Skip to main content
Top
Published in: BMC Cancer 1/2018

Open Access 01-12-2018 | Research article

Metabolic control of PPAR activity by aldehyde dehydrogenase regulates invasive cell behavior and predicts survival in hepatocellular and renal clear cell carcinoma

Authors: Diana Andrejeva, Jan-Michael Kugler, Hung Thanh Nguyen, Anders Malmendal, Mette Lind Holm, Birgitte Groenkaer Toft, Anand C. Loya, Stephen M. Cohen

Published in: BMC Cancer | Issue 1/2018

Login to get access

Abstract

Background

Changes in cellular metabolism are now recognized as potential drivers of cancer development, rather than as secondary consequences of disease. Here, we explore the mechanism by which metabolic changes dependent on aldehyde dehydrogenase impact cancer development.

Methods

ALDH7A1 was identified as a potential cancer gene using a Drosophila in vivo metastasis model. The role of the human ortholog was examined using RNA interference in cell-based assays of cell migration and invasion. 1H-NMR metabolite profiling was used to identify metabolic changes in ALDH7A1-depleted cells. Publically available cancer gene expression data was interrogated to identify a gene-expression signature associated with depletion of ALDH7A1. Computational pathway and gene set enrichment analysis was used to identify signaling pathways and cellular processes that were correlated with reduced ALDH7A1 expression in cancer. A variety of statistical tests used to evaluate these analyses are described in detail in the methods section. Immunohistochemistry was used to assess ALDH7A1 expression in tissue samples from cancer patients.

Results

Depletion of ALDH7A1 increased cellular migration and invasiveness in vitro. Depletion of ALDH7A1 led to reduced levels of metabolites identified as ligands for Peroxisome proliferator-activated receptor (PPARα). Analysis of publically available cancer gene expression data revealed that ALDH7A1 mRNA levels were reduced in many human cancers, and that this correlated with poor survival in kidney and liver cancer patients. Using pathway and gene set enrichment analysis, we establish a correlation between low ALDH7A1 levels, reduced PPAR signaling and reduced patient survival. Metabolic profiling showed that endogenous PPARα ligands were reduced in ALDH7A1-depleted cells. ALDH7A1-depletion led to reduced PPAR transcriptional activity. Treatment with a PPARα agonist restored normal cellular behavior. Low ALDH7A1 protein levels correlated with poor clinical outcome in hepatocellular and renal clear cell carcinoma patients.

Conclusions

We provide evidence that low ALDH7A1 expression is a useful prognostic marker of poor clinical outcome for hepatocellular and renal clear cell carcinomas and hypothesize that patients with low ALDH7A1 might benefit from therapeutic approaches addressing PPARα activity.
Appendix
Available only for authorised users
Literature
1.
go back to reference Pavlova NN, Thompson CB. The emerging hallmarks of Cancer metabolism. Cell Metab. 2016;23:27–47.CrossRef Pavlova NN, Thompson CB. The emerging hallmarks of Cancer metabolism. Cell Metab. 2016;23:27–47.CrossRef
2.
go back to reference Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–74.CrossRef Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell. 2011;144:646–74.CrossRef
3.
go back to reference Christofk HR, Vander Heiden MG, Harris MH, Ramanathan A, Gerszten RE, Wei R, Fleming MD, Schreiber SL, Cantley LC. The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature. 2008;452:230–3.CrossRef Christofk HR, Vander Heiden MG, Harris MH, Ramanathan A, Gerszten RE, Wei R, Fleming MD, Schreiber SL, Cantley LC. The M2 splice isoform of pyruvate kinase is important for cancer metabolism and tumour growth. Nature. 2008;452:230–3.CrossRef
4.
go back to reference Dang L, White DW, Gross S, Bennett BD, Bittinger MA, Driggers EM, Fantin VR, Jang HG, Jin S, Keenan MC, et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature. 2009;462:739–44.CrossRef Dang L, White DW, Gross S, Bennett BD, Bittinger MA, Driggers EM, Fantin VR, Jang HG, Jin S, Keenan MC, et al. Cancer-associated IDH1 mutations produce 2-hydroxyglutarate. Nature. 2009;462:739–44.CrossRef
5.
go back to reference Lu C, Ward PS, Kapoor GS, Rohle D, Turcan S, Abdel-Wahab O, Edwards CR, Khanin R, Figueroa ME, Melnick A, et al. IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature. 2012;483:474–8.CrossRef Lu C, Ward PS, Kapoor GS, Rohle D, Turcan S, Abdel-Wahab O, Edwards CR, Khanin R, Figueroa ME, Melnick A, et al. IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature. 2012;483:474–8.CrossRef
6.
go back to reference Watanabe T, Nobusawa S, Kleihues P, Ohgaki H. IDH1 mutations are early events in the development of astrocytomas and oligodendrogliomas. Am J Pathol. 2009;174:1149–53.CrossRef Watanabe T, Nobusawa S, Kleihues P, Ohgaki H. IDH1 mutations are early events in the development of astrocytomas and oligodendrogliomas. Am J Pathol. 2009;174:1149–53.CrossRef
7.
go back to reference Wu W, Zhao S. Metabolic changes in cancer: beyond the Warburg effect. Acta Biochim Biophys Sin. 2013;45:18–26.CrossRef Wu W, Zhao S. Metabolic changes in cancer: beyond the Warburg effect. Acta Biochim Biophys Sin. 2013;45:18–26.CrossRef
8.
go back to reference Jackson B, Brocker C, Thompson DC, Black W, Vasiliou K, Nebert DW, Vasiliou V. Update on the aldehyde dehydrogenase gene (ALDH) superfamily. Hum Genomics. 2011;5:283–303.CrossRef Jackson B, Brocker C, Thompson DC, Black W, Vasiliou K, Nebert DW, Vasiliou V. Update on the aldehyde dehydrogenase gene (ALDH) superfamily. Hum Genomics. 2011;5:283–303.CrossRef
9.
go back to reference Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J, Brown M, Jacquemier J, Viens P, Kleer CG, Liu S, et al. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell. 2007;1:555–67.CrossRef Ginestier C, Hur MH, Charafe-Jauffret E, Monville F, Dutcher J, Brown M, Jacquemier J, Viens P, Kleer CG, Liu S, et al. ALDH1 is a marker of normal and malignant human mammary stem cells and a predictor of poor clinical outcome. Cell Stem Cell. 2007;1:555–67.CrossRef
10.
go back to reference Yang L, Ren Y, Yu X, Qian F, Bian BS, Xiao HL, Wang WG, Xu SL, Yang J, Cui W, et al. ALDH1A1 defines invasive cancer stem-like cells and predicts poor prognosis in patients with esophageal squamous cell carcinoma. Mod Pathol. 2014;27:775–83.CrossRef Yang L, Ren Y, Yu X, Qian F, Bian BS, Xiao HL, Wang WG, Xu SL, Yang J, Cui W, et al. ALDH1A1 defines invasive cancer stem-like cells and predicts poor prognosis in patients with esophageal squamous cell carcinoma. Mod Pathol. 2014;27:775–83.CrossRef
11.
go back to reference Penumatsa K, Edassery SL, Barua A, Bradaric MJ, Luborsky JL. Differential expression of aldehyde dehydrogenase 1a1 (ALDH1) in normal ovary and serous ovarian tumors. J Ovarian Res. 2010;3:28.CrossRef Penumatsa K, Edassery SL, Barua A, Bradaric MJ, Luborsky JL. Differential expression of aldehyde dehydrogenase 1a1 (ALDH1) in normal ovary and serous ovarian tumors. J Ovarian Res. 2010;3:28.CrossRef
12.
go back to reference Tanaka K, Tomita H, Hisamatsu K, Nakashima T, Hatano Y, Sasaki Y, Osada S, Tanaka T, Miyazaki T, Yoshida K, Hara A. ALDH1A1-overexpressing cells are differentiated cells but not cancer stem or progenitor cells in human hepatocellular carcinoma. Oncotarget. 2015;6:24722–32.PubMedPubMedCentral Tanaka K, Tomita H, Hisamatsu K, Nakashima T, Hatano Y, Sasaki Y, Osada S, Tanaka T, Miyazaki T, Yoshida K, Hara A. ALDH1A1-overexpressing cells are differentiated cells but not cancer stem or progenitor cells in human hepatocellular carcinoma. Oncotarget. 2015;6:24722–32.PubMedPubMedCentral
13.
go back to reference Chakravarthy MV, Lodhi IJ, Yin L, Malapaka RR, Xu HE, Turk J, Semenkovich CF. Identification of a physiologically relevant endogenous ligand for PPARalpha in liver. Cell. 2009;138:476–88.CrossRef Chakravarthy MV, Lodhi IJ, Yin L, Malapaka RR, Xu HE, Turk J, Semenkovich CF. Identification of a physiologically relevant endogenous ligand for PPARalpha in liver. Cell. 2009;138:476–88.CrossRef
14.
go back to reference Liu S, Brown JD, Stanya KJ, Homan E, Leidl M, Inouye K, Bhargava P, Gangl MR, Dai L, Hatano B, et al. A diurnal serum lipid integrates hepatic lipogenesis and peripheral fatty acid use. Nature. 2013;502:550–4.CrossRef Liu S, Brown JD, Stanya KJ, Homan E, Leidl M, Inouye K, Bhargava P, Gangl MR, Dai L, Hatano B, et al. A diurnal serum lipid integrates hepatic lipogenesis and peripheral fatty acid use. Nature. 2013;502:550–4.CrossRef
15.
go back to reference Vitale SG, Lagana AS, Nigro A, La Rosa VL, Rossetti P, Rapisarda AM, La Vignera S, Condorelli RA, Corrado F, Buscema M, D’Anna R. Peroxisome proliferator-activated receptor modulation during metabolic diseases and cancers: master and minions. PPAR Res. 2016;2016:6517313.CrossRef Vitale SG, Lagana AS, Nigro A, La Rosa VL, Rossetti P, Rapisarda AM, La Vignera S, Condorelli RA, Corrado F, Buscema M, D’Anna R. Peroxisome proliferator-activated receptor modulation during metabolic diseases and cancers: master and minions. PPAR Res. 2016;2016:6517313.CrossRef
16.
go back to reference Ament Z, Masoodi M, Griffin JL. Applications of metabolomics for understanding the action of peroxisome proliferator-activated receptors (PPARs) in diabetes, obesity and cancer. Genome Med. 2012;4:32.CrossRef Ament Z, Masoodi M, Griffin JL. Applications of metabolomics for understanding the action of peroxisome proliferator-activated receptors (PPARs) in diabetes, obesity and cancer. Genome Med. 2012;4:32.CrossRef
17.
go back to reference Peters JM, Shah YM, Gonzalez FJ. The role of peroxisome proliferator-activated receptors in carcinogenesis and chemoprevention. Nat Rev Cancer. 2012;12:181–95.CrossRef Peters JM, Shah YM, Gonzalez FJ. The role of peroxisome proliferator-activated receptors in carcinogenesis and chemoprevention. Nat Rev Cancer. 2012;12:181–95.CrossRef
18.
go back to reference Colaprico A, Silva TC, Olsen C, Garofano L, Cava C, Garolini D, Sabedot TS, Malta TM, Pagnotta SM, Castiglioni I, et al. TCGAbiolinks: an R/Bioconductor package for integrative analysis of TCGA data. Nucleic Acids Res. 2016;44:e71.CrossRef Colaprico A, Silva TC, Olsen C, Garofano L, Cava C, Garolini D, Sabedot TS, Malta TM, Pagnotta SM, Castiglioni I, et al. TCGAbiolinks: an R/Bioconductor package for integrative analysis of TCGA data. Nucleic Acids Res. 2016;44:e71.CrossRef
19.
go back to reference Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47.CrossRef Ritchie ME, Phipson B, Wu D, Hu Y, Law CW, Shi W, Smyth GK. Limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 2015;43:e47.CrossRef
20.
go back to reference Tarca AL, Draghici S, Khatri P, Hassan SS, Mittal P, Kim JS, Kim CJ, Kusanovic JP, Romero R. A novel signaling pathway impact analysis. Bioinformatics. 2009;25:75–82.CrossRef Tarca AL, Draghici S, Khatri P, Hassan SS, Mittal P, Kim JS, Kim CJ, Kusanovic JP, Romero R. A novel signaling pathway impact analysis. Bioinformatics. 2009;25:75–82.CrossRef
21.
go back to reference Gu Z, Wang J. CePa: an R package for finding significant pathways weighted by multiple network centralities. Bioinformatics. 2013;29:658–60.CrossRef Gu Z, Wang J. CePa: an R package for finding significant pathways weighted by multiple network centralities. Bioinformatics. 2013;29:658–60.CrossRef
22.
go back to reference Sales G, Calura E, Cavalieri D, Romualdi C. Graphite - a Bioconductor package to convert pathway topology to gene network. BMC Bioinform. 2012;13:20.CrossRef Sales G, Calura E, Cavalieri D, Romualdi C. Graphite - a Bioconductor package to convert pathway topology to gene network. BMC Bioinform. 2012;13:20.CrossRef
23.
go back to reference Varemo L, Nielsen J, Nookaew I. Enriching the gene set analysis of genome-wide data by incorporating directionality of gene expression and combining statistical hypotheses and methods. Nucleic Acids Res. 2013;41:4378–91.CrossRef Varemo L, Nielsen J, Nookaew I. Enriching the gene set analysis of genome-wide data by incorporating directionality of gene expression and combining statistical hypotheses and methods. Nucleic Acids Res. 2013;41:4378–91.CrossRef
24.
go back to reference Luo W, Friedman MS, Shedden K, Hankenson KD, Woolf PJ. GAGE: generally applicable gene set enrichment for pathway analysis. BMC Bioinform. 2009;10:161.CrossRef Luo W, Friedman MS, Shedden K, Hankenson KD, Woolf PJ. GAGE: generally applicable gene set enrichment for pathway analysis. BMC Bioinform. 2009;10:161.CrossRef
25.
go back to reference Han J, Shi X, Zhang Y, Xu Y, Jiang Y, Zhang C, Feng L, Yang H, Shang D, Sun Z, et al. ESEA: discovering the dysregulated pathways based on edge set enrichment analysis. Sci Rep. 2015;5:13044.CrossRef Han J, Shi X, Zhang Y, Xu Y, Jiang Y, Zhang C, Feng L, Yang H, Shang D, Sun Z, et al. ESEA: discovering the dysregulated pathways based on edge set enrichment analysis. Sci Rep. 2015;5:13044.CrossRef
26.
go back to reference Kanehisa M, Sato Y, Kawashima M, Furumichi M, Tanabe M. KEGG as a reference resource for gene and protein annotation. Nucleic Acids Res. 2016;44:D457–62.CrossRef Kanehisa M, Sato Y, Kawashima M, Furumichi M, Tanabe M. KEGG as a reference resource for gene and protein annotation. Nucleic Acids Res. 2016;44:D457–62.CrossRef
27.
go back to reference Ogata H, Goto S, Sato K, Fujibuchi W, Bono H, Kanehisa M. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 1999;27:29–34.CrossRef Ogata H, Goto S, Sato K, Fujibuchi W, Bono H, Kanehisa M. KEGG: Kyoto encyclopedia of genes and genomes. Nucleic Acids Res. 1999;27:29–34.CrossRef
28.
go back to reference Luo W, Brouwer C. Pathview: an R/Bioconductor package for pathway-based data integration and visualization. Bioinformatics. 2013;29:1830–1.CrossRef Luo W, Brouwer C. Pathview: an R/Bioconductor package for pathway-based data integration and visualization. Bioinformatics. 2013;29:1830–1.CrossRef
29.
go back to reference Beckonert O, Keun HC, Ebbels TM, Bundy J, Holmes E, Lindon JC, Nicholson JK. Metabolic profiling, metabolomic and metabonomic procedures for NMR spectroscopy of urine, plasma, serum and tissue extracts. Nat Protoc. 2007;2:2692–703.CrossRef Beckonert O, Keun HC, Ebbels TM, Bundy J, Holmes E, Lindon JC, Nicholson JK. Metabolic profiling, metabolomic and metabonomic procedures for NMR spectroscopy of urine, plasma, serum and tissue extracts. Nat Protoc. 2007;2:2692–703.CrossRef
30.
go back to reference Gaude E, Chignola F, Spiliotopoulos D, Spitaleri A, Ghitti M, Garcia-Manteigna J, Mari S, Musco G. Muma, an R package for metabolomics univariate and multivariate statistical analysis. Curr Metabolomics. 2013;1:180–9.CrossRef Gaude E, Chignola F, Spiliotopoulos D, Spitaleri A, Ghitti M, Garcia-Manteigna J, Mari S, Musco G. Muma, an R package for metabolomics univariate and multivariate statistical analysis. Curr Metabolomics. 2013;1:180–9.CrossRef
31.
go back to reference Thanh Nguyen H, Andrejeva D, Gupta R, Choudhary C, Hong X, Eichhorn PJ, Loya AC, Cohen SM. Deubiquitylating enzyme USP9x regulates hippo pathway activity by controlling angiomotin protein turnover. Cell Discov. 2016;2:16001.CrossRef Thanh Nguyen H, Andrejeva D, Gupta R, Choudhary C, Hong X, Eichhorn PJ, Loya AC, Cohen SM. Deubiquitylating enzyme USP9x regulates hippo pathway activity by controlling angiomotin protein turnover. Cell Discov. 2016;2:16001.CrossRef
32.
go back to reference Nguyen HT, Hong X, Tan S, Chen Q, Chan L, Fivaz M, Cohen SM, Voorhoeve PM. Viral small T oncoproteins transform cells by alleviating hippo-pathway-mediated inhibition of the YAP proto-oncogene. Cell Rep. 2014;8:707–13.CrossRef Nguyen HT, Hong X, Tan S, Chen Q, Chan L, Fivaz M, Cohen SM, Voorhoeve PM. Viral small T oncoproteins transform cells by alleviating hippo-pathway-mediated inhibition of the YAP proto-oncogene. Cell Rep. 2014;8:707–13.CrossRef
33.
go back to reference Abatangelo L, Maglietta R, Distaso A, D’Addabbo A, Creanza TM, Mukherjee S, Ancona N. Comparative study of gene set enrichment methods. BMC Bioinform. 2009;10:275.CrossRef Abatangelo L, Maglietta R, Distaso A, D’Addabbo A, Creanza TM, Mukherjee S, Ancona N. Comparative study of gene set enrichment methods. BMC Bioinform. 2009;10:275.CrossRef
34.
go back to reference Bayerlova M, Jung K, Kramer F, Klemm F, Bleckmann A, Beissbarth T. Comparative study on gene set and pathway topology-based enrichment methods. BMC Bioinform. 2015;16:334.CrossRef Bayerlova M, Jung K, Kramer F, Klemm F, Bleckmann A, Beissbarth T. Comparative study on gene set and pathway topology-based enrichment methods. BMC Bioinform. 2015;16:334.CrossRef
35.
go back to reference Cui Q, Lewis IA, Hegeman AD, Anderson ME, Li J, Schulte CF, Westler WM, Eghbalnia HR, Sussman MR, Markley JL. Metabolite identification via the Madison metabolomics consortium database. Nat Biotechnol. 2008;26:162–4.CrossRef Cui Q, Lewis IA, Hegeman AD, Anderson ME, Li J, Schulte CF, Westler WM, Eghbalnia HR, Sussman MR, Markley JL. Metabolite identification via the Madison metabolomics consortium database. Nat Biotechnol. 2008;26:162–4.CrossRef
36.
go back to reference Wishart DS, Tzur D, Knox C, Eisner R, Guo AC, Young N, Cheng D, Jewell K, Arndt D, Sawhney S, et al. HMDB: the human metabolome database. Nucleic Acids Res. 2007;35:D521–6.CrossRef Wishart DS, Tzur D, Knox C, Eisner R, Guo AC, Young N, Cheng D, Jewell K, Arndt D, Sawhney S, et al. HMDB: the human metabolome database. Nucleic Acids Res. 2007;35:D521–6.CrossRef
37.
go back to reference Brocker C, Cantore M, Failli P, Vasiliou V. Aldehyde dehydrogenase 7A1 (ALDH7A1) attenuates reactive aldehyde and oxidative stress induced cytotoxicity. Chem Biol Interact. 2011;191:269–77.CrossRef Brocker C, Cantore M, Failli P, Vasiliou V. Aldehyde dehydrogenase 7A1 (ALDH7A1) attenuates reactive aldehyde and oxidative stress induced cytotoxicity. Chem Biol Interact. 2011;191:269–77.CrossRef
38.
go back to reference Brocker C, Lassen N, Estey T, Pappa A, Cantore M, Orlova VV, Chavakis T, Kavanagh KL, Oppermann U, Vasiliou V. Aldehyde dehydrogenase 7A1 (ALDH7A1) is a novel enzyme involved in cellular defense against hyperosmotic stress. J Biol Chem. 2010;285:18452–63.CrossRef Brocker C, Lassen N, Estey T, Pappa A, Cantore M, Orlova VV, Chavakis T, Kavanagh KL, Oppermann U, Vasiliou V. Aldehyde dehydrogenase 7A1 (ALDH7A1) is a novel enzyme involved in cellular defense against hyperosmotic stress. J Biol Chem. 2010;285:18452–63.CrossRef
39.
go back to reference Giacalone NJ, Den RB, Eisenberg R, Chen H, Olson SJ, Massion PP, Carbone DP, Lu B. ALDH7A1 expression is associated with recurrence in patients with surgically resected non-small-cell lung carcinoma. Future Oncol. 2013;9:737–45.CrossRef Giacalone NJ, Den RB, Eisenberg R, Chen H, Olson SJ, Massion PP, Carbone DP, Lu B. ALDH7A1 expression is associated with recurrence in patients with surgically resected non-small-cell lung carcinoma. Future Oncol. 2013;9:737–45.CrossRef
40.
go back to reference Saw YT, Yang J, Ng SK, Liu S, Singh S, Singh M, Welch WR, Tsuda H, Fong WP, Thompson D, et al. Characterization of aldehyde dehydrogenase isozymes in ovarian cancer tissues and sphere cultures. BMC Cancer. 2012;12:329.CrossRef Saw YT, Yang J, Ng SK, Liu S, Singh S, Singh M, Welch WR, Tsuda H, Fong WP, Thompson D, et al. Characterization of aldehyde dehydrogenase isozymes in ovarian cancer tissues and sphere cultures. BMC Cancer. 2012;12:329.CrossRef
41.
go back to reference Grabacka M, Pierzchalska M, Reiss K. Peroxisome proliferator activated receptor alpha ligands as anticancer drugs targeting mitochondrial metabolism. Curr Pharm Biotechnol. 2013;14:342–56.CrossRef Grabacka M, Pierzchalska M, Reiss K. Peroxisome proliferator activated receptor alpha ligands as anticancer drugs targeting mitochondrial metabolism. Curr Pharm Biotechnol. 2013;14:342–56.CrossRef
42.
go back to reference Zhang N, Chu ES, Zhang J, Li X, Liang Q, Chen J, Chen M, Teoh N, Farrell G, Sung JJ, Yu J. Peroxisome proliferator activated receptor alpha inhibits hepatocarcinogenesis through mediating NF-kappaB signaling pathway. Oncotarget. 2014;5:8330–40.PubMedPubMedCentral Zhang N, Chu ES, Zhang J, Li X, Liang Q, Chen J, Chen M, Teoh N, Farrell G, Sung JJ, Yu J. Peroxisome proliferator activated receptor alpha inhibits hepatocarcinogenesis through mediating NF-kappaB signaling pathway. Oncotarget. 2014;5:8330–40.PubMedPubMedCentral
43.
go back to reference Ament Z, West JA, Stanley E, Ashmore T, Roberts LD, Wright J, Nicholls AW, Griffin JL. PPAR-pan activation induces hepatic oxidative stress and lipidomic remodelling. Free Radic Biol Med. 2016;95:357–68.CrossRef Ament Z, West JA, Stanley E, Ashmore T, Roberts LD, Wright J, Nicholls AW, Griffin JL. PPAR-pan activation induces hepatic oxidative stress and lipidomic remodelling. Free Radic Biol Med. 2016;95:357–68.CrossRef
44.
go back to reference Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N Engl J Med. 2007;356:2457–71.CrossRef Nissen SE, Wolski K. Effect of rosiglitazone on the risk of myocardial infarction and death from cardiovascular causes. N Engl J Med. 2007;356:2457–71.CrossRef
45.
go back to reference Turner RM, Kwok CS, Chen-Turner C, Maduakor CA, Singh S, Loke YK. Thiazolidinediones and associated risk of bladder cancer: a systematic review and meta-analysis. Br J Clin Pharmacol. 2014;78:258–73.CrossRef Turner RM, Kwok CS, Chen-Turner C, Maduakor CA, Singh S, Loke YK. Thiazolidinediones and associated risk of bladder cancer: a systematic review and meta-analysis. Br J Clin Pharmacol. 2014;78:258–73.CrossRef
46.
go back to reference Bonovas S, Nikolopoulos GK, Bagos PG. Use of fibrates and cancer risk: a systematic review and meta-analysis of 17 long-term randomized placebo-controlled trials. PLoS One. 2012;7:e45259.CrossRef Bonovas S, Nikolopoulos GK, Bagos PG. Use of fibrates and cancer risk: a systematic review and meta-analysis of 17 long-term randomized placebo-controlled trials. PLoS One. 2012;7:e45259.CrossRef
47.
go back to reference Gardette V, Bongard V, Dallongeville J, Arveiler D, Bingham A, Ruidavets JB, Amouyel P, Haas B, Ducimetiere P, Ferrieres J. Ten-year all-cause mortality in presumably healthy subjects on lipid-lowering drugs (from the prospective epidemiological study of myocardial infarction [PRIME] prospective cohort). Am J Cardiol. 2009;103:381–6.CrossRef Gardette V, Bongard V, Dallongeville J, Arveiler D, Bingham A, Ruidavets JB, Amouyel P, Haas B, Ducimetiere P, Ferrieres J. Ten-year all-cause mortality in presumably healthy subjects on lipid-lowering drugs (from the prospective epidemiological study of myocardial infarction [PRIME] prospective cohort). Am J Cardiol. 2009;103:381–6.CrossRef
Metadata
Title
Metabolic control of PPAR activity by aldehyde dehydrogenase regulates invasive cell behavior and predicts survival in hepatocellular and renal clear cell carcinoma
Authors
Diana Andrejeva
Jan-Michael Kugler
Hung Thanh Nguyen
Anders Malmendal
Mette Lind Holm
Birgitte Groenkaer Toft
Anand C. Loya
Stephen M. Cohen
Publication date
01-12-2018
Publisher
BioMed Central
Published in
BMC Cancer / Issue 1/2018
Electronic ISSN: 1471-2407
DOI
https://doi.org/10.1186/s12885-018-5061-7

Other articles of this Issue 1/2018

BMC Cancer 1/2018 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