Open Access

Stomatin‑like protein 2 induces metastasis by regulating the expression of a rate‑limiting enzyme of the hexosamine biosynthetic pathway in pancreatic cancer

  • Authors:
    • Dang Chao
    • Kyohei Ariake
    • Satoko Sato
    • Hideo Ohtsuka
    • Tatsuyuki Takadate
    • Masaharu Ishida
    • Kunihiro Masuda
    • Shimpei Maeda
    • Takayuki Miura
    • Katsutaka Mitachi
    • Xun Jing Yu
    • Fumiyoshi Fujishima
    • Masamichi Mizuma
    • Kei Nakagawa
    • Takanori Morikawa
    • Takashi Kamei
    • Michiaki Unno
  • View Affiliations

  • Published online on: April 5, 2021     https://doi.org/10.3892/or.2021.8041
  • Article Number: 90
  • Copyright: © Chao 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

Stomatin‑like protein 2 (SLP‑2) is associated with poor prognosis in several types of cancer, including pancreatic cancer (PC); however, the molecular mechanism of its involvement remains elusive. The present study aimed to elucidate the role of this protein in the development of PC. Human PC cell lines AsPC‑1 and PANC‑1 were transfected by a vector expressing SLP‑2 shRNA. Analyses of cell proliferation, migration, invasion, chemosensitivity, and glucose uptake were conducted, while a mouse xenograft model was used to evaluate the functional role of SLP‑2 in PC. Immunohistochemical analysis was retrospectively performed on human tissue samples to compare expression between the primary site (n=279) and the liver metastatic site (n=22). Furthermore, microarray analysis was conducted to identify the genes correlated with SLP‑2. In vitro analysis demonstrated that cells in which SLP‑2 was suppressed exhibited reduced cell motility and glucose uptake, while in vivo analysis revealed a marked decrease in the number of liver metastases. Immunohistochemistry revealed that SLP‑2 was increased in liver metastatic sites. Microarray analysis indicated that this protein regulated the expression of glutamine‑fructose‑6‑phosphate transaminase 2 (GFPT2), a rate‑limiting enzyme of the hexosamine biosynthesis pathway. SLP‑2 contributed to the malignant character of PC by inducing liver metastasis. Cell motility and glucose uptake may be induced via the hexosamine biosynthesis pathway through the expression of GFPT2. The present study revealed a new mechanism of liver metastasis and indicated that SLP‑2 and its downstream pathway could provide novel therapeutic targets for PC.

Introduction

Pancreatic cancer (PC) is one of the most aggressive types of cancer and its prognosis is particularly poor, among gastrointestinal cancers (1). The main reasons for this are late diagnosis and early recurrence, even after curative resection (2,3). Our previous research demonstrated that, among several types of recurrence, prognosis after peritoneal recurrence and liver metastasis was markedly poor compared with other types of recurrence (3). Previous studies have also revealed that these malignant recurrences may be predicted by measuring the glucose activity of cancer cells via the maximum standardized uptake values (SUVmax) obtained from 18-fluorodeoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) (4,5). The data suggest that high glucose uptake promotes metastasis of PC cells, causing poor prognosis. However, the detailed mechanism of metastasis remains unclear.

Stomatin-like protein 2 (SLP-2) is mainly located in the mitochondrial inner membrane (6,7). It has been reported to play important roles in regulating mitochondrial membrane stability (7), the formation of mitochondrial respiratory chain super-complexes (8), and in modulating mitochondrial sodium-calcium exchange (9). SLP-2 is also required for stress-induced mitochondrial hyperfusion (SIMH) and its expression is upregulated under conditions of mitochondrial stress (10). This stress condition stimulates mitochondrial biogenesis and function (11), providing the energetic requirements of activation.

Several previous studies have demonstrated that increased SLP-2 expression induces poor prognosis (1222). An increasing number of studies have revealed that SLP-2 is implicated in tumor progression and development. The depletion of SLP-2 has been revealed to inhibit the capability of cells to proliferate in colorectal cancer and esophageal squamous cell carcinoma (15,23). Migration and invasion activities have also been revealed to be inhibited after SLP-2 suppression in glioma and liver cancer (18,24). Furthermore, SLP-2 inhibited chemotherapy-induced apoptosis in cervical cancer and in head and neck squamous cell carcinoma (22,25). These findings indicated that the role and the mechanism of SLP-2 in inducing poor prognosis differs according to the origin of the cancer.

It was previously reported by our research group that SLP-2 is a novel prognostic biomarker of PC, based on the results of proteomic analysis (26). However, the function and molecular mechanism of SLP-2 in PC had not been thoroughly explored to date, and no other correlation between SLP-2 and PC had been reported.

The aim of the present study was to explore the function of SLP-2 in PC, using in vitro and in vivo assays. The level of SLP-2 expression at metastatic sites compared to that at primary sites was analyzed, to evaluate its metastatic potential.

Materials and methods

Antibodies, reagents, and cell lines

Antibodies against SLP-2 were purchased from ProteinTech Group, Inc. (cat. no. 10348-1-AP); GAPDH from Cell Signaling Technology, Inc. (product no. 2118S); GFPT2 from Abcam (product code ab190966); and anti-rabbit IgG as a secondary antibody (cat. no. A0545) was obtained from Sigma-Aldrich; Merck KGaA. The cell lines AsPC-1, BxPC-3, SUIT-2, and SW1990 were purchased from the American Type Culture Collection. The cell line PANC-1 was obtained from RIKEN and MIA-PaCa2 was provided from Cell Resource Center for Biomedical Research, Institute of Development, Aging and Center, Tohoku University (Sendai, Japan). Cells were expanded within 3 passages after being purchased, and multiple lots were stocked at −80°C. Mycoplasma contamination check tests were performed using e-Myco plus Mycoplasma PCR Detection Kit (iNtRON Biotechnology, Inc.). Cells were used at least <20 passages, but were not independently authenticated.

Transfection with short hairpin (sh)RNA

The sequences of SLP-2 shRNAs are presented in Table SI. The shRNAs were inserted into pBAsi-hU6 NEO plasmids (cat. no. 3227; Takara Bio, Inc.), which carry a neomycin resistance gene. Lipofectamine 2000 reagent (cat. no. 11668019; Thermo Fisher Scientific, Inc.) was used for plasmid transfections into PANC-1 and AsPC-1, according to the manufacturer's protocol. In short, 20 µg plasmids with 2 ml Opti-MEM (cat. no. 31985070; Thermo Fisher Scientific, Inc.) and 30 µl Lipofectamine 2000 with Opti-MEM were produced. Then, these mediums were combined at room temperature for 20 min. After mixture, this medium was placed in a 10-mm dish and cells were cultured at 37°C for 12 h. After exposure, the medium was changed to RPMI-1640 medium supplemented with 1,000 mg/ml Geneticin® (cat. no. 10131-027; Thermo Fisher Scientific, Inc.). The blank pBAsi-hU6 NEO plasmid was also transfected as a negative control. Transfected clones, on RPMI-1640 medium supplemented with 1,000 mg/ml Geneticin®, were selected over a period of 3 weeks, after which a single colony was selected and cultured.

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR)

Total RNA was isolated from cancer cells using the Nucleospin RNA kit (cat. no. 740955; Takara Bio, Inc.). Reverse transcription reactions were set up using PrimeScript RT Master Mix (cat. no. RR036A; Takara Bio, Inc.), according to the manufacturer's protocol under the following thermocycling conditions: 37°C for 15 min, followed by 85°C for 5 sec, and the products were used as templates for RT-qPCR. The gene products were amplified using TB Green Premix Ex Taq II, ROX Plus (cat. no. RR82LR; Takara Bio, Inc.) under the following thermocycling conditions: 95°C for 30 sec, followed by 40 cycles at 95°C for 5 sec for denaturation and 60°C for 30 sec for annealing/extension. The expression levels of each target gene were calculated using the 2−ΔΔCq method (27). Relative quantities were calculated after normalizing for GAPDH expression. The primers used in the present study are presented in Table SI.

RNA preparation and microarray analyses

RNA isolates were obtained from AsPC-1cont, AsPC-1sh1, and AsPC-1sh2 cells using the Nucleospin RNA kit and assessed for quality using a NanoDrop spectrophotometer (Thermo Fisher Scientific, Inc.). The GeneChip™ WT PLUS Reagent Kit (cat. no. 902281; Thermo Fisher Scientific, Inc.) was used to prepare the RNA samples; for whole-transcriptome expression analyses, the Clariom™ S assay (cat. no. 902926; Thermo Fisher Scientific, Inc.) was used. The GeneChip was analyzed using a GeneChip™ Scanner 3000 7G system, while the gene expression was analyzed using a GeneTitan™ instrument (both from Thermo Fisher Scientific, Inc.). The transcriptomic array data set was analyzed using the Transcriptome Analysis Console software Ver 4.0 (Thermo Fisher Scientific, Inc.). Genes with fold-changes of <-20 or >20 and with a P-value <0.05 in comparisons between AsPC-1cont and AsPC-1sh1 or AsPC-1sh2 were selected as candidate genes associated with SLP-2 expression. The data of microarray analysis are available in Gene Expression Omnibus at https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE162981.

Western blotting

Cells were lysed with RIPA buffer (Thermo Fisher Scientific, Inc.), and the concentrations of protein samples were evaluated using a BCA kit (Thermo Fisher Scientific, Inc.). A total of 20 µg of proteins were separated on SDS-PAGE gels (4-15%) and subsequently transferred onto PVDF membranes (Bio-Rad Laboratories, Inc.). The membranes were then blocked at room temperature for 1 h using Western Blocking Reagent (Thermo Fisher Scientific, Inc.) and incubated with primary antibodies (SLP-2 and GFPT-2) at room temperature for 1 h at dilutions of 1:2,000. PVDF membranes were then incubated with secondary antibodies at room temperature for 30 min (HRP-conjugated) at dilutions of 1:5,000, and signals were detected using the Clarity ECL Western Substrate (cat. no. 1705062; Bio-Rad Laboratories, Inc.). Protein bands were visualized using an ImageQuant LAS 4000 mini system (GE Healthcare; Cytiva).

Cell proliferation assay

A total concentration of 5×103 cells was seeded in 96-well plates and cultured with 100 µl of RPMI-1640 medium with 10% FBS, 1% penicillin, and 1,000 mg/ml geneticin. The MTS assays were performed using CellTiter 96-well assay reagent (cat. no. G358B; Promega Corporation), according to the manufacturer's recommendations. The absorbance was measured at a wavelength of 490 nM. The experiment was performed in triplicate.

Wound-healing assay

Cells grown on petri dishes were starved in serum-free medium for 24 h and then scratched with the tip of a sterile 10-µl pipette. Following this, the cultured cells were rinsed and incubated with RPMI-1640 medium containing 10% FBS, 1% penicillin, and 1,000 mg/ml geneticin. After 12 h, the wound-closure distances were measured at three independent wound sites per group using a light microscope, and the average was calculated. The data are expressed as a relative index considering the differences in wound length at the initial time-point. The assays were performed in triplicate.

Transwell cell migration and invasion assay

Transwell assays were performed using the QCM 24-well Fluorometric Cell Migration Assay kit (cat. no. ECM509; EMD Millipore) for the migration assay and the QCM 24-well Cell Invasion Assay kit using Matrigel®-coated Transwell chambers (cat. no. ECM554; EMD Millipore) for the invasion assay. The cells were pretreated with serum-free medium for 24 h, and then harvested in a serum-free medium. Then, 300-µl cell samples were transferred to the upper chambers of the kit (1×106/ml), and 500 µl of medium containing 10% FBS for the migration assay and 20% FBS for the invasion assay was added to the lower chamber. After incubation for 24 h, the cells were dislodged using 225 µl of cell detachment solution from the underside of the upper chamber for 30 min at 37°C. Thereafter, the samples were stained with sufficient lysis buffer/dye solution for 15 min at 20–25°C. The results were quantified using a fluorescence plate reader fitted with a 480/520 nm filter. The data are expressed using a relative index, by setting the migrated or invasive control cells to 100%. This assay was performed in triplicate.

Cytotoxicity assay

Cells were plated at 5×103 cells/well in 96-well plates. After 24 h, the medium was replaced by another containing from 1×10−4 to 1×103 µM gemcitabine hydrochloride (FUJIFILM WAKO Pure Chemical Corporation). Cells were cultured for 120 h and then cell viability was measured by MTS assays using the CellTiter 96-well assay reagent (Promega Corporation), as recommended by the manufacturer. The absorbance was measured at a wavelength of 490 nM. Each test was carried out in triplicate.

Glucose uptake assay

The Glucose Uptake-Glo Assay (cat. no. J1342; Promega Corporation) was applied to cells grown in 96-well plates. Glucose-free media were used throughout the steps of this assay. A total of 50 µl of 1 mM 2-deoxyglucose was added to each well to initiate the assay. The uptake reaction was stopped according to the manufacturer's protocol, after 2 h of incubation. The luminescence was recorded using 1-sec integration on a luminometer. These experiments were performed in triplicate.

In vivo experiments

The Institutional Animal Experiment Committee of Tohoku University (Sendai, Japan) approved the present study protocol on September 20, 2017. AsPC-1 stable cells (blank or shRNA-transfected) were harvested from 80% confluent culture dishes, resuspended in PBS, and maintained on ice. Six-week-old SCID mice (C.B-17/Icr-scid/scid Jcl; male; weight, 23 g) were used for this experiment. A triple-mixed anesthetic was prepared, consisting of medetomidine (1 mg/ml), midazolam (5 mg/ml), and butorphanol (5 mg/ml); this solution was injected intraperitoneally into the mice (0.1 ml per 10 g body weight per mouse). An incision was made to exteriorize the spleens, to inject 5×105 cells per 100 µl slowly into them. A cotton swab was held over the injection site for at least 5 min to avoid leakage and bleeding. After confirming that the bleeding had stopped, the spleens were returned into the peritoneal cavities, and the abdominal wounds were closed. Six weeks after the injection, the mice were sacrificed using 60 µl of pentobarbital (50 mg/ml), and their livers were removed. The number of metastatic sites on the liver surface was counted. Three biological replicates were used for each experiment.

Immunohistochemistry

All specimens for immunohistochemistry were fixed for 24 h in 10% formalin and embedded in paraffin wax. Two specialists from the Department of Pathology of Tohoku University performed the SLP-2 immunostaining procedures and scored the immunoreactivity. The staining intensity combined with the positive cell percentages were used to obtain a semiquantitative analysis of immunoreactivity. The staining intensity was scored as 0 (negative), 1 (weak), 2 (moderate), or 3 (strong) (Fig. S1); the positive cell percentage was scored as 0 (0%), 1 (<10%), 2 (10-50%), 3 (51-80%), or 4 (>80%). The staining intensity and positive cell percentages were multiplied to evaluate the immunoreactive scores (IRS), which ranged from 0 to 12.

Clinical data concerning 279 patients (166 male and 113 female patients; aged 27–88 years old; median age, 67 years) with pancreatic ductal adenocarcinoma, who underwent surgical resection between January 1, 2006 and December 31, 2014, were obtained from records at our institute on January 1, 2020. The study included resectable to unresectable PDAC, NAC and non-NAC as well as R0 to R2 patients. Radiographic resection status was defined by the National Comprehensive Cancer Network guidelines for pancreatic cancer, Version 1 (2020) (28) and pathological status was diagnosed by the Union for International Cancer Control TNM classification (7th edition) (29). The Institutional Review Board of Tohoku University (Sendai, Japan) approved the present study design on May 25, 2016 (2016-1-151). As this was a retrospective study, the requirement for informed consent was waived and an opt-out method was used instead. The present study was conducted in accordance with the STROBE guidelines (www.strobe-statement.org) (30).

Statistical analysis

Statistical analysis was conducted on the JMP pro software v14 (SAS Institute). Data are expressed as the mean ± standard deviation (SD). Student's t-test was used to compare the means of two groups, while ANOVA followed by Dunnett's post hoc test were used to compare the means of multiple experimental groups. The binomial variables of clinicopathological factors were compared using Pearson's chi-square test. P-values <0.05 were considered to indicate a statistically significant difference.

Results

Cloning a stable SLP-2 silencing cell line

SLP-2 expression was evaluated thrice in six types of PC cell lines (PANC-1, MIA-PaCa2, AsPC-1, BXPC-3, SUIT-2, and SW1990) (Fig. 1A). SLP-2 expression in AsPC-1 and PANC-1 cells was relatively high compared with that in other cell lines; hence, these cell lines were selected for further analysis.

The designed shRNA was inserted into the pBAsi-hU6 NEO plasmid and transfected into AsPC-1 and PANC-1 cells. By RT-qPCR (Fig. 1B) and western blotting (Figs. 1C and S2A), it was revealed that SLP-2 expression was significantly decreased in shRNA-transfected cells.

SLP-2 silencing reduces cell migration and invasion abilities

To evaluate the effect of SLP-2 expression on cell motility, in vitro wound-healing assays were performed (Fig. 2A and B). The results revealed that wound widths were decreased in AsPC-1sh1 (P=0.017) and AsPC-1sh2 (P<0.001) compared with the widths observed in AsPC-1cont cells. Similar results were also revealed in PANC-1sh1 (P<0.001) and PANC-1sh2 (P<0.001) cells. In addition, Transwell cell migration assays also demonstrated that the migration abilities of AsPC-1sh1 and AsPC-1sh2 cells were decreased to 30.0% (P<0.001) and 34.2% (P<0.001), respectively, in comparison with the corresponding migration ability of AsPC-1cont cells (Fig. 2C). The same results were also demonstrated in PANC-1 cells, with migration abilities decreased to 41.6% (P<0.001) in PANC-1sh1 and 51.8% (P<0.001) in PANC-1sh2 cells. Furthermore, the results of invasion assay using Matrigel®-coated Transwell chambers revealed that the invasive activity of AsPC-1sh1 and AsPC-1sh2 cells was decreased to 67.2% (P=0.002) and 67.9% (P=0.002), respectively, compared to the corresponding activity level in control cells (Fig. 2D). The results in PANC-1 cells also confirmed a decrease in activity to 35.8% (P<0.001) in PANC-1sh1 cells and 44.7% (P<0.001) in PANC-1sh2 cells.

Inhibition of SLP-2 expression reduces the glucose uptake in PC cells

Glucose uptake was evaluated in different SLP-2-expressing PC cells. As revealed in Fig. 3A, compared to AsPC-1cont cells, glucose uptake was significantly decreased to 75.6% in AsPC-1sh1 (P=0.042) and 75.0% in AsPC-1sh2 cells (P=0.038). The same was observed in PANC-1 cells; it was decreased to 46.3% in PANC-1sh1 (P<0.001) and to 35.3% in PANC-1sh2 cells (P<0.001) (Fig. 3B).

SLP-2 does not affect cell proliferation and chemosensitivity to gemcitabine

An MTS assay demonstrated that the cell growth curves were almost identical regardless of the expression of SLP-2 in both AsPC-1 and PANC-1 cells. Additionally, no significant differences were found in the four-day growth rate of cells among AsPC-1cont, AsPC-1sh1 and AsPC-1sh2 cells (P=0.944) (Fig. S3A). Similar results were revealed among PANC-1cont cells, PANC-1sh1and PANC-1sh2 cells (P=0.532) (Fig. S3B).

To investigate whether SLP-2 affects the chemosensitivity of PC, cells were treated with different doses of gemcitabine. The present findings revealed no significant differences in the IC50 values of AsPC-1cont (0.069 µM), AsPC-1sh1 (0.066 µM), and AsPC-1sh2 cells (0.090 µM) (P=0.713) (Fig. S4A). No significant differences were also observed in the IC50 values of PANC-1cont cells (0.053 µM), PANC-1sh1 (0.053 µM), and PANC-1sh2 cells (0.053 µM) (P=0.989) (Fig. S4B).

SLP-2 expression was significantly positively correlated with liver metastasis in PC

To investigate the role of SLP-2 during liver metastasis, different SLP-2-expressing PC cells were injected into the spleen of SCID mice. A previous study revealed that only AsPC-1 could cause liver metastasis in SCID mice, among several PC cell lines such as PANC-1, MIA PaCa-2, AsPC-1, and BxPC-3 (31). In agreement with the previous study, PANC-1 cells did not cause liver metastasis, even in control cells (data not shown). Therefore, AsPC-1 cells were used to evaluate the potential of SLP-2 to cause liver metastasis. Six weeks after the injections, the mean number of liver metastases was significantly decreased from 22 in the AsPC-1cont cells, to 2.3 (P<0.001) in AsPC-1sh1, and 1.6 in AsPC-1sh2 (P<0.001) (Fig. 4A and B).

To validate the results of the in vivo analysis, SLP-2 expression level was measured from the primary sites and liver metastatic sites in PC tissue samples from patients, using immunohistochemistry. First, the SLP-2 expression of the two tissues from the group without neoadjuvant chemotherapy (non-NAC) was compared. The IRS was significantly increased in the metastatic site compared with that at the primary site (8.9 vs. 5.9; P=0.021) (Fig. 4C). Next, the IRS of the two sites was compared in patients after neoadjuvant chemotherapy (the NAC group). Similarly, the IRS was significantly increased at the metastatic site compared with that at the primary site (9.6 vs. 7.0; P=0.008) (Fig. 4D).

SLP-2 expression is associated with the expression of GFPT2

To define the SLP-2-associating factor in PC cells, a microarray analysis was performed using AsPC-1cont cells and AsPC-1sh1 or AsPC-1sh2 cells. The data of microarray analysis are available in Gene Expression Omnibus at https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE162981. It was determined that 8 candidate genes were overexpressed by >20-fold and 7 genes were downregulated by >20-fold in AsPC-1cont cells compared with the corresponding expression levels of the genes in SLP-2-suppressed cells (Fig. 5A). In the present study, the expression of glutamine-fructose-6-phosphate transaminase 2 (GFPT2) as a new candidate gene associated with SLP-2 expression was evaluated; this is because, from the literature (3234), it appears that only GFPT2 could regulate cell motility and glucose uptake activity. Expression of the GFPT2 gene was confirmed in PANC-1 cells using RT-qPCR and western blotting and was revealed to be downregulated in SLP-2-suppressed cells (Figs. 5B and C, and S2B).

SLP-2 expression is increased after NAC

Table I reveals the clinicopathological characteristics of both the non-NAC and NAC groups. The distribution between the two groups revealed a significant difference in resectability status. This is because borderline resectable or unresectable cases usually underwent preoperative treatment before surgical resection. To evaluate whether SLP-2 expression differed according to tumor progression, SLP-2 expression was analyzed for each resectability status. For patients in the non-NAC group, the mean IRS did not exhibit significant differences among the three groups (R vs. BR vs. UR, 6.1 vs. 5.8 vs. 6.0; P=0.839) (Fig. S5A). The same results were also revealed in the NAC group (R vs. BR vs. UR, 6.3 vs. 7.1 vs. 7.3; P=0.534) (Fig. S5B). These results demonstrated that SLP-2 expression did not differ according to the resectability status. However, when the SLP-2 expression profile of the non-NAC group was compared with that of the NAC group, the IRS was significantly higher in the NAC group (5.9 vs. 7.0; P=0.019) (Fig. S5C). All these results demonstrated that NAC itself may increase SLP-2 expression.

Table I.

Distribution between non-NAC and NAC patients.

Table I.

Distribution between non-NAC and NAC patients.

Distribution between non-NAC and NAC patients

Clinical/pathological variables Total (n=279)non-NAC (n=153)NAC (n=126)P-value
ResectabilityResectable1178829<0.001
Borderline1386474
Resectable
Unresectable24123
Pretreatment CA19-9 (U/ml)>10011971480.163
≤1001608278
Tumor positionph195102930.196
pbt845133
Tumor size (mm)>3013869690.108
≤301418457
UICC-T1,214950.141
3265144121
UICC-N07737400.371
120211686
UICC-M0224125990.176
1552827
Residual cancerR02441341100.944
R1,2351916

[i] The UICC-N category describes the presence of regional lymph node metastasis. The UICC-M category describes the presence of distant metastasis. NAC, neoadjuvant chemotherapy; UICC, Union for International Cancer Control. The UICC-T category describes the status of primary tumor.

Gemcitabine exposure increases SLP-2 expression in PC cells

To evaluate the hypothesis that NAC itself promotes the expression of SLP-2, its expression was evaluated after exposure to gemcitabine. SLP-2 expression was significantly increased in PANC-1 cells after exposure to 100 or 1,000 µM gemcitabine (Fig. 6A). Next, the effect of SLP-2 on GFPT2 expression was analyzed. GFPT2 expression was also significantly enhanced after gemcitabine exposure, and inhibition of SLP-2 suppressed the expression of GFPT2 (Fig. 6B). These results demonstrated that gemcitabine promoted SLP-2 expression in PC cells, and subsequently increased the expression of GFPT2. Suppression of SLP-2 could also inhibit gemcitabine-induced GFPT2 expression.

Discussion

In the present study, with regard to PC, suppression of SLP-2 did not alter the cell proliferation or chemosensitivity to gemcitabine. Conversely, it reduced cell migration and invasion activities. Suppression of SLP-2 significantly decreased the number of liver metastases in the mouse xenograft model. Furthermore, immunohistochemical results revealed that SLP-2 expression was increased at the site of liver metastasis. This confirmed that SLP-2 expression plays an important role in liver metastasis and that increased SLP-2 expression is linked to liver metastasis in PC, which may explain the poor prognosis.

SLP-2 has been revealed to regulate cell migration and invasion activities in several types of cancer. Dowling et al revealed that SLP-2 is one of the 16 most upregulated proteins in extremely invasive cancer cells, implying that SLP-2 may be active during cancer metastasis (35). However, the signaling pathway affecting cell migration and invasion activities differs according to the type of cancer. In liver cancer, SLP-2 expression was revealed to promote EMT progression (18). SLP-2 facilitated migration activity via regulating the Wnt/β-catenin pathway in colorectal cancer (15). In glioma and liver cancer, SLP-2 was revealed to regulate cell motility via the NF-κB pathway, targeting MMP2 or MMP9 (17,24). In the present study, to elucidate the mechanism in PC cells, microarray analyses were performed using SLP-2-suppressed PC cells. Among 15 candidate genes selected by the expression level of SLP-2, only GFPT2 was associated with cell death and glucose uptake activity. Therefore, GFPT2 was selected as an optimal new candidate genes regulated by SLP-2 expression to promote cell migration and invasion activities in PC cells.

GFPT2 is the first, and rate-limiting, enzyme of the hexosamine biosynthesis pathway (HBP). HBP is a branch of glucose metabolism that usually consumes approximately 2–5% of total glucose (36). It has also been revealed that GFPT2 is correlated with glucose uptake and is associated with glucose-driven metabolic pathways (32). Thus, increased GFPT2 expression could activate HBP by supplying the glucose itself and also by modulating the glucose substrate to this pathway. The HBP contributes to the provision of a substrate for glycosylation modification and has a wide range of effects on cellular function (32). Uridine diphosphate N-acetylglucosamine (UDP-GlcNAc), an end-product of the HBP, is catalyzed by O-GlcNAc transferase to form O-GlcNAc, which binds to serine/threonine residues of the target proteins in glycosylation modification (33). Increased O-GlcNAcylation can activate various cancer-related factors such as β-catenin (33), p53 (37), or c-Myc (38). In particular, O-GlcNAcylation of β-catenin enhances intranuclear β-catenin expression and facilitates signal activation, including EMT promotion and cancer invasion (33). Several previous studies have also demonstrated that GFPT2 can regulate cell migration and invasion activities by activating the EMT mechanism (32,34). Therefore, high SLP-2 expression may promote cell motility by regulating HBP through GFPT2 expression. However, the present study presented no evidence of the HBP modulation in vitro or in vivo; hence, further metabolic studies are warranted to evaluate the role of SLP-2 in affecting the GFPT2 downstream pathway in HBP.

SLP-2 has been reported to be localized in mitochondria and upregulated under several conditions of mitochondrial stress (7). This increased expression plays an important role in SIMH, which promotes mitochondrial ATP production and leads to stress resistance in cells (39). Exposure to high concentrations of cisplatin was revealed to increase SLP-2 expression and induce an anti-apoptotic effect on cells (25). This response is one possible mechanism for stress-resistant regulation of SLP-2 expression. The present study revealed that SLP-2 expression was increased after NAC. When PC cells were exposed to gemcitabine, expression was increased for SLP-2 and GFPT2. Furthermore, GFPT2 expression was decreased in SLP-2-suppressed cells, even in a stressed condition (such as exposure to anticancer drug gemcitabine). All these data indicated that SLP-2 expression may be increased by chemotherapy itself; additionally, this increase may induce activation of the HBP through upregulation of GFPT2. Recently, several studies have raised the possibility of chemotherapy-induced metastasis (40) and have suggested that cancer cells tend to become metastatically aggressive following exposure to chemotherapy (41). Although SLP-2 suppression did not alter chemosensitivity to gemcitabine, increased expression of SLP-2 after NAC may contribute to promoting liver metastasis. Further study should be conducted to investigate this hypothesis.

The present study has several limitations. First, only the function of SLP-2 was evaluated by inhibiting its expression. SLP-2 suppression did not significantly change the proliferation and chemosensitivity of PC cells to gemcitabine unlike previous findings (24,42). Overexpression of SLP-2 increased the ability of cells to produce ATP, which is necessary for proliferation and resistance to stress (11,23). Exposure of PC cells to gemcitabine also increased the expression of SLP-2, which may be a response to stress. Considering these data, overexpression may lead to diverse effects on cell functions. Second, the present study used G418 (geneticin) to select shRNA-transfected PC cells, and this agent may influence the growth and metabolism of cell lines (43). However, several examinations were performed, using two types of cell lines and two types of shRNA-transfected cell lines, to mitigate the effects of G418 and minimize this bias.

In conclusion, to the best of our knowledge, the present study is the first to analyze the effects of SLP-2 in PC. The results obtained herein demonstrated the involvement of SLP-2 in liver metastasis via regulation of the migration and invasion activities of PC cells. Furthermore, SLP-2 expression promoted glucose uptake activity and may regulate HBP, a branch of glucose metabolism, which can contribute to the activation of cancer cell motility. Furthermore, SLP-2 expression was increased under cytotoxic stress, implying that the aforementioned functions may be enhanced under the cellular stress caused by chemotherapy treatments. All these results indicated that the poor prognosis of high SLP-2 expression was caused by activated metastatic potential. Further investigations on HBP activation and its promotion of metastasis are warranted to determine whether these signals could provide novel therapeutic targets for PC.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was supported by KAKENHI Grants-in-Aid for young scientists (B) (grant no. K.A.16K19911).

Availability of data and materials

The data of microarray analysis are available in Gene Expression Omnibus at https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE162981 and can be accessed with GSE162981. The other datasets used and/or analyzed during this study are available from the corresponding author on reasonable request.

Authors' contributions

DC, KA, HO, KMi, XJY performed in vivo and in vitro analysis. KA, MM, TT, MI, KMa, SM, TMi, MM, KN, TMo, TK, MU interpreted the patient data regarding the pancreatic ductal adenocarcinoma. SS and FF performed the pathological examination of the pancreatic ductal adenocarcinoma samples. KA, TT, MI, KMa, SM, TMi, MM, KN, TMo, TK, MU were major contributors in writing the manuscript. All authors read and approved the final manuscript.

Ethics approval and consent to participate

The Institutional Animal Experiment Committee of Tohoku University (Sendai, Japan) approved the present study protocol on September 20, 2017. The Institutional Review Board of Tohoku University (Sendai, Japan) approved the present study design on May 25, 2016 (2016-1-151). This was a retrospective study, therefore, the requirement for informed consent was waived and an opt-out method was used instead.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Glossary

Abbreviations

Abbreviations:

EMT

epithelial-mesenchymal transition

GFPT2

glutamine-fructose-6-phosphate transaminase 2

HBP

hexosamine biosynthesis pathway

IRS

immunoreactive score

NAC

neoadjuvant chemotherapy

PC

pancreatic cancer

RT-qPCR

reverse transcription-quantitative polymerase chain reaction

SIMH

stress-induced mitochondrial hyperfusion

SLP-2

stomatin-like protein 2

References

1 

Siegel RL, Miller KD, Goding Sauer A, Fedewa SA, Butterly LF, Anderson JC, Cercek A, Smith RA and Jemal A: Colorectal cancer statistics, 2020. CA Cancer J Clin. 70:145–164. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Neoptolemos JP, Stocken DD, Friess H, Bassi C, Dunn JA, Hickey H, Beger H, Fernandez-Cruz L, Dervenis C, Lacaine F, et al: A randomized trial of chemoradiotherapy and chemotherapy after resection of pancreatic cancer. N Engl J Med. 350:1200–1210. 2004. View Article : Google Scholar : PubMed/NCBI

3 

Ariake K, Motoi F, Ohtsuka H, Fukase K, Masuda K, Mizuma M, Hayashi H, Nakagawa K, Morikawa T, Maeda S, et al: Predictive risk factors for peritoneal recurrence after pancreatic cancer resection and strategies for its prevention. Surg Today. 47:1434–1442. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Ariake K, Motoi F, Shimomura H, Mizuma M, Maeda S, Terao C, Tatewaki Y, Ohtsuka H, Fukase K, Masuda K, et al: 18-fluorodeoxyglucose positron emission tomography predicts recurrence in resected pancreatic ductal adenocarcinoma. J Gastrointest Surg. 22:279–287. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Yamamoto T, Sugiura T, Mizuno T, Okamura Y, Aramaki T, Endo M and Uesaka K: Preoperative FDG-PET predicts early recurrence and a poor prognosis after resection of pancreatic adenocarcinoma. Ann Surg Oncol. 22:677–684. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Hájek P, Chomyn A and Attardi G: Identification of a novel mitochondrial complex containing mitofusin 2 and stomatin-like protein 2. J Biol Chem. 282:5670–5681. 2007. View Article : Google Scholar

7 

Da Cruz S, Parone PA, Gonzalo P, Bienvenut WV, Tondera D, Jourdain A, Quadroni M and Martinou JC: SLP-2 interacts with prohibitins in the mitochondrial inner membrane and contributes to their stability. Biochim Biophys Acta. 1783:904–911. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Mitsopoulos P, Chang YH, Wai T, König T, Dunn SD, Langer T and Madrenas J: Stomatin-like protein 2 is required for in vivo mitochondrial respiratory chain supercomplex formation and optimal cell function. Mol Cell Biol. 35:1838–1847. 2015. View Article : Google Scholar : PubMed/NCBI

9 

Da Cruz S, De Marchi U, Frieden M, Parone PA, Martinou JC and Demaurex N: SLP-2 negatively modulates mitochondrial sodium-calcium exchange. Cell Calcium. 47:11–18. 2010. View Article : Google Scholar : PubMed/NCBI

10 

Tondera D, Grandemange S, Jourdain A, Karbowski M, Mattenberger Y, Herzig S, Da Cruz S, Clerc P, Raschke I, Merkwirth C, et al: SLP-2 is required for stress-induced mitochondrial hyperfusion. EMBO J. 28:1589–1600. 2009. View Article : Google Scholar : PubMed/NCBI

11 

Christie DA, Lemke CD, Elias IM, Chau LA, Kirchhof MG, Li B, Ball EH, Dunn SD, Hatch GM and Madrenas J: Stomatin-like protein 2 binds cardiolipin and regulates mitochondrial biogenesis and function. Mol Cell Biol. 31:3845–3856. 2011. View Article : Google Scholar : PubMed/NCBI

12 

Chang D, Ma K, Gong M, Cui Y, Liu ZH, Zhou XG, Zhou CN and Wang TY: SLP-2 overexpression is associated with tumour distant metastasis and poor prognosis in pulmonary squamous cell carcinoma. Biomarkers. 15:104–110. 2010. View Article : Google Scholar : PubMed/NCBI

13 

Liu D, Zhang L, Shen Z, Tan F, Hu Y, Yu J and Li G: Increased levels of SLP-2 correlate with poor prognosis in gastric cancer. Gastric Cancer. 16:498–504. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Li XH, He F, Yan SM, Li Y, Cao Y, Huang CY and Zhou ZW: Increased expression of stomatin-like protein 2 (STOML2) predicts decreased survival in gastric adenocarcinoma: A retrospective study. Med Oncol. 31:7632014. View Article : Google Scholar : PubMed/NCBI

15 

Zhou C, Li Y, Wang G, Niu W, Zhang J, Wang G, Zhao Q and Fan L: Enhanced SLP-2 promotes invasion and metastasis by regulating Wnt/β-catenin signal pathway in colorectal cancer and predicts poor prognosis. Pathol Res Pract. 215:57–67. 2019. View Article : Google Scholar : PubMed/NCBI

16 

Wang WX, Lin QF, Shen D, Liu SP, Mao WD, Ma G and Qi WD: Clinicopathological significance of SLP-2 overexpression in human gallbladder cancer. Tumour Biol. 35:419–423. 2014. View Article : Google Scholar : PubMed/NCBI

17 

Zhu W, Li W, Geng Q, Wang X, Sun W, Jiang H and Pu X: Silence of stomatin-like protein 2 represses migration and invasion ability of human liver cancer cells via inhibiting the nuclear factor kappa B (NF-κB) pathway. Med Sci Monit. 24:7625–7632. 2018. View Article : Google Scholar : PubMed/NCBI

18 

Huang Y, Chen Y, Lin X, Lin Q, Han M and Guo G: Clinical significance of SLP-2 in hepatocellular carcinoma tissues and its regulation in cancer cell proliferation, migration, and EMT. OncoTargets Ther. 10:4665–4673. 2017. View Article : Google Scholar

19 

Cao W, Zhang B, Li J, Liu Y, Liu Z and Sun B: SLP-2 overexpression could serve as a prognostic factor in node positive and HER2 negative breast cancer. Pathology. 43:713–718. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Zhang J, Song X, Li C and Tian Y: Expression and clinical significance of SLP-2 in ovarian tumors. Oncol Lett. 17:4626–4632. 2019.PubMed/NCBI

21 

Deng H, Deng Y, Liu F, Chen J, Li Z, Zhao K, Guan X and Liang W: Stomatin-like protein 2 is overexpressed in cervical cancer and involved in tumor cell apoptosis. Oncol Lett. 14:6355–6364. 2017.PubMed/NCBI

22 

Qu H, Jiang W, Wang Y and Chen P: STOML2 as a novel prognostic biomarker modulates cell proliferation, motility and chemo-sensitivity via IL6-Stat3 pathway in head and neck squamous cell carcinoma. Am J Transl Res. 11:683–695. 2019.PubMed/NCBI

23 

Wang Y, Cao W, Yu Z and Liu Z: Downregulation of a mitochondria associated protein SLP-2 inhibits tumor cell motility, proliferation and enhances cell sensitivity to chemotherapeutic reagents. Cancer Biol Ther. 8:1651–1658. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Song L, Liu L, Wu Z, Lin C, Dai T, Yu C, Wang X, Wu J, Li M and Li J: Knockdown of stomatin-like protein 2 (STOML2) reduces the invasive ability of glioma cells through inhibition of the NF-κB/MMP-9 pathway. J Pathol. 226:534–543. 2012. View Article : Google Scholar : PubMed/NCBI

25 

Hu G, Zhang J, Xu F, Deng H, Zhang W, Kang S and Liang W: Stomatin-like protein 2 inhibits cisplatin-induced apoptosis through MEK/ERK signaling and the mitochondrial apoptosis pathway in cervical cancer cells. Cancer Sci. 109:1357–1368. 2018. View Article : Google Scholar : PubMed/NCBI

26 

Takadate T, Onogawa T, Fukuda T, Motoi F, Suzuki T, Fujii K, Kihara M, Mikami S, Bando Y, Maeda S, et al: Novel prognostic protein markers of resectable pancreatic cancer identified by coupled shotgun and targeted proteomics using formalin-fixed paraffin-embedded tissues. Int J Cancer. 132:1368–1382. 2013. View Article : Google Scholar : PubMed/NCBI

27 

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

28 

National Comprehensive Cancer Network, . NCCN practice guidelines for pancreatic cancer. Version 1. (2020).https://www.nccn.org/professionals/physician_gls/PDF/pancreatic.pdfSeptember 1–2020

29 

UICC, . TNM Classification of Malignant Tumours. 7th edition. Sobin LH, Gospodarowicz MK and Wittekind C: Wiley Blackwell; Hoboken: 2009

30 

von Elm E, Altman DG, Egger M, Pocock SJ, Gøtzsche PC and Vandenbroucke JP; STROBE Initiative, : The strengthening the reporting of observational studies in epidemiology (STROBE) statement: Guidelines for reporting observational studies. J Clin Epidemiol. 61:344–349. 2008. View Article : Google Scholar : PubMed/NCBI

31 

Suemizu H, Monnai M, Ohnishi Y, Ito M, Tamaoki N and Nakamura M: Identification of a key molecular regulator of liver metastasis in human pancreatic carcinoma using a novel quantitative model of metastasis in NOD/SCID/gammacnull (NOG) mice. Int J Oncol. 31:741–751. 2007.PubMed/NCBI

32 

Zhang W, Bouchard G, Yu A, Shafiq M, Jamali M, Shrager JB, Ayers K, Bakr S, Gentles AJ, Diehn M, et al: GFPT2-expressing cancer-associated fibroblasts mediate metabolic reprogramming in human lung adenocarcinoma. Cancer Res. 78:3445–3457. 2018.PubMed/NCBI

33 

Zhou L, Luo M, Cheng LJ, Li RN, Liu B and Linghu H: Glutamine-fructose-6-phosphate transaminase 2 (GFPT2) promotes the EMT of serous ovarian cancer by activating the hexosamine biosynthetic pathway to increase the nuclear location of β-catenin. Pathol Res Pract. 215:1526812019. View Article : Google Scholar : PubMed/NCBI

34 

Szymura SJ, Zaemes JP, Allison DF, Clift SH, D'Innocenzi JM, Gray LG, McKenna BD, Morris BB, Bekiranov S, LeGallo RD, et al: NF-κB upregulates glutamine-fructose-6-phosphate transaminase 2 to promote migration in non-small cell lung cancer. Cell Commun Signal. 17:242019. View Article : Google Scholar : PubMed/NCBI

35 

Dowling P, Walsh N and Clynes M: Membrane and membrane-associated proteins involved in the aggressive phenotype displayed by highly invasive cancer cells. Proteomics. 8:4054–4065. 2008. View Article : Google Scholar : PubMed/NCBI

36 

Marshall S, Bacote V and Traxinger RR: Discovery of a metabolic pathway mediating glucose-induced desensitization of the glucose transport system. Role of hexosamine biosynthesis in the induction of insulin resistance. J Biol Chem. 266:4706–4712. 1991. View Article : Google Scholar : PubMed/NCBI

37 

Yang WH, Kim JE, Nam HW, Ju JW, Kim HS, Kim YS and Cho JW: Modification of p53 with O-linked N-acetylglucosamine regulates p53 activity and stability. Nat Cell Biol. 8:1074–1083. 2006. View Article : Google Scholar : PubMed/NCBI

38 

Itkonen HM, Minner S, Guldvik IJ, Sandmann MJ, Tsourlakis MC, Berge V, Svindland A, Schlomm T and Mills IG: O-GlcNAc transferase integrates metabolic pathways to regulate the stability of c-MYC in human prostate cancer cells. Cancer Res. 73:5277–5287. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Ghose P, Park EC, Tabakin A, Salazar-Vasquez N and Rongo C: Anoxia-reoxygenation regulates mitochondrial dynamics through the hypoxia response pathway, SKN-1/Nrf, and stomatin-like protein STL-1/SLP-2. PLOS Genet. 9:e10040632013. View Article : Google Scholar : PubMed/NCBI

40 

Karagiannis GS, Condeelis JS and Oktay MH: Chemotherapy-induced metastasis: Mechanisms and translational opportunities. Clin Exp Metastasis. 35:269–284. 2018. View Article : Google Scholar : PubMed/NCBI

41 

Shah AN, Summy JM, Zhang J, Park SI, Parikh NU and Gallick GE: Development and characterization of gemcitabine-resistant pancreatic tumor cells. Ann Surg Oncol. 14:3629–3637. 2007. View Article : Google Scholar : PubMed/NCBI

42 

Yang CT, Li JM, Li LF, Ko YS and Chen JT: Stomatin-like protein 2 regulates survivin expression in non-small cell lung cancer cells through β-catenin signaling pathway. Cell Death Dis. 9:4252018. View Article : Google Scholar : PubMed/NCBI

43 

Yallop CA and Svendsen I: The effects of G418 on the growth and metabolism of recombinant mammalian cell lines. Cytotechnology. 35:101–114. 2001. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2021
Volume 45 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
Chao D, Ariake K, Sato S, Ohtsuka H, Takadate T, Ishida M, Masuda K, Maeda S, Miura T, Mitachi K, Mitachi K, et al: Stomatin‑like protein 2 induces metastasis by regulating the expression of a rate‑limiting enzyme of the hexosamine biosynthetic pathway in pancreatic cancer. Oncol Rep 45: 90, 2021
APA
Chao, D., Ariake, K., Sato, S., Ohtsuka, H., Takadate, T., Ishida, M. ... Unno, M. (2021). Stomatin‑like protein 2 induces metastasis by regulating the expression of a rate‑limiting enzyme of the hexosamine biosynthetic pathway in pancreatic cancer. Oncology Reports, 45, 90. https://doi.org/10.3892/or.2021.8041
MLA
Chao, D., Ariake, K., Sato, S., Ohtsuka, H., Takadate, T., Ishida, M., Masuda, K., Maeda, S., Miura, T., Mitachi, K., Yu, X. J., Fujishima, F., Mizuma, M., Nakagawa, K., Morikawa, T., Kamei, T., Unno, M."Stomatin‑like protein 2 induces metastasis by regulating the expression of a rate‑limiting enzyme of the hexosamine biosynthetic pathway in pancreatic cancer". Oncology Reports 45.6 (2021): 90.
Chicago
Chao, D., Ariake, K., Sato, S., Ohtsuka, H., Takadate, T., Ishida, M., Masuda, K., Maeda, S., Miura, T., Mitachi, K., Yu, X. J., Fujishima, F., Mizuma, M., Nakagawa, K., Morikawa, T., Kamei, T., Unno, M."Stomatin‑like protein 2 induces metastasis by regulating the expression of a rate‑limiting enzyme of the hexosamine biosynthetic pathway in pancreatic cancer". Oncology Reports 45, no. 6 (2021): 90. https://doi.org/10.3892/or.2021.8041