Cargando…

The senescent mesothelial matrix accentuates colonization by ovarian cancer cells

Ovarian cancer is amongst the most morbid of gynecological malignancies due to its diagnosis at an advanced stage, a transcoelomic mode of metastasis, and rapid transition to chemotherapeutic resistance. Like all other malignancies, the progression of ovarian cancer may be interpreted as an emergent...

Descripción completa

Detalles Bibliográficos
Autores principales: Thapa, Bharat Vivan, Banerjee, Mallar, Glimm, Tilmann, Saini, Deepak K., Bhat, Ramray
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Springer International Publishing 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10694112/
https://www.ncbi.nlm.nih.gov/pubmed/38043093
http://dx.doi.org/10.1007/s00018-023-05017-x
_version_ 1785153301407334400
author Thapa, Bharat Vivan
Banerjee, Mallar
Glimm, Tilmann
Saini, Deepak K.
Bhat, Ramray
author_facet Thapa, Bharat Vivan
Banerjee, Mallar
Glimm, Tilmann
Saini, Deepak K.
Bhat, Ramray
author_sort Thapa, Bharat Vivan
collection PubMed
description Ovarian cancer is amongst the most morbid of gynecological malignancies due to its diagnosis at an advanced stage, a transcoelomic mode of metastasis, and rapid transition to chemotherapeutic resistance. Like all other malignancies, the progression of ovarian cancer may be interpreted as an emergent outcome of the conflict between metastasizing cancer cells and the natural defense mounted by microenvironmental barriers to such migration. Here, we asked whether senescence in coelom-lining mesothelia, brought about by drug exposure, affects their interaction with disseminated ovarian cancer cells. We observed that cancer cells adhered faster on senescent human and murine mesothelial monolayers than on non-senescent controls. Time-lapse epifluorescence microscopy showed that mesothelial cells were cleared by a host of cancer cells that surrounded the former, even under sub-confluent conditions. A multiscale computational model predicted that such colocalized mesothelial clearance under sub-confluence requires greater adhesion between cancer cells and senescent mesothelia. Consistent with the prediction, we observed that senescent mesothelia expressed an extracellular matrix with higher levels of fibronectin, laminins and hyaluronan than non-senescent controls. On senescent matrix, cancer cells adhered more efficiently, spread better, and moved faster and persistently, aiding the spread of cancer. Inhibition assays using RGD cyclopeptides suggested the adhesion was predominantly contributed by fibronectin and laminin. These findings led us to propose that the senescence-associated matrisomal phenotype of peritoneal barriers enhances the colonization of invading ovarian cancer cells contributing to the metastatic burden associated with the disease. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s00018-023-05017-x.
format Online
Article
Text
id pubmed-10694112
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Springer International Publishing
record_format MEDLINE/PubMed
spelling pubmed-106941122023-12-05 The senescent mesothelial matrix accentuates colonization by ovarian cancer cells Thapa, Bharat Vivan Banerjee, Mallar Glimm, Tilmann Saini, Deepak K. Bhat, Ramray Cell Mol Life Sci Original Article Ovarian cancer is amongst the most morbid of gynecological malignancies due to its diagnosis at an advanced stage, a transcoelomic mode of metastasis, and rapid transition to chemotherapeutic resistance. Like all other malignancies, the progression of ovarian cancer may be interpreted as an emergent outcome of the conflict between metastasizing cancer cells and the natural defense mounted by microenvironmental barriers to such migration. Here, we asked whether senescence in coelom-lining mesothelia, brought about by drug exposure, affects their interaction with disseminated ovarian cancer cells. We observed that cancer cells adhered faster on senescent human and murine mesothelial monolayers than on non-senescent controls. Time-lapse epifluorescence microscopy showed that mesothelial cells were cleared by a host of cancer cells that surrounded the former, even under sub-confluent conditions. A multiscale computational model predicted that such colocalized mesothelial clearance under sub-confluence requires greater adhesion between cancer cells and senescent mesothelia. Consistent with the prediction, we observed that senescent mesothelia expressed an extracellular matrix with higher levels of fibronectin, laminins and hyaluronan than non-senescent controls. On senescent matrix, cancer cells adhered more efficiently, spread better, and moved faster and persistently, aiding the spread of cancer. Inhibition assays using RGD cyclopeptides suggested the adhesion was predominantly contributed by fibronectin and laminin. These findings led us to propose that the senescence-associated matrisomal phenotype of peritoneal barriers enhances the colonization of invading ovarian cancer cells contributing to the metastatic burden associated with the disease. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1007/s00018-023-05017-x. Springer International Publishing 2023-12-03 2024 /pmc/articles/PMC10694112/ /pubmed/38043093 http://dx.doi.org/10.1007/s00018-023-05017-x 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/) .
spellingShingle Original Article
Thapa, Bharat Vivan
Banerjee, Mallar
Glimm, Tilmann
Saini, Deepak K.
Bhat, Ramray
The senescent mesothelial matrix accentuates colonization by ovarian cancer cells
title The senescent mesothelial matrix accentuates colonization by ovarian cancer cells
title_full The senescent mesothelial matrix accentuates colonization by ovarian cancer cells
title_fullStr The senescent mesothelial matrix accentuates colonization by ovarian cancer cells
title_full_unstemmed The senescent mesothelial matrix accentuates colonization by ovarian cancer cells
title_short The senescent mesothelial matrix accentuates colonization by ovarian cancer cells
title_sort senescent mesothelial matrix accentuates colonization by ovarian cancer cells
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10694112/
https://www.ncbi.nlm.nih.gov/pubmed/38043093
http://dx.doi.org/10.1007/s00018-023-05017-x
work_keys_str_mv AT thapabharatvivan thesenescentmesothelialmatrixaccentuatescolonizationbyovariancancercells
AT banerjeemallar thesenescentmesothelialmatrixaccentuatescolonizationbyovariancancercells
AT glimmtilmann thesenescentmesothelialmatrixaccentuatescolonizationbyovariancancercells
AT sainideepakk thesenescentmesothelialmatrixaccentuatescolonizationbyovariancancercells
AT bhatramray thesenescentmesothelialmatrixaccentuatescolonizationbyovariancancercells
AT thapabharatvivan senescentmesothelialmatrixaccentuatescolonizationbyovariancancercells
AT banerjeemallar senescentmesothelialmatrixaccentuatescolonizationbyovariancancercells
AT glimmtilmann senescentmesothelialmatrixaccentuatescolonizationbyovariancancercells
AT sainideepakk senescentmesothelialmatrixaccentuatescolonizationbyovariancancercells
AT bhatramray senescentmesothelialmatrixaccentuatescolonizationbyovariancancercells