Cargando…

Platelet Microparticles Enriched in miR-223 Reduce ICAM-1-Dependent Vascular Inflammation in Septic Conditions

In the process of sepsis, activated platelets shed microvesicles containing microRNAs (miRNAs), which can be internalized by distinct recipient cells in circulation, consequently eliciting a potent capability to regulate their cellular functions in different diseases. In the present study, activated...

Descripción completa

Detalles Bibliográficos
Autores principales: Szilágyi, Bernadett, Fejes, Zsolt, Rusznyák, Ágnes, Fenyvesi, Ferenc, Pócsi, Marianna, Halmi, Sándor, Griger, Zoltán, Kunapuli, Satya P., Kappelmayer, János, Nagy, Béla
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8201999/
https://www.ncbi.nlm.nih.gov/pubmed/34135769
http://dx.doi.org/10.3389/fphys.2021.658524
_version_ 1783707896901533696
author Szilágyi, Bernadett
Fejes, Zsolt
Rusznyák, Ágnes
Fenyvesi, Ferenc
Pócsi, Marianna
Halmi, Sándor
Griger, Zoltán
Kunapuli, Satya P.
Kappelmayer, János
Nagy, Béla
author_facet Szilágyi, Bernadett
Fejes, Zsolt
Rusznyák, Ágnes
Fenyvesi, Ferenc
Pócsi, Marianna
Halmi, Sándor
Griger, Zoltán
Kunapuli, Satya P.
Kappelmayer, János
Nagy, Béla
author_sort Szilágyi, Bernadett
collection PubMed
description In the process of sepsis, activated platelets shed microvesicles containing microRNAs (miRNAs), which can be internalized by distinct recipient cells in circulation, consequently eliciting a potent capability to regulate their cellular functions in different diseases. In the present study, activated human platelets transferring miR-223 into endothelial cells via platelet-derived microparticles (PMPs) was investigated in vitro during septic conditions with a proposed mechanism involving in downregulation of the enhanced expression of intercellular adhesion molecule-1 (ICAM-1). The uptake of PMPs encasing miR-223 and the adhesion of peripheral blood mononuclear cells (PBMCs) on human coronary artery endothelial cells (HCAECs) were observed by immunofluorescence microscopy upon co-culture with PMPs isolated from sepsis or control plasma. The expression of miR-223-3p and its gene target ICAM1 in HCAECs were quantified by RT-qPCR and ELISA after the cells were incubated with septic or control PMPs, whose levels were induced with thrombin-receptor activating peptide (TRAP). Leukocyte-depleted platelets (LDPs) from septic patients showed a decreased miR-223 level, while septic plasma and PMPs revealed an elevated miRNA level compared to control samples. Similarly, TRAP-activated LDPs demonstrated a reduced intracellular miR-223 expression, while increased levels in the supernatant and PMP isolates were observed vs. untreated samples. Furthermore, TNF-α alone resulted in decreased miR-223 and elevated ICAM1 levels in HCAECs, while PMPs raised the miRNA level that was associated with downregulated ICAM1 expression at both mRNA and protein levels under TNF-α treatment. Importantly, miR-223 was turned out not to be newly synthesized as shown in unchanged pre-miR-223 level, and mature miR-223 expression was also elevated in the presence of PMPs in HCAECs after transfection with Dicer1 siRNA. In addition, septic PMPs containing miR-223 decreased ICAM1 with a reduction of PBMC binding to HCAECs. In conclusion, septic platelets released PMPs carrying functional miR-223 lower ICAM1 expression in endothelial cells, which may be a protective role against excessive sepsis-induced vascular inflammation.
format Online
Article
Text
id pubmed-8201999
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-82019992021-06-15 Platelet Microparticles Enriched in miR-223 Reduce ICAM-1-Dependent Vascular Inflammation in Septic Conditions Szilágyi, Bernadett Fejes, Zsolt Rusznyák, Ágnes Fenyvesi, Ferenc Pócsi, Marianna Halmi, Sándor Griger, Zoltán Kunapuli, Satya P. Kappelmayer, János Nagy, Béla Front Physiol Physiology In the process of sepsis, activated platelets shed microvesicles containing microRNAs (miRNAs), which can be internalized by distinct recipient cells in circulation, consequently eliciting a potent capability to regulate their cellular functions in different diseases. In the present study, activated human platelets transferring miR-223 into endothelial cells via platelet-derived microparticles (PMPs) was investigated in vitro during septic conditions with a proposed mechanism involving in downregulation of the enhanced expression of intercellular adhesion molecule-1 (ICAM-1). The uptake of PMPs encasing miR-223 and the adhesion of peripheral blood mononuclear cells (PBMCs) on human coronary artery endothelial cells (HCAECs) were observed by immunofluorescence microscopy upon co-culture with PMPs isolated from sepsis or control plasma. The expression of miR-223-3p and its gene target ICAM1 in HCAECs were quantified by RT-qPCR and ELISA after the cells were incubated with septic or control PMPs, whose levels were induced with thrombin-receptor activating peptide (TRAP). Leukocyte-depleted platelets (LDPs) from septic patients showed a decreased miR-223 level, while septic plasma and PMPs revealed an elevated miRNA level compared to control samples. Similarly, TRAP-activated LDPs demonstrated a reduced intracellular miR-223 expression, while increased levels in the supernatant and PMP isolates were observed vs. untreated samples. Furthermore, TNF-α alone resulted in decreased miR-223 and elevated ICAM1 levels in HCAECs, while PMPs raised the miRNA level that was associated with downregulated ICAM1 expression at both mRNA and protein levels under TNF-α treatment. Importantly, miR-223 was turned out not to be newly synthesized as shown in unchanged pre-miR-223 level, and mature miR-223 expression was also elevated in the presence of PMPs in HCAECs after transfection with Dicer1 siRNA. In addition, septic PMPs containing miR-223 decreased ICAM1 with a reduction of PBMC binding to HCAECs. In conclusion, septic platelets released PMPs carrying functional miR-223 lower ICAM1 expression in endothelial cells, which may be a protective role against excessive sepsis-induced vascular inflammation. Frontiers Media S.A. 2021-05-31 /pmc/articles/PMC8201999/ /pubmed/34135769 http://dx.doi.org/10.3389/fphys.2021.658524 Text en Copyright © 2021 Szilágyi, Fejes, Rusznyák, Fenyvesi, Pócsi, Halmi, Griger, Kunapuli, Kappelmayer and Nagy. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Physiology
Szilágyi, Bernadett
Fejes, Zsolt
Rusznyák, Ágnes
Fenyvesi, Ferenc
Pócsi, Marianna
Halmi, Sándor
Griger, Zoltán
Kunapuli, Satya P.
Kappelmayer, János
Nagy, Béla
Platelet Microparticles Enriched in miR-223 Reduce ICAM-1-Dependent Vascular Inflammation in Septic Conditions
title Platelet Microparticles Enriched in miR-223 Reduce ICAM-1-Dependent Vascular Inflammation in Septic Conditions
title_full Platelet Microparticles Enriched in miR-223 Reduce ICAM-1-Dependent Vascular Inflammation in Septic Conditions
title_fullStr Platelet Microparticles Enriched in miR-223 Reduce ICAM-1-Dependent Vascular Inflammation in Septic Conditions
title_full_unstemmed Platelet Microparticles Enriched in miR-223 Reduce ICAM-1-Dependent Vascular Inflammation in Septic Conditions
title_short Platelet Microparticles Enriched in miR-223 Reduce ICAM-1-Dependent Vascular Inflammation in Septic Conditions
title_sort platelet microparticles enriched in mir-223 reduce icam-1-dependent vascular inflammation in septic conditions
topic Physiology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8201999/
https://www.ncbi.nlm.nih.gov/pubmed/34135769
http://dx.doi.org/10.3389/fphys.2021.658524
work_keys_str_mv AT szilagyibernadett plateletmicroparticlesenrichedinmir223reduceicam1dependentvascularinflammationinsepticconditions
AT fejeszsolt plateletmicroparticlesenrichedinmir223reduceicam1dependentvascularinflammationinsepticconditions
AT rusznyakagnes plateletmicroparticlesenrichedinmir223reduceicam1dependentvascularinflammationinsepticconditions
AT fenyvesiferenc plateletmicroparticlesenrichedinmir223reduceicam1dependentvascularinflammationinsepticconditions
AT pocsimarianna plateletmicroparticlesenrichedinmir223reduceicam1dependentvascularinflammationinsepticconditions
AT halmisandor plateletmicroparticlesenrichedinmir223reduceicam1dependentvascularinflammationinsepticconditions
AT grigerzoltan plateletmicroparticlesenrichedinmir223reduceicam1dependentvascularinflammationinsepticconditions
AT kunapulisatyap plateletmicroparticlesenrichedinmir223reduceicam1dependentvascularinflammationinsepticconditions
AT kappelmayerjanos plateletmicroparticlesenrichedinmir223reduceicam1dependentvascularinflammationinsepticconditions
AT nagybela plateletmicroparticlesenrichedinmir223reduceicam1dependentvascularinflammationinsepticconditions