Magnetic targeting increases mesenchymal stromal cell retention in lungs and enhances beneficial effects on pulmonary damage in experimental silicosis

Abstract Silicosis is a pneumoconiosis caused by inhaled crystalline silica microparticles, which trigger inflammatory responses and granuloma formation in pulmonary parenchyma, thus affecting lung function. Although systemic administration of mesenchymal stromal cells (MSCs) ameliorates lung inflam...

Full description

Bibliographic Details
Main Authors: Luisa H. A. Silva, Mariana C. Silva, Juliana B. Vieira, Emilia C. D. Lima, Renata C. Silva, Daniel J. Weiss, Marcelo M. Morales, Fernanda F. Cruz, Patricia R. M. Rocco
Format: Article
Language:English
Published: Wiley 2020-10-01
Series:Stem Cells Translational Medicine
Subjects:
Online Access:https://doi.org/10.1002/sctm.20-0004
id doaj-7c4f907b737341bdb88e48d50501fb0b
record_format Article
spelling doaj-7c4f907b737341bdb88e48d50501fb0b2020-11-25T02:55:05ZengWileyStem Cells Translational Medicine2157-65642157-65802020-10-019101244125610.1002/sctm.20-0004Magnetic targeting increases mesenchymal stromal cell retention in lungs and enhances beneficial effects on pulmonary damage in experimental silicosisLuisa H. A. Silva0Mariana C. Silva1Juliana B. Vieira2Emilia C. D. Lima3Renata C. Silva4Daniel J. Weiss5Marcelo M. Morales6Fernanda F. Cruz7Patricia R. M. Rocco8Laboratory of Pulmonary Investigation Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro Rio de Janeiro BrazilLaboratory of Pulmonary Investigation Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro Rio de Janeiro BrazilLaboratory of Pulmonary Investigation Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro Rio de Janeiro BrazilInstitute of Chemistry, Federal University of Goias Goiânia Goiás BrazilNational Institute of Metrology, Quality and Technology (INMETRO) Duque de Caxias Rio de Janeiro BrazilDepartment of Medicine University of Vermont, College of Medicine Burlington Vermont USANational Institute of Science and Technology for Regenerative Medicine Rio de Janeiro Rio de Janeiro BrazilLaboratory of Pulmonary Investigation Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro Rio de Janeiro BrazilLaboratory of Pulmonary Investigation Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro Rio de Janeiro Rio de Janeiro BrazilAbstract Silicosis is a pneumoconiosis caused by inhaled crystalline silica microparticles, which trigger inflammatory responses and granuloma formation in pulmonary parenchyma, thus affecting lung function. Although systemic administration of mesenchymal stromal cells (MSCs) ameliorates lung inflammation and attenuates fibrosis in experimental silicosis, it does not reverse collagen deposition and granuloma formation. In an attempt to improve the beneficial effects of MSCs, magnetic targeting (MT) has arisen as a potential means of prolonging MSC retention in the lungs. In this study, MSCs were incubated with magnetic nanoparticles and magnets were used for in vitro guidance of these magnetized MSCs and to enhance their retention in the lungs in vivo. In vitro assays indicated that MT improved MSC transmigration and expression of chemokine receptors. In vivo, animals implanted with magnets for 48 hours had significantly more magnetized MSCs in the lungs, suggesting improved MSC retention. Seven days after magnet removal, silicotic animals treated with magnetized MSCs and magnets showed significant reductions in static lung elastance, resistive pressure, and granuloma area. In conclusion, MT is a viable technique to prolong MSC retention in the lungs, enhancing their beneficial effects on experimentally induced silicosis. MT may be a promising strategy for enhancing MSC therapies for chronic lung diseases.https://doi.org/10.1002/sctm.20-0004magnetic fieldsmesenchymal stem cellsnanoparticlespulmonary fibrosissilicosis
collection DOAJ
language English
format Article
sources DOAJ
author Luisa H. A. Silva
Mariana C. Silva
Juliana B. Vieira
Emilia C. D. Lima
Renata C. Silva
Daniel J. Weiss
Marcelo M. Morales
Fernanda F. Cruz
Patricia R. M. Rocco
spellingShingle Luisa H. A. Silva
Mariana C. Silva
Juliana B. Vieira
Emilia C. D. Lima
Renata C. Silva
Daniel J. Weiss
Marcelo M. Morales
Fernanda F. Cruz
Patricia R. M. Rocco
Magnetic targeting increases mesenchymal stromal cell retention in lungs and enhances beneficial effects on pulmonary damage in experimental silicosis
Stem Cells Translational Medicine
magnetic fields
mesenchymal stem cells
nanoparticles
pulmonary fibrosis
silicosis
author_facet Luisa H. A. Silva
Mariana C. Silva
Juliana B. Vieira
Emilia C. D. Lima
Renata C. Silva
Daniel J. Weiss
Marcelo M. Morales
Fernanda F. Cruz
Patricia R. M. Rocco
author_sort Luisa H. A. Silva
title Magnetic targeting increases mesenchymal stromal cell retention in lungs and enhances beneficial effects on pulmonary damage in experimental silicosis
title_short Magnetic targeting increases mesenchymal stromal cell retention in lungs and enhances beneficial effects on pulmonary damage in experimental silicosis
title_full Magnetic targeting increases mesenchymal stromal cell retention in lungs and enhances beneficial effects on pulmonary damage in experimental silicosis
title_fullStr Magnetic targeting increases mesenchymal stromal cell retention in lungs and enhances beneficial effects on pulmonary damage in experimental silicosis
title_full_unstemmed Magnetic targeting increases mesenchymal stromal cell retention in lungs and enhances beneficial effects on pulmonary damage in experimental silicosis
title_sort magnetic targeting increases mesenchymal stromal cell retention in lungs and enhances beneficial effects on pulmonary damage in experimental silicosis
publisher Wiley
series Stem Cells Translational Medicine
issn 2157-6564
2157-6580
publishDate 2020-10-01
description Abstract Silicosis is a pneumoconiosis caused by inhaled crystalline silica microparticles, which trigger inflammatory responses and granuloma formation in pulmonary parenchyma, thus affecting lung function. Although systemic administration of mesenchymal stromal cells (MSCs) ameliorates lung inflammation and attenuates fibrosis in experimental silicosis, it does not reverse collagen deposition and granuloma formation. In an attempt to improve the beneficial effects of MSCs, magnetic targeting (MT) has arisen as a potential means of prolonging MSC retention in the lungs. In this study, MSCs were incubated with magnetic nanoparticles and magnets were used for in vitro guidance of these magnetized MSCs and to enhance their retention in the lungs in vivo. In vitro assays indicated that MT improved MSC transmigration and expression of chemokine receptors. In vivo, animals implanted with magnets for 48 hours had significantly more magnetized MSCs in the lungs, suggesting improved MSC retention. Seven days after magnet removal, silicotic animals treated with magnetized MSCs and magnets showed significant reductions in static lung elastance, resistive pressure, and granuloma area. In conclusion, MT is a viable technique to prolong MSC retention in the lungs, enhancing their beneficial effects on experimentally induced silicosis. MT may be a promising strategy for enhancing MSC therapies for chronic lung diseases.
topic magnetic fields
mesenchymal stem cells
nanoparticles
pulmonary fibrosis
silicosis
url https://doi.org/10.1002/sctm.20-0004
work_keys_str_mv AT luisahasilva magnetictargetingincreasesmesenchymalstromalcellretentioninlungsandenhancesbeneficialeffectsonpulmonarydamageinexperimentalsilicosis
AT marianacsilva magnetictargetingincreasesmesenchymalstromalcellretentioninlungsandenhancesbeneficialeffectsonpulmonarydamageinexperimentalsilicosis
AT julianabvieira magnetictargetingincreasesmesenchymalstromalcellretentioninlungsandenhancesbeneficialeffectsonpulmonarydamageinexperimentalsilicosis
AT emiliacdlima magnetictargetingincreasesmesenchymalstromalcellretentioninlungsandenhancesbeneficialeffectsonpulmonarydamageinexperimentalsilicosis
AT renatacsilva magnetictargetingincreasesmesenchymalstromalcellretentioninlungsandenhancesbeneficialeffectsonpulmonarydamageinexperimentalsilicosis
AT danieljweiss magnetictargetingincreasesmesenchymalstromalcellretentioninlungsandenhancesbeneficialeffectsonpulmonarydamageinexperimentalsilicosis
AT marcelommorales magnetictargetingincreasesmesenchymalstromalcellretentioninlungsandenhancesbeneficialeffectsonpulmonarydamageinexperimentalsilicosis
AT fernandafcruz magnetictargetingincreasesmesenchymalstromalcellretentioninlungsandenhancesbeneficialeffectsonpulmonarydamageinexperimentalsilicosis
AT patriciarmrocco magnetictargetingincreasesmesenchymalstromalcellretentioninlungsandenhancesbeneficialeffectsonpulmonarydamageinexperimentalsilicosis
_version_ 1724718378979950592