Skip to main content
Top
Published in: BMC Medicine 1/2022

Open Access 01-12-2022 | Prostate Cancer | Research article

The lipidomic profile of the tumoral periprostatic adipose tissue reveals alterations in tumor cell’s metabolic crosstalk

Authors: Antonio Altuna-Coy, Xavier Ruiz-Plazas, Silvia Sánchez-Martin, Helena Ascaso-Til, Manuel Prados-Saavedra, Marta Alves-Santiago, Xana Bernal-Escoté, José Segarra-Tomás, Matilde R. Chacón

Published in: BMC Medicine | Issue 1/2022

Login to get access

Abstract

Background

Periprostatic adipose tissue (PPAT) plays a role in prostate cancer (PCa) progression. PPAT lipidomic composition study may allow us to understand the tumor metabolic microenvironment and provide new stratification factors.

Methods

We used ultra-high-performance liquid chromatography-mass spectrometry-based non-targeted lipidomics to profile lipids in the PPAT of 40 patients with PCa (n = 20 with low-risk and n = 20 high-risk). Partial least squares-discriminant analysis (PLS-DA) and variable importance in projection (VIP) analysis were used to identify the most relevant features of PPAT between low- and high-risk PCa, and metabolite set enrichment analysis was used to detect disrupted metabolic pathways. Metabolic crosstalk between PPAT and PCa cell lines (PC-3 and LNCaP) was studied using ex vivo experiments. Lipid uptake and lipid accumulation were measured. Lipid metabolic-related genes (SREBP1, FASN, ACACA, LIPE, PPARG, CD36, PNPLA2, FABP4, CPT1A, FATP5, ADIPOQ), inflammatory markers (IL-6, IL-1B, TNFα), and tumor-related markers (ESRRA, MMP-9, TWIST1) were measured by RT-qPCR.

Results

Significant differences in the content of 67 lipid species were identified in PPAT samples between high- and low-risk PCa. PLS-DA and VIP analyses revealed a discriminating lipidomic panel between low- and high-risk PCa, suggesting the occurrence of disordered lipid metabolism in patients related to PCa aggressiveness. Functional analysis revealed that alterations in fatty acid biosynthesis, linoleic acid metabolism, and β-oxidation of very long-chain fatty acids had the greatest impact in the PPAT lipidome. Gene analyses of PPAT samples demonstrated that the expression of genes associated with de novo fatty acid synthesis such as FASN and ACACA were significantly lower in PPAT from high-risk PCa than in low-risk counterparts. This was accompanied by the overexpression of inflammatory markers (IL-6, IL-1B, and TNFα). Co-culture of PPAT explants with PCa cell lines revealed a reduced gene expression of lipid metabolic-related genes (CD36, FASN, PPARG, and CPT1A), contrary to that observed in co-cultured PCa cell lines. This was followed by an increase in lipid uptake and lipid accumulation in PCa cells. Tumor-related genes were increased in co-cultured PCa cell lines.

Conclusions

Disturbances in PPAT lipid metabolism of patients with high-risk PCa are associated with tumor cell metabolic changes.
Appendix
Available only for authorised users
Literature
2.
go back to reference Zadra G, Photopoulos C, Loda M. The fat side of prostate cancer. Biochim Biophys Acta. 2013;1831:1518–32.CrossRef Zadra G, Photopoulos C, Loda M. The fat side of prostate cancer. Biochim Biophys Acta. 2013;1831:1518–32.CrossRef
3.
go back to reference Galbraith L, Leung HY, Ahmad I. Lipid pathway deregulation in advanced prostate cancer. Pharmacol Res. 2018;131:177–84.CrossRef Galbraith L, Leung HY, Ahmad I. Lipid pathway deregulation in advanced prostate cancer. Pharmacol Res. 2018;131:177–84.CrossRef
4.
go back to reference Dirat B, Bochet L, Dabek M, Daviaud D, Dauvillier S, Majed B, et al. Cancer-associated adipocytes exhibit an activated phenotype and contribute to breast cancer invasion. Cancer Res. 2011;71:2455–65.CrossRef Dirat B, Bochet L, Dabek M, Daviaud D, Dauvillier S, Majed B, et al. Cancer-associated adipocytes exhibit an activated phenotype and contribute to breast cancer invasion. Cancer Res. 2011;71:2455–65.CrossRef
5.
go back to reference Laurent V, Toulet A, Attané C, Milhas D, Dauvillier S, Zaidi F, et al. Periprostatic adipose tissue favors prostate cancer cell invasion in an obesity-dependent manner: role of oxidative stress. Mol Cancer Res. 2019;17:821–35.CrossRef Laurent V, Toulet A, Attané C, Milhas D, Dauvillier S, Zaidi F, et al. Periprostatic adipose tissue favors prostate cancer cell invasion in an obesity-dependent manner: role of oxidative stress. Mol Cancer Res. 2019;17:821–35.CrossRef
6.
go back to reference Watt MJ, Clark AK, Selth LA, Haynes VR, Lister N, Rebello R, et al. Suppressing fatty acid uptake has therapeutic effects in preclinical models of prostate cancer. Sci Transl Med. 2019;11:1–14.CrossRef Watt MJ, Clark AK, Selth LA, Haynes VR, Lister N, Rebello R, et al. Suppressing fatty acid uptake has therapeutic effects in preclinical models of prostate cancer. Sci Transl Med. 2019;11:1–14.CrossRef
8.
go back to reference Figiel S, Pinault M, Domingo I, Guimaraes C, Guibon R, Besson P, et al. Fatty acid profile in peri-prostatic adipose tissue and prostate cancer aggressiveness in African-Caribbean and Caucasian patients. Eur J Cancer. 2018;91:107–15.CrossRef Figiel S, Pinault M, Domingo I, Guimaraes C, Guibon R, Besson P, et al. Fatty acid profile in peri-prostatic adipose tissue and prostate cancer aggressiveness in African-Caribbean and Caucasian patients. Eur J Cancer. 2018;91:107–15.CrossRef
9.
go back to reference Miladinovic D, Cusick T, Mahon KL, Haynes AM, Cortie CH, Meyer BJ, et al. Assessment of periprostatic and subcutaneous adipose tissue lipolysis and adipocyte size from men with localized prostate cancer. Cancers (Basel). 2020;12(6):1385.CrossRef Miladinovic D, Cusick T, Mahon KL, Haynes AM, Cortie CH, Meyer BJ, et al. Assessment of periprostatic and subcutaneous adipose tissue lipolysis and adipocyte size from men with localized prostate cancer. Cancers (Basel). 2020;12(6):1385.CrossRef
10.
go back to reference Allott EH, Arab L, Su LJ, Farnan L, Fontham ETH, Mohler JL, et al. Saturated fat intake and prostate cancer aggressiveness: results from the population-based North Carolina-Louisiana Prostate Cancer Project. Prostate Cancer Prostatic Dis. 2017;20:48–54.CrossRef Allott EH, Arab L, Su LJ, Farnan L, Fontham ETH, Mohler JL, et al. Saturated fat intake and prostate cancer aggressiveness: results from the population-based North Carolina-Louisiana Prostate Cancer Project. Prostate Cancer Prostatic Dis. 2017;20:48–54.CrossRef
11.
go back to reference Epstein JI, Egevad L, Amin MB, Delahunt B, Srigley JR, Humphrey PA. The 2014 International Society of Urological Pathology (ISUP) Consensus conference on Gleason grading of prostatic carcinoma. Am J Surg Pathol. 2016;40(2):244–52.CrossRef Epstein JI, Egevad L, Amin MB, Delahunt B, Srigley JR, Humphrey PA. The 2014 International Society of Urological Pathology (ISUP) Consensus conference on Gleason grading of prostatic carcinoma. Am J Surg Pathol. 2016;40(2):244–52.CrossRef
12.
go back to reference Kang H. Sample size determination and power analysis using the G*Power software. J Educ Eval Health Prof. 2021;18:1–12.CrossRef Kang H. Sample size determination and power analysis using the G*Power software. J Educ Eval Health Prof. 2021;18:1–12.CrossRef
13.
go back to reference Westerhuis JA, van Velzen EJJ, Hoefsloot HCJ, Smilde AK. Discriminant Q2 (DQ2) for improved discrimination in PLSDA models. Metabolomics. 2008;4:293–6.CrossRef Westerhuis JA, van Velzen EJJ, Hoefsloot HCJ, Smilde AK. Discriminant Q2 (DQ2) for improved discrimination in PLSDA models. Metabolomics. 2008;4:293–6.CrossRef
14.
go back to reference Altmann R, Hausmann M, Spöttl T, Gruber M, Bull AW, Menzel K, et al. 13-Oxo-ODE is an endogenous ligand for PPARγ in human colonic epithelial cells. Biochem Pharmacol. 2007;74:612–22.CrossRef Altmann R, Hausmann M, Spöttl T, Gruber M, Bull AW, Menzel K, et al. 13-Oxo-ODE is an endogenous ligand for PPARγ in human colonic epithelial cells. Biochem Pharmacol. 2007;74:612–22.CrossRef
15.
16.
go back to reference Gazi E, Gardner P, Lockyer NP, Hart CA, Brown MD, Clarke NW. Direct evidence of lipid translocation between adipocytes and prostate cancer cells with imaging FTIR microspectroscopy. J Lipid Res. 2007;48:1846–56.CrossRef Gazi E, Gardner P, Lockyer NP, Hart CA, Brown MD, Clarke NW. Direct evidence of lipid translocation between adipocytes and prostate cancer cells with imaging FTIR microspectroscopy. J Lipid Res. 2007;48:1846–56.CrossRef
17.
go back to reference Fontaine A, Bellanger D, Guibon R, Bruyère F, Brisson L, Fromont G. Lipophagy and prostate cancer: association with disease aggressiveness and proximity to periprostatic adipose tissue. J Pathol. 2021;255:166–76.CrossRef Fontaine A, Bellanger D, Guibon R, Bruyère F, Brisson L, Fromont G. Lipophagy and prostate cancer: association with disease aggressiveness and proximity to periprostatic adipose tissue. J Pathol. 2021;255:166–76.CrossRef
18.
go back to reference Hanson S, Thorpe G, Winstanley L, Abdelhamid AS, Hooper L, Abdelhamid A, et al. Omega-3, omega-6 and total dietary polyunsaturated fat on cancer incidence: systematic review and meta-analysis of randomised trials. Br J Cancer. 2020;122:1260–70.CrossRef Hanson S, Thorpe G, Winstanley L, Abdelhamid AS, Hooper L, Abdelhamid A, et al. Omega-3, omega-6 and total dietary polyunsaturated fat on cancer incidence: systematic review and meta-analysis of randomised trials. Br J Cancer. 2020;122:1260–70.CrossRef
20.
go back to reference Arifin SA, Falasca M. Lysophosphatidylinositol signalling and metabolic diseases. Metabolites. 2016;6:1–11.CrossRef Arifin SA, Falasca M. Lysophosphatidylinositol signalling and metabolic diseases. Metabolites. 2016;6:1–11.CrossRef
23.
go back to reference Valcarcel-Jimenez L, Macchia A, Crosas-Molist E, Schaub-Clerigue A, Camacho L, Martín-Martín N, et al. PGC1a suppresses prostate cancer cell invasion through ERRA transcriptional control. Cancer Res. 2019;79:6153–65.CrossRef Valcarcel-Jimenez L, Macchia A, Crosas-Molist E, Schaub-Clerigue A, Camacho L, Martín-Martín N, et al. PGC1a suppresses prostate cancer cell invasion through ERRA transcriptional control. Cancer Res. 2019;79:6153–65.CrossRef
25.
go back to reference Eide T, Ramberg H, Glackin C, Tindall D, Taskén KA. TWIST1, a novel androgen-regulated gene, is a target for NKX3-1 in prostate cancer cells. Cancer Cell Int. 2013;13:2–7.CrossRef Eide T, Ramberg H, Glackin C, Tindall D, Taskén KA. TWIST1, a novel androgen-regulated gene, is a target for NKX3-1 in prostate cancer cells. Cancer Cell Int. 2013;13:2–7.CrossRef
27.
go back to reference Sharma AM, Staels B. Review: Peroxisome proliferator-activated receptor γ and adipose tissue - understanding obesity-related changes in regulation of lipid and glucose metabolism. J Clin Endocrinol Metab. 2007;92:386–95.CrossRef Sharma AM, Staels B. Review: Peroxisome proliferator-activated receptor γ and adipose tissue - understanding obesity-related changes in regulation of lipid and glucose metabolism. J Clin Endocrinol Metab. 2007;92:386–95.CrossRef
28.
go back to reference Ricote M, Glass CK. PPARs and molecular mechanisms of transrepression. Biochim Biophys Acta. 2007;1771:926–35.CrossRef Ricote M, Glass CK. PPARs and molecular mechanisms of transrepression. Biochim Biophys Acta. 2007;1771:926–35.CrossRef
30.
go back to reference Thompson DA, Hammock BD. Dihydroxyoctadecamonoenoate esters inhibit the neutrophil respiratory burst. J Biosci. 2007;32:279–91.CrossRef Thompson DA, Hammock BD. Dihydroxyoctadecamonoenoate esters inhibit the neutrophil respiratory burst. J Biosci. 2007;32:279–91.CrossRef
Metadata
Title
The lipidomic profile of the tumoral periprostatic adipose tissue reveals alterations in tumor cell’s metabolic crosstalk
Authors
Antonio Altuna-Coy
Xavier Ruiz-Plazas
Silvia Sánchez-Martin
Helena Ascaso-Til
Manuel Prados-Saavedra
Marta Alves-Santiago
Xana Bernal-Escoté
José Segarra-Tomás
Matilde R. Chacón
Publication date
01-12-2022
Publisher
BioMed Central
Published in
BMC Medicine / Issue 1/2022
Electronic ISSN: 1741-7015
DOI
https://doi.org/10.1186/s12916-022-02457-3

Other articles of this Issue 1/2022

BMC Medicine 1/2022 Go to the issue