Cargando…

Liver cancer cell lines distinctly mimic the metabolic gene expression pattern of the corresponding human tumours

BACKGROUND: Although metabolism is profoundly altered in human liver cancer, the extent to which experimental models, e.g. cell lines, mimic those alterations is unresolved. Here, we aimed to determine the resemblance of hepatocellular carcinoma (HCC) cell lines to human liver tumours, specifically...

Descripción completa

Detalles Bibliográficos
Autores principales: Nwosu, Zeribe C., Battello, Nadia, Rothley, Melanie, Piorońska, Weronika, Sitek, Barbara, Ebert, Matthias P., Hofmann, Ute, Sleeman, Jonathan, Wölfl, Stefan, Meyer, Christoph, Megger, Dominik A., Dooley, Steven
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6122702/
https://www.ncbi.nlm.nih.gov/pubmed/30176945
http://dx.doi.org/10.1186/s13046-018-0872-6
_version_ 1783352706819162112
author Nwosu, Zeribe C.
Battello, Nadia
Rothley, Melanie
Piorońska, Weronika
Sitek, Barbara
Ebert, Matthias P.
Hofmann, Ute
Sleeman, Jonathan
Wölfl, Stefan
Meyer, Christoph
Megger, Dominik A.
Dooley, Steven
author_facet Nwosu, Zeribe C.
Battello, Nadia
Rothley, Melanie
Piorońska, Weronika
Sitek, Barbara
Ebert, Matthias P.
Hofmann, Ute
Sleeman, Jonathan
Wölfl, Stefan
Meyer, Christoph
Megger, Dominik A.
Dooley, Steven
author_sort Nwosu, Zeribe C.
collection PubMed
description BACKGROUND: Although metabolism is profoundly altered in human liver cancer, the extent to which experimental models, e.g. cell lines, mimic those alterations is unresolved. Here, we aimed to determine the resemblance of hepatocellular carcinoma (HCC) cell lines to human liver tumours, specifically in the expression of deregulated metabolic targets in clinical tissue samples. METHODS: We compared the overall gene expression profile of poorly-differentiated (HLE, HLF, SNU-449) to well-differentiated (HUH7, HEPG2, HEP3B) HCC cell lines in three publicly available microarray datasets. Three thousand and eighty-five differentially expressed genes in ≥2 datasets (P < 0.05) were used for pathway enrichment and gene ontology (GO) analyses. Further, we compared the topmost gene expression, pathways, and GO from poorly differentiated cell lines to the pattern from four human HCC datasets (623 tumour tissues). In well- versus poorly differentiated cell lines, and in representative models HLE and HUH7 cells, we specifically assessed the expression pattern of 634 consistently deregulated metabolic genes in human HCC. These data were complemented by quantitative PCR, proteomics, metabolomics and assessment of response to thirteen metabolism-targeting compounds in HLE versus HUH7 cells. RESULTS: We found that poorly-differentiated HCC cells display upregulated MAPK/RAS/NFkB signaling, focal adhesion, and downregulated complement/coagulation cascade, PPAR-signaling, among pathway alterations seen in clinical tumour datasets. In HLE cells, 148 downregulated metabolic genes in liver tumours also showed low gene/protein expression – notably in fatty acid β-oxidation (e.g. ACAA1/2, ACADSB, HADH), urea cycle (e.g. CPS1, ARG1, ASL), molecule transport (e.g. SLC2A2, SLC7A1, SLC25A15/20), and amino acid metabolism (e.g. PHGDH, PSAT1, GOT1, GLUD1). In contrast, HUH7 cells showed a higher expression of 98 metabolic targets upregulated in tumours (e.g. HK2, PKM, PSPH, GLUL, ASNS, and fatty acid synthesis enzymes ACLY, FASN). Metabolomics revealed that the genomic portrait of HLE cells co-exist with profound reliance on glutamine to fuel tricarboxylic acid cycle, whereas HUH7 cells use both glucose and glutamine. Targeting glutamine pathway selectively suppressed the proliferation of HLE cells. CONCLUSIONS: We report a yet unappreciated distinct expression pattern of clinically-relevant metabolic genes in HCC cell lines, which could enable the identification and therapeutic targeting of metabolic vulnerabilities at various liver cancer stages. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s13046-018-0872-6) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-6122702
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-61227022018-09-10 Liver cancer cell lines distinctly mimic the metabolic gene expression pattern of the corresponding human tumours Nwosu, Zeribe C. Battello, Nadia Rothley, Melanie Piorońska, Weronika Sitek, Barbara Ebert, Matthias P. Hofmann, Ute Sleeman, Jonathan Wölfl, Stefan Meyer, Christoph Megger, Dominik A. Dooley, Steven J Exp Clin Cancer Res Research BACKGROUND: Although metabolism is profoundly altered in human liver cancer, the extent to which experimental models, e.g. cell lines, mimic those alterations is unresolved. Here, we aimed to determine the resemblance of hepatocellular carcinoma (HCC) cell lines to human liver tumours, specifically in the expression of deregulated metabolic targets in clinical tissue samples. METHODS: We compared the overall gene expression profile of poorly-differentiated (HLE, HLF, SNU-449) to well-differentiated (HUH7, HEPG2, HEP3B) HCC cell lines in three publicly available microarray datasets. Three thousand and eighty-five differentially expressed genes in ≥2 datasets (P < 0.05) were used for pathway enrichment and gene ontology (GO) analyses. Further, we compared the topmost gene expression, pathways, and GO from poorly differentiated cell lines to the pattern from four human HCC datasets (623 tumour tissues). In well- versus poorly differentiated cell lines, and in representative models HLE and HUH7 cells, we specifically assessed the expression pattern of 634 consistently deregulated metabolic genes in human HCC. These data were complemented by quantitative PCR, proteomics, metabolomics and assessment of response to thirteen metabolism-targeting compounds in HLE versus HUH7 cells. RESULTS: We found that poorly-differentiated HCC cells display upregulated MAPK/RAS/NFkB signaling, focal adhesion, and downregulated complement/coagulation cascade, PPAR-signaling, among pathway alterations seen in clinical tumour datasets. In HLE cells, 148 downregulated metabolic genes in liver tumours also showed low gene/protein expression – notably in fatty acid β-oxidation (e.g. ACAA1/2, ACADSB, HADH), urea cycle (e.g. CPS1, ARG1, ASL), molecule transport (e.g. SLC2A2, SLC7A1, SLC25A15/20), and amino acid metabolism (e.g. PHGDH, PSAT1, GOT1, GLUD1). In contrast, HUH7 cells showed a higher expression of 98 metabolic targets upregulated in tumours (e.g. HK2, PKM, PSPH, GLUL, ASNS, and fatty acid synthesis enzymes ACLY, FASN). Metabolomics revealed that the genomic portrait of HLE cells co-exist with profound reliance on glutamine to fuel tricarboxylic acid cycle, whereas HUH7 cells use both glucose and glutamine. Targeting glutamine pathway selectively suppressed the proliferation of HLE cells. CONCLUSIONS: We report a yet unappreciated distinct expression pattern of clinically-relevant metabolic genes in HCC cell lines, which could enable the identification and therapeutic targeting of metabolic vulnerabilities at various liver cancer stages. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s13046-018-0872-6) contains supplementary material, which is available to authorized users. BioMed Central 2018-09-03 /pmc/articles/PMC6122702/ /pubmed/30176945 http://dx.doi.org/10.1186/s13046-018-0872-6 Text en © The Author(s). 2018, corrected publication October 2018 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research
Nwosu, Zeribe C.
Battello, Nadia
Rothley, Melanie
Piorońska, Weronika
Sitek, Barbara
Ebert, Matthias P.
Hofmann, Ute
Sleeman, Jonathan
Wölfl, Stefan
Meyer, Christoph
Megger, Dominik A.
Dooley, Steven
Liver cancer cell lines distinctly mimic the metabolic gene expression pattern of the corresponding human tumours
title Liver cancer cell lines distinctly mimic the metabolic gene expression pattern of the corresponding human tumours
title_full Liver cancer cell lines distinctly mimic the metabolic gene expression pattern of the corresponding human tumours
title_fullStr Liver cancer cell lines distinctly mimic the metabolic gene expression pattern of the corresponding human tumours
title_full_unstemmed Liver cancer cell lines distinctly mimic the metabolic gene expression pattern of the corresponding human tumours
title_short Liver cancer cell lines distinctly mimic the metabolic gene expression pattern of the corresponding human tumours
title_sort liver cancer cell lines distinctly mimic the metabolic gene expression pattern of the corresponding human tumours
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6122702/
https://www.ncbi.nlm.nih.gov/pubmed/30176945
http://dx.doi.org/10.1186/s13046-018-0872-6
work_keys_str_mv AT nwosuzeribec livercancercelllinesdistinctlymimicthemetabolicgeneexpressionpatternofthecorrespondinghumantumours
AT battellonadia livercancercelllinesdistinctlymimicthemetabolicgeneexpressionpatternofthecorrespondinghumantumours
AT rothleymelanie livercancercelllinesdistinctlymimicthemetabolicgeneexpressionpatternofthecorrespondinghumantumours
AT pioronskaweronika livercancercelllinesdistinctlymimicthemetabolicgeneexpressionpatternofthecorrespondinghumantumours
AT sitekbarbara livercancercelllinesdistinctlymimicthemetabolicgeneexpressionpatternofthecorrespondinghumantumours
AT ebertmatthiasp livercancercelllinesdistinctlymimicthemetabolicgeneexpressionpatternofthecorrespondinghumantumours
AT hofmannute livercancercelllinesdistinctlymimicthemetabolicgeneexpressionpatternofthecorrespondinghumantumours
AT sleemanjonathan livercancercelllinesdistinctlymimicthemetabolicgeneexpressionpatternofthecorrespondinghumantumours
AT wolflstefan livercancercelllinesdistinctlymimicthemetabolicgeneexpressionpatternofthecorrespondinghumantumours
AT meyerchristoph livercancercelllinesdistinctlymimicthemetabolicgeneexpressionpatternofthecorrespondinghumantumours
AT meggerdominika livercancercelllinesdistinctlymimicthemetabolicgeneexpressionpatternofthecorrespondinghumantumours
AT dooleysteven livercancercelllinesdistinctlymimicthemetabolicgeneexpressionpatternofthecorrespondinghumantumours