Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

1 PGC1α suppresses prostate cancer cell invasion

2 through ERRα transcriptional control

3 Lorea Valcarcel-Jimenez1,8, Alice Macchia1,8, Eva Crosas-Molist2,3, Ariane Schaub-

4 Clerigué1 , Laura Camacho1,5 , Natalia Martín-Martín1,4, Paolo Cicogna1, Cristina Viera-

5 Bardón1,4, Sonia Fernández-Ruiz1,4, Irene Rodriguez-Hernandez2,3, Ivana Hermanova1,

6 Ianire Astobiza1,4, Ana R. Cortazar1,4, Jon Corres-Mendizabal1, Antonio Gomez-Muñoz5,

7 Victoria Sanz-Moreno2,3, Verónica Torrano1,4,5,7,9, Arkaitz Carracedo1,4,5,6,7,9

8 Running title: PGC1α-ERRα suppresses MYC and prostate cancer invasion

9 1CIC bioGUNE, Bizkaia Technology Park, 801a bld., 48160 Derio, Bizkaia, Spain

10 2Barts Cancer Institute, John Vane Science Building, Charterhouse Square, Queen Mary University

11 of London, London EC1M 6BQ, UK

12 3Randall Centre for Cell and Molecular Biophysics, New Hunt's House, Guy's Campus, King's

13 College London, London SE1 1UL, UK

14 4CIBERONC

15 5Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), P.

16 O. Box 644, E-48080 Bilbao, Spain.

17 6Ikerbasque, Basque foundation for science, 48011 Bilbao, Spain

18 7Correspondence to: Verónica Torrano, [email protected] (Phone number (+34)946015925)

19 and Arkaitz Carracedo, [email protected] (Phone number (+34)944061308)

20 8 L. Valcarcel-Jimenez and A. Macchia contributed equally to this work

21 9 V. Torrano and A. Carracedo contributed equally to this work

22 No conflict of interest to declare

1

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

23 Abstract

24 The peroxisome proliferator-activated receptor gamma 1 alpha (PGC1α) is a

25 prostate tumor suppressor that controls the balance between anabolism and catabolism.

26 PGC1A downregulation in prostate cancer is causally associated with the development of

27 metastasis. Here we show that the transcriptional complex formed by PGC1α and

28 Estrogen related receptor 1 alpha (ERRα) controls the aggressive properties of prostate

29 cancer cells. PGC1α expression significantly decreased migration and invasion of various

30 prostate cancer cell lines. This phenotype was consistent with remarkable cytoskeletal

31 remodeling and inhibition of integrin alpha 1 and beta 4 expression both in vitro and in

32 vivo. CRISPR/Cas9-based deletion of ERRα suppressed PGC1α regulation of cytoskeletal

33 organization and invasiveness. Mechanistically, PGC1α expression decreased MYC levels

34 and activity prior to inhibition of invasiveness. In addition, PGC1α and ERRα associated at

35 the MYC promoter, supporting the inhibitory activity PGC1α. The inverse correlation

36 between PGC1α-ERRα activity and MYC levels was corroborated in multiple prostate

37 cancer datasets. Altogether, these results support that PGC1α-ERRα functions as a tumor

38 suppressive transcriptional complex through the regulation of metabolic and signaling

39 events.

40 Statement of significance

41 Findings describe how downregulation of the prostate tumor suppressor PGC1 drives

42 invasiveness and migration of prostate cancer cells.

2

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

43 Introduction

44 The process of cellular transformation stems from the acquisition of genomic aberrations

45 that altogether change the response of normal cells and enable them with hallmarks of

46 cancer (1,2). The mutational landscape changes within and among tumors and along time

47 following evolutionary principles (3). In addition, non-genomic alterations harness great

48 relevance in the process of cancer progression. Indeed, transcriptional regulation in cancer

49 is an emerging aspect that provides a feasible explanation to the rapid adaptation of

50 transformed cells to hostile environments (4). Yet, the control of oncogenic and tumor

51 suppressive transcriptional programs remains poorly characterized.

52 Transcriptional co-regulators encompass a family of versatile modulators of

53 expression (5). These harbor the capacity of controlling distinct transcriptional

54 programs based on their partner transcription factors. In turn, transcriptional co-regulators

55 operate in a tissue and context-specific manner, thus revealing them as major players in

56 cell and organismal homeostasis. Among this family of , the Peroxisome proliferator-

57 activator receptor (PPAR) gamma co-activator 1 alpha (PGC1α) controls biological

58 responses in health and disease (6,7). PGC1α is a tightly regulated that interacts

59 with a variety of transcription factors, including Estrogen related receptor 1 alpha (ERRα),

60 PPARs and Nuclear factor erythroid 2 like 2 (NFE2L2, NRF2) (6). As a consequence,

61 PGC1α coordinates metabolic and antioxidant responses, which account for its relevance

62 in diabetes, neurodegeneration, cardiomyopathy and cancer (7,8).

63 The role of PGC1α in cancer is largely tumor type and context-dependent. On the one

64 hand, this transcriptional co-regulator favors survival, proliferation, stem cell maintenance

65 and therapy resistance in pancreatic tumors, breast cancer and melanoma cells (9-14). On

66 the other hand, we and others have demonstrated that PGC1α expression is reduced in

3

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

67 renal and prostate carcinoma, as well as in metastatic melanoma, where it opposes the

68 acquisition of aggressive features (15-17). The predominant mechanism of action of

69 PGC1α in cancer biology is ascribed to the regulation of metabolism. This co-regulator

70 promotes the expression of genes that mediate mitochondrial biogenesis, oxidative

71 metabolism and the production of glutathione. In turn, PGC1α enhances the oxidative

72 utilization of nutrients and antioxidant production. However, emerging data suggest that a

73 fraction of the activities of PGC1α neither relies on the regulation of metabolism nor on its

74 main partner, ERRα (16).

75 In prostate cancer (PCa), PGC1α suppresses cell proliferation, anchorage-independent

76 growth, tumor burden and metastasis (17). This co-regulator is profoundly downregulated

77 in localized PCa, with a further decrease in metastatic specimens (17). Moreover, reduced

78 PGC1α expression is associated to shorter time to biochemical recurrence after surgery,

79 pointing at the relevance of this gene in the control of PCa aggressiveness.

80 Mechanistically, we previously showed that PGC1α requires the presence of ERRα to

81 suppress PCa cell proliferation and metastatic outgrowth, which was consistent with the

82 reduction of biosynthetic capacity of PGC1α re-expressing cells and the elevation of

83 nutrient catabolism (17). Moreover, a recent study revealed that the metabolic control of

84 polyamine synthesis underlies the regulation of prostate cancer aggressiveness by this co-

85 activator (18).

86 The metastatic process requires the acquisition of discreet capacities beyond cell

87 proliferation. Specifically, the motility and invasive capacity of cancer cells is paramount for

88 the achievement of metastasis (19). Stemming from this notion, in this study we evaluated

89 the contribution of PGC1α to the acquisition of these features in PCa cells. Our analysis

90 uncovers an ERRα-dependent activity of the co-activator that suppresses the acquisition of

91 invasive properties required for PCa aggressiveness.

4

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

92 Materials and Methods

93 Reagents

94 Doxycycline hyclate (Sigma #D9891) was used to induce or silencing in

95 vectors under tetracycline control. Puromycin (Sigma #P8833) and blasticidin (Invitrogen

96 #R210-01) were used for cell selection after lentiviral transfection.

97

98 Cell culture

99 Human prostate carcinoma cell lines PC3 and DU145 were purchased from Leibniz-Institut

100 DSMZ - Deutsche Sammlung von Mikroorganismen und Zellkulturen GmbH, who provided

101 authentication certificate. Cell lines where periodically subjected to microsatellite-based

102 identity validation. None of the cell lines used in this study were found in the database of

103 commonly misidentified cell lines maintained by ICLAC and NCBI Biosample. 293FT cells

104 were used for lentiviral production. All cell lines were routinely monitored for mycoplasma

105 contamination. DU145, PC3 and 293FT cell lines were maintained in DMEM media

106 supplemented with 10 % (v/v) fetal bovine serum (FBS) and 1 % (v/v) penicillin-

107 streptomycin. For PGC1A expression, cells were transduced with a modified TRIPZ

108 (Dharmacon) doxycycline inducible lentiviral construct in which the RFP and miR30 region

109 was substituted by HA-Flag-Pgc1a (9). For ESRRA deletion, sgRNA constructs targeting

110 ESRRA (sgERRα#1: 5´CTCCGGCTACCACTATGGTGTGG3´; sgERRα#2:

111 3´AGGAACCCTTTGGACTGTCAGGG5´) were designed using Crispor software

112 (crispor.tefor.net) and cloned in a lentiviral vector purchased from addgene LentiCRISPR

113 V2 (a gift from Mohan Babu, Addgene plasmid # 83480). Lentiviral vector expressing a

114 validated shRNA against human MYC from the Mission® shRNA Library

115 (TRCN0000039642) was subcloned in a Plko Tet On inducible system (following the

5

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

116 strategy provided by Dr. Dmitri Wiederschain (20), Addgene plasmid # 21915). Cells were

117 transfected with lentiviral vectors following standard procedures, and viral supernatant was

118 used to infect cells. Selection was done using puromycin (2 μg ml-1) or blasticidin (for

119 LentiCRISPR V2, 10 μg ml-1) for 3 or 5 days, respectively.

120

121 Animals

122 All mouse experiments were carried out following the ethical guidelines established by the

123 Biosafety and Welfare Committee at CIC bioGUNE. The procedures employed were

124 carried out following the recommendations from AAALAC. Xenograft experiments were

125 performed as previously described (17), injecting one million cells per tumor in two flanks

126 of Hsd:AthymicNude-Foxn1nu “Nude” mouse (Envigo). Once tumors reached an average

127 of 100mm3, animals were assigned to chow or doxycycline diet regime (Research diets,

128 D12100402) and tumor volume was monitored with external caliper. After euthanasia,

129 tumors were weighed, tissue was fresh frozen or paraffin embedded, and histological

130 evaluation of a Haematoxylin and eosin (H&E) stained sections was performed.

131 Proliferation was assessed in paraffin embedded tissue samples by using Ki67 antibody

132 (MA5-14520, Thermo Scientific).

133

134 Cellular and molecular assays

135 Cell number quantification with crystal violet was performed as referenced (21).

136 Cell morphology and stress fiber content were examined staining the cells with fluorescent

137 phalloidin (ThermoFisher F432; 1:400 dilution), a high affinity F-actin probe. Images were

138 taken with AxioImager D1 microscope at 200x for cell area analysis (FiJi Software) or at

6

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

139 400x for stress fiber quantification. Immunofluorescence detection and quantification of p-

140 MLC (Ser19) was performed as referenced (22). Briefly, cells were fixed with 4%

141 formaldehyde, permeabilised with 0.3% Triton and incubated with primary antibody (p-

142 MLC Ser19, CST #3672) overnight. Cells were then stained with secondary Alexa Fluor-

143 488 or 647 anti-rabbit (Life Technologies), Alexa Fluor 546-phalloidin for F-actin detection

144 (Life Technologies) and DAPI (Thermo Fisher D1306; 1:10,000 dilution).

145 For adhesion assays cells were plated (40,000 cells/well) on a 12-well plate previously

146 coated with rat tail collagen I (Corning 354236) at 50 μg/mL (diluted in 0.02 N of acetic

147 acid) during 1 hour. After 30 minutes, plates were washed twice with PBS, fixed with 10%

148 formalin and stained with crystal violet as previously described (17).

149 Transwell invasion assay was carried out using matrigel-coated chambers (BD CioCoatTM

150 #354480). Cells (50,000 cells/well) were re-suspended in 0.1 % FBS DMEM and seeded in

151 the upper part of the chamber. In the bottom part of the well 1.4 mL of complete DMEM

152 were added. Plates were maintained at 37 ºC and 5 % CO2 for 48 hours. Invasion was

153 stopped washing the well twice with PBS and using a cotton bud to remove the remaining

154 cell of the upper part of the membrane, being careful not to compromise the matrigel. The

155 membrane was fixed with 10 % formalin (15 minutes at 4 ºC) and stained with crystal violet

156 (Sigma C3886; 0.1% crystal violet in 20% methanol). Cells were counted under the

157 microscope. For transwell migration, chambers with membranes of 8 m pores (BD Falcon

158 351185) were used. Cell plating as well as washing and fixation conditions were the same

159 as in the invasion assay, but cells were fixed after 24 hours.

160 Spheroid cell culture and 3D invasion assays were performed as previously described

161 (23). Briefly, cells (700 cells/drop) were maintained in drops (25 L/drop) with DMEM and

162 6 % methylcellulose (Sigma M0387) on the cover of a 100 mm culture plate. Drops were

163 incubated at 37 ºC and 5 % CO2 for 48 hours. Once formed, spheroids were collected,

7

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

164 resuspended in collagen I solution (Adanced BioMatrix PureCol®) and added to 12-well

165 plates. After 4h complete media was then added on top of the well and day 0 pictures were

166 taken. For invasive growth quantification, increase in area occupied by the spheroids

167 between day 0 and day 2 was calculated using FiJi software. For 3D invasion assays, cells

168 were resuspended in a FBS-free bovine collagen I solution at 2.3 mg/mL in a 1:1

169 proportion, to a final concentration of 15000 cells per 100 µL of matrix and spin down in a

170 96-well plate. After matrix polymerization, 10% FBS-containing media was added on top.

171 Cells were fixed after 24 h. The 3D invasion index was calculated counting the number of

172 cells at 50 µm and 100 µm divided by the number of cells at the bottom. Images for 3D

173 invasion were done using a Zeiss 710 confocal microscope and cell counting was

174 analysed using FiJi Software.

175 Western blot was performed as previously described (9). Briefly, cells were seeded on 6-

176 well plates and 4 days after seeding cell lysates were prepared with RIPA buffer (50mM

177 TrisHCl pH 7.5, 150 mM NaCl, 1mM EDTA, 0.1 % SDS, 1 % Nonidet P40, 1 % sodium

178 deoxycholate, 1 mM Sodium Fluoride, 1 mM sodium orthovanadate, 1 mM beta-

179 glycerophosphate and protease inhibitor cocktail; Roche). Antibodies: PGC1α H300 (Santa

180 Cruz Technology #sc-13067), ERRα (CST #13826), ITGβ1 (CST #34981S), Caveolin-1

181 (BD Bioscience ref: 142610059), β-Actin (CST #3700S), phospho-Cofilin (CST #3313),

182 Cofilin (CST #5175), GAPDH (CST #2118), c-MYC (MYC, CST #13987S), ITGβ4 (CST

183 #14803), ITGα3 (Santa Cruz Technology #sc-374242), ITGα6 (CST #3750S), phospho-

184 Src (Life Technologies ref: 44660G; p-Src Tyr419) and Src 36D10 (CST #2109). All were

185 used at a 1:1000 dilution except from β-Actin at 1:2000. Mouse and rabbit secondary

186 antibodies were purchased from Jackson Immuno Research. After standard SDS-PAGE

187 and Western blotting techniques, proteins were visualized using the ECL system in the

188 iBrightTM FL1000 Imaging System.

8

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

189 The cytoskeleton phospho-antibody array was performed following Tebu-bio protocol

190 (https://www.tebu-bio.com). Briefly, 5 million induced and non-induced cells were collected

191 and the cell pellet was frozen for further analysis by Tebu-bio services. Over 141

192 antibodies were present in the screening for phosphorylation rate of main cytoskeleton

193 proteins.

194 RNA was extracted using NucleoSpin® RNA isolation kit from Macherey-Nagel (Ref:

195 740955.240C). For xenograft samples, a Trizol-based implementation of the NucleoSpin®

196 RNA isolation kit protocol was used as reported (24). For all cases, 1 μg of total RNA was

197 used for cDNA synthesis using qScript cDNA Supermix from Quanta (Ref: 95048).

198 Quantitative Real Time PCR (qRTPCR) was performed as previously described (9).

199 Universal Probe Library (Roche) primers and probes employed are detailed in

200 supplementary Table S1. All qRTPCR data presented were normalized using GAPDH

201 (Hs02758991_g1 from Applied Biosystems).

202

203 ChIP

204 Chromatin Immunoprecipitation (ChIP) was performed using the SimpleChIP® Enzymatic

205 Chromatin IP Kit (Cat: 9003, Cell Signalling Technology, Inc). Four million PC3 TRIPZ-

206 Pgc1a cells per immunoprecipitation were grown in 150 mm dishes either with or without

207 0.5 µg mL-1 doxycycline during 16 hours. Cells were cross-linked with 37 % formaldehyde

208 for 10 min at room temperature. Glycine was added to dishes, and cells incubated for 5

209 min at room temperature. Cells were then washed twice with ice-cold PBS and scraped

210 into PBS+PIC. Pelleted cells were lysed and nuclei were harvested following the

211 manufacturer’s instructions. Nuclear lysates were digested with micrococcal nuclease for

212 20 min at 37 °C and then sonicated in 500 μl aliquots on ice for 6 pulses of 20 s using a

9

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

213 Branson sonicator. Cells were held on ice for at least 20 s between sonications. Lysates

214 were clarified at 11000 × g for 10 min at 4 °C, and chromatin was stored at -80 °C. HA-Tag

215 polyclonal antibody (CST #3724), anti-ERRα antibody (CST #13826) and IgG antibody

216 (CST #2729), were incubated overnight (4 ºC) with rotation and protein G magnetic beads

217 were incubated 2 hours (4 ºC). Washes and elution of chromatin were performed following

218 manufacturer’s instructions. DNA quantification was carried out using a Viia7 Real-Time

219 PCR System (Applied Biosystems) with SybrGreen reagents and primers that amplify a

220 PGC1A binding region to MYC promoter (shown in supplementary table S2).

221

222 Bioinformatic analysis and statistics

223 Bioinformatic analysis containing patient data was performed using the web-based

224 interface Cancertool (25).

225 For each available patient dataset, the values of PGC1α-ERRα signature were calculated

226 from the average of the expression signal of those genes that are part of the

227 aforementioned signature. (ACACB, ACSL4, ATP1B1, GSTM4, ISCU, LAMB2, NNT,

228 PPIC, SOD2, SUCLA2). In the case of PPARGC1A /NRIP1 ratio, we calculated the

229 average expression value of PPARGC1A, and, as values are log2 scaled, subtracted the

230 average expression value of NRIP1. R software (https://cran.r-project.org/), version 3.5.1,

231 has been used for these calculations, together with ggplot2 package (https://cran.r-

232 project.org/web/packages/ggplot2) in order to perform the corresponding graphs.

233 Individual gene expression patters in Patient dataset, as well as pairwise correlation

234 information can be visualized in the Cancertool interface.

235 The differential gene expression analysis driven by PGC1α in PC3 cells can be obtained

236 from GEO with reference GSE75193.

10

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

237 In addition, pathway and network enrichment analyses of the significantly regulated genes

238 from GSE75193 (Supplementary Table S3) were performed using MetaCore from GeneGo

239 Inc. (https://portal.genego.com/).

240 No statistical method was used to predetermine sample size. The experiments were not

241 randomized. The investigators were not blinded to allocation during experiments and

242 outcome assessment. n values represent the number of independent experiments

243 performed, the number of individual mice or patient specimens. For each independent in

244 vitro experiment, normal distribution was assumed, and one sample t-test was applied for

245 one component comparisons with control and Student´s t-test for two component

246 comparisons. For in vivo experiments a non-parametric Mann-Whitney exact test was

247 used. Two-tail statistical analysis was applied for experimental design without predicted

248 result, and one-tail for validation or hypothesis-driven experiments. The confidence level

249 used for all the statistical analyses was of 95% (alpha value = 0.05). GraphPad Prism 8

250 software was used for statistical calculations.

251

11

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

252 Results

253 In order to address the role of PGC1α in the regulation of PCa features beyond

254 proliferation (17), we carried out a comprehensive evaluation of phenotypes associated to

255 cancer aggressiveness, based on an inducible system previously reported (17).

256 Interestingly, Pgc1α expression elicited a remarkable reduction in the migratory capacity of

257 PC3 and DU145 PCa cells in transwell assays (Fig. 1A; Supplementary Fig. 1A). A

258 similar effect was achieved in matrigel-coated transwell assays as a measure of invasion

259 (Fig. 1B; Supplementary Fig. 1B). In order to further characterize the regulation of

260 invasive properties by PGC1α, we applied two complementary assays in both cell lines.

261 On the one hand, we performed 3D invasion assays. We quantified the number of cells

262 invading at 50 μm and/or 100 μm of distance from the bottom of the plate. The results

263 showed a profound decrease in cells with invasive capacity upon Pgc1α induction (Fig.

264 1C; Supplementary Fig. 1C, D). On the other hand, we generated spheroids using the

265 hanging drop method in order to measure the invasive growth. The results corroborated

266 that the expression of the co-regulator inhibits the invasive capacity of PCa cells (Fig. 1D;

267 Supplementary Fig. 1E). Of note, this phenotype was observed at time points where

268 proliferation was not significantly influenced by Pgc1α or by the addition of doxycycline

269 (17) (Supplementary Fig. 1F-I). Overall, our results show that, beyond the anti-

270 proliferative capacity of PGC1α in PCa, the transcriptional co-regulator elicits a robust anti-

271 invasive phenotype.

272 The regulation of cell migration and invasion is intertwined with cell morphology and

273 adhesion (19). Hence, we characterized the effects of PGC1α on these parameters. The

274 expression of the co-regulator in PC3 cells was associated to a remarkable elevation in

275 cell area, with loss of stress fibers and with a modest increase in cell adhesion to collagen

276 I (Fig. 1E-F; Supplementary Fig. 1J). Importantly, Pgc1α induction in subcutaneous

12

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

277 xenografts of PC3 cells confirmed the antitumoral activity of this gene, and its impact on

278 PCa cell size in vivo (Fig. 1G; Supplementary Fig. 1K-M).

279 We next focused on the molecular alterations underlying the activity of PGC1α. In a

280 previous study, we analyzed a gene expression analysis in PC3 cells upon induction of

281 Pgc1α (Fig. 1) (17) (GSE75193). We sought to extend the analysis of this microarray by

282 taking advantage of bioinformatic tools, such as Metacore

283 (https://clarivate.com/products/metacore/) and Cancertool (25) that enable cancer

284 researchers to perform various functional enrichment analyses. Since functional

285 enrichment allows the integration of larger sets of data in order to identify underlying

286 molecular and functional alterations, we focused our analyses on all genes whose

287 expression was altered with a significant p-value in the transcriptomics analysis

288 (regardless of the adjusted p-value). This led to 1347 upregulated and 990 downregulated

289 unique gene IDs (Supplementary Table S3). Strikingly, functional enrichment of the

290 downregulated genes revealed a significant alteration in cytoskeleton organization,

291 migration, adhesion and integrin and Rho signaling (Fig. 2A; Supplementary Fig. 2A;

292 Supplementary Table S4-S5). Of note, we also identified other pathways with reported

293 activities in the regulation of invasion, such as p27, FAS and RAC, although their

294 prevalence in the analysis and their documented association to this phenotype were minor

295 (26-29). In line with our previous study (17), the enrichment analysis of the genes

296 upregulated upon Pgc1α expression confirmed a significant alteration of catabolic

297 pathways (Supplementary Table S6). We focused our attention in the Metacore analysis

298 of downregulated genes. The results revealed a remarkable alteration in cytoskeletal

299 remodeling upon PGC1α modulation in PCa cells, illustrated by processes regulated by

300 Rho kinase (ROCK). The axes containing ROCK-LIM kinase (LIMK)-Cofilin and ROCK-

301 Myosin Light Chain (MLC) are two key signaling pathways that regulate cytoskeletal

13

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

302 remodeling downstream of the monomeric G protein Rho and integrin signaling (30). The

303 immunostaining and quantification of phosphorylated Myosin-light chain 2 (p-MLC2)

304 revealed a significant reduction in this parameter in Pgc1α-expressing PC3 cells (Figure

305 2B). This result supports the notion that loss of PGC1α in PCa cells results in changes in

306 the actin-myosin cytoskeleton that are associated with the acquisition of invasive

307 properties. In order to ascertain which signaling pathways were modulated and affecting

308 cytoskeleton organization upon Pgc1α expression, we carried out a cytoskeleton phospho-

309 antibody array (Supplementary Table S7). The phosphorylation of Src protein was among

310 the most prominently reduced in the analysis (Supplementary Fig. 2B). We confirmed this

311 result by western blot analysis, both in vitro and in vivo, together with the reduction in

312 Cofilin phosphorylation, the final effector of actin filament polymerization downstream Src

313 (Fig 2C, D, Supplementary Fig. 2C, D).

314 Integrins are upstream regulators of the cytoskeleton with well-documented involvement in

315 cancer aggressiveness (19,31,32). The bioinformatics analysis of PGC1α downregulated

316 genes indicated an altered integrin signaling (Fig. 2A, Supplementary Fig. 2A), which

317 would be consistent with the reduction in Src, MLC2 and Cofilin phosphorylation. This,

318 together with the fact that PGC1α controls integrin expression in melanoma (16), prompted

319 us to evaluate integrin expression in our experimental systems. Interestingly, the levels of

320 various integrins and caveolin-1 (CAV1, but not CAV2) were robustly reduced at protein

321 and mRNA levels upon Pgc1α induction, an event that was not influenced by doxycycline

322 treatment (Fig. 2E; Supplementary Fig. 2E-I). Next, we analyzed extracts from

323 xenografts in which Pgc1α expression was activated (Fig. 1G). The western blot and

324 quantitative qRTPCR analysis corroborated the alterations elicited by the co-activator in

325 vivo (Fig. 2F; Supplementary Fig. 2J, K). Our results suggest that PGC1α controls a

326 transcriptional program that results in the alteration of cytoskeleton organization with the

14

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

327 concomitant reduction in integrin expression, an event that is consistent with the observed

328 reduction in migratory and invasive properties of PCa cells.

329 We then asked which effector of PGC1α could contribute to the negative regulation of

330 invasive properties. Inhibitors of differentiation (ID) are responsible for integrin repression

331 in melanoma (16). We ruled out the potential contribution of ID2-4 to our phenotype, since

332 their expression was not upregulated upon induction of the co-activator (Supplementary

333 Fig. 3A). Then, we applied promoter enrichment analysis (25) to the list of Pgc1α-

334 repressed genes. Strikingly, the results revealed a significant enrichment in MYC within

335 the promoters of the down-regulated genes (p-value = 8.5e-19; Fig. 3A; Supplementary

336 Table S3 and S8). We studied the impact of PGC1α on the expression of MYC and

337 observed that induction of the co-regulator elicited a consistent decrease in MYC

338 expression in PCa cells in a doxycycline-independent manner (Fig. 3B, Supplementary

339 Fig. 3B, C). Importantly, the effect was fully recapitulated at the transcriptional level. In

340 addition, the analysis of previously reported targets or genes contained in the promoter

341 analysis confirmed the reduction in MYC-dependent transcriptional program in the

342 aforementioned conditions (Fig. 3C). We took advantage of our Pgc1α-inducible xenograft

343 analysis to further demonstrate that the reduction in MYC expression and function was not

344 an artifact of in vitro assays (Fig. 3D, E; Supplementary Fig. 3D). These results suggest

345 that MYC repression is upstream of the molecular and cellular alterations elicited by

346 PGC1α associated to PCa invasion. We validated this notion by two different means. On

347 the one hand, a time course experiment upon PGC1α induction showed that MYC

348 repression is prior to the reduction of its targets and integrin gene expression (Fig. 3F,

349 Supplementary Fig. 3E-G). On the other hand, MYC silencing with a validated shRNA

350 (33,34) recapitulated the phenotype of Pgc1a expression in cell area, p-MLC2 and

351 invasive growth (Supplementary Fig. 3H-L).

15

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

352 The rapid repression in MYC mRNA levels prompted us to evaluate whether PGC1α could

353 exert a direct action on MYC promoter. To this end, we performed chromatin

354 immunoprecipitation (ChIP) analysis in Pgc1α-inducible PC3 cells with anti-HA antibody in

355 order to immunoprecipitate ectopic tagged Pgc1α. The ChIP analysis confirmed that the

356 co-regulator is bound to MYC promoter (Fig. 3G), thus suggesting that PGC1α represses

357 MYC expression in PCa. We next sought to ascertain whether the unprecedented

358 regulation of MYC by PGC1α in PCa could be recapitulated in human specimens. We

359 interrogated 5 PCa datasets (25,35-37) and, in agreement with our molecular and

360 mechanistic data, PGC1A expression was inversely correlated with MYC mRNA levels in

361 primary tumors from the majority (4 out of 5) of datasets analyzed (Fig. 3H and

362 Supplementary Fig. 3M).

363 Our previous studies demonstrated that the anti-proliferative activity of PGC1α in PCa is

364 dependent on its interaction with ERRα (17). In order to ascertain the requirement of

365 ERRα for the anti-invasive activity of PGC1α, we engineered Pgc1α-inducible PCa cells in

366 which ESRRA was deleted using CRISPR/Cas9. ERRα expression was undetectable in

367 PC3 cells in which ESRRA was deleted with two independent short guide RNAs

368 (sgERRα#1, sgERRα#2) (Fig. 4A). ESRRA deletion abolished the induction of target

369 genes of the transcription factor upon induction of Pgc1α, corroborating the functionality of

370 the genetic system (Supplementary Fig. 4A). Of note, we did not recapitulate the

371 regulation of ESRRA by PGC1A observed in vitro (Fig. 4A) in correlative human

372 transcriptomics analyses, suggesting that more complex ERRα-regulatory cues might

373 operate in human disease (Supplementary Fig. 4B). In line with our previous study (17),

374 ESRRA deletion hampered the growth suppressive activity of Pgc1α, rendering PC3 cells

375 insensitive to the action of the co-regulator (Fig. 4B). Strikingly, ESRRA deletion also

376 abolished the effect of Pgc1α on invasive properties and cell morphology at time points

16

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

377 prior to the reduction in cell proliferation, thus demonstrating that the regulation of invasion

378 by the co-regulator is exquisitely dependent upon its interaction with ERRα (Fig. 4C, D

379 and Supplementary Fig. 4C, D). The morphological changes and growth suppressive

380 phenotype elicited by Pgc1α were also absent in tumors in which ESRRA was deleted

381 (Fig. 4E, Supplementary Fig. 4E, F, G). It is worth noting that despite the requirement of

382 ERRα for the tumor suppressive activity of PGC1α, deletion of the alone

383 negatively influenced the establishment of tumors, suggesting that additional functions of

384 ERRα may be required for the first stages of tumor establishment (Supplementary Fig.

385 4H).

386 We next extended our analysis of ERRα dependency to the reported molecular alterations.

387 Our results showed that ESRRA deletion abrogated the reduction in protein and/or mRNA

388 levels of MYC, MYC targets, integrins, CAV1 as well as the reduced phosphorylation of

389 Src and Cofilin (Fig. 5A, B; Supplementary Fig. 5A, B). Moreover, ESRRA-ablated

390 tumors exhibited unperturbed MYC, integrin and CAV1 expression, as well as unchanged

391 Src and Cofilin phosphorylation upon Pgc1α expression (Fig. 5C, D; Supplementary Fig.

392 5C). All these data are in line with the association of ERRα to MYC promoter in Pgc1α-

393 expressing PC3 cells (Supplementary Fig. 5D).

394 Since we have observed a robust inverse correlation between PGC1A and MYC

395 expression in various PCa datasets, we asked whether the dependency on ERRα could be

396 recapitulated in this setting. To this end, we carried out two independent approaches in

397 PCa patient datasets. On the one hand, we inferred ERRα canonical activity based on the

398 equilibrium between its main co-activators (PGC1A) and co-repressors (NRIP1). We

399 calculated the ratio of abundance of PGC1A and NRIP1 transcript (PGC1A/NRIP1), which

400 provided an estimation of ERRα canonical activity towards its targets, as confirmed

401 through the analysis of ACACB and LAMB2 expression (Supplementary Fig. 6A). In line

17

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

402 with our mechanistic analysis, ERRα activity, but nor ERRα itself was consistently and

403 inversely correlated with MYC in various PCa datasets (Supplementary Fig. 6B, C). On

404 the other hand, we took advantage from a prognostic PGC1α-ERRα signature that we

405 generated previously (17). This signature was composed of 10 genes that were i)

406 regulated by PGC1α in vitro, ii) predicted to be ERRα targets and iii) correlated with

407 PGC1A in PCa datasets. In full support of our data, this PGC1α-ERRα activity signature

408 was inversely correlated with MYC expression in various PCa patient datasets (Fig. 5E;

409 Supplementary Fig. 6D).

410 Overall, our results provide solid evidence of the anti-invasive activity of the PGC1α-ERRα

411 transcriptional axis in PCa.

18

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

412 Discussion

413 Metabolic deregulation is a hallmark of cancer (2), and encompasses a variety of

414 biochemical routes, which must be coordinated in order to result in a phenotypic change.

415 We postulated in the past that this strict requirement for coordination could unveil novel

416 cancer genes. By focusing on transcriptional co-regulators that control the expression of

417 an ample set of metabolic genes, we discovered the predominant perturbation of PGC1α

418 in PCa (7,17). This metabolic regulator orchestrates the activation of catabolic and

419 antioxidant pathways, at the expense of anabolism (8). Interestingly, the contribution of

420 PGC1α to cancer biology is complex. Elegant studies have reported a role of this co-

421 regulator: i) promoting aggressiveness of breast, pancreatic, gastric tumors,

422 cholangiocarcinoma, glioma and melanoma (10-14,38-40), and ii) suppressing cancer

423 aggressiveness in prostate, kidney tumors and melanoma (9,15-18). Moreover, the

424 expression of this co-regulator is associated to the efficacy of anticancer therapies

425 (10,11,14,15,41,42).

426 PGC1α exhibits a tumor type-dependent activity, ranging from tumor suppressor to

427 advantageous for cancer cells (7). This co-activator is required for the activity of pancreatic

428 cancer stem cells (13) and for the survival of breast cancer cells in circulation (12). In

429 melanoma, the metabolic activity of PGC1α promotes cell proliferation, whereas the non-

430 metabolic function opposes metastatic dissemination (10,11,16).This study together with

431 reports by us and others demonstrate that PGC1α suppresses proliferation and invasion in

432 PCa through presumably distinct molecular pathways emanating from the regulation of

433 ERRα, consistent with its tumor and metastasis suppressive function (17,18) (Fig. 6). Our

434 results mirror the anti-invasive activity of the co-regulator in melanoma, whereas

435 proliferation is regulated in opposite sense in both tumor types. This apparent discrepancy

19

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

436 could be associated to the tissue-specific molecular cues that drive these tumors or the

437 distinct nutrient and metabolic pathways that sustain their growth.

438 Cancer cell proliferation imposes tremendous pressure to meet the bioenergetics demands

439 and to generate sufficient biomolecules to build new cells. We now possess a more

440 comprehensive view of the metabolic deregulations that sustain or accompany cancer cell

441 proliferation (43). However, beyond the relevance of cell proliferation in cancer, tumor cells

442 need to acquire additional capacities that accounts for the clinical progression of the

443 disease. The process of metastasis is the main cause of mortality in cancer, and only

444 partly depends on cell proliferation, as it requires angiogenesis, intravasation, survival in

445 circulation, extravasation and resuming cell growth in a distant organ (44). Our perspective

446 around the contribution of metabolic regulators to the acquisition of these features is

447 limited. An exciting possibility stems from the notion that factors that control metabolic

448 programs would also regulate molecular cues associated to cancer cell dissemination.

449 Little is known about the activities of PGC1α in cancer beyond proliferation. This co-

450 regulator inhibits dissemination in melanoma through the regulation of ID2-TCF4-Integrins

451 (16). In gastric cancer, a recent report suggests that PGC1α upregulation supports

452 metastasis though the regulation of SNAI1 (38). Interestingly, none of these effects are

453 ascribed to the regulation of its main transcriptional partner, ERRα. Instead, we

454 demonstrate that the PGC1α-ERRα transcriptional axis in PCa accounts for the invasive

455 phenotype. We demonstrate that PGC1α/ERRα status influences signaling pathways that

456 are important for the regulation of cytoskeletal remodeling. In turn, changes in pathways

457 related to integrin and ROCK signaling provide a feasible explanation for the anti-invasive

458 effects of the co-regulator. Interestingly, the set of genes inhibited in PGC1α-expressing

459 cells that relate to cytoskeletal remodeling are enriched in MYC promoter binding sites.

20

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

460 This data is consistent with the notion that PGC1α/ERRα represses MYC expression and

461 that silencing of this transcription factor partly phenocopies the effect of PGC1α (18).

462 Similar to PGC1α, ERRα has opposing effects in different tumor types (7). Interestingly,

463 we show that this nuclear receptor is required for the tumor suppressive activity of PGC1α,

464 whereas its deletion delays tumor onset in immunocompromised mice independently of the

465 induction of PGC1α. Our results could be explained by the differential requirement of basal

466 ERRα activity for the establishment of tumors (homing and the initial engagement of cell

467 proliferation in vivo) vs. the proliferation and invasion in later stages. Similar results were

468 reported for LKB1, which is required for the bypass of anoikis and the survival of tumor

469 cells in conditions of energetic stress, despite its tumor suppressive nature in established

470 tumors (45,46).

471 ERRα functions predominantly as a transcriptional activator, and is rarely reported to

472 repress the expression of target genes (47). However, recent studies demonstrate that a

473 subset of the genes identified by ERRα ChIP-SEQ are repressed by the nuclear receptor

474 (48). In this sense, our results demonstrating that PGC1α/ERRα inhibits the expression of

475 MYC broadens the spectrum of repressed genes by the protein complex. Interestingly,

476 work by the group of Dr. Frederic Bost reports that PGC1α regulates an alternative branch

477 of metabolism (polyamine biosynthesis) through the ERRα-dependent repression of MYC-

478 ODC1 (18), thus opening new molecular avenues connecting this co-activator to metabolic

479 pathways that coordinate proliferation and invasion.

480 In summary, our study together with recent reports (18) demonstrates that PGC1α/ERRα

481 coordinately control proliferative and invasive features in PCa, thus providing a feasible

482 explanation for its robust clinical association to biochemical recurrence and metastasis.

483

21

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

484 Acknowledgements

485 Apologies to those whose related publications were not cited due to space limitations. We

486 are grateful to the Carracedo lab for valuable input and to Dr. James D. Sutherland for

487 technical advice. V.T. is funded by Fundación Vasca de Innovación e Investigación

488 Sanitarias, BIOEF (BIO15/CA/052), the AECC J.P. Bizkaia and the Basque Department of

489 Health (2016111109) and the MINECO RTI2018-097267-B-I00. The work of A. Carracedo

490 is supported by the Basque Department of Industry, Tourism and Trade (Elkartek) and the

491 department of education (IKERTALDE IT1106-16, also participated by A. Gomez-Muñoz),

492 the BBVA foundation, the MINECO (SAF2016-79381-R (FEDER/EU); Severo Ochoa

493 Excellence Accreditation SEV-2016-0644-18-1; Excellence Networks SAF2016-81975-

494 REDT), European Training Networks Project (H2020-MSCA-ITN-308 2016 721532), the

495 AECC (IDEAS175CARR, GCTRA18006CARR), La Caixa Foundation (HR17-00094) and

496 the European Research Council (Starting Grant 336343, PoC 754627). CIBERONC was

497 co-funded with FEDER funds and funded by ISCIII. L.V-J and A.S-C. were funded by a

498 Basque Government predoctoral grant, A. M. was funded by a FPI predoctoral fellowship

499 from MINECO (PRE2018-083607) and C.V. was funded by a predoctoral grant of the

500 UPV/EHU. I.H. was funded by the Juan de la Cierva program of the MINECO. V.S-M was

501 supported by Cancer Research UK (CRUK) C33043/A12065 and C33043/A24478 (V.S-M.

502 and E.C-M); Royal Society RG110591 (V.S-M.); Barts Charity (V.S-M. and E.C-M). E.C-M

503 was funded by Fundación Ramón Areces. I.R-H was funded by Fundacion Alfonso Martin

504 Escudero and Marie Sklodowska-Curie Action (H2020-MSCA-IF-2014-EF-ST)

505

22

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

506 References

507 1. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100:57-70 508 2. Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144:646-74 509 3. Turajlic S, Sottoriva A, Graham T, Swanton C. Resolving genetic heterogeneity in cancer. 510 Nat Rev Genet 2019 511 4. Martin-Martin N, Carracedo A, Torrano V. Metabolism and Transcription in Cancer: 512 Merging Two Classic Tales. Front Cell Dev Biol 2017;5:119 513 5. Spiegelman BM, Heinrich R. Biological control through regulated transcriptional 514 coactivators. Cell 2004;119:157-67 515 6. Finck BN, Kelly DP. PGC-1 coactivators: inducible regulators of energy metabolism in 516 health and disease. J Clin Invest 2006;116:615-22 517 7. Valcarcel-Jimenez L, Gaude E, Torrano V, Frezza C, Carracedo A. Mitochondrial 518 Metabolism: Yin and Yang for Tumor Progression. Trends Endocrinol Metab 2017;28:748- 519 57 520 8. Lin J, Handschin C, Spiegelman BM. Metabolic control through the PGC-1 family of 521 transcription coactivators. Cell Metab 2005;1:361-70 522 9. Carracedo A, Weiss D, Leliaert AK, Bhasin M, de Boer VC, Laurent G, et al. A metabolic 523 prosurvival role for PML in breast cancer. The Journal of clinical investigation 524 2012;122:3088-100 525 10. Haq R, Shoag J, Andreu-Perez P, Yokoyama S, Edelman H, Rowe GC, et al. Oncogenic BRAF 526 regulates oxidative metabolism via PGC1alpha and MITF. Cancer Cell 2013;23:302-15 527 11. Vazquez F, Lim JH, Chim H, Bhalla K, Girnun G, Pierce K, et al. PGC1alpha expression 528 defines a subset of human melanoma tumors with increased mitochondrial capacity and 529 resistance to oxidative stress. Cancer Cell 2013;23:287-301 530 12. LeBleu VS, O'Connell JT, Gonzalez Herrera KN, Wikman H, Pantel K, Haigis MC, et al. PGC- 531 1alpha mediates mitochondrial biogenesis and oxidative phosphorylation in cancer cells to 532 promote metastasis. Nat Cell Biol 2014;16:992-1003, 1-15 533 13. Sancho P, Burgos-Ramos E, Tavera A, Bou Kheir T, Jagust P, Schoenhals M, et al. MYC/PGC- 534 1alpha Balance Determines the Metabolic Phenotype and Plasticity of Pancreatic Cancer 535 Stem Cells. Cell Metab 2015;22:590-605 536 14. Andrzejewski S, Klimcakova E, Johnson RM, Tabaries S, Annis MG, McGuirk S, et al. PGC- 537 1alpha Promotes Breast Cancer Metastasis and Confers Bioenergetic Flexibility against 538 Metabolic Drugs. Cell Metab 2017;26:778-87 e5 539 15. LaGory EL, Wu C, Taniguchi CM, Ding CC, Chi JT, von Eyben R, et al. Suppression of PGC- 540 1alpha Is Critical for Reprogramming Oxidative Metabolism in Renal Cell Carcinoma. Cell 541 reports 2015;12:116-27 542 16. Luo C, Lim JH, Lee Y, Granter SR, Thomas A, Vazquez F, et al. A PGC1alpha-mediated 543 transcriptional axis suppresses melanoma metastasis. Nature 2016;537:422-6 544 17. Torrano V, Valcarcel-Jimenez L, Cortazar AR, Liu X, Urosevic J, Castillo-Martin M, et al. The 545 metabolic co-regulator PGC1alpha suppresses prostate cancer metastasis. Nat Cell Biol 546 2016;18:645-56 547 18. Kaminski L, Torrino S, Dufies M, Djabari Z, Haider R, Roustan FR, et al. PGC1alpha Inhibits 548 Polyamine Synthesis to Suppress Prostate Cancer Aggressiveness. Cancer Res 549 2019;79:3268-80 550 19. Pandya P, Orgaz JL, Sanz-Moreno V. Modes of invasion during tumour dissemination. Mol 551 Oncol 2017;11:5-27

23

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

552 20. Wiederschain D, Wee S, Chen L, Loo A, Yang G, Huang A, et al. Single-vector inducible 553 lentiviral RNAi system for oncology target validation. Cell Cycle 2009;8:498-504 554 21. Carracedo A, Ma L, Teruya-Feldstein J, Rojo F, Salmena L, Alimonti A, et al. Inhibition of 555 mTORC1 leads to MAPK pathway activation through a PI3K-dependent feedback loop in 556 human cancer. J Clin Invest 2008;118:3065-74 557 22. Georgouli M, Herraiz C, Crosas-Molist E, Fanshawe B, Maiques O, Perdrix A, et al. Regional 558 Activation of Myosin II in Cancer Cells Drives Tumor Progression via a Secretory Cross-Talk 559 with the Immune Microenvironment. Cell 2019;176:757-74 e23 560 23. Crosas-Molist E, Bertran E, Rodriguez-Hernandez I, Herraiz C, Cantelli G, Fabra A, et al. The 561 NADPH oxidase NOX4 represses epithelial to amoeboid transition and efficient tumour 562 dissemination. Oncogene 2017;36:3002-14 563 24. Ugalde-Olano A, Egia A, Fernandez-Ruiz S, Loizaga-Iriarte A, Zuniga-Garcia P, Garcia S, et al. 564 Methodological aspects of the molecular and histological study of prostate cancer: focus 565 on PTEN. Methods 2015;77-78:25-30 566 25. Cortazar AR, V. T, Martín-Martín N, Caro-Maldonado A, Camacho L, Hermanova I, et al. 567 CANCERTOOL: a visualization and representation interface to exploit cancer datasets. 568 Cancer Res 2018;Accepted 569 26. Jeannot P, Nowosad A, Perchey RT, Callot C, Bennana E, Katsube T, et al. p27(Kip1) 570 promotes invadopodia turnover and invasion through the regulation of the 571 PAK1/Cortactin pathway. eLife 2017;6 572 27. Nadeem L, Brkic J, Chen YF, Bui T, Munir S, Peng C. Cytoplasmic mislocalization of p27 and 573 CDK2 mediates the anti-migratory and anti-proliferative effects of Nodal in human 574 trophoblast cells. J Cell Sci 2013;126:445-53 575 28. Steller EJ, Borel Rinkes IH, Kranenburg O. How CD95 stimulates invasion. Cell Cycle 576 2011;10:3857-62 577 29. Yoon H, Kim M, Jang K, Shin M, Besser A, Xiao X, et al. p27 transcriptionally coregulates 578 cJun to drive programs of tumor progression. Proc Natl Acad Sci U S A 2019;116:7005-14 579 30. Pandya P, Orgaz JL, Sanz-Moreno V. Actomyosin contractility and collective migration: may 580 the force be with you. Curr Opin Cell Biol 2017;48:87-96 581 31. Bravo-Cordero JJ, Magalhaes MA, Eddy RJ, Hodgson L, Condeelis J. Functions of cofilin in 582 cell locomotion and invasion. Nat Rev Mol Cell Biol 2013;14:405-15 583 32. Hood JD, Cheresh DA. Role of integrins in cell invasion and migration. Nat Rev Cancer 584 2002;2:91-100 585 33. Liu R, Zhang T, Zhu G, Xing M. Regulation of mutant TERT by BRAF V600E/MAP kinase 586 pathway through FOS/GABP in human cancer. Nat Commun 2018;9:579 587 34. Nakano T, Kanai Y, Amano Y, Yoshimoto T, Matsubara D, Shibano T, et al. Establishment of 588 highly metastatic KRAS mutant lung cancer cell sublines in long-term three-dimensional 589 low attachment cultures. PLoS One 2017;12:e0181342 590 35. Grasso CS, Wu YM, Robinson DR, Cao X, Dhanasekaran SM, Khan AP, et al. The mutational 591 landscape of lethal castration-resistant prostate cancer. Nature 2012;487:239-43 592 36. Lapointe J, Li C, Higgins JP, van de Rijn M, Bair E, Montgomery K, et al. Gene expression 593 profiling identifies clinically relevant subtypes of prostate cancer. Proc Natl Acad Sci U S A 594 2004;101:811-6 595 37. Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, Carver BS, et al. Integrative 596 genomic profiling of human prostate cancer. Cancer Cell 2010;18:11-22 597 38. Wang P, Guo X, Zong W, Li Y, Liu G, Lv Y, et al. PGC-1alpha/SNAI1 axis regulates tumor 598 growth and metastasis by targeting miR-128b in gastric cancer. J Cell Physiol 2019

24

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

599 39. Dan L, Wang C, Ma P, Yu Q, Gu M, Dong L, et al. PGC1alpha promotes cholangiocarcinoma 600 metastasis by upregulating PDHA1 and MPC1 expression to reverse the Warburg effect. 601 Cell Death Dis 2018;9:466 602 40. Gelato KA, Schockel L, Klingbeil O, Ruckert T, Lesche R, Toedling J, et al. Super-enhancers 603 define a proliferative PGC-1alpha-expressing melanoma subgroup sensitive to BET 604 inhibition. Oncogene 2018;37:512-21 605 41. Cruz-Bermudez A, Laza-Briviesca R, Vicente-Blanco RJ, Garcia-Grande A, Coronado MJ, 606 Laine-Menendez S, et al. Cisplatin resistance involves a metabolic reprogramming through 607 ROS and PGC-1alpha in NSCLC which can be overcome by OXPHOS inhibition. Free Radic 608 Biol Med 2019;135:167-81 609 42. Gentric G, Kieffer Y, Mieulet V, Goundiam O, Bonneau C, Nemati F, et al. PML-Regulated 610 Mitochondrial Metabolism Enhances Chemosensitivity in Human Ovarian Cancers. Cell 611 Metab 2019;29:156-73 e10 612 43. Zhu J, Thompson CB. Metabolic regulation of cell growth and proliferation. Nat Rev Mol 613 Cell Biol 2019 614 44. Steeg PS. Targeting metastasis. Nat Rev Cancer 2016;16:201-18 615 45. Jeon SM, Chandel NS, Hay N. AMPK regulates NADPH homeostasis to promote tumour cell 616 survival during energy stress. Nature 2012;485:661-5 617 46. Carracedo A, Cantley LC, Pandolfi PP. Cancer metabolism: fatty acid oxidation in the 618 limelight. Nat Rev Cancer 2013;13:227-32 619 47. Stein RA, McDonnell DP. Estrogen-related receptor alpha as a therapeutic target in cancer. 620 Endocr Relat Cancer 2006;13 Suppl 1:S25-32 621 48. Audet-Walsh E, Papadopoli DJ, Gravel SP, Yee T, Bridon G, Caron M, et al. The PGC- 622 1alpha/ERRalpha Axis Represses One-Carbon Metabolism and Promotes Sensitivity to 623 Anti-folate Therapy in Breast Cancer. Cell reports 2016;14:920-31

624

625 Figure legends

626 Figure 1. PGC1α expression impacts on invasive properties of PCa in vitro and in vivo. A-

627 B, Effect of Pgc1α expression on transwell migration (n=9 independent experiments) (A)

628 and on transwell invasion (n=4 independent experiments) (B) of PC3 cells. C-D, Effect of

629 Pgc1α expression on 3D invasion (C, n=3 independent experiments) and invasive growth

630 (D, n=3 independent experiments) of PC3 cells. D, the right panel shows one

631 representative experiment of invasive growth and the left panel the quantification. E-F,

632 Quantification of changes in cell area (E) and stress fibers (F) content upon Pgc1α-

633 expression in PC3 cells in vitro (n=3 independent experiments). In F, left panels show

634 representative phalloidin staining of non-expressing (No Dox) and Pgc1α –expressing PC3

25

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

635 cells and right panel shows the quantification. G, Quantification of changes in cell area

636 upon Pgc1α-expression in PC3 cells in vivo. Left images show representative

637 Hematoxilin&Eosin staining of non-expressing and Pgc1α-expressing xenograft samples

638 (n=4 tumors each condition, No Dox and Dox). Yellow line underlines cell surface. The

639 right panel shows the quantification of number of cells per field. Dox: doxycycline. Dox:

640 Pgc1α induced conditions; No Dox: Pgc1α non-expressing conditions. In A, B, C, D and F

641 data is represented as fold change relative to No Dox condition depicted by a dotted line.

642 Error bars represent the standard error mean (s.e.m). Statistic tests: one sample t-test with

643 a hypothetical value of 1 (A, B, C, D, F), two-tailed Student T test (E) and one-tailed Mann-

644 Whitney U test (G). p, p-value. *p < 0.05, **p < 0.01, ***p < 0.001.

645 Figure 2. PGC1α expression modulates integrin signalling of PCa in vitro and in vivo. A,

646 Metacore enrichment analysis of the transcriptional program downregulated regulated by

647 PGC1α in PC3 cells. B, Effect of Pgc1α expression on the phosphorylation of Myosin-light

648 chain (MLC) protein in PC3 cells. Left panels show representative images of

649 immunofluorescence staining using p-MLC antibodies. Right panel shows quantification of

650 p-MLC per cell area (n=3 independent experiments). C-D, Representative WB of the effect

651 of Pgc1α on Cofilin and Src phosphorylation in PC3 cells (C) and xenograft samples (D).

652 E-F, Representative WB of the effect of Pgc1α on ITGβ1, ITGβ4, ITGα3 and CAV1 in PC3

653 cells (E, n=3; independent experiments) and xenograft samples (F, n=4-5 tumors). Dox:

654 doxycycline. Dox: Pgc1α induced conditions; No Dox: Pgc1α non-expressing conditions.

655 Error bars and ± represent the standard error mean (s.e.m). Statistic tests: two-tailed

656 Student T test (B). *p < 0.05, **p < 0.01, ***p < 0.001.

657

658 Figure 3. PGC1α regulates c-Myc expression in prostate cancer. A, Promoter enrichment

659 analysis of the PGC1α transcriptional program in PC3 cells. B, Effect of Pgc1α expression

26

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

660 on c-Myc protein levels in PC3 cells (n=3 independent experiments). C, Quantification of

661 MYC gene expression and its target genes ODC, FASN, CAD1 and TCF4 by qRTPCR

662 upon Pgc1α expression in PC3 cells (n=4 independent experiments). Data is represented

663 as fold change relative to No Dox, depicted as a dotted line. D, Effect of Pgc1α expression

664 on c-Myc protein levels in xenograft samples (n=5 No Dox tumors, n=4 Dox tumors). E,

665 Quantification of MYC gene expression (and its target genes) by qRTPCR in xenograft

666 samples cells (n=5 No dox tumors, n=4 Dox tumors). F, qRTPCR gene expression

667 analysis of MYC, TCF4, ITGB4, ITGB1 and ITGA3 upon short acute induction of Pgc1α

668 expression (1, 2, 4 and 8h of doxycycline treatment) in PC3 cells. Data is represented as

669 fold change relative to No dox, depicted as a dotted line. G, Chromatin

670 immunoprecipitation (ChIP) of exogenous Pgc1α on MYC promoter in PC3 Pgc1α cells

671 after induction with 0.5 µg mL-1 doxycycline for 16 hours (n=5). Final data was normalized

672 to IgG (negative-immunoprecipitation control) and to No dox condition. H, Correlation

673 analysis between PGC1A and MYC expression in primary tumor specimens of different

674 PCa datasets. Sample sizes: Grasso n=45; Lapointe n=13; Taylor n=131 and TCGA

675 provisional n=495. Dox: doxycycline. Dox: Pgc1α induced conditions; No dox: Pgc1α non-

676 expressing conditions. Error bars and ± represent the standard error mean (s.e.m).

677 Statistic tests: one sample t-test with a hypothetical value of 1 (C, F), one-tailed Student T

678 test (G), one-tailed Mann-Whitney U test (E), Spearman correlation R (H). *p < 0.05, **p <

679 0.01, ***p < 0.001.

680

681 Figure 4. ERRα deletion mediates the effect of Pgc1α on invasive properties and

682 morphology of prostate cancer in vitro and in vivo. A, Representative experiment of ERRα

683 expression in PC3 Pgc1α cells after treatment with 0.5 μg/ml doxycycline (Dox) (n=3;

684 independent experiments). B, Relative cell number quantification upon ERRα deletion

27

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

685 (sgERRα#1and sgERRα#2) in PC3 Pgc1α expressing and non-expressing cells. Data is

686 represented as cell number at day 6 relative to day 0 (n=3, independent experiments). C,

687 Effect of ERRα deletion in invasive growth upon Pgc1α expression (n=3 independent

688 experiments). One representative spheroid image of each condition is shown out of 3

689 biological replicates. D, Quantification of cell area by phalloidin staining after ERRα

690 deletion alone or in combination with Pgc1α expression (n=4 independent experiments) in

691 PC3 cells. E, Effect of ERRα deletion alone or in combination of Pgc1α on the cell content

692 and size in xenograft samples (n=5 per condition). The number of cells per field is an

693 approximate representation of cell area. Dox: doxycycline. sg: short guide RNA. Dox:

694 Pgc1α induced conditions; No Dox: Pgc1α non-expressing conditions. Error bars represent

695 the standard error mean (s.e.m). Dotted line represents No Dox condition. Statistic tests:

696 paired Student t-test between Control -Dox and +Dox conditions (B) unpaired Student t-

697 test between +Dox control and sg conditions (B), one sample t-test with a hypothetical

698 value of 1 (C, D) and one-tailed Mann-Whitney U test (E). p, p-value. */$ p < 0.05, **/$$ p

699 < 0.01, ***/$$$ p < 0.001. In B, C and D asterisks indicate statistical difference between No

700 Dox and Dox conditions and dollar symbol between Control Dox and sgERRα#1/

701 sgERRα#2 Dox.

702 Figure 5. ERRα mediates the effect of Pgc1α on integrin signalling and MYC expression in

703 vitro and in vivo. A, Representative WB of the effect of ERRα deletion alone or in

704 combination with Pgc1α expression on ITGβ1, ITGβ4, CAV1 and MYC protein expression

705 as well as on Cofilin and Src phosphorylation in PC3 cells (n=3; independent experiments).

706 B, Effect of ERRα deletion alone or in combination with Pgc1α expression in the gene

707 expression (qRTPCR) of MYC, TCF4, ITGB1, ITGA3 and CAV1 (n=4 independent

708 experiments) in PC3 cells. Data is represented by fold change relative to Control No Dox

709 condition that is depicted by a dotted line. C, Effect of ERRα deletion alone or in

28

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

710 combination with Pgc1α expression on ITGβ1, ITGβ4, CAV1, and MYC protein expression

711 as well as on Cofilin and Src phosphorylation in xenograft samples (Control No Dox n=9

712 tumors, Control +Dox n=9 tumors; sgERRα#1 –Dox n=8 tumors; sgERRα#2 +Dox n=8

713 tumors). D, Effect of ERRα deletion alone or in combination with Pgc1α expression MYC,

714 TCF4, ITGB1, ITGA3 and CAV1 gene expression analysed by qRTPCR in xenograft

715 samples. (Control No Dox n=4-9 tumors, Control +Dox n=4-9 tumors; sgERRα#1 No Dox

716 n=6-8 tumors; sgERRα#2 +Dox n=5-6 tumors). E, Correlation analysis between MYC and

717 the PGC1α-ERRα transcriptional signature in primary tumor specimens of different PCa

718 datasets. Each dot correspond to a patient. Sample sizes: Grasso n=45; Lapointe n=13;

719 Glinsky n=78 and TCGA provisional n=495. Dox: doxycycline. Dox: Pgc1α induced

720 conditions; No dox: Pgc1α non-expressing conditions. Error bars and ± represent the

721 standard error mean (s.e.m). Statistic tests: one sample t-test (B), unpaired t-test (B, D),

722 Spearman correlation R (E). */$p < 0.05, **/$$p < 0.01, ***/$$$p < 0.001. Asterisks indicate

723 statistical difference between Control No Dox and the rest of the conditions and dollar

724 symbol between Control Dox and sgERRα#1/ sgERRα#2 Dox.

725 Figure 6. Schematic summary of the main findings.

29

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Figure 1. Valcarcel et al. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

A B C Transwell migration PC3 Transwell invasion PC3 3D invasion PC3

1.5 1.5

p=0.056

1.0 1.0 *** ** 0.5 0.5 ** Fold change relative to relative No Dox change Fold 0.0 to relative No Dox change Fold 0.0 Fold change relative to relative No Dox change Fold Dox Dox Distance (𝝁m) 50 100

D 0h 48h E Invasive growth PC3 Cell area PC3

1.5 * ) No Dox 1.0 3

100µm m 𝝁 ** No dox

0.5 Dox Cell area ( Cell Dox

0.0 Fold change relative to relative No Dox change Fold Dox

F

Stress fibers PC3 No Dox (i)

** Dox Fold change relative to relative No Dox change Fold Dox (ii) 20µm (i) (ii)

G

No Dox Dox

No dox Dox

100µm

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Figure 2. Valcarcel et al. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. A

TGF-beta 1-induced transactivation of membrane receptors signaling in HCC Neurophysiological process_Receptor-mediated axon growth repulsion Cytoskeleton remodeling_Regulation of actin cytoskeleton organization by the kinase effectors of Rho GTPases Cell adhesion_Role of tetraspanins in the integrin-mediated cell adhesion Chemotaxis_Lysophosphatidic acid signaling via GPCRs Cytoskeleton remodeling_Fibronectin-binding integrins in cell motility Neuroprotective action of lithium Development_Regulation of cytoskeleton proteins in oligodendrocyte differentiation and myelination Cell adhesion_Integrin-mediated cell adhesion and migration Development_Slit-Robo signaling

0246810 -Log(p-val)

B C -+Dox pMLC2 F-actin pMLC2 / F-actin / Dapi PGC1α No dox 80KDa 70KDa Dox p-Src

No dox 1 0.67 ± 0.05 70KDa 50um total-Src

GAPDH 35KDa pMLC/Area Dox -+Dox 25KDa p-Cofilin

1 0.61 ± 0.04 25KDa total-Cofilin

D E GAPDH 35KDa -+Dox -+Dox

PGC1α 140KDa 80 KDa ITGβ1

F -+Dox 35 KDa GAPDH 1 0.7 ± 0.08

HSP90 ITGβ4 80KDa 160 KDa -+Dox 1 ± 0.11 0.30 ± 0.16 70KDa -+Dox p-Src 140 KDa ITGβ1 ITGβ4 1 ± 0.12 0.62 ± 0.11 160KDa 1 ± 0.04 0.40 ± 0.15 70KDa 1 0.55 ± 0.13 total-Src GAPDH 45 KDa βactin 35KDa 45 KDa βactin -+Dox -+Dox 160KDa CAV1 -+Dox ITGα3 17 KDa 25 KDa 1 ± 0.18 0.33 ± 0.08 p-Cofilin 1 0.61 ± 0.04 GAPDH GAPDH 35 KDa 1 ± 0.19 0.27 ± 0.09 35KDa 25 KDa total-Cofilin -+Dox CAV1 GAPDH 17KDa 35 KDa 1 0.21 ± 0.05 GAPDH 35KDa

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. H DE BC A FG 70 KDa 35KDa 80KDa 0.0 0.5 1.0 1.5 2.0 Downloaded from R= -0.3764p=0.03371 70 KDa 35KDa 80KDa Author manuscriptshavebeenpeerreviewedandacceptedforpublicationbutnotyetedited. 012345678 Author ManuscriptPublishedOnlineFirstonOctober8,2019;DOI:10.1158/0008-5472.CAN-19-1231 Grasso TGGAAA_V$NFAT_Q4_01 GGGTGGRR_V$PAX4_03 GGGAGGRR_V$MAZ_Q6 0.14 1 ± SCGGAAGY_V$ELK1_02 - MYC TCF4 GGGCGGR_V$SP1_Q6 *** TTGTTT_V$FOXO4_01 CACGTG_V$MYC_Q2 CTTTGT_V$LEF1_Q2 CAGGTG_V$E12_Q6 AACTTT_UNKNOWN -+ Time (h) cancerres.aacrjournals.org ** ITGB1 ITGA3 .3 0.02 0.037 ± + GAPDH MYC PGC1α Dox R= -0.3295p=0.2717 04 08 0 2 4 6 180 160 140 120 100 80 60 40 20 0 GAPDH MYC PGC1α Dox ITGB4 ODC Lapointe ** * * Promoter analysis on September 29, 2021. © 2019American Association for Cancer Research.

Gene expression -Log (Fold change relative to No dox) 2 (p-val) R= -0.2683p=0.00203 10 15 0 5 Taylor * Figure 3. Valcarcel et al. Valcarcel et Figure 3. R= -0.2214p=6.5e-7 No dox No Dox TCGA No dox No Dox Figure 4 . Valcarcel et al. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. A B $ $$$ Control sgERRα#1 sgERRα#2 Control No Dox ** - -- +++ ---+++ ---+++ Dox ** Control +Dox sgERRα#1 No Dox PGC1α 80KDa sgERRα#1 +Dox sgERRα#2 No Dox ERRα 45KDa sgERRα#2 +Dox GAPDH

35KDa to 0 relative Day change Fold

C Invasive growth Control sgERRα#1 sgERRα#2 $$ No Dox Dox No Dox Dox No Dox Dox

$$$ 0h 10µm Dox

*** 48h Fold change relative to No relative each change Fold Control +Dox sgERRα#1 +Dox sgERRα#2 +Dox

D E

$ $ ** p=0.059 Control +Dox 500 **

sgERRα#1 +Dox 400 Control No Dox

to each No Dox) sgERRα#2 +Dox Control +Dox 300 sgERRα#1 No Dox Cell area Cell 200 sgERRα#1 +Dox

100

Number of cells per field (x20) field per cells of Number 0 (Fold change relative (Fold change

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Figure 5. Valcarcel et al. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. A Cont sgERRα#1 sgERRα#2 B - + - +-+Dox MYC TCF4 $$$ PGC1α 2.5 4 80KDa $$ $ ERRα 2.0 ** 45 KDa ** 3 * $ 160KDa ITGβ4 1.5

1 0.34 ± 0.03 1.3 ± 0.15 1.4 ± 0.16 0.9 ± 0.26 1.01 ± 0.3 2 1.0 140KDa ITGβ1 0.5 1 *** 1 0.59 ± 0.12 0.97 ± 0.14 1.1 ± 0.26 1.48 ± 0.22 1.48 ± 0.15 *** 0.0 0 17 KDa CAV1 70 KDa MYC ITGB1 ITGA3 70KDa p-Src

1 0.44 ± 0.09 0.68 ± 0.07 0.68 ± 0.14 0.93 ± 0.24 0.76 ± 0.08 70KDa total-Src

35 KDa GAPDH

Cont sgERRα#1 sgERRα#2 - + - +-+Dox 17 KDa p-Cofilin Gene expression (Fold change) 1 0.67 ± 0.11 0.81 ± 0.21 1.1 ± 0.17 0.98 ± 0.1 0.96 ± 0.15 17 KDa total-Cofilin CAV1 Control No Dox 35 KDa GAPDH Control +Dox sgERRα#1 No Dox C Control sgERRα#1 sgERRα#1 +Dox - Dox + Dox - Dox + Dox sgERRα#2 No Dox

80 KDa PGC1α sgERRα#2 +Dox

45 KDa ERRα 140 KDa ITGβ1

1 ± 0.1 0.75 ± 0.09 1 ± 0.13 1.2 ± 0.11 CAV1 17 KDa 17 KDa p-Cofilin D 1.± 0.11 0.23 ± 0.05 1 ± 0.08 0.94 ± 0.20 MYC TCF4 17 KDa total-Cofilin 45 KDa βactin

- Dox + Dox - Dox + Dox ITGβ4 160 KDa 1 ± 0.13 0.67 ± 0.17 1 ± 0.22 0.95 ± 0.28 70 KDa MYC 70 KDa p-Src

1± 0.12 0.73 ± 0.11 1 ± 0.16 0.91 ± 0.15 70 KDa total-Src 45 KDa βactin ITGB1 ITGA3

E Grasso Lapointe R= -0.4883 p=0.0043 R= -0.4011 p=0.175 Gene expression (Fold change) CAV1 $$$ TCGA Glinsky 2.0 *** R= -0.2464 p= 3e-8 R= -0.2759 p=0.014 1.5 Control No Dox

1.0 Control +Dox sgERRα#1 No Dox 0.5 sgERRα#1 +Dox 0.0 Gene expression (Fold change)

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Figure 6. Valcarcel et al. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

Metastasis PGC1α /ERRα

Catabolism Anabolism

MYC expression Integrin signaling • This study Polyamine synthesis • Torrano et al., Nature Cell Biol. 2016 Contractility • Kaminski et al., Cancer Research 2019

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research. Author Manuscript Published OnlineFirst on October 8, 2019; DOI: 10.1158/0008-5472.CAN-19-1231 Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited.

PGC1α suppresses prostate cancer cell invasion through ERRα transcriptional control

Lorea Valcarcel-Jimenez, Alice Macchia, Eva Crosas-Molist, et al.

Cancer Res Published OnlineFirst October 8, 2019.

Updated version Access the most recent version of this article at: doi:10.1158/0008-5472.CAN-19-1231

Supplementary Access the most recent supplemental material at: Material http://cancerres.aacrjournals.org/content/suppl/2019/10/08/0008-5472.CAN-19-1231.DC1

Author Author manuscripts have been peer reviewed and accepted for publication but have not yet been Manuscript edited.

E-mail alerts Sign up to receive free email-alerts related to this article or journal.

Reprints and To order reprints of this article or to subscribe to the journal, contact the AACR Publications Subscriptions Department at [email protected].

Permissions To request permission to re-use all or part of this article, use this link http://cancerres.aacrjournals.org/content/early/2019/10/08/0008-5472.CAN-19-1231. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC) Rightslink site.

Downloaded from cancerres.aacrjournals.org on September 29, 2021. © 2019 American Association for Cancer Research.