Cargando…

TRAIL attenuates RANKL-mediated osteoblastic signalling in vascular cell mono-culture and co-culture models

BACKGROUND AND OBJECTIVES: Vascular calcification (VC) is a major risk factor for elevated cardiovascular morbidity/mortality. Underlying this process is osteoblastic signalling within the vessel wall involving complex and interlinked roles for receptor-activator of nuclear factor-κB ligand (RANKL),...

Descripción completa

Detalles Bibliográficos
Autores principales: Harper, Emma, Rochfort, Keith D., Forde, Hannah, Davenport, Colin, Smith, Diarmuid, Cummins, Philip M.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Public Library of Science 2017
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5690591/
https://www.ncbi.nlm.nih.gov/pubmed/29145460
http://dx.doi.org/10.1371/journal.pone.0188192
_version_ 1783279632696475648
author Harper, Emma
Rochfort, Keith D.
Forde, Hannah
Davenport, Colin
Smith, Diarmuid
Cummins, Philip M.
author_facet Harper, Emma
Rochfort, Keith D.
Forde, Hannah
Davenport, Colin
Smith, Diarmuid
Cummins, Philip M.
author_sort Harper, Emma
collection PubMed
description BACKGROUND AND OBJECTIVES: Vascular calcification (VC) is a major risk factor for elevated cardiovascular morbidity/mortality. Underlying this process is osteoblastic signalling within the vessel wall involving complex and interlinked roles for receptor-activator of nuclear factor-κB ligand (RANKL), osteoprotegerin (OPG), and tumour necrosis factor-related apoptosis-inducing ligand (TRAIL). RANKL promotes vascular cell osteoblastic differentiation, whilst OPG acts as a neutralizing decoy receptor for RANKL (and TRAIL). With respect to TRAIL, much recent evidence points to a vasoprotective role for this ligand, albeit via unknown mechanisms. In order to shed more light on TRAILs vasoprotective role therefore, we employed in vitro cell models to test the hypothesis that TRAIL can counteract the RANKL-mediated signalling that occurs between the vascular cells that comprise the vessel wall. METHODS AND RESULTS: Human aortic endothelial and smooth muscle cell mono-cultures (HAECs, HASMCs) were treated with RANKL (0–25 ng/mL ± 5 ng/mL TRAIL) for 72 hr. Furthermore, to better recapitulate the paracrine signalling that exists between endothelial and smooth muscle cells within the vessel wall, non-contact transwell HAEC:HASMC co-cultures were also employed and involved RANKL treatment of HAECs (±TRAIL), subsequently followed by analysis of pro-calcific markers in the underlying subluminal HASMCs. RANKL elicited robust osteoblastic signalling across both mono- and co-culture models (e.g. increased BMP-2, alkaline phosphatase/ALP, Runx2, and Sox9, in conjunction with decreased OPG). Importantly, several RANKL actions (e.g. increased BMP-2 release from mono-cultured HAECs or increased ALP/Sox9 levels in co-cultured HASMCs) could be strongly blocked by co-incubation with TRAIL. In summary, this paper clearly demonstrates that RANKL can elicit pro-osteoblastic signalling in HAECs and HASMCs both directly and across paracrine signalling axes. Moreover, within these contexts we present clear evidence that TRAIL can block several key signalling actions of RANKL in vascular cells, providing further evidence of its vasoprotective potential.
format Online
Article
Text
id pubmed-5690591
institution National Center for Biotechnology Information
language English
publishDate 2017
publisher Public Library of Science
record_format MEDLINE/PubMed
spelling pubmed-56905912017-11-30 TRAIL attenuates RANKL-mediated osteoblastic signalling in vascular cell mono-culture and co-culture models Harper, Emma Rochfort, Keith D. Forde, Hannah Davenport, Colin Smith, Diarmuid Cummins, Philip M. PLoS One Research Article BACKGROUND AND OBJECTIVES: Vascular calcification (VC) is a major risk factor for elevated cardiovascular morbidity/mortality. Underlying this process is osteoblastic signalling within the vessel wall involving complex and interlinked roles for receptor-activator of nuclear factor-κB ligand (RANKL), osteoprotegerin (OPG), and tumour necrosis factor-related apoptosis-inducing ligand (TRAIL). RANKL promotes vascular cell osteoblastic differentiation, whilst OPG acts as a neutralizing decoy receptor for RANKL (and TRAIL). With respect to TRAIL, much recent evidence points to a vasoprotective role for this ligand, albeit via unknown mechanisms. In order to shed more light on TRAILs vasoprotective role therefore, we employed in vitro cell models to test the hypothesis that TRAIL can counteract the RANKL-mediated signalling that occurs between the vascular cells that comprise the vessel wall. METHODS AND RESULTS: Human aortic endothelial and smooth muscle cell mono-cultures (HAECs, HASMCs) were treated with RANKL (0–25 ng/mL ± 5 ng/mL TRAIL) for 72 hr. Furthermore, to better recapitulate the paracrine signalling that exists between endothelial and smooth muscle cells within the vessel wall, non-contact transwell HAEC:HASMC co-cultures were also employed and involved RANKL treatment of HAECs (±TRAIL), subsequently followed by analysis of pro-calcific markers in the underlying subluminal HASMCs. RANKL elicited robust osteoblastic signalling across both mono- and co-culture models (e.g. increased BMP-2, alkaline phosphatase/ALP, Runx2, and Sox9, in conjunction with decreased OPG). Importantly, several RANKL actions (e.g. increased BMP-2 release from mono-cultured HAECs or increased ALP/Sox9 levels in co-cultured HASMCs) could be strongly blocked by co-incubation with TRAIL. In summary, this paper clearly demonstrates that RANKL can elicit pro-osteoblastic signalling in HAECs and HASMCs both directly and across paracrine signalling axes. Moreover, within these contexts we present clear evidence that TRAIL can block several key signalling actions of RANKL in vascular cells, providing further evidence of its vasoprotective potential. Public Library of Science 2017-11-16 /pmc/articles/PMC5690591/ /pubmed/29145460 http://dx.doi.org/10.1371/journal.pone.0188192 Text en © 2017 Harper et al http://creativecommons.org/licenses/by/4.0/ This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0/) , which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
spellingShingle Research Article
Harper, Emma
Rochfort, Keith D.
Forde, Hannah
Davenport, Colin
Smith, Diarmuid
Cummins, Philip M.
TRAIL attenuates RANKL-mediated osteoblastic signalling in vascular cell mono-culture and co-culture models
title TRAIL attenuates RANKL-mediated osteoblastic signalling in vascular cell mono-culture and co-culture models
title_full TRAIL attenuates RANKL-mediated osteoblastic signalling in vascular cell mono-culture and co-culture models
title_fullStr TRAIL attenuates RANKL-mediated osteoblastic signalling in vascular cell mono-culture and co-culture models
title_full_unstemmed TRAIL attenuates RANKL-mediated osteoblastic signalling in vascular cell mono-culture and co-culture models
title_short TRAIL attenuates RANKL-mediated osteoblastic signalling in vascular cell mono-culture and co-culture models
title_sort trail attenuates rankl-mediated osteoblastic signalling in vascular cell mono-culture and co-culture models
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC5690591/
https://www.ncbi.nlm.nih.gov/pubmed/29145460
http://dx.doi.org/10.1371/journal.pone.0188192
work_keys_str_mv AT harperemma trailattenuatesranklmediatedosteoblasticsignallinginvascularcellmonocultureandcoculturemodels
AT rochfortkeithd trailattenuatesranklmediatedosteoblasticsignallinginvascularcellmonocultureandcoculturemodels
AT fordehannah trailattenuatesranklmediatedosteoblasticsignallinginvascularcellmonocultureandcoculturemodels
AT davenportcolin trailattenuatesranklmediatedosteoblasticsignallinginvascularcellmonocultureandcoculturemodels
AT smithdiarmuid trailattenuatesranklmediatedosteoblasticsignallinginvascularcellmonocultureandcoculturemodels
AT cumminsphilipm trailattenuatesranklmediatedosteoblasticsignallinginvascularcellmonocultureandcoculturemodels