Cytidine monophosphate N-acetylneuraminic acid synthetase enhances invasion of human triple-negative breast cancer cells

Elizabeth M O’Day,1,2 Greg E Idos,3 Collin Hill,4 Joan W Chen,5 Gerhard Wagner1 1Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; 2Olaris Therapeutics, Cambridge, MA 02138, USA; 3Division of Gastroenterology and Liver Disease, K...

Full description

Bibliographic Details
Main Authors: O'Day EM, Idos GE, Hill C, Chen JW, Wagner G
Format: Article
Language:English
Published: Dove Medical Press 2018-10-01
Series:OncoTargets and Therapy
Subjects:
Online Access:https://www.dovepress.com/cytidine-monophosphate-n-acetylneuraminic-acid-synthetase-enhances-inv-peer-reviewed-article-OTT
id doaj-8890dff34e78442396adeabfbf95f32d
record_format Article
spelling doaj-8890dff34e78442396adeabfbf95f32d2020-11-25T00:33:39ZengDove Medical PressOncoTargets and Therapy1178-69302018-10-01Volume 116827683841363Cytidine monophosphate N-acetylneuraminic acid synthetase enhances invasion of human triple-negative breast cancer cellsO'Day EMIdos GEHill CChen JWWagner GElizabeth M O’Day,1,2 Greg E Idos,3 Collin Hill,4 Joan W Chen,5 Gerhard Wagner1 1Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; 2Olaris Therapeutics, Cambridge, MA 02138, USA; 3Division of Gastroenterology and Liver Disease, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA; 4PerkinElmer, Waltham, MA 02451, USA; 5Rancho Biosciences, San Diego, CA 92127, USA Background: Cancer cells have altered bioenergetics, which contributes to their ability to proliferate, survive in unusual microenvironments, and invade other tissues. Changes in glucose metabolism can have pleomorphic effects on tumor cells. Methods: To investigate potential mechanisms responsible for the increased malignancy associated with altered glucose metabolism, we used an unbiased nuclear magnetic resonance spectroscopy screening method to identify glucose metabolites differentially produced in a highly malignant human triple-negative breast cancer (TNBC) cell line (BPLER) and a less malignant isogenic TNBC cell line (HMLER). Results: N-acetylneuraminic acid (Neu5Ac), the predominant sialic acid derivative in mammalian cells, which forms the terminal sugar on mucinous cell surface glycoproteins, was the major glucose metabolite that differed. Neu5Ac was ~7-fold more abundant in BPLER than HMLER. Loss of Neu5Ac by enzymatic removal or siRNA knockdown of cytidine monophosphate N-acetylneuraminic acid synthetase (CMAS), which activates cellular sialic acids for glycoprotein conjugation, had no significant effect on cell proliferation, but decreased the ability of BPLER to invade through a basement membrane. Conversely, overexpressing CMAS in HMLER increased invasivity. TNBCs in The Cancer Genome Atlas also had significantly more CMAS copy number variations and higher mRNA expression than non-TNBC, which have a better prognosis. CMAS knockdown in BPLER ex vivo blocked xenograft formation in mice. Conclusion: Neu5Ac is selectively highly enriched in aggressive TNBC, and CMAS, the enzyme required for sialylation, may play an important role in TNBC tumor formation and invasivity. Keywords: triple-negative breast cancer, metabolism, invasion, cytidine monophosphate N-acetylneuraminic acid, sialic acid, NMR spectroscopy, metabolomicshttps://www.dovepress.com/cytidine-monophosphate-n-acetylneuraminic-acid-synthetase-enhances-inv-peer-reviewed-article-OTTTriple negative breast cancer (TNBC)metabolisminvasionCytidine monophosphate N-acetylneuraminic acid (CMAS)N-acetylneuraminic acid (Neu5Ac)sialic acid.
collection DOAJ
language English
format Article
sources DOAJ
author O'Day EM
Idos GE
Hill C
Chen JW
Wagner G
spellingShingle O'Day EM
Idos GE
Hill C
Chen JW
Wagner G
Cytidine monophosphate N-acetylneuraminic acid synthetase enhances invasion of human triple-negative breast cancer cells
OncoTargets and Therapy
Triple negative breast cancer (TNBC)
metabolism
invasion
Cytidine monophosphate N-acetylneuraminic acid (CMAS)
N-acetylneuraminic acid (Neu5Ac)
sialic acid.
author_facet O'Day EM
Idos GE
Hill C
Chen JW
Wagner G
author_sort O'Day EM
title Cytidine monophosphate N-acetylneuraminic acid synthetase enhances invasion of human triple-negative breast cancer cells
title_short Cytidine monophosphate N-acetylneuraminic acid synthetase enhances invasion of human triple-negative breast cancer cells
title_full Cytidine monophosphate N-acetylneuraminic acid synthetase enhances invasion of human triple-negative breast cancer cells
title_fullStr Cytidine monophosphate N-acetylneuraminic acid synthetase enhances invasion of human triple-negative breast cancer cells
title_full_unstemmed Cytidine monophosphate N-acetylneuraminic acid synthetase enhances invasion of human triple-negative breast cancer cells
title_sort cytidine monophosphate n-acetylneuraminic acid synthetase enhances invasion of human triple-negative breast cancer cells
publisher Dove Medical Press
series OncoTargets and Therapy
issn 1178-6930
publishDate 2018-10-01
description Elizabeth M O’Day,1,2 Greg E Idos,3 Collin Hill,4 Joan W Chen,5 Gerhard Wagner1 1Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA; 2Olaris Therapeutics, Cambridge, MA 02138, USA; 3Division of Gastroenterology and Liver Disease, Keck School of Medicine of University of Southern California, Los Angeles, CA 90033, USA; 4PerkinElmer, Waltham, MA 02451, USA; 5Rancho Biosciences, San Diego, CA 92127, USA Background: Cancer cells have altered bioenergetics, which contributes to their ability to proliferate, survive in unusual microenvironments, and invade other tissues. Changes in glucose metabolism can have pleomorphic effects on tumor cells. Methods: To investigate potential mechanisms responsible for the increased malignancy associated with altered glucose metabolism, we used an unbiased nuclear magnetic resonance spectroscopy screening method to identify glucose metabolites differentially produced in a highly malignant human triple-negative breast cancer (TNBC) cell line (BPLER) and a less malignant isogenic TNBC cell line (HMLER). Results: N-acetylneuraminic acid (Neu5Ac), the predominant sialic acid derivative in mammalian cells, which forms the terminal sugar on mucinous cell surface glycoproteins, was the major glucose metabolite that differed. Neu5Ac was ~7-fold more abundant in BPLER than HMLER. Loss of Neu5Ac by enzymatic removal or siRNA knockdown of cytidine monophosphate N-acetylneuraminic acid synthetase (CMAS), which activates cellular sialic acids for glycoprotein conjugation, had no significant effect on cell proliferation, but decreased the ability of BPLER to invade through a basement membrane. Conversely, overexpressing CMAS in HMLER increased invasivity. TNBCs in The Cancer Genome Atlas also had significantly more CMAS copy number variations and higher mRNA expression than non-TNBC, which have a better prognosis. CMAS knockdown in BPLER ex vivo blocked xenograft formation in mice. Conclusion: Neu5Ac is selectively highly enriched in aggressive TNBC, and CMAS, the enzyme required for sialylation, may play an important role in TNBC tumor formation and invasivity. Keywords: triple-negative breast cancer, metabolism, invasion, cytidine monophosphate N-acetylneuraminic acid, sialic acid, NMR spectroscopy, metabolomics
topic Triple negative breast cancer (TNBC)
metabolism
invasion
Cytidine monophosphate N-acetylneuraminic acid (CMAS)
N-acetylneuraminic acid (Neu5Ac)
sialic acid.
url https://www.dovepress.com/cytidine-monophosphate-n-acetylneuraminic-acid-synthetase-enhances-inv-peer-reviewed-article-OTT
work_keys_str_mv AT odayem cytidinemonophosphatenacetylneuraminicacidsynthetaseenhancesinvasionofhumantriplenegativebreastcancercells
AT idosge cytidinemonophosphatenacetylneuraminicacidsynthetaseenhancesinvasionofhumantriplenegativebreastcancercells
AT hillc cytidinemonophosphatenacetylneuraminicacidsynthetaseenhancesinvasionofhumantriplenegativebreastcancercells
AT chenjw cytidinemonophosphatenacetylneuraminicacidsynthetaseenhancesinvasionofhumantriplenegativebreastcancercells
AT wagnerg cytidinemonophosphatenacetylneuraminicacidsynthetaseenhancesinvasionofhumantriplenegativebreastcancercells
_version_ 1725315585104936960