Cargando…

Autoantibody against β(1)-adrenoceptor promotes the differentiation of natural regulatory T cells from activated CD4(+) T cells by up-regulating AMPK-mediated fatty acid oxidation

Therapeutic adoptive transfer of natural regulatory T cells (nTreg, CD4(+) CD25(+) Foxp3(+) T cells) or in vivo selective expansion of nTreg cells has been demonstrated to improve the cardiac function in various cardiovascular disease models. The differentiation of nTreg cells is mediated by catecho...

Descripción completa

Detalles Bibliográficos
Autores principales: Xu, Wenli, Wu, Ye, Wang, Li, Bai, Yan, Du, Yunhui, Li, Yang, Cao, Ning, Zhao, Yuhui, Zhang, Youyi, Liu, Huirong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2019
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6377640/
https://www.ncbi.nlm.nih.gov/pubmed/30770790
http://dx.doi.org/10.1038/s41419-018-1209-2
_version_ 1783395778670100480
author Xu, Wenli
Wu, Ye
Wang, Li
Bai, Yan
Du, Yunhui
Li, Yang
Cao, Ning
Zhao, Yuhui
Zhang, Youyi
Liu, Huirong
author_facet Xu, Wenli
Wu, Ye
Wang, Li
Bai, Yan
Du, Yunhui
Li, Yang
Cao, Ning
Zhao, Yuhui
Zhang, Youyi
Liu, Huirong
author_sort Xu, Wenli
collection PubMed
description Therapeutic adoptive transfer of natural regulatory T cells (nTreg, CD4(+) CD25(+) Foxp3(+) T cells) or in vivo selective expansion of nTreg cells has been demonstrated to improve the cardiac function in various cardiovascular disease models. The differentiation of nTreg cells is mediated by catecholamines via β(1)-adrenergic receptor (β(1)-AR) activation. Autoantibody against β(1)-adrenoceptor (β(1)-AA) as a β(1)-AR agonist is closely associated with the occurrence and deterioration of cardiac dysfunction. However, whether β(1)-AA has any impact on nTreg cells has not been reported. The aim of the present study was intended to assess the potential impact of β(1)-AA on nTreg cell differentiation and explore the underlying mechanism. It was found that the expression of multiple proteins involved in nTreg cell differentiation, immunosuppressive function, and migration was up-regulated in mice after β(1)-AA administration, suggesting that β(1)-AA may promote nTreg cell activation. In vitro, β(1)-AA promoted nTreg cell differentiation by up-regulating mitochondrial fatty acid oxidation (FAO) in activated CD4(+) T cells via AMP-activated protein kinase (AMPK) activation and mitochondrial membrane potential reduction. In addition, the AMPK agonist facilitated β(1)-AA-mediated FAO and nTreg cell differentiation. To further confirm the role of AMPK in β(1)-AA-mediated nTreg cell differentiation, β(1)-AA was acted on the CD4(+) T cells isolated from AMPK-deficient (AMPK(−/−)) mice. The result showed that the effect of β(1)-AA on nTreg cell differentiation was attenuated markedly after AMPK knockout. In conclusion, AMPK-mediated metabolic regulation targeting for nTreg cell restoration may be a promising therapeutic target for β(1)-AA-positive patients with cardiac dysfunction.
format Online
Article
Text
id pubmed-6377640
institution National Center for Biotechnology Information
language English
publishDate 2019
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-63776402019-02-19 Autoantibody against β(1)-adrenoceptor promotes the differentiation of natural regulatory T cells from activated CD4(+) T cells by up-regulating AMPK-mediated fatty acid oxidation Xu, Wenli Wu, Ye Wang, Li Bai, Yan Du, Yunhui Li, Yang Cao, Ning Zhao, Yuhui Zhang, Youyi Liu, Huirong Cell Death Dis Article Therapeutic adoptive transfer of natural regulatory T cells (nTreg, CD4(+) CD25(+) Foxp3(+) T cells) or in vivo selective expansion of nTreg cells has been demonstrated to improve the cardiac function in various cardiovascular disease models. The differentiation of nTreg cells is mediated by catecholamines via β(1)-adrenergic receptor (β(1)-AR) activation. Autoantibody against β(1)-adrenoceptor (β(1)-AA) as a β(1)-AR agonist is closely associated with the occurrence and deterioration of cardiac dysfunction. However, whether β(1)-AA has any impact on nTreg cells has not been reported. The aim of the present study was intended to assess the potential impact of β(1)-AA on nTreg cell differentiation and explore the underlying mechanism. It was found that the expression of multiple proteins involved in nTreg cell differentiation, immunosuppressive function, and migration was up-regulated in mice after β(1)-AA administration, suggesting that β(1)-AA may promote nTreg cell activation. In vitro, β(1)-AA promoted nTreg cell differentiation by up-regulating mitochondrial fatty acid oxidation (FAO) in activated CD4(+) T cells via AMP-activated protein kinase (AMPK) activation and mitochondrial membrane potential reduction. In addition, the AMPK agonist facilitated β(1)-AA-mediated FAO and nTreg cell differentiation. To further confirm the role of AMPK in β(1)-AA-mediated nTreg cell differentiation, β(1)-AA was acted on the CD4(+) T cells isolated from AMPK-deficient (AMPK(−/−)) mice. The result showed that the effect of β(1)-AA on nTreg cell differentiation was attenuated markedly after AMPK knockout. In conclusion, AMPK-mediated metabolic regulation targeting for nTreg cell restoration may be a promising therapeutic target for β(1)-AA-positive patients with cardiac dysfunction. Nature Publishing Group UK 2019-02-15 /pmc/articles/PMC6377640/ /pubmed/30770790 http://dx.doi.org/10.1038/s41419-018-1209-2 Text en © The Author(s) 2019 Open Access This 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 license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license 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 license, visit http://creativecommons.org/licenses/by/4.0/.
spellingShingle Article
Xu, Wenli
Wu, Ye
Wang, Li
Bai, Yan
Du, Yunhui
Li, Yang
Cao, Ning
Zhao, Yuhui
Zhang, Youyi
Liu, Huirong
Autoantibody against β(1)-adrenoceptor promotes the differentiation of natural regulatory T cells from activated CD4(+) T cells by up-regulating AMPK-mediated fatty acid oxidation
title Autoantibody against β(1)-adrenoceptor promotes the differentiation of natural regulatory T cells from activated CD4(+) T cells by up-regulating AMPK-mediated fatty acid oxidation
title_full Autoantibody against β(1)-adrenoceptor promotes the differentiation of natural regulatory T cells from activated CD4(+) T cells by up-regulating AMPK-mediated fatty acid oxidation
title_fullStr Autoantibody against β(1)-adrenoceptor promotes the differentiation of natural regulatory T cells from activated CD4(+) T cells by up-regulating AMPK-mediated fatty acid oxidation
title_full_unstemmed Autoantibody against β(1)-adrenoceptor promotes the differentiation of natural regulatory T cells from activated CD4(+) T cells by up-regulating AMPK-mediated fatty acid oxidation
title_short Autoantibody against β(1)-adrenoceptor promotes the differentiation of natural regulatory T cells from activated CD4(+) T cells by up-regulating AMPK-mediated fatty acid oxidation
title_sort autoantibody against β(1)-adrenoceptor promotes the differentiation of natural regulatory t cells from activated cd4(+) t cells by up-regulating ampk-mediated fatty acid oxidation
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6377640/
https://www.ncbi.nlm.nih.gov/pubmed/30770790
http://dx.doi.org/10.1038/s41419-018-1209-2
work_keys_str_mv AT xuwenli autoantibodyagainstb1adrenoceptorpromotesthedifferentiationofnaturalregulatorytcellsfromactivatedcd4tcellsbyupregulatingampkmediatedfattyacidoxidation
AT wuye autoantibodyagainstb1adrenoceptorpromotesthedifferentiationofnaturalregulatorytcellsfromactivatedcd4tcellsbyupregulatingampkmediatedfattyacidoxidation
AT wangli autoantibodyagainstb1adrenoceptorpromotesthedifferentiationofnaturalregulatorytcellsfromactivatedcd4tcellsbyupregulatingampkmediatedfattyacidoxidation
AT baiyan autoantibodyagainstb1adrenoceptorpromotesthedifferentiationofnaturalregulatorytcellsfromactivatedcd4tcellsbyupregulatingampkmediatedfattyacidoxidation
AT duyunhui autoantibodyagainstb1adrenoceptorpromotesthedifferentiationofnaturalregulatorytcellsfromactivatedcd4tcellsbyupregulatingampkmediatedfattyacidoxidation
AT liyang autoantibodyagainstb1adrenoceptorpromotesthedifferentiationofnaturalregulatorytcellsfromactivatedcd4tcellsbyupregulatingampkmediatedfattyacidoxidation
AT caoning autoantibodyagainstb1adrenoceptorpromotesthedifferentiationofnaturalregulatorytcellsfromactivatedcd4tcellsbyupregulatingampkmediatedfattyacidoxidation
AT zhaoyuhui autoantibodyagainstb1adrenoceptorpromotesthedifferentiationofnaturalregulatorytcellsfromactivatedcd4tcellsbyupregulatingampkmediatedfattyacidoxidation
AT zhangyouyi autoantibodyagainstb1adrenoceptorpromotesthedifferentiationofnaturalregulatorytcellsfromactivatedcd4tcellsbyupregulatingampkmediatedfattyacidoxidation
AT liuhuirong autoantibodyagainstb1adrenoceptorpromotesthedifferentiationofnaturalregulatorytcellsfromactivatedcd4tcellsbyupregulatingampkmediatedfattyacidoxidation