Modulated TRPC1 expression predicts sensitivity of breast cancer to doxorubicin and magnetic field therapy: segue towards a precision medicine approach

Chemotherapy is the mainstream treatment modality for invasive breast cancer. Unfortunately, chemotherapy-associated adverse events can result in early termination of treatment. Paradoxical effects of chemotherapy are also sometimes observed, whereby prolonged exposure to high doses of chemotherapeu...

Full description

Saved in:
Bibliographic Details
Main Authors: Tai, Yee Kit, Chan, Karen Ka Wing, Fong, Charlene Hui Hua, Ramanan, Sharanya, Yap, Jasmine Lye Yee, Yin, Jocelyn Naixin, Yip, Yun Sheng, Tan, Wei Ren, Koh, Angele Pei Fern, Tan, Nguan Soon, Chan, Ching Wan, Huang, Ruby Yun Ju, Li, Jing Ze, Fröhlich, Jürg, Franco-Obregón, Alfredo
Other Authors: Lee Kong Chian School of Medicine (LKCMedicine)
Format: Article
Language:English
Published: 2022
Subjects:
Online Access:https://hdl.handle.net/10356/163189
Tags: Add Tag
No Tags, Be the first to tag this record!
Institution: Nanyang Technological University
Language: English
id sg-ntu-dr.10356-163189
record_format dspace
institution Nanyang Technological University
building NTU Library
continent Asia
country Singapore
Singapore
content_provider NTU Library
collection DR-NTU
language English
topic Science::Medicine
Breast Cancer
Patient-Derived Xenograft
spellingShingle Science::Medicine
Breast Cancer
Patient-Derived Xenograft
Tai, Yee Kit
Chan, Karen Ka Wing
Fong, Charlene Hui Hua
Ramanan, Sharanya
Yap, Jasmine Lye Yee
Yin, Jocelyn Naixin
Yip, Yun Sheng
Tan, Wei Ren
Koh, Angele Pei Fern
Tan, Nguan Soon
Chan, Ching Wan
Huang, Ruby Yun Ju
Li, Jing Ze
Fröhlich, Jürg
Franco-Obregón, Alfredo
Modulated TRPC1 expression predicts sensitivity of breast cancer to doxorubicin and magnetic field therapy: segue towards a precision medicine approach
description Chemotherapy is the mainstream treatment modality for invasive breast cancer. Unfortunately, chemotherapy-associated adverse events can result in early termination of treatment. Paradoxical effects of chemotherapy are also sometimes observed, whereby prolonged exposure to high doses of chemotherapeutic agents results in malignant states resistant to chemotherapy. In this study, potential synergism between doxorubicin (DOX) and pulsed electromagnetic field (PEMF) therapy was investigated in: 1) MCF-7 and MDA-MB-231 cells in vitro; 2) MCF-7 tumors implanted onto a chicken chorioallantoic membrane (CAM) and; 3) human patient-derived and MCF-7 and MDA-MB-231 breast cancer xenografts implanted into NOD-SCID gamma (NSG) mice. In vivo, synergism was observed in patient-derived and breast cancer cell line xenograft mouse models, wherein PEMF exposure and DOX administration individually reduced tumor size and increased apoptosis and could be augmented by combined treatments. In the CAM xenograft model, DOX and PEMF exposure also synergistically reduced tumor size as well as reduced Transient Receptor Potential Canonical 1 (TRPC1) channel expression. In vitro, PEMF exposure alone impaired the survival of MCF-7 and MDA-MB-231 cells, but not that of non-malignant MCF10A breast cells; the selective vulnerability of breast cancer cells to PEMF exposure was corroborated in human tumor biopsy samples. Stable overexpression of TRPC1 enhanced the vulnerability of MCF-7 cells to both DOX and PEMF exposure and promoted proliferation, whereas TRPC1 genetic silencing reduced sensitivity to both DOX and PEMF treatments and mitigated proliferation. Chronic exposure to DOX depressed TRPC1 expression, proliferation, and responses to both PEMF exposure and DOX in a manner that was reversible upon removal of DOX. TRPC1 channel overexpression and silencing positively correlated with markers of epithelial-mesenchymal transition (EMT), including SLUG, SNAIL, VIMENTIN, and E-CADHERIN, indicating increased and decreased EMT, respectively. Finally, PEMF exposure was shown to attenuate the invasiveness of MCF-7 cells in correlation with TRPC1 expression. We thus demonstrate that the expression levels of TRPC1 consistently predicted breast cancer sensitivity to DOX and PEMF interventions and positively correlated to EMT status, providing an initial rationale for the use of PEMF-based therapies as an adjuvant to DOX chemotherapy for the treatment of breast cancers characterized by elevated TRPC1 expression levels.
author2 Lee Kong Chian School of Medicine (LKCMedicine)
author_facet Lee Kong Chian School of Medicine (LKCMedicine)
Tai, Yee Kit
Chan, Karen Ka Wing
Fong, Charlene Hui Hua
Ramanan, Sharanya
Yap, Jasmine Lye Yee
Yin, Jocelyn Naixin
Yip, Yun Sheng
Tan, Wei Ren
Koh, Angele Pei Fern
Tan, Nguan Soon
Chan, Ching Wan
Huang, Ruby Yun Ju
Li, Jing Ze
Fröhlich, Jürg
Franco-Obregón, Alfredo
format Article
author Tai, Yee Kit
Chan, Karen Ka Wing
Fong, Charlene Hui Hua
Ramanan, Sharanya
Yap, Jasmine Lye Yee
Yin, Jocelyn Naixin
Yip, Yun Sheng
Tan, Wei Ren
Koh, Angele Pei Fern
Tan, Nguan Soon
Chan, Ching Wan
Huang, Ruby Yun Ju
Li, Jing Ze
Fröhlich, Jürg
Franco-Obregón, Alfredo
author_sort Tai, Yee Kit
title Modulated TRPC1 expression predicts sensitivity of breast cancer to doxorubicin and magnetic field therapy: segue towards a precision medicine approach
title_short Modulated TRPC1 expression predicts sensitivity of breast cancer to doxorubicin and magnetic field therapy: segue towards a precision medicine approach
title_full Modulated TRPC1 expression predicts sensitivity of breast cancer to doxorubicin and magnetic field therapy: segue towards a precision medicine approach
title_fullStr Modulated TRPC1 expression predicts sensitivity of breast cancer to doxorubicin and magnetic field therapy: segue towards a precision medicine approach
title_full_unstemmed Modulated TRPC1 expression predicts sensitivity of breast cancer to doxorubicin and magnetic field therapy: segue towards a precision medicine approach
title_sort modulated trpc1 expression predicts sensitivity of breast cancer to doxorubicin and magnetic field therapy: segue towards a precision medicine approach
publishDate 2022
url https://hdl.handle.net/10356/163189
_version_ 1759857094107856896
spelling sg-ntu-dr.10356-1631892023-02-28T17:12:33Z Modulated TRPC1 expression predicts sensitivity of breast cancer to doxorubicin and magnetic field therapy: segue towards a precision medicine approach Tai, Yee Kit Chan, Karen Ka Wing Fong, Charlene Hui Hua Ramanan, Sharanya Yap, Jasmine Lye Yee Yin, Jocelyn Naixin Yip, Yun Sheng Tan, Wei Ren Koh, Angele Pei Fern Tan, Nguan Soon Chan, Ching Wan Huang, Ruby Yun Ju Li, Jing Ze Fröhlich, Jürg Franco-Obregón, Alfredo Lee Kong Chian School of Medicine (LKCMedicine) School of Biological Sciences Science::Medicine Breast Cancer Patient-Derived Xenograft Chemotherapy is the mainstream treatment modality for invasive breast cancer. Unfortunately, chemotherapy-associated adverse events can result in early termination of treatment. Paradoxical effects of chemotherapy are also sometimes observed, whereby prolonged exposure to high doses of chemotherapeutic agents results in malignant states resistant to chemotherapy. In this study, potential synergism between doxorubicin (DOX) and pulsed electromagnetic field (PEMF) therapy was investigated in: 1) MCF-7 and MDA-MB-231 cells in vitro; 2) MCF-7 tumors implanted onto a chicken chorioallantoic membrane (CAM) and; 3) human patient-derived and MCF-7 and MDA-MB-231 breast cancer xenografts implanted into NOD-SCID gamma (NSG) mice. In vivo, synergism was observed in patient-derived and breast cancer cell line xenograft mouse models, wherein PEMF exposure and DOX administration individually reduced tumor size and increased apoptosis and could be augmented by combined treatments. In the CAM xenograft model, DOX and PEMF exposure also synergistically reduced tumor size as well as reduced Transient Receptor Potential Canonical 1 (TRPC1) channel expression. In vitro, PEMF exposure alone impaired the survival of MCF-7 and MDA-MB-231 cells, but not that of non-malignant MCF10A breast cells; the selective vulnerability of breast cancer cells to PEMF exposure was corroborated in human tumor biopsy samples. Stable overexpression of TRPC1 enhanced the vulnerability of MCF-7 cells to both DOX and PEMF exposure and promoted proliferation, whereas TRPC1 genetic silencing reduced sensitivity to both DOX and PEMF treatments and mitigated proliferation. Chronic exposure to DOX depressed TRPC1 expression, proliferation, and responses to both PEMF exposure and DOX in a manner that was reversible upon removal of DOX. TRPC1 channel overexpression and silencing positively correlated with markers of epithelial-mesenchymal transition (EMT), including SLUG, SNAIL, VIMENTIN, and E-CADHERIN, indicating increased and decreased EMT, respectively. Finally, PEMF exposure was shown to attenuate the invasiveness of MCF-7 cells in correlation with TRPC1 expression. We thus demonstrate that the expression levels of TRPC1 consistently predicted breast cancer sensitivity to DOX and PEMF interventions and positively correlated to EMT status, providing an initial rationale for the use of PEMF-based therapies as an adjuvant to DOX chemotherapy for the treatment of breast cancers characterized by elevated TRPC1 expression levels. Published version This work is supported by Lee Kong Chian MedTech Initiative, Singapore (N-176-000-045-001), SMART Ignition Grant (R-176-000-206-592) and the Institute for Health Innovation & Technology, iHealthtech, at the National University of Singapore. The publication cost of this article is funded by Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore. 2022-11-28T07:05:54Z 2022-11-28T07:05:54Z 2022 Journal Article Tai, Y. K., Chan, K. K. W., Fong, C. H. H., Ramanan, S., Yap, J. L. Y., Yin, J. N., Yip, Y. S., Tan, W. R., Koh, A. P. F., Tan, N. S., Chan, C. W., Huang, R. Y. J., Li, J. Z., Fröhlich, J. & Franco-Obregón, A. (2022). Modulated TRPC1 expression predicts sensitivity of breast cancer to doxorubicin and magnetic field therapy: segue towards a precision medicine approach. Frontiers in Oncology, 11, 783803-. https://dx.doi.org/10.3389/fonc.2021.783803 2234-943X https://hdl.handle.net/10356/163189 10.3389/fonc.2021.783803 35141145 2-s2.0-85124221315 11 783803 en N-176-000-045-001 R-176-000-206-592 Frontiers in Oncology © 2022 Tai, Chan, Fong, Ramanan, Yap, Yin, Yip, Tan, Koh, Tan, Chan, Huang, Li, Fröhlich and Franco-Obregon. This is an open-access article distributed ́ under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms. application/pdf