Expression of LPA2 is associated with poor prognosis in human breast cancer and regulates HIF-1α expression and breast cancer cell growth

  • Authors:
    • Min Li
    • Dong Xiao
    • Jia Zhang
    • Hangying Qu
    • Ya Yang
    • Yan Yan
    • Xu Liu
    • Jizhao Wang
    • Lin Liu
    • Jiansheng Wang
    • Xiaoyi Duan
  • View Affiliations

  • Published online on: October 26, 2016     https://doi.org/10.3892/or.2016.5206
  • Pages: 3479-3487
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Breast cancer (BC) generally exhibits poor prognosis owing to its invasive and metastatic characteristics and is the leading cause of cancer-related deaths in women worldwide. Lysophosphatidic acid receptor 2 (LPA2) and hypoxia inducible factor-1α (HIF-1α) were found to be correlated with BC invasion and metastasis, respectively. However, the effect of LPA2 on BC in Chinese women has not yet been reported, nor have the overall survival and prognostic significance of LPA2 or its association with HIF-1α in BC. In the present study, we assessed the effect of LPA2 on HIF-1α expression, on overall survival and prognostic significance in BC in Chinese women, and on cell proliferation, migration and invasion in MCF-7 BC cells in vitro. The data showed that LPA2 and HIF-1α protein expression levels were higher in the BC tissue specimens and that LPA2 expression was significantly associated with menopausal status (postmenopausal), nodal metastasis and tumor-node-metastasis (TNM) stage, whereas HIF-1α expression was significantly associated with estrogen receptor status, nodal metastasis and TNM stage. Furthermore, LPA2 and HIF-1α expression were positively correlated (r=0.562; P<0.001). LPA2 and HIF-1α protein levels were associated with shorter overall patient survival according to univariate analysis (log-rank test; P<0.001), and nodal metastasis, TNM stage, LPA2 and HIF-1α expression were independent prognostic predictors in patients as determined by multivariate analysis (P<0.05). In vitro, the data suggested that LPA2 affected HIF-1α expression and LPA2 overexpression or knockdown resulted in increased or inhibited tumor cell proliferation, migration and invasion, respectively. In conclusion, our data demonstrated that LPA2 expression was associated with HIF-1α expression and that a high level of LPA2 was associated with shorter overall survival and was an independent prognostic predictor for BC in Chinese women. Furthermore, LPA2 showed the ability to regulate the cell proliferation, migration and invasion of BC cells.

Introduction

Cancer represents a major public health issue worldwide and constitutes an enormous burden on society (1,2). Breast cancer (BC) is the most commonly diagnosed cancer and the leading cause of cancer-related deaths among women (3). Although current therapies have proven effective in reducing primary tumors, the 5-year survival rate of BC is still poor mainly due to its intensive tendency for invasion and metastasis (4). Thus, further study is needed to identify and evaluate biomarkers for prognostic prediction as well as to develop novel strategies for the prevention and control of BC.

Lysophosphatidic acid (LPA) receptors are specific G protein-coupled receptors that bind with lysophosphatidic acid to mediate a variety of biological processes such as cell proliferation, migration, invasion and differentiation (5). At least six LPA receptors (LPA1-6) have been identified (6). Notably, LPAs have been correlated with the invasive and metastatic behaviors of several malignant cancers including BC (79). LPA2 is associated with a more malignant phenotype in carcinomas of the breast (10). The hypoxic microenvironment of a tumor also serves as an independent predictor of a poor prognosis in various types of cancer including BC (11); the effects of such hypoxia are mediated by the transcription factor hypoxia inducible factor-1α (HIF-1α) (12). Consistent with this, HIF-1α is closely associated with the occurrence and progression of tumors (1315), and HIF-1α activation represents a final common event in the pathogenesis of different types of tumors 12,1618. Thus, HIF-1α is considered to be specifically responsible for hypoxia-mediated progression of cancer (19,20). However, the association between LPA2 and HIF-1α in BC has not yet been described.

To address these issues, in the present study, we first detected the expression of LPA2 and HIF-1α in BC tissue specimens to assess their association with patient clinicopathological and survival data. Subsequently, we assessed the role of LPA2 and HIF-1α in BC in vitro and explored the effects of LPA2 overexpression or knockdown on HIF-1α expression and cell proliferation, migration and invasion.

Materials and methods

Tissue specimens

Tissue specimens from 156 patients with BC were collected from consecutive surgical cases at the Department of Surgical Oncology, the 3201st Hospital of Shaanxi Province, between 2005 and 2009. The histological type for all cases was breast ductal cancer. All tissues were pathologically examined. The patients were all females, ranging between the ages of 26 and 77 years. The clinicopathological data of the patients are documented in Table II. None of the 156 patients received neoadjuvant or adjuvant chemotherapy before surgery. The present study was approved by the Protection of Human Subjects Committee of the 3201st Hospital and complies with the Declaration of Helsinki.

Table II.

Association of LPA2 and HIF-1α expression with clinicopathological data from the BC patients.

Table II.

Association of LPA2 and HIF-1α expression with clinicopathological data from the BC patients.

LPA2 HIF-1α


VariableNHighLowP-valueHighLowP-value
Age (years)
  <50794831 4039
  ≥507743340.53443340.514
Menopausal status
  Premenopausal632934 3132
  Postmenopausal9362310.01052410.410
Tumor site
  Left875136 4641
  Right6940290.93537320.926
Tumor diameter (cm)
  ≤2411922 2120
  >2 to ≤5986335 5147
  >517980.1311160.600
ER status
  Negative704228 4525
  Positive8649370.70338480.012
PR status
  Negative704030 3634
  Positive8651350.78647390.688
Her-2 status
  Negative1116645 6051
  Positive4525200.65423220.739
Nodal metastasis
  Negative502228 1238
  Positive10669370.0137135<0.001
Histologic grade
  1322111 1418
  2784038 3939
  34630160.20330160.126
TNM stage
  I23914 815
  II603228 2832
  III483018 2622
  IV252050.0262140.003

[i] ER, estrogen receptor; PR, progesterone receptor; Her-2, human epidermal growth factor receptor-2; TNM, tumor-node-metastasis.

Immunohistochemical (IHC) staining

Formalin-fixed paraffin-embedded tumor sections (5-µm thickness) were analyzed by IHC staining using an anti-LPA2 rabbit polyclonal antibody (ab38322), anti-HIF-1α (ab82832) (1:200 dilution; Abcam, Cambridge, MA, USA), and a biotin-conjugated secondary antibody. The staining was performed following the SP kit procedure (Golden Bridge International, Beijing, China). As a control, the primary antibody was replaced by phosphate-buffered saline (PBS). IHC staining results were independently assessed by two pathologists in a semi-quantitative manner, and graded in accordance with an immunoreactive score (IRS): IRS is equal to the percentage of positive cells (0, negative; 1, <10%; 2, 10–50%; 3, 51–80%; 4, >80%) multiplied by the staining intensity (1, weak; 2, moderate; 3, strong).

The scores ranged from 0 to 12. A score of 6 or higher was considered high expression and below 6 was considered low expression. A score of 0 represented a negative staining result.

Cell culture

The non-tumorigenic MCF-10A and BC MCF-7 cell lines were obtained from the Cell Bank of Shanghai (Shanghai, China) and were cultured in Dulbecco's modified Eagle's medium (HyClone, Logan, UT, USA) supplemented with 10% heat-inactivated fetal bovine serum (Hzsjq Co. Ltd., Hangzhou, China) at 37°C in a humidified incubator under an atmosphere of 5% carbon dioxide. All experiments were performed with cells in the logarithmic phase of growth.

Plasmid and siRNA transfection

The plasmid (pENTR223-LPA2) was obtained from the DNASU Plasmid Repository (Tempe, AZ, USA) and small interfering RNA (siRNA) molecules targeting human LPA2 (si-LPA2) were obtained from Santa Cruz Biotechnology (sc-39926; Santa Cruz, CA, USA). The MCF-7 cells were grown and transfected with the negative control (NC) (GenePharma Co., Ltd., Shanghai, China), pENTR223-LPA2 or si-LPA2 using Lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA) according to the manufacturer's protocol. MCF-7 cells were cultured in normal medium as a control group (normal group).

Cell proliferation assessment

Following plasmid and siRNA transfection for 48 h, the colorimetric 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay was used to assess the proliferation of the MCF-7 cells. The MCF-7 cells (100 µl) were seeded at a density of 4×104 cells/ml in 96-well plates and were cultured for 24, 48 and 72 h. An aliquot of 10 µl MTT solution in PBS (5 mg/ml) was added to each well and the plates were incubated for 4 h at 37°C. Then, 150 µl dimethyl sulfoxide (Sigma, St. Louis, MO, USA) was added to each well and the plates were shaken for 15 min before reading the optical density of each well at 570 nm on a microplate reader. MCF-7 cells were cultured in normal medium as a control group. All experiments were conducted in triplicate.

Cell migration and invasion assays

Following plasmid and siRNA transfection for 48 h, migration and invasion assays were conducted in MCF-7 cells using Transwell plates with 8-µm pore size membranes (Millipore Inc., Billerica, MA, USA) as previously described (21). After incubation for 12 h (for migration assays) or 24 h (for invasion assays), cells remaining in the upper side of the filter were removed with cotton swabs. The cells attached on the lower surface were fixed, stained using crystal violet and washed with PBS. Cells were counted in 5-high power fields/membrane. The results are presented as the mean number of cells that migrated/field/membrane. MCF-7 cells were cultured in normal medium as a control group. All experiments were conducted in triplicate.

Western blot analyses

Following plasmid and siRNA transfection of MCF-7 cells for 48 h, the proteins were extracted according to standard procedures of RIPA lysis buffer kit (Santa Cruz Biotechnology). Equal amounts of protein samples were separated onto a 10% sodium dodecyl sulfate-polyacrylamide gel using sodium dodecyl sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and were then transferred onto a polyvinylidene difluoride (PVDF) membrane (Millipore). The membranes were examined with the following specific primary antibodies: anti-LPA2 rabbit polyclonal or anti-HIF-1α rabbit polyclonal (1:1,000 dilution) or anti-β-actin rabbit polyclonal antibodies (bs-0061R; 1:1,000 dilution) followed by the conjugated secondary antibody goat-anti-rabbit IgG (1:3,000 dilution) (both from Bioss, Woburn, MA, USA). The reactions were visualized using an enhanced chemiluminescence (ECL) reagent (Millipore). The band intensities of the western blotting were measured by densitometry using Quantity One software (Bio-Rad Laboratories, Hercules, CA, USA). MCF-7 cells were cultured in normal medium as a control group (normal group). The protein levels were normalized to those of β-actin protein level used as a loading control.

Statistical analysis

The Chi-square test was used to analyze the association of LPA2 and HIF-1α expression with the clinicopathological variables. The Spearman's rank correlation coefficient test was performed to associate LPA2 expression with HIF-1α expression in tissue specimens. Overall survival was defined as the time from the date of surgery to the date of the last follow-up or of patient death. Survival curves were calculated using the Kaplan-Meier method and compared using the log-rank test. Multivariate analysis using Cox's proportional hazard model was used to assess prognostic factors. The Student's t-test or one-way ANOVA was used to compare normally distributed variables. P<0.05 was considered significant. All analyses were performed using SPSS software version 18.0 (IBM Corp., New York, NY, USA).

Results

Differential LPA2 and HIF-1α protein expression is associated with the clinicopathological data of the patients with BC

We first assessed the protein expression of LPA2 and HIF-1α using IHC in 156 BC tissue specimens (Fig. 1A as a negative control) and found that the LPA2 protein was localized in both the cell membrane and cytoplasm (Fig. 1B) whereas the HIF-1α protein was predominantly localized in the nuclei (Fig. 1C). The expression levels of these two proteins significantly differed between the cancer and para-cancer normal tissues (Table I). High expression of the LPA2 protein was significantly associated with menopausal status, nodal metastasis and TNM stage, yet not with age, tumor site, tumor diameter, estrogen or progesterone receptor and Her-2 status, and histologic grade (Table II). In comparison, HIF-1α expression was associated with estrogen receptor status, nodal metastasis and TNM stage, but not with age, menopausal status, tumor site, tumor diameter, progesterone receptor and Her-2 status and grade. In addition, LPA2 expression was associated with HIF-1α expression in the BC tissues. Specifically, Spearman's rank correlation coefficient test showed that there were 91 cancer cases expressing a high level of LPA2 vs. 83 cases with HIF-1α expression (Table III), revealing a positive association (r=0.562; P<0.001).

Table I.

Differential expression of LPA2 and HIF-1α proteins in breast cancer (BC) tissues vs. para-cancer normal tissues.

Table I.

Differential expression of LPA2 and HIF-1α proteins in breast cancer (BC) tissues vs. para-cancer normal tissues.

LPA2 HIF-1α


+P-value+P-value
Cancer11046 11640
Para-carcinoma30126<0.00133123<0.001
Total140172 149163

[i] BC, breast cancer; LPA2, lysophosphatidic acid receptor 2; HIF-1α, hypoxia inducible factor-1α.

Table III.

Association of LPA2 and HIF-1α expression.

Table III.

Association of LPA2 and HIF-1α expression.

LPA2

HighLowrP-value
HIF-1α
  High7013
  Low21520.562<0.001

[i] LPA2, lysophosphatidic acid receptor 2; HIF-1α, hypoxia inducible factor-1α.

Association of LPA2 and HIF-1α expression with the survival of patients with BC

The Kaplan-Meier curves and the log-rank test data showed that patients with a high LPA2-expressing tumor had a statistically significantly poorer overall survival compared to those with a low LPA2-expressing tumor (Fig. 2A and Table IV) of 41 months [95% confidence interval (CI), 38.053–45.531] vs. 55 months (95% CI, 53.497–58.356; P<0.001). The same was true for HIF-1α expression (Fig. 2B and Table IV). Furthermore, tumor diameter, nodal metastasis and TNM stage were all associated with poor overall patient survival (P<0.05). The multivariate Cox regression model showed that nodal metastasis [P=0.007, hazard ratio (HR)=7.516, 95% CI, 1.748–32.327], TNM stage (P<0.001, HR=2.436, 95% CI, 1.703–3.486), LPA2 expression (P=0.036 HR=2.294, 95% CI, 1.054–4.992) and HIF-1α (P=0.029, HR=2.369, 95% CI, 1.090–5.150) were independent prognostic indicators for the survival of patients with BC (Table V).

Table IV.

Univariate analysis of overall survival.

Table IV.

Univariate analysis of overall survival.

Overall survival

VariableNMedian95% CIP-value
LPA2
  High9141.00±1.90838.053–45.531
  Low6555.00±1.23953.497–58.356<0.001
HIF-1α
  High8339.00±2.01735.839–43.746
  Low7356.00±0.98154.728–58.575<0.001
Age (years)
  <507947.00±1.89144.052–51.465
  ≥507747.00±1.92143.837–51.3680.920
Menopausal status
  Premenopausal6349.00±2.12845.431–53.771
  Postmenopausal9346.00±1.72942.993–49.7690.123
Tumor site
  Left8748.00±1.72544.981–51.742
  Right6946.00±2.13042.650–50.9990.597
Tumor diameter (cm)
  ≤24154.00±1.86051.191–58.480
  >2 to ≤59845.00±1.77641.581–48.544
  >51745.00±4.25737.182–53.8690.007
ER status
  Negative7046.00±2.19342.581–51.179
  Positive8648.00±1.66745.067–51.6010.978
PR status
  Negative7048.00±1.98644.959–52.746
  Positive8646.00±1.82743.148–50.3100.363
Her-2 status
  Negative11146.00±1.62743.092–49.468
  Positive4551.00±2.31446.603–55.6740.111
Nodal metastasis
  Negative5058.00±1.05356.544–60.672
  Positive10642.00±1.70439.187–45.868<0.001
Histologic grade
  13253.00 ±2.56948.704–58.775
  27845.00±2.00441.984–49.840
  34646.00±2.33341.895–51.0410.057
TNM stage
  I2358.00±1.15756.550–61.084
  II6053.00±1.86949.397–56.723
  III4845.00±2.32040.795–49.888
  IV2529.00±2.81923.496–34.547<0.001

[i] CI, confidence interval; LPA2, lysophosphatidic acid receptor 2; HIF-1α, hypoxia inducible factor-1α; ER, estrogen receptor; PR, progesterone receptor; Her-2, human epidermal growth factor receptor-2; TNM, tumor-node-metastasis.

Table V.

Multivariate Cox proportional hazard analysis of overall survival.

Table V.

Multivariate Cox proportional hazard analysis of overall survival.

Overall survival

VariableHR95% CIP-value
Age (years)1.3890.755–2.5560.290
Menopausal status1.2810.651–2.5200.473
Tumor site0.9620.555–1.6670.890
Tumor diameter (cm)1.3810.850–2.2420.192
ER status0.9310.528–1.6420.805
PR status1.2880.753–2.2010.355
Her-2 status0.6970.341–1.4260.323
Nodal metastasis7.5161.748–32.3270.007
Grade0.8030.535–1.2070.292
TNM stage2.4361.703–3.4860.000
LPA22.2941.054–4.9920.036
HIF-1α2.3691.090–5.1500.029

[i] HR, hazard ratio; CI, confidence interval; ER, estrogen receptor; PR, progesterone receptor; Her-2, human epidermal growth factor receptor-2; TNM, tumor-node-metastasis; LPA2, lysophosphatidic acid receptor 2; HIF-1α, hypoxia inducible factor-1α.

LPA2 and HIF-1α are expressed in human BC cell lines

Our ex vivo data demonstrated that LPA2 and HIF-1α protein expression were positively correlated (r=0.562) and were associated with a poor overall survival; however, this result had not previously been reported with regard to both LPA2 and HIF-1α in BC. Therefore, we further assessed the role of LPA2 and HIF-1α in a human BC cell line. We first explored the expression levels of LPA2 and HIF-1α in non-tumorigenic MCF-10A and MCF-7 BC cells. As shown in Fig. 3, the levels of both LPA2 and HIF-1α were high in MCF-7 and low in MCF-10A as assessed by western blot analysis; this difference was statistically significant (P<0.001).

Effects of LPA2 overexpression in BC cells

At 48 h following transfection of MCF-7 cells with the plasmid (pENTR223-LPA2), western blot analysis showed that LPA2 protein was overexpressed in the LPA2-overexpressing cells compared to that in the normal and NC group (control groups) (P<0.001; Fig. 4A). Upon LPA2 overexpression, the level of HIF-1α was elevated in the MCF-7 cells compared to that in the controls (P<0.05; Fig. 4A). The MTT assay showed that overexpression of LPA2 promoted the proliferation of MCF-7 cells in a time-dependent manner (P<0.01 at 72 h; Fig. 4B). To further analyze the effect of LPA2 overexpression on MCF-7 cells, cell migration and invasion assays were conducted, demonstrating that these characteristics of MCF-7 cells were enhanced following LPA2 overexpression (P<0.001; Fig. 4C and D).

Effects of LPA2 knockdown on BC cells

The effects of LPA2 knockdown were directly inverse to those of its upregulation. At 48 h following transfection of MCF-7 cells with si-LPA2, both LPA2 (P<0.001; Fig. 5A) and HIF-1α (P<0.05; Fig. 5A) expression was downregulated compared to the controls as assessed by western blot analysis. Similarly, MCF-7 cell proliferation was inhibited in a time-dependent manner (P<0.05 at 72 h; Fig. 5B) and their migration and invasion were reduced following LPA2 downregulation (P<0.001 and P<0.01; Fig. 5C and D).

Discussion

The invasive and metastatic properties of breast cancer (BC) play a considerable role in its poor prognosis, requiring the development of novel strategies to combat its position as the leading cause of cancer-related deaths among women worldwide. The results of the present study illustrated the role of LPA2 in the regulation of HIF-1α expression and BC progression, suggesting its potential for BC diagnostic and therapeutic application to help facilitate the prevention and control of this disease.

LPA receptors are specific G protein-coupled receptors and multifunctional signaling molecules that control the proliferation, motility, and differentiation of many cell types including cancer cells (5,22). Although the expression and functions of LPA2 have been the subject of only a few studies, increased LPA2 expression has been reported in invasive BC (10,23). However, the study of the roles of LPA2 in BC has mainly focused on western populations; in contrast, data related to the effect of LPA2 on BC in Chinese women or to the overall survival and prognostic significance of LPA2 in BC are not available. In the present study, we first analyzed the association of LPA2 expression in BC tissue specimens with the clinicopathological and survival data of Chinese women. Our date showed that LPA2 was more highly expressed in BC tissues than in para-cancer tissues, consistent with some prior reports from other populations (8,10). We also found that LPA2 was upregulated more frequently in postmenopausal than in premenopausal women, suggesting that LPA2 overexpression was more strongly related to the carcinogenesis of postmenopausal BC, consistent with the study by Kitayama et al (10). In addition, our data showed that LPA2 overexpression was significantly associated with nodal metastasis and later clinical TNM stage and that patients with high LPA2-expressing tumors had a statistically significant poorer overall survival compared to those with tumors expressing low levels of LPA2. These results confirmed the validity of LPA2 as an independent prognostic indicator for patient survival in BC.

HIF-1α has a close association with tumor occurrence and progression and its activation represents a final common event in the pathogenesis of multiple tumor types (12,18,2427). Data from the present study showed that BC tumors exhibiting high HIF-1α expression correlated with statistically significant poorer overall patient survival compared to those with low levels of HIF-1α expression, which is consistent with other studies (28). The association between LPA2 and HIF-1α had not been previously reported; however, the results of the present study indicated that a high level of LPA2 expression was associated with a high level of HIF-1α expression in BC clinical tissue specimens, revealing a positive association (r=0.562, P<0.001). This suggested the existence of a potential gene regulatory effect between LPA2 and HIF-1α. In the present study we aimed to explore whether LPA2 was able to regulate HIF-1α in BC cells.

To address this issue, we performed in vitro experiments to assess the effect of LPA2 on HIF-1α expression in the MCF-7 BC cell line. Consistent with an earlier study, we found that both LPA2 and HIF-1α exhibited higher expression (MCF-7) than non-tumorigenic breast (MCF-10A) cells (8,29). In addition, we found when the level of LPA2 was artificially increased through exogenous overexpression, the level of HIF-1α was concurrently elevated compared to MCF-7 controls (P<0.05) and the proliferation (P<0.01), migration and invasion of MCF-7 cells (P<0.001) was enhanced. Conversely, we also found when the level of LPA2 was decreased via siRNA knockdown, HIF-1α also exhibited low expression compared to MCF-7 controls (P<0.05) and that MCF-7 proliferation (P<0.05), migration (P<0.001) and invasion (P<0.01) were also inhibited. Together, these data indicated that LPA2 was able to regulate HIF-1α expression and the proliferation, migration and invasion of BC cells.

In summary, the present study demonstrated that LPA2 was able to serve as an independent prognostic indicator for survival in BC and that high LPA2 expression was associated with poor overall patient survival among Chinese women. LPA2 and HIF-1α protein expression exhibited a positive association, and LPA2 showed the ability to regulate HIF-1α expression and proliferation, migration and invasion in BC cells. Thus, our data suggest that LPA2 may function as a useful prognostic marker of BC and may also represent a valid therapeutic target for BC management.

Acknowledgements

The present study was supported by a grant from the National Natural Science Foundation of China (no. 81471710).

References

1 

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

2 

Long J, Luo GP, Xiao ZW, Liu ZQ, Guo M, Liu L, Liu C, Xu J, Gao YT, Zheng Y, et al: Cancer statistics: Current diagnosis and treatment of pancreatic cancer in Shanghai, China. Cancer Lett. 346:273–277. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Torre LA, Bray F, Siegel RL, Ferlay J, Lortet-Tieulent J and Jemal A: Global cancer statistics, 2012. CA Cancer J Clin. 65:87–108. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Petekkaya I, Ayyildiz V, Kizilarslanoglu MC, Sahin U, Gezgen G, Roach EC, Karcaaltincaba M and Altundag K: Prognosis of breast cancer in patients with peritoneal metastasis. Breast. 21:420–421. 2012. View Article : Google Scholar : PubMed/NCBI

5 

Gotoh M, Fujiwara Y, Yue J, Liu J, Lee S, Fells J, Uchiyama A, Murakami-Murofushi K, Kennel S, Wall J, et al: Controlling cancer through the autotaxin-lysophosphatidic acid receptor axis. Biochem Soc Trans. 40:31–36. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Chun J, Hla T, Lynch KR, Spiegel S and Moolenaar WH: International Union of Basic and Clinical Pharmacology. LXXVIII. Lysophospholipid receptor nomenclature. Pharmacol Rev. 62:579–587. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Sedláková I, Vávrová J, Tošner J and Hanousek L: Lysophosphatidic acid (LPA) - a perspective marker in ovarian cancer. Tumour Biol. 32:311–316. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Sun K, Cai H, Duan X, Yang Y, Li M, Qu J, Zhang X and Wang J: Aberrant expression and potential therapeutic target of lysophosphatidic acid receptor 3 in triple-negative breast cancers. Clin Exp Med. 15:371–380. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Sun K, Duan X, Cai H, Liu X, Yang Y, Li M, Zhang X and Wang J: Curcumin inhibits LPA-induced invasion by attenuating RhoA/ROCK/MMPs pathway in MCF7 breast cancer cells. Clin Exp Med. 16:37–47. 2016. View Article : Google Scholar : PubMed/NCBI

10 

Kitayama J, Shida D, Sako A, Ishikawa M, Hama K, Aoki J, Arai H and Nagawa H: Over-expression of lysophosphatidic acid receptor-2 in human invasive ductal carcinoma. Breast Cancer Res. 6:R640–R646. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Bos R, van der Groep P, Greijer AE, Shvarts A, Meijer S, Pinedo HM, Semenza GL, van Diest PJ and van der Wall E: Levels of hypoxia-inducible factor-1alpha independently predict prognosis in patients with lymph node negative breast carcinoma. Cancer. 97:1573–1581. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Semenza GL: Targeting HIF-1 for cancer therapy. Nat Rev Cancer. 3:721–732. 2003. View Article : Google Scholar : PubMed/NCBI

13 

Span PN and Bussink J: Biology of hypoxia. Semin Nucl Med. 45:101–109. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Fransén K, Fenech M, Fredrikson M, Dabrosin C and Söderkvist P: Association between ulcerative growth and hypoxia inducible factor-1alpha polymorphisms in colorectal cancer patients. Mol Carcinog. 45:833–840. 2006. View Article : Google Scholar : PubMed/NCBI

15 

Putra AC, Tanimoto K, Arifin M and Hiyama K: Hypoxia-inducible factor-1α polymorphisms are associated with genetic aberrations in lung cancer. Respirology. 16:796–802. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Amelio I and Melino G: The ‘Sharp’ blade against HIF-mediated metastasis. Cell Cycle. 11:4530–4535. 2012. View Article : Google Scholar : PubMed/NCBI

17 

Piccolo S, Enzo E and Montagner M: p63, Sharp1, and HIFs: Master regulators of metastasis in triple-negative breast cancer. Cancer Res. 73:4978–4981. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Ke Q and Costa M: Hypoxia-inducible factor-1 (HIF-1). Mol Pharmacol. 70:1469–1480. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Bernaudin M, Tang Y, Reilly M, Petit E and Sharp FR: Brain genomic response following hypoxia and re-oxygenation in the neonatal rat. Identification of genes that may contribute to hypoxia-induced ischemic tolerance. J Biol Chem. 277:39728–39738. 2002. View Article : Google Scholar : PubMed/NCBI

20 

Badowska-Kozakiewicz AM, Budzik MP and Przybylski J: Hypoxia in breast cancer. Pol J Pathol. 66:337–346. 2015. View Article : Google Scholar : PubMed/NCBI

21 

Huanna T, Tao Z, Xiangfei W, Longfei A, Yuanyuan X, Jianhua W, Cuifang Z, Manjing J, Wenjing C, Shaochuan Q, et al: GALNT14 mediates tumor invasion and migration in breast cancer cell MCF-7. Mol Carcinog. 54:1159–1171. 2015. View Article : Google Scholar : PubMed/NCBI

22 

Choi JW, Herr DR, Noguchi K, Yung YC, Lee CW, Mutoh T, Lin ME, Teo ST, Park KE, Mosley AN, et al: LPA receptors: Subtypes and biological actions. Annu Rev Pharmacol Toxicol. 50:157–186. 2010. View Article : Google Scholar : PubMed/NCBI

23 

Liu S, Umezu-Goto M, Murph M, Lu Y, Liu W, Zhang F, Yu S, Stephens LC, Cui X, Murrow G, et al: Expression of autotaxin and lysophosphatidic acid receptors increases mammary tumorigenesis, invasion, and metastases. Cancer Cell. 15:539–550. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Lee JW, Bae SH, Jeong JW, Kim SH and Kim KW: Hypoxia-inducible factor (HIF-1)α: Its protein stability and biological functions. Exp Mol Med. 36:1–12. 2004. View Article : Google Scholar : PubMed/NCBI

25 

Birner P, Gatterbauer B, Oberhuber G, Schindl M, Rössler K, Prodinger A, Budka H and Hainfellner JA: Expression of hypoxia-inducible factor-1 alpha in oligodendrogliomas: Its impact on prognosis and on neoangiogenesis. Cancer. 92:165–171. 2001. View Article : Google Scholar : PubMed/NCBI

26 

Lu H, Forbes RA and Verma A: Hypoxia-inducible factor 1 activation by aerobic glycolysis implicates the Warburg effect in carcinogenesis. J Biol Chem. 277:23111–23115. 2002. View Article : Google Scholar : PubMed/NCBI

27 

Montagner M, Enzo E, Forcato M, Zanconato F, Parenti A, Rampazzo E, Basso G, Leo G, Rosato A, Bicciato S, et al: SHARP1 suppresses breast cancer metastasis by promoting degradation of hypoxia-inducible factors. Nature. 487:380–384. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Bertout JA, Patel SA and Simon MC: The impact of O2 availability on human cancer. Nat Rev Cancer. 8:967–975. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Chen M, Towers LN and O'Connor KL: LPA2 (EDG4) mediates Rho-dependent chemotaxis with lower efficacy than LPA1 (EDG2) in breast carcinoma cells. Am J Physiol Cell Physiol. 292:C1927–C1933. 2007. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2016
Volume 36 Issue 6

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
Li M, Xiao D, Zhang J, Qu H, Yang Y, Yan Y, Liu X, Wang J, Liu L, Wang J, Wang J, et al: Expression of LPA2 is associated with poor prognosis in human breast cancer and regulates HIF-1α expression and breast cancer cell growth. Oncol Rep 36: 3479-3487, 2016
APA
Li, M., Xiao, D., Zhang, J., Qu, H., Yang, Y., Yan, Y. ... Duan, X. (2016). Expression of LPA2 is associated with poor prognosis in human breast cancer and regulates HIF-1α expression and breast cancer cell growth. Oncology Reports, 36, 3479-3487. https://doi.org/10.3892/or.2016.5206
MLA
Li, M., Xiao, D., Zhang, J., Qu, H., Yang, Y., Yan, Y., Liu, X., Wang, J., Liu, L., Wang, J., Duan, X."Expression of LPA2 is associated with poor prognosis in human breast cancer and regulates HIF-1α expression and breast cancer cell growth". Oncology Reports 36.6 (2016): 3479-3487.
Chicago
Li, M., Xiao, D., Zhang, J., Qu, H., Yang, Y., Yan, Y., Liu, X., Wang, J., Liu, L., Wang, J., Duan, X."Expression of LPA2 is associated with poor prognosis in human breast cancer and regulates HIF-1α expression and breast cancer cell growth". Oncology Reports 36, no. 6 (2016): 3479-3487. https://doi.org/10.3892/or.2016.5206