, AR (L-003400-00), DECR1 (L-009642-00-005), as well as non-targeting siRNA (D-001810-01-20) were purchased from Dharmacon (Dharmacon, Horizon inspired cell solutions, transfection. Cells were seeded in 6-well plates to reach 70% confluence and allowed to attach overnight. ON-TARGETplus smartpool siRNAs against AMPKa (L-005027-00)

, USA) using a nucleofector (Amaxa Biosystems, Lonza, Basel, Switzerland) according to the manufacturer's instructions. Cells were then cultured in the presence of 1 µg ml ?1 of puromycin, Generation of DECR1 stable KO cells. DECR1 deletion was performed using the CRISPR CAS9 gene editing technology

, Final cell number was normalised to the initial cell count obtained at T0. qPCR analysis. RNA was extracted from cells (70-80% confluence) using the RNeasy Mini Kit (Qiagen, Hilden, Germany) with on-column DNase digestion (RNase-Free DNase Set, Cell proliferation. 6-8 × 10 5 cells were seeded in 6-well plates and allowed to attach for 16 h

. Immunoblotting, Membrane was blocked in 5% milk-TBST for 1 h and probed overnight with primary antibodies (see Supplementary Table 2) diluted in 5% BSA-TBST. The membrane was then washed three times with TBST, incubated with respective HRP-conjugated secondary antibodies diluted in 5% milk-TBST, washed another three times with TBST and revealed using the ECL kit, SDS buffer (1% SDS supplemented with protease and phosphatase inhibitors) and protein concentration was determined using the BCA protein assay kit

, The next day, coverslips were washed three times with PBS-Tween, incubated with fluorophore-coupled secondary antibodies (Abcam, Cambridge, UK) and washed again three times with PBS. Coverslips were mounted using Diamond Prolong with DAPI, Immunofluorescence. Cells were seeded on coverslips in 24-well plates to reach 50% confluence and allowed to attach overnight

M. Burlington, M. A. , -. Cat-c-3?-[r]-and, and 5. Gtt-g-3?, The DNA/protein complexes were washed four times in IP Wash buffer (100 mM Tris-HCl pH 8.0; 500 mM LiCl 1%; Triton X100; 1% deoxycholic acid. After reversal of crosslinking, the immunoprecipitated DNA was purified by a regular DNA extraction protocol and analysed employing RT-qPCR with the SYBR-Green Takara, Chromatin immunoprecipitation (ChIP). Chromatin was prepared with the tru-ChIP? Chromatin Shearing Kit (Covaris, Brighton, UK) according to manufacturer's instructions. Each sample was sonicated for 10 min using Covaris sonicator. ChIP were performed using the IP-Star Compact Automated System

, Statistical analysis. Statistical analyses were performed using GraphPad PRISM software v7.05

, 51 (ENZ) organoids examined over two independent biological experiments. Panels d, e: n = 3 independent biological experiments, Data reproducibility. Figure 1: Panels a, b, f, h: representative image from three independent biological experiments. Panel c (top): n = 6 independent biological experiments. Panel c (bottom): n = 55 (LN), 40 (BIC), vol.51

R. L. Siegel, K. D. Miller, and A. Jemal, Cancer statistics, vol.68, pp.7-30, 2018.

P. A. Watson, V. K. Arora, and C. L. Sawyers, Emerging mechanisms of resistance to androgen receptor inhibitors in prostate cancer, Nat. Rev. Cancer, vol.15, pp.701-711, 2015.

G. J. Kolvenbag, G. R. Blackledge, and K. Gotting-smith, Bicalutamide (Casodex) in the treatment of prostate cancer: history of clinical development, Prostate, vol.34, pp.61-72, 1998.

C. Tran, Development of a second-generation antiandrogen for treatment of advanced prostate cancer, Science, vol.324, pp.787-790, 2009.

D. F. Penson, Enzalutamide versus bicalutamide in castration-resistant prostate cancer: the STRIVE trial, J. Clin. Oncol, vol.34, pp.2098-2106, 2016.

N. D. Shore, Efficacy and safety of enzalutamide versus bicalutamide for patients with metastatic prostate cancer (TERRAIN): a randomised, doubleblind, phase 2 study, Lancet Oncol, vol.17, pp.153-163, 2016.

T. M. Beer, Enzalutamide in metastatic prostate cancer before chemotherapy, N. Engl. J. Med, vol.371, pp.424-433, 2014.
URL : https://hal.archives-ouvertes.fr/hal-02169779

H. I. Scher, Increased survival with enzalutamide in prostate cancer after chemotherapy, N. Engl. J. Med, vol.367, pp.1187-1197, 2012.

M. Hussain, Enzalutamide in men with nonmetastatic, castrationresistant prostate cancer, N. Engl. J. Med, vol.378, pp.2465-2474, 2018.

M. R. Smith, Apalutamide treatment and metastasis-free survival in prostate cancer, N. Engl. J. Med, vol.378, pp.1408-1418, 2018.

T. Yoshida, Antiandrogen bicalutamide promotes tumor growth in a novel androgen-dependent prostate cancer xenograft model derived from a bicalutamide-treated patient, Cancer Res, vol.65, pp.9611-9616, 2005.

M. D. Balbas, Overcoming mutation-based resistance to antiandrogens with rational drug design, vol.2, p.499, 2013.

J. D. Joseph, A clinically relevant androgen receptor mutation confers resistance to second-generation antiandrogens enzalutamide and ARN-509, Cancer Discov, vol.3, pp.1020-1029, 2013.

M. Korpal, An F876L mutation in androgen receptor confers genetic and phenotypic resistance to MDV3100 (enzalutamide), Cancer Discov, vol.3, pp.1030-1043, 2013.

T. Visakorpi, In vivo amplification of the androgen receptor gene and progression of human prostate cancer, Nat. Genet, vol.9, pp.401-406, 1995.

S. Sun, Castration resistance in human prostate cancer is conferred by a frequently occurring androgen receptor splice variant, J. Clin. Investig, vol.120, pp.2715-2730, 2010.

V. K. Arora, Glucocorticoid receptor confers resistance to antiandrogens by bypassing androgen receptor blockade, Cell, vol.155, pp.1309-1322, 2013.

C. E. Massie, The androgen receptor fuels prostate cancer by regulating central metabolism and biosynthesis, EMBO J, vol.30, pp.2719-2733, 2011.

E. Eidelman, J. Twum-ampofo, J. Ansari, and M. M. Siddiqui, The metabolic phenotype of prostate cancer, Front. Oncol, vol.7, p.131, 2017.

G. Zadra and M. Loda, Metabolic vulnerabilities of prostate cancer: diagnostic and therapeutic opportunities. Cold Spring Harbor Perspect, 2018.

X. Wu, G. Daniels, P. Lee, and M. E. Monaco, Lipid metabolism in prostate cancer, Am. J. Clin. Exp. Urol, vol.2, pp.111-120, 2014.

F. Pampaloni, E. G. Reynaud, and E. H. Stelzer, The third dimension bridges the gap between cell culture and live tissue, Nat. Rev. Mol. Cell Biol, vol.8, pp.839-845, 2007.

F. Bovenga, C. Sabba, and A. Moschetta, Uncoupling nuclear receptor LXR and cholesterol metabolism in cancer, Cell Metab, vol.21, pp.517-526, 2015.

H. G. Nguyen, Targeting autophagy overcomes Enzalutamide resistance in castration-resistant prostate cancer cells and improves therapeutic response in a xenograft model, Oncogene, vol.33, pp.4521-4530, 2014.

S. J. Barfeld, c-Myc antagonises the transcriptional activity of the androgen receptor in prostate cancer affecting key gene networks, EBioMedicine, vol.18, pp.83-93, 2017.

R. Patel, Sprouty2 loss-induced IL6 drives castration-resistant prostate cancer through scavenger receptor B1, EMBO Mol. Med, 2018.

D. Robinson, Integrative clinical genomics of advanced prostate cancer, Cell, vol.162, p.454, 2015.

B. S. Taylor, Integrative genomic profiling of human prostate cancer, Cancer Cell, vol.18, pp.11-22, 2010.

W. H. Kim, C. H. Choi, S. K. Kang, C. H. Kwon, and Y. K. Kim, Ceramide induces non-apoptotic cell death in human glioma cells, Neurochem. Res, vol.30, pp.969-979, 2005.

L. Magtanong, Exogenous monounsaturated fatty acids promote a ferroptosis-resistant cell state, Cell Chem. Biol, vol.26, pp.420-432, 2019.

W. S. Yang, Peroxidation of polyunsaturated fatty acids by lipoxygenases drives ferroptosis, Proc. Natl Acad. Sci. USA, vol.113, pp.4966-4975, 2016.

M. J. Hangauer, Drug-tolerant persister cancer cells are vulnerable to GPX4 inhibition, Nature, vol.551, pp.247-250, 2017.

S. Koljenovic, Tissue characterization using high wave number Raman spectroscopy, J. Biomed. Opt, vol.10, p.31116, 2005.

E. G. Bluemn, Androgen receptor pathway-independent prostate cancer is sustained through FGF signaling, Cancer Cell, vol.32, pp.474-489, 2017.

N. L. Sharma, The androgen receptor induces a distinct transcriptional program in castration-resistant prostate cancer in man, Cancer Cell, vol.23, pp.35-47, 2013.

J. Hoefer, Critical role of androgen receptor level in prostate cancer cell resistance to new generation antiandrogen enzalutamide, Oncotarget, vol.7, pp.59781-59794, 2016.

S. Kregel, Acquired resistance to the second-generation androgen receptor antagonist enzalutamide in castration-resistant prostate cancer, Oncotarget, vol.7, pp.26259-26274, 2016.

J. Li, Aberrant corticosteroid metabolism in tumor cells enables GR takeover in enzalutamide resistant prostate cancer, 2017.

Y. Cui, Upregulation of glucose metabolism by NF-kappaB2/p52 mediates enzalutamide resistance in castration-resistant prostate cancer cells, Endocr. Relat. Cancer, vol.21, pp.435-442, 2014.

L. Wang, Co-targeting hexokinase 2-mediated Warburg effect and ULK1-dependent autophagy suppresses tumor growth of PTEN-and TP53-deficiency-driven castration-resistant prostate cancer, vol.7, pp.50-61, 2016.

D. A. Bader, Mitochondrial pyruvate import is a metabolic vulnerability in androgen receptor-driven prostate cancer, Nat. Metab, vol.1, pp.70-85, 2019.

F. Rohrig and A. Schulze, The multifaceted roles of fatty acid synthesis in cancer, Nat. Rev. Cancer, vol.16, pp.732-749, 2016.

A. Carracedo, L. C. Cantley, and P. P. Pandolfi, Cancer metabolism: fatty acid oxidation in the limelight, Nat. Rev. Cancer, vol.13, pp.227-232, 2013.

S. Shah, Targeting ACLY sensitizes castration-resistant prostate cancer cells to AR antagonism by impinging on an ACLY-AMPK-AR feedback mechanism, Oncotarget, vol.7, pp.43713-43730, 2016.

K. Zaugg, Carnitine palmitoyltransferase 1C promotes cell survival and tumor growth under conditions of metabolic stress, Genes Dev, vol.25, pp.1041-1051, 2011.

Z. T. Schafer, Antioxidant and oncogene rescue of metabolic defects caused by loss of matrix attachment, Nature, vol.461, pp.109-113, 2009.

R. Camarda, Inhibition of fatty acid oxidation as a therapy for MYCoverexpressing triple-negative breast cancer, Nat. Med, vol.22, pp.427-432, 2016.

I. Samudio, Pharmacologic inhibition of fatty acid oxidation sensitizes human leukemia cells to apoptosis induction, J. Clin. Investig, vol.120, pp.142-156, 2010.

H. M. Itkonen, Lipid degradation promotes prostate cancer cell survival, Oncotarget, vol.8, pp.38264-38275, 2017.

I. R. Schlaepfer, Lipid catabolism via CPT1 as a therapeutic target for prostate cancer, Mol. Cancer Ther, vol.13, pp.2361-2371, 2014.

T. W. Flaig, Lipid catabolism inhibition sensitizes prostate cancer cells to antiandrogen blockade, Oncotarget, vol.8, pp.56051-56065, 2017.

J. S. Myers, A. K. Von-lersner, and Q. X. Sang, Proteomic upregulation of fatty acid synthase and fatty acid binding protein 5 and identification of cancerand race-specific pathway associations in human prostate cancer tissues, J. Cancer, vol.7, pp.1452-1464, 2016.

A. P. Khan, Quantitative proteomic profiling of prostate cancer reveals a role for miR-128 in prostate cancer, Mol. Cell. Proteom, vol.9, pp.298-312, 2010.

J. Ursini-siegel, Elevated expression of DecR1 impairs ErbB2/Neuinduced mammary tumor development, Mol. Cell. Biol, vol.27, pp.6361-6371, 2007.

I. J. Miinalainen, Mitochondrial 2,4-dienoyl-CoA reductase deficiency in mice results in severe hypoglycemia with stress intolerance and unimpaired ketogenesis, PLoS Genet, vol.5, p.1000543, 2009.

K. Vriens, Evidence for an alternative fatty acid desaturation pathway increasing cancer plasticity, Nature, vol.566, pp.403-406, 2019.

M. M. Gilligan, Aspirin-triggered proresolving mediators stimulate resolution in cancer, Proc. Natl Acad. Sci. USA, vol.116, pp.6292-6297, 2019.

R. Volmer, K. Van-der-ploeg, and D. Ron, Membrane lipid saturation activates endoplasmic reticulum unfolded protein response transducers through their transmembrane domains, Proc. Natl Acad. Sci. USA, vol.110, pp.4628-4633, 2013.

N. Ho, C. Xu, and G. Thibault, From the unfolded protein response to metabolic diseases-lipids under the spotlight, J. Cell Sci, 2018.

D. J. Naylor, N. J. Hoogenraad, and P. B. Hoj, Characterisation of several Hsp70 interacting proteins from mammalian organelles, Biochim. et. Biophys. Acta, vol.1431, pp.443-450, 1999.

J. Rappsilber, M. Mann, and Y. Ishihama, Protocol for micro-purification, enrichment, pre-fractionation and storage of peptides for proteomics using StageTips, Nat. Protoc, vol.2, pp.1896-1906, 2007.

J. Cox and M. Mann, MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification, Nat. Biotechnol, vol.26, pp.1367-1372, 2008.

J. Cox, Andromeda: a peptide search engine integrated into the MaxQuant environment, J. Proteome Res, vol.10, pp.1794-1805, 2011.

C. Uniprot, The Universal Protein Resource (UniProt) in 2010, Nucleic Acids Res, vol.38, pp.142-148, 2010.

J. Cox, Accurate proteome-wide label-free quantification by delayed normalization and maximal peptide ratio extraction, termed MaxLFQ, Mol. Cell. Proteom, vol.13, pp.2513-2526, 2014.

S. Tyanova, The Perseus computational platform for comprehensive analysis of (prote)omics data, Nat. Methods, vol.13, pp.731-740, 2016.

N. Rath, Rho kinase inhibition by AT13148 blocks pancreatic ductal adenocarcinoma invasion and tumor growth, Cancer Res, vol.78, pp.3321-3336, 2018.

G. M. Mackay, L. Zheng, N. J. Van-den-broek, and E. Gottlieb, Analysis of cell metabolism using LC-MS and isotope tracers, Methods Enzymol, vol.561, pp.171-196, 2015.

H. Tsugawa, Comprehensive identification of sphingolipid species by in silico retention time and tandem mass spectral library, J. Cheminform, vol.9, p.19, 2017.

T. Kind, LipidBlast in silico tandem mass spectrometry database for lipid identification, Nat. Methods, vol.10, pp.755-758, 2013.

P. D. Hutchins, J. D. Russell, and J. J. Coon, LipiDex: an integrated software package for high-confidence lipid identification, Cell Syst, vol.6, pp.621-625, 2018.

S. Christen, Breast cancer-derived lung metastases show increased pyruvate carboxylase-dependent anaplerosis, Cell Rep, vol.17, pp.837-848, 2016.

D. Lorendeau, Dual loss of succinate dehydrogenase (SDH) and complex I activity is necessary to recapitulate the metabolic phenotype of SDH mutant tumors, Metab. Eng, vol.43, pp.187-197, 2017.

M. Van-gorsel, I. Elia, and S. M. Fendt, 13)C tracer analysis and metabolomics in 3D cultured cancer cells, Methods Mol. Biol, vol.1862, pp.53-66, 2019.

C. A. Fernandez, C. Des-rosiers, S. F. Previs, F. David, and H. Brunengraber, Correction of 13C mass isotopomer distributions for natural stable isotope abundance, J. Mass Spectrom, vol.31, pp.255-262, 1996.

C. Kilkenny, Animal research: reporting in vivo experiments-the ARRIVE guidelines, J. Cereb. Blood Flow. Metab, vol.31, pp.991-993, 2011.