Skip to main content

Contribution of Pyk2 pathway and reactive oxygen species (ROS) to the anti-cancer effects of eicosapentaenoic acid (EPA) in PC3 prostate cancer cells

Abstract

Background

n-3 polyunsaturated fatty acids (n-3 PUFAs), including eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), are thought to exert protective effects in cardiovascular diseases. In addition, n-3 PUFAs have demonstrated anti-cancer effects in vitro and in vivo.

Objective

We investigated the anti-cancer effects and mechanism of action of EPA on PC3 prostate cancer cells in vitro.

Methods

PC3 cells were treated with various concentrations of EPA, and cell survival and the abilities of migration and invasion were evaluated. The time course of the growth inhibitory effect of EPA on PC3 cells was also assessed. The mechanism underlying the anti-cancer effects of EPA was investigated by human phosphokinase and human apoptosis antibody arrays, and confirmed by western blot analysis. We also examined the contribution of reactive oxygen species (ROS) to the effects of EPA using the ROS inhibitor N-acetyl cysteine.

Results

EPA decreased the survival of PC3 cells in a dose-dependent manner within 3 h of application, with an effective concentration of 500 μmol/L. EPA inhibited proline-rich tyrosine kinase (Pyk)2 and extracellular signal-regulated kinase 1/2 phosphorylation as determined by western blotting and the antibody arrays. The growth of PC3 cells was inhibited by EPA, which was dependent on ROS induction, while EPA inhibited Pyk2 phosphorylation independent of ROS production.

Conclusions

Inhibition of Pyk2 phosphorylation and ROS production contribute to the anticancer effects of EPA on PC3 cells.

Introduction

Prostate cancer (PC) is the second most common cancer in men worldwide, with the incidence increasing in Asian countries, including Japan [1, 2]. Standard treatments for PC include surgery, radiation, hormone therapy, and chemotherapy. Advanced PC with metastasis is treated with androgen deprivation therapy (ADT) in association with medical or surgical castration [3]. However, after several years of treatment, patients ultimately develop castration resistance; non-metastatic or metastatic castration-resistant (CR) PC is refractory to ADT and develops mechanisms to proliferate irrespective of castration. Furthermore, anti-androgen therapy can worsen the patient’s condition by stimulating PC growth. This has been demonstrated with flutamide therapy and is known as anti-androgen withdrawal syndrome [4]. Only a few chemotherapeutics and radiation have been developed for the treatment of CRPC [5], and these have not improved the poor prognosis of this disease [6].

There is growing evidence that n-3 polyunsaturated fatty acids (n-3 PUFAs) found in fish oil—especially eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)—can improve lipid metabolism and blood lipid profiles, prevent the progression of atherosclerosis, and reduce the incidence of cardiovascular diseases [7] as well as liver and plasma triglyceride levels [8]. We previously demonstrated that chronic oral administration of EPA prevented endothelial dysfunction in a mouse model of type 2 diabetes [9]. n-3 PUFAs have also been reported to have various anti-cancer effects in several types of malignancy in vitro and in vivo. n-3 PUFAs inhibit extracellular signal-regulated kinase (ERK) and Akt signaling pathways and show anti-cancer effects in breast cancer [10], reduce the incidence of liver cancer in hepatitis virus-infected patients [11], and lower the risk of pancreatic cancer [12]. Cis-unsaturated fatty acids including EPA are easily incorporated into cancer cells and induce free radical generation, inducing tumoricidal action [13,14,15,16]. However, other studies have suggested that there is insufficient evidence for a significant association between n-3 PUFAs and cancer incidence [17].

We previously reported that EPA and DHA have anti-proliferative, −migratory, and -invasive effects in the PC3 CRPC cell line [18]. We also observed that the combination of anti-cancer drugs and n-3 PUFAs synergistically inhibited the proliferation of PC3 cells (unpublished data). However, the mechanism of action of n-3 PUFAs, and specifically, the molecular basis for the inhibitory effects of EPA on PC3 cell proliferation, migration, and invasion has yet to be defined.

Various signaling pathways control cell proliferation; their dysregulation can lead to over-proliferation and aberrant migration and invasion. Tyrosine kinase (TK) is an important mediator of cell proliferation. Receptor (R) TK is a transmembrane receptor with TK activity, whereas non-receptor (NR) TK is present in the cytoplasm and comprises Abl, Src, and proline-rich TK (Pyk)2 families. Various NRTKs are expressed in PC3 cells, and Src plays a role in multiple biological processes in PC cells [19]. Activation of these TKs leads to cell proliferation through activation of transcription factors that promote cell cycle progression.

NRTKs enhance the proliferation of cancer cells to promote their survival, so suppressing these pathways can lead to cell growth inhibition or death. Most cancer cells are resistant to apoptosis and activate pathways that suppress the pro-apoptotic active form of caspases [20]. Moderate ROS levels may cause tumorigenesis through DNA damage in pro-tumorigenic cells [21]. Thus, eliminating or reducing ROS production is a potential strategy for cancer therapy, since ROS also regulate the activation of several RTKs and NRTKs [22]. On the other hand, EPA-induced ROS overproduction stimulates apoptotic signals in HepG2 liver cancer cells [23], and oxidative stress causes PC3 cell death by stimulating mitochondrial ROS production and apoptosis [24]. DHA has been found to cause cell death by inducing ROS production in PC3 and DU145 PC cells [25].

In the present study, we investigated the mechanism underlying the anti-cancer effects of n-3 PUFA EPA, first by characterizing the optimal concentration and time required for these effects in PC3 cells, and then by analyzing protein expression by western blotting and antibody arrays. To clarify the effects of ROS production induced by EPA, we used the ROS inhibitor N-acetyl cysteine (NAC).

Materials and methods

Reagents

EPA (Sigma-Aldrich, St. Louis, MO, USA) was dissolved in 99% isopropanol (Wako, Tokyo, Japan) and stored at − 30 °C. For experiments, EPA was dissolved in serum-free Dulbecco’s modified Eagle’s medium (DMEM; Gibco, New York, NY, USA) containing 3% bovine serum albumin (BSA) (Sigma-Aldrich), and NAC (Wako) was dissolved in phosphate-buffered saline (PBS). Human phospho-kinase array (R&D Systems, Minnesota, MN, USA) and the human apoptosis antibody Array Membranes (Abcam, Cambridge, UK) were used for protein analysis. Antibodies against ERK1/2, phosphorylated (p)-ERK1/2, Pyk2, p-Pyk2, and β-actin were from Cell Signaling Technology Japan (Tokyo, Japan) and were diluted with Can Get Signal reagent (Toyobo, Osaka, Japan).

Cell culture

PC3 cells (Riken BRC Cell Bank, Tsukuba, Japan) were grown in DMEM (Gibco) supplemented with 10% heat-inactivated fetal bovine serum (FBS), 100 U/mL of penicillin, and 100 μg/mL of streptomycin (Gibco). The cells were cultured at 37 °C in a humidified 5% CO2 atmosphere.

Cell proliferation assay

PC3 cells were seeded in 35-mm dishes at a density of 3 × 105 cells/dish in DMEM containing 10% FBS and incubated for 24 h, and the culture medium was replaced with serum-free DMEM after washing with PBS. After an additional 24 h, the cells were washed with PBS, and serum-free medium containing 3% BSA and various concentrations of EPA (100–500 μM) were added. After 24 h, the number of cells was counted with a hemocytometer.

Time course of cell proliferation

PC3 cells were seeded in a 35-mm high μ-Dish Grid-500 (Ibidi, Planegg, Germany) at a density of 3 × 105 cells/dish in DMEM containing 10% FBS. After 24 h, the cells were washed with PBS, and serum-free DMEM was added; after another 24 h, the cells were washed with PBS, and serum-free medium containing 3% BSA with or without EPA (500 μM) was added. Cells were imaged with a microscope and counted at the indicated times.

Migration and invasion assay

Migration and invasion were assessed as described in our previous study [18]. Briefly, we used Transwell membranes (8-μm pore size; Corning Inc., Corning, NY, USA) with or without Matrigel coating. PC3 cells were cultured in the upper chamber of a Transwell insert in serum-free medium containing various concentrations of EPA. The lower chamber was filled with DMEM containing 10% FBS. For the migration assay, Matrigel-uncoated upper chambers were used; cells were seeded at a density of 2 × 105 cells/mL, and after 24 h, the number of cells that had migrated into the lower chamber was counted. For the invasion assay, Matrigel-coated upper chambers were used. Before seeding the cells, Matrigel was fixed by incubation for 90 min, and cells were seeded at a density of 1 × 105 cells/mL. After 48 h, cells that had invaded into the lower chamber were counted.

Antibody arrays

PC3 cells were cultured in DMEM containing 10% FBS for 24 h, then cultured in serum-free medium for an additional 24 h before treatment with EPA (500 μM) or vehicle as a control. After 2 h of incubation with EPA or vehicle, samples were prepared for the antibody array according to the manufacturers’ protocols. Chemiluminescence detection was performed on a Lumi Cube (Liponics, Tokyo, Japan) using Clarity Western ECL Substrate (Bio-Rad, Hercules, CA, USA). Images of the arrays were analyzed using Image J software (National Institutes of Health, Bethesda, MD, USA).

Inhibition of ROS production

PC3 cells were cultured in a 35-mm dish at a density of 3 × 105 cells/dish in DMEM containing 10% FBS for 24 h, and then washed with PBS and incubated in serum-free DMEM for an additional 24 h. The cells were washed with PBS, and EPA (500 μM) with or without 5 mM of NAC (Wako) was added, cells were incubated for 24 h, and cell numbers were counted with a hemocytometer.

Western blot analysis

PC3 cells were cultured in 60-mm dishes at a density of 8 × 105 cells/dish in 10% FBS-containing DMEM for 24 h. PC3 cells were cultured with serum starvation for an additional 24 h, then treated with EPA (500 μM) or vehicle. The cells were lysed in M-PER (Thermo Fisher Scientific, Waltham, MA, USA) and centrifuged at 13,000 rpm (30,250 g) at 4 °C for 10 min. Protein samples were diluted in 2× Laemmli sample buffer (Bio-Rad), then boiled at 100 °C, and 20 μg of protein were loaded in each lane of a TGX precast gel (Bio-Rad) and resolved by electrophoresis (100 V, 0.3 A). The proteins were transferred to an Immun-Blot polyvinylidene difluoride membrane (Bio-Rad) at 100 V and 0.3 A for 1 h. The membrane was blocked with Block Ace (KAC, Kyoto, Japan) for 1 h and probed with antibody solution for 1 h at room temperature. The membrane was washed with Tris-buffered saline (Bio-Rad) containing 1% Tween 20. Lumi Cube and Clarity Western ECL substrate were used for signal detection, and protein band intensity was analyzed using Image J software.

Statistical analysis

Statistical analyses were performed using R v.3.4.3 (https://www.r-project.org/). Data are presented as means ± standard error, and differences between means were evaluated with Dunnett’s test. A P value less than 0.05 was considered statistically significant.

Results

EPA suppresses PC3 cell proliferation

EPA inhibited the proliferation of PC3 cells in a dose-dependent manner (Fig. 1). However, only the highest concentration of EPA (500 μM) significantly reduced the number of cells to 50% of the control value.

Fig. 1
figure 1

Effect of EPA on PC3 cell proliferation. After 24 h of culture in serum-free medium, various concentrations of EPA (0, 100, 300, and 500 μM) were added to the cultures. Data represent mean + SEM (n = 3). **P < 0.01 vs. 0 μM

In serum-free medium, the number of PC3 cells in the control group did not change over a 24-h period, but gradually decreased with EPA treatment, with significant differences detected after 3 h. After 12 h, the number of cells was 50% of the control value and remained constant up to 24 h (Fig. 2).

Fig. 2
figure 2

Effect of EPA on the time course of PC3 cell proliferation. After 24 h of culture in serum-free medium, EPA (500 μM), or vehicle was added to the cells. Data represent mean ± SEM (n = 3). **P < 0.001 vs 0 h; #P < 0.05, ##P < 0.01 v. vehicle

EPA inhibits PC3 cell migration and invasion

EPA inhibited both PC3 cell migration and invasion in a dose-dependent manner, and the rate of migration and invasion relative to the control was 43% (Fig. 3a) and 26% (Fig. 3b), respectively, at 200 μmol/L EPA. Thus, PC3 cell invasion was inhibited to a greater extent than migration by EPA.

Fig. 3
figure 3

Effect of EPA on EPA on PC3 (a) cell migration and (b) invasion. Double-chambered cell culture dishes with a transwell insert separating the two chambers were used to evaluate PC3 cell migration and invasion. Cells were seeded in the upper chamber, which was uncoated (migration) or coated (invasion) with Matrigel, while the lower chamber was filled with DMEM containing 10% FBS. Data represent mean + SEM (n = 3). *P < 0.05

Proteins associated with phosphokinase and apoptosis pathways are activated by EPA treatment

Using two types of antibody arrays, we determined that proteins involved in the phosphokinase and apoptosis pathways were activated by EPA treatment. Based on the results shown in Fig. 2, protein samples for antibody arrays and western blotting were prepared 2 h after adding 500 μmol/L EPA. More significant results were obtained between the EPA-treated and untreated control groups using the phosphokinase as compared to the apoptosis array (Figs. 4 and 5). The expression of apoptosis-related proteins including soluble tumor necrosis factor receptor (sTNF-R)2, second mitochondria-derived activators of caspase (SMAC), Survivin, tumor necrosis factor related apoptosis-inducing ligand receptor (TRAIL-R)1, caspase 8, B-cell lymphoma (Bcl)-2, and p53 differed by more than 20% between the two groups (Table 1), with sTNF-R2 and Bcl-2 being downregulated and the other proteins being upregulated by EPA treatment.

Fig. 4
figure 4

Results of the apoptosis antibody array. PC3 cells with or without EPA treatment were analyzed. The y axis shows the pixel density (%). The expression level of each protein in the control was adjusted to 100%

Fig. 5
figure 5

Results of two experiments using the human phosphokinase array. The y axis shows pixel density (%). For the control groups in experiments 1 and 2 (Expt1 Control and Expt2 Control, respectively), the expression level of each protein was adjusted to 100%. For the EPA-treated groups in experiments 1 and 2 (Expt1 EPA and Expt2 EPA, respectively), the expression level of each protein was determined relative to that in the corresponding control group

Table 1 Difference in expression levels of various proteins relative to the control in PC3 cells treated with EPA

The phosphokinase array data showed increased or decreased phosphorylation of several kinases (Fig. 5 and Table 2). EPA reduced the phosphorylation of several proteins by over 20% relative to the control, including Pyk2, endothelial nitric oxide synthase (eNOS), c-Jun, and ribosomal S6 kinases (RSK)1/2/3.

Table 2 Differences in the expression levels of various proteins in EPA-treated PC3 cells relative to the control (related to Fig. 5)

Contribution of ROS to PC3 cell death

We next analyzed the contribution of ROS to the effects of EPA on PC3 cell survival and found that EPA treatment decreased survival to 60% of the control rate, whereas NAC (5 mmol/L) had no effect. Treatment with both EPA and NAC restored the survival rate to the control level. Thus, NAC abrogates the anti-cancer effect of EPA against PC3 cells (Fig. 6).

Fig. 6
figure 6

Effect of NAC on the anti-cancer effect of EPA against PC3 cells. After 24 h of culture in serum-free medium, cells were treated with EPA (500 μM) or NAC or both. Data represent mean + SEM (n = 3). *P < 0.05

EPA inhibits ERK and Pyk2 phosphorylation

To confirm the mechanism of action of EPA, cells were treated at a concentration of 500 μmol/L, and the expression of native and phosphorylated ERK and Pyk2 was examined by western blotting. EPA significantly suppressed ERK phosphorylation at 1 and 2 h, although there was no difference relative to the control group after 3 h (Fig. 7). A similar significant inhibitory effect was observed on Pyk2 phosphorylation at 2 and 4 h (Fig. 8). Treatment with the ROS inhibitor NAC (5 mM) alone did not affect Pyk2 phosphorylation, while NAC slightly enhanced the inhibitory effect of EPA on Pyk2 phosphorylation (Fig. 9).

Fig. 7
figure 7

Effect of EPA on ERK1/2 phosphorylation. Upper panel: Representative results of western blot analysis. Lower panel: Pixel density of the control group at the indicated times was adjust to 100%; relative pixel density of EPA-treated PC3 cells is shown. Data represent mean + SEM (n = 3). *P < 0.05

Fig. 8
figure 8

Effect of EPA on Pyk2 phosphorylation. Upper panel: Representative result of western blot analysis. Lower panel: Pixel density of the control group at the indicated times was adjusted to 100%; relative pixel density of EPA-treated PC3 cells is shown. Data represent mean + SEM (n = 3). *P < 0.05

Fig. 9
figure 9

NAC enhances the inhibition effect of EPA on Pyk2 phosphorylation. After 24 h of culture in serum-free mediumm, PC3 cells were treated with EPA (500 μM) or NAC or both. Data represent mean + SEM (n + 3). *P < 005

Discussion

The anti-cancer effects of n-3 PUFAs in vitro and in vivo are well known [13, 14, 18, 26,27,28,29,30,31], and these effects have been observed in PC cells. Our previous study showed that the n-3 PUFAs EPA and DHA inhibit PC3 cell proliferation, migration, and invasion [18]. Several reports have indicated that n-3 PUFAs-induced apoptosis or oxidative stress suppresses the growth of various types of cancer cells, including PC cells [32, 33]. Activation of NRTKs may be related to PC cell growth, and clinical trials of therapeutics that target NRTK are currently underway [34, 35]. Other mechanisms of action have been proposed for the anti-cancer effects of n-3 PUFAs [36, 37]. In this study, we addressed this question using phosphokinase and apoptosis antibody arrays and examined the contribution of ROS to the anti-cancer effect of EPA on PC3 cells.

For the antibody array analysis, the cell cycle of PC3 cells was synchronized by culturing in serum-free medium for 24 h. PC3 cells are relatively resistant to the anti-cancer effects of EPA. We treated cells with a moderately high concentration of EPA that was nonetheless within the physiological range (500 μM). PC3 cell proliferation did not change over 24 h in the control group, which is consistent with the notion that the rates of proliferation and death are equivalent in PC3 cells grown under conditions of moderate nutrient starvation. In contrast, the number of cells in the EPA-treated group decreased over time, with significant differences relative to the control group observed from 3 h to 24 h.

The results of the apoptosis array showed that EPA increased the expression of the pro-apoptotic proteins TRAIL-R1, caspase 8, and p53, and decreased that of the anti-apoptotic protein Bcl-2. However, EPA also increased the levels of the anti-apoptotic proteins SMAC and Survivin while having no effect on caspase 3. We speculated that ROS induced cell death via a pathway independent of caspase-induced apoptosis. To test this hypothesis, we evaluated the effect of the ROS inhibitor NAC on EPA-treated cells. First, we measured ROS generation at two time points, 2 and 24 h. In these experiments, we observed relevant (not significant) ROS generation induced by EPA only at 2 h, and NAC reduced the ROS level to the control (data not shown). We observed that the addition of NAC abrogated the anti-cancer effects of EPA (Fig. 6), suggesting that ROS induced by EPA suppressed PC3 cell proliferation. This is supported by previous reports showing that the anti-cancer effects of n-3 PUFAs are mediated by ROS in liver cancer and PC cells [23, 24].

We also investigated the contribution of cell proliferation pathways to the anti-cancer effects of EPA using a phosphokinase antibody array. The results showed that Pyk2, eNOS, c-Jun, and RSK1/2/3 were downregulated by over 20% by EPA treatment as compared to the control group. Western blot analysis revealed that ERK and Pyk2 phosphorylation were inhibited by EPA (Figs. 7 and 8). Suppressing ROS production with NAC did not affect the inhibitory effects of EPA on Pyk2 phosphorylation, suggesting that the suppression of Pyk2 phosphorylation by EPA does not involve modulation of ROS. Pyk2 is a member of the focal adhesion kinase (FAK) family of NRTK and plays important roles in cell survival, proliferation, and migration [38,39,40,41]. In our experiments, FAK phosphorylation was also suppressed by EPA treatment (Table 2). FAK is localized at the site of cell adhesion with the extracellular matrix along with integrins, whereas Pyk2 associates with integrin within the cell [42, 43]. Since several types of integrins are expressed in PC3 cells and interact with extracellular matrix, inhibition of activated FAK at these sites may prevent cell adhesion and migration [44]. The N-terminal domain of FAK has phosphorylation sites for the cell proliferation-related factors epidermal growth factor receptor, c-Met, and Src [45]. The antibody used in the antibody array detected phosphorylated tyrosine 402 of Pyk2, which is necessary for Src activation [40] and was slightly reduced in our experiment (Table 2). It was reported that Pyk2 activation induced smooth muscle contraction by activating Rho-associated kinase signaling [46], and FAK and Pyk2 show similar activities on this pathway in the regulation of cell migration [38, 47, 48]. Activation of both factors leads to abnormalities in cell morphology and movement associated with malignant transformation. Our results suggest that EPA negatively regulates the proliferation, migration, and invasion of PC3 cells via inhibition of Pyk2 and FAK [18].

Pyk2 is an NRTK that is activated by increased intracellular Ca2+ concentrations [49, 50], which are in turn increased by n-3 PUFAs and ROS [23, 51,52,53]. In our results, the n-3 PUFA EPA inhibited Pyk2 phosphorylation, whereas ROS inhibition by NAC did not affect the inhibitory effects of EPA, suggesting that EPA blocks Pyk2 activation independent of intracellular Ca2+ levels.

Conclusions

Our results demonstrate that EPA induced cell death by stimulating ROS production in PC3 cells, and suppressed Pyk2 phosphorylation inducing inhibition of cell proliferation, migration, and invasion partly controlled by downstream Pyk2 signaling. Our findings were summarized in Fig. 10. These results provide a basis for the development of novel therapeutics for the treatment of PC.

Fig. 10
figure 10

EPA-induce anti- cancer signaling in PC3 cells

Availability of data and materials

Available.

Abbreviations

ADT:

Androgen deprivation therapy

Bcl-2:

B-cell lymphoma 2

CRPC:

Castration-resistant prostate cancer

DHA:

Docosahexaenoic acid

eNOS:

Endothelial nitric oxide synthase

EPA:

Eicosapentaenoic acid

ERK:

Extracellular signal-regulated kinase

FAK:

Focal adhesion kinase

n-3 PUFA:

n-3 polyunsaturated fatty acid

NAC:

N-acetyl cysteine

NRTK:

Non-receptor tyrosine kinase

PBS:

Phosphate-buffered saline

PC:

Prostate cancer

Pyk2:

Proline-rich tyrosine kinase-2

ROS:

Reactive oxygen species

RSK1/2/3:

Ribosomal S6 kinase 1/2/3

RTK:

Receptor tyrosine kinase

SMAC:

Second mitochondria-derived activators of caspase

sTNF-R2:

Soluble tumor necrosis factor receptor 2

TRAIL-R1:

Tumor necrosis factor-related apoptosis-inducing ligand and receptor 1

References

  1. Global Burden of Disease Cancer Collaboration, Fitzmaurice C, Akinyemiju TF, Al Lami FH, Alam T, Alizadeh-Navaei R, Allen C, Alsharif U, Alvis-Guzman N, Amini E, et al. Global, regional, and national cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life-years for 29 cancer groups, 1990 to 2016: a systematic analysis for the global burden of disease study. JAMA Oncol. 2018;4(11):1553–68.

    Article  Google Scholar 

  2. Kimura T, Egawa S. Epidemiology of prostate cancer in Asian countries. Int J Urol. 2018;25(6):524–31.

    Article  PubMed  Google Scholar 

  3. Ritch C, Cookson M. Recent trends in the management of advanced prostate cancer. F1000Res. 2018;21:7.

    Google Scholar 

  4. Small EJ, Srinivas S. The antiandrogen withdrawal syndrome. Experience in a large cohort of unselected patients with advanced prostate cancer. Cancer. 1995;76(8):1428–34.

    Article  CAS  PubMed  Google Scholar 

  5. Hurwitz MD. Chemotherapy and radiation for prostate cancer. Transl Androl Urol. 2018;7(3):390–8.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Hussain M, Fizazi K, Saad F, Rathenborg P, Shore N, Ferreia U, Ivashchenko P, Demirthan E, Modelska K, Phung D, et al. Enzalutamide in men with nonmetastatic, castration-resistant prostate cancer. N Engl J Med. 2018;378(26):2465–74.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Balk EM, Lichtenstein AH. Omega-3 fatty acids and cardiovascular disease: summary of the 2016 agency of healthcare research and quality evidence review. Nutrients. 2017;9(8). https://doi.org/10.3390/nu9080865.

    Article  PubMed Central  CAS  Google Scholar 

  8. Harris WS. Fish oils and plasma lipid and lipoprotein metabolism in humans: a critical review. J Lipid Res. 1989;30(6):785–807.

    CAS  PubMed  Google Scholar 

  9. Takenouchi Y, Ohtake K, Nobe K, Kasono K. Eicosapentaenoic acid ethyl ester improves endothelial dysfunction in type 2 diabetic mice. Lipids Health Dis. 2018;17(1):118.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  10. Serini S, Calviello G. Modulation of Ras/ERK and phosphoinositide signaling by long-chain n-3 PUFA in breast cancer and their potential complementary role in combination with targeted drugs. Nutrients. 2017;9(3). https://doi.org/10.3390/nu9030185.

    Article  PubMed Central  CAS  Google Scholar 

  11. Sawada N, Inoue M, Iwasaki M, Sasazuki S, Shimazu T, Yamaji T, Takachi R, Tanaka Y, Mizokami M, Tsugane S, et al. Consumption of n-3 fatty acids and fish reduces risk of hepatocellular carcinoma. Gastroenterology. 2012;142(7):1468–75.

    Article  CAS  PubMed  Google Scholar 

  12. Hidaka A, Shimazu T, Sawada N, Yamaji T, Iwasaki M, Sasazuki S, Inoue M, Tsugane S. Japan public health center-based prospective study group. Fish, n-3 PUFA consumption, and pancreatic cancer risk in Japanese: a large, population-based, prospective cohort study. Am J Clin Nutr. 2015;102(6):1490–7.

    Article  CAS  PubMed  Google Scholar 

  13. Das UN. Tumoricidal action of cis-unsaturated fatty acids and their relationship to free radicals and lipid peroxidation. Cancer Lett. 1991;56(3):235–43.

    Article  CAS  PubMed  Google Scholar 

  14. Bégin ME, Das UN, Ells G, Horrobin DF. Selective killing of human cancer cells by polyunsaturated fatty acids. Prostaglandins Leukot Med. 1985;19(2):177–86.

    Article  PubMed  Google Scholar 

  15. Das UN, Begin ME, Ells G, Huang YS, Horrobin DF. Polyunsaturated fatty acids augment free radical generation in tumor cells in vitro. Biochem Biophys Res Commun. 1987;145(1):15–24.

    Article  CAS  PubMed  Google Scholar 

  16. Das UN, Huang YS, Begin ME, Ells G, Horrobin DF. Uptake and distribution of cis-unsaturated fatty acids and their effect on free radical generation in normal and tumor cells in vitro. Free Radic Biol Med. 1987;3(1):9–14.

    Article  CAS  PubMed  Google Scholar 

  17. MacLean CH, Newberry SJ, Mojica WA, Khanna P, Issa AM, Suttorp MJ, Lim YW, Traina SB, Hilton L, Garland R, et al. Effects of omega-3 fatty acids on cancer risk: a systematic review. JAMA. 2006;295(4):403–15.

    Article  CAS  PubMed  Google Scholar 

  18. Oono K, Takahashi K, Sukehara S, Kurosawa H, Matsumura T, Taniguchi S, Ohta S. Inhibition of PC3 human prostate cancer cell proliferation, invasion and migration by eicosapentaenoic acid and docosahexaenoic acid. Mol Clin Oncol. 2017;7(2):217–20.

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Chang YM, Kung HJ, Evans CP. Nonreceptor tyrosine kinases in prostate cancer. Neoplasia. 2007;9(2):90–100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Watson RW, Fitzpatrick JM. Targeting apoptosis in prostate cancer: focus on caspases and inhibitors of apoptosis proteins. BJU Int. 2005;96(Suppl 2):30–4.

    Article  PubMed  Google Scholar 

  21. Moloney JN, Cotter TG. ROS signaling in the biology of cancer. Semin Cell Dev Biol. 2018;80:50–64.

    Article  CAS  PubMed  Google Scholar 

  22. Thannickal VJ, Fanburg BL. Reactive oxygen species in cell signaling. Am J Physiol Lung Cell Mol Physiol. 2000;279(6):1005–28.

    Article  Google Scholar 

  23. Zhang Y, Han L, Qi W, Cheng D, Ma X, Hou L, Cao X, Wang C. Eicosapentaenoic acid (EPA) induced apoptosis in HepG2 cells through ROS-Ca2+-JNK mitochondrial pathways. Biochem Biophys Res Commun. 2015;456(4):926–32.

    Article  CAS  PubMed  Google Scholar 

  24. Ding Y, Ren K, Dong H, Song F, Chen J, Guo Y, Liu Y, Tao W, Zhang Y. Flavonoids from persimmon (Diospyros kaki L.) leaves inhibit proliferation and induce apoptosis in PC-3 cells by activation of oxidative stress and mitochondrial apoptosis. Chem Biol Interact. 2017;275:210–7.

    Article  CAS  PubMed  Google Scholar 

  25. Shin S, Jing K, Jeong S, Kim N, Song KS, Heo JY, Park JH, Seo KS, Han J, Park JI, et al. The omega-3 polyunsaturated fatty acid DHA induces simultaneous apoptosis and autophagy via mitochondrial ROS-mediated Akt-mTOR signaling in prostate cancer cells expressing mutant p53. Biomed Res Int. 2013;2013:568671.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  26. Bégin ME, Ells G, Das UN, Horrobin DF. Differential killing of human carcinoma cells supplemented with n-3 and n-6 polyunsaturated fatty acids. J Natl Cancer Inst. 1986;77(5):1053–62.

    PubMed  Google Scholar 

  27. Corsetto PA, Montorfano G, Zava S, Jovenitti IE, Cremona A, Berra B, Rizzo AM. Effects of n-3 PUFAs on breast cancer cells through their incorporation in plasma membrane. Lipids Health Dis. 2011;10:73–89.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. McEntee MF, Ziegler C, Reel D, Tomer K, Shoieb A, Ray M, Li X, Neilsen N, Lih FB, O’Rourke D, et al. Dietary n-3 polyunsaturated fatty acids enhance hormone ablation therapy in androgen therapy in androgen-dependent prostate cancer. Am J Pathol. 2008;173(1):229–41.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Altenburg JD, Siddiqui RA. Omega-3 polyunsaturated fatty acids down-modulate CXCR4 expression and function in MDA-MB-231 breast cancer cells. Mol Cancer Res. 2009;7(7):1013–20.

    Article  CAS  PubMed  Google Scholar 

  30. Berquin IM, Edwards IJ, Chen YQ. Multi-targeted therapy of cancer by omega-3 fatty acids. Cancer Lett. 2008;269(2):363–77.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Davidson LA, Nguyen DV, Hokanson RM, Callaway ES, Isett RB, Turner ND, Dougherty ER, Wang N, Lupton JR, Carroll RJ, et al. Chemopreventive n-3 polyunsaturated fatty acids reprogram genetic signatures during colon cancer initiation and progression in the rat. Cancer Res. 2004;64(18):6797–804.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Song EA, Kim H. Docosahexaenoic acid induces oxidative DNA damage and apoptosis, and enhances the chemosensitivity of cancer cells. Int J Mol Sci. 2016;17(8). https://doi.org/10.3390/ijms17081257.

    Article  PubMed Central  CAS  Google Scholar 

  33. Camargo CQ, Brunetta HS, Nunes EA. Effects of cotreatment with omega-3 polyunsaturated fatty acids and anticancer agents on oxidative stress parameters: a systematic review of in vitro, animal, and human studies. Nutr Rev. 2018;76(10):765–77.

    Article  PubMed  Google Scholar 

  34. Arora A, Scholar EM. Role of tyrosine kinase inhibitors in cancer therapy. J Pharmacol Exp Ther. 2005;315(3):971–9.

    Article  CAS  PubMed  Google Scholar 

  35. Araujo JC, Trudel GC, Saad F, Armstrong AJ, Yu EY, Bellmunt J, Wilding G, McCaffrey J, Serrano SV, Matveev VB, et al. Docetaxel and dasatinib or placebo in men with metastatic castration-resistant prostate cancer (READY): a randomized, double-blind phase 3 trial. Lancet Oncol. 2013;14(13):1307–16.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. D’Eliseo D, Velotti F. Omega-3 fatty acids and cancer cell cytotoxicity: implications for multi-targeted cancer therapy. J Clin Med. 2016;5(2). https://doi.org/10.3390/jcm5020015.

    Article  PubMed Central  CAS  Google Scholar 

  37. Menedez JA, Lupu R. Fatty acid synthase and the lipogenetic phenotype in cancer pathogenesis. Nat Rev Cancer. 2007;7(10):763–77.

    Article  CAS  Google Scholar 

  38. Avraham H, Park SY, Schinkmann K, Avraham S. RAFTK/Pyk2-mediated cellular signaling. Call Signal. 2000;12(3):123–33.

    Article  CAS  Google Scholar 

  39. Sun CK, Man K, Ng KT, Ho JW, Lim ZX, Cheng Q, Lo CM, Poon RT, Fan ST. Proline-rich tyrosine kinase 2 (Pyk2) promotes proliferation and invasiveness of hepatocellular carcinoma cells through c-Src/ERK activation. Carcinogenesis. 2008;29(11):2096–105.

    Article  CAS  PubMed  Google Scholar 

  40. Zhao M, Finlay D, Zharkikh I, Vuori K. Novel role of Src in priming Pyk2 phosphorylation. PLoS One. 2016;11(2):e0149231.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Perez J, Torres RA, Rocic P, Cismowski MJ, Weber DS, Darley-Usmar VM, Lucchesi PA. PYK2 signaling is required for PDGF-dependent vascular smooth muscle cell proliferation. Am J Physiol Cell Physiol. 2011;301(1):C242–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Matsuya M, Sasaki H, Aoto H, Mitaka T, Nagura K, Ohba T, Ishino M, Takahashi S, Suzuki R, Sasaki T. Cell adhesion kinase beta forms a complex with a new member, hic-, of proteins localized at focal adhesions. J Biol Chem. 1998;273(2):1003–14.

    Article  CAS  PubMed  Google Scholar 

  43. Murphy JM, Park H, Lim STS. FAK and Pyk2 in disease. Front Biol. 2016;11(1):1–9.

    Article  CAS  Google Scholar 

  44. Juan-Rivera MC, Martinez-Ferrer M. Integrin inhibitors in prostate cancer. Cancers (Basel). 2018;10(2). https://doi.org/10.3390/cancers10020044.

    Article  PubMed Central  CAS  Google Scholar 

  45. Lee BY, Trimpson P, Horvath LG, Daly RJ. FAK signaling in human cancer as a target for therapeutics. Pharmacol Ther. 2015;146:132–49.

    Article  CAS  PubMed  Google Scholar 

  46. Mills RD, Mita M, Walsh MP. A role for the Ca2+-dependent tyrosine kinase Pyk2 in tonic depolarization-induced vascular smooth muscle contraction. J Muscle Res Cell Motil. 2015;36(6):479–89.

    Article  CAS  PubMed  Google Scholar 

  47. Miller NL, Kleinschmidt EG, Schlaepfer DD. Rho GEFs in cell motility: novel links between Rgnef and focal adhesion kinase. Curr Mol Med. 2014;14(2):221–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Tornin J, Hermida-Prado F, Padda RS, Gonzalez MV, Alvarez-Fernandez C, Rey V, Martinez-Cruzado L, Estuinan O, Menendez ST, Fernandez-Nevado L, et al. FUS-CHOP promotes invasion in myxoid liposarcoma through a SRC/FAKRHO/ROCK-dependent pathway. Neoplasia. 2018;20(1):44–56.

    Article  CAS  PubMed  Google Scholar 

  49. Rozengurt E. Mitogenic signaling pathways induced by G protein-coupled receptors. J Cell Physiol. 2007;213(3):589–602.

    Article  CAS  PubMed  Google Scholar 

  50. Hirschler-Laszkiewica I, Chen SJ, Bao L, Wang J, Zhang XQ, Shanmughapriya S, Keefer K, Madesh M, Cheung JY, Miller BA. The human ion channel TRPM2 modulates neuroblastoma cell survival and mitochondrial function through Pyk2, CREB, and MCU activation. Am J Physiol Cell Physiol. 2018;315(4):C571–86.

    Article  CAS  Google Scholar 

  51. Jakobsen CH, Størvold GL, Bremseth H, Follestad T, Sand K, Mack M, Olsen KS, Lundemo AG, Iversen JG, Krokan HE, et al. DHA induces ER stress and growth arrest in human colon cancer cells: associations with cholesterol and calcium homeostasis. J Lipid Res. 2008;49(10):2089–100.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Hempel N, Trebak M. Crosstalk between calcium and reactive oxygen species signaling in cancer. Cell Calcium. 2017;63:70–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Yamamoto S, Shimazu S, Kiyonaka S, Takahashi N, Wajima T, Hara Y, Negoro T, Hiroi T, Kiuchi Y, Okada T, et al. TRPM2-mediated Ca2+ influx induces chemokine production in monocytes that aggravates inflammatory neutrophil infiltration. Nat Med. 2008;14(7):738.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgments

We would like to thank FORTE (https://www.forte-science.co.jp/) for their language review of the manuscript.

Adherence to national and international regulations

Not applicable.

Funding

This work was partly supported by JSPS KAKENHI (Grant Number: JP26350904).

Author information

Authors and Affiliations

Authors

Contributions

KO conceived and designed the study. KO performed the experiments. SS performed several experiments according to the reviewer’s comments. KO, CW and KK wrote the paper. KO, TS, KO and SS designed the figures and tables. KO, CW, TS and KK reviewed and edited the manuscript. All authors read and approved the manuscript and agree to be accountable for all aspects of the research in ensuring that the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Corresponding author

Correspondence to Keizo Kasono.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Oono, K., Ohtake, K., Watanabe, C. et al. Contribution of Pyk2 pathway and reactive oxygen species (ROS) to the anti-cancer effects of eicosapentaenoic acid (EPA) in PC3 prostate cancer cells. Lipids Health Dis 19, 15 (2020). https://doi.org/10.1186/s12944-019-1122-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12944-019-1122-4

Keywords