Microvesicles released from pneumolysin-stimulated lung epithelial cells carry mitochondrial cargo and suppress neutrophil oxidative burst

Abstract Microvesicles (MVs) are cell-derived extracellular vesicles that have emerged as markers and mediators of acute lung injury (ALI). One of the most common pathogens in pneumonia-induced ALI is Streptococcus pneumoniae (Spn), but the role of MVs during Spn lung infection is largely unknown. I...

Descripción completa

Guardado en:
Detalles Bibliográficos
Autores principales: E. Letsiou, L. G. Teixeira Alves, D. Fatykhova, M. Felten, T. J. Mitchell, H.C. Müller-Redetzky, A. C. Hocke, M. Witzenrath
Formato: article
Lenguaje:EN
Publicado: Nature Portfolio 2021
Materias:
R
Q
Acceso en línea:https://doaj.org/article/5c41aea01663482497d68444e77e884d
Etiquetas: Agregar Etiqueta
Sin Etiquetas, Sea el primero en etiquetar este registro!
id oai:doaj.org-article:5c41aea01663482497d68444e77e884d
record_format dspace
spelling oai:doaj.org-article:5c41aea01663482497d68444e77e884d2021-12-02T15:37:59ZMicrovesicles released from pneumolysin-stimulated lung epithelial cells carry mitochondrial cargo and suppress neutrophil oxidative burst10.1038/s41598-021-88897-y2045-2322https://doaj.org/article/5c41aea01663482497d68444e77e884d2021-05-01T00:00:00Zhttps://doi.org/10.1038/s41598-021-88897-yhttps://doaj.org/toc/2045-2322Abstract Microvesicles (MVs) are cell-derived extracellular vesicles that have emerged as markers and mediators of acute lung injury (ALI). One of the most common pathogens in pneumonia-induced ALI is Streptococcus pneumoniae (Spn), but the role of MVs during Spn lung infection is largely unknown. In the first line of defense against Spn and its major virulence factor, pneumolysin (PLY), are the alveolar epithelial cells (AEC). In this study, we aim to characterize MVs shed from PLY-stimulated AEC and explore their contribution in mediating crosstalk with neutrophils. Using in vitro cell and ex vivo (human lung tissue) models, we demonstrated that Spn in a PLY-dependent manner stimulates AEC to release increased numbers of MVs. Spn infected mice also had higher levels of epithelial-derived MVs in their alveolar compartment compared to control. Furthermore, MVs released from PLY-stimulated AEC contain mitochondrial content and can be taken up by neutrophils. These MVs then suppress the ability of neutrophils to produce reactive oxygen species, a critical host-defense mechanism. Taken together, our results demonstrate that AEC in response to pneumococcal PLY release MVs that carry mitochondrial cargo and suggest that these MVs regulate innate immune responses during lung injury.E. LetsiouL. G. Teixeira AlvesD. FatykhovaM. FeltenT. J. MitchellH.C. Müller-RedetzkyA. C. HockeM. WitzenrathNature PortfolioarticleMedicineRScienceQENScientific Reports, Vol 11, Iss 1, Pp 1-13 (2021)
institution DOAJ
collection DOAJ
language EN
topic Medicine
R
Science
Q
spellingShingle Medicine
R
Science
Q
E. Letsiou
L. G. Teixeira Alves
D. Fatykhova
M. Felten
T. J. Mitchell
H.C. Müller-Redetzky
A. C. Hocke
M. Witzenrath
Microvesicles released from pneumolysin-stimulated lung epithelial cells carry mitochondrial cargo and suppress neutrophil oxidative burst
description Abstract Microvesicles (MVs) are cell-derived extracellular vesicles that have emerged as markers and mediators of acute lung injury (ALI). One of the most common pathogens in pneumonia-induced ALI is Streptococcus pneumoniae (Spn), but the role of MVs during Spn lung infection is largely unknown. In the first line of defense against Spn and its major virulence factor, pneumolysin (PLY), are the alveolar epithelial cells (AEC). In this study, we aim to characterize MVs shed from PLY-stimulated AEC and explore their contribution in mediating crosstalk with neutrophils. Using in vitro cell and ex vivo (human lung tissue) models, we demonstrated that Spn in a PLY-dependent manner stimulates AEC to release increased numbers of MVs. Spn infected mice also had higher levels of epithelial-derived MVs in their alveolar compartment compared to control. Furthermore, MVs released from PLY-stimulated AEC contain mitochondrial content and can be taken up by neutrophils. These MVs then suppress the ability of neutrophils to produce reactive oxygen species, a critical host-defense mechanism. Taken together, our results demonstrate that AEC in response to pneumococcal PLY release MVs that carry mitochondrial cargo and suggest that these MVs regulate innate immune responses during lung injury.
format article
author E. Letsiou
L. G. Teixeira Alves
D. Fatykhova
M. Felten
T. J. Mitchell
H.C. Müller-Redetzky
A. C. Hocke
M. Witzenrath
author_facet E. Letsiou
L. G. Teixeira Alves
D. Fatykhova
M. Felten
T. J. Mitchell
H.C. Müller-Redetzky
A. C. Hocke
M. Witzenrath
author_sort E. Letsiou
title Microvesicles released from pneumolysin-stimulated lung epithelial cells carry mitochondrial cargo and suppress neutrophil oxidative burst
title_short Microvesicles released from pneumolysin-stimulated lung epithelial cells carry mitochondrial cargo and suppress neutrophil oxidative burst
title_full Microvesicles released from pneumolysin-stimulated lung epithelial cells carry mitochondrial cargo and suppress neutrophil oxidative burst
title_fullStr Microvesicles released from pneumolysin-stimulated lung epithelial cells carry mitochondrial cargo and suppress neutrophil oxidative burst
title_full_unstemmed Microvesicles released from pneumolysin-stimulated lung epithelial cells carry mitochondrial cargo and suppress neutrophil oxidative burst
title_sort microvesicles released from pneumolysin-stimulated lung epithelial cells carry mitochondrial cargo and suppress neutrophil oxidative burst
publisher Nature Portfolio
publishDate 2021
url https://doaj.org/article/5c41aea01663482497d68444e77e884d
work_keys_str_mv AT eletsiou microvesiclesreleasedfrompneumolysinstimulatedlungepithelialcellscarrymitochondrialcargoandsuppressneutrophiloxidativeburst
AT lgteixeiraalves microvesiclesreleasedfrompneumolysinstimulatedlungepithelialcellscarrymitochondrialcargoandsuppressneutrophiloxidativeburst
AT dfatykhova microvesiclesreleasedfrompneumolysinstimulatedlungepithelialcellscarrymitochondrialcargoandsuppressneutrophiloxidativeburst
AT mfelten microvesiclesreleasedfrompneumolysinstimulatedlungepithelialcellscarrymitochondrialcargoandsuppressneutrophiloxidativeburst
AT tjmitchell microvesiclesreleasedfrompneumolysinstimulatedlungepithelialcellscarrymitochondrialcargoandsuppressneutrophiloxidativeburst
AT hcmullerredetzky microvesiclesreleasedfrompneumolysinstimulatedlungepithelialcellscarrymitochondrialcargoandsuppressneutrophiloxidativeburst
AT achocke microvesiclesreleasedfrompneumolysinstimulatedlungepithelialcellscarrymitochondrialcargoandsuppressneutrophiloxidativeburst
AT mwitzenrath microvesiclesreleasedfrompneumolysinstimulatedlungepithelialcellscarrymitochondrialcargoandsuppressneutrophiloxidativeburst
_version_ 1718386174005870592