Cargando…

Increase of aerobic glycolysis mediated by activated T helper cells drives synovial fibroblasts towards an inflammatory phenotype: new targets for therapy?

BACKGROUND: A dysregulated glucose metabolism in synovial fibroblasts (SF) has been associated with their aggressive phenotype in rheumatoid arthritis (RA). Even though T helper (Th) cells are key effector cells in the propagation and exacerbation of synovitis in RA, little is known about their infl...

Descripción completa

Detalles Bibliográficos
Autores principales: Kvacskay, Peter, Yao, Nina, Schnotz, Jürgen-Heinz, Scarpone, Roberta, Carvalho, Rui de Albuquerque, Klika, Karel D., Merkt, Wolfgang, Tretter, Theresa, Lorenz, Hanns-Martin, Tykocinski, Lars-Oliver
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7883459/
https://www.ncbi.nlm.nih.gov/pubmed/33588937
http://dx.doi.org/10.1186/s13075-021-02437-7
_version_ 1783651215815475200
author Kvacskay, Peter
Yao, Nina
Schnotz, Jürgen-Heinz
Scarpone, Roberta
Carvalho, Rui de Albuquerque
Klika, Karel D.
Merkt, Wolfgang
Tretter, Theresa
Lorenz, Hanns-Martin
Tykocinski, Lars-Oliver
author_facet Kvacskay, Peter
Yao, Nina
Schnotz, Jürgen-Heinz
Scarpone, Roberta
Carvalho, Rui de Albuquerque
Klika, Karel D.
Merkt, Wolfgang
Tretter, Theresa
Lorenz, Hanns-Martin
Tykocinski, Lars-Oliver
author_sort Kvacskay, Peter
collection PubMed
description BACKGROUND: A dysregulated glucose metabolism in synovial fibroblasts (SF) has been associated with their aggressive phenotype in rheumatoid arthritis (RA). Even though T helper (Th) cells are key effector cells in the propagation and exacerbation of synovitis in RA, little is known about their influence on the metabolism of SF. Thus, this study investigates the effect of Th cells on the glucose metabolism and phenotype of SF and how this is influenced by the blockade of cytokines, janus kinases (JAKs) and glycolysis. METHODS: SF from patients with RA or osteoarthritis (OA) were cultured in the presence of a stable glucose isotopomer ([U-(13)C]-glucose) and stimulated with the conditioned media of activated Th cells (ThCM). Glucose consumption and lactate production were measured by proton nuclear magnetic resonance ((1)H NMR) spectroscopy. Cytokine secretion was quantified by ELISA. The expression of glycolytic enzymes was analysed by PCR, western blot and immunofluorescence. JAKs were blocked using either baricitinib or tofacitinib and glycolysis by using either 3-bromopyruvate or FX11. RESULTS: Quiescent RASF produced significantly higher levels of lactate, interleukin (IL)-6 and matrix metalloproteinase (MMP) 3 than OASF. Stimulation by ThCM clearly changed the metabolic profile of both RASF and OASF by inducing a shift towards aerobic glycolysis with strongly increased lactate production together with a rise in IL-6 and MMP3 secretion. Interestingly, chronic stimulation of OASF by ThCM triggered an inflammatory phenotype with significantly increased glycolytic activity compared to unstimulated, singly stimulated or re-stimulated OASF. Finally, in contrast to cytokine-neutralizing biologics, inhibition of JAKs or glycolytic enzymes both significantly reduced lactate production and cytokine secretion by Th cell-stimulated SF. CONCLUSIONS: Soluble mediators released by Th cells drive SF towards a glycolytic and pro-inflammatory phenotype. Targeting of JAKs or glycolytic enzymes both potently modulate SF’s glucose metabolism and decrease the release of IL-6 and MMP3. Thus, manipulation of glycolytic pathways could represent a new therapeutic strategy to decrease the pro-inflammatory phenotype of SF. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13075-021-02437-7.
format Online
Article
Text
id pubmed-7883459
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-78834592021-02-17 Increase of aerobic glycolysis mediated by activated T helper cells drives synovial fibroblasts towards an inflammatory phenotype: new targets for therapy? Kvacskay, Peter Yao, Nina Schnotz, Jürgen-Heinz Scarpone, Roberta Carvalho, Rui de Albuquerque Klika, Karel D. Merkt, Wolfgang Tretter, Theresa Lorenz, Hanns-Martin Tykocinski, Lars-Oliver Arthritis Res Ther Research Article BACKGROUND: A dysregulated glucose metabolism in synovial fibroblasts (SF) has been associated with their aggressive phenotype in rheumatoid arthritis (RA). Even though T helper (Th) cells are key effector cells in the propagation and exacerbation of synovitis in RA, little is known about their influence on the metabolism of SF. Thus, this study investigates the effect of Th cells on the glucose metabolism and phenotype of SF and how this is influenced by the blockade of cytokines, janus kinases (JAKs) and glycolysis. METHODS: SF from patients with RA or osteoarthritis (OA) were cultured in the presence of a stable glucose isotopomer ([U-(13)C]-glucose) and stimulated with the conditioned media of activated Th cells (ThCM). Glucose consumption and lactate production were measured by proton nuclear magnetic resonance ((1)H NMR) spectroscopy. Cytokine secretion was quantified by ELISA. The expression of glycolytic enzymes was analysed by PCR, western blot and immunofluorescence. JAKs were blocked using either baricitinib or tofacitinib and glycolysis by using either 3-bromopyruvate or FX11. RESULTS: Quiescent RASF produced significantly higher levels of lactate, interleukin (IL)-6 and matrix metalloproteinase (MMP) 3 than OASF. Stimulation by ThCM clearly changed the metabolic profile of both RASF and OASF by inducing a shift towards aerobic glycolysis with strongly increased lactate production together with a rise in IL-6 and MMP3 secretion. Interestingly, chronic stimulation of OASF by ThCM triggered an inflammatory phenotype with significantly increased glycolytic activity compared to unstimulated, singly stimulated or re-stimulated OASF. Finally, in contrast to cytokine-neutralizing biologics, inhibition of JAKs or glycolytic enzymes both significantly reduced lactate production and cytokine secretion by Th cell-stimulated SF. CONCLUSIONS: Soluble mediators released by Th cells drive SF towards a glycolytic and pro-inflammatory phenotype. Targeting of JAKs or glycolytic enzymes both potently modulate SF’s glucose metabolism and decrease the release of IL-6 and MMP3. Thus, manipulation of glycolytic pathways could represent a new therapeutic strategy to decrease the pro-inflammatory phenotype of SF. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13075-021-02437-7. BioMed Central 2021-02-15 2021 /pmc/articles/PMC7883459/ /pubmed/33588937 http://dx.doi.org/10.1186/s13075-021-02437-7 Text en © The Author(s) 2021 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
Kvacskay, Peter
Yao, Nina
Schnotz, Jürgen-Heinz
Scarpone, Roberta
Carvalho, Rui de Albuquerque
Klika, Karel D.
Merkt, Wolfgang
Tretter, Theresa
Lorenz, Hanns-Martin
Tykocinski, Lars-Oliver
Increase of aerobic glycolysis mediated by activated T helper cells drives synovial fibroblasts towards an inflammatory phenotype: new targets for therapy?
title Increase of aerobic glycolysis mediated by activated T helper cells drives synovial fibroblasts towards an inflammatory phenotype: new targets for therapy?
title_full Increase of aerobic glycolysis mediated by activated T helper cells drives synovial fibroblasts towards an inflammatory phenotype: new targets for therapy?
title_fullStr Increase of aerobic glycolysis mediated by activated T helper cells drives synovial fibroblasts towards an inflammatory phenotype: new targets for therapy?
title_full_unstemmed Increase of aerobic glycolysis mediated by activated T helper cells drives synovial fibroblasts towards an inflammatory phenotype: new targets for therapy?
title_short Increase of aerobic glycolysis mediated by activated T helper cells drives synovial fibroblasts towards an inflammatory phenotype: new targets for therapy?
title_sort increase of aerobic glycolysis mediated by activated t helper cells drives synovial fibroblasts towards an inflammatory phenotype: new targets for therapy?
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7883459/
https://www.ncbi.nlm.nih.gov/pubmed/33588937
http://dx.doi.org/10.1186/s13075-021-02437-7
work_keys_str_mv AT kvacskaypeter increaseofaerobicglycolysismediatedbyactivatedthelpercellsdrivessynovialfibroblaststowardsaninflammatoryphenotypenewtargetsfortherapy
AT yaonina increaseofaerobicglycolysismediatedbyactivatedthelpercellsdrivessynovialfibroblaststowardsaninflammatoryphenotypenewtargetsfortherapy
AT schnotzjurgenheinz increaseofaerobicglycolysismediatedbyactivatedthelpercellsdrivessynovialfibroblaststowardsaninflammatoryphenotypenewtargetsfortherapy
AT scarponeroberta increaseofaerobicglycolysismediatedbyactivatedthelpercellsdrivessynovialfibroblaststowardsaninflammatoryphenotypenewtargetsfortherapy
AT carvalhoruidealbuquerque increaseofaerobicglycolysismediatedbyactivatedthelpercellsdrivessynovialfibroblaststowardsaninflammatoryphenotypenewtargetsfortherapy
AT klikakareld increaseofaerobicglycolysismediatedbyactivatedthelpercellsdrivessynovialfibroblaststowardsaninflammatoryphenotypenewtargetsfortherapy
AT merktwolfgang increaseofaerobicglycolysismediatedbyactivatedthelpercellsdrivessynovialfibroblaststowardsaninflammatoryphenotypenewtargetsfortherapy
AT trettertheresa increaseofaerobicglycolysismediatedbyactivatedthelpercellsdrivessynovialfibroblaststowardsaninflammatoryphenotypenewtargetsfortherapy
AT lorenzhannsmartin increaseofaerobicglycolysismediatedbyactivatedthelpercellsdrivessynovialfibroblaststowardsaninflammatoryphenotypenewtargetsfortherapy
AT tykocinskilarsoliver increaseofaerobicglycolysismediatedbyactivatedthelpercellsdrivessynovialfibroblaststowardsaninflammatoryphenotypenewtargetsfortherapy