Cargando…

The role of the oncostatin M/OSM receptor β axis in activating dermal microvascular endothelial cells in systemic sclerosis

BACKGROUND: Scleroderma (SSc) is a rare autoimmune disease characterized by vascular impairment and progressive fibrosis of the skin and other organs. Oncostatin M, a member of the IL-6 family, is elevated in SSc serum and was recognized as a significant player in various stages of fibrosis. The goa...

Descripción completa

Detalles Bibliográficos
Autores principales: Marden, G., Wan, Q., Wilks, J., Nevin, K., Feeney, M., Wisniacki, N., Trojanowski, M., Bujor, A., Stawski, L., Trojanowska, M.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7393919/
https://www.ncbi.nlm.nih.gov/pubmed/32736577
http://dx.doi.org/10.1186/s13075-020-02266-0
_version_ 1783565131255382016
author Marden, G.
Wan, Q.
Wilks, J.
Nevin, K.
Feeney, M.
Wisniacki, N.
Trojanowski, M.
Bujor, A.
Stawski, L.
Trojanowska, M.
author_facet Marden, G.
Wan, Q.
Wilks, J.
Nevin, K.
Feeney, M.
Wisniacki, N.
Trojanowski, M.
Bujor, A.
Stawski, L.
Trojanowska, M.
author_sort Marden, G.
collection PubMed
description BACKGROUND: Scleroderma (SSc) is a rare autoimmune disease characterized by vascular impairment and progressive fibrosis of the skin and other organs. Oncostatin M, a member of the IL-6 family, is elevated in SSc serum and was recognized as a significant player in various stages of fibrosis. The goal of this study was to assess the contribution of the OSM/OSMRβ pathway to endothelial cell (EC) injury and activation in SSc. METHODS: IHC and IF were used to assess the distribution of OSM and OSMRβ in SSc (n = 14) and healthy control (n = 7) skin biopsies. Cell culture experiments were performed in human dermal microvascular endothelial cells (HDMECs) and included mRNA and protein analysis, and cell migration and proliferation assays. Ex vivo skin organoid culture was used to evaluate the effect of OSM on perivascular fibrosis. RESULTS: OSMRβ protein was elevated in dermal ECs and in fibroblasts of SSc patients. Treatments of HDMECs with OSM or IL-6+sIL-6R have demonstrated that both cytokines similarly stimulated proinflammatory genes and genes related to endothelial to mesenchymal transition (EndMT). OSM was more effective than IL-6+sIL-6R in inducing cell migration, while both treatments similarly induced cell proliferation. The effects of OSM were mediated via OSMRβ and STAT3, while the LIFR did not contribute to these responses. Both OSM and IL-6+sIL-6R induced profibrotic gene expression in HDMECs, as well as expansion of the perivascular PDGFRβ(+) cells in the ex vivo human skin culture system. Additional studies in HDMECs showed that siRNA-mediated downregulation of FLI1 and its close homolog ERG resulted in increased expression of OSMRβ in HDMECs. CONCLUSIONS: This work provides new insights into the role of the OSM/OSMRβ axis in activation/injury of dermal ECs and supports the involvement of this pathway in SSc vascular disease.
format Online
Article
Text
id pubmed-7393919
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-73939192020-08-04 The role of the oncostatin M/OSM receptor β axis in activating dermal microvascular endothelial cells in systemic sclerosis Marden, G. Wan, Q. Wilks, J. Nevin, K. Feeney, M. Wisniacki, N. Trojanowski, M. Bujor, A. Stawski, L. Trojanowska, M. Arthritis Res Ther Research Article BACKGROUND: Scleroderma (SSc) is a rare autoimmune disease characterized by vascular impairment and progressive fibrosis of the skin and other organs. Oncostatin M, a member of the IL-6 family, is elevated in SSc serum and was recognized as a significant player in various stages of fibrosis. The goal of this study was to assess the contribution of the OSM/OSMRβ pathway to endothelial cell (EC) injury and activation in SSc. METHODS: IHC and IF were used to assess the distribution of OSM and OSMRβ in SSc (n = 14) and healthy control (n = 7) skin biopsies. Cell culture experiments were performed in human dermal microvascular endothelial cells (HDMECs) and included mRNA and protein analysis, and cell migration and proliferation assays. Ex vivo skin organoid culture was used to evaluate the effect of OSM on perivascular fibrosis. RESULTS: OSMRβ protein was elevated in dermal ECs and in fibroblasts of SSc patients. Treatments of HDMECs with OSM or IL-6+sIL-6R have demonstrated that both cytokines similarly stimulated proinflammatory genes and genes related to endothelial to mesenchymal transition (EndMT). OSM was more effective than IL-6+sIL-6R in inducing cell migration, while both treatments similarly induced cell proliferation. The effects of OSM were mediated via OSMRβ and STAT3, while the LIFR did not contribute to these responses. Both OSM and IL-6+sIL-6R induced profibrotic gene expression in HDMECs, as well as expansion of the perivascular PDGFRβ(+) cells in the ex vivo human skin culture system. Additional studies in HDMECs showed that siRNA-mediated downregulation of FLI1 and its close homolog ERG resulted in increased expression of OSMRβ in HDMECs. CONCLUSIONS: This work provides new insights into the role of the OSM/OSMRβ axis in activation/injury of dermal ECs and supports the involvement of this pathway in SSc vascular disease. BioMed Central 2020-07-31 2020 /pmc/articles/PMC7393919/ /pubmed/32736577 http://dx.doi.org/10.1186/s13075-020-02266-0 Text en © The Author(s) 2020 Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Research Article
Marden, G.
Wan, Q.
Wilks, J.
Nevin, K.
Feeney, M.
Wisniacki, N.
Trojanowski, M.
Bujor, A.
Stawski, L.
Trojanowska, M.
The role of the oncostatin M/OSM receptor β axis in activating dermal microvascular endothelial cells in systemic sclerosis
title The role of the oncostatin M/OSM receptor β axis in activating dermal microvascular endothelial cells in systemic sclerosis
title_full The role of the oncostatin M/OSM receptor β axis in activating dermal microvascular endothelial cells in systemic sclerosis
title_fullStr The role of the oncostatin M/OSM receptor β axis in activating dermal microvascular endothelial cells in systemic sclerosis
title_full_unstemmed The role of the oncostatin M/OSM receptor β axis in activating dermal microvascular endothelial cells in systemic sclerosis
title_short The role of the oncostatin M/OSM receptor β axis in activating dermal microvascular endothelial cells in systemic sclerosis
title_sort role of the oncostatin m/osm receptor β axis in activating dermal microvascular endothelial cells in systemic sclerosis
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7393919/
https://www.ncbi.nlm.nih.gov/pubmed/32736577
http://dx.doi.org/10.1186/s13075-020-02266-0
work_keys_str_mv AT mardeng theroleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis
AT wanq theroleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis
AT wilksj theroleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis
AT nevink theroleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis
AT feeneym theroleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis
AT wisniackin theroleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis
AT trojanowskim theroleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis
AT bujora theroleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis
AT stawskil theroleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis
AT trojanowskam theroleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis
AT mardeng roleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis
AT wanq roleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis
AT wilksj roleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis
AT nevink roleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis
AT feeneym roleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis
AT wisniackin roleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis
AT trojanowskim roleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis
AT bujora roleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis
AT stawskil roleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis
AT trojanowskam roleoftheoncostatinmosmreceptorbaxisinactivatingdermalmicrovascularendothelialcellsinsystemicsclerosis