Cargando…

N-cadherin mediates the migration of bone marrow-derived mesenchymal stem cells toward breast tumor cells

Rationale: Bone marrow-derived mesenchymal stem cells (BM-MSCs) recruited into breast tumors regulate the behavior of tumor cells via various mechanisms and affect clinical outcomes. Although signaling molecules, such as transforming growth factor β (TGF-β), are known to transmit signals between BM-...

Descripción completa

Detalles Bibliográficos
Autores principales: Choi, Sanghyuk, Yu, Jinyeong, Kim, Wootak, Park, Ki-Sook
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8171089/
https://www.ncbi.nlm.nih.gov/pubmed/34093853
http://dx.doi.org/10.7150/thno.59703
_version_ 1783702364178350080
author Choi, Sanghyuk
Yu, Jinyeong
Kim, Wootak
Park, Ki-Sook
author_facet Choi, Sanghyuk
Yu, Jinyeong
Kim, Wootak
Park, Ki-Sook
author_sort Choi, Sanghyuk
collection PubMed
description Rationale: Bone marrow-derived mesenchymal stem cells (BM-MSCs) recruited into breast tumors regulate the behavior of tumor cells via various mechanisms and affect clinical outcomes. Although signaling molecules, such as transforming growth factor β (TGF-β), are known to transmit signals between BM-MSCs and breast tumor cells for recruiting BM-MSCs, it is unclear which specific intrinsic molecules involved in cell motility mediate the migration of BM-MSCs into breast tumor. It is also unclear as to how specific intrinsic molecules contribute to the migration. Methods: Conditioned medium (CM) from breast tumor cells (MCF-7 and MDA-MB-231) that simulates breast tumor secreting TGF-β was used to examine the migration of BM-MSCs into breast tumors. A three-dimensional migration assay was performed to investigate the collective migration of BM-MSCs, maintaining cell-cell adhesion, toward breast tumor cells. Results: N-cadherin formed adherens junction-like structures on the intercellular borders of BM-MSCs, and TGF-β increased the expression of N-cadherin on these borders. Knockdown of Smad4 impaired the TGF-β-mediated increase in N-cadherin expression in BM-MSCs, but inhibitors of non-canonical TGF-β pathways, such as extracellular signal‐regulated kinases, Akt, and p38, did not affect it. siRNA-mediated knockdown of N-cadherin and Smad4 impaired the migration of BM-MSCs in response to TGF-β. Conditioned medium from breast tumor cells also enhanced the expression of N-cadherin in BM-MSCs, but inactivation of TGF-β type 1 receptor (TGFBR1) with SB505124 and TGFBR1 knockdown abolished the increase in N-cadherin expression. BM-MSCs collectively migrated toward CM from MDA-MB-231 in vitro while maintaining cell-cell adhesion through N-cadherin. Knockdown of N-cadherin abolished the migration of BM-MSCs toward the CM from breast tumor cells. Conclusion: In the present study, we identified N-cadherin, an intrinsic transmembrane molecule in adherens junction-like structures, on BM-MSCs as a mediator for the migration of these cells toward breast tumor. The expression of N-cadherin increases on the intercellular borders of BM-MSCs through the TGF-β canonical signaling and they collectively migrate in response to breast tumor cells expressing TGF-β via N-cadherin-dependent cell-cell adhesion. We, herein, introduce a novel promising strategy for controlling and re-engineering the breast tumor microenvironment.
format Online
Article
Text
id pubmed-8171089
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-81710892021-06-03 N-cadherin mediates the migration of bone marrow-derived mesenchymal stem cells toward breast tumor cells Choi, Sanghyuk Yu, Jinyeong Kim, Wootak Park, Ki-Sook Theranostics Research Paper Rationale: Bone marrow-derived mesenchymal stem cells (BM-MSCs) recruited into breast tumors regulate the behavior of tumor cells via various mechanisms and affect clinical outcomes. Although signaling molecules, such as transforming growth factor β (TGF-β), are known to transmit signals between BM-MSCs and breast tumor cells for recruiting BM-MSCs, it is unclear which specific intrinsic molecules involved in cell motility mediate the migration of BM-MSCs into breast tumor. It is also unclear as to how specific intrinsic molecules contribute to the migration. Methods: Conditioned medium (CM) from breast tumor cells (MCF-7 and MDA-MB-231) that simulates breast tumor secreting TGF-β was used to examine the migration of BM-MSCs into breast tumors. A three-dimensional migration assay was performed to investigate the collective migration of BM-MSCs, maintaining cell-cell adhesion, toward breast tumor cells. Results: N-cadherin formed adherens junction-like structures on the intercellular borders of BM-MSCs, and TGF-β increased the expression of N-cadherin on these borders. Knockdown of Smad4 impaired the TGF-β-mediated increase in N-cadherin expression in BM-MSCs, but inhibitors of non-canonical TGF-β pathways, such as extracellular signal‐regulated kinases, Akt, and p38, did not affect it. siRNA-mediated knockdown of N-cadherin and Smad4 impaired the migration of BM-MSCs in response to TGF-β. Conditioned medium from breast tumor cells also enhanced the expression of N-cadherin in BM-MSCs, but inactivation of TGF-β type 1 receptor (TGFBR1) with SB505124 and TGFBR1 knockdown abolished the increase in N-cadherin expression. BM-MSCs collectively migrated toward CM from MDA-MB-231 in vitro while maintaining cell-cell adhesion through N-cadherin. Knockdown of N-cadherin abolished the migration of BM-MSCs toward the CM from breast tumor cells. Conclusion: In the present study, we identified N-cadherin, an intrinsic transmembrane molecule in adherens junction-like structures, on BM-MSCs as a mediator for the migration of these cells toward breast tumor. The expression of N-cadherin increases on the intercellular borders of BM-MSCs through the TGF-β canonical signaling and they collectively migrate in response to breast tumor cells expressing TGF-β via N-cadherin-dependent cell-cell adhesion. We, herein, introduce a novel promising strategy for controlling and re-engineering the breast tumor microenvironment. Ivyspring International Publisher 2021-05-03 /pmc/articles/PMC8171089/ /pubmed/34093853 http://dx.doi.org/10.7150/thno.59703 Text en © The author(s) https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Choi, Sanghyuk
Yu, Jinyeong
Kim, Wootak
Park, Ki-Sook
N-cadherin mediates the migration of bone marrow-derived mesenchymal stem cells toward breast tumor cells
title N-cadherin mediates the migration of bone marrow-derived mesenchymal stem cells toward breast tumor cells
title_full N-cadherin mediates the migration of bone marrow-derived mesenchymal stem cells toward breast tumor cells
title_fullStr N-cadherin mediates the migration of bone marrow-derived mesenchymal stem cells toward breast tumor cells
title_full_unstemmed N-cadherin mediates the migration of bone marrow-derived mesenchymal stem cells toward breast tumor cells
title_short N-cadherin mediates the migration of bone marrow-derived mesenchymal stem cells toward breast tumor cells
title_sort n-cadherin mediates the migration of bone marrow-derived mesenchymal stem cells toward breast tumor cells
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8171089/
https://www.ncbi.nlm.nih.gov/pubmed/34093853
http://dx.doi.org/10.7150/thno.59703
work_keys_str_mv AT choisanghyuk ncadherinmediatesthemigrationofbonemarrowderivedmesenchymalstemcellstowardbreasttumorcells
AT yujinyeong ncadherinmediatesthemigrationofbonemarrowderivedmesenchymalstemcellstowardbreasttumorcells
AT kimwootak ncadherinmediatesthemigrationofbonemarrowderivedmesenchymalstemcellstowardbreasttumorcells
AT parkkisook ncadherinmediatesthemigrationofbonemarrowderivedmesenchymalstemcellstowardbreasttumorcells