The extracellular matrix supports breast cancer cell growth under amino acid starvation by promoting tyrosine catabolism.

Breast tumours are embedded in a collagen I-rich extracellular matrix (ECM) network, where nutrients are scarce due to limited blood flow and elevated tumour growth. Metabolic adaptation is required for cancer cells to endure these conditions. Here, we demonstrated that the presence of ECM supported...

Full description

Bibliographic Details
Main Authors: Mona Nazemi, Bian Yanes, Montserrat Llanses Martinez, Heather J Walker, Khoa Pham, Mark O Collins, Frederic Bard, Elena Rainero
Format: Article
Language:English
Published: Public Library of Science (PLoS) 2024-01-01
Series:PLoS Biology
Online Access:https://journals.plos.org/plosbiology/article/file?id=10.1371/journal.pbio.3002406&type=printable
_version_ 1797315919561621504
author Mona Nazemi
Bian Yanes
Montserrat Llanses Martinez
Heather J Walker
Khoa Pham
Mark O Collins
Frederic Bard
Elena Rainero
author_facet Mona Nazemi
Bian Yanes
Montserrat Llanses Martinez
Heather J Walker
Khoa Pham
Mark O Collins
Frederic Bard
Elena Rainero
author_sort Mona Nazemi
collection DOAJ
description Breast tumours are embedded in a collagen I-rich extracellular matrix (ECM) network, where nutrients are scarce due to limited blood flow and elevated tumour growth. Metabolic adaptation is required for cancer cells to endure these conditions. Here, we demonstrated that the presence of ECM supported the growth of invasive breast cancer cells, but not non-transformed mammary epithelial cells, under amino acid starvation, through a mechanism that required macropinocytosis-dependent ECM uptake. Importantly, we showed that this behaviour was acquired during carcinoma progression. ECM internalisation, followed by lysosomal degradation, contributed to the up-regulation of the intracellular levels of several amino acids, most notably tyrosine and phenylalanine. This resulted in elevated tyrosine catabolism on ECM under starvation, leading to increased fumarate levels, potentially feeding into the tricarboxylic acid (TCA) cycle. Interestingly, this pathway was required for ECM-dependent cell growth and invasive cell migration under amino acid starvation, as the knockdown of p-hydroxyphenylpyruvate hydroxylase-like protein (HPDL), the third enzyme of the pathway, opposed cell growth and motility on ECM in both 2D and 3D systems, without affecting cell proliferation on plastic. Finally, high HPDL expression correlated with poor prognosis in breast cancer patients. Collectively, our results highlight that the ECM in the tumour microenvironment (TME) represents an alternative source of nutrients to support cancer cell growth by regulating phenylalanine and tyrosine metabolism.
first_indexed 2024-03-08T03:11:47Z
format Article
id doaj.art-73bd8338597b4be8864562ecae43c95e
institution Directory Open Access Journal
issn 1544-9173
1545-7885
language English
last_indexed 2024-03-08T03:11:47Z
publishDate 2024-01-01
publisher Public Library of Science (PLoS)
record_format Article
series PLoS Biology
spelling doaj.art-73bd8338597b4be8864562ecae43c95e2024-02-13T05:30:47ZengPublic Library of Science (PLoS)PLoS Biology1544-91731545-78852024-01-01221e300240610.1371/journal.pbio.3002406The extracellular matrix supports breast cancer cell growth under amino acid starvation by promoting tyrosine catabolism.Mona NazemiBian YanesMontserrat Llanses MartinezHeather J WalkerKhoa PhamMark O CollinsFrederic BardElena RaineroBreast tumours are embedded in a collagen I-rich extracellular matrix (ECM) network, where nutrients are scarce due to limited blood flow and elevated tumour growth. Metabolic adaptation is required for cancer cells to endure these conditions. Here, we demonstrated that the presence of ECM supported the growth of invasive breast cancer cells, but not non-transformed mammary epithelial cells, under amino acid starvation, through a mechanism that required macropinocytosis-dependent ECM uptake. Importantly, we showed that this behaviour was acquired during carcinoma progression. ECM internalisation, followed by lysosomal degradation, contributed to the up-regulation of the intracellular levels of several amino acids, most notably tyrosine and phenylalanine. This resulted in elevated tyrosine catabolism on ECM under starvation, leading to increased fumarate levels, potentially feeding into the tricarboxylic acid (TCA) cycle. Interestingly, this pathway was required for ECM-dependent cell growth and invasive cell migration under amino acid starvation, as the knockdown of p-hydroxyphenylpyruvate hydroxylase-like protein (HPDL), the third enzyme of the pathway, opposed cell growth and motility on ECM in both 2D and 3D systems, without affecting cell proliferation on plastic. Finally, high HPDL expression correlated with poor prognosis in breast cancer patients. Collectively, our results highlight that the ECM in the tumour microenvironment (TME) represents an alternative source of nutrients to support cancer cell growth by regulating phenylalanine and tyrosine metabolism.https://journals.plos.org/plosbiology/article/file?id=10.1371/journal.pbio.3002406&type=printable
spellingShingle Mona Nazemi
Bian Yanes
Montserrat Llanses Martinez
Heather J Walker
Khoa Pham
Mark O Collins
Frederic Bard
Elena Rainero
The extracellular matrix supports breast cancer cell growth under amino acid starvation by promoting tyrosine catabolism.
PLoS Biology
title The extracellular matrix supports breast cancer cell growth under amino acid starvation by promoting tyrosine catabolism.
title_full The extracellular matrix supports breast cancer cell growth under amino acid starvation by promoting tyrosine catabolism.
title_fullStr The extracellular matrix supports breast cancer cell growth under amino acid starvation by promoting tyrosine catabolism.
title_full_unstemmed The extracellular matrix supports breast cancer cell growth under amino acid starvation by promoting tyrosine catabolism.
title_short The extracellular matrix supports breast cancer cell growth under amino acid starvation by promoting tyrosine catabolism.
title_sort extracellular matrix supports breast cancer cell growth under amino acid starvation by promoting tyrosine catabolism
url https://journals.plos.org/plosbiology/article/file?id=10.1371/journal.pbio.3002406&type=printable
work_keys_str_mv AT monanazemi theextracellularmatrixsupportsbreastcancercellgrowthunderaminoacidstarvationbypromotingtyrosinecatabolism
AT bianyanes theextracellularmatrixsupportsbreastcancercellgrowthunderaminoacidstarvationbypromotingtyrosinecatabolism
AT montserratllansesmartinez theextracellularmatrixsupportsbreastcancercellgrowthunderaminoacidstarvationbypromotingtyrosinecatabolism
AT heatherjwalker theextracellularmatrixsupportsbreastcancercellgrowthunderaminoacidstarvationbypromotingtyrosinecatabolism
AT khoapham theextracellularmatrixsupportsbreastcancercellgrowthunderaminoacidstarvationbypromotingtyrosinecatabolism
AT markocollins theextracellularmatrixsupportsbreastcancercellgrowthunderaminoacidstarvationbypromotingtyrosinecatabolism
AT fredericbard theextracellularmatrixsupportsbreastcancercellgrowthunderaminoacidstarvationbypromotingtyrosinecatabolism
AT elenarainero theextracellularmatrixsupportsbreastcancercellgrowthunderaminoacidstarvationbypromotingtyrosinecatabolism
AT monanazemi extracellularmatrixsupportsbreastcancercellgrowthunderaminoacidstarvationbypromotingtyrosinecatabolism
AT bianyanes extracellularmatrixsupportsbreastcancercellgrowthunderaminoacidstarvationbypromotingtyrosinecatabolism
AT montserratllansesmartinez extracellularmatrixsupportsbreastcancercellgrowthunderaminoacidstarvationbypromotingtyrosinecatabolism
AT heatherjwalker extracellularmatrixsupportsbreastcancercellgrowthunderaminoacidstarvationbypromotingtyrosinecatabolism
AT khoapham extracellularmatrixsupportsbreastcancercellgrowthunderaminoacidstarvationbypromotingtyrosinecatabolism
AT markocollins extracellularmatrixsupportsbreastcancercellgrowthunderaminoacidstarvationbypromotingtyrosinecatabolism
AT fredericbard extracellularmatrixsupportsbreastcancercellgrowthunderaminoacidstarvationbypromotingtyrosinecatabolism
AT elenarainero extracellularmatrixsupportsbreastcancercellgrowthunderaminoacidstarvationbypromotingtyrosinecatabolism