Cargando…

Matrix stiffness mediates pancreatic cancer chemoresistance through induction of exosome hypersecretion in a cancer associated fibroblasts-tumor organoid biomimetic model

In pancreatic ductal adenocarcinoma (PDAC), the abundant stromal cells which comprise the tumor microenvironment constitute more than 90% of the primary tumor bulk. Moreover, this desmoplastic environment has been found to be three times stiffer than normal pancreas tissue. Despite the importance of...

Descripción completa

Detalles Bibliográficos
Autores principales: Xiao, Weikun, Pahlavanneshan, Mahsa, Eun, Chae-Young, Zhang, Xinyu, DeKalb, Charlene, Mahgoub, Bayan, Knaneh-Monem, Hanaa, Shah, Sana, Sohrabi, Alireza, Seidlits, Stephanie K., Hill, Reginald
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Elsevier 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9126837/
https://www.ncbi.nlm.nih.gov/pubmed/35619988
http://dx.doi.org/10.1016/j.mbplus.2022.100111
_version_ 1784712216540348416
author Xiao, Weikun
Pahlavanneshan, Mahsa
Eun, Chae-Young
Zhang, Xinyu
DeKalb, Charlene
Mahgoub, Bayan
Knaneh-Monem, Hanaa
Shah, Sana
Sohrabi, Alireza
Seidlits, Stephanie K.
Hill, Reginald
author_facet Xiao, Weikun
Pahlavanneshan, Mahsa
Eun, Chae-Young
Zhang, Xinyu
DeKalb, Charlene
Mahgoub, Bayan
Knaneh-Monem, Hanaa
Shah, Sana
Sohrabi, Alireza
Seidlits, Stephanie K.
Hill, Reginald
author_sort Xiao, Weikun
collection PubMed
description In pancreatic ductal adenocarcinoma (PDAC), the abundant stromal cells which comprise the tumor microenvironment constitute more than 90% of the primary tumor bulk. Moreover, this desmoplastic environment has been found to be three times stiffer than normal pancreas tissue. Despite the importance of studying the desmoplastic environment of PDAC, there is still a lack of models designed to adequately recapitulate this complex stiff microenvironment, a critical hallmark of the disease that has been shown to induce chemoresistance. Here, we present a bio-mimetic, 3-dimensional co-culture system that integrates tumor organoids and host-matching stromal cancer associated-fibroblasts (CAFs) that recapitulates the complex, fibrotic matrix of PDAC using advanced biomaterials. With this model, we show that matrix-activated CAFs are able to “re-engineer” the fibrotic environment into a significantly stiffer environment through lysyl-oxidase dependent crosslinking. Moreover, we show that culture of CAFs in this model leads to an increase of exosomes; extracellular vesicles known to promote chemoresistance. Finally, using previously identified exosome inhibitors, climbazole and imipramine, we demonstrate how abrogation of exosome hypersecretion can reduce matrix stiffness-induced chemoresistance. These data highlight the importance of the development of new models that recapitulate not only the cellular composition found in PDAC tumors, but also the biophysical stresses, like stiffness, that the cells are exposed to in order to identify therapies that can overcome this critical feature which can contribute to the chemoresistance observed in patients. We believe that the 3D bio-mimetic model we have developed will be a valuable tool for the development, testing, and optimization of “mechano-medicines” designed to target the biophysical forces that lead to tumor growth and chemoresistance.
format Online
Article
Text
id pubmed-9126837
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Elsevier
record_format MEDLINE/PubMed
spelling pubmed-91268372022-05-25 Matrix stiffness mediates pancreatic cancer chemoresistance through induction of exosome hypersecretion in a cancer associated fibroblasts-tumor organoid biomimetic model Xiao, Weikun Pahlavanneshan, Mahsa Eun, Chae-Young Zhang, Xinyu DeKalb, Charlene Mahgoub, Bayan Knaneh-Monem, Hanaa Shah, Sana Sohrabi, Alireza Seidlits, Stephanie K. Hill, Reginald Matrix Biol Plus Short Communication In pancreatic ductal adenocarcinoma (PDAC), the abundant stromal cells which comprise the tumor microenvironment constitute more than 90% of the primary tumor bulk. Moreover, this desmoplastic environment has been found to be three times stiffer than normal pancreas tissue. Despite the importance of studying the desmoplastic environment of PDAC, there is still a lack of models designed to adequately recapitulate this complex stiff microenvironment, a critical hallmark of the disease that has been shown to induce chemoresistance. Here, we present a bio-mimetic, 3-dimensional co-culture system that integrates tumor organoids and host-matching stromal cancer associated-fibroblasts (CAFs) that recapitulates the complex, fibrotic matrix of PDAC using advanced biomaterials. With this model, we show that matrix-activated CAFs are able to “re-engineer” the fibrotic environment into a significantly stiffer environment through lysyl-oxidase dependent crosslinking. Moreover, we show that culture of CAFs in this model leads to an increase of exosomes; extracellular vesicles known to promote chemoresistance. Finally, using previously identified exosome inhibitors, climbazole and imipramine, we demonstrate how abrogation of exosome hypersecretion can reduce matrix stiffness-induced chemoresistance. These data highlight the importance of the development of new models that recapitulate not only the cellular composition found in PDAC tumors, but also the biophysical stresses, like stiffness, that the cells are exposed to in order to identify therapies that can overcome this critical feature which can contribute to the chemoresistance observed in patients. We believe that the 3D bio-mimetic model we have developed will be a valuable tool for the development, testing, and optimization of “mechano-medicines” designed to target the biophysical forces that lead to tumor growth and chemoresistance. Elsevier 2022-05-16 /pmc/articles/PMC9126837/ /pubmed/35619988 http://dx.doi.org/10.1016/j.mbplus.2022.100111 Text en © 2022 The Author(s) https://creativecommons.org/licenses/by-nc-nd/4.0/This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
spellingShingle Short Communication
Xiao, Weikun
Pahlavanneshan, Mahsa
Eun, Chae-Young
Zhang, Xinyu
DeKalb, Charlene
Mahgoub, Bayan
Knaneh-Monem, Hanaa
Shah, Sana
Sohrabi, Alireza
Seidlits, Stephanie K.
Hill, Reginald
Matrix stiffness mediates pancreatic cancer chemoresistance through induction of exosome hypersecretion in a cancer associated fibroblasts-tumor organoid biomimetic model
title Matrix stiffness mediates pancreatic cancer chemoresistance through induction of exosome hypersecretion in a cancer associated fibroblasts-tumor organoid biomimetic model
title_full Matrix stiffness mediates pancreatic cancer chemoresistance through induction of exosome hypersecretion in a cancer associated fibroblasts-tumor organoid biomimetic model
title_fullStr Matrix stiffness mediates pancreatic cancer chemoresistance through induction of exosome hypersecretion in a cancer associated fibroblasts-tumor organoid biomimetic model
title_full_unstemmed Matrix stiffness mediates pancreatic cancer chemoresistance through induction of exosome hypersecretion in a cancer associated fibroblasts-tumor organoid biomimetic model
title_short Matrix stiffness mediates pancreatic cancer chemoresistance through induction of exosome hypersecretion in a cancer associated fibroblasts-tumor organoid biomimetic model
title_sort matrix stiffness mediates pancreatic cancer chemoresistance through induction of exosome hypersecretion in a cancer associated fibroblasts-tumor organoid biomimetic model
topic Short Communication
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9126837/
https://www.ncbi.nlm.nih.gov/pubmed/35619988
http://dx.doi.org/10.1016/j.mbplus.2022.100111
work_keys_str_mv AT xiaoweikun matrixstiffnessmediatespancreaticcancerchemoresistancethroughinductionofexosomehypersecretioninacancerassociatedfibroblaststumororganoidbiomimeticmodel
AT pahlavanneshanmahsa matrixstiffnessmediatespancreaticcancerchemoresistancethroughinductionofexosomehypersecretioninacancerassociatedfibroblaststumororganoidbiomimeticmodel
AT eunchaeyoung matrixstiffnessmediatespancreaticcancerchemoresistancethroughinductionofexosomehypersecretioninacancerassociatedfibroblaststumororganoidbiomimeticmodel
AT zhangxinyu matrixstiffnessmediatespancreaticcancerchemoresistancethroughinductionofexosomehypersecretioninacancerassociatedfibroblaststumororganoidbiomimeticmodel
AT dekalbcharlene matrixstiffnessmediatespancreaticcancerchemoresistancethroughinductionofexosomehypersecretioninacancerassociatedfibroblaststumororganoidbiomimeticmodel
AT mahgoubbayan matrixstiffnessmediatespancreaticcancerchemoresistancethroughinductionofexosomehypersecretioninacancerassociatedfibroblaststumororganoidbiomimeticmodel
AT knanehmonemhanaa matrixstiffnessmediatespancreaticcancerchemoresistancethroughinductionofexosomehypersecretioninacancerassociatedfibroblaststumororganoidbiomimeticmodel
AT shahsana matrixstiffnessmediatespancreaticcancerchemoresistancethroughinductionofexosomehypersecretioninacancerassociatedfibroblaststumororganoidbiomimeticmodel
AT sohrabialireza matrixstiffnessmediatespancreaticcancerchemoresistancethroughinductionofexosomehypersecretioninacancerassociatedfibroblaststumororganoidbiomimeticmodel
AT seidlitsstephaniek matrixstiffnessmediatespancreaticcancerchemoresistancethroughinductionofexosomehypersecretioninacancerassociatedfibroblaststumororganoidbiomimeticmodel
AT hillreginald matrixstiffnessmediatespancreaticcancerchemoresistancethroughinductionofexosomehypersecretioninacancerassociatedfibroblaststumororganoidbiomimeticmodel