Cargando…

Citrate Promotes Excessive Lipid Biosynthesis and Senescence in Tumor Cells for Tumor Therapy

Metabolic disorder is one of the hallmarks of cancers, and reprogramming of metabolism is becoming a novel strategy for cancer treatment. Citrate is a key metabolite and critical metabolic regulator linking glycolysis and lipid metabolism in cellular energy homeostasis. Here it is reported that citr...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhao, Yangjing, Liu, Xia, Si, Fusheng, Huang, Lan, Gao, Aiqin, Lin, Wenli, Hoft, Daniel F., Shao, Qixiang, Peng, Guangyong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8728847/
https://www.ncbi.nlm.nih.gov/pubmed/34747157
http://dx.doi.org/10.1002/advs.202101553
_version_ 1784626816278855680
author Zhao, Yangjing
Liu, Xia
Si, Fusheng
Huang, Lan
Gao, Aiqin
Lin, Wenli
Hoft, Daniel F.
Shao, Qixiang
Peng, Guangyong
author_facet Zhao, Yangjing
Liu, Xia
Si, Fusheng
Huang, Lan
Gao, Aiqin
Lin, Wenli
Hoft, Daniel F.
Shao, Qixiang
Peng, Guangyong
author_sort Zhao, Yangjing
collection PubMed
description Metabolic disorder is one of the hallmarks of cancers, and reprogramming of metabolism is becoming a novel strategy for cancer treatment. Citrate is a key metabolite and critical metabolic regulator linking glycolysis and lipid metabolism in cellular energy homeostasis. Here it is reported that citrate treatment (both sodium citrate and citric acid) significantly suppresses tumor cell proliferation and growth in various tumor types. Mechanistically, citrate promotes excessive lipid biosynthesis and induces disruption of lipid metabolism in tumor cells, resulting in tumor cell senescence and growth inhibition. Furthermore, ATM‐associated DNA damage response cooperates with MAPK and mTOR signaling pathways to control citrate‐induced tumor cell growth arrest and senescence. In vivo studies further demonstrate that citrate administration dramatically inhibits tumor growth and progression in a colon cancer xenograft model. Importantly, citrate administration combined with the conventional chemotherapy drugs exhibits synergistic antitumor effects in vivo in the colon cancer models. These results clearly indicate that citrate can reprogram lipid metabolism and cell fate in cancer cells, and targeting citrate can be a promising therapeutic strategy for tumor treatment.
format Online
Article
Text
id pubmed-8728847
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-87288472022-01-11 Citrate Promotes Excessive Lipid Biosynthesis and Senescence in Tumor Cells for Tumor Therapy Zhao, Yangjing Liu, Xia Si, Fusheng Huang, Lan Gao, Aiqin Lin, Wenli Hoft, Daniel F. Shao, Qixiang Peng, Guangyong Adv Sci (Weinh) Research Articles Metabolic disorder is one of the hallmarks of cancers, and reprogramming of metabolism is becoming a novel strategy for cancer treatment. Citrate is a key metabolite and critical metabolic regulator linking glycolysis and lipid metabolism in cellular energy homeostasis. Here it is reported that citrate treatment (both sodium citrate and citric acid) significantly suppresses tumor cell proliferation and growth in various tumor types. Mechanistically, citrate promotes excessive lipid biosynthesis and induces disruption of lipid metabolism in tumor cells, resulting in tumor cell senescence and growth inhibition. Furthermore, ATM‐associated DNA damage response cooperates with MAPK and mTOR signaling pathways to control citrate‐induced tumor cell growth arrest and senescence. In vivo studies further demonstrate that citrate administration dramatically inhibits tumor growth and progression in a colon cancer xenograft model. Importantly, citrate administration combined with the conventional chemotherapy drugs exhibits synergistic antitumor effects in vivo in the colon cancer models. These results clearly indicate that citrate can reprogram lipid metabolism and cell fate in cancer cells, and targeting citrate can be a promising therapeutic strategy for tumor treatment. John Wiley and Sons Inc. 2021-11-07 /pmc/articles/PMC8728847/ /pubmed/34747157 http://dx.doi.org/10.1002/advs.202101553 Text en © 2021 The Authors. Advanced Science published by Wiley‐VCH GmbH https://creativecommons.org/licenses/by/4.0/This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
spellingShingle Research Articles
Zhao, Yangjing
Liu, Xia
Si, Fusheng
Huang, Lan
Gao, Aiqin
Lin, Wenli
Hoft, Daniel F.
Shao, Qixiang
Peng, Guangyong
Citrate Promotes Excessive Lipid Biosynthesis and Senescence in Tumor Cells for Tumor Therapy
title Citrate Promotes Excessive Lipid Biosynthesis and Senescence in Tumor Cells for Tumor Therapy
title_full Citrate Promotes Excessive Lipid Biosynthesis and Senescence in Tumor Cells for Tumor Therapy
title_fullStr Citrate Promotes Excessive Lipid Biosynthesis and Senescence in Tumor Cells for Tumor Therapy
title_full_unstemmed Citrate Promotes Excessive Lipid Biosynthesis and Senescence in Tumor Cells for Tumor Therapy
title_short Citrate Promotes Excessive Lipid Biosynthesis and Senescence in Tumor Cells for Tumor Therapy
title_sort citrate promotes excessive lipid biosynthesis and senescence in tumor cells for tumor therapy
topic Research Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8728847/
https://www.ncbi.nlm.nih.gov/pubmed/34747157
http://dx.doi.org/10.1002/advs.202101553
work_keys_str_mv AT zhaoyangjing citratepromotesexcessivelipidbiosynthesisandsenescenceintumorcellsfortumortherapy
AT liuxia citratepromotesexcessivelipidbiosynthesisandsenescenceintumorcellsfortumortherapy
AT sifusheng citratepromotesexcessivelipidbiosynthesisandsenescenceintumorcellsfortumortherapy
AT huanglan citratepromotesexcessivelipidbiosynthesisandsenescenceintumorcellsfortumortherapy
AT gaoaiqin citratepromotesexcessivelipidbiosynthesisandsenescenceintumorcellsfortumortherapy
AT linwenli citratepromotesexcessivelipidbiosynthesisandsenescenceintumorcellsfortumortherapy
AT hoftdanielf citratepromotesexcessivelipidbiosynthesisandsenescenceintumorcellsfortumortherapy
AT shaoqixiang citratepromotesexcessivelipidbiosynthesisandsenescenceintumorcellsfortumortherapy
AT pengguangyong citratepromotesexcessivelipidbiosynthesisandsenescenceintumorcellsfortumortherapy