Skip to main content
Top
Published in: BMC Cancer 1/2019

Open Access 01-12-2019 | Leukemia | Research article

Cancer cell lipid class homeostasis is altered under nutrient-deprivation but stable under hypoxia

Authors: Jan Lisec, Carsten Jaeger, Rida Rashid, Rimsha Munir, Nousheen Zaidi

Published in: BMC Cancer | Issue 1/2019

Login to get access

Abstract

Background

Cancer cells modify the balance between fatty acid (FA) synthesis and uptake under metabolic stress, induced by oxygen/nutrient deprivation. These modifications were shown to alter the levels of individual triglyceride (TG) or phospholipid sub-species. To attain a holistic overview of the lipidomic profiles of cancer cells under stress we performed a broad lipidomic assay, comprising 244 lipids from six major classes. This assay allowed us to perform robust analyses and assess the changes in averages of broader lipid-classes, stratified on the basis of saturation index of their fatty-acyl side chains.

Methods

Global lipidomic profiling using Liquid Chromatography-Mass Spectrometry was performed to assess lipidomic profiles of biologically diverse cancer cell lines cultivated under metabolically stressed conditions.

Results

Neutral lipid compositions were markedly modified under serum-deprived conditions and, strikingly, the cellular level of triglyceride subspecies decreased with increasing number of double bonds in their fatty acyl chains. In contrast and unexpectedly, no robust changes were observed in lipidomic profiles of hypoxic (2% O2) cancer cells despite concurrent changes in proliferation rates and metabolic gene expression.

Conclusions

Serum-deprivation significantly affects lipidomic profiles of cancer cells. Although, the levels of individual lipid moieties alter under hypoxia (2% O2), the robust averages of broader lipid classes remain unchanged.
Appendix
Available only for authorised users
Literature
1.
go back to reference Beloribi-Djefaflia S, Vasseur S, Guillaumond F. Lipid metabolic reprogramming in cancer cells. Oncogenesis. 2016;5:e189.CrossRef Beloribi-Djefaflia S, Vasseur S, Guillaumond F. Lipid metabolic reprogramming in cancer cells. Oncogenesis. 2016;5:e189.CrossRef
2.
go back to reference Mullen PJ, Yu R, Longo J, Archer MC, Penn LZ. The interplay between cell signalling and the mevalonate pathway in cancer. Nat Rev Cancer. 2016;16(11):718–31.CrossRef Mullen PJ, Yu R, Longo J, Archer MC, Penn LZ. The interplay between cell signalling and the mevalonate pathway in cancer. Nat Rev Cancer. 2016;16(11):718–31.CrossRef
3.
go back to reference Rohrig F, Schulze A. The multifaceted roles of fatty acid synthesis in cancer. Nat Rev Cancer. 2016;16(11):732–49.CrossRef Rohrig F, Schulze A. The multifaceted roles of fatty acid synthesis in cancer. Nat Rev Cancer. 2016;16(11):732–49.CrossRef
4.
go back to reference Menendez JA, Lupu R. Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis. Nat Rev Cancer. 2007;7(10):763–77.CrossRef Menendez JA, Lupu R. Fatty acid synthase and the lipogenic phenotype in cancer pathogenesis. Nat Rev Cancer. 2007;7(10):763–77.CrossRef
5.
go back to reference Zhang F, Du G. Dysregulated lipid metabolism in cancer. World J Biol Chem. 2012;3(8):167–74.CrossRef Zhang F, Du G. Dysregulated lipid metabolism in cancer. World J Biol Chem. 2012;3(8):167–74.CrossRef
6.
go back to reference Zaidi N, Lupien L, Kuemmerle NB, Kinlaw WB, Swinnen JV, Smans K. Lipogenesis and lipolysis: the pathways exploited by the cancer cells to acquire fatty acids. Prog Lipid Res. 2013;52(4):585–9.CrossRef Zaidi N, Lupien L, Kuemmerle NB, Kinlaw WB, Swinnen JV, Smans K. Lipogenesis and lipolysis: the pathways exploited by the cancer cells to acquire fatty acids. Prog Lipid Res. 2013;52(4):585–9.CrossRef
7.
go back to reference Brusselmans K, Timmermans L, Van de Sande T, Van Veldhoven PP, Guan G, Shechter I, Claessens F, Verhoeven G, Swinnen JV. Squalene synthase, a determinant of raft-associated cholesterol and modulator of cancer cell proliferation. J Biol Chem. 2007;282(26):18777–85.CrossRef Brusselmans K, Timmermans L, Van de Sande T, Van Veldhoven PP, Guan G, Shechter I, Claessens F, Verhoeven G, Swinnen JV. Squalene synthase, a determinant of raft-associated cholesterol and modulator of cancer cell proliferation. J Biol Chem. 2007;282(26):18777–85.CrossRef
8.
go back to reference Sebti SM. Protein farnesylation: implications for normal physiology, malignant transformation, and cancer therapy. Cancer Cell. 2005;7(4):297–300.CrossRef Sebti SM. Protein farnesylation: implications for normal physiology, malignant transformation, and cancer therapy. Cancer Cell. 2005;7(4):297–300.CrossRef
9.
go back to reference Kuemmerle NB, Rysman E, Lombardo PS, Flanagan AJ, Lipe BC, Wells WA, Pettus JR, Froehlich HM, Memoli VA, Morganelli PM, et al. Lipoprotein lipase links dietary fat to solid tumor cell proliferation. Mol Cancer Ther. 2011;10(3):427–36.CrossRef Kuemmerle NB, Rysman E, Lombardo PS, Flanagan AJ, Lipe BC, Wells WA, Pettus JR, Froehlich HM, Memoli VA, Morganelli PM, et al. Lipoprotein lipase links dietary fat to solid tumor cell proliferation. Mol Cancer Ther. 2011;10(3):427–36.CrossRef
10.
go back to reference Chushi L, Wei W, Kangkang X, Yongzeng F, Ning X, Xiaolei C. HMGCR is up-regulated in gastric cancer and promotes the growth and migration of the cancer cells. Gene. 2016;587(1):42–7.CrossRef Chushi L, Wei W, Kangkang X, Yongzeng F, Ning X, Xiaolei C. HMGCR is up-regulated in gastric cancer and promotes the growth and migration of the cancer cells. Gene. 2016;587(1):42–7.CrossRef
11.
go back to reference Chajes V, Cambot M, Moreau K, Lenoir GM, Joulin V. Acetyl-CoA carboxylase alpha is essential to breast cancer cell survival. Cancer Res. 2006;66(10):5287–94.CrossRef Chajes V, Cambot M, Moreau K, Lenoir GM, Joulin V. Acetyl-CoA carboxylase alpha is essential to breast cancer cell survival. Cancer Res. 2006;66(10):5287–94.CrossRef
12.
go back to reference Migita T, Narita T, Nomura K, Miyagi E, Inazuka F, Matsuura M, Ushijima M, Mashima T, Seimiya H, Satoh Y, et al. ATP citrate lyase: activation and therapeutic implications in non-small cell lung cancer. Cancer Res. 2008;68(20):8547–54.CrossRef Migita T, Narita T, Nomura K, Miyagi E, Inazuka F, Matsuura M, Ushijima M, Mashima T, Seimiya H, Satoh Y, et al. ATP citrate lyase: activation and therapeutic implications in non-small cell lung cancer. Cancer Res. 2008;68(20):8547–54.CrossRef
13.
go back to reference Mashima T, Seimiya H, Tsuruo T. De novo fatty-acid synthesis and related pathways as molecular targets for cancer therapy. Br J Cancer. 2009;100(9):1369–72.CrossRef Mashima T, Seimiya H, Tsuruo T. De novo fatty-acid synthesis and related pathways as molecular targets for cancer therapy. Br J Cancer. 2009;100(9):1369–72.CrossRef
14.
go back to reference Hatzivassiliou G, Zhao F, Bauer DE, Andreadis C, Shaw AN, Dhanak D, Hingorani SR, Tuveson DA, Thompson CB. ATP citrate lyase inhibition can suppress tumor cell growth. Cancer Cell. 2005;8(4):311–21.CrossRef Hatzivassiliou G, Zhao F, Bauer DE, Andreadis C, Shaw AN, Dhanak D, Hingorani SR, Tuveson DA, Thompson CB. ATP citrate lyase inhibition can suppress tumor cell growth. Cancer Cell. 2005;8(4):311–21.CrossRef
15.
go back to reference Beckers A, Organe S, Timmermans L, Scheys K, Peeters A, Brusselmans K, Verhoeven G, Swinnen JV. Chemical inhibition of acetyl-CoA carboxylase induces growth arrest and cytotoxicity selectively in cancer cells. Cancer Res. 2007;67(17):8180–7.CrossRef Beckers A, Organe S, Timmermans L, Scheys K, Peeters A, Brusselmans K, Verhoeven G, Swinnen JV. Chemical inhibition of acetyl-CoA carboxylase induces growth arrest and cytotoxicity selectively in cancer cells. Cancer Res. 2007;67(17):8180–7.CrossRef
16.
go back to reference Brusselmans K, De Schrijver E, Verhoeven G, Swinnen JV. RNA interference-mediated silencing of the acetyl-CoA-carboxylase-alpha gene induces growth inhibition and apoptosis of prostate cancer cells. Cancer Res. 2005;65(15):6719–25.CrossRef Brusselmans K, De Schrijver E, Verhoeven G, Swinnen JV. RNA interference-mediated silencing of the acetyl-CoA-carboxylase-alpha gene induces growth inhibition and apoptosis of prostate cancer cells. Cancer Res. 2005;65(15):6719–25.CrossRef
17.
go back to reference De Schrijver E, Brusselmans K, Heyns W, Verhoeven G, Swinnen JV. RNA interference-mediated silencing of the fatty acid synthase gene attenuates growth and induces morphological changes and apoptosis of LNCaP prostate cancer cells. Cancer Res. 2003;63(13):3799–804.PubMed De Schrijver E, Brusselmans K, Heyns W, Verhoeven G, Swinnen JV. RNA interference-mediated silencing of the fatty acid synthase gene attenuates growth and induces morphological changes and apoptosis of LNCaP prostate cancer cells. Cancer Res. 2003;63(13):3799–804.PubMed
18.
go back to reference Kamphorst JJ, Cross JR, Fan J, de Stanchina E, Mathew R, White EP, Thompson CB, Rabinowitz JD. Hypoxic and Ras-transformed cells support growth by scavenging unsaturated fatty acids from lysophospholipids. Proc Natl Acad Sci U S A. 2013;110(22):8882–7.CrossRef Kamphorst JJ, Cross JR, Fan J, de Stanchina E, Mathew R, White EP, Thompson CB, Rabinowitz JD. Hypoxic and Ras-transformed cells support growth by scavenging unsaturated fatty acids from lysophospholipids. Proc Natl Acad Sci U S A. 2013;110(22):8882–7.CrossRef
19.
go back to reference Bensaad K, Favaro E, Lewis CA, Peck B, Lord S, Collins JM, Pinnick KE, Wigfield S, Buffa FM, Li JL, et al. Fatty acid uptake and lipid storage induced by HIF-1alpha contribute to cell growth and survival after hypoxia-reoxygenation. Cell Rep. 2014;9(1):349–65.CrossRef Bensaad K, Favaro E, Lewis CA, Peck B, Lord S, Collins JM, Pinnick KE, Wigfield S, Buffa FM, Li JL, et al. Fatty acid uptake and lipid storage induced by HIF-1alpha contribute to cell growth and survival after hypoxia-reoxygenation. Cell Rep. 2014;9(1):349–65.CrossRef
20.
go back to reference Lee JM, Lee H, Kang S, Park WJ. Fatty acid desaturases, polyunsaturated fatty acid regulation, and biotechnological advances. Nutrients. 2016;8(1).CrossRef Lee JM, Lee H, Kang S, Park WJ. Fatty acid desaturases, polyunsaturated fatty acid regulation, and biotechnological advances. Nutrients. 2016;8(1).CrossRef
21.
go back to reference Rysman E, Brusselmans K, Scheys K, Timmermans L, Derua R, Munck S, Van Veldhoven PP, Waltregny D, Daniels VW, Machiels J, et al. De novo lipogenesis protects cancer cells from free radicals and chemotherapeutics by promoting membrane lipid saturation. Cancer Res. 2010;70(20):8117–26.CrossRef Rysman E, Brusselmans K, Scheys K, Timmermans L, Derua R, Munck S, Van Veldhoven PP, Waltregny D, Daniels VW, Machiels J, et al. De novo lipogenesis protects cancer cells from free radicals and chemotherapeutics by promoting membrane lipid saturation. Cancer Res. 2010;70(20):8117–26.CrossRef
22.
go back to reference Bensaad K, Favaro E, Lewis CA, Peck B, Lord S, Collins JM, Pinnick KE, Wigfield S, Buffa FM, Li J-L. Fatty acid uptake and lipid storage induced by HIF-1α contribute to cell growth and survival after hypoxia-reoxygenation. Cell Rep. 2014;9(1):349–65.CrossRef Bensaad K, Favaro E, Lewis CA, Peck B, Lord S, Collins JM, Pinnick KE, Wigfield S, Buffa FM, Li J-L. Fatty acid uptake and lipid storage induced by HIF-1α contribute to cell growth and survival after hypoxia-reoxygenation. Cell Rep. 2014;9(1):349–65.CrossRef
23.
go back to reference Schlaepfer IR, Nambiar DK, Ramteke A, Kumar R, Dhar D, Agarwal C, Bergman B, Graner M, Maroni P, Singh RP. Hypoxia induces triglycerides accumulation in prostate cancer cells and extracellular vesicles supporting growth and invasiveness following reoxygenation. Oncotarget. 2015;6(26):22836.CrossRef Schlaepfer IR, Nambiar DK, Ramteke A, Kumar R, Dhar D, Agarwal C, Bergman B, Graner M, Maroni P, Singh RP. Hypoxia induces triglycerides accumulation in prostate cancer cells and extracellular vesicles supporting growth and invasiveness following reoxygenation. Oncotarget. 2015;6(26):22836.CrossRef
24.
go back to reference Valli A, Rodriguez M, Moutsianas L, Fischer R, Fedele V, Huang H-L, Van Stiphout R, Jones D, Mccarthy M, Vinaxia M. Hypoxia induces a lipogenic cancer cell phenotype via HIF1α-dependent and-independent pathways. Oncotarget. 2015;6(4):1920.CrossRef Valli A, Rodriguez M, Moutsianas L, Fischer R, Fedele V, Huang H-L, Van Stiphout R, Jones D, Mccarthy M, Vinaxia M. Hypoxia induces a lipogenic cancer cell phenotype via HIF1α-dependent and-independent pathways. Oncotarget. 2015;6(4):1920.CrossRef
25.
go back to reference Yu Y, Vidalino L, Anesi A, Macchi P, Guella G. A lipidomics investigation of the induced hypoxia stress on HeLa cells by using MS and NMR techniques. Mol BioSyst. 2014;10(4):878–90.CrossRef Yu Y, Vidalino L, Anesi A, Macchi P, Guella G. A lipidomics investigation of the induced hypoxia stress on HeLa cells by using MS and NMR techniques. Mol BioSyst. 2014;10(4):878–90.CrossRef
26.
go back to reference Schug ZT, Peck B, Jones DT, Zhang Q, Grosskurth S, Alam IS, Goodwin LM, Smethurst E, Mason S, Blyth K, et al. Acetyl-CoA synthetase 2 promotes acetate utilization and maintains cancer cell growth under metabolic stress. Cancer Cell. 2015;27(1):57–71.CrossRef Schug ZT, Peck B, Jones DT, Zhang Q, Grosskurth S, Alam IS, Goodwin LM, Smethurst E, Mason S, Blyth K, et al. Acetyl-CoA synthetase 2 promotes acetate utilization and maintains cancer cell growth under metabolic stress. Cancer Cell. 2015;27(1):57–71.CrossRef
27.
go back to reference Ackerman D, Tumanov S, Qiu B, Michalopoulou E, Spata M, Azzam A, Xie H, Simon MC, Kamphorst JJ. Triglycerides promote lipid homeostasis during hypoxic stress by balancing fatty acid saturation. Cell Rep. 2018;24(10):2596–2605.e2595.CrossRef Ackerman D, Tumanov S, Qiu B, Michalopoulou E, Spata M, Azzam A, Xie H, Simon MC, Kamphorst JJ. Triglycerides promote lipid homeostasis during hypoxic stress by balancing fatty acid saturation. Cell Rep. 2018;24(10):2596–2605.e2595.CrossRef
28.
go back to reference Klein D. The tumor vascular endothelium as decision maker in Cancer therapy. Front Oncol. 2018;8:367.CrossRef Klein D. The tumor vascular endothelium as decision maker in Cancer therapy. Front Oncol. 2018;8:367.CrossRef
29.
go back to reference Daster S, Amatruda N, Calabrese D, Ivanek R, Turrini E, Droeser RA, Zajac P, Fimognari C, Spagnoli GC, Iezzi G, et al. Induction of hypoxia and necrosis in multicellular tumor spheroids is associated with resistance to chemotherapy treatment. Oncotarget. 2017;8(1):1725–36.CrossRef Daster S, Amatruda N, Calabrese D, Ivanek R, Turrini E, Droeser RA, Zajac P, Fimognari C, Spagnoli GC, Iezzi G, et al. Induction of hypoxia and necrosis in multicellular tumor spheroids is associated with resistance to chemotherapy treatment. Oncotarget. 2017;8(1):1725–36.CrossRef
30.
go back to reference Tsugawa H, Cajka T, Kind T, Ma Y, Higgins B, Ikeda K, Kanazawa M, VanderGheynst J, Fiehn O, Arita M. MS-DIAL: data-independent MS/MS deconvolution for comprehensive metabolome analysis. Nat Methods. 2015;12(6):523–6.CrossRef Tsugawa H, Cajka T, Kind T, Ma Y, Higgins B, Ikeda K, Kanazawa M, VanderGheynst J, Fiehn O, Arita M. MS-DIAL: data-independent MS/MS deconvolution for comprehensive metabolome analysis. Nat Methods. 2015;12(6):523–6.CrossRef
31.
go back to reference Daniels VW, Smans K, Royaux I, Chypre M, Swinnen JV, Zaidi N. Cancer cells differentially activate and thrive on de novo lipid synthesis pathways in a low-lipid environment. PLoS One. 2014;9(9):e106913.CrossRef Daniels VW, Smans K, Royaux I, Chypre M, Swinnen JV, Zaidi N. Cancer cells differentially activate and thrive on de novo lipid synthesis pathways in a low-lipid environment. PLoS One. 2014;9(9):e106913.CrossRef
32.
go back to reference Roongta UV, Pabalan JG, Wang X, Ryseck R-P, Fargnoli J, Henley BJ, Yang W-P, Zhu J, Madireddi MT, Lawrence RM. Cancer cell dependence on unsaturated fatty acids implicates stearoyl-CoA desaturase as a target for cancer therapy. Mol Cancer Res. 2011;9(11):1551–61.CrossRef Roongta UV, Pabalan JG, Wang X, Ryseck R-P, Fargnoli J, Henley BJ, Yang W-P, Zhu J, Madireddi MT, Lawrence RM. Cancer cell dependence on unsaturated fatty acids implicates stearoyl-CoA desaturase as a target for cancer therapy. Mol Cancer Res. 2011;9(11):1551–61.CrossRef
33.
go back to reference Cabodevilla AG, Sanchez-Caballero L, Nintou E, Boiadjieva VG, Picatoste F, Gubern A, Claro E. Cell survival during complete nutrient deprivation depends on lipid droplet-fueled beta-oxidation of fatty acids. J Biol Chem. 2013;288(39):27777–88.CrossRef Cabodevilla AG, Sanchez-Caballero L, Nintou E, Boiadjieva VG, Picatoste F, Gubern A, Claro E. Cell survival during complete nutrient deprivation depends on lipid droplet-fueled beta-oxidation of fatty acids. J Biol Chem. 2013;288(39):27777–88.CrossRef
34.
go back to reference Gubern A, Barcelo-Torns M, Casas J, Barneda D, Masgrau R, Picatoste F, Balsinde J, Balboa MA, Claro E. Lipid droplet biogenesis induced by stress involves triacylglycerol synthesis that depends on group VIA phospholipase A2. J Biol Chem. 2009;284(9):5697–708.CrossRef Gubern A, Barcelo-Torns M, Casas J, Barneda D, Masgrau R, Picatoste F, Balsinde J, Balboa MA, Claro E. Lipid droplet biogenesis induced by stress involves triacylglycerol synthesis that depends on group VIA phospholipase A2. J Biol Chem. 2009;284(9):5697–708.CrossRef
35.
go back to reference Gubern A, Casas J, Barcelo-Torns M, Barneda D, de la Rosa X, Masgrau R, Picatoste F, Balsinde J, Balboa MA, Claro E. Group IVA phospholipase A2 is necessary for the biogenesis of lipid droplets. J Biol Chem. 2008;283(41):27369–82.CrossRef Gubern A, Casas J, Barcelo-Torns M, Barneda D, de la Rosa X, Masgrau R, Picatoste F, Balsinde J, Balboa MA, Claro E. Group IVA phospholipase A2 is necessary for the biogenesis of lipid droplets. J Biol Chem. 2008;283(41):27369–82.CrossRef
36.
go back to reference Guillaumond F, Bidaut G, Ouaissi M, Servais S, Gouirand V, Olivares O, Lac S, Borge L, Roques J, Gayet O, et al. Cholesterol uptake disruption, in association with chemotherapy, is a promising combined metabolic therapy for pancreatic adenocarcinoma. Proc Natl Acad Sci U S A. 2015;112(8):2473–8.CrossRef Guillaumond F, Bidaut G, Ouaissi M, Servais S, Gouirand V, Olivares O, Lac S, Borge L, Roques J, Gayet O, et al. Cholesterol uptake disruption, in association with chemotherapy, is a promising combined metabolic therapy for pancreatic adenocarcinoma. Proc Natl Acad Sci U S A. 2015;112(8):2473–8.CrossRef
37.
go back to reference Zhang X, Saarinen AM, Hitosugi T, Wang Z, Wang L, Ho TH, Liu J. Inhibition of intracellular lipolysis promotes human cancer cell adaptation to hypoxia. eLife. 2017;6. Zhang X, Saarinen AM, Hitosugi T, Wang Z, Wang L, Ho TH, Liu J. Inhibition of intracellular lipolysis promotes human cancer cell adaptation to hypoxia. eLife. 2017;6.
Metadata
Title
Cancer cell lipid class homeostasis is altered under nutrient-deprivation but stable under hypoxia
Authors
Jan Lisec
Carsten Jaeger
Rida Rashid
Rimsha Munir
Nousheen Zaidi
Publication date
01-12-2019
Publisher
BioMed Central
Keyword
Leukemia
Published in
BMC Cancer / Issue 1/2019
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-019-5733-y

Other articles of this Issue 1/2019

BMC Cancer 1/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine