Open Access

Rapamycin suppresses the PI3K/AKT/mTOR signaling pathway by targeting <em>SIRT1</em> in esophageal cancer

Retraction in: /10.3892/etm.2023.12288

  • Authors:
    • Tao Liu
    • Xiangsen Liang
    • Yu Sun
    • Shengzhuang Yang
  • View Affiliations

  • Published online on: August 17, 2021     https://doi.org/10.3892/etm.2021.10624
  • Article Number: 1190
  • Copyright: © Liu et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Rapamycin, a secondary metabolite produced by Streptomyces hygroscopicus, is known for its pharmacological effects, especially antitumor and immunosuppressive activities. However, the antitumoral effects of rapamycin in human esophageal cancer (EC) are still poorly understood. To investigate the potential of rapamycin in EC treatment, sirtuin 1 (SIRT1) mRNA expression was quantified in the tissue of patients with EC or in EC cell lines using reverse transcription‑quantitative PCR. The protein levels of SIRT1 and PI3K/AKT/mTOR were measured via western blotting. Furthermore, cell viability, migration and invasion were investigated by Cell Counting Kit‑8, wound healing and Transwell assays, respectively. The present results suggested that SIRT1 expression was upregulated in EC. In vitro, the inhibitory effect of rapamycin on cell viability in EC was strengthened or weakened after small interfering (si)‑SIRT1 or pcDNA3.1/SIRT1 transfection. Furthermore, SIRT1 rescued the inhibitory effect of rapamycin on the migration and invasion of EC cells. In vivo, si‑SIRT1 or SIRT1 overexpression in mice could enhance or rescue the inhibitory effects of rapamycin on tumor growth. In addition, SIRT1 transfection rescued the decreased level of phosphorylated (p)‑PI3K, p‑AKT and p‑mTOR induced by rapamycin treatment. Taken together, the present results suggested that rapamycin suppressed the cell viability, migration, invasion and PI3K/AKT/mTOR signaling pathway in EC by negatively regulating SIRT1.

Introduction

Esophageal cancer (EC), a digestive tract malignancy, is the 9th most common cancer worldwide (1). The incidence and mortality of EC rank 8 and 5th in developing countries, respectively (2). EC is characterized by rapid growth, early metastasis and low response to medication (3). Squamous cell carcinoma and adenocarcinoma are the two major subtypes of EC. Currently, surgery, radiotherapy, chemotherapy and combination therapy are the main treatments for EC (4). However, most patients are diagnosed at an advanced stage, due to the atypical symptoms of EC at the early stage (5). As a result, although the survival rate of patients has increased with the development of medicine and technology, the long-term prognosis of patients with EC is still unsatisfactory (6). Therefore, there is an urgent need to identify drugs with satisfactory therapeutic effects to effectively improve clinical treatment and reduce mortality of EC.

Rapamycin exhibits antitumor and immunosuppressive properties. As a mTOR-targeting molecule, rapamycin has been approved to prevent the rejection of transplanted organs and to block restenosis after angioplasty (7). A combination of rapamycin and cisplatin inhibited the growth of various cancer cell lines, such as endometrial ECC-1 cells and cervical carcinoma HeLa cells (8). As of today, although rapamycin has been used as an anti-EC drug, the role of rapamycin in EC remains controversial (9). Therefore, it is necessary to explore the molecular mechanisms of rapamycin in EC treatment.

The conserved serine/threonine kinase mTOR is a mammalian target of rapamycin, and a downstream effector of the PI3K/AKT pathway (10). mTOR forms two distinct multiprotein complexes, mTOR complex (C)1 and mTORC2(11). mTORC1 is sensitive to rapamycin and can regulate cell growth, proliferation and survival by activating the PI3K signaling pathway. mTORC2 is considered resistant to rapamycin and activates AKT. In addition, the role of the PI3K/AKT/mTOR signaling pathway has been well characterized in cell proliferation (8). Aberrant activation of this signaling pathway has been reported in multiple human cancers, including EC (12). Hence, exploring the association between rapamycin and the PI3K/AKT/mTOR signaling pathway is important to understand the therapeutic mechanism of rapamycin on EC.

Sirtuin-1 (SIRT1), a highly conserved NAD+-dependent class III histone deacetylase, is a member of the mammalian sirtuin family (13). Accumulating evidence suggests that SIRT1 is a key regulator of life extension, DNA damage, metabolic stress, inflammation and cancer (14). For example, SIRT1 acts as a tumor suppressor against infiltrated immune cells and tumoral cells in the tumoral microenvironment (such as gastric, bladder and liver cancer) (15). Furthermore, SIRT1 upregulation has been confirmed in a variety of solid tumors, including EC (4,10). Although the association of SIRT1 in tumorigenesis has been indicated in these studies, its mechanism in EC has not been sufficiently studied.

In the present study, SIRT1 expression and its effects with rapamycin on cell viability, migration and invasion were detected in EC tissues and cells. Protein levels of the PI3K/AKT/mTOR signaling pathway were evaluated to investigate the underlying mechanism of rapamycin in EC.

Materials and methods

Tissue samples

A total of 30 EC and their corresponding adjacent normal tissue samples from were collected from patients with EC. The collected tissues were washed twice with PBS and stored at -70˚C. The present study was approved by the Ethics Committee of The Second Affiliated Hospital of Guangxi Medical University (approval no. SYXK Gui 2018-0004), and all patients provided written informed consent. The clinical pathological features of all patients, including sex, age, tumor diameter, lymph node metastasis and TNM stage were summarized in Table I.

Table I

Association between SIRT1 expression and clinical pathological features of 30 patients with esophageal cancer.

Table I

Association between SIRT1 expression and clinical pathological features of 30 patients with esophageal cancer.

 SIRT1 expression 
CharacteristicAll cases (n=30)High (n=15)Low (n=15)P-value
Sex, (n)   0.724
     Male1376 
     Female1789 
Age, years   0.712
     <501165 
     ≥5019910 
Tumor diameter, cm   0.676
     <3201010 
     ≥31055 
Lymph node metastasis, n   0.031a
     No14311 
     Yes16124 
TNM stage, n   0.026a
     I/II14410 
     III/IV18115 

[i] aP<0.05 vs. high-risk group. SIRT1, sirtuin 1.

Cell culture

Human EC cell lines (KYSE30 and KYSE150) and a healthy esophageal cell line (Het-1A) were obtained from the American Type Culture Collection. Het-1A, KYSE30 and KYSE150 cells were cultured in DMEM (Gibco; Thermo Fisher Scientific, Inc.), supplemented with 10% FBS (Beyotime Institute of Biotehcnology) and 1% streptomycin-penicillin. All cell lines were maintained at 37˚C in a 5% CO2 atmosphere incubator.

Western blot analysis

Protein from KYSE30 and KYSE150 cells was extracted using RIPA lysis buffer (MilliporeSigma) according to the manufacturer's instructions. The total protein concentration was measured with a BCA protein assay kit (Takara Biotechnology Co., Ltd.). The samples (20 µg) were separated via 10% SDS-PAGE, then transferred onto PVDF membranes. Subsequently, the membranes were blocked with 5% non-fat milk for 2 h at room temperature, then treated with the following primary antibodies overnight at 4˚C: Anti-SIRT1 (1:1,000; cat. no. ab110304; Abcam), anti-AKT (1:1,000; cat. no. ab8805; Abcam), anti-PI3K (1:1,000; cat. no. ab278545; Abcam), anti-mTOR (1:1,000; cat. no. ab134903; Abcam), anti-GAPDH (1:1,000; cat. no. ab181602; Abcam), anti-phosphorylated (p)-PI3K (1:1,000; Abcam), anti-p-AKT (1:1,000; Abcam) and anti-p-mTOR (1:1,000; Abcam). After washing three times with Tris-buffered saline-0.1% Tween-20 (TBST), the membranes were incubated with the HRP-labeled secondary antibody (anti-rat IgG; 1:2,000; cat. no. ab6728; Abcam) for 2 h at 4˚C. Finally, the protein bands were visualized with ECL system (Thermo Fisher Scientific, Inc.) and analyzed by densitometry using software ImageJ (version 1.52; National Institutes of Health), with GAPDH as a loading control.

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA was extracted from EC tissues or cells using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.) according to the manufacturer's instructions. RNA concentration was detected by NanoDrop™ ND-1000 spectrophotometer (NanoDrop Technologies; Thermo Fisher Scientific, Inc.). Subsequently, RNA was reverse transcribed into cDNA using the PrimeScript RT reagent kit (Takara Biotechnology Co., Ltd.) and analyzed via qPCR using the SYBR Green PCR kit (Takara Biotechnology Co., Ltd.) according to the manufacturers' instructions. The following thermocycling conditions were used for qPCR: Initial denaturation of 95˚C for 1 min, 40 cycles of 94˚C for 15 sec followed by 60˚C for 30 sec. The primer sequences were as follows: SIRT1 forward, 5'-GCCGATGGTCATGCAGTCAG-3' and reverse, 5'-CAGGTGGCAGGTCATTTTTCT-3'; GAPDH forward, 5'-GAGTCAACGGATTTGGTCGT-3' and reverse, 5'-TGATATTTGGAGCGATCTCG-3'. Relative target gene expression was calculated using the 2-ΔΔCq method (16). Human GAPDH was used as an internal control.

Small interfering RNA (siRNA) and cell plasmid transfection

KYSE30 and KYSE150 cells (1x105 cells/well) were seeded in 6-well plates. siRNA targeting SIRT1 (si-SIRT1; 5'-GACUCCUGGCAAGAATT-3') and a control non-targeting siRNA [si-negative control (NC); 5'-ATGGCAGAAGGAGGAGGG-3'] were designed and synthesized by BioTeke Corporation. The full-length sequence of SIRT1 was synthesized and cloned into a pcDNA3.1 plasmid (Invitrogen; Thermo Fisher Scientific, Inc.) to produce a pcDNA3.1/SIRT1 vector (SIRT1). The empty pcDNA3.1 vector was used as the NC. The siRNAs (50 nmol/l) and plasmids (50 nmol/l) were transfected into KYSE30 and KYSE150 cells using Lipofectamine® 3000 Reagent (Invitrogen; Thermo Fisher Scientific, Inc.) overnight at 37˚C, according to the manufacturer's instructions. KYSE30 and KYSE150 cells transfected with si-NC, si-SIRT1, empty pcDNA3.1 vector or pcDNA3.1/SIRT1 for 24 h were treated with rapamycin at different concentrations (0, 10, 50, 100 and 200 nM) at room temperature (6).

Cell viability assay

Cell Counting Kit-8 (CCK-8; MilliporeSigma) assays were performed to evaluate cell viability. KYSE30 and KYSE150 cells were treated with rapamycin (MilliporeSigma) at different concentrations (0, 10, 50, 100 and 200 nM) for 48 h. After transfection and treatment, cells (1x105 cells/well) were seeded into 96-well plates. A total of 10 µl CCK-8 was added into each well and the cells were cultured at room temperature for 1 h. Finally, the absorbance was measured at 450 nm using a microplate reader (Bio-Rad Laboratories, Inc.).

Wound healing assay

After SIRT1 transfection and rapamycin treatment, KYSE30 and KYSE150 cells (8x105; serum starved) were seeded in a 6-well plate. A scratch was made in the middle of the well using a P200 pipette tip. Mitomycin (10 µg/ml; Morey Biosciences, Inc.) was added to inhibit cell proliferation, in order to eliminate the interference of cell proliferation according to previous research (17). Cell migration was assessed on the basis of images captured at 0 and 24 h using an optical microscope (magnification, x40; Bio-Rad Laboratories, Inc.) and analyzed using ImageJ (version 2.0). The relative distance of migration was calculated as: (Scratch area at 0 h-scratch area at 24 h)/scratch area at 0 h x100%.

Transwell assay

KYSE30 and KYSE150 cells, resuspended in serum-free media (5x104 cells/ml) were inoculated into the upper chamber of the Transwell inserts (Sigma-Aldrich; Merck KGaA), which were pre-embedded with Matrigel (Sigma-Aldrich; Merck KGaA). DMEM with 10% FBS was added to the lower chamber and incubated at 37˚C for 24 h. Cells below the membrane were fixed with 4% paraformaldehyde for 30 min at 37˚C, and stained with 0.5% crystal violet dye (MilliporeSigma) for 30 min at 37˚C. Finally, the lower chamber cells were counted under an optical microscope (magnification, x40; Bio-Rad Laboratories, Inc.).

In vivo xenograft study

BALB/c male nude mice (4-6 weeks old; 25±5 g; 10/group) were housed in laminar flow cabinets under specific pathogen-free conditions, with a 12/12 h light/dark cycle and 60% humidity, and free access to food and water. Mice were divided into four groups: i) Control group; ii) rapamycin group; iii) rapamycin + SIRT1 group and iv) rapamycin + si-SIRT1 group. The control group was subcutaneously inoculated with KYSE30 cells (5x106 in 200 µl). The rapamycin group was subcutaneously inoculated with KYSE30 cells (5x106 in 200 µl), and the mice were then intraperitoneally injected with rapamycin (50 mg/kg) (6). The rapamycin + si-SIRT1 or rapamycin + SIRT1 groups were subcutaneously inoculated with KYSE30 cells (5x106 in 200 µl) transfected with si-SIRT1 or pcDNA3.1/SIRT1, after which the mice were intraperitoneally injected with rapamycin (50 mg/kg) every other day. For tumor growth analysis, the tumor size weas measured every 5 days with a sliding caliper, and the tumor volume was defined as (longest diameter) x (shortest diameter)2/2. After 4 weeks of treatment, tumor-bearing mice were euthanized with an overdose of intraperitoneal pentobarbital (200 mg/kg) and the tumors were removed, weighed and stored (-80˚C) for further analysis. The study was approved by the Ethics Committee of The Second Affiliated Hospital of Guangxi Medical University (approval no. SYXK Gui 2018-0004), and animal care and euthanasia were carried out in strict accordance with the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health (18).

Statistical analysis

The data in the current study were presented as the mean ± SD. The data of two groups were assessed using paired Student's t-test (for tumor and adjacent non-tumor samples) and an unpaired Student's t-test (for cell samples). Moreover, multiple comparisons were analyzed via one-way ANOVA followed by Tukey's multiple comparison post hoc test. All experiments were carried out in triplicate. The correlation between SIRT1 and AKT expression was measured using Spearman correlation analysis. Associations between gene expression and clinicopathological features were analyzed using Fisher's exact test. All statistical analyses were performed using SPSS 22.0 Statistical Software (IBM Corp.) and GraphPad Prism 7.0 software (GraphPad Software, Inc.). P<0.05 was considered to indicate a statistically significant difference.

Results

SIRT1 is upregulated in EC

RT-qPCR and western blotting were carried out to characterize SIRT1 in EC tissues at the mRNA and protein level, respectively. As indicated in Fig. 1A, SIRT1 mRNA levels were significantly increased in EC tissues compared with adjacent normal tissues (P<0.05). Similarly, SIRT1 mRNA expression in KYSE30 and KYSE150 cells was upregulated compared with Het-1A cells (P<0.05; Fig. 1B). Furthermore, SIRT1 protein levels were significantly increased in EC tissues and cells when compared with their respective normal controls (P<0.05; Fig. 1C and D). Based on the mean of SIRT1 expression in tissues samples, the patients were divided into high- and low-SIRT1 expression groups. Fisher's exact test showed that high SIRT1 expression was significantly associated with lymph node metastasis (P=0.031) and III/IV TNM stage (P=0.026); however, there were no significant differences indicated in other clinical pathological features, including sex, age and diameter between the high- and low-SIRT1 expression groups (Table I). The present results suggested that SIRT1 expression was upregulated in EC, and it may serve as an oncogene in the progression of EC.

In addition, SIRT1 protein expression in KYSE30 and KYSE150 cells decreased gradually as the concentration of rapamycin increased (Fig. S1), indicating that SIRT1 expression was associated with rapamycin treatment in EC development.

SIRT1 rescues the inhibitory effect of rapamycin on cell viability in EC cells

To further validate the role of SIRT1 in EC development, SIRT1 expression was knocked down by transfection with si-SIRT1 in KYSE30 and KYSE150 cells. Similarly, for SIRT1 overexpression, the full-length sequence of SIRT1 was synthesized and cloned into pcDNA3.1 plasmid to produce the SIRT1 plasmid in KYSE30 and KYSE150 cells. The present results indicated that the expression level of SIRT1 in the si-SIRT1 group was significantly lower than that in the si-NC group at both mRNA and protein levels (P<0.05; Fig. 2A and B). Additionally, SIRT1 expression was higher in the SIRT1 group compared with the negative control vector group at both mRNA and protein levels (P<0.05; Fig. 2C and D).

KYSE30 and KYSE150 transfected with si-NC, si-SIRT1 or pcDNA3.1/SIRT1 for 24 h were treated with rapamycin at different concentrations (0, 10, 50, 100 and 200 nM) (6) for 48 h. The results of the CCK-8 assay indicated that the inhibitory effect of rapamycin on cell viability was increased in a dose-dependent manner in the si-NC group (≤100 nM). This inhibitory effect weakened along with the increase of rapamycin concentration (≥100 nM). After cells were transfected with si-SIRT1, the inhibitory effect of rapamycin on cell viability was enhanced (P<0.05 at 50, 100 and 200 nM rapamycin; Fig. 2E and F); however, the inhibitory effect of rapamycin on cell viability was significantly rescued after cells were transfected with SIRT1 (all, P<0.05). The present results indicated that SIRT1 could rescue the inhibitory effect of rapamycin on the viability of EC cells.

SIRT1 rescues the inhibitory effect of rapamycin on the migration and invasion of EC cells

After KYSE30 and KYSE150 cells were treated with 100 nM rapamycin for 48 h (6), the mRNA and protein levels of SIRT1 were analyzed. As presented in Fig. 3, SIRT1 expression levels were significantly decreased in the rapamycin group compared with the control group (P<0.05; Fig. 3A and B). Furthermore, SIRT1 expression was significantly decreased in the rapamycin + si-SIRT1 group and increased in the rapamycin + SIRT1 group when compared with the rapamycin group (P<0.05). To further investigate whether SIRT1 could affect the pathological progression of EC, the migration and invasion abilities of EC cells were studied. Wound healing and Transwell assays revealed that the migration and invasion of cells treated with rapamycin was decreased (P<0.05). Moreover, the inhibition of migration and invasion in cells treated with rapamycin was enhanced by si-SIRT1 transfection, while pcDNA3.1/SIRT1 transfection rescued the decrease in migration and invasion induced by rapamycin treatment (P<0.05; Fig. 3C-F).

SIRT1 rescues the inhibitory effect of rapamycin on the PI3K/AKT/mTOR signaling pathway of EC cells

Changes in the phosphorylation of key activation proteins of the PI3K/AKT/mTOR signaling pathway (including PI3K, AKT, mTOR and their phosphorylated forms) were evaluated via western blot analysis. Compared with the control group, rapamycin reduced the expression levels of p-PI3K/PI3K, p-AKT/AKT and p-mTOR/mTOR in KYSE30 and KYSE150 cells (P<0.05), while SIRT1 transfection could reverse this decrease (P<0.05; Fig. 4A and B). In particular, as a key regulator of cell proliferation, AKT was determined to be upregulated in EC tissues compared with adjacent healthy tissues (P<0.05; Fig. S2A). Additionally, a significant correlation between SIRT1 and AKT expression was identified (r=0.4873, P<0.01; Fig. S2B).

Inhibition or overexpression of SIRT1 enhances or weakens the inhibitory effect of rapamycin on xenograft growth from KYSE30 cells, respectively

The effect of si-SIRT1 on cell sensitivity to rapamycin was investigated by xenograft experiments. As presented in Fig. 5A, tumor growth in rapamycin or rapamycin + si-SIRT1 groups was inhibited compared with the control group (P<0.05). In terms of tumor size and weight, rapamycin combined with si-SIRT1 had a significantly stronger inhibitory effect on xenografts (P<0.05; Fig. 5B). On the contrary, the inhibitory effect of rapamycin on tumor growth could be suppressed by SIRT1 overexpression (P<0.05; Fig. S3). To further clarify the role of SIRT1 in xenotransplantation, the mRNA and protein expression of SIRT1 was observed in transplanted tumors. In xenografts, the expression levels of SIRT1 in the rapamycin + si-SIRT1 group were lower than that in rapamycin group at the mRNA and protein levels (Fig. 5C and D). The present data suggested that SIRT1 could rescue the inhibitory effects of rapamycin on tumor growth in vivo.

Discussion

Due to the potential malignancy and poor prognosis of EC, the local control effect of radiotherapy and chemotherapy and three-field lymph node dissection has become limited (19). Therefore, there is an urgent need for technical support and drug mechanism improvement in existing therapeutic approaches.

Rapamycin, a macrolide antibiotic, is found in Streptomyces hygroscopicus. Its anticancer effect was first reported in 2002(20). Rapamycin is an inhibitor of serine/threonine protein kinase mTOR, which is the mechanical target of rapamycin (7). During the past decade, the majority of studies have focused on the potential anti-aging role of rapamycin in age-related diseases, including cancer and Alzheimer's disease, as well as improving cardiovascular and cerebrovascular cognitive impairment (11,21). Due to mutations of oncogenes (including PI3K, AKT or Ras) or loss of function of tumor suppressors [including PTEN, liver kinase B1 or tuberous sclerosis 1 (TSC1)] in cancer, increased activation of rapamycin-sensitive complex mTORC1 can be observed (12). mTORC1 activation can not only drive the expression of proteins related to energy metabolism and nutrient absorption in cancer cells, but also promote the transcription of oncogenes (22). In addition, although numerous studies have reported the antitumoral effects of rapamycin in preclinical models of human tumors, their efficacy as a broad monotherapy was unsatisfactory in patients with cancer (15,23). Therefore, rapamycin may need to be combined with other factors or drugs, such as resveratrol and cisplatin, to exhibit a higher efficiency in cancer, including EC (19). In the present study, rapamycin treatment inhibited SIRT1 expression in EC cells (KYSE30 and KYSE150), suggesting that rapamycin could regulate the expression of SIRT1 in EC. More importantly, upon rapamycin treatment, SIRT1 could significantly rescue the viability of EC in vitro; in vivo, SIRT1 could also rescue the inhibitory effects of rapamycin on tumor growth.

SIRT1 was previously indicated to activate stress defense and DNA repair mechanisms, thus contributing to genomic integrity (14). SIRT1 has been reported to serve a role in the regulation of metabolism and the maintenance of genomic integrity, thus being described as a potential tumor suppressor (24). Selective SIRT1 inhibitors, such as EX527, could significantly inhibit cell migration and epithelial mesenchymal transition, thus changing the invasive and metastatic potential of esophageal cancer cell lines (25). SIRT1 has been demonstrated to be upregulated in a number of human tumors, including colon, renal and lung cancers (26). Similarly, in the present study, SIRT1 was demonstrated to have a notably high expression in EC tissues and cells, indicating that dysregulation of SIRT1 may be involved in the pathogenesis of human EC (27). A previous study indicated that downregulation of SIRT1 expression could lead to significant changes in the invasive and metastatic potential of resistant EC cell lines (28). In the present study, after overexpressing SIRT1 siRNA in EC cell lines, the effect of rapamycin on cell viability was inhibited. Furthermore, SIRT1 overexpression could promote cell invasion and migration, and alleviate the inhibitory effect of rapamycin. Taken together, the present results revealed that rapamycin could inhibit tumor development through the inhibition of cell viability, migration and invasion in EC cells by regulating SIRT1 expression.

The PI3K/AKT/mTOR signaling pathway has been reported to be involved in cell proliferation, differentiation, apoptosis and metastasis (29). The activation of the PI3K signaling pathway may lead to the occurrence of certain cancers, such as breast cancer (30). AKT is a key regulator of cell growth that mediates cell proliferation and apoptosis (31). mTOR can activate AKT and the AKT/mTOR signaling pathway (30). Rapamycin can also suppress the assembly and function of mTORC2 to inhibit AKT signaling (32). A previous study has demonstrated the activation of the mTOR signaling pathway in esophageal squamous cell carcinoma. Rapamycin specifically blocked the mTOR pathway in esophageal squamous cell carcinoma cells (33). Hence, in the present study, to further investigate the mechanisms of rapamycin and SIRT in EC, proteins of the PI3K/AKT/mTOR signaling pathway were quantified in treated cells. The present results indicated that rapamycin could reduce the protein levels of p-PI3K/PI3K, p-AKT/AKT and p-mTOR/mTOR, while SIRT1 overexpression could rescue the rapamycin-induced decrease in p-PI3K/PI3K, p-AKT/AKT and p-mTOR/mTOR levels. The present data indicated that in EC cells, rapamycin could inhibit the activation of the PI3K/AKT/mTOR signaling pathway by regulating SIRT1. SIRT1 inhibitor nicotinamide has been reported to increase mTOR activity in a dose-dependent manner and interact with TSC2, a component of the upstream mTOR inhibition complex mTORC1(32). SIRT1 may negatively regulate mTOR signaling through the TSC1/2 complex to inhibit EC cell proliferation. Therefore, it is hypothesized that rapamycin, by targeting SIRT1, may inhibit the PI3K/AKT/mTOR signaling pathway by blocking its binding to TSC2. However, this hypothesis needs to be further tested in future research.

In conclusion, the expression of SIRT1 was increased in EC, and rapamycin treatment could inhibit SIRT1 expression. In addition, rapamycin suppressed cell viability, migration, invasion and the PI3K/AKT/mTOR signaling pathway in EC by targeting SIRT1. These findings not only provided novel insights into rapamycin as a potential anticancer drug, but also emphasized SIRT1 as a potential molecular target of rapamycin for EC. However, the specific molecular mechanism and clinical effect of rapamycin in EC need to be explored in further research.

Supplementary Material

SIRT1 expression in esophageal cancer. SIRT1 protein expression in (A) KYSE30 and (B) KYSE150 cells. SIRT1 expression decreased gradually as the concentration of rapamycin increased. **P<0.01, ***P<0.001 vs. 0 nM. SIRT1, sirtuin 1.
AKT expression in esophageal cancer. (A) AKT in esophageal cancer tissue was upregulated compared with adjacent normal control tissue. *P<0.05 as indicated. (B) Significant correlation between SIRT1 expression and AKT expression was observed (r=0.4873; P<0.01). SIRT1, sirtuin 1.
SIRT1 rescues the inhibitory effect of rapamycin on xenograft growth from esophageal cancer cells. (A) Tumor volume and (B) weight from the xenografts of each group are presented. (C) Tumor weight in each group. (D) Reverse transcription-quantitative PCR was conducted to measure SIRT1 mRNA expression in the tumor tissue of each group. SIRT1, sirtuin 1. *P<0.05.

Acknowledgements

Not applicable.

Funding

Funding: This study was supported by the Scientific Research Project of Guangxi Administration of Traditional Chinese Medicine (grant no. GZZC2019033); the Development and Application Project of Guangxi Medical and Health Appropriate Technology (grant no. S2019012); the Scientific Research Project of Guangxi International Zhuang Hospital (grant no. GZ202003); the Scientific Research Basic Ability Improvement Project of Young and Middle-aged Universities in Guangxi (grant no. 2021KY0282); and the Guangxi Traditional Chinese Medicine Appropriate Technology Development and Application Project (grant no. GZSY21-43).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

All authors contributed to the study conception and design. Material preparation, data collection, experiment design, experiment implementation and analysis were performed by TL and XL, who also wrote the first draft of the manuscript. TL and YS confirm the authenticity of all the raw data. SY and YS provided valuable opinions and reviewed the draft of the paper. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

All patients provided written informed consent for their tissues to be used for clinical research. The present study was approved by the Ethics Committee of The Second Affiliated Hospital of Guangxi Medical University (approval no. SYXK Gui 2018-0004x).

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Short MW, Burgers K and Fry V: Esophageal cancer. Am Fam Physician. 95:22–28. 2017.PubMed/NCBI

2 

North BJ and Verdin E: Sirtuins: Sir2-related NAD-dependent protein deacetylases. Genome Biol. 5(224)2004.PubMed/NCBI View Article : Google Scholar

3 

Zhang Y: Epidemiology of esophageal cancer. World J Gastroenterol. 19(5598)2013.PubMed/NCBI View Article : Google Scholar

4 

Grabowska W, Sikora E and Bielak-Zmijewska A: Sirtuins, a promising target in slowing down the ageing process. Biogerontology. 18:447–476. 2017.PubMed/NCBI View Article : Google Scholar

5 

Huang FL and Yu SJ: Esophageal cancer: Risk factors, genetic association, and treatment. Asian J Surg. 41:210–215. 2018.PubMed/NCBI View Article : Google Scholar

6 

Lu Z, Peng K, Wang N, Liu HM and Hou G: Downregulation of p70S6K enhances cell sensitivity to rapamycin in esophageal squamous cell carcinoma. J Immunol Res. 2016(7828916)2016.PubMed/NCBI View Article : Google Scholar

7 

Du Z, Wang G, Cao Y, Hu CM, Yang K, Liu YZ, Zhang CZ, Zhang WH, Zhu ZT, Sun HZ, et al: Everolimus-inhibited multiple isoforms of UDP-glucuronosyltransferases (UGTs). Xenobiotica. 48:452–458. 2018.PubMed/NCBI View Article : Google Scholar

8 

Liu C, Ma T, Jiang T, Jia G, Yang C, Peng Y, Qian Y, Wang R and Wang S: Abnormal increase of miR-4262 promotes cell proliferation and migration by targeting large tumor suppressor 1 in gliomas. Pathol Res Pract. 216(152778)2020.PubMed/NCBI View Article : Google Scholar

9 

Boland PM and Burtness B: Esophageal carcinoma: Are modern targeted therapies shaking the rock? Curr Opin Oncol. 25:417–424. 2013.PubMed/NCBI View Article : Google Scholar

10 

Pópulo H, Lopes J and Soares P: The mTOR signalling pathway in human cancer. Int J Mol Sci. 13:1886–1918. 2012.PubMed/NCBI View Article : Google Scholar

11 

Guo W, Qian L, Zhang J, Zhang W, Morrison A, Hayes P, Wilson S, Chen T and Zhao J: Sirt1 overexpression in neurons promotes neurite outgrowth and cell survival through inhibition of the mTOR signaling. J Neurosci Res. 89:1723–1736. 2011.PubMed/NCBI View Article : Google Scholar

12 

Chueh S and Kahan B: Clinical application of sirolimus in renal transplantation: An update. Transpl Int. 18:261–277. 2005.PubMed/NCBI View Article : Google Scholar

13 

Hirashima K, Baba Y, Watanabe M, Karashima RI, Sato N, Imamura Y, Nagai Y, Hayashi N, Iyama KI and Baba H: Aberrant activation of the mTOR pathway and anti-tumour effect of everolimus on oesophageal squamous cell carcinoma. Br J Cancer. 106:876–882. 2012.PubMed/NCBI View Article : Google Scholar

14 

Zhang J, Peng J, Kong D, Wang X, Wang Z, Liu J, Yu W, Wu H, Long Z, Zhang W, et al: Silent information regulator 1 suppresses epithelial-to-mesenchymal transition in lung cancer cells via its regulation of mitochondria status. Life Sci. 280(119716)2021.PubMed/NCBI View Article : Google Scholar

15 

Yang H, Bi Y, Xue L, Wang J, Lu Y, Zhang Z, Chen X, Chu Y, Yang R, Wang R and Liu G: Multifaceted modulation of SIRT1 in cancer and inflammation. Crit Rev Oncog. 20:49–64. 2015.PubMed/NCBI View Article : Google Scholar

16 

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.PubMed/NCBI View Article : Google Scholar

17 

Milovic V, Teller I, Murphy G, Caspary W and Stein J: Deoxycholic acid stimulates migration in colon cancer cells. Eur J Gastroenterol Hepatol. 13:945–949. 2001.PubMed/NCBI View Article : Google Scholar

18 

Institute of Laboratory Animal Research, Commission on Life Sciences, National Research Council. Guide for the Care and Use of Laboratory Animals. Publication. 327:963–965. 1996.

19 

Majumder S, Caccamo A, Medina DX, Benavides AD, Javors MA, Kraig E, Strong R, Richardson A and Oddo S: Lifelong rapamycin administration ameliorates age-dependent cognitive deficits by reducing IL-1β and enhancing NMDA signaling. Aging Cell. 11:326–335. 2012.PubMed/NCBI View Article : Google Scholar

20 

Lv L, Shen Z, Zhang J, Zhang H, Dong J, Yan Y, Liu F, Jiang K, Ye Y and Wang S: Clinicopathological significance of SIRT1 expression in colorectal adenocarcinoma. Med Oncol. 31(965)2014.PubMed/NCBI View Article : Google Scholar

21 

He Z, Yi J, Jin L, Pan B, Chen L and Song H: Overexpression of Sirtuin-1 is associated with poor clinical outcome in esophageal squamous cell carcinoma. Tumor Biol. 37:7139–7148. 2016.PubMed/NCBI View Article : Google Scholar

22 

Guertin DA and Sabatini DM: An expanding role for mTOR in cancer. Trends Mol Med. 11:353–361. 2005.PubMed/NCBI View Article : Google Scholar

23 

Hudes G, Carducci M, Tomczak P, Dutcher J, Figlin R, Kapoor A, Staroslawska E, Sosman J, McDermott D, Bodrogi I, et al: Temsirolimus, interferon alfa, or both for advanced renal-cell carcinoma. N Engl J Med. 356:2271–2281. 2007.PubMed/NCBI View Article : Google Scholar

24 

Hou G, Zhang Q, Wang L, Liu M, Wang J and Xue L: mTOR inhibitor rapamycin alone or combined with cisplatin inhibits growth of esophageal squamous cell carcinoma in nude mice. Cancer Lett. 290:248–254. 2010.PubMed/NCBI View Article : Google Scholar

25 

Shen Z, Xu L, Li J and Zhang N: Capilliposide C sensitizes esophageal squamous carcinoma cells to oxaliplatin by inducing apoptosis through the PI3K/Akt/mTOR pathway. Med Sci Monit. 23:2096–2103. 2017.PubMed/NCBI View Article : Google Scholar

26 

Sohda M and Kuwano H: Current status and future prospects for esophageal cancer treatment. Ann Thorac Cardiovasc Surg. 23:1–11. 2017.PubMed/NCBI View Article : Google Scholar

27 

Hanigan MH and Devarajan P: Cisplatin nephrotoxicity: Molecular mechanisms. Cancer Ther. 1:47–61. 2003.PubMed/NCBI

28 

Xu Y, Xie Z, Shi Y, Zhang M, Pan J, Li Y and Lu H: Gefitinib single drug in treatment of advanced esophageal cancer. J Cancer Res Ther. 12 (Suppl):C295–C297. 2016.PubMed/NCBI View Article : Google Scholar

29 

Fire A, Xu S, Montgomery M, Kostas S, Driver S and Mello C: Potent and specific genetic interference by double-stranded RNA in Caenorhabditis elegans. Nature. 391:806–811. 1998.PubMed/NCBI View Article : Google Scholar

30 

Hwang B, Madabushi A, Jin J, Lin S and Lu A: Histone/protein deacetylase SIRT1 is an anticancer therapeutic target. Am J Cancer Res. 4:211–221. 2014.PubMed/NCBI

31 

Qin T, Liu W, Huo J, Li L, Zhang X, Shi X, Zhou J and Wang C: SIRT1 expression regulates the transformation of resistant esophageal cancer cells via the epithelial-mesenchymal transition. Biomed Pharmacother. 103:308–316. 2018.PubMed/NCBI View Article : Google Scholar

32 

Sarbassov D, Ali S, Sengupta S, Sheen JH, Hsu PP, Bagley AF, Markhard AL and Sabatini DM: Prolonged rapamycin treatment inhibits mTORC2 assembly and Akt/PKB. Mol Cell. 22:159–168. 2006.PubMed/NCBI View Article : Google Scholar

33 

Chan T, Rittenhouse S and Tsichlis P: AKT/PKB and other D3 phosphoinositide-regulated kinases: Kinase activation by phosphoinositide-dependent phosphorylation. Ann Rev Biochem. 68:965–1014. 1999.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

October-2021
Volume 22 Issue 4

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu T, Liang X, Sun Y and Yang S: Rapamycin suppresses the PI3K/AKT/mTOR signaling pathway by targeting <em>SIRT1</em> in esophageal cancer Retraction in /10.3892/etm.2023.12288. Exp Ther Med 22: 1190, 2021
APA
Liu, T., Liang, X., Sun, Y., & Yang, S. (2021). Rapamycin suppresses the PI3K/AKT/mTOR signaling pathway by targeting <em>SIRT1</em> in esophageal cancer Retraction in /10.3892/etm.2023.12288. Experimental and Therapeutic Medicine, 22, 1190. https://doi.org/10.3892/etm.2021.10624
MLA
Liu, T., Liang, X., Sun, Y., Yang, S."Rapamycin suppresses the PI3K/AKT/mTOR signaling pathway by targeting <em>SIRT1</em> in esophageal cancer Retraction in /10.3892/etm.2023.12288". Experimental and Therapeutic Medicine 22.4 (2021): 1190.
Chicago
Liu, T., Liang, X., Sun, Y., Yang, S."Rapamycin suppresses the PI3K/AKT/mTOR signaling pathway by targeting <em>SIRT1</em> in esophageal cancer Retraction in /10.3892/etm.2023.12288". Experimental and Therapeutic Medicine 22, no. 4 (2021): 1190. https://doi.org/10.3892/etm.2021.10624