The University of Southampton
University of Southampton Institutional Repository

Lysine methyltransferase 2D regulates pancreatic carcinogenesis through metabolic reprogramming

Lysine methyltransferase 2D regulates pancreatic carcinogenesis through metabolic reprogramming
Lysine methyltransferase 2D regulates pancreatic carcinogenesis through metabolic reprogramming

Objective: Despite advances in the identification of epigenetic alterations in pancreatic cancer, their biological roles in the pathobiology of this dismal neoplasm remain elusive. Here, we aimed to characterise the functional significance of histone lysine methyltransferases (KMTs) and demethylases (KDMs) in pancreatic tumourigenesis. Design: DNA methylation sequencing and gene expression microarrays were employed to investigate CpG methylation and expression patterns of KMTs and KDMs in pancreatic cancer tissues versus normal tissues. Gene expression was assessed in five cohorts of patients by reverse transcription quantitative-PCR. Molecular analysis and functional assays were conducted in genetically modified cell lines. Cellular metabolic rates were measured using an XF24-3 Analyzer, while quantitative evaluation of lipids was performed by liquid chromatography-mass spectrometry (LC-MS) analysis. Subcutaneous xenograft mouse models were used to evaluate pancreatic tumour growth in vivo. Results: We define a new antitumorous function of the histone lysine (K)-specific methyltransferase 2D (KMT2D) in pancreatic cancer. KMT2D is transcriptionally repressed in human pancreatic tumours through DNA methylation. Clinically, lower levels of this methyltransferase associate with poor prognosis and significant weight alterations. RNAi-based genetic inactivation of KMT2D promotes tumour growth and results in loss of H3K4me3 mark. In addition, KMT2D inhibition increases aerobic glycolysis and alters the lipidomic profiles of pancreatic cancer cells. Further analysis of this phenomenon identified the glucose transporter SLC2A3 as a mediator of KMT2D-induced changes in cellular, metabolic and proliferative rates. Conclusion: Together our findings define a new tumour suppressor function of KMT2D through the regulation of glucose/fatty acid metabolism in pancreatic cancer.

gene regulation, molecular oncology, pancreatic cancer
0017-5749
Koutsioumpa, Marina
23caa6e6-2a7f-475f-8bb2-5c54ca2b1efe
Hatziapostolou, Maria
c210f495-e959-4813-a530-6a316f0b221a
Polytarchou, Christos
82cbf33a-b136-49b0-ba59-90d5f30586e5
Tolosa, Ezequiel J.
8aaa4a15-e510-4b71-bd5e-605bf0bc78f9
Almada, Luciana L.
a2570d09-b6be-4476-8d1e-a1831174e343
Mahurkar-Joshi, Swapna
d23ee691-7f1b-4787-902e-3edd92aa8148
Williams, Jennifer
80d203da-2d6c-4d37-9177-ce58df412082
Tirado-Rodriguez, Ana Belen
24ca2a12-0ef5-4059-8c8c-c66f200be550
Huerta-Yepez, Sara
6fff097d-f0ba-410b-aab1-f9bfdd022532
Karavias, Dimitrios
ec27bb13-8e06-45f2-9ea0-2dc5b4c45fce
Kourea, Helen
dbab8942-6599-4bab-95f2-d317855e2b73
Poultsides, George A.
d336cd2f-1a89-488f-b9c3-5782f65b4c36
Struhl, Kevin
c0f05d3b-32d2-4d18-ad83-9dd5c331e7c0
Dawson, David W.
4b0d64bb-f17e-4f1c-9277-9f37b00a8e8a
Donahue, Timothy R.
6ffc15e5-1565-4f16-b839-9d2a97520b8c
Fernández-Zapico, Martín E.
38c289a2-64fc-48d0-812f-eae4c9ca9aab
Iliopoulos, Dimitrios
ed053f0c-6049-4b9c-9984-0a7367bf6d61
Koutsioumpa, Marina
23caa6e6-2a7f-475f-8bb2-5c54ca2b1efe
Hatziapostolou, Maria
c210f495-e959-4813-a530-6a316f0b221a
Polytarchou, Christos
82cbf33a-b136-49b0-ba59-90d5f30586e5
Tolosa, Ezequiel J.
8aaa4a15-e510-4b71-bd5e-605bf0bc78f9
Almada, Luciana L.
a2570d09-b6be-4476-8d1e-a1831174e343
Mahurkar-Joshi, Swapna
d23ee691-7f1b-4787-902e-3edd92aa8148
Williams, Jennifer
80d203da-2d6c-4d37-9177-ce58df412082
Tirado-Rodriguez, Ana Belen
24ca2a12-0ef5-4059-8c8c-c66f200be550
Huerta-Yepez, Sara
6fff097d-f0ba-410b-aab1-f9bfdd022532
Karavias, Dimitrios
ec27bb13-8e06-45f2-9ea0-2dc5b4c45fce
Kourea, Helen
dbab8942-6599-4bab-95f2-d317855e2b73
Poultsides, George A.
d336cd2f-1a89-488f-b9c3-5782f65b4c36
Struhl, Kevin
c0f05d3b-32d2-4d18-ad83-9dd5c331e7c0
Dawson, David W.
4b0d64bb-f17e-4f1c-9277-9f37b00a8e8a
Donahue, Timothy R.
6ffc15e5-1565-4f16-b839-9d2a97520b8c
Fernández-Zapico, Martín E.
38c289a2-64fc-48d0-812f-eae4c9ca9aab
Iliopoulos, Dimitrios
ed053f0c-6049-4b9c-9984-0a7367bf6d61

Koutsioumpa, Marina, Hatziapostolou, Maria, Polytarchou, Christos, Tolosa, Ezequiel J., Almada, Luciana L., Mahurkar-Joshi, Swapna, Williams, Jennifer, Tirado-Rodriguez, Ana Belen, Huerta-Yepez, Sara, Karavias, Dimitrios, Kourea, Helen, Poultsides, George A., Struhl, Kevin, Dawson, David W., Donahue, Timothy R., Fernández-Zapico, Martín E. and Iliopoulos, Dimitrios (2018) Lysine methyltransferase 2D regulates pancreatic carcinogenesis through metabolic reprogramming. Gut. (doi:10.1136/gutjnl-2017-315690).

Record type: Article

Abstract

Objective: Despite advances in the identification of epigenetic alterations in pancreatic cancer, their biological roles in the pathobiology of this dismal neoplasm remain elusive. Here, we aimed to characterise the functional significance of histone lysine methyltransferases (KMTs) and demethylases (KDMs) in pancreatic tumourigenesis. Design: DNA methylation sequencing and gene expression microarrays were employed to investigate CpG methylation and expression patterns of KMTs and KDMs in pancreatic cancer tissues versus normal tissues. Gene expression was assessed in five cohorts of patients by reverse transcription quantitative-PCR. Molecular analysis and functional assays were conducted in genetically modified cell lines. Cellular metabolic rates were measured using an XF24-3 Analyzer, while quantitative evaluation of lipids was performed by liquid chromatography-mass spectrometry (LC-MS) analysis. Subcutaneous xenograft mouse models were used to evaluate pancreatic tumour growth in vivo. Results: We define a new antitumorous function of the histone lysine (K)-specific methyltransferase 2D (KMT2D) in pancreatic cancer. KMT2D is transcriptionally repressed in human pancreatic tumours through DNA methylation. Clinically, lower levels of this methyltransferase associate with poor prognosis and significant weight alterations. RNAi-based genetic inactivation of KMT2D promotes tumour growth and results in loss of H3K4me3 mark. In addition, KMT2D inhibition increases aerobic glycolysis and alters the lipidomic profiles of pancreatic cancer cells. Further analysis of this phenomenon identified the glucose transporter SLC2A3 as a mediator of KMT2D-induced changes in cellular, metabolic and proliferative rates. Conclusion: Together our findings define a new tumour suppressor function of KMT2D through the regulation of glucose/fatty acid metabolism in pancreatic cancer.

This record has no associated files available for download.

More information

Accepted/In Press date: 12 September 2018
e-pub ahead of print date: 18 October 2018
Keywords: gene regulation, molecular oncology, pancreatic cancer

Identifiers

Local EPrints ID: 427497
URI: http://eprints.soton.ac.uk/id/eprint/427497
ISSN: 0017-5749
PURE UUID: ec6be023-a50d-41f7-a067-d316bcdf673a

Catalogue record

Date deposited: 21 Jan 2019 17:30
Last modified: 15 Mar 2024 22:40

Export record

Altmetrics

Contributors

Author: Marina Koutsioumpa
Author: Maria Hatziapostolou
Author: Christos Polytarchou
Author: Ezequiel J. Tolosa
Author: Luciana L. Almada
Author: Swapna Mahurkar-Joshi
Author: Jennifer Williams
Author: Ana Belen Tirado-Rodriguez
Author: Sara Huerta-Yepez
Author: Dimitrios Karavias
Author: Helen Kourea
Author: George A. Poultsides
Author: Kevin Struhl
Author: David W. Dawson
Author: Timothy R. Donahue
Author: Martín E. Fernández-Zapico
Author: Dimitrios Iliopoulos

Download statistics

Downloads from ePrints over the past year. Other digital versions may also be available to download e.g. from the publisher's website.

View more statistics

Atom RSS 1.0 RSS 2.0

Contact ePrints Soton: eprints@soton.ac.uk

ePrints Soton supports OAI 2.0 with a base URL of http://eprints.soton.ac.uk/cgi/oai2

This repository has been built using EPrints software, developed at the University of Southampton, but available to everyone to use.

We use cookies to ensure that we give you the best experience on our website. If you continue without changing your settings, we will assume that you are happy to receive cookies on the University of Southampton website.

×