Leptin promotes proliferation and metastasis of human gallbladder cancer through OB-Rb leptin receptor

  • Authors:
    • Hao Zou
    • Yunxia Liu
    • Dong Wei
    • Tao Wang
    • Kun Wang
    • Songquan Huang
    • Lixin Liu
    • Yuehua Li
    • Jiayun Ge
    • Xiao Li
    • Hong Zhu
    • Lianmin Wang
    • Songling Zhao
    • Xiaowen Zhang
    • Lin Wang
  • View Affiliations

  • Published online on: May 18, 2016     https://doi.org/10.3892/ijo.2016.3530
  • Pages: 197-206
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Emerging evidence has shown that leptin, an adipocyte-derived cytokine that is closely associated with obesity, play a significant role in carcinogenesis and tumorigenesis. However, its impact on gallbladder cancer (GBC) remains unclear. In this study, we firstly found that leptin and its functional receptor OB-Rb were significantly co-expressed in human GBC tissues and cell lines, the content of which were higher than those in normal human gallbladder tissues. Treatment with leptin promoted the proliferation, migration and invasion of GBC cells, which were attenuated by OB-Rb shRNA. Blocking in the G2/M period of cell cycle, increasing of MMP3 and MMP9, increasing of VEGF-C/D, activation of SOCS3/JAK2/p-STAT3 pathway was demonstrated after treatment with leptin. All of these positive responses were attenuated by OB-Rb receptor shRNA. Taken together, our findings suggest that leptin promoted the proliferation, migration and invasion of GBC cells by increasing OB-Rb expression through the SOCS3/JAK2/p-STAT3 signal pathway. Targeting the leptin/OB-Rb axis could be an attractive therapeutic strategy for treatment of GBC.

Introduction

Gallbladder cancer (GBC) is a rare but highly aggressive malignancy. The lack of severe symptoms makes the diagnosis very difficult (1). Even though there are some therapies such as cholecystectomy or radical resection, chemotherapy, or radiotherapy (2,3), they are not as effective as expected. The 5-year survival rate is extremely low (4). So far there is no systemic therapy with a satisfactory outcomes. Thus, studying novel signal molecules involved in GBC margin and metastasis may provide new effective therapeutic strategies.

Leptin, the product of the OB gene, is a 16 kDa non-glycosylated peptide hormone which is synthesized almost exclusively by adipocytes that regulates appetite and energy expenditure at the hypothalamic level (5,6). In recent years, accumulating evidence suggests that leptin plays an important role in tumorigenesis, angiogenesis and metastasis of many cancers, including breast (7), pancreatic (8), and stomach cancer (9). Previous studies have shown that leptin could activate Janus kinase 2 (JAK2) when leptin was bound to one form of the receptor, OB-Rb. Then JAK2 initiated downstream signaling including members of the signal transducers and activators of transcription (STAT) family of transcription factors (10). However, the expression of leptin and OB-Rb in GBCs has not been fully investigated, and the precise role of leptin in the development and promotion of GBC remains unknown.

In this study, we investigated the clinical implications of leptin and OB-Rb in GBC patients. Moreover, we explored the role of leptin and one form of its receptor OB-Rb in GBC cells through in vitro and in vivo studies. SOCS3/JAK2/p-STAT3 signaling pathways were also assessed and these pathways may be involved in cell migration and metastasis by leptin.

Materials and methods

Immunohistochemistry and evaluation

Forty paraffin-embedded specimens of normal gallbladder tissues and 40 specimens of gallbladder cancer (GBC) tissues were collected from January 1, 2005 to June 30, 2010 at Department of Hepatopancreatobiliary Surgery, the Second Affiliated Hospital of Kunming Medical University, Kunming, China. No patients had received chemotherapy or radiotherapy before biopsy. The prior patient's consents and approval from the Institutional Research Ethics Committee were obtained. Rabbit anti-human polyclonal leptin and OB-Rb antibody (Sigma, St. Louis, MO, USA) was used for immunohistochemistry assay, which was performed following the protocol of Universal SP kit (MXB Biotechnology, Fujian, China). Positive staining of leptin protein is brown in the cytoplasm, partly in the nucleus, and positive staining of OB-Rb protein is brown in the cytomembrane. The human GBC tissue sections were blindly examined and scored concurrently by two observers. The intensity of the immunostaining was scored as 0 (negative), 1 (weak), 2 (moderate), or 3 (strong). Four visual fields were selected randomly under high power lens (x400). The number of positive cells was counted and the average positive rate was calculated. The percentage of positive tumor cells was scored as ‘+’ (<25%), ‘++’ (26–50%), ‘+++’ (51–75%), or ‘++++’, (76–100%), and that without any positive cells scored as ‘−’.

Cell culture

The human GBC cell sublines (GBC-SD) were obtained from the Type Culture Collection of the Chinese Academy of Sciences (Shanghai, China). The cells were cultured in Dulbecco's modified Eagle's medium/α-modified Eagle medium (DMEM) (Gibco Life Technology, Gaithersburg, MD, USA) supplemented with 10% fetal bovine serum (FBS) (Gibco Life Technology) and maintained at 37°C in a humidified atmosphere of 5% CO2.

shRNA synthesis and transfection

Four different template oligonucleotides targeting OB-Rb (Table II) were synthesized by Ribobio Inc. (Guangzhou, Guangdong, China), and were annealed and ligated into pGPH1/GFP/Neo plasmid. The shRNAs inserted vectors were named as pGPH-GFP-s1, pGPH-GFP-s2, pGPH-GFP-s3, pGPH-GFP-s4 and pGPH-GFP-NC, respectively.

Table II

Uni- and multivariate analyses of factors associated with survival.

Table II

Uni- and multivariate analyses of factors associated with survival.

FactorOSDFS


Univariate, P-valueMultivariateUnivariate, P-valueMultivariate


HR95% CIP-valueHR95% CIP-value
Gender (male vs. female)0.117NA0.328NA
Age, years (≥60 vs. <60)0.056NA0.070NA
BMI, kg/m2 (≥30 vs. <30)0.584NA0.326NA
CA199, U/ml (≥35 vs. <35)0.0023.5321.485–13.4060.0120.0043.8761.514–12.7110.027
T classification (T1 vs. T2)0.091NA0.242NA
N classification (N0 vs. N1)0.093NA0.167NA
Tumor differentiation (I/II vs. III/IV)0.782NA0.765NA
AJCC (I vs. II)0.575NA0.648NA
Leptin expression (low vs. high)0.0282.2711.865–18.6150.0010.0442.8741.196–19.6820.003
OB-Rb expression (low vs. high)0.0163.4612.043–18.2920.0010.0202.9311.970–11.4380.001

[i] Cox's proportional hazards regression model. OS, overall survival; DFS, disease-free survival; HR, hazard ratio; 95% CI, 95% confidence interval; NA, not adopted.

Transfection of shRNAs was performed using Lipofectamine 2000 reagent (Invitrogen Co., Carlsbad, CA) according to the manufacturer's instructions. Briefly, GBC-SD cells were applied for OB-Rb silence, which were cultured in 6-well plates at a density of 5×104 cells/well. Then GBC-SD cells were subject to shR-OB-Rb and shR-Con treatment for 24, 48 and 96 h. To evaluate the infection efficiency, cells were observed under a fluorescence microscope and the percentage of GFP-positive cells was counted.

Quantitative real-time PCR analysis

Total RNA from GBC-SD cells was extracted using the TRIzol reagent (Invitrogen). Then, 2 μg of total RNA was subjected to reverse transcription for cDNA synthesis by using MMLV (MBI Fermentas, Euromedex, Souffelweyersheim, France). Real-time PCR was performed with the manufacturer's (Kapa Biosystem, Hercules, CA, USA) instructions. The primer sequences listed below were used. A mathematical model, 2−ΔΔCT method, was used for relative quantification in real-time PCR (11). GAPDH was used as internal control gene to normalize the variability at mRNA expression levels.

Western blot analysis

The GBC-SD cell pellet was washed twice with ice-cold phosphate buffered saline (PBS) and lysed with lysis buffer (Beyotime Institute of Biotechnology, Jiangsu, China). Protein (30 mg) was loaded and separated in 12% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) gel and transferred to polyvinylidene difluoride membranes (Millipore, Bedford, MA, USA). The following antibodies were used to probe the alterations of protein: JAK2 (Abcam, Cambridge, UK), SOCS3 (Abcam), STAT3 and p-STAT3 (Cell Signaling Technology, Inc., Danvers, MA, USA), MMP-3 and MMP-9 (Santa Cruz Biotechnology, Santa Cruz, CA, USA), VEGF-C/D (Santa Cruz Biotechnology). GAPDH (Santa Cruz Biotechnology) was used as loading control. Signal was detected by enhanced chemiluminescence techniques (Pierce Thermo Scientific, Rockford, IL, USA).

Cell proliferation assay

Cells (2×104) per well were seeded into 96-well plate and incubated overnight. Then the medium was removed. Medium (100 μl) with the final concentration of 100 nM OB-Rb shRNA was added to each well with or without leptin (250 ng/ml). Scramble shRNA or untreated cells were used as the control group. All groups were in triplicate. After 24, 48, and 72 h transfection, cell proliferation was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay (Beyotime Institute of Biotechnology).

Flow cytometry analysis

Cells (2×105) were seeded in 6-well plates and incubated overnight to 50–60% confluence. OB-Rb shRNA was added into the medium at a final concentration of 100 nM with or without leptin (250 ng/ml). Scramble shRNA or untreated cells were used as the control group. The cells were incubated with leptin for 24 h, then treated into single cell suspension with cold PBS. The experiment was performed following manufacturer's protocol of Annexin V-FITC Apoptosis and Cells cycle Detection kit (Beyotime Biotechnology, Jiangsu, China). Then, rates of apoptosis were analyzed with FACScan system (BD Biosciences, Franklin Lakes, NJ, USA). Each experiment was performed in triplicate, independently.

Cell migration and invasion assay

Transwell chambers and Matrigel Invasion Chambers (8 μm pore size, Corning Inc., Corning, NY, USA) were used for cell migration and invasion assay, respectively. GBC-SD cells transfected with OB-Rb shRNA or scramble shRNA were treated with or without leptin (250 ng/ml). After 24 h, cells were detached. Then 500 μl medium with 20% FBS was added into each lower chamber which was incubated at 37°C. Incubation periods were 2 h for migration, and 4 h for invasion. Then, the surface of the upper chamber was swabbed with cotton-tipped applicators to remove the cells that did not migrate. The lower membrane surface was fixed in methanol and stained by crystal violet. Migrating cells were counted using light microscopy (five random 100× fields per well) or a spectrophotometer. Results were calculated from three independent experiments.

Immunofluorescence

shRNA-transfected cells (2×105) and untreated cells were all seeded on coverslips. Cells were cultured in a 6-well plate and incubated with leptin for 24 h, then rinsed twice in PBS and fixed with methanol at −20°C for 20 min. Following fixation, the coverslips were directly washed in PBS for 5 min, followed by incubation with PBS, 0.2% Triton X-100 and 5% bovine serum albumin for 20 min at room temperature. Following rinsing with PBS, the cells were incubated with primary antibody at 4°C in a humidity box. Primary antibodies included mouse anti-JAK2 (1:200; Abcam), rabbit anti-STAT3 or p-STAT3 (1:200; CST) and rabbit polyclonal to SOCS3 (1:200; Abcam). Coverslips were subsequently washed 3 times with cold PBS and incubated with the corresponding secondary antibodies (diluted to 1:500, Santa Cruz Biotechnology) for 2 h at room temperature in the dark, humid box. DAPI staining was then performed to identify the nuclei.

Gelatin zymography assay

Cells (2×105) transfected with OB-Rb shRNA or scramble shRNA were seeded in a 6-well plates and incubated for 6 h. Then leptin (250 ng/ml) was added and the cells incubated for another 24 h. After each treatment, the cells were washed twice with serum-free medium, and used for a zymogram according to the protocol of Zymography kit (Genmed, Shanghai, China).

Enzyme-linked immunosorbent assay (ELISA)

VEGF-C and VEGF-D levels were detected using the ELISA kit according to the manufacturer's manual (Keygentec, Shanghai, China). Colorimetric measurement was recorded as OD 450 readings.

Xenograft model assay

All animal procedures were previously approved by the Kunming Medical University ethics committee. Female BALB/c nu/nu mice (4–5 weeks old, 15–18 g), from Vital River Laboratory Animal Technology Co., Ltd. (Peking, China), were randomly assigned into five groups as described above: control, shR-NC, leptin (1 mg/kg), shR-OB-Rb, and shR-OB-Rb + Leptin (1 mg/kg), groups. Approximately 5×106 cells were suspended in 0.1 ml PBS and injected subcutaneously into each mouse. The tumors were monitored every 5 days beginning at day 5 by measuring two perpendicular diameters with a caliper. The mice were sacrificed on the 30th day after injection. The tumors were dissected and weighed.

Statistical analysis

Statistical analysis was performed with SPSS software (17.0; SPSS, Inc., Chicago, IL, USA). Values are expressed as mean ± standard deviation (SD). The Student's t-test was used for comparisons between groups. Categorical data were analyzed by the chi-square or Fisher's exact tests. Correlation analysis was performed between leptin and OB-Rb. Cumulative recurrence and survival rates were analyzed using Kaplan-Meier's method and the log-rank test. Cox's proportional hazards regression model was used to analyze independent prognostic factors. Statistical significance was defined as P-value <0.05.

Results

Expression levels of leptin and OB-Rb in GBC tissues

To evaluate the possible roles of leptin in GBC, we investigated the expression levels of leptin and OB-Rb in human normal gallbladder and GBC tissues by immunohistochemistry. Compared to normal gallbladder tissues, expression levels of leptin were significantly upregulated in GBC tissues (P=0.000). Moreover, OB-Rb density was significantly higher in GBC tissues than in normal gallbladder tissues (P=0.001) (Fig. 1A). In addition, a scatter plot of leptin and OB-Rb expression revealed a significantly positive correlation between leptin and OB-Rb levels in cancerous tissues (r=0.797, P=0.000). The characteristics of the study participants including age, gender, BMI, T classification, N classification, Tumor differentiation, AJCC stage are shown in Table I. Results demonstrated that GBC patients with leptinhigh had high BMI (P<0.001), elevated CA199 (P<0.001), high T (P=0.030), N (P=0.003) classification and AJCC stage (P=0.001), poor differentiation (P=0.026). Moreover, GBC patients with OB-Rbhigh had high BMI (P<0.001), elevated CA199 (P<0.001), high T (P=0.012), N (P=0.001) classification and AJCC stage (P<0.001), poor differentiation (P=0.026). We then analyzed the prognostic implication of leptin and OB-Rb expression. Importantly, we found that patients with leptinhigh and OB-Rbhigh expression had significantly worse prognosis than those with leptinlow and OB-Rblow expression (Fig. 1B). Multivariate analysis identified leptin and OB-Rb expression as an independent predictor for disease-free survival and overall survival (OS; Table II). These results indicate that leptin and OB-Rb is likely involved in tumorigenesis and progression of GBC.

Table I

Clinical characteristics of 40 gallbladder carcinoma (GBC) patients and leptin and OB-Rb expression.

Table I

Clinical characteristics of 40 gallbladder carcinoma (GBC) patients and leptin and OB-Rb expression.

LeptinOB-Rb


CharacteristicsLow (n=15)High (n=25)P-valueLow (n=17)High (n=23)P-value
Age
 ≥608110.5671180.061
 <60714615
Gender
 Male390.285570.994
 Female12161216
BMI, kg/m2
 <30121<0.001130<0.001
 ≥30324423
CA199, U/ml
 <3591<0.001100<0.001
 ≥35624723
T classification (1)
 T1b/T214150.03016130.012
 T3110110
N classification (1)
 N015140.00317120.001
 N1011011
Tumor differentiation
 I/II13130.02613130.191
 III/IV212410
AJCC (1)
 I14100.001168<0.001
 II115115

[i] P<0.05 was considered statistically significant. Pearson's Chi-square test and Fisher exact test (when 1<T<5) were used.

Involvement of OB-Rb receptor in leptin-mediated growth, migration and invasion of GBC-SD cells

Leptin exhibits its effects on cancer through interaction with specific leptin receptors (OB-Rb and OB-Rs) (12). In this study, we examined the effect of leptin on GBC-SD cell growth, migration and invasion. The result from MTT assay showed leptin (40 ng/ml) significantly increased the proliferation of GBC-SD cells after 24-h incubation compared with basal values, which were significantly inhibited by shR-OB-Rb transfection (Fig. 2A).

Leptin was able to promote GBC-SD cell migration, which was significantly suppressed in GBC-SD cell transfected with OB-Rb shRNA at 24 h compared with the control group as shown in Fig. 2B. Transwell matrix penetration assay showed that leptin treatment increased the mean of GBC-SD cell invasive number, which could be repressed by transfection with OB-Rb shRNA (Fig. 2C). Flow cytometry was used to analyze cell apoptosis in GBC-SD treated with leptin or OB-Rb shRNA for 24 h. As shown in Fig. 2D–G, leptin treatment or OB-Rb knockdown significantly induced G2/M-phase cell cycle arrest, and decreased cells number of G0/G1 and S-phase. OB-Rb knockdown significantly induced apoptosis in GBC-SD cell line, compared with the other groups (P<0.05).

Leptin promoting growth and metastasis were retarded by OB-Rb RNAi

In vivo, the volumes and weight of xenograft tumors removed from nude mice which were injected with leptin and shR-NC were both higher than the other groups. In addition, they were retarded apparently in Leptin + shR-OB-Rb group after 30 days. As shown in Fig. 2H–J, the growth of xenograft tumors in shR-OB-Rb group obviously less than the other groups.

Signaling pathways of JAK2/STAT3/SOCS3 were involved in leptin stimulation

The leptin action was by signaling via JAK2 and phosphorylation of STAT3 or other pathways such as SOCS3 (13,14). In GBC-SD cells, we found that leptin increased JAK2 expression levels and STAT3 phosphorylation, and decreased SOCS3 expression levels. Such an effect was blocked by shR-OB-Rb treatment (Fig. 3A and B). The immunofluorescence experiments also confirmed similar results (Fig. 3C). These results indicate that the JAK2/STAT3/SOCS3 pathway is involved in leptin-induced migration of human GBC cells.

shR-OB-Rb downregulated MMP-3/9 activity and expression of VEGF-C/D increased by leptin

Numerous studies have mechanistically associated the invasive and metastasis ability of cancer cells with expression of VEGF factors (15) and activation of MMP family (16). To understand the mechanism by which leptin promoted the invasiveness and migration of GBC-SD cells, we investigated the expression of VEGF-C/D and activity of MMP-9 in GBC-SD cells treated with leptin and/or transfected with shR-OB-Rb. As shown in Fig. 4A, gelatin zymography assay results show leptin increases MMP-3 and MMP-9 activity in GBC-SD cells, which are attenuated by transfecting shR-OB-Rb. ELISA also confirmed similar result in expression of VEGF-C and VEGF-D (Fig. 4B). Then we detected activity of MMP-3/9 and expression of VEGF-C/D in vivo, which were confirmed by western blot (Fig. 4C and D) and immunofluorescence (Fig. 4E–J) analysis. Taken together, these data suggested that leptin upregulated VEGF-C/D levels and activated MMP-3/9 in vivo and in vitro.

Discussion

Our results indicate that leptin and OB-Rb are expressed at a high level in GBC patients, compare with normal gallbladder tissue. In addition, a high level of leptin and OB-Rb is a poor prognostic marker for GBC patient. Furthermore, this study shows for the first time that leptin and OB-Rb mediates migration of human gallbladder cancer (GBC) cells. We show that in vitro: i) leptin stimulates growth and migration of GBC-SD cells; ii) the enhancement of GBC-SD cell growth by leptin is associated with G2/M cell cycle arrest, iii) activity of MMP-3/9 and expression VEGF-C/D is determined. iv) JAK2/STAT3/SOCS3 pathway is involved in this process. Moreover, in vivo v) genetic ablation of leptin-mediated signaling enhanced cancer growth in an animal model of GBC. Moreover, these effect could all be attenuated by OB-Rb receptor shRNA.

Several studies have shown strong epidemiologic evidence suggesting the existence of a close link between obesity, a clinical condition characterized by high levels of circulating leptin (17) and a multitude of cancers, such as prostate (18), mammary (19), endometrial (20), hepatocellular (21), colon (22), pancreatic (23), adenocarcinoma of esophagus (24), and cholangiocarcinoma (25). In this study, consistent data show that leptin enhanced progression of GBC. Leptin is usually related to binding with its receptor OB-Rb, which belongs to the cytokine receptor superfamily (26). It has been reported that human cancer cells expressed OB-Rb and other OB-Rs leptin receptors (27). However, the role of OB-Rb in human GBC is mostly unknown. In this setting, we found that cell migration and integrin upregulation induced by leptin were attenuated by OB-Rb knockdown. Upon leptin binding, OB-Rb could activate JAK2, which in turn phosphorylated tyrosine residues in the receptor tails, leading to the recruitment and activation of STAT-3 (28). The leptin receptor, through the activation of JAK2, was also able to downregulate SOCS3 proteins and stimulate the downstream signaling pathway (13,29). Herein, we used the OB-Rb shRNA to determine its role and found that it inhibited leptin-induced migration and JAK2/STAT-3/SOCS3 upregulation, indicating the possible involvement of OB-Rb in leptin-induced cell growth and migration in GBC-SD cells.

Human MMPs, also known as collagenase, are a matrix metalloproteinase originally identified in breast carcinomas (30). Recent studies have revealed that this enzyme was also produced by a variety of malignant tumors (31). In all of the cases, the expression of MMPs was associated with aggressive tumors. GBC is known to have a high potential for invasion and metastasis. In the present study, we compared the expression levels of MMP-3 and MMP-9 in GBC cells with and without leptin and/or OB-Rb shRNA treatment. We found MMP-3/9 was activated by treating with leptin, and downregulated by OB-Rb interruption suggesting that leptin's regulation of MMPs is in a tissue-specific manner.

In addition, cell migration, and metastatic colonization must be successful with angiogenesis, which is essential for metastasized tumors in distant sites (32). Thus, we detected VEGF-C/D, which is the most important mediator of tumor angiogenesis, inducing formation of new blood vessels (33). Our study indicated that leptin could increase VEGF-C/D expression in vivo and in vitro. However, further research is necessary to clarify the mechanism underlying this process. Considering previous studies and the results described above, leptin and its receptor OB-Rb could be a potential therapeutic target for GBC.

In summary, our data showed that leptin and its receptor OB-Rb may be implicate in growth and metastasis of gall-bladder carcinoma, which may involve the regulation of MMPs and VEGF family through SOCS3/JAK2/STAT3 pathways. Regulation of leptin and its receptor OB-Rb could serve as a promising intervention strategy for gene therapy of gallbladder carcinoma.

Acknowledgements

This work is supported by grants from the Basic Research for Application Fund of Yunnan China (nos. 2012FB050 and 2011FZ124). The work is also funded by National Natural Science Foundation of China (83160360).

References

1 

Dwivedi AN, Jain S and Dixit R: Gall bladder carcinoma: Aggressive malignancy with protean loco-regional and distant spread. World J Clin Cases. 3:231–244. 2015. View Article : Google Scholar : PubMed/NCBI

2 

Boutros C, Gary M, Baldwin K and Somasundar P: Gallbladder cancer: Past, present and an uncertain future. Surg Oncol. 21:e183–e191. 2012. View Article : Google Scholar : PubMed/NCBI

3 

Caldow Pilgrim CH, Groeschl RT, Quebbeman EJ and Gamblin TC: Recent advances in systemic therapies and radiotherapy for gallbladder cancer. Surg Oncol. 22:61–67. 2013. View Article : Google Scholar : PubMed/NCBI

4 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2015. CA Cancer J Clin. 65:5–29. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Halaas JL, Gajiwala KS, Maffei M, Cohen SL, Chait BT, Rabinowitz D, Lallone RL, Burley SK and Friedman JM: Weight-reducing effects of the plasma protein encoded by the obese gene. Science. 269:543–546. 1995. View Article : Google Scholar : PubMed/NCBI

6 

Vona-Davis L and Rose DP: Adipokines as endocrine, paracrine, and autocrine factors in breast cancer risk and progression. Endocr Relat Cancer. 14:189–206. 2007. View Article : Google Scholar : PubMed/NCBI

7 

Surmacz E: Obesity hormone leptin: A new target in breast cancer? Breast Cancer Res. 9:3012007.PubMed/NCBI

8 

Fan Y, Gan Y, Shen Y, Cai X, Song Y, Zhao F, Yao M, Gu J and Tu H: Leptin signaling enhances cell invasion and promotes the metastasis of human pancreatic cancer via increasing MMP-13 production. Oncotarget. 6:16120–16134. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Dong Z, Xu X, Du L, Yang Y, Cheng H, Zhang X, Li Z, Wang L, Li J, Liu H, et al: Leptin-mediated regulation of MT1-MMP localization is KIF1B dependent and enhances gastric cancer cell invasion. Carcinogenesis. 34:974–983. 2013. View Article : Google Scholar : PubMed/NCBI

10 

Kloek C, Haq AK, Dunn SL, Lavery HJ, Banks AS and Myers MG Jr: Regulation of Jak kinases by intracellular leptin receptor sequences. J Biol Chem. 277:41547–41555. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method. Methods. 25:402–408. 2001. View Article : Google Scholar

12 

Barone I, Catalano S, Gelsomino L, Marsico S, Giordano C, Panza S, Bonofiglio D, Bossi G, Covington KR, Fuqua SA, et al: Leptin mediates tumor-stromal interactions that promote the invasive growth of breast cancer cells. Cancer Res. 72:1416–1427. 2012. View Article : Google Scholar : PubMed/NCBI

13 

Frühbeck G: Intracellular signalling pathways activated by leptin. Biochem J. 393:7–20. 2006. View Article : Google Scholar :

14 

Yang R and Barouch LA: Leptin signaling and obesity: Cardiovascular consequences. Circ Res. 101:545–559. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Goel HL and Mercurio AM: VEGF targets the tumour cell. Nat Rev Cancer. 13:871–882. 2013. View Article : Google Scholar : PubMed/NCBI

16 

Gong Y, Chippada-Venkata UD and Oh WK: Roles of matrix metalloproteinases and their natural inhibitors in prostate cancer progression. Cancers (Basel). 6:1298–1327. 2014. View Article : Google Scholar

17 

Garofalo C and Surmacz E: Leptin and cancer. J Cell Physiol. 207:12–22. 2006. View Article : Google Scholar

18 

Saglam K, Aydur E, Yilmaz M and Göktaş S: Leptin influences cellular differentiation and progression in prostate cancer. J Urol. 169:1308–1311. 2003. View Article : Google Scholar : PubMed/NCBI

19 

O'brien SN, Welter BH and Price TM: Presence of leptin in breast cell lines and breast tumors. Biochem Biophys Res Commun. 259:695–698. 1999. View Article : Google Scholar : PubMed/NCBI

20 

Petridou E, Belechri M, Dessypris N, Koukoulomatis P, Diakomanolis E, Spanos E and Trichopoulos D: Leptin and body mass index in relation to endometrial cancer risk. Ann Nutr Metab. 46:147–151. 2002. View Article : Google Scholar : PubMed/NCBI

21 

Saxena NK, Sharma D, Ding X, Lin S, Marra F, Merlin D and Anania FA: Concomitant activation of the JAK/STAT, PI3K/AKT, and ERK signaling is involved in leptin-mediated promotion of invasion and migration of hepatocellular carcinoma cells. Cancer Res. 67:2497–2507. 2007. View Article : Google Scholar : PubMed/NCBI

22 

Bahceci M, Tuzcu A, Akkus M, Yaldiz M and Ozbay A: The effect of high-fat diet on the development of obesity and serum leptin level in rats. Eat Weight Disord. 4:128–132. 1999. View Article : Google Scholar

23 

Calle EE, Rodriguez C, Walker-Thurmond K and Thun MJ: Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N Engl J Med. 348:1625–1638. 2003. View Article : Google Scholar : PubMed/NCBI

24 

Somasundar P, Riggs D, Jackson B, Vona-Davis L and McFadden DW: Leptin stimulates esophageal adenocarcinoma growth by nonapoptotic mechanisms. Am J Surg. 186:575–578. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Fava G, Alpini G, Rychlicki C, Saccomanno S, DeMorrow S, Trozzi L, Candelaresi C, Venter J, Di Sario A, Marzioni M, et al: Leptin enhances cholangiocarcinoma cell growth. Cancer Res. 68:6752–6761. 2008. View Article : Google Scholar : PubMed/NCBI

26 

Yang W-H, Liu S-C, Tsai C-H, Fong YC, Wang SJ, Chang YS and Tang CH: Leptin induces IL-6 expression through OBRl receptor signaling pathway in human synovial fibroblasts. PLoS One. 8:e755512013. View Article : Google Scholar : PubMed/NCBI

27 

Otvos L Jr and Surmacz E: Targeting the leptin receptor: A potential new mode of treatment for breast cancer. Expert Rev Anticancer Ther. 11:1147–1150. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Oh HK, Choi YS, Yang Y-I, Kim J-H, Leung PC and Choi J-H: Leptin receptor is induced in endometriosis and leptin stimulates the growth of endometriotic epithelial cells through the JAK2/STAT3 and ERK pathways. Mol Hum Reprod. 19:160–168. 2013. View Article : Google Scholar

29 

Benomar Y, Roy AF, Aubourg A, Djiane J and Taouis M: Cross down-regulation of leptin and insulin receptor expression and signalling in a human neuronal cell line. Biochem J. 388:929–939. 2005. View Article : Google Scholar : PubMed/NCBI

30 

Shuman Moss LA, Jensen-Taubman S and Stetler-Stevenson WG: Matrix metalloproteinases: Changing roles in tumor progression and metastasis. Am J Pathol. 181:1895–1899. 2012. View Article : Google Scholar : PubMed/NCBI

31 

Deryugina EI and Quigley JP: Tumor angiogenesis: MMP-mediated induction of intravasation- and metastasis-sustaining neovasculature. Matrix Biol. 44–46:94–112. 2015. View Article : Google Scholar

32 

Valastyan S and Weinberg RA: Tumor metastasis: Molecular insights and evolving paradigms. Cell. 147:275–292. 2011. View Article : Google Scholar : PubMed/NCBI

33 

Hoeben A, Landuyt B, Highley MS, Wildiers H, Van Oosterom AT and De Bruijn EA: Vascular endothelial growth factor and angiogenesis. Pharmacol Rev. 56:549–580. 2004. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2016
Volume 49 Issue 1

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zou H, Liu Y, Wei D, Wang T, Wang K, Huang S, Liu L, Li Y, Ge J, Li X, Li X, et al: Leptin promotes proliferation and metastasis of human gallbladder cancer through OB-Rb leptin receptor. Int J Oncol 49: 197-206, 2016
APA
Zou, H., Liu, Y., Wei, D., Wang, T., Wang, K., Huang, S. ... Wang, L. (2016). Leptin promotes proliferation and metastasis of human gallbladder cancer through OB-Rb leptin receptor. International Journal of Oncology, 49, 197-206. https://doi.org/10.3892/ijo.2016.3530
MLA
Zou, H., Liu, Y., Wei, D., Wang, T., Wang, K., Huang, S., Liu, L., Li, Y., Ge, J., Li, X., Zhu, H., Wang, L., Zhao, S., Zhang, X., Wang, L."Leptin promotes proliferation and metastasis of human gallbladder cancer through OB-Rb leptin receptor". International Journal of Oncology 49.1 (2016): 197-206.
Chicago
Zou, H., Liu, Y., Wei, D., Wang, T., Wang, K., Huang, S., Liu, L., Li, Y., Ge, J., Li, X., Zhu, H., Wang, L., Zhao, S., Zhang, X., Wang, L."Leptin promotes proliferation and metastasis of human gallbladder cancer through OB-Rb leptin receptor". International Journal of Oncology 49, no. 1 (2016): 197-206. https://doi.org/10.3892/ijo.2016.3530