Cargando…

Gastric cancer-derived LBP promotes liver metastasis by driving intrahepatic fibrotic pre-metastatic niche formation

BACKGROUND: Liver metastasis (LM) is one of the most common distant metastases of gastric cancer (GC). However, the mechanisms underlying the LM of GC (GC-LM) remain poorly understood. This study aimed to identify the tumour-secreted protein associated with GC-LM and to investigate the mechanisms by...

Descripción completa

Detalles Bibliográficos
Autores principales: Xie, Li, Qiu, Shengkui, Lu, Chen, Gu, Chao, Wang, Jihuan, Lv, Jialun, Fang, Lang, Chen, Zetian, Li, Ying, Jiang, Tianlu, Xia, Yiwen, Wang, Weizhi, Li, Bowen, Xu, Zekuan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10546721/
https://www.ncbi.nlm.nih.gov/pubmed/37789385
http://dx.doi.org/10.1186/s13046-023-02833-8
_version_ 1785114918888931328
author Xie, Li
Qiu, Shengkui
Lu, Chen
Gu, Chao
Wang, Jihuan
Lv, Jialun
Fang, Lang
Chen, Zetian
Li, Ying
Jiang, Tianlu
Xia, Yiwen
Wang, Weizhi
Li, Bowen
Xu, Zekuan
author_facet Xie, Li
Qiu, Shengkui
Lu, Chen
Gu, Chao
Wang, Jihuan
Lv, Jialun
Fang, Lang
Chen, Zetian
Li, Ying
Jiang, Tianlu
Xia, Yiwen
Wang, Weizhi
Li, Bowen
Xu, Zekuan
author_sort Xie, Li
collection PubMed
description BACKGROUND: Liver metastasis (LM) is one of the most common distant metastases of gastric cancer (GC). However, the mechanisms underlying the LM of GC (GC-LM) remain poorly understood. This study aimed to identify the tumour-secreted protein associated with GC-LM and to investigate the mechanisms by which this secreted protein remodels the liver microenvironment to promote GC-LM. METHODS: Data-independent acquisition mass spectrometry (DIA-MS), mRNA expression microarray, quantitative real-time PCR, enzyme-linked immunosorbent assay (ELISA) and immunohistochemistry (IHC) were performed to identify and validate the GC-secreted proteins associated with GC-LM. A modified intrasplenic injection mouse model of LM was used to evaluate the progression and tumour burden of LM in vivo. Flow cytometry, immunofluorescence (IF), western blots (WB) and IHC were performed to validate the pre-metastatic niche (PMN) formation in the pre-modelling mouse models. mRNA sequencing of PMA-treated THP-1 cells with or without lipopolysaccharide binding protein (LBP) treatment was used to identify the functional target genes of LBP in macrophages. Co-immunoprecipitation (Co-IP), WB, ELISA, IF and Transwell assays were performed to explore the underlying mechanism of LBP in inducing intrahepatic PMN formation. RESULTS: LBP was identified as a critical secreted protein associated with GC-LM and correlated with a worse prognosis in patients with GC. LBP activated the TLR4/NF-κB pathway to promote TGF-β1 secretion in intrahepatic macrophages, which, in turn, activated hepatic satellite cells (HSCs) to direct intrahepatic fibrotic PMN formation. Additionally, TGF-β1 enhanced the migration and invasion of incoming metastatic GC cells in the liver. Consequently, selective targeting of the TGF-β/Smad signaling pathway with galunisertib demonstrated its efficacy in effectively preventing GC-LM in vivo. CONCLUSIONS: The results of this study provide compelling evidence that serological LBP can serve as a valuable diagnostic biomarker for the early detection of GC-LM. Mechanistically, GC-derived LBP mediates the crosstalk between primary GC cells and the intrahepatic microenvironment by promoting TGF-β1 secretion in intrahepatic macrophages, which induces intrahepatic fibrotic PMN formation to promote GC-LM. Importantly, selectively targeting the TGF-β/Smad signaling pathway with galunisertib represents a promising preventive and therapeutic strategy for GC-LM. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13046-023-02833-8.
format Online
Article
Text
id pubmed-10546721
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-105467212023-10-04 Gastric cancer-derived LBP promotes liver metastasis by driving intrahepatic fibrotic pre-metastatic niche formation Xie, Li Qiu, Shengkui Lu, Chen Gu, Chao Wang, Jihuan Lv, Jialun Fang, Lang Chen, Zetian Li, Ying Jiang, Tianlu Xia, Yiwen Wang, Weizhi Li, Bowen Xu, Zekuan J Exp Clin Cancer Res Research BACKGROUND: Liver metastasis (LM) is one of the most common distant metastases of gastric cancer (GC). However, the mechanisms underlying the LM of GC (GC-LM) remain poorly understood. This study aimed to identify the tumour-secreted protein associated with GC-LM and to investigate the mechanisms by which this secreted protein remodels the liver microenvironment to promote GC-LM. METHODS: Data-independent acquisition mass spectrometry (DIA-MS), mRNA expression microarray, quantitative real-time PCR, enzyme-linked immunosorbent assay (ELISA) and immunohistochemistry (IHC) were performed to identify and validate the GC-secreted proteins associated with GC-LM. A modified intrasplenic injection mouse model of LM was used to evaluate the progression and tumour burden of LM in vivo. Flow cytometry, immunofluorescence (IF), western blots (WB) and IHC were performed to validate the pre-metastatic niche (PMN) formation in the pre-modelling mouse models. mRNA sequencing of PMA-treated THP-1 cells with or without lipopolysaccharide binding protein (LBP) treatment was used to identify the functional target genes of LBP in macrophages. Co-immunoprecipitation (Co-IP), WB, ELISA, IF and Transwell assays were performed to explore the underlying mechanism of LBP in inducing intrahepatic PMN formation. RESULTS: LBP was identified as a critical secreted protein associated with GC-LM and correlated with a worse prognosis in patients with GC. LBP activated the TLR4/NF-κB pathway to promote TGF-β1 secretion in intrahepatic macrophages, which, in turn, activated hepatic satellite cells (HSCs) to direct intrahepatic fibrotic PMN formation. Additionally, TGF-β1 enhanced the migration and invasion of incoming metastatic GC cells in the liver. Consequently, selective targeting of the TGF-β/Smad signaling pathway with galunisertib demonstrated its efficacy in effectively preventing GC-LM in vivo. CONCLUSIONS: The results of this study provide compelling evidence that serological LBP can serve as a valuable diagnostic biomarker for the early detection of GC-LM. Mechanistically, GC-derived LBP mediates the crosstalk between primary GC cells and the intrahepatic microenvironment by promoting TGF-β1 secretion in intrahepatic macrophages, which induces intrahepatic fibrotic PMN formation to promote GC-LM. Importantly, selectively targeting the TGF-β/Smad signaling pathway with galunisertib represents a promising preventive and therapeutic strategy for GC-LM. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s13046-023-02833-8. BioMed Central 2023-10-03 /pmc/articles/PMC10546721/ /pubmed/37789385 http://dx.doi.org/10.1186/s13046-023-02833-8 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
Xie, Li
Qiu, Shengkui
Lu, Chen
Gu, Chao
Wang, Jihuan
Lv, Jialun
Fang, Lang
Chen, Zetian
Li, Ying
Jiang, Tianlu
Xia, Yiwen
Wang, Weizhi
Li, Bowen
Xu, Zekuan
Gastric cancer-derived LBP promotes liver metastasis by driving intrahepatic fibrotic pre-metastatic niche formation
title Gastric cancer-derived LBP promotes liver metastasis by driving intrahepatic fibrotic pre-metastatic niche formation
title_full Gastric cancer-derived LBP promotes liver metastasis by driving intrahepatic fibrotic pre-metastatic niche formation
title_fullStr Gastric cancer-derived LBP promotes liver metastasis by driving intrahepatic fibrotic pre-metastatic niche formation
title_full_unstemmed Gastric cancer-derived LBP promotes liver metastasis by driving intrahepatic fibrotic pre-metastatic niche formation
title_short Gastric cancer-derived LBP promotes liver metastasis by driving intrahepatic fibrotic pre-metastatic niche formation
title_sort gastric cancer-derived lbp promotes liver metastasis by driving intrahepatic fibrotic pre-metastatic niche formation
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10546721/
https://www.ncbi.nlm.nih.gov/pubmed/37789385
http://dx.doi.org/10.1186/s13046-023-02833-8
work_keys_str_mv AT xieli gastriccancerderivedlbppromoteslivermetastasisbydrivingintrahepaticfibroticpremetastaticnicheformation
AT qiushengkui gastriccancerderivedlbppromoteslivermetastasisbydrivingintrahepaticfibroticpremetastaticnicheformation
AT luchen gastriccancerderivedlbppromoteslivermetastasisbydrivingintrahepaticfibroticpremetastaticnicheformation
AT guchao gastriccancerderivedlbppromoteslivermetastasisbydrivingintrahepaticfibroticpremetastaticnicheformation
AT wangjihuan gastriccancerderivedlbppromoteslivermetastasisbydrivingintrahepaticfibroticpremetastaticnicheformation
AT lvjialun gastriccancerderivedlbppromoteslivermetastasisbydrivingintrahepaticfibroticpremetastaticnicheformation
AT fanglang gastriccancerderivedlbppromoteslivermetastasisbydrivingintrahepaticfibroticpremetastaticnicheformation
AT chenzetian gastriccancerderivedlbppromoteslivermetastasisbydrivingintrahepaticfibroticpremetastaticnicheformation
AT liying gastriccancerderivedlbppromoteslivermetastasisbydrivingintrahepaticfibroticpremetastaticnicheformation
AT jiangtianlu gastriccancerderivedlbppromoteslivermetastasisbydrivingintrahepaticfibroticpremetastaticnicheformation
AT xiayiwen gastriccancerderivedlbppromoteslivermetastasisbydrivingintrahepaticfibroticpremetastaticnicheformation
AT wangweizhi gastriccancerderivedlbppromoteslivermetastasisbydrivingintrahepaticfibroticpremetastaticnicheformation
AT libowen gastriccancerderivedlbppromoteslivermetastasisbydrivingintrahepaticfibroticpremetastaticnicheformation
AT xuzekuan gastriccancerderivedlbppromoteslivermetastasisbydrivingintrahepaticfibroticpremetastaticnicheformation