G‑protein‑coupled receptor 120 regulates the development and progression of human esophageal cancer

  • Authors:
    • Zhen Cui
    • Duojie Li
    • Jingjing Liu
    • Yajun Zhang
    • Hongbo Xu
    • Hongmei Yin
    • Hongwei Li
    • Gengming Wang
    • Hanfei Cai
    • Lei Zhang
    • Shimiao Duan
    • Hao Jiang
  • View Affiliations

  • Published online on: May 31, 2018     https://doi.org/10.3892/or.2018.6470
  • Pages: 1147-1155
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The aim of the present study was to investigate the role of G‑protein coupled receptor 120 (GPR120) in esophageal cancer and explore the related mechanisms. The expression of GPR120 in esophageal cancer tissues was examined by immunohistochemistry. Correlation analysis was performed to investigate the association between the level of GPR120 and clinical parameters. The expression of GPR120 was evaluated in esophageal cancer cell lines and the effects of GPR120 on cell proliferation, clone formation, migration and invasion were evaluated in an in vitro cell model and an in vivo ectopic tumor nude mice model. In addition, the effect of GPR120 on epithelial‑mesenchymal transition (EMT), PI3K and I‑κB pathway, as well as angiogenesis and inflammation‑related cytokines was explored in order to elucidate the underlying mechanisms. Significantly increased expression of GPR120 was observed in esophageal cancer tissues compared to normal tissues. The expression of GPR120 was significantly related with histological grade, TNM stage and lymph node metastasis. GPR120 knockdown significantly decreased cell proliferation, clone formation, migration and invasion in vitro and decreased tumor growth in vivo. Furthermore significantly increased levels of E‑cadherin and decreased levels of N‑cadherin and vimentin, decreased level of Akt phosphorylation and I‑κB phosphorylation, as well as decreased levels of vascular endothelial growth factor (VEGF), interleukin‑8 (IL‑8) and cyclooxygenase‑2 (Cox‑2) and its corresponding protein PGE2 were observed as the underlying mechanisms. In conclusion, we observed an increased level of GPR120 in esophageal cancer tissues, which served as a positive regulator of the development and progression of esophageal cancer. Multiple mechanisms including EMT, PI3K and I‑κB pathway, as well as angiogenesis and inflammation‑related cytokines were involved.

Introduction

Esophageal cancer, a serious malignancy with respect to prognosis and mortality rate, accounts for more than 40,000 deaths worldwide annually (1). According to the statistical data (24), esophageal carcinoma is the eighth most common cancer and the sixth most common cause of cancer-related deaths worldwide with more than 80% of total cases and deaths in developing nations, while the prevalence of esophageal cancer is expected to increase by 140% by 2025. Despite many advances in diagnosis and treatment, the 5-year survival rate for patients diagnosed with esophageal cancer ranges only from 15 to 20% (5,6). Therefore, it is necessary to explore novel therapeutic targets to achieve an improved treatment. Exploration and understanding of the molecular mechanisms involved in the development and progression of esophageal cancer provide possible clues for an improved treatment goal.

Dysregulated expression of G-protein-coupled receptor (GPCR) and dysregulation of GPCR signaling have been recognized as a hallmark of cancer (7). Several studies have revealed that GPCR could affect the multiple biological processes of cancer cells, including proliferation, migration and invasion (810), while these processes were involved in the development and progression of cancer. Recently, several GPCRs identified as free fatty acid receptors have emerged as key players in various physiological homeostasis mechanisms, and GPR120 is one of the receptors (11). Oh et al (12) have demonstrated that GPR120 could function as functional ω-3 PUFA receptor that mediates potent insulin sensitizing and anti-diabetic effects in vivo by supressing macrophage-induced adipose tissue inflammation in obese mice. Since the inflammation effects and macrophage-related properties are cancer related (13), GPR120 was considered to play a role in tumorigenesis. However, the role of GPR120 in esophageal cancer has not yet been explored.

In the present study, we explored the relationship between GPR120 and esophageal cancer and investigated the function and mechanisms of GPR120 in esophageal cancer cells in order to elucidate the role of GPR120 in the development and progression of esophageal cancer.

Materials and methods

Ethical approval of the study protocol

All research involving human participants was approved by the Institutional Review Board of The First Affiliated Hospital of Bengbu Medical School (Bengbu, China). Written informed consent was obtained from the participating individuals. The study protocol on animal research was approved by the Institutional Animal Care and Use Committee of The First Affiliated Hospital of Bengbu Medical School which is adherent to the accepted international guidelines for animal experimentation.

Tissue collection

A total of 100 specimens surgically derived from 100 esophageal cancer patients treated from January 2012 to December 2014 at The First Affiliated Hospital of Bengbu Medical College were collected. Among them, 50 cases belonged to matched tumor and normal mucosae, which were taken at least 5–10 cm away from the edges of a tumor of the same patient. All the enrolled patients did not receive any neoadjuvant chemotherapy or radiation therapy prior to esophagectomy. Fresh tumor tissues or corresponding normal esophageal mucosae were immediately frozen in liquid nitrogen after dissection, then stored at −80°C until further analysis. Tumor specimens were carefully microdissected to ensure that at least 90% of the analyzed tissue contained cancer cells. The clinical diagnosis, tumor stage, histological differentiation and resection margin were determined by routine histopathological examination of hematoxylin and eosin (H&E) stained specimens by an experienced pathologist.

Immunohistochemistry

The paraffin specimen of each patient was stained by H&E staining and the pathological type of the tissue was analyzed by an experienced pathologist. The immunohistochemistry was performed using streptavidin-peroxidase method. The section was deparaffinated by the Leica TP1020 tissue processor (Leica Instruments, Mussloch, Germany) and pre-treated with microwave antigen retrieval procedure at 100°C for 5 min in 10 mM citrate buffer (pH 6.0). After incubation in 3% hydrogen peroxide for 6 min, washing with phosphate-buffered saline (PBS) for 3 times, the slide was blocked using 50 µl goat serum at room temperature (RT) for 30 min. Subsequently, the primary anti-GPR120 antibody (dilution 1:100; cat. no. ab118757; Abcam, Cambridge, MA, USA) was added and the slide was incubated at 4°C overnight. Subsequently, the HRP-conjugated sreptavidin was added followed by washing 3 times with PBS. To visualize the immunostaining, DAB (Dako, Carpinteria, CA, USA) was used. After the tissue turned yellow, the sections were washed, re-stained with hematoxylin, dehydrated and covered.

Assessment of the immunohistochemistry

The intensity of the immunostaining was evaluated by two pathologists without knowing the clinical history of the patients. The cells with dyed membrane or cytoplasm were considered as positive. Five representative regions of ×200 magnification were selected to observe and at least 200 cancer cells were presented at that regions. Frequency and staining intensity of GPR120 by tumor cells were analyzed, and the expression of GPR120 was quantified using the modified Histo-score (H-score) (14), with a range of possible scores from 0 to 300. The expression of GPR120 was categorized into two groups according to the frequency distributions of the H-scores, using a cut-off score of >100 (H-score, 0–99=negative/low expression and 100–300=positive/high expression).

Cell culture

Human esophageal cancer cell lines Eca-109, TE-1 and KYSE450 and human colorectal cancer cell line SW480 were obtained from the American Type Culture Collection (ATCC, Manassas, VA, USA). The cells were maintained as monolayer cultures in cell culture flasks with RPMI-1640 medium containing 10% (v/v) fetal bovine serum (FBS) and 1% antibiotics. Cells were cultured at 37°C in a humidified atmosphere with 5% CO2. All the cell culture media and additives were purchased from Invitrogen (Thermo Fisher Scientific, Inc., Waltham, MA, USA).

Lentiviral shRNA particles

Recombinant lentiviral particles expressing GPR120 or control siRNA were obtained from GenePharma Co. Ltd. (Shanghai, China). Cells were grown to a certain degree of confluency ~40% and then infected with lentiviral particles in complete medium for 48 h. To increase infection efficiency, cells were co-treated with the cationic polymer Polybrene (Sigma-Aldrich; Merck KGaA, Darmstadt, Germany; 8 µg/ml in water). Neither shRNA nor Polybrene affected cell viability. The siRNA and shRNA had no off-target effects and at the indicated multiplicity of infection (MOI) and duration, failed to modulate cell adherence, shape and viability.

Real-time quantitative PCR

Total cellular RNA of human esophageal cancer cell line Eca-109 was extracted using TRIzol reagent (Invitrogen; Thermo Fisher Scientific, Inc.). RT-PCR was performed using a One Step SYBR® PrimeScript™ RT-PCR kit (Takara Biotechnology, Co., Ltd., Dalian, China) and an iQ5 real-time PCR Detection system (Bio-Rad Laboratories, Hercules, CA, USA). The expression of glyceraldehyde 3-phosphate dehydrogenase (GAPDH) gene was assessed simultaneously in all samples as an internal control. Relative gene expression was determined by the 2−ΔΔCt method (15). Oligonucleotide primers specific for GPR120, vascular endothelial growth factor (VEGF), interleukin-8 (IL-8), cyclooxygenase-2 (Cox-2) and GAPDH are listed in Table I.

Table I.

Primer sequences.

Table I.

Primer sequences.

GeneForward primerReverse primer
VEGF 5′-TGCAATGGATCAAGGACCAGAGG-3′ 5′-TGCAGCCAGCAAGAAGCATCAG-3′
IL-8 5′-CCGAGGATCTGATGACGATTA-3′ 5′-GGCTCCCAGAAATAGCTTCAA-3′
Cox-2 5′-CACAGCACAGCCAGGAAGG-3′ 5′-GTTCCCTGGCTCTGAGTAGTCGA-3′
GAPDH 5′-GGATTTGGTCGTATTGGG-3′ 5′-GGAAGATGGTGATGGGATT-3′

[i] VEGF, vascular endothelial growth factor; IL-8, interleukin-8; Cox-2, cyclooxygenase-2; GAPDH, glyceraldehyde 3-phosphate dehydrogenase.

Western blotting

Cells obtained from the above-mentioned treatment were lysed in RIPA buffer, followed by high-speed centrifugation and protein quantification using a bicinchoninic acid assay (Thermo Fisher Scientific). Cellular proteins were separated by sodium dodecyl sulfate-polyacrylamide gel electrophoresis and transferred onto polyvinylidenedifluoride membranes. After blocking, the membranes were incubated with anti-total-(1:1,000; cat. no. 9272) or -phospho-Akt (1:1,000; cat. no. 5012), phospho-IκB (1:1,000; cat. no. 2859), E-cadherin (1:1,000; cat. no. 3195), N-cardherin (1:1,000; cat. no. 13116), vimentin (1:1,000; cat. no. 5741) (Cell Signaling Technology, Inc., Danvers, MA, USA) and GPR120 monoclonal primary antibodies (dilution 1:1,000; cat. no. ab118757; Abcam, Cambridge, MA, USA). GAPDH (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) was used as the loading control. Appropriate horseradish peroxidase-conjugated secondary antibodies (goat anti-rabbit HRP conjugate antibody: Dilution 1:2,000; cat. no. 7074; Cell Signaling Technology; and goat anti-mouse HRP-conjugate antibody: 1:2,000; cat. no. 7076; Cell Signaling Technology) were applied to detect labeled proteins. The protein bands were developed with SuperSignal Ultra Chemiluminescent Substrate (Pierce; Thermo Fisher Scientific) on X-ray films (Kodak Japan Ltd., Tokyo, Japan).

Cell proliferation

Human esophageal cancer cell line Eca-109 (3×103 cells) were seeded in 96-well plates in complete medium and infected with GPR120 or control siRNA lentivirus particles. Two days later, cell proliferation was evaluated by Cell Counting Kit-8 (CCK-8) method according to the manufacturer's instructions using a microplate reader (Molecular Devices, Sunnyvale, CA, USA) to assess the absorbance.

Clone formation

Human esophageal cancer cell line Eca-109 (800 cells) were seeded in 6-well plates in complete medium and infected with GPR120 or control siRNA lentivirus particles. After medium replacement at 24 h post-infection, the cells were maintained at 37°C in a humidified atmosphere with 5% CO2 for 7 days, and then they were stained with crystal violet. The colony survival with a definition of >50 cells were counted under a light microscope (DM4000B; Leica Microsystems, Benshein, Germany). The whole process was performed 3 times to obtain a mean number of colony formation.

Scratch assay

Human esophageal cancer cell line Eca-109 infected with GPR120 or control siRNA lentivirus particles were plated at 70,000 cells/well in a 12-well plate. Cells were grown to 90% confluency and scratched once using a sterile 1-ml pipette tip, washed twice with complete medium to remove floating cells and cell components. Images were captured at a ×40 magnification using a Leica inverted phase contrast microscope (DM IRB; Leica Microsystems). The area of the gap at 24 h was assessed and subtracted from that at 0 h to quantify the migrated cells. The experiments were repeated at least 3 times with similar results.

Cell invasion

A Transwell system was employed to perform the cell invasion assay. Briefly, resuspended Eca-109 cells (2×105 cells) infected with GPR120 or control siRNA lentivirus particles were seeded into the upper chamber prefilled with Matrigel and RPMI-1640 medium supplemented with 20% FBS was added to the lower chamber. After the Transwell plate was maintained in a routine cell culture incubator for a specific time-point, the upper chamber was retained and the membranes were obtained for hematoxylin staining. The cell number of each membrane was determined in 3 randomly picked fields (magnification, ×200) under a light microscope. All the experiments were performed in triplicate.

Nude mice model of ectopic tumor

Athymic nude (nu/nu) 6-weeks old mice were purchased from Shanghai SLAC Laboratory Animal Co. Ltd. (Shanghai, China). The tumors were generated by subcutaneous injection of 2×106 GPR120 or control siRNA lentivirus particles infected Eca-109 cells suspended in 50 µl PBS into the dorsal region near the thigh. Mice were then weighted and assessed for tumor size every other day by measuring tumor length and tumor width. At week 4 post-treatment, all mice were sacrificed by cervical dislocation and the tumors were excised, weighted and imaged. For histological analysis, organs from the treated groups and the control group were fixed in 4% formalin, and then conducted with paraffin-embedded sections for H&E staining. The slices were examined by a digital microscope (Leica QWin Plus v3 software; Leica Microsystems).

Enzyme-linked immunosorbent assay (ELISA)

Condition medium was obtained from the above-described cell culture at 1,500 × g centrifugation for 10 min and was stored at −80°C before further processing. Angiogenesis and inflammation-related cytokines including VEGF, IL-8 and PGE2, were determined by ELISA kit (Invitrogen; Thermo Fisher Scientific) according to the manufacturer's instructions.

Statistical analysis

All statistical analyses were performed using SPSS version 18 (SPSS, Inc., Chicago, IL, USA). Data are presented as the mean ± SD. The Student's t-test or one-way analysis of variance (ANOVA) were used to examine differences between groups. A P-value of <0.05 was considered to indicate a statistically significant difference.

Results

Correlation analysis between GPR120 level and clinical parameters in esophageal cancer

In order to investigate the role of GPR120 in esophageal cancer, we firstly performed the correlation analysis between the expression level of GPR120 and the clinical parameters of the esophageal cancer patients. As displayed in Table II, the expression level of GPR120 was significantly elevated in esophageal cancer tissues and correlated with histological grade (P<0.001), lymph node metastasis (P=0.003) and metastasis depth (P<0.001). These results indicated that GPR120 affected the progression of esophageal cancer.

Table II.

Correlation analysis between the level of GPR-120 and the clinical parameters.

Table II.

Correlation analysis between the level of GPR-120 and the clinical parameters.

GPR-120

ParametersCases (n=100)LowHighχ2P-value
Age (years) 0.2190.640
  ≤60431132
  >60571740
Sex 0.1180.732
  Female421131
  Male581741
Tissue type 112.5<0.001
  Normal1001000
  Tumor1002872
Size (cm) 0.8420.359
  ≤5571839
  >5431033
Histological grade 43.96<0.001
  I16151
  II641351
  III20020
Location 0.1210.942
  Neck and upperthoracic1037
  Mid-thoracic481434
  Lower-thoracic421131
Lymph node metastasis 8.730.003
  Yes45639
  No552233
Metastasis depth 16.053<0.001
  T1+T2311714
  T3+T4691158

[i] GPR-120, G-protein coupled protein-120.

Effects of GPR120 on cell proliferation, clone formation, cell migration and invasion in esophageal cancer

Since the expression of GPR120 in esophageal cancer was elevated, we obtained some esophageal cancer cell lines to investigate the biological function of GPR120. In order to mimic the clinical status, we first examined the expression level of GPR120 in esophageal cancer cell lines. As displayed in Fig. 1, significantly increased level of GPR120 expression was found in Eca-109 cells compared to TE-1 and KYSE450 cells. Therefore, Eca-109 cell line was selected as the cell line for the following experiments. According to the results displayed in Fig. 2, GPR120-knockdown esophageal cancer cell line Eca-109 exhibited a significantly decreased degree of cell proliferation, clone formation, cell migration and invasion compared to the control cells. These results indicated that GPR120 affected the biological function of esophageal cancer cells via proliferation, clone formation, migration and invasion.

Effects of GPR120 on tumor growth in vivo

Due to the tumor-promoting effects of GPR120 observed in vitro, we further established an ectopic tumor nude mice model to evaluate the effects of GPR120 in vivo. As displayed in Fig. 3A and B, GPR120-knockdown esophageal cancer cell line Eca-109 exhibited a decreased level of tumor growth in vivo according to tumor size and weight. Furthermore, the immunostaining results also confirmed the effects of GPR120 knockdown in esophageal cancer cells (Fig. 3C).

Mechanism involved in the effects of GPR120

We further explored the mechanism involved in the effects of GPR120 in esophageal cancer. Our observations indicated that GPR120 knockdown in esophageal cancer cell line Eca-109 resulted in increased level of the EMT marker E-cadherin and decreased level of N-cadherin and vimentin, decreased level of Akt phosphorylation and I-κB phosphorylation compared to the control cells (Fig. 4). These results indicated the possible involvement of EMT process, PI3K/Akt pathway and NF-κB in the role of GPR120 in esophageal cancer. Furthermore, we also examined the possible role of angiogenesis and inflammatory cytokines on the effects of GPR120 and we observed decreased mRNA and protein levels of angiogenesis cytokine VEGF, inflammatory cytokine IL-8 and Cox-2 (protein PGE2) in GPR120-knockdown Eca-109 cells compared to control cells (Fig. 5).

Discussion

Practical strategies have been proposed to prevent the harmful sequelae of the worldwide obesity epidemic in order to reduce the future medical burden to society. Research has indicated the association between obesity and the overall risk for multiple cancers, including endometrial, colorectal, prostate, pancreatic and postmenopausal breast cancer (1620). However, the exact role of obesity in cancer risk has not been fully explored. According to a previous study (21), the physiological effects related to obesity, including increased tissue inflammation, insulin resistance and/or hyperinsulinemia are considered to play a critical role in cancer risk. Therefore, dietary intervention is a potential mean to decrease this type of risk in our daily life. Altering the balance between dietary ω-3 and ω-6 polyunsaturated fatty acids (PUFAs) has been considered as an approach for disease prevention (22,23) and several epidemiological and preclinical studies have revealed an antitumor effect of ω-3 PUFAs in cancer patients (2426). The detailed mechanisms mediated by which ω-3 PUFAs, particularly eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA), exert their anticancer effects are not well understood despite multiple targets regulating cell proliferation and survival, inflammation, angiogenesis and metastasis may be involved (27). Recently, several GPCRs identified as free fatty acid receptors have emerged as key players in various physiological homeostasis mechanisms, and GPR120 has been demonstrated to function as a receptor for ω-3 PUFAs (11), and molecular and cellular effects could be generated following the ligand-receptor interaction.

In the present study, we firstly evaluated the expression of GPR120 in esophageal cancer tissue and observed significantly increased GPR120 in esophageal cancer tissues compared to the normal tissues. Based on this observation, we performed in vitro and in vivo experiments to investigate the role of GPR120 in esophageal cancer development and progression. Our results indicated that GPR120 served as tumor-promoting regulator in esophageal cancer according to cell model and nude mice ectopic model. In addition, the investigation of the underlying mechanism indicated that EMT, PI3K and I-κB pathway, as well as angiogenesis and inflammation-related cytokines secretion attributed to the phenotype resulted by GPR120. To the best of our knowledge, this is the first study to elucidate the role of GPR120 in esophageal cancer.

Oh et al (12) have demonstrated that GPR120 is a functional ω-3 PUFA receptor that mediates potent insulin sensitizing and anti-diabetic effects in vivo by suppressing macrophage-induced adipose tissue inflammation in obese mice. The state of chronic, low grade inflammation arising in obesity is characterized by infiltration of M1-type adipose tissue macrophages, cells that secrete high levels of proinflammatory cytokines, including TNF-α, IL1β and IL-6, which are considered to be major contributors to tissue inflammation and insulin resistance in obesity (28,29). In cancer patients, increased inflammation levels are positively correlated with tumor cell proliferation, tumor stage and lymph node metastasis (30,31). In the present study, we also demonstrated that GPR120 promoted tumor cell proliferation, migration and invasion, and its expression level was associated with tumor stage and lymph node metastasis.

According to a previous study, the PI3K and NFκB pathways are involved in the inflammatory signaling pathway and ω-3 PUFAs can inhibit these pathways by sequestering TAB1 in obese mice (32). In the presents study, we also demonstrated that GPR120 knockdown resulted in decreased activity of Akt and I-κB phosphorylation. In a study by Wu et al (33), they also revealed that GPR120 exerted its functions via the PI3K and NFκB pathways in colorectal cancer. However, controversial results also existed. In a recently published study, Chung et al (34) demonstrated that obesity promoted mammary tumor progression in a model of postmenopausal breast cancer and that ω-3 PUFAs inhibited mammary tumor progression in obese mice, independently of GPR120. We believe that the differences may be attributed to the type of cancer.

Besides the aforementioned signaling pathways, GPR120 is considered to enhance cell motility by inducing EMT. In the present study, we observed significantly increased levels of E-cadherin and decreased level of N-cadherin and vimentin in GPR120 knockdown esophageal cancer cells compared to control cells, which is consistent with previous research.

In conclusion, in the present study, we demonstrated that increased level of GPR120 in esophageal cancer tissues, functioned as a positive regulator of the development and progression of esophageal cancer. Furthermore, multiple mechanisms including EMT, PI3K and I-κB pathway, as well as and angiogenesis and inflammation-related cytokines secretion were involved.

Acknowledgements

Not applicable.

Funding

This study was supported by the ‘12th Five-year’ Clinical Medical Key Construction Foundation of Anhui province (no. 01Z33).

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors' contributions

ZC, DL, JL and HJ conceived and designed the experiments; ZC, DL, JL, YZ, HX, HY, HL, GW, HC, LZ and SY performed the experiments; ZC, DL, JL, YZ, HX, HY, HL, GW, HC, LZ, SY and HJ analyzed the data; ZC, DL, JL, YZ, HX, HY, HL, GW, HC, LZ, SY and HJ contributed reagents/materials/analysis tools; ZC, DL, JL and HJ contributed to the writing of 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.

Ethics approval and consent to participate

All experimental protocols were approved by the Institutional Review Board of The First Affiliated Hospital of Bengbu Medical School (Bengbu, China).

Patient consent for publication

Not applicable.

Competing interests

The authors state that they have no competing interests.

References

1 

Siegel R, Ma J, Zou Z and Jemal A: Cancer statistics, 2014. CA Cancer J Clin. 64:9–29. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Napier KJ, Scheerer M and Misra S: Esophageal cancer: A review of epidemiology, pathogenesis, staging workup and treatment modalities. World J Gastrointest Oncol. 6:112–120. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Global Burden of Disease Cancer Collaboration; Fitzmaurice C, Allen C, Barber RM, Barregard L, Bhutta ZA, Brenner H, Dicker DJ, Chimed-Orchir O, Dandona R, Dandona L, et al: Global, regional, and national cancer incidence, mortality, years of life lost, years lived with disability, and disability-adjusted life-years for 32 cancer groups, 1990 to 2015: A systematic analysis for the global burden of disease study. JAMA Oncol. 3:524–548. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Liang H, Fan JH and Qiao YL: Epidemiology, etiology, and prevention of esophageal squamous cell carcinoma in China. Cancer Biol Med. 14:33–41. 2017. View Article : Google Scholar : PubMed/NCBI

5 

Miller KD, Siegel RL, Lin CC, Mariotto AB, Kramer JL, Rowland JH, Stein KD, Alteri R and Jemal A: Cancer treatment and survivorship statistics, 2016. CA Cancer J Clin. 66:271–289. 2016. View Article : Google Scholar : PubMed/NCBI

6 

Pennathur A, Gibson MK, Jobe BA and Luketich JD: Oesophageal carcinoma. Lancet. 381:400–412. 2013. View Article : Google Scholar : PubMed/NCBI

7 

Steury MD, McCabe LR and Parameswaran N: G protein-coupled receptor kinases in the inflammatory response and signaling. Adv Immunol. 136:227–277. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Nohata N, Goto Y and Gutkind JS: Onco-GPCR signaling and dysregulated expression of microRNAs in human cancer. J Hum Genet. 62:87–96. 2017. View Article : Google Scholar : PubMed/NCBI

9 

Nogués L, Palacios-García J, Reglero C, Rivas V, Neves M, Ribas C, Penela P and Mayor F Jr: G protein-coupled receptor kinases (GRKs) in tumorigenesis and cancer progression: GPCR regulators and signaling hubs. Semin Cancer Biol. 78–90. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Khalil BD, Hsueh C, Cao Y, Abi Saab WF, Wang Y, Condeelis JS, Bresnick AR and Backer JM: GPCR signaling mediates tumor metastasis via PI3Kβ. Cancer Res. 76:2944–2953. 2016. View Article : Google Scholar : PubMed/NCBI

11 

Hara T, Kashihara D, Ichimura A, Kimura I, Tsujimoto G and Hirasawa A: Role of free fatty acid receptors in the regulation of energy metabolism. Biochim Biophys Acta. 1841:1292–1300. 2014. View Article : Google Scholar : PubMed/NCBI

12 

Oh DY, Talukdar S, Bae EJ, Imamura T, Morinaga H, Fan W, Li P, Lu WJ, Watkins SM and Olefsky JM: GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects. Cell. 142:687–698. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Diakos CI, Charles KA, McMillan DC and Clarke SJ: Cancer-related inflammation and treatment effectiveness. Lancet Oncol. 15:e493–e503. 2014. View Article : Google Scholar : PubMed/NCBI

14 

McCarty KS Jr, Miller LS, Cox EB, Konrath J and McCarty KS Sr: Estrogen receptor analyses. Correlation of biochemical and immunohistochemical methods using monoclonal antireceptor antibodies. Arch Pathol Lab Med. 109:716–721. 1985.

15 

Ji Y, Strawn TL, Grunz EA, Stevenson MJ, Lohman AW, Lawrence DA and Fay WP: Multifaceted role of plasminogen activator inhibitor-1 in regulating early remodeling of vein bypass grafts. Arterioscler Thromb Vasc Biol. 31:1781–1787. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Golabek T, Bukowczan J, Chłosta P, Powroźnik J, Dobruch J and Borówka A: Obesity and prostate cancer incidence and mortality: A systematic review of prospective cohort studies. Urol Int. 92:7–14. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Kolodecik T, Shugrue C, Ashat M and Thrower EC: Risk factors for pancreatic cancer: Underlying mechanisms and potential targets. Front Physiol. 4:4152014. View Article : Google Scholar : PubMed/NCBI

18 

Ligibel JA and Strickler HD: Obesity and its impact on breast cancer: Tumor incidence, recurrence, survival, and possible interventions. Am Soc Clin Oncol Educ Book. 2013:52–59. 2013. View Article : Google Scholar

19 

Patterson RE, Rock CL, Kerr J, Natarajan L, Marshall SJ, Pakiz B and Cadmus-Bertram LA: Metabolism and breast cancer risk: Frontiers in research and practice. J Acad Nutr Diet. 113:288–296. 2013. View Article : Google Scholar : PubMed/NCBI

20 

Van Kruijsdijk RC, van der Wall E and Visseren FL: Obesity and cancer: The role of dysfunctional adipose tissue. Cancer Epidemiol Biomarkers Prev. 18:2569–2578. 2009. View Article : Google Scholar : PubMed/NCBI

21 

James FR, Wootton S, Jackson A, Wiseman M, Copson ER and Cutress RI: Obesity in breast cancer-what is the risk factor? Eur J Cancer. 51:705–720. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Fares H, Lavie CJ, DiNicolantonio JJ, O'Keefe JH and Milani RV: Omega-3 fatty acids: A growing ocean of choices. Curr Atheroscler Rep. 16:3892014. View Article : Google Scholar : PubMed/NCBI

23 

Simopoulos AP: An increase in the omega-6/omega-3 fatty acid ratio increases the risk for obesity. Nutrients. 8:1282016. View Article : Google Scholar : PubMed/NCBI

24 

Farvid MS, Eliassen AH, Cho E, Liao X, Chen WY and Willett WC: Dietary fiber intake in young adults and breast cancer risk. Pediatrics. 137:e201512262016. View Article : Google Scholar : PubMed/NCBI

25 

Fabian CJ, Kimler BF and Hursting SD: Omega-3 fatty acids for breast cancer prevention and survivorship. Breast Cancer Res. 17:622015. View Article : Google Scholar : PubMed/NCBI

26 

Zheng JS, Hu XJ, Zhao YM, Yang J and Li D: Intake of fish and marine n-3 polyunsaturated fatty acids and risk of breast cancer: Meta-analysis of data from 21 independent prospective cohort studies. BMJ. 346:f37062013. View Article : Google Scholar : PubMed/NCBI

27 

Nabavi SF, Bilotto S, Russo GL, Orhan IE, Habtemariam S, Daglia M, Devi KP, Loizzo MR, Tundis R and Nabavi SM: Omega-3 polyunsaturated fatty acids and cancer: Lessons learned from clinical trials. Cancer Metastasis Rev. 34:359–380. 2015. View Article : Google Scholar : PubMed/NCBI

28 

Kraakman MJ, Murphy AJ, Jandeleit-Dahm K and Kammoun HL: Macrophage polarization in obesity and type 2 diabetes: Weighing down our understanding of macrophage function? Front Immunol. 5:4702014. View Article : Google Scholar : PubMed/NCBI

29 

Ballak DB, Van Diepen JA, Moschen AR, Jansen HJ, Hijmans A, Groenhof GJ, Leenders F, Bufler P, Boekschoten MV, Müller M, et al: IL-37 protects against obesity-induced inflammation and insulin resistance. Nat Commun. 5:47112014. View Article : Google Scholar : PubMed/NCBI

30 

Katanov C, Lerrer S, Liubomirski Y, Leider-Trejo L, Meshel T, Bar J, Feniger-Barish R, Kamer I, Soria-Artzi G, Kahani H, et al: Regulation of the inflammatory profile of stromal cells in human breast cancer: Prominent roles for TNF-α and the NF-κB pathway. Stem Cell Res Ther. 6:872015. View Article : Google Scholar : PubMed/NCBI

31 

Zhu X, Du L, Feng J, Ling Y and Xu S: Clinicopathological and prognostic significance of serum cytokine levels in breast cancer. Clin Lab. 60:1145–1151. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Oh DY, Walenta E, Akiyama TE, Lagakos WS, Lackey D, Pessentheiner AR, Sasik R, Hah N, Chi TJ, Cox JM, et al: A Gpr120-selective agonist improves insulin resistance and chronic inflammation in obese mice. Nat Med. 20:942–947. 2014. View Article : Google Scholar : PubMed/NCBI

33 

Wu Q, Wang H, Zhao X, Shi Y, Jin M, Wan B, Xu H, Cheng Y, Ge H and Zhang Y: Identification of G-protein-coupled receptor 120 as a tumor-promoting receptor that induces angiogenesis and migration in human colorectal carcinoma. Oncogene. 32:5541–5550. 2013. View Article : Google Scholar : PubMed/NCBI

34 

Chung H, Lee YS, Mayoral R, Oh DY, Siu JT, Webster NJ, Sears DD, Olefsky JM and Ellies LG: Omega-3 fatty acids reduce obesity-induced tumor progression independent of GPR120 in a mouse model of postmenopausal breast cancer. Oncogene. 34:3504–3513. 2015. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2018
Volume 40 Issue 2

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
Cui Z, Li D, Liu J, Zhang Y, Xu H, Yin H, Li H, Wang G, Cai H, Zhang L, Zhang L, et al: G‑protein‑coupled receptor 120 regulates the development and progression of human esophageal cancer. Oncol Rep 40: 1147-1155, 2018
APA
Cui, Z., Li, D., Liu, J., Zhang, Y., Xu, H., Yin, H. ... Jiang, H. (2018). G‑protein‑coupled receptor 120 regulates the development and progression of human esophageal cancer. Oncology Reports, 40, 1147-1155. https://doi.org/10.3892/or.2018.6470
MLA
Cui, Z., Li, D., Liu, J., Zhang, Y., Xu, H., Yin, H., Li, H., Wang, G., Cai, H., Zhang, L., Duan, S., Jiang, H."G‑protein‑coupled receptor 120 regulates the development and progression of human esophageal cancer". Oncology Reports 40.2 (2018): 1147-1155.
Chicago
Cui, Z., Li, D., Liu, J., Zhang, Y., Xu, H., Yin, H., Li, H., Wang, G., Cai, H., Zhang, L., Duan, S., Jiang, H."G‑protein‑coupled receptor 120 regulates the development and progression of human esophageal cancer". Oncology Reports 40, no. 2 (2018): 1147-1155. https://doi.org/10.3892/or.2018.6470