#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Role of GDF15 in methylseleninic acid-mediated inhibition of cell proliferation and induction of apoptosis in prostate cancer cells


Authors: Wenbo Zhang aff001;  Cheng Hu aff001;  Xiaojie Wang aff001;  Shanshan Bai aff001;  Subing Cao aff002;  Margaret Kobelski aff002;  James R. Lambert aff003;  Jingkai Gu aff001;  Yang Zhan aff001
Authors place of work: National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun, Jilin, China aff001;  Department of Structural and Cellular Biology, Tulane Cancer Center, School of Medicine, Tulane University, New Orleans, Louisiana, United States of America aff002;  Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado, United States of America aff003
Published in the journal: PLoS ONE 14(9)
Category: Research Article
doi: https://doi.org/10.1371/journal.pone.0222812

Summary

The growth inhibitory efficacy of methylseleninic acid (MSA) in prostate cancer cells has been documented extensively. However, our understanding of the immediate targets that are key to the growth inhibitory effects of MSA remains limited. Here, using multiple preclinical prostate cancer models, we demonstrated in vitro and in vivo that GDF15 is a most highly induced, immediate target of MSA. We further showed that knockdown of GDF15 mitigates MSA inhibition of cell proliferation and induction of apoptosis. Analysis of gene expression data from over 1000 primary and 200 metastatic prostate cancer samples revealed that GDF15 expression is decreased in metastatic prostate cancers compared to primary tumors and that lower GDF15 levels in primary tumors are associated with higher Gleason scores and shorter survival of the patients. Additionally, pathways that are negatively correlated with GDF15 levels in clinical samples are also negatively correlated with MSA treatment in cultured cells. Since most, if not all, of these pathways have been implicated in prostate cancer progression, suppressing their activities by inducing GDF15 is consistent with the anticancer effects of MSA in prostate cancer. Overall, this study provides support for GDF15 as an immediate target of MSA in prostate cancer cells.

Keywords:

Biology and life sciences – Cell biology – Physical sciences – Chemistry – Research and analysis methods – Cell processes – Anatomy – Medicine and health sciences – Urology – oncology – Cancer treatment – Cancers and neoplasms – Cell death – Chemical elements – Immunologic techniques – Immunoassays – Enzyme-linked immunoassays – apoptosis – Bioassays and physiological analysis – Microarrays – Exocrine glands – Cell proliferation – Genitourinary tract tumors – prostate cancer – Prostate diseases – Prostate gland – Selenium

Introduction

Prostate cancer is the most common non-skin cancer and the second leading cause of cancer death in American men. Surgery and radiation therapy are effective regimens for localized prostate cancer, but there is no cure for metastatic disease, for which androgen deprivation therapy is the first-line therapy. While androgen deprivation therapy is effective initially, progression to castration-resistant prostate cancer (CRPC) is almost inevitable [1]. Docetaxel-based chemotherapy is a standard of care for patients with metastatic CRPC [2]. However, about half of the patients do not respond to the treatment, and those who do respond become refractory within one year [2]. Several new therapies have been recently developed and approved for treating docetaxel-resistant metastatic CRPC, such as the new taxane cabazitaxel [3], the CYP17A1 inhibitor abiraterone that suppresses androgen biosynthesis [4], and the potent AR antagonist enzalutamide [5]. Nonetheless, the survival benefits remain modest (<5 months), and resistance develops in essentially all patients [35]. Thus, developing effective therapeutic modality for prostate cancer remains an urgent task.

Methylseleninic acid (MSA) and methylselenocysteine (MSC) are two methyl-selenium compounds that have been demonstrated by many in vitro and in vivo studies to have potent anticancer activities against prostate cancer [624], and the safety profile of MSC has been established in humans [25]. It is important to appreciate that the anticancer efficacy of selenium compounds depends on the form and dosage administered [13, 16, 26, 27]. For example, selenomethionine, the first-generation selenium compound that was used in the Selenium and Vitamin E Chemoprevention Trial and showed no protection against prostate cancer [28], has distinct biological and pharmacological properties from MSA and MSC [13, 16, 26, 27, 29]. Unlike MSA and MSC, selenomethionine is ineffective in suppressing the growth of prostate tumors in animal studies [13, 16]. This could be attributed to non-specific incorporation of selenomethionine into proteins in place of methionine, limiting its further metabolism [27]. In contrast, as monomethylated forms of selenium, MSA and MSC can be easily metabolized to the active anticancer metabolite methylselenol [27]. With regard to selenium dosage, most of the preclinical studies that showed a positive association between selenium administration and tumor inhibition were conducted using pharmacological doses of selenium, not the nutritional dose that was used in the Selenium and Vitamin E Chemoprevention Trial. Therefore, the use of potent selenium compounds, such as MSA and MSC, at pharmacological doses is essential for further developing selenium compounds for prostate cancer intervention.

The metabolism of MSC to the active metabolite methylselenol requires the activity of β-lyase, which is mainly expressed in the liver and kidney [27]. In contrast, MSA, as an oxidized form of methylselenol, is readily reduced to methylselenol through a non-enzymatic reaction in cells [26]. Due to the fact that prostate epithelial cells express a low level of β-lyase, MSA is 10 times more potent than MSC in affecting biological processes in vitro, and it can inhibit the growth of prostate cancer cells at in-vivo-relevant concentrations (2–10 μM) [8, 11, 26]. MSA also has excellent anticancer activity in animals [13, 1618, 20].

In order to gain insights into the mechanisms underlying the anticancer activity of monomethylated selenium compounds in prostate cancer, microarray studies have been conducted previously to profile MSA-induced gene expression changes in PC-3 and LNCaP human prostate cancer cells [8, 21]. In the present study, we reanalyzed these microarray datasets to compile the list of most highly modulated genes in both cell models, and the analysis and downstream studies led to the identification of growth differentiation factor 15 (GDF15) as a rapidly induced target of MSA. GDF15 is a divergent member of the transforming growth factor-β (TGF-β) superfamily, and it is also known as macrophage inhibitory cytokine-1, prostate differentiation factor, prostate-derived factor, non-steroidal anti-inflammatory drug-activated gene-1, placental bone morphogenic protein, and placental TGF-β [30]. Similar to the role of TGF-β in cancer development and progression, both tumor-suppressing and tumor-promoting functions of GDF15 have been reported in different cancer types, including prostate cancer [30]. In the present study, we analyzed several publicly available clinical prostate cancer datasets encompassing over 1000 primary prostate cancer samples and over 200 metastatic CRPC samples to assess the role of GDF15 in prostate cancer progression, and we characterized the functional significance of GDF15 induction in mediating the effects of MSA in inhibiting cell proliferation and inducing apoptosis in prostate cancer cells.

Materials and methods

Cell lines and reagents

LNCaP and PC-3 cells were obtained from ATCC. LAPC4 cells [31] were provided by Dr. Charles Sawyers, then at the University of California, Los Angeles. All cell lines are cultured in RPMI1640 supplemented with penicillin, streptomycin, and 10% fetal bovine serum. Cells used in all experiments were within 3 months of resuscitation of frozen cell stocks established within 3 passages after receipt of the cells. Cell authentication was performed at the Genetica DNA Laboratories, and cells were regularly evaluated for mycoplasma contamination. MSA and MSC were purchased from PharmaSe.

Microarray, RNA-seq, and proteomics data analyses

The microarray profiles of MSA-modulated genes in PC-3 and LNCaP cells were from previously published studies [8, 21]. The MSA-PC-3 dataset was generated using the Affymetrix U95A chip [8], and the MSA-LNCaP dataset was generated using a 3K custom cDNA microarray containing the genes that are modulated by MSA in the MSA-PC-3 dataset [21]. The data analysis was conducted following the previous publications [8, 21].

The TCGA RNA-seq data of 500 primary prostate cancer samples were downloaded from Genomic Data Commons. The RNA-seq data from 3 cohorts of a total of 159 metastatic CRPC samples were downloaded from dbGaP, including 51 samples from the “Stand Up To Cancer East Coast Prostate Cancer Research Group” (SU2C) project (dbGaP accession pht004946.v1.p1) [32], 74 samples from the Prostate Cancer Medically Optimized Genome-Enhanced Therapy (PROMOTE) study (dbGaP accession phs001141.v1.p1) [33], and 34 samples from the Beltran study (dbGaP accession phs000909.v1.p1) [34]. RNA-seq gene expression analysis was performed using RSEM [35]. The transcripts per million (TPM) values from the RSEM output were used for downstream analysis.

The Grasso [36], Taylor [37], and Erho [38] microarray datasets were downloaded from Gene Expression Omnibus with accession numbers GSE35988, GSE21034, and GSE46691, respectively. The data were normalized using the Single Channel Array Normalization algorithm [39].

Gene Set Enrichment Analysis (GSEA) was performed with 1000 permutations. For analysis of the MSA-PC-3 microarray dataset, the genes were ranked using the Manhattan matrix with a continuous phenotype profile that shows steadily increasing gene expression across the time series. For analysis of the compiled RNA-seq data of 500 primary prostate tumors and the 159 metastatic CRPC samples, the genes were ranked using the Manhattan matrix with continuous phenotype labels for GDF15. Both datasets were run against the hallmark gene sets in the Molecular Signatures Database.

The Excel file containing processed protein expression data from the Latonen proteomics study [40] was downloaded from the Nature Communications website.

Quantitative Reverse Transcription-PCR (qRT-PCR)

qRT-PCR was performed using the TaqMan method. The TaqMan PCR primers and probes for GDF15 and β-actin were from Applied Biosystems. The qRT-PCR analysis was performed at least three times in triplicate, and GDF15 levels were normalized by β-actin levels.

Western blot analysis

Western blot analysis was performed using a standard protocol. After blocking in blocking buffer, the membrane was incubated with a primary antibody overnight at 4°C, followed by incubation with a fluorescent-labeled secondary antibody for 1 hour at room temperature. Membranes were scanned and analyzed using an Odyssey® Infrared scanner (LI-COR Bioscience). The following antibodies were used: anti-glyceraldehyde-3-phosphate dehydrogenase (GAPDH, Millipore) and anti-GDF15 (US Biological). The Western blot analysis was done at least three times, and GDF15 levels were normalized by GAPDH levels.

22Rv1 xenograft tumor model

The 22Rv1 xenograft tumors were from a previous study [20]. The mixture of Ketamine (100 mg/kg) and Xylazine (10 mg/kg) was intraperitoneally injected for anesthesia before tumor cell inoculation. The mice were inspected daily to determine any untoward effect and avoid any unnecessary suffering. Daily administration of MSA or MSC at 3 mg selenium/kg/day by an oral route was started when the tumors reached ~100 mm3. On Day 15, mice were euthanized by CO2 asphyxiation and tumors excised for molecular analysis. All animal procedures were approved by Tulane University Institutional Animal Care and Use Committee.

shGDF15 transfection

The shGDF15 construct was generated as previously described [41]. LNCaP cells were transfected with the shGDF15 or the scrambled control (shCtrl) construct using the Lipofectamine 2000 and Plus reagent (Invitrogen) per instruction of the manufacturer. The cells were treated with MSA at 24 h after transfection. BrdU ELISA, Cell Death ELISA, and Western blot analysis were conducted at 24 h post-MSA treatment.

Cell proliferation assay

Proliferation was measured by using the BrdU Cell Proliferation ELISA kit (Roche) per instruction of the manufacturer with minor modifications. Briefly, after labeling the cells with BrdU for 2 h, the WST-1 reagent (Roche), which quantitatively monitors metabolic activity of the cells, was added to the wells to a final concentration of 10%. The cells were incubated for an additional 2 h. The amount of formazan converted from WST-1 by the metabolically active cells was quantitated at 450 nm. After removing the medium, the cells were fixed and the DNA denatured for the incorporated BrdU to bind to a peroxidase-conjugated anti-BrdU antibody. The immune complexes were detected by the subsequent substrate reaction, and the reaction product was quantified by absorbance at 370 nm (reference wavelength at 492 nm). Culture medium without cells and cells incubated with the anti-BrdU-peroxidase antibody in the absence of BrdU were used as controls for nonspecific binding. The BrdU ELISA result was normalized by the WST-1 reading, which correlates directly with cell number. The experiment was performed three times in triplicate.

Apoptosis detection

Detached cells were precipitated by centrifugation and pooled with attached cells. Cytoplasmic histone-associated DNA fragments were quantified by using the Cell Death Detection ELISAPLUS Kit (Roche) per manufacturer’s protocol. The absorbance was measured at 405 nm (reference wavelength at 492 nm). The experiment was performed three times in triplicate.

Results

GDF15 is one of the most highly induced targets of MSA

To identify key targets of MSA in prostate cancer, we analyzed the microarray data from the previous studies in PC-3 and LNCaP human prostate cancer cells treated with or without 10 μM MSA for 3, 6, 12, 24, 36, or 48 hours [8, 21]. Twenty-one genes (16 upregulated and 5 downregulated) were modulated by MSA by an average of more than 2 fold in both cell models, and GDF15 was among the most induced genes (Fig 1A). We then conducted qRT-PCR analysis to verify the microarray finding. In concordance with the pattern of changes observed in the microarray analysis, GDF15 mRNA was induced early in response to MSA treatment (Fig 1B and 1C). A significant increase was already evident at the 3-hour time point in LNCaP cells and at the 6-hour time point in PC-3 and LAPC-4 cells (Fig 1B). A similar induction was observed in vivo in 22Rv1 human prostate cancer xenograft tumors (Fig 1C). Moreover, MSC, which generates the same active metabolite as MSA in animals [27], exerted the same effect on GDF15 expression in 22Rv1 tumors (Fig 1C). The induction of GDF15 also occurred at the protein level (Fig 1D). Together, the above data from different preclinical models of prostate cancer support GDF15 as one of the highly induced and immediate targets of MSA.

<h2>MSA upregulates the expression of GDF15.</h2>
Fig. 1.

MSA upregulates the expression of GDF15.


A. Microarray data showing GDF15 as one of the most highly induced genes by MSA in both PC-3 and LNCaP cells. Shown are all the genes that are modulated by MSA by at least a mean of 2 fold in both cell models. Colors indicate log2-transformed fold of change after MSA treatment. B. qRT-PCR analysis confirming MSA induction of GDF15 mRNA level in cultured cells. C. qRT-PCR showing induction of GDF15 mRNA level by MSA and MSC in 22Rv1 xenograft tumors (n = 3 for the control group; n = 4 for the MSA and MSC groups). D. Western blotting showing MSA induction of GDF15 protein level in LNCaP cells. All MSA treatment of cultured cells is at 10 μM. Bars, SEM. *, P < 0.05 from control using Student’s t test.

GDF15 contributes to MSA inhibition of cell proliferation and induction of apoptosis

To investigate the functional significance of GDF15 induction by MSA, we knocked down the expression of GDF15 in LNCaP cells and assessed the impact on MSA inhibition of cell proliferation and induction of apoptosis. We lowered the dose of MSA from 10 μM to 5 μM for the BrdU ELISA assay in order to gauge the effect of MSA on cell proliferation in the absence of marked apoptosis. The BrdU ELISA data are presented as relative to the untreated shCtrl group or percentage of inhibition by MSA in shCtrl and shGDF15 cells (Fig 2A), and the apoptosis ELISA data are presented as mean OD reading at 405 nm subtracted by the OD reading of incubation buffer for individual groups (Fig 2B). MSA became less effective in inhibiting cell proliferation and inducing apoptosis after GDF15 knockdown, supporting an important role of GDF15 upregulation in mediating these biological activities of MSA. Additionally, the GDF15 knockdown cells proliferated faster than the control cells in the absence of MSA (Fig 2A), indicating a growth suppressive function of GDF15 in these cells.

<h2>GDF15 knockdown attenuates MSA inhibition of cell proliferation and induction of apoptosis.</h2>
Fig. 2.

GDF15 knockdown attenuates MSA inhibition of cell proliferation and induction of apoptosis.


LNCaP cells were transfected with the shGDF15 or the shCtrl construct. The cells were treated with MSA at 24 h after transfection. BrdU ELISA, Cell Death ELISA, and Western blot analysis were conducted at 24 h post-MSA treatment. A. BrdU cell proliferation ELISA showing GDF15 knockdown promoting cell proliferation and attenuating the inhibitory effect of MSA on cell proliferation in LNCaP cells and attenuating the inhibitory effect of MSA on cell proliferation in LNCaP cells. Data are presented as relative to the untreated shCtrl group (left panel) or percentage of inhibition by MSA in control and GDF15 knockdown cells (right panel). Δ, P < 0.05 using Student’s t test. B. Cell death ELISA showing GDF15 attenuating MSA induction of apoptosis in LNCaP cells. Data are presented as mean OD reading at 405 nm subtracted by the OD reading of incubation buffer for individual groups. C. Western blotting showing the efficacy of GDF15 knockdown. The numbers below the blots denote relative normalized intensities of the GDF15 protein bands compared to the untreated shCtrl value of 1. Treatment duration, 24 h. Bars, SEM. *, P < 0.05 from control using Student’s t test.

GDF15 RNA and protein levels are higher in primary human prostate cancers compared to metastatic tissues

To assess the clinical relevance of increased GDF15 expression in prostate cancer, we examined a number of published cohorts for GDF15 expression. Collating the RNA-seq data from 500 primary tumors in TCGA cohort and the RNA-seq data of 159 metastatic CRPC samples from the SU2C project (n = 51) [32], the PROMOTE study (n = 74) [33], and the Beltran cohort (n = 34) [34] showed significantly higher levels of GDF15 mRNA in primary prostate cancers compared to metastatic CRPC samples (Fig 3A). Analysis of the Grasso and the Taylor microarray datasets [36, 37] yielded the same result (Fig 3B and 3C). We further evaluated GDF15 protein levels in the 33 primary prostate cancer specimens and 18 metastatic CRPC samples included in the proteomics study by Latonen et al. [40] and found that GDF15 protein expression is also significantly higher in primary prostate cancers than in metastatic CRPC samples (Fig 3D). These results indicate that elevated levels of GDF15 are associated with less advanced prostate cancer.

<h2>GDF15 RNA and protein levels are higher in primary human prostate cancer compared to metastatic samples.</h2>
Fig. 3.

GDF15 RNA and protein levels are higher in primary human prostate cancer compared to metastatic samples.


A, GDF15 RNA levels from RNA-seq data of 500 TCGA primary prostate cancer (primary) samples and 159 metastatic CRPC (mCRPC) samples from the SU2C, PROMOTE, and Beltran cohorts. B, GDF15 RNA levels from the Grasso microarray dataset (GSE35988) including 59 primary prostate cancer and 35 mCRPC samples. C, GDF15 RNA levels from the Taylor microarray dataset (GSE21034) including 131 primary and 19 metastatic prostate cancer samples. D, GDF15 protein levels from the Latonen proteomics dataset (PASS01126) including 33 primary prostate cancer and 18 mCRPC samples. TPM, transcripts per million. Numbers in parentheses, number of samples in each group. P values are from Mann-Whitney test.

High GDF15 levels in primary prostate cancers are associated with better prognosis

To assess the prognostic value of GDF15 expression in primary prostate cancer, we analyzed the Erho microarray dataset, which contains 545 radical prostatectomy samples with a median follow-up of 16.9 years [38], for association with clinicopathologic parameters. As shown in Fig 4A, GDF15 mRNA level was lower in Gleason score 9 and 10 tumors than in Gleason score 7 tumors. Moreover, lower GDF15 levels were associated with shorter time to distant metastasis (Fig 4B) and to prostate cancer-specific death (Fig 4C). Together, the association between lower GDF15 expression and worse clinical outcome supports a role of GDF15 downregulation in prostate cancer progression.

<h2>Low GDF15 levels are associated with high Gleason scores in primary tumors and shorter survival of the patients.</h2>
Fig. 4.

Low GDF15 levels are associated with high Gleason scores in primary tumors and shorter survival of the patients.


Data are from the Erho GenomeDx Biosciences cohort. A, association of low GDF15 expression with high Gleason scores. P values are from Mann-Whitney test. B & C, Kaplan-Meier plots showing association of low GDF15 expression with shorter time to distant metastasis (B) and to prostate cancer-specific death (C) after radical prostatectomy. P values and hazard ratios (HR) are from Log-rank test. Numbers in parentheses, number of samples in each group.

Same pathways are shared by GDF15 and MSA

To explore the mechanism by which GDF15 contributes to the antiproliferative and apoptosis-inducing activities of MSA, we conducted GSEA on the microarray data from PC-3 cells treated with or without MSA for different durations and the collated RNA-seq data of 500 primary tumors and 159 metastatic CRPC samples. We found that the pathways that were highly enriched in the transcriptional profiles of tumors that express a low level of GDF15 were similarly enriched in untreated control PC-3 cells compared to MSA-treated cells (Fig 5). Since most, if not all, of these pathways have been implicated in prostate cancer progression, suppressing the activities of these pathways by inducing GDF15 is consistent with the antitumor effect of MSA in prostate cancer.

<h2>Same pathways are affected GDF15 and MSA.</h2>
Fig. 5.

Same pathways are affected GDF15 and MSA.


GSEA was conducted on the combined RNA-seq data of 500 TCGA primary prostate cancer (primary) samples and 159 metastatic CRPC (mCRPC) samples from the SU2C, PROMOTE, and Beltran cohorts and the microarray data from PC-3 cells treated with or without MSA. The same pathways that are negatively correlated with GDF15 levels in the combined RNA-seq data (A) are negatively correlated with MSA treatment in the PC-3-MSA microarray dataset (B). FDR < 0.25 is considered significant.

Discussion

Since the first report of the growth inhibitory activities of MSA in prostate cancer in 2001 [11], numerous studies have been conducted to unravel the underlying mechanisms. However, our understanding of the immediate targets that are key to the growth inhibitory effects of MSA is still limited. Here, we showed that GDF15 is one of the most highly induced genes by MSA in multiple preclinical prostate cancer models in vitro and in vivo and that the induction occurred rapidly. GDF15 has been shown to function either in the nucleus or as a secreted protein [30, 42]. Nuclear GDF15 was shown to inhibit TGF-β1-induced Smad signaling to mitigate cell migration and invasion [42]. Consistently, we found that TGF-β signaling is negatively correlated with GDF15 levels in clinical prostate cancer samples. Moreover, TGF-β signaling is also negatively correlated with MSA treatment in cultured cells. The data indicate that nuclear GDF15 may be involved in mediating the growth-suppressive effects of MSA through inhibiting TGF-β signaling. As for secreted GDF15, the only receptor that has been identified to date is the orphan receptor glial-derived neurotrophic factor-family receptor α-like (GFRAL), which was reported to mediate the metabolic effects of GDF15 [43]. However, through surveying our collated RNA-seq data, we found that the levels of GFRAL mRNA in benign prostate tissues, primary prostate cancer, and metastatic CRPC samples are very low, with mean TPM values of 0.22, 0.13, and 0.13, respectively, as opposed to 134, 459, and 127, respectively, for GDF15 (S3 Fig). Thus, secreted GDF15 is likely to signal through a different receptor in the prostate.

Several transcription factors have been indicated to regulate the expression of GDF15 [44]. Of particular interest are p53 [45, 46] and androgen receptor (AR) [47] as they are also immediate targets of MSA [6, 7, 22, 48]. However, since MSA is able to induce GDF15 expression in both p53 wild-type and p53-null cells and in both AR-expressing and AR-null cells, GDF15 upregulation by MSA is likely to be independent of p53 and AR. Endoplasmic reticulum (ER) stress has also been shown to increase GDF15 expression [49], and MSA is known to induce ER stress in prostate cancer cells [19]. Nonetheless, when we overexpressed GRP78, the rheostat of ER stress transducers and the overexpression of which mitigates MSA induction of ER stress [19], in LNCaP cells, the effect on MSA upregulation of GDF15 was minimal (S4 Fig), excluding ER stress as an underlying mechanism. Understanding how MSA induces GDF15 expression is an area of our ongoing research.

Both tumor-suppressing and tumor-promoting functions of GDF15 in the prostate have been implicated [30]. For example, immunohistochemical staining of prostatectomy specimens showed an inverse association between GDF-15 levels and prostatic inflammation, a known prostate tumor-promoting factor [50]. TRAMP mice bearing a germline deletion of GDF15 develop larger prostatic tumors than TRAMP mice with wild-type GDF15 [51]. On the other hand, both tissue and serum levels of GDF15 have been shown to be elevated in prostate cancer patients relative to non-cancerous individuals [5254]. Ectopic expression of GDF15 has been shown to decrease cell growth and induce apoptosis in some prostate cancer cell models [55, 56] but increase cell growth and metastatic potential in some other models or other conditions [5759].

The seemingly paradoxical role of GDF-15 in prostate cancer might be attributed to the biphasic regulation of GDF15 expression in early-stage of tumor development versus during tumor progression. Albeit GDF15 expression is downregulated in metastatic CRPCs compared to primary tumors, we found that the levels of GDF15 RNA and protein are higher in primary prostate tumors than in benign prostate tissues (S1 and S2 Figs). This is consistent with the report from a previous study, in which immunohistochemical staining of tissue microarrays containing over 1600 cores of benign prostate tissues, low- and high-grade prostatic intraepithelial neoplasia, and primary prostate cancer samples showed a progressive increase of GDF15 protein levels from benign tissues to low- and high-grade prostatic intraepithelial neoplasia and to prostate cancer [60]. However, the same study showed that in prostate cancer samples, lower GDF15 expression is associated with higher Gleason pattern and pathologic stage and increased risk of relapse after surgery [60]. This biphasic regulation of GDF15 expression was also observed in another study, which demonstrated a higher level of GDF15 mRNA in prostate cancer than in benign prostate tissues but a lower level of GDF15 mRNA in moderately and poorly differentiated adenocarcinomas than in well-differentiated prostate cancer [61]. Therefore, the role of GDF15 in prostate cancer is likely to be stage specific. It may promote early stages of tumorigenesis but suppress the progression of advanced prostate cancer. Consequently, the role of GDF15 in mediating the growth-suppressive effects of MSA is likely to be specific to prostate cancer cells that are at an advanced stage.

In summary, the work described herein demonstrates that GDF15 is an immediate target of MSA in prostate cancer cells and that GDF15 induction contributes to MSA inhibition of cell proliferation and induction of apoptosis. Since low GDF15 expression is associated with more aggressive prostate cancer and worse clinical outcome, induction of GDF15 may be a viable approach to treat advanced prostate cancer and prevent prostate cancer progression. By improving our understanding of the mechanisms of action of MSA in prostate cancer, these findings will help future design of clinical trials using MSA to treat prostate cancer.

Supporting information

S1 Fig [mcrpc]
GDF15 mRNA levels are higher in primary prostate cancer compared to benign prostate tissues.

S2 Fig [tif]
GDF15 protein levels are higher in primary human prostate cancer compared to benign prostate tissues.

S3 Fig [mcrpc]
GFRAL mRNA levels are low in the prostate.

S4 Fig [tif]
GRP78 knockdown does not affect MSA upregulation of GDF15.


Zdroje

1. Knudsen KE, Scher HI. Starving the addiction: new opportunities for durable suppression of AR signaling in prostate cancer. ClinCancer Res. 2009;15(15):4792–8.

2. Tannock IF, de Wit R, Berry WR, Horti J, Pluzanska A, Chi KN, et al. Docetaxel plus Prednisone or Mitoxantrone plus Prednisone for Advanced Prostate Cancer. New England Journal of Medicine. 2004;351(15):1502–12. doi: 10.1056/NEJMoa040720 15470213

3. de Bono JS, Oudard S, Ozguroglu M, Hansen S, Machiels JP, Kocak I, et al. Prednisone plus cabazitaxel or mitoxantrone for metastatic castration-resistant prostate cancer progressing after docetaxel treatment: a randomised open-label trial. Lancet. 2010;376(9747):1147–54. doi: 10.1016/S0140-6736(10)61389-X 20888992

4. Fizazi K, Scher HI, Molina A, Logothetis CJ, Chi KN, Jones RJ, et al. Abiraterone acetate for treatment of metastatic castration-resistant prostate cancer: final overall survival analysis of the COU-AA-301 randomised, double-blind, placebo-controlled phase 3 study. Lancet Oncol. 2012;13(10):983–92. doi: 10.1016/S1470-2045(12)70379-0 22995653

5. Scher HI, Fizazi K, Saad F, Taplin ME, Sternberg CN, Miller K, et al. Increased survival with enzalutamide in prostate cancer after chemotherapy. NEnglJMed. 2012;367(13):1187–97.

6. Cho SD, Jiang C, Malewicz B, Dong Y, Young CY, Kang KS, et al. Methyl selenium metabolites decrease prostate-specific antigen expression by inducing protein degradation and suppressing androgen-stimulated transcription. MolCancer Ther. 2004;3(5):605–11.

7. Dong Y, Lee SO, Zhang H, Marshall J, Gao AC, Ip C. Prostate specific antigen expression is down-regulated by selenium through disruption of androgen receptor signaling. Cancer Res. 2004;64(1):19–22. 14729601

8. Dong Y, Zhang H, Hawthorn L, Ganther HE, Ip C. Delineation of the Molecular Basis for Selenium-induced Growth Arrest in Human Prostate Cancer Cells by Oligonucleotide Array. Cancer Research. 2003;63(1):52–9. 12517777

9. Gasparian AV, Yao YJ, Lu J, Yemelyanov AY, Lyakh LA, Slaga TJ, et al. Selenium compounds inhibit I kappa B kinase (IKK) and nuclear factor-kappa B (NF-kappa B) in prostate cancer cells. MolCancer Ther. 2002;1(12):1079–87.

10. Gundimeda U, Schiffman JE, Chhabra D, Wong J, Wu A, Gopalakrishna R. Locally generated methylseleninic acid induces specific inactivation of protein kinase C isoenzymes: relevance to selenium-induced apoptosis in prostate cancer cells. J Biol Chem. 2008;283(50):34519–31. doi: 10.1074/jbc.M807007200 18922790.

11. Jiang C, Wang Z, Ganther H, Lu J. Caspases as key executors of methyl selenium-induced apoptosis (anoikis) of DU-145 prostate cancer cells. Cancer Res. 2001;61(7):3062–70. 11306488

12. Lee SO, Yeon CJ, Nadiminty N, Trump DL, Ip C, Dong Y, et al. Monomethylated selenium inhibits growth of LNCaP human prostate cancer xenograft accompanied by a decrease in the expression of androgen receptor and prostate-specific antigen (PSA). The Prostate. 2006;66(10):1070–5. doi: 10.1002/pros.20329 16637076

13. Li GX, Lee HJ, Wang Z, Hu H, Liao JD, Watts JC, et al. Superior in vivo inhibitory efficacy of methylseleninic acid against human prostate cancer over selenomethionine or selenite. Carcinogenesis. 2008;29(5):1005–12. doi: 10.1093/carcin/bgn007 18310093

14. Liu Y, Liu X, Guo Y, Liang Z, Tian Y, Lu L, et al. Methylselenocysteine preventing castration-resistant progression of prostate cancer. Prostate. 2015;75(9):1001–8. doi: 10.1002/pros.22987 25754033.

15. Pinto JT, Sinha R, Papp K, Facompre ND, Desai D, El-Bayoumy K. Differential effects of naturally occurring and synthetic organoselenium compounds on biomarkers in androgen responsive and androgen independent human prostate carcinoma cells. Int J Cancer. 2007;120(7):1410–7. doi: 10.1002/ijc.22500 17205524.

16. Wang L, Bonorden MJ, Li GX, Lee HJ, Hu H, Zhang Y, et al. Methyl-selenium compounds inhibit prostate carcinogenesis in the transgenic adenocarcinoma of mouse prostate model with survival benefit. Cancer PrevRes(Phila Pa). 2009;2(5):484–95.

17. Wang L, Guo X, Wang J, Jiang C, Bosland MC, Lu J, et al. Methylseleninic Acid Superactivates p53-Senescence Cancer Progression Barrier in Prostate Lesions of Pten-Knockout Mouse. Cancer Prev Res (Phila). 2016;9(1):35–42. doi: 10.1158/1940-6207.CAPR-15-0236 26511486.

18. Wang L, Zhang J, Zhang Y, Nkhata K, Quealy E, Liao JD, et al. Lobe-specific lineages of carcinogenesis in the transgenic adenocarcinoma of mouse prostate and their responses to chemopreventive selenium. Prostate. 2011;71(13):1429–40. doi: 10.1002/pros.21360 21360561.

19. Wu Y, Zhang H, Dong Y, Park YM, Ip C. Endoplasmic reticulum stress signal mediators are targets of selenium action. Cancer Res. 2005;65(19):9073–9. doi: 10.1158/0008-5472.CAN-05-2016 16204082

20. Zhan Y, Cao B, Qi Y, Liu S, Zhang Q, Zhou W, et al. Methylselenol prodrug enhances MDV3100 efficacy for treatment of castration-resistant prostate cancer. Int J Cancer. 2013;133(9):2225–33. doi: 10.1002/ijc.28202 23575870

21. Zhang H, Dong Y, Zhao H, Brooks JD, Hawthorn L, Nowak N, et al. Microarray data mining for potential selenium targets in chemoprevention of prostate cancer. Cancer Genomics and Proteomics. 2005;2:97–114. 18548127

22. Zhao H, Whitfield ML, Xu T, Botstein D, Brooks JD. Diverse effects of methylseleninic acid on the transcriptional program of human prostate cancer cells. MolBiolCell. 2004;15(2):506–19.

23. Sinha I, Allen JE, Pinto JT, Sinha R. Methylseleninic acid elevates REDD1 and inhibits prostate cancer cell growth despite AKT activation and mTOR dysregulation in hypoxia. Cancer Med. 2014;3(2):252–64. doi: 10.1002/cam4.198 24515947.

24. Sinha I, Null K, Wolter W, Suckow MA, King T, Pinto JT, et al. Methylseleninic acid downregulates hypoxia-inducible factor-1alpha in invasive prostate cancer. Int J Cancer. 2012;130(6):1430–9. doi: 10.1002/ijc.26141 21500193.

25. Marshall JR, Burk RF, Payne Ondracek R, Hill KE, Perloff M, Davis W, et al. Selenomethionine and methyl selenocysteine: multiple-dose pharmacokinetics in selenium-replete men. Oncotarget. 2017;8(16):26312–22. doi: 10.18632/oncotarget.15460 28412747.

26. Ip C, Thompson HJ, Zhu Z, Ganther HE. In vitro and in vivo studies of methylseleninic acid: evidence that a monomethylated selenium metabolite is critical for cancer chemoprevention. Cancer Res. 2000;60(11):2882–6. 10850432

27. Ip C. Lessons from basic research in selenium and cancer prevention. J Nutr. 1998;128(11):1845–54. doi: 10.1093/jn/128.11.1845 9808633

28. Lippman SM, Klein EA, Goodman PJ, Lucia MS, Thompson IM, Ford LG, et al. Effect of selenium and vitamin E on risk of prostate cancer and other cancers: the Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA. 2009;301(1):39–51. doi: 10.1001/jama.2008.864 19066370

29. Ohta Y, Kobayashi Y, Konishi S, Hirano S. Speciation analysis of selenium metabolites in urine and breath by HPLC- and GC-inductively coupled plasma-MS after administration of selenomethionine and methylselenocysteine to rats. ChemRes Toxicol. 2009;22(11):1795–801.

30. Wang X, Baek SJ, Eling TE. The diverse roles of nonsteroidal anti-inflammatory drug activated gene (NAG-1/GDF15) in cancer. Biochem Pharmacol. 2013;85(5):597–606. doi: 10.1016/j.bcp.2012.11.025 23220538.

31. Klein KA, Reiter RE, Redula J, Moradi H, Zhu XL, Brothman AR, et al. Progression of metastatic human prostate cancer to androgen independence in immunodeficient SCID mice. NatMed. 1997;3(4):402–8.

32. Robinson D, Van Allen EM, Wu YM, Schultz N, Lonigro RJ, Mosquera JM, et al. Integrative clinical genomics of advanced prostate cancer. Cell. 2015;161(5):1215–28. doi: 10.1016/j.cell.2015.05.001 26000489

33. Wang L, Dehm SM, Hillman DW, Sicotte H, Tan W, Gormley M, et al. A prospective genome-wide study of prostate cancer metastases reveals association of wnt pathway activation and increased cell cycle proliferation with primary resistance to abiraterone acetate-prednisone. Ann Oncol. 2018;29(2):352–60. doi: 10.1093/annonc/mdx689 29069303.

34. Beltran H, Prandi D, Mosquera JM, Benelli M, Puca L, Cyrta J, et al. Divergent clonal evolution of castration-resistant neuroendocrine prostate cancer. Nat Med. 2016;22(3):298–305. doi: 10.1038/nm.4045 26855148.

35. Li B, Dewey CN. RSEM: accurate transcript quantification from RNA-Seq data with or without a reference genome. BMCBioinformatics. 2011;12:323.

36. Grasso CS, Wu YM, Robinson DR, Cao X, Dhanasekaran SM, Khan AP, et al. The mutational landscape of lethal castration-resistant prostate cancer. Nature. 2012;487(7406):239–43. doi: 10.1038/nature11125 22722839

37. Taylor BS, Schultz N, Hieronymus H, Gopalan A, Xiao Y, Carver BS, et al. Integrative genomic profiling of human prostate cancer. Cancer Cell. 2010;18(1):11–22. doi: 10.1016/j.ccr.2010.05.026 20579941

38. Erho N, Crisan A, Vergara IA, Mitra AP, Ghadessi M, Buerki C, et al. Discovery and validation of a prostate cancer genomic classifier that predicts early metastasis following radical prostatectomy. PLoS One. 2013;8(6):e66855. doi: 10.1371/journal.pone.0066855 23826159.

39. Piccolo SR, Sun Y, Campbell JD, Lenburg ME, Bild AH, Johnson WE. A single-sample microarray normalization method to facilitate personalized-medicine workflows. Genomics. 2012;100(6):337–44. doi: 10.1016/j.ygeno.2012.08.003 22959562.

40. Latonen L, Afyounian E, Jylha A, Nattinen J, Aapola U, Annala M, et al. Integrative proteomics in prostate cancer uncovers robustness against genomic and transcriptomic aberrations during disease progression. Nat Commun. 2018;9(1):1176. doi: 10.1038/s41467-018-03573-6 29563510.

41. Appierto V, Villani MG, Cavadini E, Gariboldi M, De Cecco L, Pierotti MA, et al. Analysis of gene expression identifies PLAB as a mediator of the apoptotic activity of fenretinide in human ovarian cancer cells. Oncogene. 2007;26(27):3952–62. doi: 10.1038/sj.onc.1210171 17213814.

42. Min KW, Liggett JL, Silva G, Wu WW, Wang R, Shen RF, et al. NAG-1/GDF15 accumulates in the nucleus and modulates transcriptional regulation of the Smad pathway. Oncogene. 2016;35(3):377–88. doi: 10.1038/onc.2015.95 25893289.

43. Emmerson PJ, Wang F, Du Y, Liu Q, Pickard RT, Gonciarz MD, et al. The metabolic effects of GDF15 are mediated by the orphan receptor GFRAL. Nat Med. 2017;23(10):1215–9. doi: 10.1038/nm.4393 28846098.

44. Vanhara P, Hampl A, Kozubik A, Soucek K. Growth/differentiation factor-15: prostate cancer suppressor or promoter? Prostate Cancer Prostatic Dis. 2012;15(4):320–8. doi: 10.1038/pcan.2012.6 22370725.

45. Cheng JC, Chang HM, Leung PC. Wild-type p53 attenuates cancer cell motility by inducing growth differentiation factor-15 expression. Endocrinology. 2011;152(8):2987–95. doi: 10.1210/en.2011-0059 21586550.

46. Kelly JA, Lucia MS, Lambert JR. p53 controls prostate-derived factor/macrophage inhibitory cytokine/NSAID-activated gene expression in response to cell density, DNA damage and hypoxia through diverse mechanisms. Cancer Lett. 2009;277(1):38–47. doi: 10.1016/j.canlet.2008.11.013 19100681.

47. Kakehi Y, Segawa T, Wu XX, Kulkarni P, Dhir R, Getzenberg RH. Down-regulation of macrophage inhibitory cytokine-1/prostate derived factor in benign prostatic hyperplasia. Prostate. 2004;59(4):351–6. doi: 10.1002/pros.10365 15065082.

48. Smith ML, Lancia JK, Mercer TI, Ip C. Selenium compounds regulate p53 by common and distinctive mechanisms. Anticancer Res. 2004;24(3a):1401–8. 15274301

49. Park SH, Choi HJ, Yang H, Do KH, Kim J, Kim HH, et al. Two in-and-out modulation strategies for endoplasmic reticulum stress-linked gene expression of pro-apoptotic macrophage-inhibitory cytokine 1. J Biol Chem. 2012;287(24):19841–55. doi: 10.1074/jbc.M111.330639 22511768.

50. Lambert JR, Whitson RJ, Iczkowski KA, La Rosa FG, Smith ML, Wilson RS, et al. Reduced expression of GDF-15 is associated with atrophic inflammatory lesions of the prostate. Prostate. 2015;75(3):255–65. doi: 10.1002/pros.22911 25327758.

51. Husaini Y, Lockwood GP, Nguyen TV, Tsai VW, Mohammad MG, Russell PJ, et al. Macrophage inhibitory cytokine-1 (MIC-1/GDF15) gene deletion promotes cancer growth in TRAMP prostate cancer prone mice. PLoS One. 2015;10(2):e0115189. doi: 10.1371/journal.pone.0115189 25695521.

52. Cheung PK, Woolcock B, Adomat H, Sutcliffe M, Bainbridge TC, Jones EC, et al. Protein profiling of microdissected prostate tissue links growth differentiation factor 15 to prostate carcinogenesis. Cancer Res. 2004;64(17):5929–33. doi: 10.1158/0008-5472.CAN-04-1216 15342369.

53. Li J, Veltri RW, Yuan Z, Christudass CS, Mandecki W. Macrophage inhibitory cytokine 1 biomarker serum immunoassay in combination with PSA is a more specific diagnostic tool for detection of prostate cancer. PLoS One. 2015;10(4):e0122249. doi: 10.1371/journal.pone.0122249 25853582.

54. Hood BL, Darfler MM, Guiel TG, Furusato B, Lucas DA, Ringeisen BR, et al. Proteomic analysis of formalin-fixed prostate cancer tissue. Mol Cell Proteomics. 2005;4(11):1741–53. doi: 10.1074/mcp.M500102-MCP200 16091476.

55. Lambert JR, Kelly JA, Shim M, Huffer WE, Nordeen SK, Baek SJ, et al. Prostate derived factor in human prostate cancer cells: gene induction by vitamin D via a p53-dependent mechanism and inhibition of prostate cancer cell growth. J Cell Physiol. 2006;208(3):566–74. doi: 10.1002/jcp.20692 16741990.

56. Liu T, Bauskin AR, Zaunders J, Brown DA, Pankhurst S, Russell PJ, et al. Macrophage inhibitory cytokine 1 reduces cell adhesion and induces apoptosis in prostate cancer cells. Cancer Res. 2003;63(16):5034–40. 12941831.

57. Senapati S, Rachagani S, Chaudhary K, Johansson SL, Singh RK, Batra SK. Overexpression of macrophage inhibitory cytokine-1 induces metastasis of human prostate cancer cells through the FAK-RhoA signaling pathway. Oncogene. 2010;29(9):1293–302. doi: 10.1038/onc.2009.420 19946339.

58. Chen SJ, Karan D, Johansson SL, Lin FF, Zeckser J, Singh AP, et al. Prostate-derived factor as a paracrine and autocrine factor for the proliferation of androgen receptor-positive human prostate cancer cells. Prostate. 2007;67(5):557–71. doi: 10.1002/pros.20551 17221842.

59. Tsui KH, Chang YL, Feng TH, Chung LC, Lee TY, Chang PL, et al. Growth differentiation factor-15 upregulates interleukin-6 to promote tumorigenesis of prostate carcinoma PC-3 cells. J Mol Endocrinol. 2012;49(2):153–63. doi: 10.1530/JME-11-0149 22872134.

60. Rasiah KK, Kench JG, Gardiner-Garden M, Biankin AV, Golovsky D, Brenner PC, et al. Aberrant neuropeptide Y and macrophage inhibitory cytokine-1 expression are early events in prostate cancer development and are associated with poor prognosis. Cancer Epidemiol Biomarkers Prev. 2006;15(4):711–6. doi: 10.1158/1055-9965.EPI-05-0752 16614113.

61. Kawahara T, Ishiguro H, Hoshino K, Teranishi J, Miyoshi Y, Kubota Y, et al. Analysis of NSAID-activated gene 1 expression in prostate cancer. Urol Int. 2010;84(2):198–202. doi: 10.1159/000277599 20215826.


Článok vyšiel v časopise

PLOS One


2019 Číslo 9
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#