Neutrophil-derived microvesicles enter cartilage and protect the joint in inflammatory arthritis

Detalhes bibliográficos
Autor(a) principal: Headland, Sarah E.
Data de Publicação: 2015
Outros Autores: Jones, Hefin R., Norling, Lucy V., Kim, Andrew, Souza, Patricia R., Corsiero, Elisa, Gil, Cristiane D. [UNESP], Nerviani, Alessandra, Dell'accio, Francesco, Pitzalis, Costantino, Oliani, Sonia M. [UNESP], Jan, Lily Y., Perretti, Mauro
Tipo de documento: Artigo
Idioma: eng
Título da fonte: Repositório Institucional da UNESP
Texto Completo: http://dx.doi.org/10.1126/scitranslmed.aac5608
http://hdl.handle.net/11449/172413
Resumo: Microvesicles (MVs) are emerging as a new mechanism of intercellular communication by transferring cellular lipid and protein components to target cells, yet their function in disease is only now being explored. We found that neutrophilderived MVs were increased in concentration in synovial fluid from rheumatoid arthritis patients compared to paired plasma. Synovial MVs overexpressed the proresolving, anti-inflammatory protein annexin A1 (AnxA1). Mice deficient in TMEM16F, a lipid scramblase required for microvesiculation, exhibited exacerbated cartilage damage when subjected to inflammatory arthritis. To determine the function of MVs in inflammatory arthritis, toward the possibility of MV-based therapeutics, we examined the role of immune cell-derived MVs in rodent models and in human primary chondrocytes. In vitro, exogenous neutrophil-derived AnxA1+ MVs activated anabolic gene expression in chondrocytes, leading to extracellular matrix accumulation and cartilage protection through the reduction in stress-adaptive homeostatic mediators interleukin-8 and prostaglandin E2. In vivo, intra-articular injection of AnxA1+ MV lessened cartilage degradation caused by inflammatory arthritis. Arthritic mice receiving adoptive transfer of whole neutrophils displayed abundant MVs within cartilage matrix and revealed that MVs, but not neutrophils themselves, can penetrate cartilage. Mechanistic studies support a model whereby MV-associated AnxA1 interacts with its receptor FPR2 (formyl peptide receptor 2)/ALX, increasing transforming growth factor-p production by chondrocytes, ultimately leading to cartilage protection. We envisage that MVs, either directly or loaded with therapeutics, can be harnessed as a unique therapeutic strategy for protection in diseases associated with cartilage degeneration.
id UNSP_4c09da3abcf7798665d84c7f37b5e59d
oai_identifier_str oai:repositorio.unesp.br:11449/172413
network_acronym_str UNSP
network_name_str Repositório Institucional da UNESP
repository_id_str 2946
spelling Neutrophil-derived microvesicles enter cartilage and protect the joint in inflammatory arthritisMicrovesicles (MVs) are emerging as a new mechanism of intercellular communication by transferring cellular lipid and protein components to target cells, yet their function in disease is only now being explored. We found that neutrophilderived MVs were increased in concentration in synovial fluid from rheumatoid arthritis patients compared to paired plasma. Synovial MVs overexpressed the proresolving, anti-inflammatory protein annexin A1 (AnxA1). Mice deficient in TMEM16F, a lipid scramblase required for microvesiculation, exhibited exacerbated cartilage damage when subjected to inflammatory arthritis. To determine the function of MVs in inflammatory arthritis, toward the possibility of MV-based therapeutics, we examined the role of immune cell-derived MVs in rodent models and in human primary chondrocytes. In vitro, exogenous neutrophil-derived AnxA1+ MVs activated anabolic gene expression in chondrocytes, leading to extracellular matrix accumulation and cartilage protection through the reduction in stress-adaptive homeostatic mediators interleukin-8 and prostaglandin E2. In vivo, intra-articular injection of AnxA1+ MV lessened cartilage degradation caused by inflammatory arthritis. Arthritic mice receiving adoptive transfer of whole neutrophils displayed abundant MVs within cartilage matrix and revealed that MVs, but not neutrophils themselves, can penetrate cartilage. Mechanistic studies support a model whereby MV-associated AnxA1 interacts with its receptor FPR2 (formyl peptide receptor 2)/ALX, increasing transforming growth factor-p production by chondrocytes, ultimately leading to cartilage protection. We envisage that MVs, either directly or loaded with therapeutics, can be harnessed as a unique therapeutic strategy for protection in diseases associated with cartilage degeneration.William Harvey Research Institute Barts and The London School of Medicine Queen Mary University of London, Charterhouse SquareDepartment of Physiology Howard Hughes Medical Institute University of California San FranciscoDepartment of Biology Instituto de Biociências Letras e Ciências Exatas São Paulo State University (UNESP)Department of Rheumatology Barts Health Trust, Bancroft RoadDepartment of Biology Instituto de Biociências Letras e Ciências Exatas São Paulo State University (UNESP)Queen Mary University of LondonSan FranciscoUniversidade Estadual Paulista (Unesp)Barts Health TrustHeadland, Sarah E.Jones, Hefin R.Norling, Lucy V.Kim, AndrewSouza, Patricia R.Corsiero, ElisaGil, Cristiane D. [UNESP]Nerviani, AlessandraDell'accio, FrancescoPitzalis, CostantinoOliani, Sonia M. [UNESP]Jan, Lily Y.Perretti, Mauro2018-12-11T17:00:10Z2018-12-11T17:00:10Z2015-11-25info:eu-repo/semantics/publishedVersioninfo:eu-repo/semantics/articleapplication/pdfhttp://dx.doi.org/10.1126/scitranslmed.aac5608Science Translational Medicine, v. 7, n. 315, 2015.1946-62421946-6234http://hdl.handle.net/11449/17241310.1126/scitranslmed.aac56082-s2.0-849541970242-s2.0-84954197024.pdfScopusreponame:Repositório Institucional da UNESPinstname:Universidade Estadual Paulista (UNESP)instacron:UNESPengScience Translational Medicine9,7009,700info:eu-repo/semantics/openAccess2023-12-25T06:20:30Zoai:repositorio.unesp.br:11449/172413Repositório InstitucionalPUBhttp://repositorio.unesp.br/oai/requestopendoar:29462024-08-05T21:15:29.464206Repositório Institucional da UNESP - Universidade Estadual Paulista (UNESP)false
dc.title.none.fl_str_mv Neutrophil-derived microvesicles enter cartilage and protect the joint in inflammatory arthritis
title Neutrophil-derived microvesicles enter cartilage and protect the joint in inflammatory arthritis
spellingShingle Neutrophil-derived microvesicles enter cartilage and protect the joint in inflammatory arthritis
Headland, Sarah E.
title_short Neutrophil-derived microvesicles enter cartilage and protect the joint in inflammatory arthritis
title_full Neutrophil-derived microvesicles enter cartilage and protect the joint in inflammatory arthritis
title_fullStr Neutrophil-derived microvesicles enter cartilage and protect the joint in inflammatory arthritis
title_full_unstemmed Neutrophil-derived microvesicles enter cartilage and protect the joint in inflammatory arthritis
title_sort Neutrophil-derived microvesicles enter cartilage and protect the joint in inflammatory arthritis
author Headland, Sarah E.
author_facet Headland, Sarah E.
Jones, Hefin R.
Norling, Lucy V.
Kim, Andrew
Souza, Patricia R.
Corsiero, Elisa
Gil, Cristiane D. [UNESP]
Nerviani, Alessandra
Dell'accio, Francesco
Pitzalis, Costantino
Oliani, Sonia M. [UNESP]
Jan, Lily Y.
Perretti, Mauro
author_role author
author2 Jones, Hefin R.
Norling, Lucy V.
Kim, Andrew
Souza, Patricia R.
Corsiero, Elisa
Gil, Cristiane D. [UNESP]
Nerviani, Alessandra
Dell'accio, Francesco
Pitzalis, Costantino
Oliani, Sonia M. [UNESP]
Jan, Lily Y.
Perretti, Mauro
author2_role author
author
author
author
author
author
author
author
author
author
author
author
dc.contributor.none.fl_str_mv Queen Mary University of London
San Francisco
Universidade Estadual Paulista (Unesp)
Barts Health Trust
dc.contributor.author.fl_str_mv Headland, Sarah E.
Jones, Hefin R.
Norling, Lucy V.
Kim, Andrew
Souza, Patricia R.
Corsiero, Elisa
Gil, Cristiane D. [UNESP]
Nerviani, Alessandra
Dell'accio, Francesco
Pitzalis, Costantino
Oliani, Sonia M. [UNESP]
Jan, Lily Y.
Perretti, Mauro
description Microvesicles (MVs) are emerging as a new mechanism of intercellular communication by transferring cellular lipid and protein components to target cells, yet their function in disease is only now being explored. We found that neutrophilderived MVs were increased in concentration in synovial fluid from rheumatoid arthritis patients compared to paired plasma. Synovial MVs overexpressed the proresolving, anti-inflammatory protein annexin A1 (AnxA1). Mice deficient in TMEM16F, a lipid scramblase required for microvesiculation, exhibited exacerbated cartilage damage when subjected to inflammatory arthritis. To determine the function of MVs in inflammatory arthritis, toward the possibility of MV-based therapeutics, we examined the role of immune cell-derived MVs in rodent models and in human primary chondrocytes. In vitro, exogenous neutrophil-derived AnxA1+ MVs activated anabolic gene expression in chondrocytes, leading to extracellular matrix accumulation and cartilage protection through the reduction in stress-adaptive homeostatic mediators interleukin-8 and prostaglandin E2. In vivo, intra-articular injection of AnxA1+ MV lessened cartilage degradation caused by inflammatory arthritis. Arthritic mice receiving adoptive transfer of whole neutrophils displayed abundant MVs within cartilage matrix and revealed that MVs, but not neutrophils themselves, can penetrate cartilage. Mechanistic studies support a model whereby MV-associated AnxA1 interacts with its receptor FPR2 (formyl peptide receptor 2)/ALX, increasing transforming growth factor-p production by chondrocytes, ultimately leading to cartilage protection. We envisage that MVs, either directly or loaded with therapeutics, can be harnessed as a unique therapeutic strategy for protection in diseases associated with cartilage degeneration.
publishDate 2015
dc.date.none.fl_str_mv 2015-11-25
2018-12-11T17:00:10Z
2018-12-11T17:00:10Z
dc.type.status.fl_str_mv info:eu-repo/semantics/publishedVersion
dc.type.driver.fl_str_mv info:eu-repo/semantics/article
format article
status_str publishedVersion
dc.identifier.uri.fl_str_mv http://dx.doi.org/10.1126/scitranslmed.aac5608
Science Translational Medicine, v. 7, n. 315, 2015.
1946-6242
1946-6234
http://hdl.handle.net/11449/172413
10.1126/scitranslmed.aac5608
2-s2.0-84954197024
2-s2.0-84954197024.pdf
url http://dx.doi.org/10.1126/scitranslmed.aac5608
http://hdl.handle.net/11449/172413
identifier_str_mv Science Translational Medicine, v. 7, n. 315, 2015.
1946-6242
1946-6234
10.1126/scitranslmed.aac5608
2-s2.0-84954197024
2-s2.0-84954197024.pdf
dc.language.iso.fl_str_mv eng
language eng
dc.relation.none.fl_str_mv Science Translational Medicine
9,700
9,700
dc.rights.driver.fl_str_mv info:eu-repo/semantics/openAccess
eu_rights_str_mv openAccess
dc.format.none.fl_str_mv application/pdf
dc.source.none.fl_str_mv Scopus
reponame:Repositório Institucional da UNESP
instname:Universidade Estadual Paulista (UNESP)
instacron:UNESP
instname_str Universidade Estadual Paulista (UNESP)
instacron_str UNESP
institution UNESP
reponame_str Repositório Institucional da UNESP
collection Repositório Institucional da UNESP
repository.name.fl_str_mv Repositório Institucional da UNESP - Universidade Estadual Paulista (UNESP)
repository.mail.fl_str_mv
_version_ 1808129301739995136