018
Altmetric Badge

Citations

dimensions_citation
29 Dimensions

Readers on

mendeley
31 Mendeley
You are seeing a free-to-access but limited selection of the activity Altmetric has collected about this research output. Click here to find out more.
Title
Use of cilomilast-loaded phosphatiosomes to suppress neutrophilic inflammation for attenuating acute lung injury: the effect of nanovesicular surface charge
Published in
Journal of Nanobiotechnology, March 2018
DOI 10.1186/s12951-018-0364-z
Pubmed ID
Authors

Fu-Chao Liu, Huang-Ping Yu, Cheng-Yu Lin, Ahmed O. Elzoghby, Tsong-Long Hwang, Jia-You Fang

Abstract

Cilomilast is a phosphodiesterase 4 (PDE4) inhibitor for treating inflammatory lung diseases. This agent has a narrow therapeutic index with significant adverse effects on the nervous system. This study was conducted to entrap cilomilast into PEGylated phosphatidylcholine-rich niosomes (phosphatiosomes) to improve pulmonary delivery via the strong affinity to pulmonary surfactant film. Neutrophils were used as a cell model to test the anti-inflammatory activity of phosphatiosomes. In an in vivo approach, mice were given lipopolysaccharide to produce acute lung injury. The surface charge in phosphatiosomes that influenced the anti-inflammatory potency is discussed in this study. The average diameter of the phosphatiosomes was about 100 nm. The zeta potential of anionic and cationic nanovesicles was - 35 and 32 mV, respectively. Cilomilast in both its free and nanocapsulated forms inhibited superoxide anion production but not elastase release in activated neutrophils. Cationic phosphatiosomes mitigated calcium mobilization far more effectively than the free drug. In vivo biodistribution evaluated by organ imaging demonstrated a 2-fold ameliorated lung uptake after dye encapsulation into the phosphatiosomes. The lung/brain distribution ratio increased from 3 to 11 after nanocarrier loading. The intravenous nanocarriers deactivated the neutrophils in ALI, resulting in the elimination of hemorrhage and alveolar wall damage. Only cationic phosphatiosomes could significantly suppress IL-1β and TNF-α in the inflamed lung tissue. These results suggest that phosphatiosomes should further be investigated as a potential nanocarrier for the treatment of pulmonary inflammation.

Mendeley readers

Mendeley readers

The data shown below were compiled from readership statistics for 31 Mendeley readers of this research output. Click here to see the associated Mendeley record.

Geographical breakdown

Country Count As %
Unknown 31 100%

Demographic breakdown

Readers by professional status Count As %
Student > Doctoral Student 4 13%
Researcher 3 10%
Student > Bachelor 3 10%
Student > Ph. D. Student 3 10%
Other 2 6%
Other 5 16%
Unknown 11 35%
Readers by discipline Count As %
Pharmacology, Toxicology and Pharmaceutical Science 4 13%
Agricultural and Biological Sciences 3 10%
Nursing and Health Professions 2 6%
Biochemistry, Genetics and Molecular Biology 2 6%
Medicine and Dentistry 2 6%
Other 5 16%
Unknown 13 42%