Cargando…

FAP(high) α‐SMA(low) cancer‐associated fibroblast‐derived SLPI protein encapsulated in extracellular vesicles promotes ovarian cancer development via activation of PI3K/AKT and downstream signaling pathways

Ovarian cancer is the most lethal gynecological malignancy worldwide with high metastasis and poor prognosis rates. Cancer‐associated fibroblasts (CAFs), a heterogeneous population of cells that constitutes a major component of the tumor microenvironment, secrete extracellular vesicles (EVs) loading...

Descripción completa

Detalles Bibliográficos
Autores principales: Sun, Luyao, Ke, Miaola, Wang, Xin, Yin, Mengyuan, Wei, Junni, Xu, Lu, Tian, Xing, Wang, Fei, Zhang, He, Fu, Songbin, Zhang, Chunyu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: John Wiley and Sons Inc. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9541539/
https://www.ncbi.nlm.nih.gov/pubmed/35801406
http://dx.doi.org/10.1002/mc.23445
_version_ 1784803948019843072
author Sun, Luyao
Ke, Miaola
Wang, Xin
Yin, Mengyuan
Wei, Junni
Xu, Lu
Tian, Xing
Wang, Fei
Zhang, He
Fu, Songbin
Zhang, Chunyu
author_facet Sun, Luyao
Ke, Miaola
Wang, Xin
Yin, Mengyuan
Wei, Junni
Xu, Lu
Tian, Xing
Wang, Fei
Zhang, He
Fu, Songbin
Zhang, Chunyu
author_sort Sun, Luyao
collection PubMed
description Ovarian cancer is the most lethal gynecological malignancy worldwide with high metastasis and poor prognosis rates. Cancer‐associated fibroblasts (CAFs), a heterogeneous population of cells that constitutes a major component of the tumor microenvironment, secrete extracellular vesicles (EVs) loading with proteins, lipids, and RNAs to promote tumorigenesis. However, the specific roles of CAF‐derived proteins contained in EVs in ovarian cancer remain poorly understood at present. Using the gene expression microarray analysis, we identified a list of dysregulated genes between the α‐SMA(+)CAF and FAP(+)CAF subpopulations, from which secretory leukocyte protease inhibitor (SLPI) was chosen for further validation. Quantitative PCR, western blot, immunohistochemistry, and enzyme‐linked immunosorbent assays were used to assess SLPI expression in ovarian cancer cells, tissues, CAFs, and EVs. Additionally, we evaluated the effects of exogenous SLPI on proliferation, migration, invasion, and adhesion of ovarian cancer cells in vitro. Our results showed SLPI protein was upregulated in CAFs, particularly in the FAP(high)α‐SMA(low)CAF subpopulation, and associated with increased tumor grade and decreased overall survival (OS). Importantly, CAF‐derived SLPI protein could be encapsulated in EVs for delivery to ovarian cancer cells, thus facilitating cell proliferation, migration, invasion, and adhesion via activating the PI3K/AKT and downstream signaling pathways. Moreover, high plasma expression of SLPI encapsulated in EVs was closely correlated with tumor stage in ovarian cancer patients. Our collective results highlight an oncogenic role of plasma EV‐encapsulated SLPI secreted by CAFs in tumor progression for the first time, supporting its potential utility as a prognostic biomarker of ovarian cancer.
format Online
Article
Text
id pubmed-9541539
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher John Wiley and Sons Inc.
record_format MEDLINE/PubMed
spelling pubmed-95415392022-10-14 FAP(high) α‐SMA(low) cancer‐associated fibroblast‐derived SLPI protein encapsulated in extracellular vesicles promotes ovarian cancer development via activation of PI3K/AKT and downstream signaling pathways Sun, Luyao Ke, Miaola Wang, Xin Yin, Mengyuan Wei, Junni Xu, Lu Tian, Xing Wang, Fei Zhang, He Fu, Songbin Zhang, Chunyu Mol Carcinog Research Articles Ovarian cancer is the most lethal gynecological malignancy worldwide with high metastasis and poor prognosis rates. Cancer‐associated fibroblasts (CAFs), a heterogeneous population of cells that constitutes a major component of the tumor microenvironment, secrete extracellular vesicles (EVs) loading with proteins, lipids, and RNAs to promote tumorigenesis. However, the specific roles of CAF‐derived proteins contained in EVs in ovarian cancer remain poorly understood at present. Using the gene expression microarray analysis, we identified a list of dysregulated genes between the α‐SMA(+)CAF and FAP(+)CAF subpopulations, from which secretory leukocyte protease inhibitor (SLPI) was chosen for further validation. Quantitative PCR, western blot, immunohistochemistry, and enzyme‐linked immunosorbent assays were used to assess SLPI expression in ovarian cancer cells, tissues, CAFs, and EVs. Additionally, we evaluated the effects of exogenous SLPI on proliferation, migration, invasion, and adhesion of ovarian cancer cells in vitro. Our results showed SLPI protein was upregulated in CAFs, particularly in the FAP(high)α‐SMA(low)CAF subpopulation, and associated with increased tumor grade and decreased overall survival (OS). Importantly, CAF‐derived SLPI protein could be encapsulated in EVs for delivery to ovarian cancer cells, thus facilitating cell proliferation, migration, invasion, and adhesion via activating the PI3K/AKT and downstream signaling pathways. Moreover, high plasma expression of SLPI encapsulated in EVs was closely correlated with tumor stage in ovarian cancer patients. Our collective results highlight an oncogenic role of plasma EV‐encapsulated SLPI secreted by CAFs in tumor progression for the first time, supporting its potential utility as a prognostic biomarker of ovarian cancer. John Wiley and Sons Inc. 2022-07-08 2022-10 /pmc/articles/PMC9541539/ /pubmed/35801406 http://dx.doi.org/10.1002/mc.23445 Text en © 2022 The Authors. Molecular Carcinogenesis published by Wiley Periodicals LLC. https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc-nd/4.0/ (https://creativecommons.org/licenses/by-nc-nd/4.0/) License, which permits use and distribution in any medium, provided the original work is properly cited, the use is non‐commercial and no modifications or adaptations are made.
spellingShingle Research Articles
Sun, Luyao
Ke, Miaola
Wang, Xin
Yin, Mengyuan
Wei, Junni
Xu, Lu
Tian, Xing
Wang, Fei
Zhang, He
Fu, Songbin
Zhang, Chunyu
FAP(high) α‐SMA(low) cancer‐associated fibroblast‐derived SLPI protein encapsulated in extracellular vesicles promotes ovarian cancer development via activation of PI3K/AKT and downstream signaling pathways
title FAP(high) α‐SMA(low) cancer‐associated fibroblast‐derived SLPI protein encapsulated in extracellular vesicles promotes ovarian cancer development via activation of PI3K/AKT and downstream signaling pathways
title_full FAP(high) α‐SMA(low) cancer‐associated fibroblast‐derived SLPI protein encapsulated in extracellular vesicles promotes ovarian cancer development via activation of PI3K/AKT and downstream signaling pathways
title_fullStr FAP(high) α‐SMA(low) cancer‐associated fibroblast‐derived SLPI protein encapsulated in extracellular vesicles promotes ovarian cancer development via activation of PI3K/AKT and downstream signaling pathways
title_full_unstemmed FAP(high) α‐SMA(low) cancer‐associated fibroblast‐derived SLPI protein encapsulated in extracellular vesicles promotes ovarian cancer development via activation of PI3K/AKT and downstream signaling pathways
title_short FAP(high) α‐SMA(low) cancer‐associated fibroblast‐derived SLPI protein encapsulated in extracellular vesicles promotes ovarian cancer development via activation of PI3K/AKT and downstream signaling pathways
title_sort fap(high) α‐sma(low) cancer‐associated fibroblast‐derived slpi protein encapsulated in extracellular vesicles promotes ovarian cancer development via activation of pi3k/akt and downstream signaling pathways
topic Research Articles
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9541539/
https://www.ncbi.nlm.nih.gov/pubmed/35801406
http://dx.doi.org/10.1002/mc.23445
work_keys_str_mv AT sunluyao faphighasmalowcancerassociatedfibroblastderivedslpiproteinencapsulatedinextracellularvesiclespromotesovariancancerdevelopmentviaactivationofpi3kaktanddownstreamsignalingpathways
AT kemiaola faphighasmalowcancerassociatedfibroblastderivedslpiproteinencapsulatedinextracellularvesiclespromotesovariancancerdevelopmentviaactivationofpi3kaktanddownstreamsignalingpathways
AT wangxin faphighasmalowcancerassociatedfibroblastderivedslpiproteinencapsulatedinextracellularvesiclespromotesovariancancerdevelopmentviaactivationofpi3kaktanddownstreamsignalingpathways
AT yinmengyuan faphighasmalowcancerassociatedfibroblastderivedslpiproteinencapsulatedinextracellularvesiclespromotesovariancancerdevelopmentviaactivationofpi3kaktanddownstreamsignalingpathways
AT weijunni faphighasmalowcancerassociatedfibroblastderivedslpiproteinencapsulatedinextracellularvesiclespromotesovariancancerdevelopmentviaactivationofpi3kaktanddownstreamsignalingpathways
AT xulu faphighasmalowcancerassociatedfibroblastderivedslpiproteinencapsulatedinextracellularvesiclespromotesovariancancerdevelopmentviaactivationofpi3kaktanddownstreamsignalingpathways
AT tianxing faphighasmalowcancerassociatedfibroblastderivedslpiproteinencapsulatedinextracellularvesiclespromotesovariancancerdevelopmentviaactivationofpi3kaktanddownstreamsignalingpathways
AT wangfei faphighasmalowcancerassociatedfibroblastderivedslpiproteinencapsulatedinextracellularvesiclespromotesovariancancerdevelopmentviaactivationofpi3kaktanddownstreamsignalingpathways
AT zhanghe faphighasmalowcancerassociatedfibroblastderivedslpiproteinencapsulatedinextracellularvesiclespromotesovariancancerdevelopmentviaactivationofpi3kaktanddownstreamsignalingpathways
AT fusongbin faphighasmalowcancerassociatedfibroblastderivedslpiproteinencapsulatedinextracellularvesiclespromotesovariancancerdevelopmentviaactivationofpi3kaktanddownstreamsignalingpathways
AT zhangchunyu faphighasmalowcancerassociatedfibroblastderivedslpiproteinencapsulatedinextracellularvesiclespromotesovariancancerdevelopmentviaactivationofpi3kaktanddownstreamsignalingpathways