Cargando…

A comprehensive multiomics approach reveals that high levels of sphingolipids in cardiac cachexia adipose tissue are associated with inflammatory and fibrotic changes

Cardiac cachexia is a deadly consequence of advanced heart failure that is characterised by the dysregulation of adipose tissue homeostasis. Once cachexia occurs with heart failure, it prevents the normal treatment of heart failure and increases the risk of death. Targeting adipose tissue is an impo...

Descripción completa

Detalles Bibliográficos
Autores principales: Qu, Yiwei, Wang, Yong, Wu, Tao, Liu, Xue, Wang, Huaizhe, Ma, Dufang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10691093/
https://www.ncbi.nlm.nih.gov/pubmed/38041133
http://dx.doi.org/10.1186/s12944-023-01967-0
_version_ 1785152668397731840
author Qu, Yiwei
Wang, Yong
Wu, Tao
Liu, Xue
Wang, Huaizhe
Ma, Dufang
author_facet Qu, Yiwei
Wang, Yong
Wu, Tao
Liu, Xue
Wang, Huaizhe
Ma, Dufang
author_sort Qu, Yiwei
collection PubMed
description Cardiac cachexia is a deadly consequence of advanced heart failure that is characterised by the dysregulation of adipose tissue homeostasis. Once cachexia occurs with heart failure, it prevents the normal treatment of heart failure and increases the risk of death. Targeting adipose tissue is an important approach to treating cardiac cachexia, but the pathogenic mechanisms are still unknown, and there are no effective therapies available. Transcriptomics, metabolomics, and lipidomics were used to examine the underlying mechanisms of cardiac cachexia. Transcriptomics investigation of cardiac cachexia adipose tissue revealed that genes involved in fibrosis and monocyte/macrophage migration were increased and strongly interacted. The ECM-receptor interaction pathway was primarily enriched, as shown by KEGG enrichment analysis. In addition, gene set enrichment analysis revealed that monocyte chemotaxis/macrophage migration and fibrosis gene sets were upregulated in cardiac cachexia. Metabolomics enrichment analysis demonstrated that the sphingolipid signalling pathway is important for adipose tissue remodelling in cardiac cachexia. Lipidomics analysis showed that the adipose tissue of rats with cardiac cachexia had higher levels of sphingolipids, including Cer and S1P. Moreover, combined multiomics analysis suggested that the sphingolipid metabolic pathway was associated with inflammatory-fibrotic changes in adipose tissue. Finally, the key indicators were validated by experiments. In conclusion, this study described a mechanism by which the sphingolipid signalling pathway was involved in adipose tissue remodelling by inducing inflammation and fat fibrosis in cardiac cachexia. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12944-023-01967-0.
format Online
Article
Text
id pubmed-10691093
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-106910932023-12-02 A comprehensive multiomics approach reveals that high levels of sphingolipids in cardiac cachexia adipose tissue are associated with inflammatory and fibrotic changes Qu, Yiwei Wang, Yong Wu, Tao Liu, Xue Wang, Huaizhe Ma, Dufang Lipids Health Dis Research Cardiac cachexia is a deadly consequence of advanced heart failure that is characterised by the dysregulation of adipose tissue homeostasis. Once cachexia occurs with heart failure, it prevents the normal treatment of heart failure and increases the risk of death. Targeting adipose tissue is an important approach to treating cardiac cachexia, but the pathogenic mechanisms are still unknown, and there are no effective therapies available. Transcriptomics, metabolomics, and lipidomics were used to examine the underlying mechanisms of cardiac cachexia. Transcriptomics investigation of cardiac cachexia adipose tissue revealed that genes involved in fibrosis and monocyte/macrophage migration were increased and strongly interacted. The ECM-receptor interaction pathway was primarily enriched, as shown by KEGG enrichment analysis. In addition, gene set enrichment analysis revealed that monocyte chemotaxis/macrophage migration and fibrosis gene sets were upregulated in cardiac cachexia. Metabolomics enrichment analysis demonstrated that the sphingolipid signalling pathway is important for adipose tissue remodelling in cardiac cachexia. Lipidomics analysis showed that the adipose tissue of rats with cardiac cachexia had higher levels of sphingolipids, including Cer and S1P. Moreover, combined multiomics analysis suggested that the sphingolipid metabolic pathway was associated with inflammatory-fibrotic changes in adipose tissue. Finally, the key indicators were validated by experiments. In conclusion, this study described a mechanism by which the sphingolipid signalling pathway was involved in adipose tissue remodelling by inducing inflammation and fat fibrosis in cardiac cachexia. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12944-023-01967-0. BioMed Central 2023-12-01 /pmc/articles/PMC10691093/ /pubmed/38041133 http://dx.doi.org/10.1186/s12944-023-01967-0 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/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 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/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://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
Qu, Yiwei
Wang, Yong
Wu, Tao
Liu, Xue
Wang, Huaizhe
Ma, Dufang
A comprehensive multiomics approach reveals that high levels of sphingolipids in cardiac cachexia adipose tissue are associated with inflammatory and fibrotic changes
title A comprehensive multiomics approach reveals that high levels of sphingolipids in cardiac cachexia adipose tissue are associated with inflammatory and fibrotic changes
title_full A comprehensive multiomics approach reveals that high levels of sphingolipids in cardiac cachexia adipose tissue are associated with inflammatory and fibrotic changes
title_fullStr A comprehensive multiomics approach reveals that high levels of sphingolipids in cardiac cachexia adipose tissue are associated with inflammatory and fibrotic changes
title_full_unstemmed A comprehensive multiomics approach reveals that high levels of sphingolipids in cardiac cachexia adipose tissue are associated with inflammatory and fibrotic changes
title_short A comprehensive multiomics approach reveals that high levels of sphingolipids in cardiac cachexia adipose tissue are associated with inflammatory and fibrotic changes
title_sort comprehensive multiomics approach reveals that high levels of sphingolipids in cardiac cachexia adipose tissue are associated with inflammatory and fibrotic changes
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10691093/
https://www.ncbi.nlm.nih.gov/pubmed/38041133
http://dx.doi.org/10.1186/s12944-023-01967-0
work_keys_str_mv AT quyiwei acomprehensivemultiomicsapproachrevealsthathighlevelsofsphingolipidsincardiaccachexiaadiposetissueareassociatedwithinflammatoryandfibroticchanges
AT wangyong acomprehensivemultiomicsapproachrevealsthathighlevelsofsphingolipidsincardiaccachexiaadiposetissueareassociatedwithinflammatoryandfibroticchanges
AT wutao acomprehensivemultiomicsapproachrevealsthathighlevelsofsphingolipidsincardiaccachexiaadiposetissueareassociatedwithinflammatoryandfibroticchanges
AT liuxue acomprehensivemultiomicsapproachrevealsthathighlevelsofsphingolipidsincardiaccachexiaadiposetissueareassociatedwithinflammatoryandfibroticchanges
AT wanghuaizhe acomprehensivemultiomicsapproachrevealsthathighlevelsofsphingolipidsincardiaccachexiaadiposetissueareassociatedwithinflammatoryandfibroticchanges
AT madufang acomprehensivemultiomicsapproachrevealsthathighlevelsofsphingolipidsincardiaccachexiaadiposetissueareassociatedwithinflammatoryandfibroticchanges
AT quyiwei comprehensivemultiomicsapproachrevealsthathighlevelsofsphingolipidsincardiaccachexiaadiposetissueareassociatedwithinflammatoryandfibroticchanges
AT wangyong comprehensivemultiomicsapproachrevealsthathighlevelsofsphingolipidsincardiaccachexiaadiposetissueareassociatedwithinflammatoryandfibroticchanges
AT wutao comprehensivemultiomicsapproachrevealsthathighlevelsofsphingolipidsincardiaccachexiaadiposetissueareassociatedwithinflammatoryandfibroticchanges
AT liuxue comprehensivemultiomicsapproachrevealsthathighlevelsofsphingolipidsincardiaccachexiaadiposetissueareassociatedwithinflammatoryandfibroticchanges
AT wanghuaizhe comprehensivemultiomicsapproachrevealsthathighlevelsofsphingolipidsincardiaccachexiaadiposetissueareassociatedwithinflammatoryandfibroticchanges
AT madufang comprehensivemultiomicsapproachrevealsthathighlevelsofsphingolipidsincardiaccachexiaadiposetissueareassociatedwithinflammatoryandfibroticchanges