Next Article in Journal
X-Linked Duchenne-Type Muscular Dystrophy in Jack Russell Terrier Associated with a Partial Deletion of the Canine DMD Gene
Next Article in Special Issue
The Genetic Complexity of Prostate Cancer
Previous Article in Journal
A Label-Free Assay for Aminoacylation of tRNA
Previous Article in Special Issue
Convergence of Prognostic Gene Signatures Suggests Underlying Mechanisms of Human Prostate Cancer Progression
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Characterization of Hormone-Dependent Pathways in Six Human Prostate-Cancer Cell Lines: A Gene-Expression Study

by
Andras Franko
1,2,3,
Lucia Berti
2,3,*,
Alke Guirguis
4,
Jörg Hennenlotter
5,
Robert Wagner
1,2,3,
Marcus O. Scharpf
6,
Martin Hrabĕ de Angelis
3,7,
Katharina Wißmiller
8,9,10,
Heiko Lickert
3,8,9,10,
Arnulf Stenzl
5,
Andreas L. Birkenfeld
1,2,3,
Andreas Peter
2,3,4,
Hans-Ulrich Häring
1,2,3,
Stefan Z. Lutz
1,11,† and
Martin Heni
1,2,3,4,†
1
Department of Internal Medicine IV, Division of Diabetology, Endocrinology, and Nephrology, University Hospital Tübingen, 72076 Tübingen, Germany
2
Institute for Diabetes Research and Metabolic Diseases of the Helmholtz Centre Munich at the University of Tübingen, 72076 Tübingen, Germany
3
German Center for Diabetes Research (DZD), 85764 Neuherberg, Germany
4
Department for Diagnostic Laboratory Medicine, Institute for Clinical Chemistry and Pathobiochemistry, University Hospital Tübingen, 72076 Tübingen, Germany
5
Department of Urology, University Hospital Tübingen, 72076 Tübingen, Germany
6
Institute of Pathology, University Hospital Tübingen, 72076 Tübingen, Germany
7
Institute of Experimental Genetics, Helmholtz Zentrum München, 85764 Neuherberg, Germany
8
Institute of Diabetes and Regeneration Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany
9
Institute of Stem Cell Research, Helmholtz Zentrum München, 85764 Neuherberg, Germany
10
Department of Medicine, Technical University of Munich, 81675 München, Germany
11
Clinic for Geriatric and Orthopedic Rehabilitation Bad Sebastiansweiler, 72116 Mössingen, Germany
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Genes 2020, 11(10), 1174; https://doi.org/10.3390/genes11101174
Submission received: 25 August 2020 / Revised: 26 September 2020 / Accepted: 30 September 2020 / Published: 7 October 2020
(This article belongs to the Special Issue Genetic Complexity of Hormone Sensitive Cancers)

Abstract

:
Prostate cancer (PCa), the most incident cancer in men, is tightly regulated by endocrine signals. A number of different PCa cell lines are commonly used for in vitro experiments, but these are of diverse origin, and have very different cell-proliferation rates and hormone-response capacities. By analyzing the gene-expression pattern of main hormone pathways, we systematically compared six PCa cell lines and parental primary cells. We compared these cell lines (i) with each other and (ii) with PCa tissue samples from 11 patients. We found major differences in the gene-expression levels of androgen, insulin, estrogen, and oxysterol signaling between PCa tissue and cell lines, and between different cell lines. Our systematic characterization gives researchers a solid basis to choose the appropriate PCa cell model for the hormone pathway of interest.

1. Introduction

Genetic and metabolic alterations can both drive the development of prostate cancer (PCa) [1,2]. Metabolic alterations can influence autocrine and paracrine signaling pathways, which are crucial in the carcinogenic processes of PCa [3]. A tumor is a complex milieu and, in addition to prostate-cancer cells, it contains fibroblasts, endothelial cells, and mesenchymal stem cells [4]. These various cells communicate with PCa cells via paracrine factors that drive carcinogenesis [4,5]. For instance, paracrine factors secreted by PCa cells were shown to stimulate osteoblasts, which led to excess bone formation [6]. Metabolic alterations, including obesity, metabolic syndrome, insulin resistance, and diabetes, are known to exacerbate PCa [7,8]. These conditions mediate the aggressivity of PCa via endocrine signaling [1]. The major hormone pathways in PCa are androgen, insulin, estrogen, and oxysterol signaling [1,9,10]. In vitro studies use different cell models that are of diverse origin, and have very different cell-proliferation rates and hormone-response capacities [11,12,13]. Although all these cell lines represent valuable tools to study the development of PCa in vitro, whether these cell lines are suitable for studying endocrine pathways, which regulate PCa development in humans, is poorly understood. Therefore, it is vital to use a suitable in vitro model that resembles the physiology of endocrine pathways in human PCa. To characterize the major hormone pathways in human PCa cell lines, we systematically compared six commonly applied PCa cell lines and parental primary cells with (i) each other and (ii) PCa tissue samples of patients with PCa by analyzing their gene-expression patterns.

2. Materials and Methods

2.1. Cell Culture

Human prostate adenocarcinoma cell lines PC3 and LNCaP, isolated from bone and lymph-node metastasis respectively, were purchased from CLS-Cell line services (Eppelheim, Baden-Württemberg, Germany). DU145 cells (isolated from brain metastasis) were obtained from the DSMZ-German collection of microorganisms and cell cultures (Braunschweig, Niedersachsen, Germany). NCI-H660 cells (isolated from lymph-node metastasis) and MDA-PCa-2b (isolated from bone metastasis) were purchased from the ATCC-American type culture collection (Manassas, VA, USA). CWR-R1ca cells (isolated from human xenograft tumors) and human prostate epithelial cells (HPEC) were purchased from Merck (Darmstadt, Hessen, Germany). HPEC cells were used at passages 3 and 4. All cell lines were propagated according to the supplier’s instructions and maintained in a medium supplemented with 100 IU/mL penicillin, 0.1 mg/mL streptomycin, and 2 mM glutamine (Gibco/Thermo Fisher Scientific, Karlsruhe, Baden-Württemberg, Germany) in a 5% CO2 humidified atmosphere at 37 °C. LNCaP cells for 14 days of androgen deprivation were cultured in a basal medium (ATCC #CRL-1740, ThermoFisher Scientific #21875034) containing 2 mM L-glutamine and 10% FBS Good (Pan-Biotech P40-37500, Aidenbach, Freistaat Bayern, Germany). The medium used for androgen deprivation (ThermoFisher Scientific #11835030) was supplemented with 10% charcoal-stripped FBS (Sigma-Aldrich, F6765, Munich, Freistaat Bayern, Germany) and 10 nM dihydrotestosterone (Sigma-Aldrich, D-073) for 14 days. Detailed information regarding culture medium and supplements is given in Table 1. Cells were routinely cultivated until 90% confluency before harvesting.

2.2. Human Samples

Newly diagnosed PCa patients who had not received treatment before surgery were recruited prior to radical prostatectomy. Tissue sampling was performed by an experienced uropathologist. PCa tissues were immediately snap-frozen in liquid nitrogen and stored at −80 °C. Hematoxylin and eosin staining was performed on paraffinized samples for histological confirmation. Histopathological features were assessed, and pT- and postoperative Gleason scores were determined [14,15]. Informed written consent was obtained from all participants, and the Ethics Committee of the University of Tübingen (575/2018BO2) approved the protocol according to the Declaration of Helsinki.

2.3. Real-Time qPCR

Total RNA was isolated using an AllPrep DNA/RNA/miRNA kit (Qiagen, Hilden, Germany) according to the manufacturer’s description, and cDNA was synthesized (Roche, Basel, Switzerland). Real-time PCR was performed with LightCycler 480 Probes Master (Roche) with universal probe library using LightCycler 480 (Roche) [16]. Delta–delta crossing-point (Cp) values were calculated, and data were normalized to ubiquitin c (UBC) [8,15]. For real-time PCR analysis, the following primers and probes were applied: ESR1 5′-TCCTAACTTGCTCTTGGACAGG and 3′-GTAGCCAGCAGCATGTCG (probe nr 22), ESRRA 5′-GTGGGCGGCAGAAGTACA and 3′-TCAACCACCAGCAGATGAGA (probe nr 3), CYP46A1 5′-GCTGGACAACTTCGTCACCT and 3′-CATCACTGTGAACGCCAAGT (probe nr 53), CCND1 5′-GACCTTCGTTGCCCTCTGT and 3′-GGTTCAGGCCTTGCACTG (probe nr 87), TP53 5′-AAGTCTAGAGCCACCGTCCA and 3′-AGTCTGGCTGCCAATCCA (probe nr 3), GLUT1 5′-GGTTGTGCCATACTCATGACC and 3′-CAGATAGGACATCCAGGGTAGC (probe nr 67), GLUT12 5′-TGCTGCTTTTTCAATTGGTCT and 3′-AGGAAAGATCTCGCTGAGCA (probe nr 37), IRS1 5′-GCCTATGCCAGCATCAGTTT and 3′-TTGCTGAGGTCATTTAGGTCTTC (probe nr 71), IRS2 5′-TGACTTCTTGTCCCACCACTT and 3′-CATCCTGGTGATAAAGCCAGA (probe nr 49), FGFR1 5′-GGCAGCATCAACCACACATA and 3′-TACCCAGGGCCACTGTTTT (probe nr 42), CH25H 5′-CCTTCTTCCCGGTCATCTTC and 3′-GATATCCAGGACCACGAAGG (probe nr 9), CDKN1A 5′-CCGAAGTCAGTTCCTTGTGG and 3′-CATGGGTTCTGACGGACAT (probe nr 82), CDKN1B 5′-GAGAGCCAGGATGTCAGCG and 3′-TTGTTTTGAGTAGAAGAATCGTCGGT (probe CCTTTAATTGGGGCTCCGGCTAACT), NOS2 5′-GCTCAAATCTCGGCAGAATC and 3′-GCCATCCTCACAGGAGAGTT (probe nr 42), SEPP1 5′-GGAGCTGCCAGAGTAAAGCA and 3′-ACATTGCTGGGGTTGTCAC (probe nr 38). Primer sequences for AR, CYP27A1, CYP7B1, ESR2, FOLH1, IGF1R, INSRA, INSRB, KLK3, UBC and MKI67 are given in [9]. Primer sequences for HIF1A, RELA and BIRC5 are given in [14]. For the human PCa samples, the gene-expression levels of GLUT1, HIF1A, BIRC5, NOS2, RELA and MKI67 were previously studied [15].

2.4. Western Blot

Total cellular protein was extracted using RIPA buffer (50 mmol/L Tris-HCl pH 7.4, 150 mmol/L NaCl, 1% NP-40, 0.25% Na-deoxycholate, 1 mmol/L phenyl-methyl-sulfonyl-fluoride, 1 mmol/L dithiothreitol) containing a protease and phosphatase inhibitor cocktail (Roche Molecular Biochemicals, Mannheim, Baden-Württemberg, Germany), and cleared by centrifugation. Protein concentration was determined using a BCA protein assay from Bio-Rad (Hercules, CA, USA). The 20 µg protein lysates were separated on a 4–12% Bis-Tris gel (Invitrogen, Carlsbad, CA, USA). After electrophoresis, proteins were transferred using nitrocellulose ministacks and the iBlot dry-blotting system (Invitrogen, Carlsbad, CA, USA). Membranes were blocked for two hours in Odyssey Blocking Buffer (LiCor, Lincoln, NE, USA) and further incubated with antibodies against androgen receptor (AR, ab133273), prostate-specific antigen (PSA, KLK3, ab53774), β-tubulin (ab21057), prostate-specific membrane antigen (PSMA, FOLH1, ab19071) (Abcam, Cambridge, Cambridgeshire, UK), IGF1R β subunit (D23H3, #9750), and insulin receptor β subunit (L55B10, #3020) (Cell Signaling Technologies, Danvers, MA, USA). IRDye® or AlexaFluor® secondary antibodies (LiCor or Abcam, Cambridge, UK) were used, and signals were detected and quantified using the iBright device (Invitrogen, Carlsbad, CA, USA).

3. Results

For our comprehensive analysis, we chose six commonly investigated human PCa cell lines (CWR-R1ca, DU145, LNCaP, NCI-H660, MDA-PCa-2b, and PC3). Human prostate epithelial cells (HPEC) were included as parental, nontumorous primary prostate cells. To compare gene expression for hormone pathways in the PCa cell lines to the human situation, we analyzed 11 PCa samples isolated from patients who underwent radical prostatectomy due to their tumor. Histopathological screening confirmed the presence of prostate cancer in the collected tissues. As prostate-cancer metabolism could be different at various tumor stages, we specifically selected patients at a similar tumor stage with comparable Gleason scores (7a and 7b) and without lymph-node metastasis (Table 2). Data for the 11 human samples are shown as pooled values (mean ± standard deviation) in Figure 1, Figure 2 and Figure 3.
As a first step, we quantified the transcripts of the main hormone receptors. The expression levels of androgen receptor (AR) and its target genes, prostate-specific antigen (PSA, KLK3), and prostate-specific membrane antigen (PSMA, FOLH1) were most pronounced in LNCaP and MDA-PCa-2b cells (Figure 1A–C). The insulin receptor isoform A (INSRA)/insulin receptor isoform B (INSRB) ratio was highest in MDA-PCa-2b cells (Figure 1D–F), suggesting the activation of a mitogenic insulin pathway [17]. PC3 cells demonstrated the largest insulin receptor substrate (IRS) IRS1/IRS2 ratio (Figure 1G–I).
The highest gene levels of estrogen receptors α (ESR1) and β (ESR2) were observed in NCI-H660 and PC3 cells, whereas the expression of estrogen-related receptor α (ESRRA) was the highest in LNCaP cells (Figure 2A–C). The gene-expression levels of insulin-like growth factor 1 receptor (IGF1R) were the highest in HPEC and DU145, whereas fibroblast growth factor receptor 1 (FGFR1) showed the strongest expression in CWR-R1ca and DU145 cells (Figure 2D–E).
As a second step, we quantified the mRNA levels of further major intracellular regulators of PCa. Among the three investigated enzymes of oxysterol metabolism, the strongest expression for cytochrome P450 family 27 subfamily A member 1 (CYP27A1) was observed in DU145 and PC3 cells (Figure 2F), whereas cytochrome P450 family 7 subfamily B member 1 (CYP7B1) was highly represented in CWR-R1ca cells (Figure 2G). The transcript for cytochrome P450 family 46 subfamily A member 1 (CYP46A1) was detected only in CWR-R1ca, DU145, and NCI-H660 cells (Figure 2H). In order to use a statistical test among the analyzed cells lines, we examined cell-type-dependent gene expressions in a mixed model by bundling different genes to pathways: androgen pathway: AR, PSA, and PSMA; insulin pathway: INSRA, INSRB, IRS1, and IRS2; estrogen pathway: ESR1, ESR2, and ESRRA; oxysterols: CYP7B1, CYP46A1, and CYP27A1. The expressed genes were considered as random effects, and the pathways, cell types, and their interactions as fixed effects. We observed a significant interaction between cell type MDA-PCa-2b and the androgen pathway, which suggested that the MDA-PCa-2b cell line has a different gene expression for this pathway. Cholesterol 25-hydroxylase (CH25H) was only identified in four cell lines (Figure 2I).
Next, we analyzed three genes that are involved in proliferative pathways [18,19]. The expression of Ki-67 (MKI67) was the highest in NCI-H660 and MDA-PCa-2b cells (Figure 3A). CWR-R1ca cells showed the strongest expression of cyclin D1 (CCND1) (Figure 3B). The mRNA level of tumor protein p53 (TP53) was high in LNCaP and MDA-PCa-2b cells (Figure 3C). Hypoxia inducible factor 1 subunit α (HIF1A) and RELA proto-oncogene NF-kB subunit (RELA), as well as the NF-kB target genes, including baculoviral IAP repeat containing 5 (BIRC5), nitric oxide synthase 2 (NOS2), and selenoprotein P (SEPP1), are important regulators of carcinogenic processes in the development of PCa [20,21,22,23]. HIF1A was strongly expressed in PC3 cells (Figure 3D). mRNA levels of RELA and BIRC5 were comparable among the cell lines (Figure 3E,F). NOS2 was detected in three cell lines (Figure 3G). The transcript levels of SEPP1 were the largest in LNCaP and MDA-PCa-2b cells (Figure 3H). Cyclin-dependent kinases and their inhibitors are implicated in mitogenic signaling and were demonstrated to be regulated by androgen signaling [18]. Cyclin-dependent kinase inhibitor 1A (CDKN1A) showed strong gene expression in HPEC cells (Figure 3I). The mRNA levels of CDKN1B were the highest in LNCaP and MDA-PCa-2b cells (Figure 3J). Glucose transporters regulate crucial tumorigenic pathways, which have particular interest in the term of diabetes [24]. Among the analyzed glucose transporters, solute carrier family 2 member 1 (GLUT1) showed the strongest expression in HPEC cells (Figure 3K). The strongest gene expression for solute carrier family 2 member 12 (GLUT12) was found in CWR-R1ca cells (Figure 3L).
Ghandi and colleagues recently assessed the transcript pattern of five PCa cell lines using RNA sequencing [25]. The observed differences of gene-expression pattern among the analyzed cells lines in our study resemble these previous data for DU145, LNCaP, NCI-H660, MDA-PCa-2b, and PC3 cells (Table 3, https://www.cbioportal.org/, and [25]).
Furthermore, we measured the protein levels for AR, PSMA, IR, IGF1R, and PSA using Western blotting (Figure 4A). Most of the observed changes at the gene-expression level among HPEC, LNCaP, and PC3 cells were mirrored well on the protein level. The gene-expression profiles of CWR-R1ca, DU145, NCI-H660, and MDA-PCa-2b cells are in line with protein-expression data of previous studies [11,12,26,27,28,29]. As LNCaP cells showed profound PSA and PSMA expressions at the protein level, we treated these cells with dihydrotestosterone (DHT) after serum deprivation. DHT treatment increased the protein level of PSA (Figure 4B), suggesting that LNCaP cells are partly sensitive for hormone treatment after serum deprivation.

4. Discussion

All analyzed PCa cell lines had lower insulin-receptor expression levels than those of PCa samples obtained from PCa patients. On the other hand, transcript levels of potential oncogenic mediators and proliferation markers in different prostate-cancer cell lines and in human prostate-cancer tissue have similar expression. These data indicate that certain PCa cell lines can serve as an appropriate model for investigating the main hormone pathways on the transcript level.
Our comprehensive data regarding the mRNA levels of endocrine-signaling components identified major differences among the six human PCa cell lines and the parental nontumorous primary cells. Androgen-receptor signaling is the best-studied pathway in PCa [1,22]. LNCaP and MDA-PCa-2b cells have very high transcript levels for androgen-signaling components; the present data suggest that, out of the six analyzed PCa cell lines, LNCaP and MDA-PCa-2b cells are probably the best choice for studies on androgen signaling (Table 4).
In contrast to the androgen-signaling pathway, there is less knowledge about insulin, estrogen, and oxysterol signaling in PCa cell models. A high insulin-receptor isoform A to insulin-receptor isoform B ratio in PCa suggests the prevailing activation of the mitogenic insulin pathway [17,30]. Our analysis indicates that MDA-PCa-2b cells could be highly responsive to mitogenic insulin signaling (Table 4). On the other hand, NCI-H660 and PC3 cells appear to be suitable for analyzing estrogen signaling due to the high expression levels of ESR1 and ESR2 (Table 4). Cholesterol derivates, including oxysterols [31], were recently implicated in the regulation of PCa growth by modulating androgen-receptor signaling [9]. Our current data indicate that, on the mRNA level, the major enzymes of oxysterol metabolism show significant differences in the analyzed PCa cell lines.
Furthermore, the activation of HIF1A and NF-kB pathways plays a pivotal role in oncogenic processes [24,32]. In human prostate samples, we observed a positive correlation of the transcript levels of HIF1A and NF-kB pathways to the oncometabolite fumarate [14]. These real-time PCR data indicate that, for investigating HIF1A pathways in vitro, PC3 cells could serve as an appropriate model (Table 4). Patients with Type 2 diabetes develop more aggressive PCa [33,34] and show increased risk of PCa mortality [35]; therefore, altered glucose metabolism and the upregulation of glucose transporters could be involved in the progression of PCa in patients with diabetes [7,24]. HPEC cells had high GLUT1 expression levels, whereas CWR-R1ca cells showed the highest expression for GLUT12 transcripts, indicating that these cell lines could be used to analyze the involvement of glucose metabolism in cancer progression.
Androgen-deprivation therapy (ADT) is one of the common treatment options for localized PCa [36] and was shown to modify intracellular hormonal metabolism [37]. PCa cell lines represent in vitro models studying the consequences of ADT and hormone stimulus [13]. In LNCaP cells, PSA showed high androgen sensitivity, which is in line with previous studies [38,39]. Nevertheless, ADT has additional effects on PCa cells, as it also stimulates neuroendocrine differentiation in LNCaP cells [40]; thus, the in vitro application of hormone treatment after serum deprivation should be carefully interpreted.
The cell lines in our study were cultivated in various culture conditions, such as different media, hormonal supplements, and FBS concentrations, following suggestions for each cell line from the provider. Nevertheless, these different conditions could have created a bias on the gene expressions that we could not control.
In summary, our systematic characterization gives researchers a solid basis to choose the most appropriate PCa cell-culture model to characterize the hormone pathway of interest (Table 4).

Author Contributions

Conceptualization, S.Z.L. and H.-U.H.; methodology, A.F., L.B., J.H., A.G., K.W., and R.W.; formal analysis, A.F., L.B., and R.W.; resources, H.L., A.S., A.L.B., M.H.d.A., and H.-U.H.; writing—original-draft preparation, A.F.; writing—review and editing, L.B., A.G., J.H., R.W., M.O.S., M.H.d.A., K.W., H.L., A.S., A.L.B., A.P., H.-U.H., S.Z.L., and M.H.; All authors have read and agreed to the published version of the manuscript.

Funding

The study was supported in part by a grant from the German Federal Ministry of Education and Research (BMBF) to the German Center for Diabetes Research (DZD e.V.).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Grossmann, M.; Wittert, G. Androgens, diabetes and prostate cancer. Endocr. Relat. Cancer 2012, 19, F47–F62. [Google Scholar] [CrossRef] [Green Version]
  2. Elia, I.; Schmieder, R.; Christen, S.; Fendt, S.M. Organ-Specific Cancer Metabolism and Its Potential for Therapy. Handb. Exp. Pharmacol. 2016, 233, 321–353. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Bushman, W. Hedgehog Signaling in Prostate Development, Regeneration and Cancer. J. Dev. Biol. 2016, 4, 30. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Ridge, S.M.; Sullivan, F.J.; Glynn, S.A. Mesenchymal stem cells: Key players in cancer progression. Mol. Cancer 2017, 16, 31. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Ishii, K.; Takahashi, S.; Sugimura, Y.; Watanabe, M. Role of Stromal Paracrine Signals in Proliferative Diseases of the Aging Human Prostate. J. Clin. Med. 2018, 7, 68. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Hensel, J.; Thalmann, G.N. Biology of Bone Metastases in Prostate Cancer. Urology 2016, 92, 6–13. [Google Scholar] [CrossRef] [Green Version]
  7. Di Sebastiano, K.M.; Pinthus, J.H.; Duivenvoorden, W.C.M.; Mourtzakis, M. Glucose impairments and insulin resistance in prostate cancer: The role of obesity, nutrition and exercise. Obes. Rev. 2018, 19, 1008–1016. [Google Scholar] [CrossRef]
  8. Franko, A.; Berti, L.; Hennenlotter, J.; Rausch, S.; Scharpf, M.; De Angelis, M.H.; Stenzl, A.; Birkenfeld, A.L.; Peter, A.; Lutz, S.Z.; et al. Prostate tissue of patients with type 2 diabetes is characterized by increased gene expressions of Matrix metalloproteinases (MMPs) and CC chemokine ligands (CCLs). 2020. In Preparation. [Google Scholar]
  9. Lutz, S.Z.; Hennenlotter, J.; Scharpf, M.O.; Sailer, C.; Fritsche, L.; Schmid, V.; Kantartzis, K.; Wagner, R.; Lehmann, R.; Berti, L.; et al. Androgen receptor overexpression in prostate cancer in type 2 diabetes. Mol. Metab. 2018, 8, 158–166. [Google Scholar] [CrossRef]
  10. Nelles, J.L.; Hu, W.-Y.; Prins, G.S. Estrogen action and prostate cancer. Expert Rev. Endocrinol. Metab. 2011, 6, 437–451. [Google Scholar] [CrossRef] [Green Version]
  11. Wu, X.; Gong, S.; Roy-Burman, P.; Lee, P.; Culig, Z. Current mouse and cell models in prostate cancer research. Endocr. Relat. Cancer 2013, 20, R155–R170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Sobel, R.E.; Sadar, M.D. Cell Lines Used in Prostate Cancer Research: A Compendium of Old and New Lines—Part 1. J. Urol. 2005, 173, 342–359. [Google Scholar] [CrossRef] [PubMed]
  13. Namekawa, T.; Ikeda, K.; Horie-Inoue, K.; Inoue, S. Application of Prostate Cancer Models for Preclinical Study: Advantages and Limitations of Cell Lines, Patient-Derived Xenografts, and Three-Dimensional Culture of Patient-Derived Cells. Cells 2019, 8, 74. [Google Scholar] [CrossRef] [Green Version]
  14. Franko, A.; Shao, Y.; Heni, M.; Hennenlotter, J.; Hoene, M.; Hu, C.; Liu, X.; Zhao, X.; Wang, Q.; Birkenfeld, A.L.; et al. Human Prostate Cancer is Characterized by an Increase in Urea Cycle Metabolites. Cancers 2020, 12, 1814. [Google Scholar] [CrossRef] [PubMed]
  15. Franko, A.; Berti, L.; Hennenlotter, J.; Rausch, S.; Scharpf, M.O.; De Angelis, M.H.; Stenzl, A.; Birkenfeld, A.L.; Peter, A.; Lutz, S.Z.; et al. Transcript Levels of Aldo-Keto Reductase Family 1 Subfamily C (AKR1C) Are Increased in Prostate Tissue of Patients with Type 2 Diabetes. J. Pers. Med. 2020, 10, 124. [Google Scholar] [CrossRef] [PubMed]
  16. Franko, A.; Kovarova, M.; Feil, S.; Feil, R.; Wagner, R.; Heni, M.; Königsrainer, A.; Ruoss, M.; Nussler, A.K.; Weigert, C.; et al. cGMP-dependent protein kinase I (cGKI) modulates human hepatic stellate cell activation. Metabolism 2018, 88, 22–30. [Google Scholar] [CrossRef] [Green Version]
  17. Heni, M.; Hennenlotter, J.; Scharpf, M.; Lutz, S.Z.; Schwentner, C.; Todenhöfer, T.; Schilling, D.; Kühs, U.; Gerber, V.; Machicao, F.; et al. Insulin Receptor Isoforms A and B as well as Insulin Receptor Substrates-1 and -2 Are Differentially Expressed in Prostate Cancer. PLoS ONE 2012, 7, e50953. [Google Scholar] [CrossRef] [Green Version]
  18. Balk, S.P.; Knudsen, K.E. AR, the cell cycle, and prostate cancer. Nucl. Recept. Signal. 2008, 6. [Google Scholar] [CrossRef] [Green Version]
  19. Bjartell, A.; Montironi, R.; Berney, D.M.; Egevad, L. Tumour markers in prostate cancer II: Diagnostic and prognostic cellular biomarkers. Acta Oncol. 2011, 50 (Suppl. 1), 76–84. [Google Scholar] [CrossRef]
  20. Debelec-Butuner, B.; Alapinar, C.; Varisli, L.; Erbaykent-Tepedelen, B.; Hamid, S.M.; Gonen-Korkmaz, C.; Korkmaz, K.S. Inflammation-mediated abrogation of androgen signaling: An in vitro model of prostate cell inflammation. Mol. Carcinog. 2014, 53, 85–97. [Google Scholar] [CrossRef] [Green Version]
  21. McIlwain, D.W.; Fishel, M.L.; Boos, A.; Kelley, M.R.; Jerde, T.J. APE1/Ref-1 redox-specific inhibition decreases survivin protein levels and induces cell cycle arrest in prostate cancer cells. Oncotarget 2018, 9, 10962–10977. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Russo, M.A.; Ravenna, L.; Pellegrini, L.; Petrangeli, E.; Salvatori, L.; Magrone, T.; Fini, M.; Tafani, M. Hypoxia and Inflammation in Prostate Cancer Progression. Cross-talk with Androgen and Estrogen Receptors and Cancer Stem Cells. Endocr. Metab. Immune Disord. Drug Targets 2016, 16, 235–248. [Google Scholar] [CrossRef] [PubMed]
  23. Sciacovelli, M.; Frezza, C. Oncometabolites: Unconventional triggers of oncogenic signalling cascades. Free. Radic. Biol. Med. 2016, 100, 175–181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Ramteke, P.; Deb, A.; Shepal, V.; Bhat, M.K. Hyperglycemia Associated Metabolic and Molecular Alterations in Cancer Risk, Progression, Treatment, and Mortality. Cancers 2019, 11, 1402. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Ghandi, M.; Huang, F.W.; Jané-Valbuena, J.; Kryukov, G.V.; Lo, C.C.; McDonald, E.R., 3rd; Barretina, J.; Gelfand, E.T.; Bielski, C.M.; Li, H.; et al. Next-generation characterization of the Cancer Cell Line Encyclopedia. Nature 2019, 569, 503–508. [Google Scholar] [CrossRef]
  26. Shourideh, M.; DePriest, A.; Mohler, J.L.; Wilson, E.M.; Koochekpour, S. Characterization of fibroblast-free CWR-R1ca castration-recurrent prostate cancer cell line. Prostate 2016, 76, 1067–1077. [Google Scholar] [CrossRef]
  27. Hsieh, T.C.; Wu, J.M. Cell growth and gene modulatory activities of Yunzhi (Windsor Wunxi) from mushroom Trametes versicolor in androgen-dependent and androgen-insensitive human prostate cancer cells. Int. J. Oncol. 2001, 18, 81–88. [Google Scholar] [CrossRef]
  28. Li, Y.; Chen, R.; Bowden, M.; Mo, F.; Lin, Y.Y.; Gleave, M.; Collins, C.; Dong, X. Establishment of a neuroendocrine prostate cancer model driven by the RNA splicing factor SRRM4. Oncotarget 2017, 8, 66878–66888. [Google Scholar] [CrossRef]
  29. Thomas-Jardin, S.E.; Kanchwala, M.S.; Jacob, J.; Merchant, S.; Meade, R.K.; Gahnim, N.M.; Nawas, A.F.; Xing, C.; Delk, N.A. Identification of an IL-1-induced gene expression pattern in AR(+)PCa cells that mimics the molecular phenotype of AR(−)PCa cells. Prostate 2018, 78, 595–606. [Google Scholar] [CrossRef]
  30. Belfiore, A.; Malaguarnera, R.; Vella, V.; Lawrence, M.C.; Sciacca, L.; Frasca, F.; Morrione, A.; Vigneri, R. Insulin Receptor Isoforms in Physiology and Disease: An Updated View. Endocr. Rev. 2017, 38, 379–431. [Google Scholar] [CrossRef]
  31. Alfaqih, M.A.; Nelson, E.R.; Liu, W.; Safi, R.; Jasper, J.S.; Macias, E.; Geradts, J.; Thompson, J.W.; Dubois, L.G.; Freeman, M.R.; et al. CYP27A1 Loss Dysregulates Cholesterol Homeostasis in Prostate Cancer. Cancer Res. 2017, 77, 1662–1673. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Kimbro, K.S.; Simons, J.W. Hypoxia-inducible factor-1 in human breast and prostate cancer. Endocr. Relat. Cancer 2006, 13, 739–749. [Google Scholar] [CrossRef] [PubMed]
  33. Lutz, S.Z.; Todenhöfer, T.; Wagner, R.; Hennenlotter, J.; Ferchl, J.M.; Scharpf, M.O.; Martus, P.; Staiger, H.; Fritsche, A.; Stenzl, A.; et al. Higher prevalence of lymph node metastasis in prostate cancer in patients with diabetes. Endocr. Relat. Cancer 2018, 25, L19–L22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Kim, H.S.; Presti, J.C., Jr.; Aronson, W.J.; Terris, M.K.; Kane, C.J.; Amling, C.L.; Freedland, S.J. Glycemic control and prostate cancer progression: Results from the SEARCH database. Prostate 2010, 70, 1540–1546. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Bensimon, L.; Yin, H.; Suissa, S.; Pollak, M.N.; Azoulay, L. Type 2 diabetes and the risk of mortality among patients with prostate cancer. Cancer Causes Control. 2014, 25, 329–338. [Google Scholar] [CrossRef]
  36. Jhan, J.H.; Yeh, H.C.; Chang, Y.H.; Guu, S.J.; Wu, W.J.; Chou, Y.H.; Li, C.C. New-onset diabetes after androgen-deprivation therapy for prostate cancer: A nationwide propensity score-matched four-year longitudinal cohort study. J. Diabetes Complicat. 2018, 32, 688–692. [Google Scholar] [CrossRef]
  37. Shiota, M.; Fujimoto, N.; Kashiwagi, E.; Eto, M. The Role of Nuclear Receptors in Prostate Cancer. Cells 2019, 8, 602. [Google Scholar] [CrossRef] [Green Version]
  38. Zhao, H.; Ma, T.F.; Lin, J.; Liu, L.L.; Sun, W.J.; Guo, L.X.; Wang, S.Q.; Otecko, N.O.; Zhang, Y.P. Identification of valid reference genes for mRNA and microRNA normalisation in prostate cancer cell lines. Sci. Rep. 2018, 8, 1949. [Google Scholar] [CrossRef] [Green Version]
  39. Olsen, J.R.; Azeem, W.; Hellem, M.R.; Marvyin, K.; Hua, Y.; Qu, Y.; Li, L.; Lin, B.; Ke, X.; Øyan, A.M.; et al. Context dependent regulatory patterns of the androgen receptor and androgen receptor target genes. BMC Cancer 2016, 16, 377. [Google Scholar] [CrossRef] [Green Version]
  40. Yuan, T.C.; Veeramani, S.; Lin, F.F.; Kondrikou, D.; Zelivianski, S.; Igawa, T.; Karan, D.; Batra, S.K.; Lin, M.F. Androgen deprivation induces human prostate epithelial neuroendocrine differentiation of androgen-sensitive LNCaP cells. Endocr. Relat. Cancer 2006, 13, 151–167. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Transcript levels of hormone receptors and downstream substrates in prostate-cancer cell lines and in human prostate-cancer tissue. Transcript levels of indicated genes measured using real-time PCR. (A) AR, (B) KLK3, (C) FOLH1, (D) INSRA, (E) INSRB, (F) INSRA/INSRB ratio, (G) IRS1, (H) IRS2, (I) IRS1/IRS2 ratio. PCa: prostate cancer, HPEC: parental primary prostate cells, CWR-R1ca: xenograft PCa cells, DU145: brain metastasis PCa cells, LNCaP: lymph-node metastasis PCa cells, NCI-H660: lymph-node metastasis PCa cells, MDA-PCa-2b: bone metastasis PCa cells, PC3: bone metastasis PCa cells, nd: not detected. For human PCa samples, data shown as pooled samples: mean ± standard deviation (n = 11).
Figure 1. Transcript levels of hormone receptors and downstream substrates in prostate-cancer cell lines and in human prostate-cancer tissue. Transcript levels of indicated genes measured using real-time PCR. (A) AR, (B) KLK3, (C) FOLH1, (D) INSRA, (E) INSRB, (F) INSRA/INSRB ratio, (G) IRS1, (H) IRS2, (I) IRS1/IRS2 ratio. PCa: prostate cancer, HPEC: parental primary prostate cells, CWR-R1ca: xenograft PCa cells, DU145: brain metastasis PCa cells, LNCaP: lymph-node metastasis PCa cells, NCI-H660: lymph-node metastasis PCa cells, MDA-PCa-2b: bone metastasis PCa cells, PC3: bone metastasis PCa cells, nd: not detected. For human PCa samples, data shown as pooled samples: mean ± standard deviation (n = 11).
Genes 11 01174 g001
Figure 2. Transcript levels of hormone receptors and potential oncogenic mediators in prostate-cancer cell lines and in human prostate-cancer tissue. Transcript levels of indicated genes were measured using real-time PCR. (A) ESR1, (B) ESR2, (C) ESRRA, (D) IGF1R, (E) FGFR1, (F) CYP27A1, (G) CYP7B1, (H) CYP46A1, (I) CH25H. PCa: prostate cancer, HPEC: parental primary prostate cells, CWR-R1ca: xenograft PCa cells, DU145: brain metastasis PCa cells, LNCaP: lymph-node metastasis PCa cells, NCI-H660: lymph-node metastasis PCa cells, MDA-PCa-2b: bone metastasis PCa cells, PC3: bone metastasis PCa cells, nd: not detected. For human PCa samples, data shown as pooled samples: mean ± standard deviation (n = 11).
Figure 2. Transcript levels of hormone receptors and potential oncogenic mediators in prostate-cancer cell lines and in human prostate-cancer tissue. Transcript levels of indicated genes were measured using real-time PCR. (A) ESR1, (B) ESR2, (C) ESRRA, (D) IGF1R, (E) FGFR1, (F) CYP27A1, (G) CYP7B1, (H) CYP46A1, (I) CH25H. PCa: prostate cancer, HPEC: parental primary prostate cells, CWR-R1ca: xenograft PCa cells, DU145: brain metastasis PCa cells, LNCaP: lymph-node metastasis PCa cells, NCI-H660: lymph-node metastasis PCa cells, MDA-PCa-2b: bone metastasis PCa cells, PC3: bone metastasis PCa cells, nd: not detected. For human PCa samples, data shown as pooled samples: mean ± standard deviation (n = 11).
Genes 11 01174 g002
Figure 3. Transcript levels of potential oncogenic mediators in prostate-cancer cell lines and in human prostate-cancer tissue. Transcript levels of indicated genes measured using real-time PCR. (A) MKI67, (B) CCND1, (C) TP53, (D) HIF1A, (E) RELA, (F) BIRC5, (G) NOS2, (H) SEPP1, (I) CDKN1A, (J) CDKN1B, (K) GLUT1, (L) GLUT12. PCa: prostate cancer, HPEC: parental primary prostate cells, CWR-R1ca: xenograft PCa cells, DU145: brain metastasis PCa cells, LNCaP: lymph-node metastasis PCa cells, NCI-H660: lymph-node metastasis PCa cells, MDA-PCa-2b: bone metastasis PCa cells, PC3: bone metastasis PCa cells, nd: not detected. For human PCa samples, data shown as pooled samples: mean ± standard deviation (n = 11).
Figure 3. Transcript levels of potential oncogenic mediators in prostate-cancer cell lines and in human prostate-cancer tissue. Transcript levels of indicated genes measured using real-time PCR. (A) MKI67, (B) CCND1, (C) TP53, (D) HIF1A, (E) RELA, (F) BIRC5, (G) NOS2, (H) SEPP1, (I) CDKN1A, (J) CDKN1B, (K) GLUT1, (L) GLUT12. PCa: prostate cancer, HPEC: parental primary prostate cells, CWR-R1ca: xenograft PCa cells, DU145: brain metastasis PCa cells, LNCaP: lymph-node metastasis PCa cells, NCI-H660: lymph-node metastasis PCa cells, MDA-PCa-2b: bone metastasis PCa cells, PC3: bone metastasis PCa cells, nd: not detected. For human PCa samples, data shown as pooled samples: mean ± standard deviation (n = 11).
Genes 11 01174 g003
Figure 4. Protein levels of androgen receptor (AR), PSA, prostate-specific membrane antigen (PSMA), insulin receptor (IR), and insulin-like growth factor 1 receptor (IGF1R) in prostate-cancer cell lines. Proteins detected using SDS-PAGE and Western blot. (A) For HPEC, LNCaP, and PC3 cell lysates, AR, PSA, PSMA, IR, and IGF1R antibodies were applied. (B) For LNCaP cell lysates, PSA, PSMA, and IR antibodies were applied. As loading control, β-tubulin was used. Protein intensities were normalized to (A) β-tubulin and HPEC cells or (B) control condition. These relative protein intensities are under the pictures. For quantifying PSA intensity, upper protein bands were evaluated. Control: LNCaP cells grown in growth medium; DHT: dihydrotestosterone treatment of LNCaP cells after serum deprivation. Numbers on the right side represent molecular-weight markers.
Figure 4. Protein levels of androgen receptor (AR), PSA, prostate-specific membrane antigen (PSMA), insulin receptor (IR), and insulin-like growth factor 1 receptor (IGF1R) in prostate-cancer cell lines. Proteins detected using SDS-PAGE and Western blot. (A) For HPEC, LNCaP, and PC3 cell lysates, AR, PSA, PSMA, IR, and IGF1R antibodies were applied. (B) For LNCaP cell lysates, PSA, PSMA, and IR antibodies were applied. As loading control, β-tubulin was used. Protein intensities were normalized to (A) β-tubulin and HPEC cells or (B) control condition. These relative protein intensities are under the pictures. For quantifying PSA intensity, upper protein bands were evaluated. Control: LNCaP cells grown in growth medium; DHT: dihydrotestosterone treatment of LNCaP cells after serum deprivation. Numbers on the right side represent molecular-weight markers.
Genes 11 01174 g004
Table 1. Characteristics of the cell lines used in the study. If not otherwise specified, fetal bovine serum (FBS) was purchased from Bio&SELL (FBS Superior, #S 0615, Feucht, Germany).
Table 1. Characteristics of the cell lines used in the study. If not otherwise specified, fetal bovine serum (FBS) was purchased from Bio&SELL (FBS Superior, #S 0615, Feucht, Germany).
Cell LineOriginCatalog NrMediumSupplements
(Final Concentration)
PC3Adult, male Caucasian.
Prostate, from metastatic site: bone.
Adenocarcinoma, Grade IV
CLS, cell line service
#300312
ThermoFisher Scientific
#11039047
2 mM L-glutamine,
5% FBS
LNCaPAdult, male Caucasian.
Prostate, from metastatic site: left supraclavicular lymph node.
Carcinoma
CLS, cell line service
#300265
ThermoFisher Scientific
#51200046
2 mM L-glutamine,
10% FBS, 0.1 mM nonessential amino acids, 1 mM sodium pyruvate
MDA-PCa-2bAdult, male African American. Prostate, from metastatic site: boneATCC
#CRL-2422
ATCC
#30-2004
20% FBS, 25 ng/mL cholera toxin, 10 ng/mL mouse EGF, 0.005 mM phosphoethanolamine, 100 pg/mL hydrocortisone, 45 nM sodium selenite, 0.005 mg/mL human recombinant insulin
CWR-R1caFibroblast-free cell line derived from the castration-resistant or recurrent CWR-R1 human prostate-cancer cell lineMerck-Millipore
#SCC118
ThermoFisher Scientific
#32404014
2 mM L-glutamine,
10% FBS
NCI-H660Adult, male Caucasian.
Prostate, from metastatic site: lymph node
ATCC
#CRL-5813
ThermoFisher Scientific
#32404014
4 mM L-glutamine,
5% FBS, 0.005 mg/mL insulin, 0.01 mg/mL transferrin, 30 nM sodium selenite, 10 nM hydrocortisone, 10 nM β-estradiol
DU145Adult, male Caucasian.
Prostate, from metastatic site: brain
DSMZ
#ACC 261
ThermoFisher Scientific
#32404014
2 mM L-glutamine,
10% FBS
HPECAdult, normalMerck-Millipore
#SCCE019
Merck-Millipore
#SCMP001 (Kit)
6 mM L-glutamine, 0.4% epiFactor P, 1 µM epinephrine, 0.5 ng/mL rh TGF-α, 100 ng/mL hydrocortisone hemisuccinate, 5 µg/mL human recombinant insulin, 5 µg/mL apo-transferrin
Table 2. Patient characteristics. Abbreviations: BMI: body-mass index, PSA: prostate-specific antigen, N: number of patients, Stdev: standard deviation. pT and Gleason scores represent prostate-cancer (PCa) pathological stages; pN denotes lymph-node status.
Table 2. Patient characteristics. Abbreviations: BMI: body-mass index, PSA: prostate-specific antigen, N: number of patients, Stdev: standard deviation. pT and Gleason scores represent prostate-cancer (PCa) pathological stages; pN denotes lymph-node status.
Patient CharacteristicsMeanStdevN
Age (y)65811
BMI (kg/m2)26.53.211
PSA (ng/mL)12.18.110
pT–2c 6
pT–3a 4
pT–3b 1
pN 0
GLEASON 7a 7
GLEASON 7b 4
Table 3. RNA sequencing data from https://www.cbioportal.org/ [25].
Table 3. RNA sequencing data from https://www.cbioportal.org/ [25].
Cell LineDU145LNCaPNCI-H660MDA-PCa-2bPC3
Gene ID
AR0.093452.350.010424.770.0098
KLK30.01523066.7803292.290
FOLH10.0447473.170.0119308.790.1089
INSR *1.275.686.415.391.87
IRS13.592.502.155.407.05
IRS23.440.80802.806.190.7902
ESR10.01840.00150.91320.00160.2013
ESR20.16130.08670.51530.06320.2337
ESRRA11.1927.0412.6529.4119.22
IGF1R5.419.466.2122.711.58
FGFR113.302.680.33943.325.23
CYP27A13.420.06500.17430.02645.67
CYP7B10.055000.32310.00920.7905
CYP46A10.04460.25180.19620.13490.1359
CH25H0.04550.01510.028200.0157
MKI6758.0916.1538.5226.2224.39
CCND1129.71110.320.601798.12232.60
TP5332.2139.175.8517.031.01
HIF1A111.9540.7254.63123.3283.55
RELA27.0419.1820.1819.5316.76
BIRC56.842.614.122.576.09
NOS20.508000.046200.0081
SEPP10.673727.1834.1044.551.32
CDKN1A18.7392.3952.2540.944.49
CDKN1B17.7228.1540.7032.498.18
GLUT168.7719.1710.5325.1563.88
GLUT120.563326.020.90426.980.6577
Numbers denote median values.* Applied method did not distinguish between INSRA and INSRB isoforms, which were, however, differentiated by our qPCR analysis.
Table 4. Summary table.
Table 4. Summary table.
Pathway of InterestProposed Useful PCa Cell Lines
Androgen signalingLNCaP, MDA-PCa-2b
Mitogenic insulin signalingMDA-PCa-2b
Estrogen signalingNCI-H660, PC3
HIF1A pathwayPC3

Share and Cite

MDPI and ACS Style

Franko, A.; Berti, L.; Guirguis, A.; Hennenlotter, J.; Wagner, R.; Scharpf, M.O.; de Angelis, M.H.; Wißmiller, K.; Lickert, H.; Stenzl, A.; et al. Characterization of Hormone-Dependent Pathways in Six Human Prostate-Cancer Cell Lines: A Gene-Expression Study. Genes 2020, 11, 1174. https://doi.org/10.3390/genes11101174

AMA Style

Franko A, Berti L, Guirguis A, Hennenlotter J, Wagner R, Scharpf MO, de Angelis MH, Wißmiller K, Lickert H, Stenzl A, et al. Characterization of Hormone-Dependent Pathways in Six Human Prostate-Cancer Cell Lines: A Gene-Expression Study. Genes. 2020; 11(10):1174. https://doi.org/10.3390/genes11101174

Chicago/Turabian Style

Franko, Andras, Lucia Berti, Alke Guirguis, Jörg Hennenlotter, Robert Wagner, Marcus O. Scharpf, Martin Hrabĕ de Angelis, Katharina Wißmiller, Heiko Lickert, Arnulf Stenzl, and et al. 2020. "Characterization of Hormone-Dependent Pathways in Six Human Prostate-Cancer Cell Lines: A Gene-Expression Study" Genes 11, no. 10: 1174. https://doi.org/10.3390/genes11101174

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop