Cargando…

Gastric cancer cell-originated small extracellular vesicle induces metabolic reprogramming of BM-MSCs through ERK-PPARγ-CPT1A signaling to potentiate lymphatic metastasis

Tumor microenvironment and metabolic reprogramming are critical for tumor metastasis. Bone marrow-derived mesenchymal stem cells (BM-MSCs) are widely involved in the formation of tumor microenvironment and present oncogenic phenotypes to facilitate lymph node metastasis (LNM) in response to small ex...

Descripción completa

Detalles Bibliográficos
Autores principales: Huang, Jiaying, Wang, Xiang, Wen, Jing, Zhao, Xinxin, Wu, Chen, Wang, Lin, Cao, Xiaoli, Dong, Haibo, Xu, Xuejing, Huang, Feng, Zhu, Wei, Wang, Mei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10169337/
https://www.ncbi.nlm.nih.gov/pubmed/37158903
http://dx.doi.org/10.1186/s12935-023-02935-5
_version_ 1785039033648283648
author Huang, Jiaying
Wang, Xiang
Wen, Jing
Zhao, Xinxin
Wu, Chen
Wang, Lin
Cao, Xiaoli
Dong, Haibo
Xu, Xuejing
Huang, Feng
Zhu, Wei
Wang, Mei
author_facet Huang, Jiaying
Wang, Xiang
Wen, Jing
Zhao, Xinxin
Wu, Chen
Wang, Lin
Cao, Xiaoli
Dong, Haibo
Xu, Xuejing
Huang, Feng
Zhu, Wei
Wang, Mei
author_sort Huang, Jiaying
collection PubMed
description Tumor microenvironment and metabolic reprogramming are critical for tumor metastasis. Bone marrow-derived mesenchymal stem cells (BM-MSCs) are widely involved in the formation of tumor microenvironment and present oncogenic phenotypes to facilitate lymph node metastasis (LNM) in response to small extracellular vesicles (sEV) released by gastric cancer (GC) cells. However, whether metabolic reprograming mediates transformation of BM-MSCs remains elusive. Herein, we revealed that the capacity of LNM-GC-sEV educating BM-MSCs was positively correlated with the LNM capacity of GC cells themselves. Fatty acid oxidation (FAO) metabolic reprogramming was indispensable for this process. Mechanistically, CD44 was identified as a critical cargo for LNM-GC-sEV enhancing FAO via ERK/PPARγ/CPT1A signaling. ATP was shown to activate STAT3 and NF-κB signaling to induce IL-8 and STC1 secretion by BM-MSCs, thereby in turn facilitating GC cells metastasis and increasing CD44 levels in GC cells and sEV to form a persistent positive feedback loop between GC cells and BM-MSCs. The critical molecules were abnormally expressed in GC tissues, sera and stroma, and correlated with the prognosis and LNM of GC patients. Together, our findings uncover the role of metabolic reprogramming mediated BM-MSCs education by LNM-GC-sEV, which presents a novel insight into the mechanism underlying LNM and provides candidate targets for GC detection and therapy. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12935-023-02935-5.
format Online
Article
Text
id pubmed-10169337
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-101693372023-05-11 Gastric cancer cell-originated small extracellular vesicle induces metabolic reprogramming of BM-MSCs through ERK-PPARγ-CPT1A signaling to potentiate lymphatic metastasis Huang, Jiaying Wang, Xiang Wen, Jing Zhao, Xinxin Wu, Chen Wang, Lin Cao, Xiaoli Dong, Haibo Xu, Xuejing Huang, Feng Zhu, Wei Wang, Mei Cancer Cell Int Research Tumor microenvironment and metabolic reprogramming are critical for tumor metastasis. Bone marrow-derived mesenchymal stem cells (BM-MSCs) are widely involved in the formation of tumor microenvironment and present oncogenic phenotypes to facilitate lymph node metastasis (LNM) in response to small extracellular vesicles (sEV) released by gastric cancer (GC) cells. However, whether metabolic reprograming mediates transformation of BM-MSCs remains elusive. Herein, we revealed that the capacity of LNM-GC-sEV educating BM-MSCs was positively correlated with the LNM capacity of GC cells themselves. Fatty acid oxidation (FAO) metabolic reprogramming was indispensable for this process. Mechanistically, CD44 was identified as a critical cargo for LNM-GC-sEV enhancing FAO via ERK/PPARγ/CPT1A signaling. ATP was shown to activate STAT3 and NF-κB signaling to induce IL-8 and STC1 secretion by BM-MSCs, thereby in turn facilitating GC cells metastasis and increasing CD44 levels in GC cells and sEV to form a persistent positive feedback loop between GC cells and BM-MSCs. The critical molecules were abnormally expressed in GC tissues, sera and stroma, and correlated with the prognosis and LNM of GC patients. Together, our findings uncover the role of metabolic reprogramming mediated BM-MSCs education by LNM-GC-sEV, which presents a novel insight into the mechanism underlying LNM and provides candidate targets for GC detection and therapy. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12935-023-02935-5. BioMed Central 2023-05-09 /pmc/articles/PMC10169337/ /pubmed/37158903 http://dx.doi.org/10.1186/s12935-023-02935-5 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/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/ (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
Huang, Jiaying
Wang, Xiang
Wen, Jing
Zhao, Xinxin
Wu, Chen
Wang, Lin
Cao, Xiaoli
Dong, Haibo
Xu, Xuejing
Huang, Feng
Zhu, Wei
Wang, Mei
Gastric cancer cell-originated small extracellular vesicle induces metabolic reprogramming of BM-MSCs through ERK-PPARγ-CPT1A signaling to potentiate lymphatic metastasis
title Gastric cancer cell-originated small extracellular vesicle induces metabolic reprogramming of BM-MSCs through ERK-PPARγ-CPT1A signaling to potentiate lymphatic metastasis
title_full Gastric cancer cell-originated small extracellular vesicle induces metabolic reprogramming of BM-MSCs through ERK-PPARγ-CPT1A signaling to potentiate lymphatic metastasis
title_fullStr Gastric cancer cell-originated small extracellular vesicle induces metabolic reprogramming of BM-MSCs through ERK-PPARγ-CPT1A signaling to potentiate lymphatic metastasis
title_full_unstemmed Gastric cancer cell-originated small extracellular vesicle induces metabolic reprogramming of BM-MSCs through ERK-PPARγ-CPT1A signaling to potentiate lymphatic metastasis
title_short Gastric cancer cell-originated small extracellular vesicle induces metabolic reprogramming of BM-MSCs through ERK-PPARγ-CPT1A signaling to potentiate lymphatic metastasis
title_sort gastric cancer cell-originated small extracellular vesicle induces metabolic reprogramming of bm-mscs through erk-pparγ-cpt1a signaling to potentiate lymphatic metastasis
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10169337/
https://www.ncbi.nlm.nih.gov/pubmed/37158903
http://dx.doi.org/10.1186/s12935-023-02935-5
work_keys_str_mv AT huangjiaying gastriccancercelloriginatedsmallextracellularvesicleinducesmetabolicreprogrammingofbmmscsthrougherkppargcpt1asignalingtopotentiatelymphaticmetastasis
AT wangxiang gastriccancercelloriginatedsmallextracellularvesicleinducesmetabolicreprogrammingofbmmscsthrougherkppargcpt1asignalingtopotentiatelymphaticmetastasis
AT wenjing gastriccancercelloriginatedsmallextracellularvesicleinducesmetabolicreprogrammingofbmmscsthrougherkppargcpt1asignalingtopotentiatelymphaticmetastasis
AT zhaoxinxin gastriccancercelloriginatedsmallextracellularvesicleinducesmetabolicreprogrammingofbmmscsthrougherkppargcpt1asignalingtopotentiatelymphaticmetastasis
AT wuchen gastriccancercelloriginatedsmallextracellularvesicleinducesmetabolicreprogrammingofbmmscsthrougherkppargcpt1asignalingtopotentiatelymphaticmetastasis
AT wanglin gastriccancercelloriginatedsmallextracellularvesicleinducesmetabolicreprogrammingofbmmscsthrougherkppargcpt1asignalingtopotentiatelymphaticmetastasis
AT caoxiaoli gastriccancercelloriginatedsmallextracellularvesicleinducesmetabolicreprogrammingofbmmscsthrougherkppargcpt1asignalingtopotentiatelymphaticmetastasis
AT donghaibo gastriccancercelloriginatedsmallextracellularvesicleinducesmetabolicreprogrammingofbmmscsthrougherkppargcpt1asignalingtopotentiatelymphaticmetastasis
AT xuxuejing gastriccancercelloriginatedsmallextracellularvesicleinducesmetabolicreprogrammingofbmmscsthrougherkppargcpt1asignalingtopotentiatelymphaticmetastasis
AT huangfeng gastriccancercelloriginatedsmallextracellularvesicleinducesmetabolicreprogrammingofbmmscsthrougherkppargcpt1asignalingtopotentiatelymphaticmetastasis
AT zhuwei gastriccancercelloriginatedsmallextracellularvesicleinducesmetabolicreprogrammingofbmmscsthrougherkppargcpt1asignalingtopotentiatelymphaticmetastasis
AT wangmei gastriccancercelloriginatedsmallextracellularvesicleinducesmetabolicreprogrammingofbmmscsthrougherkppargcpt1asignalingtopotentiatelymphaticmetastasis