Lipid degradation promotes prostate cancer cell survival

Harri M Itkonen, Michael Brown, Alfonso Urbanucci, Gregory Tredwell, Chung Ho Lau, Stefan Barfeld, Claire Hart, Ingrid J Guldvik, Mandeep Takhar, Hannelore V Heemers, Nicholas Erho, Katarzyna Bloch, Elai Davicioni, Rita Derua, Etienne Waelkens, James L Mohler, Noel Clarke, Johan V Swinnen, Hector C Keun, Ole P Rekvig, Ian G Mills, Harri M Itkonen, Michael Brown, Alfonso Urbanucci, Gregory Tredwell, Chung Ho Lau, Stefan Barfeld, Claire Hart, Ingrid J Guldvik, Mandeep Takhar, Hannelore V Heemers, Nicholas Erho, Katarzyna Bloch, Elai Davicioni, Rita Derua, Etienne Waelkens, James L Mohler, Noel Clarke, Johan V Swinnen, Hector C Keun, Ole P Rekvig, Ian G Mills

Abstract

Prostate cancer is the most common male cancer and androgen receptor (AR) is the major driver of the disease. Here we show that Enoyl-CoA delta isomerase 2 (ECI2) is a novel AR-target that promotes prostate cancer cell survival. Increased ECI2 expression predicts mortality in prostate cancer patients (p = 0.0086). ECI2 encodes for an enzyme involved in lipid metabolism, and we use multiple metabolite profiling platforms and RNA-seq to show that inhibition of ECI2 expression leads to decreased glucose utilization, accumulation of fatty acids and down-regulation of cell cycle related genes. In normal cells, decrease in fatty acid degradation is compensated by increased consumption of glucose, and here we demonstrate that prostate cancer cells are not able to respond to decreased fatty acid degradation. Instead, prostate cancer cells activate incomplete autophagy, which is followed by activation of the cell death response. Finally, we identified a clinically approved compound, perhexiline, which inhibits fatty acid degradation, and replicates the major findings for ECI2 knockdown. This work shows that prostate cancer cells require lipid degradation for survival and identifies a small molecule inhibitor with therapeutic potential.

Keywords: ECI2; androgen receptor; cell cycle; lipid degradation; metabolism.

Conflict of interest statement

CONFLICTS OF INTEREST

None.

Figures

Figure 1. Enoyl-CoA delta isomerase 2 (ECI2)…
Figure 1. Enoyl-CoA delta isomerase 2 (ECI2) is over-expressed in prostate cancer
(A) ECI2 expression was evaluated in prostate cancer tissue samples. The data shown represents matched normal epithelium and adenocarcinoma from 20 radical prostatectomy specimens. Relative expression of the different transcripts were calculated using the comparative CT method, where the matched benign tissue of the same patient were set to 1 and normalized to the geometric mean CT value of GAPDH, TBP and 18s. Wilcoxon matched-pairs signed rank test was used to test for significance in the differential expression of ECI2 between the matched benign and cancer tissue. (B) Kaplan Meier curves for the low/medium group versus the high ECI2 expressing group. We evaluated whether ECI2 expression levels are associated with survival in prostate cancer patients. The difference in overall survival between the low/medium expressing group and high expressing group was 77 months vs 115 months, p = 0.0086. Here stating that an overview of the clinical cohorts use in Figures 1A and 1B and the statistical analysis are to be found in Supplementary Tables 2, 4 and 5.
Figure 2. Androgen receptor (AR) regulates Enoyl-CoA…
Figure 2. Androgen receptor (AR) regulates Enoyl-CoA delta isomerase 2 (ECI2) expression
(A) Chromatin immunoprecipitation (ChIP) of androgen receptor (AR) in VCaP cells. Cells were deprived of androgens for 3 days and treated either with 1nM R1881 or vehicle, as indicated. The putative AR binding site for ECI2 was identified from a published AR ChIP-seq data set [3]. The data shown is representative of two biological replicates. (B) LNCaP and VCaP cells were treated as in A. Total mRNA was isolated at 12 hours and the expression of ECI2 and actin was evaluated using RT-qPCR. The data shown are an average of three independent experiments with SEM, and significance was evaluated using paired samples Student's t-test, *< 0.05. (C) LNCaP and VCaP cells were deprived of androgens for 3 days, treated either with 1nM R1881 or vehicle (−) and protein lysates were collected. Densitometry was used to evaluate ECI2 levels. (D) Western blot was used to confirm ECI2 knockdown in LNCaP and RWPE-1 cells after 96 hours. Densitometry was used to evaluate ECI2 levels. The growth rate of cells was evaluated with life-cell imaging. The data shown are an average of three independent biological replicates with SEM. The significance was evaluated using paired samples Student's t-test, **< 0.01, ***< 0.001. (E) Cell death activation after ECI2 knockdown. LNCaP cells were reverse-transfected and allowed to attach for 24 hours. At this point, a dye detecting caspase 3/7 activation was added and cumulative activation of caspase 3/7 was followed in real-time using life-cell imaging. Data shown is an average of three biological replicates with SEM.
Figure 3. Metabolomic profiling after ECI2 knockdown…
Figure 3. Metabolomic profiling after ECI2 knockdown in LNCaP cells
(A) Reaction catalyzed by Enoyl-CoA Delta Isomerase 2 (ECI2). (B) The level of extra-cellular lactate, glucose and choline were determined using nuclear magnetic resonance after 72 hours of ECI2 knockdown. The data are presented as % of complete media without cells and are an average (with SEM) of five biological replicates. The significance was evaluated using paired samples Student's t-test, *< 0.05, ***< 0.001. (C) The levels of water-soluble intra-cellular metabolites were determined using mass-spectrometry after 72 hours of ECI2 knockdown. Only metabolites that were affected significantly by at least one siRNA are shown. Data shown are an average (with SEM) of five biological replicates. The significance was evaluated using paired samples Student's t-test, *< 0.05, **< 0.01. (D) ECI2 knockdown in LNCaP (for 72 hours) and VCaP cells (for 96 hours). The data shown are representative of at least three biological replicates for LNCaP cells and two replicates for VCaP cells. Densitometry was used to evaluate signal intensity. (E) The levels of intra-cellular phosphatidylserine (PS), phosphatidylcholine (PC), phosphatidylethanolamine (PE) and phosphatidylinositol (PI) were determined using mass-spectrometry after 72 hours of ECI2 knockdown. Identification of lipid composition is provided in Supplementary Figure 2. Data shown are an average (with SEM) of three biological replicates. The significance was evaluated using paired samples Student's t-test, *< 0.05, **< 0.01. (F) The levels of saturated and un-saturated lipids were determined using mass-spectrometry after 72 hours of ECI2 knockdown (same data-set as presented in Figure 3E). Scrambled samples were set to 1 for all four lipid classes, and siECI2 samples were normalized to this. Data shown are an average (with SEM) of three biological replicates. The significance was evaluated using paired samples Student's t-test, *< 0.05, **< 0.01.
Figure 4. RNA-seq after ECI2 knockdown in…
Figure 4. RNA-seq after ECI2 knockdown in LNCaP and RWPE-1 cells
The expression of ECI2 was reduced by treating LNCaP and RWPE-1 cells for 48 hours with siRNA and RNA was collected and used for RNA-seq. (A) Venn diagram shows the number of genes that were differentially regulated by both siRNAs in either LNCaP or RWPE-1 cells, and regulated differentially between the two cell lines. (B) Validation of the RNA-seq data using RT-qPCR. The data shown are an average of at least two biological replicates for both RNA-seq and validation, and the significance was evaluated using paired samples Student's t-test, *< 0.05, **< 0.01, ***< 0.001.
Figure 5. Lipid degradation inhibitor perhexiline activates…
Figure 5. Lipid degradation inhibitor perhexiline activates cell death in prostate cancer cells
(A) Oil red O staining of LNCaP cells after 24 hours of perhexiline treatment. Data shown are an average (with SEM) of five biological replicates. The significance was evaluated using paired samples Student's t-test, *< 0.05. (B) LNCaP cells were treated with increasing doses of perhexiline and growth rate was followed using life-cell imaging. Data shown is an average of four biological replicates with SEM. Significance was evaluated using paired samples Student's t-test, *< 0.05, **< 0.01, ***< 0.001. (C) LNCaP cells were treated with perhexiline for 4 hours or 24 hours, and protein lysates were collected for western blotting. The data shown are representative of three biological replicates. Densitometry was used to evaluate signal intensity. (D) LNCaP cells were treated with perhexiline as indicated and cell death activation was evaluated using life-cell imaging detecting activation of caspases 3 and 7. Data shown is an average of four biological replicates with SEM. Significance was evaluated with paired samples Student's t-test, *< 0.05, **< 0.01. (E) LNCaP cells were treated with perhexiline alone or in combination with androgen deprivation therapy (either Abiraterone or MDV-3100) and growth rate was followed using life-cell imaging. Data shown are an average of four biological replicates with SEM. Significance was evaluated using paired samples Student's t-test, *< 0.05. Red stars indicate comparison between perhexiline and combinatorial treatment, while green stars indicate comparison between androgen deprivation and combinatorial treatment.

References

    1. Mills IG. Maintaining and reprogramming genomic androgen receptor activity in prostate cancer. Nature reviews Cancer. 2014;14:187–198.
    1. Rathkopf D, Scher HI. Androgen receptor antagonists in castration-resistant prostate cancer. Cancer journal. 2013;19:43–49.
    1. Sharma NL, Massie CE, Ramos-Montoya A, Zecchini V, Scott HE, Lamb AD, MacArthur S, Stark R, Warren AY, Mills IG, Neal DE. The androgen receptor induces a distinct transcriptional program in castration-resistant prostate cancer in man. Cancer cell. 2013;23:35–47.
    1. Geisbrecht BV, Zhang D, Schulz H, Gould SJ. Characterization of PECI, a novel monofunctional Delta(3), Delta(2)-enoyl-CoA isomerase of mammalian peroxisomes. The Journal of biological chemistry. 1999;274:21797–21803.
    1. Chang KH, Ercole CE, Sharifi N. Androgen metabolism in prostate cancer: from molecular mechanisms to clinical consequences. British journal of cancer. 2014;111:1249–1254.
    1. Ryan CJ, Smith MR, de Bono JS, Molina A, Logothetis CJ, de Souza P, Fizazi K, Mainwaring P, Piulats JM, Ng S, Carles J, Mulders PF, Basch E, et al. Abiraterone in metastatic prostate cancer without previous chemotherapy. The New England journal of medicine. 2013;368:138–148.
    1. Barfeld SJ, Itkonen HM, Urbanucci A, Mills IG. Androgen-regulated metabolism and biosynthesis in prostate cancer. Endocrine-related cancer. 2014;21:T57–66.
    1. Dimitrakopoulou-Strauss A, Strauss LG. PET imaging of prostate cancer with 11C-acetate. Journal of nuclear medicine. 2003;44:556–558.
    1. Zadra G, Photopoulos C, Loda M. The fat side of prostate cancer. Biochimica et biophysica acta. 2013;1831:1518–1532.
    1. Moon H, Hill MM, Roberts MJ, Gardiner RA, Brown AJ. Statins: protectors or pretenders in prostate cancer? Trends in endocrinology and metabolism. 2014;25:188–196.
    1. Baron A, Migita T, Tang D, Loda M. Fatty acid synthase: a metabolic oncogene in prostate cancer? Journal of cellular biochemistry. 2004;91:47–53.
    1. Kridel SJ, Axelrod F, Rozenkrantz N, Smith JW. Orlistat is a novel inhibitor of fatty acid synthase with antitumor activity. Cancer research. 2004;64:2070–2075.
    1. Mizushima N. Autophagy: process and function. Genes & development. 2007;21:2861–2873.
    1. Johansen T, Lamark T. Selective autophagy mediated by autophagic adapter proteins. Autophagy. 2011;7:279–296.
    1. Huang L, Min JN, Masters S, Mivechi NF, Moskophidis D. Insights into function and regulation of small heat shock protein 25 (HSPB1) in a mouse model with targeted gene disruption. Genesis. 2007;45:487–501.
    1. Kuner R, Falth M, Pressinotti NC, Brase JC, Puig SB, Metzger J, Gade S, Schafer G, Bartsch G, Steiner E, Klocker H, Sultmann H. The maternal embryonic leucine zipper kinase (MELK) is upregulated in high-grade prostate cancer. Journal of molecular medicine. 2013;91:237–248.
    1. Fernando HS, Sanders AJ, Kynaston HG, Jiang WG. WAVE3 is associated with invasiveness in prostate cancer cells. Urologic oncology. 2010;28:320–327.
    1. Teng Y, Ren MQ, Cheney R, Sharma S, Cowell JK. Inactivation of the WASF3 gene in prostate cancer cells leads to suppression of tumorigenicity and metastases. British journal of cancer. 2010;103:1066–1075.
    1. Huang da W, Sherman BT, Lempicki RA. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic acids research. 2009;37:1–13.
    1. Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nature protocols. 2009;4:44–57.
    1. Jung YS, Qian Y, Chen X. Examination of the expanding pathways for the regulation of p21 expression and activity. Cellular signalling. 2010;22:1003–1012.
    1. Ayad NG. CDKs give Cdc6 a license to drive into S phase. Cell. 2005;122:825–827.
    1. Erho N, Crisan A, Vergara IA, Mitra AP, Ghadessi M, Buerki C, Bergstralh EJ, Kollmeyer T, Fink S, Haddad Z, Zimmermann B, Sierocinski T, Ballman KV, et al. Discovery and validation of a prostate cancer genomic classifier that predicts early metastasis following radical prostatectomy. PloS one. 2013;8:e66855.
    1. Lionetti V, Stanley WC, Recchia FA. Modulating fatty acid oxidation in heart failure. Cardiovascular research. 2011;90:202–209.
    1. Kolwicz SC, Jr, Purohit S, Tian R. Cardiac metabolism and its interactions with contraction, growth, and survival of cardiomyocytes. Circulation research. 2013;113:603–616.
    1. Sorensen LB, Sorensen RN, Miners JO, Somogyi AA, Grgurinovich N, Birkett DJ. Polymorphic hydroxylation of perhexiline in vitro. British journal of clinical pharmacology. 2003;55:635–638.
    1. Ashrafian H, Horowitz JD, Frenneaux MP. Perhexiline. Cardiovascular drug reviews. 2007;25:76–97.
    1. Itkonen HM, Engedal N, Babaie E, Luhr M, Guldvik IJ, Minner S, Hohloch J, Tsourlakis MC, Schlomm T, Mills IG. UAP1 is overexpressed in prostate cancer and is protective against inhibitors of N-linked glycosylation. Oncogene. 2015;34:3744–3750.
    1. Itkonen HM, Minner S, Guldvik IJ, Sandmann MJ, Tsourlakis MC, Berge V, Svindland A, Schlomm T, Mills IG. O-GlcNAc transferase integrates metabolic pathways to regulate the stability of c-MYC in human prostate cancer cells. Cancer research. 2013;73:5277–5287.
    1. Mallik I, Davila M, Tapia T, Schanen B, Chakrabarti R. Androgen regulates Cdc6 transcription through interactions between androgen receptor and E2F transcription factor in prostate cancer cells. Biochimica et biophysica acta. 2008;1783:1737–1744.
    1. Beloribi-Djefaflia S, Vasseur S, Guillaumond F. Lipid metabolic reprogramming in cancer cells. Oncogenesis. 2016;5:e189.
    1. Itkonen HM, Gorad SS, Duveau DY, Martin SE, Barkovskaya A, Bathen TF, Moestue SA, Mills IG. Inhibition of O-GlcNAc transferase activity reprograms prostate cancer cell metabolism. Oncotarget. 2016;7:12464–12476. doi: 10.18632/oncotarget.7039.
    1. Camarda R, Zhou AY, Kohnz RA, Balakrishnan S, Mahieu C, Anderton B, Eyob H, Kajimura S, Tward A, Krings G, Nomura DK, Goga A. Inhibition of fatty acid oxidation as a therapy for MYC-overexpressing triple-negative breast cancer. Nature medicine. 2016;22:427–432.
    1. Ren XR, Wang J, Osada T, Mook RA, Jr, Morse MA, Barak LS, Lyerly HK, Chen W. Perhexiline promotes HER3 ablation through receptor internalization and inhibits tumor growth. Breast cancer research. 2015;17:20.
    1. Balgi AD, Fonseca BD, Donohue E, Tsang TC, Lajoie P, Proud CG, Nabi IR, Roberge M. Screen for chemical modulators of autophagy reveals novel therapeutic inhibitors of mTORC1 signaling. PloS one. 2009;4:e7124.
    1. Liu PP, Liu J, Jiang WQ, Carew JS, Ogasawara MA, Pelicano H, Croce CM, Estrov Z, Xu RH, Keating MJ, Huang P. Elimination of chronic lymphocytic leukemia cells in stromal microenvironment by targeting CPT with an antiangina drug perhexiline. Oncogene. 2016;35:5663–5673.
    1. Rodriguez-Enriquez S, Hernandez-Esquivel L, Marin-Hernandez A, El Hafidi M, Gallardo-Perez JC, Hernandez-Resendiz I, Rodriguez-Zavala JS, Pacheco-Velazquez SC, Moreno-Sanchez R. Mitochondrial free fatty acid beta-oxidation supports oxidative phosphorylation and proliferation in cancer cells. The international journal of biochemistry & cell biology. 2015;65:209–221.
    1. Foster BJ, Grotzinger KR, McKoy WM, Rubinstein LV, Hamilton TC. Modulation of induced resistance to adriamycin in two human breast cancer cell lines with tamoxifen or perhexiline maleate. Cancer chemotherapy and pharmacology. 1988;22:147–152.
    1. Ramu A, Fuks Z, Gatt S, Glaubiger D. Reversal of acquired resistance to doxorubicin in P388 murine leukemia cells by perhexiline maleate. Cancer research. 1984;44:144–148.
    1. Vella S, Penna I, Longo L, Pioggia G, Garbati P, Florio T, Rossi F, Pagano A. Perhexiline maleate enhances antitumor efficacy of cisplatin in neuroblastoma by inducing over-expression of NDM29 ncRNA. Scientific reports. 2015;5:18144.
    1. Nieman KM, Kenny HA, Penicka CV, Ladanyi A, Buell-Gutbrod R, Zillhardt MR, Romero IL, Carey MS, Mills GB, Hotamisligil GS, Yamada SD, Peter ME, Gwin K, et al. Adipocytes promote ovarian cancer metastasis and provide energy for rapid tumor growth. Nature medicine. 2011;17:1498–1503.
    1. Martinez-Outschoorn UE, Lin Z, Whitaker-Menezes D, Howell A, Lisanti MP, Sotgia F. Ketone bodies and two-compartment tumor metabolism: stromal ketone production fuels mitochondrial biogenesis in epithelial cancer cells. Cell cycle. 2012;11:3956–3963.
    1. Martinez-Outschoorn UE, Sotgia F, Lisanti MP. Power surge: supporting cells “fuel” cancer cell mitochondria. Cell metabolism. 2012;15:4–5.
    1. Horowitz JD, Sia ST, Macdonald PS, Goble AJ, Louis WJ. Perhexiline maleate treatment for severe angina pectoris—correlations with pharmacokinetics. International journal of cardiology. 1986;13:219–229.
    1. Cole PL, Beamer AD, McGowan N, Cantillon CO, Benfell K, Kelly RA, Hartley LH, Smith TW, Antman EM. Efficacy and safety of perhexiline maleate in refractory angina. A double-blind placebo-controlled clinical trial of a novel antianginal agent. Circulation. 1990;81:1260–1270.
    1. Horowitz JD, Chirkov YY. Perhexiline and hypertrophic cardiomyopathy: a new horizon for metabolic modulation. Circulation. 2010;122:1547–1549.
    1. Fang YH, Piao L, Hong Z, Toth PT, Marsboom G, Bache-Wiig P, Rehman J, Archer SL. Therapeutic inhibition of fatty acid oxidation in right ventricular hypertrophy: exploiting Randle's cycle. Journal of molecular medicine. 2012;90:31–43.
    1. Trapnell C, Roberts A, Goff L, Pertea G, Kim D, Kelley DR, Pimentel H, Salzberg SL, Rinn JL, Pachter L. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nature protocols. 2012;7:562–578.

Source: PubMed

3
Abonneren