Mead acid inhibits the growth of KPL-1 human breast cancer cells in vitro and in vivo

  • Authors:
    • Yuichi Kinoshita
    • Katsuhiko Yoshizawa
    • Kei Hamazaki
    • Yuko Emoto
    • Takashi Yuri
    • Michiko Yuki
    • Nobuaki Shikata
    • Hiroshi Kawashima
    • Airo Tsubura
  • View Affiliations

  • Published online on: August 7, 2014     https://doi.org/10.3892/or.2014.3390
  • Pages: 1385-1394
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The effects of mead acid (MA; 5,8,11-eicosatrienoic acid) on the suppression of breast cancer cell growth and metastasis were examined in vitro and in vivo by using the KPL-1 human breast cancer cell line. MA suppressed KPL-1 cell growth in culture with an IC50 value of 214.2 µM (65.7 µg/ml) for 72 h, and MA significantly suppressed transplanted KPL-1 tumor growth (tumor volume and tumor weight: 872±103 mm3 and 1,000±116 mg vs. 376±66 mm3 and 517±84 mg) and regional (axillary) lymph node metastasis (67%, 10/15 vs. 10%, 1/10) in female athymic mice fed an MA-rich diet for 8 weeks. Tumor suppression was due to the suppression of cell proliferation. In ELISA, although vascular endothelial growth factor (VEGF) levels were unchanged, VEGF receptor (VEGFR)1 and VEGFR2 levels were significantly decreased after treatment with a 214.2-µM dose of MA for 72 h; E-cadherin levels were unchanged. As VEGF, VEGFR1 and VEGFR2 expression was co-localized in KPL-1 cells, the mechanism leading to cell growth suppression was VEGF signaling directly to KPL-1 cells by an autocrine process. In contrast, MA did not influence angiogenesis. The mechanisms of action were through VEGF signaling directly to cancer cells.

Introduction

Breast cancer is one of the most common types of cancer in industrial countries. Epidemiological studies indicate that the incidence and mortality of breast cancer is up to 5-fold higher in Western countries than in some Asian countries, and Asian migrants to the USA eventually acquire the breast cancer incidence of their host country, suggesting the importance of environmental factors (1). Among the environmental factors, it is clear that dietary exposure influences breast cancer risk, and particularly dietary intake of fat may play an important role in the genesis of breast cancer (25). Not only the amount of fat consumption, but also the type of dietary fat may have different effects on breast carcinogenesis. Long-chain polyunsaturated fatty acids (PUFAs) have different effects on mammary tumorigenesis based on the double bond position (68). The first double bond located at the 3rd, 6th or 9th carbon from the terminal methyl group of a fatty acid is called an n-3, n-6 or n-9 series fatty acid, respectively. Epidemiological studies and preclinical data indicate a positive association between dietary n-6 PUFA and breast cancer risk, while n-3 PUFA possesses chemopreventive properties (9,10). n-3 PUFAs such as eicosapentaenoic acid (EPA; 20:5n-3) suppress human breast cancer cell growth (11). Among n-3 PUFAs, docosahexaenoic acid (DHA; 22:6n-2) suppresses mammary carcinogenesis more effectively than EPA (7). In contrast, n-6 PUFAs such as linoleic acid (LA; 18:2n-6) and arachidonic acid (AA; 20:4n-6) promote the growth of human breast cancer cells (11,12). The typical American high-fat diet contains high levels of LA (13). Olive oil rich in oleic acid (OA; 18:1n-9) suppresses mammary carcinogenesis (14). However, it is difficult to determine if the effects of OA on experimental carcinogenesis are due to specific biochemical properties of OA or simply due to the substitution for n-6 PUFA (15). The role of n-9 PUFA in relation to breast cancer has not been studied in detail.

Mead acid (MA; also referred to as 5,8,11-eicosatrienoic acid), which was first characterized by James F. Mead, is a carboxylic acid with a 20-carbon chain and three methylene-interrupted cis double bonds, in which the first double bond is located at the ninth carbon from the terminal methyl group of a fatty acid (20:3n-9; Fig. 1). MA is a minor constituent of plasma and tissue in adult mammals. Elongation and desaturation of OA take place to form MA when n-6 and n-3 essential fatty acids, particularly LA, are deficient. Therefore, MA elevation in the blood is an indication of essential fatty acid deficiency. MA is found in large quantities in cartilage; MA decreases osteoblastic activity for the maintenance of cartilage to prevent ossification and suppresses angiogenesis to maintain avascular status (16,17). Angiogenesis plays an important role in the growth of breast cancer (18), and anti-angiogenic agents [inhibitors of vascular endothelial growth factor (VEGF)] and the VEGF receptor (VEGFR) may be promising targets for breast cancer control (19,20). The loss of cell adhesion is related to cancer invasion and metastasis. MA is related to the expression of the cell-cell adhesion molecule E-cadherin in human cancer cell lines including breast cancer cells (21,22). E-cadherin-mediated signaling can influence invasive and metastatic behavior (23,24). VEGF and/or E-cadherin signaling may modulate breast cancer growth at the primary site, and invasion and metastasis in the MA-rich condition. In addition, leukotriene B4 (LTB4) enhances tumor growth in human cancer cells including breast cancer cells (25,26). Dietary supplementation with MA suppresses LTB4 in rats (27,28). Moreover, a nested case-control study revealed an inverse association between MA and breast cancer risk as well as overall cancer risk (29); in the present study, neither the n-6/n-3 ratio nor AA intake correlated with breast cancer risk. Collectively, MA may exert cancer preventive properties. However, MA shows different effects on different types of cancer cells (21,22). The present study was designed to explore the effect of MA on KPL-1 human breast cancer cell growth and metastasis. The mechanisms of action were investigated based on VEGF and E-cadherin signaling and the modulation of fatty acid composition.

Materials and methods

Cell line

KPL-1 is a human breast cancer cell line established from the malignant effusion of a breast cancer patient (30). This cell line was derived from a patient with recurrent breast cancer that appeared during postsurgical adjuvant chemoendocrine therapy including tamoxifen and medroxy-progesterone acetate. The KPL-1 cell line is estrogen receptor (ER)-positive, progesterone receptor (PgR)-negative, and human epidermal growth factor receptor-2 (HER2)-negative (luminal A subtype) (31). Xenograft athymic mice injected with KPL-1 cells often develop lymph node metastasis (30). KPL-1 cells were cultured in Dulbecco’s modified Eagle’s medium (DMEM; Sigma, St. Louis, MO, USA) with 10% fetal bovine serum (FBS; Gibco-BRL, Grand Island, NY, USA) in 5% CO2/95% humidified air at 37°C.

MA preparation

MA used for in vitro experiments was purchased from Sigma and dissolved to 5 mg/500 μl (32.6 mM) in ethanol and stored at −30°C. MA was diluted in DMEM containing 10% FBS to achieve final concentrations of 16.25, 32.5, 75, 150 and 300 μM.

MTT assay

KPL-1 cells were seeded at 2×103 cells/well in 96-well plates in DMEM with 10% FBS. The cells were treated with ethanol (final concentration, 0.9%) or the indicated concentration of MA for up to 72 h. Cell proliferation was monitored by the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay as previously described (32), and IC50 value was calculated.

Animal experiments

Animals were housed in groups of four or five in plastic cages with paper bedding (Paper Clean, SLC, Hamamatsu, Japan) in a specific pathogen-free room maintained at 22±2°C and 60±10% relative humidity with a 12-h light/dark cycle (lights on at 8:00 AM and lights off at 8:00 PM). The mice were randomly divided into two groups, the control diet group (n=15) and the MA diet group (n=10). Both experimental diets were modifications of the AIN-76 diet (33) and contained the same amount of nutrients but had different fatty acid compositions (Table I). The MA diet contained 5% SUNTGM33 (a kind gift from Suntory Wellness, Tokyo, Japan), which contains 48.0% MA (Table II). SUNTGM33 is a microbial oil obtained by fungal fermentation (34). Olive oil purchased from Nacalai Tesque (Kyoto, Japan) was used for the control diet. The fatty acid compositions of SUNTGM33 and olive oil are listed in Table II. Each experimental diet was formulated by Oriental Yeast (Tokyo, Japan). Four-week old female athymic BALB/c mice purchased from Charles River Japan (Kyoto, Japan) were fed either a control or MA diet starting at 4 weeks of age. In comparison to the control diet, the MA diet for 8 days starting at 4 weeks of age did not cause weight gain. Therefore, the MA diet group was switched to the control diet for 4 successive days to accelerate weight gain; thereafter, the MA diet group was treated with a cyclic feeding regime of 5 days on the MA diet followed by 2 days on the control diet throughout the experiment. The control group was treated with the control diet throughout the experiment. Schematic representation of the in vivo experiment is shown in Fig. 2. At 6 weeks of age, both groups were inoculated with 2.5×105 KPL-1 cells in 100 μl DMEM supplemented with 10% FBS into the thoracic mammary fat pad (MA diet group was on the third day of MA diet). During the experiment, the dose of diet ingested, body weight and locally growing tumor volumes were measured once per week. Tumor volume was calculated by using the standard formula: width2 × length × 0.5. Thirty-seven days after tumor cell inoculation (fifth day on MA diet), body weight was measured, and bromodeoxyuridine (BrdU; Invitrogen, Camarillo, CA, USA) was injected (50 mg/kg animal body weight) via the abdominal cavity, blood was sampled and then animals were sacrificed by exsanguination from aortic transection. At autopsy, all organs were examined macroscopically, and the primary tumors and local axillary lymph nodes were examined histologically; half of the primary tumor was snap frozen and used for fatty acid analysis. Tissues were fixed in 10% neutral buffered formalin, embedded in paraffin, and stained with hematoxylin and eosin (H&E). The study protocol and animal procedures were approved by the Animal Care and Use Committee of Kansai Medical University (permit no. 13-060). Throughout the experiments, animals were cared for in accordance with the Guidelines for Animal Experimentation of Kansai Medical University.

Table I

Composition of experimental diets.

Table I

Composition of experimental diets.

MA dietControl diet
Casein2020
DL-methionine0.30.3
Cornstarch4343
α-cornstarch1212
Sucrose1010
Cellulose55
AIN-76 mineral mix3.53.5
AIN-76 vitamin mix11
Choline bitartrate0.20.2
SUNTGM3350
Olive oil05

[i] Values are expressed in g/100 g diet. MA, mead acid.

Table II

Fatty acid composition of SUNTGM33 and olive oil.

Table II

Fatty acid composition of SUNTGM33 and olive oil.

Fatty acid composition (%)SUNTGM33Olive oil
Myristic acid (14:0)0.120.00
Palmitic acid (16:0)3.3510.88
Palmitoleic (16:1n-7)0.000.85
Stearic acid (18:0)4.963.32
Oleic acid (18:1n-9)25.1074.71
Vaccenic acid (18:1n-7)0.141.89
Linoleic acid (18:2n-6)0.426.90
α-Linolenic acid (18:3n-3)0.000.68
Arachidic acid (20:0)0.000.46
Gondoic acid (20:1n-9)4.950.31
Mead acid (20:3n-9)48.020.00
Arachidonic acid (20:4n-6)1.230.00
Eicosapentaenoic acid (20:5n-3)0.470.00
Behenic acid (22:0)2.150.00
Lignoceric acid (24:0)5.340.00
Nervonic acid (24:1n-9)2.420.00
Total n-980.4975.02
Total n-30.470.68
Total n-61.656.90
n-6/n-33.5310.19
Total SFA15.9114.66
Total MUFA32.6177.76
Total PUFA50.157.58

[i] SFA, saturated fatty acid; MUFA, monounsaturated fatty acid; PUFA, polyunsaturated fatty acid.

Fatty acid analysis

The fatty acid composition of the total phospholipid fraction of serum was determined. Total lipids were extracted by the method of Bligh and Dyer (35). The total phospholipid fraction was separated by thin-layer chromatography. For an internal standard, 1,2-diheptadecanoyl- sn-glycero-3-phosphocholine (Avanti Polar Lipids, Inc., Alabaster, AL, USA) was added. Total phospholipid fractions were transmethylated with HCl-methanol and then the fatty acid composition was analyzed by gas chromatography (GC-2014; Shimadzu Corporation, Kyoto, Japan) with a capillary column DB-225 (0.25 mm × 30 m × 0.25 μM; J&M Scientific, Folsom, CA, USA). The entire system was controlled with gas chromatography software (GCsolution; Shimadzu Corporation). The fatty acid composition of the total lipid fraction of KPL-1 tumors was determined. In brief, frozen tumor tissues were thawed, minced and homogenized three times in 8 ml chloroform-methanol (2:1) by a polytron homogenizer (Kinematica, Lucerne, Switzerland) for 10 sec. The fatty acid analysis of total lipids in the tumor was performed by the same method as previously mentioned (35,36).

Microvessel density and cell kinetics

Microvessel density of primary KPL-1 tumors was evaluated by anti-CD34 antiserum (Abcam, Cambridge, UK). The cell kinetics (cell proliferation and cell death) in primary KPL-1 tumors were evaluated. Cell proliferation was evaluated by anti-BrdU antibody (clone B44, 1:50; Becton-Dickinson, Franklin Lakes, NJ, USA) by using an LSAB staining kit (Dako, Glostrup, Denmark). Cell death was evaluated by anti-phospho-histone H2A.X (γ-H2AX) antibody (Ser139, 1:100; Cell Signaling, Danvers, MA, USA), an immunomarker of the DNA damage response. Each slide was scanned with a high-resolution digital slide scanner (NanoZoomer 2.0 Digital Pathology; Hamamatsu Photonics, Hamamatsu, Japan) to prepare digital images. The ndpi image files were opened in color mode with NDP.view software (Hamamatsu Photonics). The images were changed to jpeg files at ×40 magnification in five randomly selected areas within each tumor that were used to analyze immunohistochemical staining (3739).

CD34, VEGF, VEGFR1, VEGFR2 and E-cadherin immunohistochemistry

Cell blocks were prepared from KPL-1 cells cultured with or without 214.2 μM MA for 72 h (IC50 value for 72 h). Cell blocks were prepared from cultured KPL-1 cells; cells were centrifuged at 1,000 rpm for 5 min, and cell pellets were fixed in 10% neutral buffer formalin and embedded in paraffin. Antisera used to detect CD34, VEGF, VEGFR1, VEGFR2 and E-cadherin were anti-CD34 antiserum (1:20; Abcam), anti-VEGF antiserum (1:50), anti-VEGFR1 antiserum (1:500) (both from Santa Cruz Biotechnology, Santa Cruz, CA, USA) and anti-VEGFR2 antiserum (1:400) (Cell Signaling Technology, Danvers, MA, USA), respectively, by using an LSAB staining kit (Dako), and anti-E-cadherin antibody (NCH38 ready to use) by using Histofine MAX-PO (both from Nichirei Biosciences, Tokyo, Japan) according to the manufacturer’s instructions. The reaction products were visualized with 3-3′-diaminobenzidine tetrahydrochloride. CD34 immunohistochemistry was applied for angiogenesis evaluation in KPL-1 tumor grown in athymic mice, and VEGF, VEGFR1, VEGFR2 and E-cadherin immunohistochemistry, respectively, were applied in cultured KPL-1 cells in cell pellets.

Quantitation of VEGF, VEGFR1, VEGFR2 and E-cadherin

KPL-1 cells cultured with or without 214.2 μM MA for 72 h (IC50 value for 72 h) were sampled. VEGF was quantified in the culture supernatant, and VEGFR1, VEGFR2 and E-cadherin were measured in the cell lysate. For cell lysate preparation, after washing cells with PBS (−), cell pellets were homogenized with RIPA buffer (Wako, Osaka, Japan), and the cell lysate was centrifuged at 14,000 rpm for 15 min at 4°C to obtain supernatant containing cell lysate. Protein concentrations were measured by the DC protein assay kit (Bio-Rad, Hercules, CA, USA). VEGF, VEGFR1, VEGFR2 and E-cadherin levels were measured by enzyme-linked immunoabsorbent assay (ELISA) with a human VEGF assay kit (IL-226; IBL, Fujioka, Japan), VEGFR1 human ELISA kit (ab119613), VEGFR2 human ELISA kit (ab100665) (both from Abcam), and E-cadherin human ELISA kit (R&D Systems, Minneapolis, MN, USA), respectively, according to the manufacturers’ protocols. Each protein standard or protein purified from each group was incubated overnight at 4°C against VEGF or E-cadherin; 90 min at 37°C against VEGFR1; or 150 min at room temperature against VEGFR2. After the final colors were developed with the addition of each color reagent, absorbance was measured at 450 nm. Cell sample concentration was calculated from a standard curve and corrected for protein concentration. The limits for detection for VEGF, VEGFR1, VEGFR2 and E-cadherin were 8, 156, 70 and 39 pg/ml, respectively.

Statistical analysis

Values are expressed as the means ± standard error of the mean (SEM). Body weight, tumor volume, tumor weight, fatty acid composition, CD34-positive area and the number of BrdU- and γ-H2AX- positive cells per 1 mm2 among the groups were analyzed by t-test. The incidence of metastasis was analyzed with the χ2 test.

Results

KPL-1 cell growth inhibition in vitro

The KPL-1 cells were treated with 5 concentrations (16.25–300 μM) of MA for up to 72 h. The MTT assay revealed that MA induced growth inhibition in a dose- and time-dependent manner (Fig. 3). The IC50 value of MA against KPL-1 cells was 214.2 μM (65.7 μg/ml) for a 72-h treatment.

Host animals

During the experiment, the daily dose of MA ingestion was 63–80 mg/mouse/day (mean, 74 mg/mouse/day). Body weight gain in the MA group was significantly smaller during the experiment but at the end of the experiment (eating the respective diet for 8 weeks), the difference in body weight between the MA and control diet group was not statistically significant (Fig. 4). No organs or tissues were macroscopically abnormal. Throughout the experiment, the MA diet group ingested less food than the control diet group (average, 92.1%).

Primary KPL-1 tumor growth and metastasis

Locally growing KPL-1 tumors in the MA diet group grew slower than in the control diet group. The final tumor volume was significantly smaller (Fig. 5A) and the final tumor weight was significantly lighter (Fig. 5B) in the MA diet group as compared to the control diet group (p<0.01 and p<0.05, respectively). Tumor volume two days before the termination of the experiment was 872±103 and 376±66 mm3, and the final tumor weight was 1,000±116 and 517±84 mg in the control and MA group, respectively. Regional (axillary) lymph node metastasis was found in both groups (Fig. 5C). Similarly, as compared with the control diet group, metastasis was significantly suppressed in the MA diet group (66.7%: 10/15 vs. 10%: 1/10).

Morphology and angiogenesis of KPL-1 tumor

Although the growth of KPL-1 tumors was suppressed in the MA diet group, the morphology was comparable in both groups (data not shown). The angiogenesis of KPL-1 tumors at the primary site (Fig. 6A) as well as the CD34-stained area was compatible between the control and MA diet groups (Fig. 6B).

Proliferation and apoptotic ratio of KPL-1 tumor

To compare the cell kinetics of KPL-1 tumors (cell proliferation and cell death), the number of BrdU-positive cells and γ-H2AX-positive cells per 1,000 cells in primary tumors from the control and MA diet groups of mice was compared. The proliferation and apoptotic ratios are shown in Fig. 7A and B, respectively. The proliferation ratio in the control and MA diet groups was 11.8±0.7 and 8.7±0.9%, respectively (p<0.05), while the apoptotic ratio in the control and MA diet groups was 8.7±1.3 and 9.6±1.2% (not statistically significant).

Fatty acid analysis

Fatty acid composition of sera and KPL-1 tumors in the control and MA diet group is shown in Fig. 8A and B, respectively. In the sera, the concentration of MA was significantly higher in the MA diet group as compared to controls (237.2±17.6 vs. 23.8±1.1 μg/ml). In contrast, LA and AA were significantly lower in the MA diet group as compared with controls. In the KPL-1 tumor, the fatty acid profile was generally similar to that of the fatty acid composition in the serum. However, the serum OA composition was significantly higher in the control diet group, as MA was replaced by OA in this diet, while the OA concentration in KPL-1 tumors was comparable. The MA diet significantly decreased the n-6/n-3 ratio in the sera and the tumors (Fig. 9).

Expression of VEGF, VEGFR1, VEGFR2 and E-cadherin in KPL-1 cells in culture

KPL-1 cells strongly expressed VEGF, VEGFR1 and VEGFR2 in the cytoplasm and E-cadherin at the cell surface. Although VEGF expression was unchanged, VEGFR1 and VEGFR2 expression tended to diminish and E-cadherin expression tended to increase in KPL-1 cells treated with the IC50 dose of MA for 72 h (214.2 μM) (Fig. 10).

Quantification of VEGF, VEGFR1, VEGFR2 and E-cadherin levels in KPL-1 cells in culture

Levels of VEGF, VEGFR1, VEGFR2 and E-cadherin in KPL-1 cells cultured with or without the IC50 dose of MA for 72 h were compared. Although the VEGF levels in KPL-1 tumor cells were similar (Fig. 11A), MA treatment significantly decreased VEGFR1 and VEGFR2 levels and tended to increase E-cadherin levels (Fig. 11B–D).

Discussion

In the present study, MA suppressed KPL-1 human breast cancer cell growth in culture with an IC50 value of 214.2 μM (65.7 μg/ml) for 72 h and significantly suppressed KPL-1 tumor growth and regional (axillary) lymph node metastasis in female athymic mice. Tumor suppression was due to decreased cell proliferation. The body weight of the MA diet group, although it was similar to the control diet-fed mice at the termination of the present study, was significantly lighter during the experimental period; the dose of food intake was less in the MA-fed mice. Thus, the lighter body weight in the MA diet group may be due to consuming a low amount of the MA diet. The serum MA level in MA diet-fed mice was 237.2 μg/ml, which is higher than the IC50 value. Thus, the IC50 dose of MA used in the in vitro experiments is achievable and may not cause serious side-effects in female mice. In contrast to n-3 PUFA, which invariably exerts antiproliferation action on human tumor cell lines, the n-9 series of MA causes different actions on different human tumor cells, depending on their origin and cellular types (21,22). MA may function in a cell-specific manner. However, in agreement with MCF-7 human breast cancer cells, the MA diet suppressed KPL-1 tumor growth and metastasis in female athymic mice. Tumor growth is a balance between cell proliferation and cell death. Mammary tumors of the VEGF knockout mice exhibit cell-cycle arrest and induction of apoptosis (37). With the present dose of MA, the MA diet suppressed KPL-1 tumor growth cytostatically by diminishing cell proliferation; however, it was below the dose needed to cause apoptosis.

Tumor angiogenesis is closely related to the growth of breast cancer, and VEGF and its receptors are essential for breast cancer growth (1820). Cartilage is an avascular tissue and contains high levels of MA; MA dose-dependently inhibits VEGF-stimulated angiogenesis (17). However, MA did not alter angiogenesis as evaluated by microvessel density in KPL-1 tumors in athymic female mice. Although VEGF is well known for its key roles in blood vessel growth, it also promotes a range of other functions, such as cell adhesion, survival, migration and invasion (40). VEGF, VEGFR1 and VEGFR2 were immunohistochemically detected in KPL-1 cells. The presence of VEGFR1 and VEGFR2 as well as VEGF on KPL-1 cells may raise the possibility that VEGF may promote tumor growth not only by inducing angiogenesis but directly through the activation of VEGFR1 and VEGFR2 (4143). In the present study, VEGF levels in cultured KPL-1 cells treated with MA were comparable to the levels in KPL-1 cells without MA treatment. However, VEGFR1 and VEGFR2 levels were significantly decreased in KPL-1 cells treated with MA. As VEGF, VEGFR1 and VEGFR2 were co-localized in KPL-1 cells, the present results suggest that VEGF signaling did not modulate angiogenesis, however it directly modulated the growth of VEGFR-positive tumor cells via an autocrine process, an endothelial cell-independent pathway (44). Expression of VEGF and its receptors (VEGF1 and VEGFR2) is associated with poor outcome in breast cancer patients (45). VEGF knockout directly decreases the proliferation of breast cancer cells (37).

Downregulation of E-cadherin is associated with loss of cellular adhesiveness and initiates metastatic outgrowth resulting in poor prognosis (46,47). Downregulation of E-cadherin is required to initiate metastatic outgrowth of breast cancer (47). VEGF increases transcription factor Snail, which is associated with breast cancer metastasis and reduces E-cadherin expression in breast cancer cells (48). Although MA tended to increase E-cadherin levels, the difference did not reach statistical significance. VEGF increases the cellular invasion of T-47D breast cancer cells on Matrigel/fibronectin-coated transwell membranes (41). Therefore, VEGF, but not E-cadherin, may contribute to the mechanisms of suppression of invasion and metastasis in the KPL-1 tumor system.

In addition to affecting the VEGF pathway, MA may function via other unknown mechanisms. In a human case-control study of breast fat tissues, increased n-6/n-3 PUFA ratios and decreased n-3 PUFA levels were found in breast cancer patients as compared with controls (49,50). Alteration in the n-6/n-3 PUFA ratio appears to be important (51) and n-3 PUFA, namely EPA and DHA (4) or n-6 PUFA, such as LA and AA (11,12), is important. The MA diet-fed mice had significantly decreased LA and AA levels, and the n-6/n-3 PUFA ratio was significantly decreased in both the sera and the KPL-1 tumors, which may lead to growth suppression of KPL-1 cells. The essential fatty acid LA cannot be synthesized in the body and must be derived from the diet. Dietary LA shows a negative effect on the incorporation of dietary MA into the plasma lipid fraction and membrane phospholipids (52). LA is converted via intermediates to dihomo-γ-linolenic acid and to AA. Thus, an inverse relationship between n-9 PUFA MA and n-6 PUFA LA and AA may exist. In the present MA diet, decreased expression of n-6 PUFA, namely LA and AA, in the MA-fed mice, may suppress breast cancer cell proliferation and invasion (4). However, n-6 content in the control diet was considerably below 4%; n-6 PUFA (LA) at a dose of ~4% is required for maximal acceleration of mammary tumorigenesis (53).

In conclusion, MA effectively suppressed the growth and metastasis of KPL-1 human breast cancer cells via VEGF signaling.

Acknowledgements

The authors thank Ms. S Takebe (University of Toyama) for her technical assistance and Ms. A Shudo (Kansai Medical University) for manuscript preparation. This study was supported in part by a Grant-in-Aid for Scientific Research (C) (24592611) from the Ministry of Education, Culture, Sports, Science and Technology of Japan, and by a grant from the Kiyoko Wada Foundation of the Kansai Medical University Alumni Association (2012).

References

1 

Adami HO, Signorello LB and Trichopoulos D: Towards an understanding of breast cancer etiology. Semin Cancer Biol. 8:255–262. 1998. View Article : Google Scholar : PubMed/NCBI

2 

Tsubura A, Uehara N, Kiyozuka Y and Shikata N: Dietary factors modifying breast cancer risk and relation to time of intake. J Mammary Gland Biol Neoplasia. 10:87–100. 2005. View Article : Google Scholar : PubMed/NCBI

3 

Tsubura A, Yuri T, Yoshizawa K, Uehara N and Takada H: Role of fatty acids in malignancy and visual impairment: epidemiological evidence and experimental studies. Histol Histopathol. 24:223–234. 2009.PubMed/NCBI

4 

Chénais B and Blanckaert V: The Janus face of lipids in human breast cancer: how polyunsaturated fatty acids affect tumor cell hallmarks. Int J Breast Cancer. 2012:7125362012.PubMed/NCBI

5 

de Lorgeril M and Salen P: New insights into the health effects of dietary saturated and omega-6 and omega-3 polyunsaturated fatty acids. BMC Med. 10:502012.PubMed/NCBI

6 

Bartsch H, Nair J and Owen RW: Dietary polyunsaturated fatty acids and cancers of the breast and colorectum: emerging evidence for their role as risk modifiers. Carcinogenesis. 20:2209–2218. 1999. View Article : Google Scholar : PubMed/NCBI

7 

Yuri T, Danbara N, Tsujita-Kyutoku M, Fukunaga K, Takada H, Inoue Y, Hada T and Tsubura A: Dietary docosahexaenoic acid suppresses N-methyl-N-nitrosourea-induced mammary carcinogenesis in rats more effectively than eicosapentaenoic acid. Nutr Cancer. 45:211–217. 2003. View Article : Google Scholar : PubMed/NCBI

8 

Wen ZH, Su YC, Lai PL, Zhang Y, Xu YF, Zhao A, Yao GY, Jia CH, Lin J, Xu S, Wang L, Wang XK, Liu AL, Jiang Y, Dai YF and Bai XC: Critical role of arachidonic acid-activated mTOR signaling in breast carcinogenesis and angiogenesis. Oncogene. 32:160–170. 2013. View Article : Google Scholar : PubMed/NCBI

9 

Patterson RE, Flatt SW, Newman VA, Natarajan L, Rock CL, Thomson CA, Caan BJ, Parker BA and Pierce JP: Marine fatty acid intake is associated with breast cancer prognosis. J Nutr. 141:201–206. 2011. View Article : Google Scholar : PubMed/NCBI

10 

Signori C, El-Bayoumy K, Russo J, Thompson HJ, Richie JP, Hartman TJ and Manni A: Chemoprevention of breast cancer by fish oil in preclinical models: trials and tribulations. Cancer Res. 71:6091–6096. 2011. View Article : Google Scholar : PubMed/NCBI

11 

Senzaki H, Iwamoto S, Ogura E, Kiyozuka Y, Arita S, Kurebayashi J, Takada H, Hioki K and Tsubura A: Dietary effects of fatty acids on growth and metastasis of KPL-1 human breast cancer cells in vivo and in vitro. Anticancer Res. 18:1621–1627. 1998.PubMed/NCBI

12 

Chang NW, Wu CT, Chen DR, Yeh CY and Lin C: High levels of arachidonic acid and peroxisome proliferator-activated receptor-alpha in breast cancer tissues are associated with promoting cancer cell proliferation. J Nutr Biochem. 24:274–281. 2013. View Article : Google Scholar

13 

Taioli E, Nicolosi A and Wynder EL: Dietary habits and breast cancer: a comparative study of United States and Italian data. Nutr Cancer. 16:259–265. 1991. View Article : Google Scholar : PubMed/NCBI

14 

Cohen LA, Epstein M, Pittman B and Rivenson A: The influence of different varieties of olive oil on N-methylnitrosourea (NMU)-induced mammary tumorigenesis. Anticancer Res. 20:2307–2312. 2000.PubMed/NCBI

15 

Rose DP: Dietary fatty acids and cancer. Am J Clin Nutr. 66(Suppl 4): 998S–1003S. 1997.PubMed/NCBI

16 

Hamazaki T, Suzuki N, Widyowati R, Miyahara T, Kadota S, Ochiai H and Hamazaki K: The depressive effects of 5,8,11-eicosatrienoic acid (20:3n-9) on osteoblasts. Lipids. 44:97–102. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Hamazaki T, Nagasawa T, Hamazaki K and Itomura M: Inhibitory effect of 5,8,11-eicosatrienoic acid on angiogenesis. Prostaglandins Leukot Essent Fatty Acids. 86:221–224. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Hicklin DJ and Ellis LM: Role of the vascular endothelial growth factor pathway in tumor growth and angiogenesis. J Clin Oncol. 23:1011–1027. 2005. View Article : Google Scholar : PubMed/NCBI

19 

Fox SB, Generali DG and Harris AL: Breast tumour angiogenesis. Breast Cancer Res. 9:2162007. View Article : Google Scholar

20 

Sharma PS, Sharma R and Tyagi T: VEGF/VEGFR pathway inhibitors as anti-angiogenic agents: present and future. Curr Cancer Drug Targets. 11:624–653. 2011. View Article : Google Scholar : PubMed/NCBI

21 

Eynard AR, Jiang WG and Mansel RE: Eicosatrienoic acid (20:3 n-9) inhibits the expression of E-cadherin and desmoglein in human squamous cell carcinoma in vitro. Prostaglandins Leukot Essent Fatty Acids. 59:371–377. 1998. View Article : Google Scholar : PubMed/NCBI

22 

Heyd VL and Eynard AR: Effects of eicosatrienoic acid (20:3 n-9, Mead’s acid) on some promalignant-related properties of three human cancer cell lines. Prostaglandins Other Lipid Mediat. 71:177–188. 2003.

23 

Jones J and Walker R: Cell-cell and cell-stromal interactions in breast cancer invasion and metastasis. Int J Oncol. 11:609–616. 1997.PubMed/NCBI

24 

Scully OJ, Bay BH, Yip G and Yu Y: Breast cancer metastasis. Cancer Genomics Proteomics. 9:311–320. 2012.

25 

Pidgeon GP, Lysaght J, Krishnamoorthy S, Reynolds JV, O’Byrne K, Nie D and Honn KV: Lipoxygenase metabolism: roles in tumor progression and survival. Cancer Metastasis Rev. 26:503–524. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Hu N, Li Y, Zhao Y, Wang Q, You JC, Zhang XD and Ye LH: A novel positive feedback loop involving FASN/p-ERK1/2/5-LOX/LTB4/FASN sustains high growth of breast cancer cells. Acta Pharmacol Sin. 32:921–929. 2011. View Article : Google Scholar : PubMed/NCBI

27 

James MJ, Gibson RA, Neumann MA and Cleland LG: Effect of dietary supplementation with n-9 eicosatrienoic acid on leukotriene B4 synthesis in rats: a novel approach to inhibition of eicosanoid synthesis. J Exp Med. 178:2261–2265. 1993. View Article : Google Scholar : PubMed/NCBI

28 

Cleland LG, Gibson RA, Neumann MA, Hamazaki T, Akimoto K and James MJ: Dietary (n-9) eicosatrienoic acid from a cultured fungus inhibits leukotriene B4 synthesis in rats and the effect is modified by dietary linoleic acid. J Nutr. 126:1534–1540. 1996.PubMed/NCBI

29 

Pouchieu C, Chajès V, Laporte F, Kesse-Guyot E, Galan P, Hercberg S, Latino-Martel P and Touvier M: Prospective associations between plasma saturated, monounsaturated and polyunsaturated fatty acids and overall and breast cancer risk - modulation by antioxidants: a nested case-control study. PLoS One. 9:e904422014. View Article : Google Scholar

30 

Kurebayashi J, Kurosumi M and Sonoo H: A new human breast cancer cell line, KPL-1 secretes tumour-associated antigens and grows rapidly in female athymic nude mice. Brit J Cancer. 71:845–853. 1995. View Article : Google Scholar : PubMed/NCBI

31 

Kurebayashi J, Kanomata N, Moriya T, Kozuka Y, Watanabe M and Sonoo H: Preferential antitumor effect of the Src inhibitor dasatinib associated with a decreased proportion of aldehyde dehydrogenase 1-positive cells in breast cancer cells of the basal B subtype. BMC Cancer. 10:5682010. View Article : Google Scholar

32 

Kanematsu S, Uehara N, Miki H, Yoshizawa K, Kawanaka A, Yuri T and Tsubura A: Autophagy inhibition enhances sulforaphane-induced apoptosis in human breast cancer cells. Anticancer Res. 30:3381–3390. 2010.PubMed/NCBI

33 

No authors listed. Report of the American Institute of Nutrition Ad Hoc Committee on Standards for Nutritional Studies. J Nutr. 107:1340–1348. 1977.PubMed/NCBI

34 

Sakuradani E, Kamada N, Hirano Y, Nishihara M, Kawashima H, Akimoto K, Higashiyama K, Ogawa J and Shimizu S: Production of 5,8,11-eicosatrienoic acid by a Δ5 and Δ6 desaturation activity-enhanced mutant derived from a Δ12 desaturation activity-defective mutant of Mortierella alpina 1S-4. Appl Microbiol Biotechnol. 60:281–287. 2002.

35 

Bligh EG and Dyer WJ: A rapid method of total lipid extraction and purification. Can J Biochem Physiol. 3:911–917. 1959. View Article : Google Scholar : PubMed/NCBI

36 

Lai YC, Hamazaki K, Yoshizawa K, Kawanaka A, Kuwata M, Kanematsu S, Hamazaki T, Takada H and Tsubura A: Short-term pregnancy hormone treatment of N-methyl-N-nitrosourea-induced mammary carcinogenesis in relation to fatty acid composition of serum phospholipids in female Lewis rats. In Vivo. 24:553–560. 2010.PubMed/NCBI

37 

Schoeffner DJ, Matheny SL, Akahane T, Factor V, Berry A, Merlino G and Thorgeirsson UP: VEGF contributes to mammary tumor growth in transgenic mice through paracrine and autocrine mechanisms. Lab Invest. 85:608–623. 2005. View Article : Google Scholar : PubMed/NCBI

38 

Kanematsu S, Yoshizawa K, Uehara N, Miki H, Sasaki T, Kuro M, Lai YC, Kimura A, Yuri T and Tsubura A: Sulforaphane inhibits the growth of KPL-1 human breast cancer cells in vitro and suppresses the growth and metastasis of orthotopically transplanted KPL-1 cells in female athymic mice. Oncol Rep. 26:603–608. 2011.PubMed/NCBI

39 

Redon CE, Weyemi U, Parekh PR, Huang D, Burrell AS and Bonner WM: γ-H2AX and other histone post-translational modifications in the clinic. Biochim Biophys Acta. 1819:743–756. 2012.

40 

Perrot-Applanat M and Di Benedetto M: Autocrine functions of VEGF in breast tumor cells: adhesion, survival, migration and invasion. Cell Adh Migr. 6:547–553. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Price DJ, Miralem T, Jiang S, Steinberg R and Avraham H: Role of vascular endothelial growth factor in the stimulation of cellular invasion and signaling of breast cancer cells. Cell Growth Differ. 12:129–135. 2001.PubMed/NCBI

42 

Weigand M, Hantel P, Kreienberg R and Waltenberger J: Autocrine vascular endothelial growth factor signalling in breast cancer. Evidence from cell lines and primary breast cancer cultures in vitro. Angiogenesis. 8:197–204. 2005. View Article : Google Scholar : PubMed/NCBI

43 

Wu Y, Hooper AT, Zhong Z, Witte L, Bohlen P, Rafii S and Hicklin DJ: The vascular endothelial growth factor receptor (VEGFR-1) supports growth and survival of human breast carcinoma. Int J Cancer. 119:1519–1529. 2006. View Article : Google Scholar : PubMed/NCBI

44 

Guo S, Colbert LS, Fuller M, Zhang Y and Gonzalez-Perez RR: Vascular endothelial growth factor receptor-2 in breast cancer. Biochim Biophys Acta. 1806:108–121. 2010.PubMed/NCBI

45 

Ghosh S, Sullivan CA, Zerkowski MP, Molinaro AM, Rimm DL, Camp RL and Chung GG: High levels of vascular endothelial growth factor and its receptors (VEGFR-1, VEGFR-2, neuropilin-1) are associated with worse outcome in breast cancer. Hum Pathol. 39:1835–1843. 2008. View Article : Google Scholar : PubMed/NCBI

46 

Howard EM, Lau SK, Lyles RH, Birdsong GG, Umbreit JN and Kochhar R: Expression of e-cadherin in high-risk breast cancer. J Cancer Res Clin Oncol. 131:14–18. 2005. View Article : Google Scholar : PubMed/NCBI

47 

Wendt MK, Taylor MA, Schiemann BJ and Schiemann WP: Downregulation of epithelial cadherin is required to initiate metastatic outgrowth of breast cancer. Mol Biol Cell. 22:2423–2435. 2011. View Article : Google Scholar : PubMed/NCBI

48 

Wanami LS, Chen HY, Peiró S, García de Herreros A and Bachelder RE: Vascular endothelial growth factor-A stimulates Snail expression in breast tumor cells: implications for tumor progression. Exp Cell Res. 314:2448–2453. 2008. View Article : Google Scholar : PubMed/NCBI

49 

Zhu ZR, Agren J, Männistö S, Pietinen P, Eskelinen M, Syrjänen K and Uusitupa M: Fatty acid composition of breast adipose tissue in breast cancer patients and in patients with benign breast disease. Nutr Cancer. 24:151–160. 1995. View Article : Google Scholar : PubMed/NCBI

50 

Maillard V, Bougnoux P, Ferrari P, Jourdan ML, Pinault M, Lavillonnière F, Body G, Le Floch O and Chajès V: N-3 and N-6 fatty acids in breast adipose tissue and relative risk of breast cancer in a case-control study in Tours, France. Int J Cancer. 98:78–83. 2002. View Article : Google Scholar : PubMed/NCBI

51 

Okuyama H, Kobayashi T and Watanabe S: Dietary fatty acids - the n-6/n-3 balance and chronic elderly diseases. Excess linoleic acid and relative n-3 deficiency syndrome seen in Japan. Prog Lipid Res. 35:409–457. 1996. View Article : Google Scholar : PubMed/NCBI

52 

Cleland LG, Neumann MA, Gibson RA, Hamazaki T, Akimoto K and James MJ: Effect of dietary n-9 eicosatrienoic acid on the fatty acid composition of plasma lipid fractions and tissue phospholipids. Lipids. 31:829–837. 1996. View Article : Google Scholar : PubMed/NCBI

53 

Ip C, Carter CA and Ip MM: Requirement of essential fatty acid for mammary tumorigenesis in the rat. Cancer Res. 45:1997–2001. 1985.PubMed/NCBI

Related Articles

Journal Cover

October 2014
Volume 32 Issue 4

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kinoshita Y, Yoshizawa K, Hamazaki K, Emoto Y, Yuri T, Yuki M, Shikata N, Kawashima H and Tsubura A: Mead acid inhibits the growth of KPL-1 human breast cancer cells in vitro and in vivo. Oncol Rep 32: 1385-1394, 2014
APA
Kinoshita, Y., Yoshizawa, K., Hamazaki, K., Emoto, Y., Yuri, T., Yuki, M. ... Tsubura, A. (2014). Mead acid inhibits the growth of KPL-1 human breast cancer cells in vitro and in vivo. Oncology Reports, 32, 1385-1394. https://doi.org/10.3892/or.2014.3390
MLA
Kinoshita, Y., Yoshizawa, K., Hamazaki, K., Emoto, Y., Yuri, T., Yuki, M., Shikata, N., Kawashima, H., Tsubura, A."Mead acid inhibits the growth of KPL-1 human breast cancer cells in vitro and in vivo". Oncology Reports 32.4 (2014): 1385-1394.
Chicago
Kinoshita, Y., Yoshizawa, K., Hamazaki, K., Emoto, Y., Yuri, T., Yuki, M., Shikata, N., Kawashima, H., Tsubura, A."Mead acid inhibits the growth of KPL-1 human breast cancer cells in vitro and in vivo". Oncology Reports 32, no. 4 (2014): 1385-1394. https://doi.org/10.3892/or.2014.3390