• Users Online: 680
  • Print this page
  • Email this page

 
Table of Contents
ORIGINAL ARTICLE
Year : 2023  |  Volume : 66  |  Issue : 2  |  Page : 111-117

Stanniocalcin 2 is induced by estrogen and promotes growth in endometrial cancer via AMPK pathway


Center for Reproductive Medicine, Department of Obstetrics, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang Province, China

Date of Submission05-Sep-2022
Date of Decision22-Nov-2022
Date of Acceptance01-Dec-2022
Date of Web Publication20-Apr-2023

Correspondence Address:
Dr. Qiqi Wang
Center for Reproductive Medicine, Department of Obstetrics, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, No. 158, Shangtang Road, Hangzhou City, Zhejiang Province
China
Login to access the Email id

Source of Support: None, Conflict of Interest: None


DOI: 10.4103/cjop.CJOP-D-22-00077

Rights and Permissions
  Abstract 


Stanniocalcin 2 (STC2) is identified as a glycosylated peptide hormone and estrogen-responsive gene in cancer cells. STC2 participates in angiogenesis, cell development, cytoprotection, and calcium and phosphate regulation during the development of cancer. The role of STC2 in endometrial cancer (EC) remains unclear. The data from the bioinformatic and immunohistochemical analysis showed that STC2 was upregulated in the EC tissues. The EC cells were treated with 17β-estradiol (E2), and 0.1 μmol/L E2 increased the expression of STC2 in the EC cells. E2 also increased cell viability, promoted proliferation, and inhibited apoptosis of EC. However, the knockdown of STC2 decreased cell viability, reduced proliferation, and promoted apoptosis of E2-stimulated EC. Moreover, silencing of STC2 attenuated E2-induced downregulation of phosphorylated-AMP-activated protein kinase (AMPK) in the EC cells. The loss of STC2 reduced E2-stimulated tumor growth EC in vivo. In conclusion, STC2 deficiency suppressed E2-stimulated proliferation and tumor growth of EC through the activation of AMPK signaling.

Keywords: AMP-activated protein kinase, apoptosis, endometrial cancer, estrogen, proliferation, stanniocalcin 2


How to cite this article:
Wang Q, Wang Q, Zhao Y. Stanniocalcin 2 is induced by estrogen and promotes growth in endometrial cancer via AMPK pathway. Chin J Physiol 2023;66:111-7

How to cite this URL:
Wang Q, Wang Q, Zhao Y. Stanniocalcin 2 is induced by estrogen and promotes growth in endometrial cancer via AMPK pathway. Chin J Physiol [serial online] 2023 [cited 2023 May 29];66:111-7. Available from: https://www.cjphysiology.org/text.asp?2023/66/2/111/374407




  Introduction Top


Endometrial cancer (EC) is a common gynecological malignancy worldwide with an increasing incidence.[1] EC includes Type I and Type II based on estrogen dependency, and Type I, with increased estrogen dependency, is the major type of EC.[2] Therapeutic strategies, including radiotherapy, chemotherapy, and standard surgery, have been widely used in the treatment of patients with early-stage EC.[3] The 5-year survival rate for patients with early-stage EC is approximately 80%, while the prognosis for patients with advanced EC remains poor.[4] Therefore, exploring the novel diagnostic and prognostic biomarkers of EC and understanding the underlying mechanisms involved in tumor progression are essential for the improvement of patients with advanced EC.[5]

Estrogen has been widely known as a risk factor for tumorigenesis of the endometrium.[6] Estrogen signaling through ERs (ERs), including ER-α and ER-β, is an oncogenic signaling in the development of EC.[7] Estrogen signaling enhanced cell proliferation, invasion, and epithelial–mesenchymal transformation of EC.[6],[7],[8] Estrogen also inhibited autophagy and promoted tumor growth of EC.[9] The suppression of estrogen-induced endometrial growth showed promising inhibitory effects on the EC progression.[7]

Stanniocalcin 2 (STC2) is identified as a glycosylated peptide hormone and plays a pivotal role in angiogenesis, cell development, cytoprotection, and calcium and phosphate regulation during the development of cancer.[10] A high expression of STC2 was associated with a poor prognosis of solid cancers,[11] and overexpression of STC2 promoted cell proliferation, metastasis, and chemo- or radioresistance of cancer cells.[12] Therefore, STC2 is a potential therapeutic target for distinct tumors.[13] It has been reported that STC2 was upregulated in EC, and an elevated expression of STC2 was associated with the recurrence and predicted poor prognosis in EC.[14] However, the role of STC2 in EC progression remains unclear. Moreover, estrogen exposure stimulated the upregulation of STC2 in breast cancer cells, and STC2 was regarded as an estrogen-responsive gene in tumorigenesis.[15] STC2 might also be triggered by estrogen and contributes to cell proliferation and metastasis of EC.

In this study, the effects of STC2 on cell proliferation and apoptosis of estrogen-induced EC were investigated, and the effect of STC2 on in vivo tumor growth of EC was also assessed.


  Materials and Methods Top


Bioinformatic analysis

The expression of STC2 in the EC tissues was analyzed by TIMER (https://cistrome.shinyapps.io/timer/), GEPIA (http://gepia.cancer-pku.cn/index.html), and UALCAN (http://ualcan.path.uab.edu/cgi-bin/TCGAExResultNew2.pl?genenam=MED27andctype=STAD).

Human tissues and immunohistochemistry

Patients with type I estrogen-related EC (n = 30) and type II estrogen-independent EC (n = 15) were recruited at the Medical Ethics Committee of Zhejiang Provincial People's Hospital. Tissues were surgically collected from these patients. Benign endometrial tissues were also harvested from the volunteers (n = 20). The experiment was approved by the Medical Ethics Committee of Zhejiang Provincial People's Hospital (approval no. 20220063) and in accordance with those of the 1964 Helsinki Declaration and its later amendments for the ethical research involving human subjects. Tissues were fixed with 10% formalin and embedded in paraffin. The paraffin-embedded tissues were sliced into 4-μm thick sections. Sections were deparaffinized, rehydrated, and then immersed in tris-ethylenediaminetetraacetic acid buffer containing 0.05% Tween 20. After incubated with 3% hydrogen peroxide and blocked in 5% dry milk, the sections were probed overnight with rabbit antihuman STC2 antibody (1:50; Santa Cruz Biotechnology, Santa Cruz, CA, USA). Sections were then incubated with horseradish peroxidase-labeled goat anti-rabbit secondary antibody (Santa Cruz Biotechnology, Santa Cruz, CA, USA). Slides were incubated with 3,3'-diaminobenzidine and counterstained with hematoxylin before measurement under microscope (Olympus, Tokyo, Japan).

Cell culture and transfection

Ishikawa and RL95-2 cells were grown in Dulbecco's Modified Eagle's Medium containing 10% fetal bovine serum (Gibco, Gaithersburg, MD, USA). Cells were transfected with sh-STC2 or shNC (RiboBio, Guangzhou, China) using lipofectamine 2000 (Invitrogen, Carlsbad, CA, USA) for 48 h and then treated with 0.001, 0.01, 0.1, 1, or 10 μmol/L 17β-estradiol (E2; Sigma-Aldrich, St. Louis, MO, USA) or 10 μmol/L compound C for 24 h.

Cell viability assay

Ishikawa and RL95-2 cells were seeded in 96-well plates for 24 h and cultured in medium for 24, 48, 72, or 96 h. Cells were treated with CCK8 solution (Beyotime, Beijing, China) for 2 h. Absorbance at 450 nm was examined through a microplate autoreader (Thermo Fisher, Waltham, MA, USA).

Colony formation assay

Ishikawa was seeded in 6-well plates and cultured in medium for 10 days. Cells were fixed in methanol and then stained with crystal violet. The colonies were photographed under the light microscope.

Flow cytometry

Ishikawa was harvested after trypsin digestion and resuspended in 100 μL binding buffer (KeyGEN BioTech, Jiangning, Nanjing, China). Cells were treated with 100 μg/mL propidium iodide and then incubated with fluorescein isothiocyanate-conjugated annexin V. Cells were analyzed by FACS flow cytometer (Attune, Life Technologies, Darmstadt, Germany).

Western blot

Cells and tumor tissues were lysed in RIPA buffer (KeyGEN BioTech, Jiangning, Nanjing, China) and centrifuged at 12,000 × g for 60 min to collect the protein samples. Samples were separated by SDS-PAGE, and electrotransferred onto PVDF membrane. Membranes were blocked with 5% skim milk and probed with primary antibodies, including anti-STC2, anti-β-actin (1:3000), antiphosphorylated-AMP-activated protein kinase (anti-p-AMPK), and anti-AMPK (1:4000). Membranes were incubated with horseradish peroxidase-labeled secondary antibody (1:5000) and subjected to enhanced chemiluminescence (KeyGEN BioTech) to detect the immunoreactivities. All the proteins were purchased from Santa Cruz Biotechnology.

Mouse xenograft assay

To stable silencing STC2, shRNA targeting STC2 or shNC was inserted into the pAAV-U6-GFP vector (Cell Biolabs, San Diego, CA, USA). HEK-293T cells were co-transfected with pAAV-shSTC2 or pAAV-shNC with pHelper and pAAV-DJ Rep-Cap. Viruses were harvested and then infected with Ishikawa cells to obtain stable cells. BALB/c nude mice (n = 20; 4–6 weeks old, 18–20 g weight) were divided into four groups: sh-NC (n = 5), sh-STC2 (n = 5), sh-NC with E2 (n = 5), and sh-STC2 with E2 (n = 5). The experiment was approved by the Medical Ethics Committee of Zhejiang Provincial People's Hospital (Approval no. 20220063) and in accordance with the National Institutes of Health Laboratory Animal Care and Use Guidelines. Stable STC2-silencing Ishikawa cells or the negative control was subcutaneously injected into the nude mice. Mice in the sh-NC with E2 or sh-STC2 with E2 groups were subcutaneously injected with 500 mg/kg E2 once a week 2 days after cell injection. The tumor volume was calculated every week and weighed 5 weeks later.

Statistical analysis

All the data were expressed as the mean ± standard error of the mean and analyzed via the Student's t-test or one-way analysis of variance in GraphPad Prism software (GraphPad Software, San Diego, CA, USA). P < 0.05 was considered statistically significant.


  Results Top


Stanniocalcin 2 was elevated in the estrogen-related endometrial cancer tissues

To investigate the role of STC2 in EC, bioinformatic analysis was performed to detect the expression of STC2 in EC. Results showed that STC2 was upregulated in the EC tissues compared to the normal tissues based on TIMER [Figure 1]a, GEPIA [Figure 1]b, and UALCAN [Figure 1]c. Immunohistochemistry confirmed the upregulation of STC2 in the EC tissues [Figure 1]d. The type I estrogen-related EC tissues showed a higher expression of STC2 than the type II estrogen-independent EC tissues [Figure 1]d. Ishikawa and RL95-2 cells were then treated with increasing concentrations of E2 in 0.001, 0.01, 0.1, 1, or 10 μM. Treatment with E2 significantly increased STC2 in Ishikawa and RL95-2 cells [Figure 1]e compared to that in cells without E2 treatment, and E2 treatment (0.1 μM) showed the highest expression of STC2 [Figure 1]e.
Figure 1: STC2 was elevated in the estrogen-related EC tissues. (a) The expression level of STC2 in the EC tissues and normal tissues based on TIMER analysis (https://cistrome.shinyapps.io/timer/). (b) STC2 was upregulated in the EC tissues (n = 174) compared to the normal tissues (n = 91) based on GEPIA analysis (http://gepia.cancer-pku.cn/index.html). Red color represents the EC tissues, and black color represents the normal tissues. UCEC represents uterine corpus endometrial cancer. (c) The expression levels of STC2 in the EC tissues (n = 546) and normal tissues (n = 35) based on UALCAN analysis (http://ualcan.path.uab.edu/cgi-bin/TCGAExResultNew2.pl?genenam=MED27&ctype=STAD). (d) Immunohistochemistry confirmed the upregulation of STC2 in the EC tissues, and the type I estrogen-related EC tissues demonstrated a higher expression of STC2 than the type II estrogen-independent EC tissues. (e) Treatment with E2 significantly increased STC2 in Ishikawa and RL95-2 compared to that without E2 treatment, and E2 treatment (0.1 μM) showed the highest expression of STC2. (n = 3). *P < 0.05, **P < 0.01, ***P < 0.001. STC2: Stanniocalcin 2, EC: Endometrial cancer.

Click here to view


Knockdown of stanniocalcin 2 reduced the proliferation of 17β-estradiol-induced endometrial cancer cells

Loss-of-functional assays were used to detect the effect of STC2 on the proliferation of EC. Ishikawa [Figure 2]a and RL95-2 cells, with or without 0.1 μM E2 treatment, were transfected with sh-STC2 to decrease the protein expression of STC2. Treatment with E2 increased the cell viability of Ishikawa and RL95-2 cells [Figure 2]b and promoted the proliferation of Ishikawa cells [Figure 2]c. However, silencing of STC2 decreased the cell viability of E2-induced Ishikawa and RL95-2 cells [Figure 2]b and inhibited the proliferation of E2-induced Ishikawa [Figure 2]c.
Figure 2: Knockdown of STC2 reduced the proliferation of E2-induced EC cells. (a) Ishikawa cells were transfected with sh-STC2 or sh-NC for 48 h, then treated with E2 or vehicle for 24 h, and STC2 protein expression was detected by western blot. (b) Silencing of STC2 reduced the cell viability of Ishikawa and RL95-2 and attenuated E2-induced increase of the cell viability in Ishikawa and RL95-2 based on the CCK8 assay. (c) Silencing of STC2 reduced the cell proliferation of Ishikawa and attenuated E2-induced increase of cell proliferation in Ishikawa based on the colony formation assay. (n = 3). *P < 0.05, **P < 0.01, ***P < 0.001. STC2: Stanniocalcin 2, EC: Endometrial cancer, E2: 17β-estradiol.

Click here to view


Knockdown of stanniocalcin 2 promoted apoptosis of 17β-estradiol-induced endometrial cancer cells

The effect of STC2 on apoptosis of EC was assessed using flow cytometry. Knockdown of STC2 promoted cell apoptosis of Ishikawa cells [Figure 3], while treatment with E2 reduced cell apoptosis of Ishikawa cells [Figure 3]. However, the loss of STC2 promoted cell apoptosis of E2-induced Ishikawa cells [Figure 3].
Figure 3: Knockdown of STC2 promoted apoptosis of E2-induced EC cells. Silencing of STC2 promoted cell apoptosis of Ishikawa and attenuated E2-induced decrease of cell apoptosis in Ishikawa based on the flow cytometry assay. (n = 3). *P < 0.05, **P < 0.01, ***P < 0.001. STC2: Stanniocalcin 2, EC: Endometrial cancer, E2: 17β-estradiol.

Click here to view


Knockdown of stanniocalcin 2 promoted the activation of AMP-activated protein kinase in 17β-estradiol-induced endometrial cancer cells

Underlying mechanism involved in STC2-mediated EC was evaluated. Transfection with sh-STC2 increased the protein expression of p-AMPK in Ishikawa cells [Figure 4]a. However, E2 reduced p-AMPK expression in Ishikawa cells [Figure 4]a. Moreover, silencing of STC2 attenuated E2-induced decrease in p-AMPK in Ishikawa cells [Figure 4]a. Treatment with AMPK inhibitor, compound C, attenuated STC2 defciency-induced decrease of cell viability in E2-induced Ishikawa cells [Figure 4]b.
Figure 4: Knockdown of STC2 promoted the activation of AMPK in E2-induced EC cells. (a) Silencing of STC2 enhanced the protein expression of p-AMPK in Ishikawa, and attenuated E2-induced decrease of p-AMPK expression in Ishikawa. (b) Treatment with AMPK inhibitor and compound C attenuated an STC2 deficiency-induced decrease of the cell viability in E2-induced Ishikawa. (n = 3). ***P < 0.001. AMPK: AMP-activated protein kinase, p-AMPK: Phosphorylated-AMPK, STC2: Stanniocalcin 2, EC: Endometrial cancer, E2: 17β-estradiol.

Click here to view


Knockdown of stanniocalcin 2 suppressed 17β-estradiol-induced tumor growth of endometrial cancer

Mouse xenograft assay was conducted to evaluate the effect of STC2 on the tumor growth of EC. Mice were injected with stable STC2-silencing Ishikawa cells, and then treated with E2. The loss of STC2 inhibited in vivo tumor growth of EC through decreasing of tumor weight and volume [Figure 5]a. E2 treatment promoted the tumor weight and volume of EC [Figure 5]a. However, the loss of STC2 repressed E2-induced tumor growth of EC with the reduction of tumor weight and volume [Figure 5]a. Transfection with sh-STC2 increased the protein expression of p-AMPK in tumor tissues [Figure 5]b. However, E2 reduced p-AMPK expression [Figure 5]b. Moreover, silencing of STC2 attenuated an E2-induced decrease in p-AMPK in the tumor tissues [Figure 5]b.
Figure 5: Knockdown of STC2 suppressed the E2-induced tumor growth of EC. (a) E2 treatment promoted the tumor weight and volume of EC, while the loss of STC2 repressed the E2-induced tumor growth of EC with a reduction of tumor weight and volume. (b) Silencing of STC2 enhanced the protein expression of p-AMPK in the tumor tissues and attenuated an E2-induced decrease of p-AMPK expression. (n = 3). ***P < 0.001. p-AMPK: Phosphorylated-AMP-activated protein kinase, STC2: Stanniocalcin 2, EC: Endometrial cancer, E2: 17β-estradiol.

Click here to view



  Discussion Top


Estrogen signaling functions as oncogenic signaling in EC.[7] Estrogen stimulated the upregulation of UBE2C through ER-α, and UBE2C promoted cell proliferation, metastasis, and epithelial-mesenchymal transformation of EC.[16] RIZ1 functioned as a tumor suppressor in EC in an estrogen-dependent way.[17] This study showed that STC2 was an estrogen-responsive gene, and STC2 deficiency inhibited estrogen-stimulated tumor growth of EC.

The previous study has shown that STC-1 was reduced in the EC tissues.[18] STC-1 also suppressed cervical cancer proliferation and metastasis.[19] Nuclear factor kappa-B (NF-κB) binds to the promoter region of STC-1 and promotes STC-1 expression in cervical cancer.[19] NF-κB is activated in cervical cancer with a reduced level of nonphosphorylated p65 protein, and reduced p65 resulted in a decrease in STC-1.[20] Considering that NF-κB is also activated in EC,[21] reduced p65 might also inhibit the expression of STC-1 in EC. However, immunohistochemical analysis showed that STC2 was upregulated in the EC tissues.[14] STC2 was identified as an oncogene in the distinct tumors.[11],[12],[13],[15] Results in this study also confirmed the upregulation of STC2 in the EC tissues. Type I estrogen-related EC tissues demonstrated a higher expression of STC2 than type II estrogen-independent EC tissues, and estrogen-stimulated STC2 expression in EC cells suggesting that STC2 was an estrogen-responsive gene in EC. Knockdown of STC2 attenuated estrogen-induced decrease in cell apoptosis and increase in the proliferation in EC cells. Moreover, silencing of STC2 also inhibited estrogen-stimulated in vivo tumor growth of EC. Therefore, STC2 functioned as an oncogene in EC. However, the effects of STC2 on estrogen-induced migration, invasion, and epithelial-mesenchymal transformation of EC should be investigated in further research.

AMPK signaling is known as an energy sensor, and an increased AMP/ATP ratio initiates the activation of AMPK signaling and stimulates the catabolic pathways to maintain energy homeostasis.[22],[23] Therefore, AMPK signaling functioned as a metabolic tumor suppressor to inhibit tumor progression through the regulation of cell growth and metabolism.[24] Estrogen has been reported to mediate glucose homeostasis and energy balance through AMPK signaling,[25] and to induce the downregulation of p-AMPK in EC cells.[26] Furthermore, the activation of AMPK signaling contributed to the suppression of estrogen-dependent EC progression.[27] STC2 modulated AMPK signaling to mediate autophagy and epithelial-mesenchymal transition of trophoblasts.[28] In this study, estrogen reduced the protein expression of p-AMPK to stimulate EC progression. However, the loss of STC2 increased the p-AMPK expression in estrogen-induced EC cells, indicating that STC2 might promote the proliferation and growth of EC through the inactivation of AMPK signaling.


  Conclusion Top


Collectively, STC2 was an estrogen-responsive gene in EC. STC2 was upregulated by estrogen, and the loss of STC2 repressed the proliferation and growth of EC. The activation of AMPK signaling was involved in the suppressive effect of STC2 deficiency on EC progression. Therefore, STC2 might be a potential target for type I estrogen-related EC. However, the mechanism involved in the STC2-mediated type II estrogen-independent EC should be investigated in further research.

Data availability

The authors declare that all data supporting the findings of this study are available within the paper and any raw data can be obtained from the corresponding author upon request.

Financial support and sponsorship

This work was supported by the Zhejiang Medical and Health Science and Technology Project (Grant No. 2021KY533).

Conflicts of interest

There are no conflicts of interest.



 
  References Top

1.
Ozgul N, Akgor U, Salman MC, Kuru O, Gultekın M. Lymph node dissection in intermediate and high-intermediate risk endometrial cancer. EJGO 2021;42:1146-52.  Back to cited text no. 1
    
2.
Zhang K, Li H, Yan Y, Zang Y, Li K, Wang Y, et al. Identification of key genes and pathways between type I and type II endometrial cancer using bioinformatics analysis. Oncol Lett 2019;18:2464-76.  Back to cited text no. 2
    
3.
Ruel-Laliberté J, Bessette P, Turcotte É, Belissant O, Lapointe-Milot K. High diagnostic value of 18F-FDG PET/CT in detecting endometrial cancer in patients with precancerous endometrial lesions. EJGO 2021;42:1130-7.  Back to cited text no. 3
    
4.
Yamamoto M, Yoshida H. Prognosis of positive peritoneal cytology in minimally invasive surgery for early-stage endometrial cancer. EJGO 2021;42:1124-9.  Back to cited text no. 4
    
5.
Szewczuk W, Szewczuk O, Czajkowski K, Górnicka B, Ilczuk T, Semczuk A. ARID1A and GPR30 expression patterns characterize the different histological subtypes of endometrial carcinoma. EJGO 2021;42:1138-45.  Back to cited text no. 5
    
6.
Watanabe J, Kamata Y, Seo N, Okayasu I, Kuramoto H. Stimulatory effect of estrogen on the growth of endometrial cancer cells is regulated by cell-cycle regulators. J Steroid Biochem Mol Biol 2007;107:163-71.  Back to cited text no. 6
    
7.
Rodriguez AC, Blanchard Z, Maurer KA, Gertz J. Estrogen signaling in endometrial cancer: A key oncogenic pathway with several open questions. Horm Cancer 2019;10:51-63.  Back to cited text no. 7
    
8.
Tian W, Teng F, Gao J, Gao C, Liu G, Zhang Y, et al. Estrogen and insulin synergistically promote endometrial cancer progression via crosstalk between their receptor signaling pathways. Cancer Biol Med 2019;16:55-70.  Back to cited text no. 8
    
9.
Zhou WJ, Zhang J, Yang HL, Wu K, Xie F, Wu JN, et al. Estrogen inhibits autophagy and promotes growth of endometrial cancer by promoting glutamine metabolism. Cell Commun Signal 2019;17:99.  Back to cited text no. 9
    
10.
Joshi AD. New insights into physiological and pathophysiological functions of stanniocalcin 2. Front Endocrinol (Lausanne) 2020;11:172.  Back to cited text no. 10
    
11.
Hu L, Zha Y, Kong F, Pan Y. Prognostic value of high stanniocalcin 2 expression in solid cancers: A meta-analysis. Medicine (Baltimore) 2019;98:e17432.  Back to cited text no. 11
    
12.
Qie S, Sang N. Stanniocalcin 2 (STC2): A universal tumour biomarker and a potential therapeutical target. J Exp Clin Cancer Res 2022;41:161.  Back to cited text no. 12
    
13.
Li S, Huang Q, Li D, Lv L, Li Y, Wu Z. The significance of stanniocalcin 2 in malignancies and mechanisms. Bioengineered 2021;12:7276-85.  Back to cited text no. 13
    
14.
Aydin HA, Toptas T, Bozkurt S, Aydin A, Erdogan G, Pestereli E, et al. Stanniocalcin-2 may be a potentially valuable prognostic marker in endometrial cancer: A preliminary study. Pathol Oncol Res 2019;25:751-7  Back to cited text no. 14
    
15.
Bouras T, Southey MC, Chang AC, Reddel RR, Willhite D, Glynne R, et al. Stanniocalcin 2 is an estrogen-responsive gene coexpressed with the estrogen receptor in human breast cancer. Cancer Res 2002;62:1289-95.  Back to cited text no. 15
    
16.
Liu Y, Zhao R, Chi S, Zhang W, Xiao C, Zhou X, et al. UBE2C is upregulated by estrogen and promotes epithelial-mesenchymal transition via p53 in endometrial cancer. Mol Cancer Res 2020;18:204-15.  Back to cited text no. 16
    
17.
Yang T, Ren C, Jiang A, Yu Z, Li G, Wang G, et al. RIZ1 is regulated by estrogen and suppresses tumor progression in endometrial cancer. Biochem Biophys Res Commun 2017;489:96-102.  Back to cited text no. 17
    
18.
Khatun M, Urpilainen E, Ahtikoski A, Arffman RK, Pasanen A, Puistola U, et al. Low expression of stanniocalcin 1 (STC-1) protein is associated with poor clinicopathologic features of endometrial cancer. Pathol Oncol Res 2021;27:1609936.  Back to cited text no. 18
    
19.
Guo F, Li Y, Wang J, Li Y, Li Y, Li G. Stanniocalcin1 (STC1) inhibits cell proliferation and invasion of cervical cancer cells. PLoS One 2013;8:e53989.  Back to cited text no. 19
    
20.
Pan X, Jiang B, Liu J, Ding J, Li Y, Sun R, et al. STC1 promotes cell apoptosis via NF-κB phospho-P65 Ser536 in cervical cancer cells. Oncotarget 2017;8:46249-61.  Back to cited text no. 20
    
21.
Caruana BT, Byrne FL. The NF-κB signalling pathway regulates GLUT6 expression in endometrial cancer. Cell Signal 2020;73:109688.  Back to cited text no. 21
    
22.
Zhong H, Hao L, Li X, Wang C, Wu X. Anti-inflammatory role of trilobatin on lipopolysaccharide-induced acute lung injury through activation of AMPK/GSK3β-Nrf2 pathway. Signa Vitae 2020;16:160-6.  Back to cited text no. 22
    
23.
Li W, Saud SM, Young MR, Chen G, Hua B. Targeting AMPK for cancer prevention and treatment. Oncotarget 2015;6:7365-78.  Back to cited text no. 23
    
24.
Luo Z, Zang M, Guo W. AMPK as a metabolic tumor suppressor: Control of metabolism and cell growth. Future Oncol 2010;6:457-70.  Back to cited text no. 24
    
25.
Mauvais-Jarvis F, Clegg DJ, Hevener AL. The role of estrogens in control of energy balance and glucose homeostasis. Endocr Rev 2013;34:309-38.  Back to cited text no. 25
    
26.
Yin X, Liu Y, Qin J, Wu Y, Huang J, Zhao Q, et al. Artesunate suppresses the proliferation and development of estrogen receptor-α-positive endometrial cancer in HAND2-dependent pathway. Front Cell Dev Biol 2020;8:606969.  Back to cited text no. 26
    
27.
Zou J, Hong L, Luo C, Li Z, Zhu Y, Huang T, et al. Metformin inhibits estrogen-dependent endometrial cancer cell growth by activating the AMPK-FOXO1 signal pathway. Cancer Sci 2016;107:1806-17.  Back to cited text no. 27
    
28.
Lai R, Ji L, Zhang X, Xu Y, Zhong Y, Chen L, et al. Stanniocalcin2 inhibits the epithelial-mesenchymal transition and invasion of trophoblasts via activation of autophagy under high-glucose conditions. Mol Cell Endocrinol 2022;547:111598.  Back to cited text no. 28
    


    Figures

  [Figure 1], [Figure 2], [Figure 3], [Figure 4], [Figure 5]



 

Top
 
  Search
 
    Similar in PUBMED
   Search Pubmed for
   Search in Google Scholar for
 Related articles
    Access Statistics
    Email Alert *
    Add to My List *
* Registration required (free)  

 
  In this article
Abstract
Introduction
Materials and Me...
Results
Discussion
Conclusion
References
Article Figures

 Article Access Statistics
    Viewed362    
    Printed8    
    Emailed0    
    PDF Downloaded54    
    Comments [Add]    

Recommend this journal