Cancer stem cells possess self-renewal and chemoresistance activities. However, the manner in which these features are maintained remains obscure. We sought to identify cell surface protein(s) that mark self-renewing and chemoresistant gastric cancer cells using the explorer antibody microarray. We identified PMP22, a target gene of the Wnt/β-catenin pathway, as the most upregulated cell surface protein in gastric cancer xenografts exposed to cisplatin (DDP). PMP22 expression was markedly upregulated in tumorspheric cells and declined with differentiation. Infecting gastric cancer cells with lentivirus expressing PMP22 shRNAs reduced proliferation, tumorsphere formation, and chemoresistance to cisplatin in vitro and in NOD/SCID mice. When combined with bortezomib, a PMP22 inhibitor, the chemotherapeutic sensitivity to cisplatin treatment was dramatically increased by inducing cell apoptosis in cultured cells and xenograft mouse models. Finally, mRNA expression levels of PMP22 were detected in 38 tumor specimens from patients who received six cycles of perioperative chemotherapy. A strong correlation between PMP22 level and tumor recurrence was revealed, thus showing a pivotal role of PMP22 in the clinical chemoresistance of gastric cancer. Our study is the first to show the role of PMP22 in gastric cancer stemness and chemoresistance and reveals a potential new target for the diagnosis and treatment of recurrent gastric cancer. Mol Cancer Ther; 16(6); 1187–98. ©2017 AACR.

Gastric cancer is a major health burden worldwide. Its incidence ranks second among men and third among women, and the associated mortality still remains second for both males and females despite a downward trend in 2015 (1). The delivery of chemotherapeutic agents, including cisplatin (DDP) and fluoropyrimidine, following surgical resection is currently the standard treatment for advanced gastric cancer patients (2). Although multiagent chemotherapy can improve gastric cancer outcomes, almost all patients develop chemotherapy resistance, and the 5-year survival rate continues to be poor (3).

The cancer stem cell (CSC) hypothesis proposes that CSCs are a group of heterogeneous cells that exhibit self-renewal, multiple differentiation potential, high tumorigenicity, and drug resistance (4, 5). CSCs may have a central role in tumor initiation/maintenance, progression/metastasis, and recurrence (6–10). Conventional anticancer chemotherapies typically kill only differentiated tumor cells, whereas CSCs could be selected for recurrence. Therefore, the development of anticancer drugs that can target all CSC subsets within a tumor to control tumor growth and prevent recurrence is promising. Gastric cancer stem cells (GCSC) have received great attention, and some GCSC candidate surface markers were reported as potential novel therapeutic targets in the treatment of gastric cancer (11). Takaishi and colleagues found that CD44 can be used as a GCSC marker (12). Furthermore, ALDH1+, CD90+, CD71, CD133+, CD44+/CD24+, EpCAM+/CD44+, and CD44+/CD54+ were also identified as potential GCSC markers (13–18). Despite many laboratory efforts, there is no highly specific marker for both GCSCs and chemoresistant gastric cancer cells, and the mechanisms regulating the self-renewal and chemoresistance of gastric cancer cells are not fully understood.

PMP22 is an integral membrane glycoprotein of peripheral nervous system myelin (19). Mutations in the PMP22 gene are the most common causes of Charcot–Marie–Tooth disease (CMT), which is an inherited peripheral nerve disorder (20). The precise expression and function of PMP22 in tumors remain to be clarified. Several studies indicate that PMP22 is a potential tumor suppressor (21, 22), whereas some observations suggest a potential oncogenic function of PMP22 in tumors (23–28). Studies on the role of PMP22 in regulating gastric cancer have not been reported, and we describe here that PMP22 is required for both self-renewal and chemoresistance in gastric cancer.

Bortezomib, a proteasome inhibitor, was approved for the treatment of multiple myeloma by the FDA in 2003 (29, 30). A proposed mechanism for its action is that it prevents the proteasomal degradation of proapoptotic proteins, leading to enhanced apoptosis (31). Jang and colleagues found that bortezomib exhibited marked reduction of endogenous PMP22 mRNA and protein (32). We showed that bortezomib reduced PMP22 expression levels in gastric cancer and significantly suppressed tumor growth when combined with traditional chemotherapy.

Our study provides strong evidence suggesting that PMP22 may act as a therapeutic target mediating the self-renewal and chemoresistance functions in gastric cancer. We also provide a new anticancer regimen that inhibits PMP22 expression to enhance the anticancer effect.

Cell culture

The MGC-803, SGC-7901, and SGC-7901/DDP human gastric cancer cell lines were purchased from the Institute of Cell Biology (Shanghai, China; http://www.cellbank.org.cn). The 293T human embryonic kidney cell line was obtained from the ATCC. Short tandem repeat sequencing has been done for cell authentication. All cell lines used in this study have been validated within the last 6 months. MGC-803, SGC-7901, and SGC-7901/DDP cells were maintained in RPMI1640 medium, whereas 293T cells were maintained in DMEM. All media contained 10% FBS (Gibco) and 1% penicillin/streptomycin (Invitrogen), and cells were maintained at 37°C in a 5% CO2 humidified atmosphere. PCR tests for mycoplasma were negative. Cisplatin (Qilu Pharmaceutical Co., Ltd.) and bortezomib (Cell Signaling Technology, catalog no. 2204) were used for gastric cancer cell chemotherapy. PCR tests for mycoplasma were negative.

Lentivirus-mediated RNA interference

The pLKO.1 lentiviral vector was used to express short hairpin RNA directed against the PMP22 or LacZ control sequence (GTCTCCGAACGTGTCACGTT). Human PMP22 target sequences of PMP22 shRNA1 and PMP22 shRNA2 were CCAAACTCAAACCAAACCAAA and CGGTGTCATCTATGTGATCTT, respectively. The generation of lentiviral vectors was performed by cotransfecting pLKO.1 carrying the expression cassette with helper plasmids pVSV-G and pHR into 293T cells using Lipofectamine 3000 transfection reagent (Invitrogen Life Technologies). The viral supernatant was collected 48 hours after transfection, and cells at 60% to 70% confluence were infected with viral supernatants containing 10 mg/mL polybrene for 24 hours. Then, fresh medium with puromycin was used to select a pool of stably expressed cells.

RNA extraction and quantitative PCR analysis

Total RNA from cells or tissues was extracted using TRIzol reagent (Invitrogen) and reverse-transcribed with the SuperScript First-Strand Synthesis System for RT-PCR (Invitrogen) to produce cDNA according to the manufacturer's protocol. Platinum SYBR Green qPCR SuperMix (Invitrogen) was used for the qPCR reaction, and the expression levels were quantified using the −ΔΔCt method. GAPDH was used as the internal control. The following primer sequences for quantitative PCR were used:

PMP22: 5′-CTGGTCTGTGCGTGATGAGTG-3′ (forward),

5′-ATGTAGGCGAAACCGTAGGAG-3′ (reverse);

MYC: 5′-CTTCTCTCCGTCCTCGGATTCT-3′ (forward),

5′- GAAGGTGATCCAGACTCTGACCTT -3′ (reverse);

SOX2: 5′-GCCGAGTGGAAACTTTTGTCG-3′ (forward),

5′-GGCAGCGTGTACTTATCCTTCT-3′ (reverse);

DDR1: 5′-GCGTCTGTCTGCGGGTAGAG-3′ (forward),

5′-ACGGCCTCAGATAAATACATTGTCT-3′ (reverse);

GAPDH: 5′-TCTCCTCTGACTTCAACAGCGA-3′ (forward),

5′- GTCCACCACCCTGTTGCTGT -3′ (reverse).

Western blotting

Cells were lysed with NP-40 lysis buffer. Anti-PMP22 (Sigma-Aldrich, catalog no. P0081), anti-claudin-7 (Santa Cruz Biotechnology, catalog no. sc-33532), anti-HLA-DR (Santa Cruz Biotechnology, catalog no. sc-51617), anti-CD16 (Santa Cruz Biotechnology, catalog no. sc-20052), anti-PARP (Sigma-Aldrich, catalog no. P248), anti-CD24 (R&D Systems, catalog no. AF5247), anti-CD44 (Cell Signaling Technology, catalog no. 5640), anti-ALDH1 (R&D Systems, catalog no. AF5869), and anti-β-actin (Sigma-Aldrich, catalog no. A1978) were used for immunoblotting. β-Actin protein was used as the internal control.

Tumorsphere culture

The cells (500 cells per well) were cultured in ultra-low-attachment 12-well plates (Corning Life Sciences) in 1 mL of serum-free DMEM/F-12 (Invitrogen) supplemented with B-27 (1:50; Invitrogen), 20 ng/mL EGF (BD Biosciences), 20 ng/mL basic fibroblast growth factor (bFGF; BD Biosciences), and 4 mg/mL insulin (Sigma-Aldrich). Cells were fed every 3 days. The sphere number and size were measured on day 14.

For spheroid colony formation, virus-infected cells were inoculated in each well (three cells per well) of ultra-low-attachment 96-well plates (Corning Life Sciences) in sphere culture medium. After 2 weeks, each well was examined using a light microscope, and the total number of wells with spheres was counted. Images of the spheres were recorded using a Model 1×71 S8F-2 inverted microscope (Olympus America Inc.) and a MicroPublisher 5.0 RTV camera (QImaging). Relative sphere volumes were calculated at the same time points using the following equation: volume = length × (width)2 × π/6. Relative sphere length and width were assessed using Adobe Photoshop CS5 software under identical settings.

Colony formation assays

For the colony formation assay, stable transfected cells were plated in 6-well dishes at 400 cells per well. After 12 days, the cultures were washed twice with PBS, incubated with methanol for 20 minutes, stained with crystal violet for 30 minutes, and washed with tap water. The colonies were counted under a low-magnification microscope, and a group of more than 10 cells was defined as a colony.

For soft agar colony formation, agar (Nacalai Tesque) was dissolved in culture medium to 0.5% and plated in 6-well plates (bottom layer). Then, cells were seeded at 400 cells per well in 0.3% agar (top layer) over the bottom layer. Cells were covered with liquid growth media and fed every 3 days. The colony number was measured on day 14.

Cell analysis and sorting of PMP22+ and PMP22 populations

Confluent cells were washed once with PBS and then harvested with 0.05% trypsin/0.025% EDTA. Detached cells were washed with PBS containing 2% FBS (FACS buffer) and resuspended in the FACS buffer. The cell suspensions were incubated with isotype controls (BD Biosciences) or an mAb against human PMP22 (Sigma-Aldrich), followed by the secondary antibodies anti-mouse IgG/IgM conjugated with FITC (BD Biosciences). For cell analysis of the PMP22+ population, 106 cells were analyzed using a Beckman EPICS XL flow cytometer that assessed 10,000 events. For cell sorting of PMP22+ and PMP22 populations, cells were sorted with a Beckman MoFlo XDP cell sorter. The purity of the sorted cells was estimated to be greater than 95%. Cell apoptosis was evaluated by FACS using the Annexin V-FITC/PI Apoptosis Detection Kit (Sigma-Aldrich) following the manufacturer's instructions.

Cell viability assays

Cell growth rates and cytotoxicity were determined using MTS (Promega) following the manufacturer's instructions. The absorbance at 490 nm was measured with a Multiskan GO microplate spectrophotometer (Thermo Scientific).

Antibody microarray analysis

The antibody microarray (ASB600) was obtained from Full Moon BioSystems. Each glass slide contains 656 highly specific and well-characterized antibodies in duplicate. Proteins (whole-cell lysates) were extracted, biotinylated, hybridized to the microarray, and detected with fluorescent-labeled strepatavidin using the Antibody Microarray Detection Kit (Spring Bioscience) according to the manufacturer's protocol. A change of approximately 2-fold was used as the cut-off value to evaluate the differential expression of proteins between consecutive DDP-treated and control xenograft cells.

Gene expression microarray analysis

Microarray experiments to investigate the transcriptional profiles of the MGC-803 Ctrl shRNA and PMP22 shRNA2 cells were performed by Shanghai KangChen Biotech Company (http://www.kangchen.com.cn). Sample labeling and microarray (Agilent Human 4×44K Gene Expression Microarrays, >41,000 probes) hybridization were performed according to the Agilent One-Color Microarray-Based Gene Expression Analysis protocol (Agilent Technology). Corresponding GEO accession numbers were GSE94714.

Mouse xenograft model and chemotherapy

Experiments were conducted in male BALB/c athymic nude mice aged 5 weeks old. Animal care and handling procedures were performed in accordance with the Guide for the Care and Use of Laboratory Animals, and the animal study protocol was approved by the Institutional Animal Care and Use Committee of Xiamen University (reference no. XMULAC20150166). MGC-803 gastric cancer cells (3 × 106 cells for each mouse) were injected subcutaneously into the back fat pad of nude mice. After tumors reached approximately 100 mm3 in size, mice were randomized to treatment with PBS or different doses of cisplatin for 4 days of a 20-day period. For GCSC enrichment, consecutively passaged equal numbers (3 × 106) of MGC-803 xenograft cells from the 3 mg/kg DDP group and 0.9% NaCl group were treated in vivo with the same dose of DDP or NaCl for 4 days of a 32-day period as one passage.

For in vivo chemotherapy experiments, nude mice were subcutaneously inoculated with SGC-7901/DDP cells (2 × 106 cells per mouse). After 8 days, tumor-bearing mice were treated with 5 mg/kg DDP, 0.4 mg/kg bortezomib, or 2.5 mg/kg DDP combined with 0.25 mg/kg bortezomib for 3 days over a 15-day period (n = 6 mice per group). The tumor volumes were monitored and recorded every three days. The investigator who monitored the tumor volumes was blinded to group allocation. The tumor volumes were estimated using the following formula: 0.5 × length × width2.

Patients

The cohort of 38 eligible patients with resectable locally advanced adenocarcinoma of the stomach (AJCC, stage III) diagnosed from 2010 to 2013 was assigned to receive 6 cycles of perioperative epirubicin, cisplatin and 5-fluorouracil (ECF) chemotherapy. PMP22 expression levels were analyzed in the tumor samples obtained by surgery after preoperative chemotherapy. According to the follow-up survey, the patients were divided into two groups based on whether they experienced recurrence within 2 years. The tumor material was collected after informed consent was obtained, in agreement with the Institutional Review Board of Zhongshan Hospital of Xiamen University. The investigator who performed the examination of PMP22 expression was blinded to the sample information.

Statistical analysis

The data were analyzed using GraphPad Prism software. The data are presented as the mean ± SD, n ≥ 3. All data were analyzed using two-sided Student t tests, and P < 0.05 was considered statistically significant.

Chemotherapy selectively enriches for self-renewing gastric cancer cells

Resistance to chemotherapy distinguishes CSCs from other cancer cells (10). Therefore, GCSCs could be enriched by in vivo consecutive passage of gastric cancer cells in NOD/SCID mice treated with chemotherapy. First, we treated mice with xenograft MGC-803 cell tumors by tail vein injection of different doses of cisplatin (DDP). As shown in Fig. 1A, xenograft growth was effectively inhibited only when the concentration of DDP reached 3 mg/kg. Then, we chose this dosage (3 mg/kg) for the treatment of two subsequent passages of xenograft tumors. The relative tumor weight ratio in the 0.9% NaCl control group versus the DDP-treated group was 30% for the second-passage xenografts, whereas this relative tumor weight ratio reached 70% in the third-passage xenografts, which suggests the gain of a chemoresistance phenotype in the process of consecutive chemotherapy (Fig. 1B). Moreover, the mRNA levels of the self-renewal–related and drug resistance–related genes MYC, SOX2, and DDR1 were dramatically increased in the third-passage xenografts treated with consecutive chemotherapy (Fig. 1C).

Figure 1.

Gastric cancer cells exposed to chemotherapy are enriched for self-renewing cancer cells. A, A total of 3 × 106 MGC-803 cells were xenografted. After tumors reached approximately 100 mm3 in size, mice were randomized to treatment with PBS or different doses of cisplatin for 4 days of a 20-day period. The time course of the estimated mean tumor volume was quantified. B, Consecutively passaged equal numbers (3 × 106) of MGC-803 xenograft cells from the 3 mg/kg DDP and 0.9% NaCl groups were treated in vivo with the same dose of DDP or NaCl for 4 days of a 32-day period as one passage. Tumor appearance in each passage was photographed (left), and the tumor masses were weighed (right). C, Stem cell- and chemoresistance-related gene expression was compared between the 3 mg/kg DDP and 0.9% NaCl groups of the third-passage xenografts using quantitative PCR. D, Consecutive DDP-treated xenograft cells of the third passage formed more and larger tumorspheres than the normal saline group. The number (bottom, left) and size (bottom, right) of spheres were quantified. Representative photographs of tumorspheres were obtained on day 14 (top). Scale bar, 50 μm. E, Relative cell surface protein expression was compared between consecutive DDP-treated xenograft cells of the third passage and the normal saline group using Western blotting. The data are presented as the mean ± SD. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 1.

Gastric cancer cells exposed to chemotherapy are enriched for self-renewing cancer cells. A, A total of 3 × 106 MGC-803 cells were xenografted. After tumors reached approximately 100 mm3 in size, mice were randomized to treatment with PBS or different doses of cisplatin for 4 days of a 20-day period. The time course of the estimated mean tumor volume was quantified. B, Consecutively passaged equal numbers (3 × 106) of MGC-803 xenograft cells from the 3 mg/kg DDP and 0.9% NaCl groups were treated in vivo with the same dose of DDP or NaCl for 4 days of a 32-day period as one passage. Tumor appearance in each passage was photographed (left), and the tumor masses were weighed (right). C, Stem cell- and chemoresistance-related gene expression was compared between the 3 mg/kg DDP and 0.9% NaCl groups of the third-passage xenografts using quantitative PCR. D, Consecutive DDP-treated xenograft cells of the third passage formed more and larger tumorspheres than the normal saline group. The number (bottom, left) and size (bottom, right) of spheres were quantified. Representative photographs of tumorspheres were obtained on day 14 (top). Scale bar, 50 μm. E, Relative cell surface protein expression was compared between consecutive DDP-treated xenograft cells of the third passage and the normal saline group using Western blotting. The data are presented as the mean ± SD. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal

To examine whether chemotherapy may enrich for GCSCs, freshly isolated cells from the third-passage DDP-treated xenografts were cultured in suspension to generate tumor spheres. The number of spheres reflects the quantity of cells capable of in vitro self-renewal, whereas the number of cells per sphere indicates the self-renewal capacity of each sphere-generating cell (33, 34). As shown in Fig. 1D, freshly isolated cells from the third passage of DDP-treated xenografts generated an increased number of spheres that were larger than the controls. Finally, we analyzed the protein expression profiles of the third-passage DDP-treated xenografts versus the 0.9% NaCl-treated controls using the explorer antibody microarray. Notably, several cell surface proteins, including Claudin7, PMP22, HLA-DR, and CD16, were significantly altered (Supplementary Table S1), and Western blotting confirmed these results (Fig. 1E). Together, these results indicated that chemotherapy selectively enriches for self-renewing gastric cancer cells.

Cell surface protein PMP22 is upregulated in tumorspheric cells and marks self-renewing gastric cancer cells

Given that PMP22 is the most upregulated cell surface protein in DDP-selected xenografts, we determined whether PMP22 is also upregulated in tumorspheric cells. Higher levels of PMP22+ cells were detected in tumorspheric cells than in adherent cultures of MGC-803 cells according to FACS analysis (Fig. 2A). Consistently, PMP22 mRNA and protein levels were also increased in tumorspheric cells (Fig. 2B). Next, PMP22+ and PMP22 cells sorted from tumorspheric MGC-803 cells cultured in suspension were plated on collagen under differentiating conditions in serum. Only approximately 5.97% remained PMP22+ by day 5 in the PMP22+ fraction, whereas the PMP22 fraction remained >97% by day 5 (Fig. 2C). Therefore, PMP22+ cells not only have self-renewing capability, but, at the same time, PMP22+ cell levels were negatively correlated with GCSC differentiation process in vitro.

Figure 2.

PMP22 is upregulated in tumorspheric cells and marks self-renewing gastric cancer cells. A, FACS analysis of PMP22+ cell levels in tumorspheres or corresponding adherent MGC-803 cells (right). Representative photographs of spheres and adherent MGC-803 cells were obtained on day 14 (left). B, qPCR and Western blot analysis of PMP22 mRNA (top) and protein (bottom) levels in tumorspheres or corresponding adherent MGC-803 cells. C, FACS analysis of the differentiation ability of PMP22+ and PMP22 cells sorted from tumorspheric MGC-803 cells under adherent culture conditions. D, Western blot analysis of CD24, CD44, and ALDH1 protein levels in PMP22+ and PMP22 MGC-803 cells. E, The expression levels of stem cell-related and chemoresistance-related genes were compared between PMP22+ and PMP22 MGC-803 cells using qPCR. F, Analysis of soft agar colony formation ability in PMP22+ and PMP22 MGC-803 cells. The number of soft agar colonies was quantified (right). Representative photographs of soft agar colonies were obtained on day 14 (left). G, Analysis of sphere formation ability of PMP22+ and PMP22 MGC-803 cells. The number of spheres was quantified (right). Representative photographs of spheres were obtained on day 14 (left). Scale bar, 100 μm. H, Analysis of PMP22+ and PMP22 MGC-803 cell growth rate by the MTS assay. In B, E, F, G, and H, the data are expressed as the mean ± SD of three independent experiments. *, P < 0.05; **, P < 0.01.

Figure 2.

PMP22 is upregulated in tumorspheric cells and marks self-renewing gastric cancer cells. A, FACS analysis of PMP22+ cell levels in tumorspheres or corresponding adherent MGC-803 cells (right). Representative photographs of spheres and adherent MGC-803 cells were obtained on day 14 (left). B, qPCR and Western blot analysis of PMP22 mRNA (top) and protein (bottom) levels in tumorspheres or corresponding adherent MGC-803 cells. C, FACS analysis of the differentiation ability of PMP22+ and PMP22 cells sorted from tumorspheric MGC-803 cells under adherent culture conditions. D, Western blot analysis of CD24, CD44, and ALDH1 protein levels in PMP22+ and PMP22 MGC-803 cells. E, The expression levels of stem cell-related and chemoresistance-related genes were compared between PMP22+ and PMP22 MGC-803 cells using qPCR. F, Analysis of soft agar colony formation ability in PMP22+ and PMP22 MGC-803 cells. The number of soft agar colonies was quantified (right). Representative photographs of soft agar colonies were obtained on day 14 (left). G, Analysis of sphere formation ability of PMP22+ and PMP22 MGC-803 cells. The number of spheres was quantified (right). Representative photographs of spheres were obtained on day 14 (left). Scale bar, 100 μm. H, Analysis of PMP22+ and PMP22 MGC-803 cell growth rate by the MTS assay. In B, E, F, G, and H, the data are expressed as the mean ± SD of three independent experiments. *, P < 0.05; **, P < 0.01.

Close modal

Next, we detected other known GCSC markers as well as the mRNA levels of the self-renewal and drug resistance genes MYC, SOX2, and DDR1 in PMP22+ and PMP22 MGC-803 cells. As shown in Fig. 2D, proteins levels of GCSC marker ALDH1 in PMP22+ MGC-803 cells was more than 3-fold in PMP22 cells, but the CD24 and CD44 proteins levels had no obvious change. The qPCR results showed that the expression of MYC, SOX2, and DDR1 mRNA was significantly increased in PMP22+ cells compared with PMP22 cells (Fig. 2E). Moreover, PMP22+ cells formed more colonies and spheres than PMP22 cells in the soft agar colony formation (Fig. 2F) and sphere formation (Fig. 2G) assays. Using the MTS assay, we also observed an increased cell growth rate in PMP22+ cells compared with PMP22 cells (Fig. 2H); this phenotype may be due to a combination of factors such as PMP22 induced stem cell expansion and increased expression of some oncogenes in gastric cancer cells. Collectively, these results demonstrate that PMP22 is upregulated in self-renewing gastric cancer cells and that PMP22+ cells possess the features of self-renewal.

PMP22 is involved in gastric cancer initiation

To determine the role of PMP22 in gastric cancer cells, PMP22 was significantly knocked down in the MGC-803 cells by lentivirus-expressed shRNAs (Fig. 3A). First, we identified PMP22 downstream-regulated genes by screening a microarray system. Remarkably, by performing ANOVA of normalized microarray data (Supplementary Table S2), we identified a set of genes associated with drug resistance, stemness, and tumorigenesis whose expression levels in PMP22 knockdown MGC-803 cells significantly differed from those in the control cells (Fig. 3B). These results indicate that PMP22 may play functional roles in regulating the carcinogenesis, self-renewal, and chemoresistance of gastric cancer cells.

Figure 3.

PMP22 knockdown significantly reduces the self-renewal and tumorigenic ability of gastric cancer cells. A, qPCR analysis of PMP22 mRNA levels in MGC-803 cells stably expressing control shRNA and two different PMP22 shRNAs (n = 3). B, qPCR analysis showing different expression of drug resistance-, stemness-, and tumorigenesis-related genes in PMP22-knockdown MGC-803 cells. n = 3, P < 0.05. C, Knockdown of PMP22 expression in MGC-803 cells resulted in a reduced number of colonies in a plate colony formation assay (n = 4; right). Representative photographs of plate colonies were obtained on day 12 (left). D, Knockdown of PMP22 expression in MGC-803 cells resulted in a reduced number and size of colonies in a spheroid colony formation assay (n = 60; right). Representative photographs of spheroid colonies were obtained on day 14 (left). Scale bar, 100 μm. E, Knockdown of PMP22 expression in MGC-803 cells resulted in a reduced cell growth rate based on the MTS assay (n = 4). F, Knockdown of PMP22 expression in MGC-803 cells resulted in reduced subcutaneous tumorigenic ability in nude mice. The dissected xenograft tumors were photographed (top) and weighed (bottom) on day 28 (n = 5). The data are presented as the mean ± SD. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.

Figure 3.

PMP22 knockdown significantly reduces the self-renewal and tumorigenic ability of gastric cancer cells. A, qPCR analysis of PMP22 mRNA levels in MGC-803 cells stably expressing control shRNA and two different PMP22 shRNAs (n = 3). B, qPCR analysis showing different expression of drug resistance-, stemness-, and tumorigenesis-related genes in PMP22-knockdown MGC-803 cells. n = 3, P < 0.05. C, Knockdown of PMP22 expression in MGC-803 cells resulted in a reduced number of colonies in a plate colony formation assay (n = 4; right). Representative photographs of plate colonies were obtained on day 12 (left). D, Knockdown of PMP22 expression in MGC-803 cells resulted in a reduced number and size of colonies in a spheroid colony formation assay (n = 60; right). Representative photographs of spheroid colonies were obtained on day 14 (left). Scale bar, 100 μm. E, Knockdown of PMP22 expression in MGC-803 cells resulted in a reduced cell growth rate based on the MTS assay (n = 4). F, Knockdown of PMP22 expression in MGC-803 cells resulted in reduced subcutaneous tumorigenic ability in nude mice. The dissected xenograft tumors were photographed (top) and weighed (bottom) on day 28 (n = 5). The data are presented as the mean ± SD. *, P < 0.05; **, P < 0.01; ***, P < 0.001; ****, P < 0.0001.

Close modal

We also performed plate colony formation (Fig. 3C) and spheroid colony formation (Fig. 3D) assays, and the results showed that PMP22 knockdown cells generated significantly fewer and smaller colonies in both experiments. The MTS assay results also showed that PMP22 knockdown significantly inhibited the cell growth rate in MGC-803 cells (Fig. 3E). Next, we subcutaneously injected these cells into SCID mice. After 4 weeks, we observed that the PMP22 knockdown cells produced much smaller tumors than those derived from the control cells (Fig. 3F). These results imply that PMP22 is involved in tumor initiation of gastric cancer.

Reduced PMP22 inhibits chemoresistance of gastric cancer cells

Studies have suggested that CSC-enriched cells in several solid tumors exhibited greater resistance to anticancer chemotherapy drugs than the non-CSC population (35, 36). To investigate whether PMP22 is associated with the chemoresistance of gastric cancer cells, we performed cell survival assays using different doses of DDP. The PMP22+ fraction of MGC-803 cells exhibited increased resistance to the anticancer drug DDP compared with the PMP22 fraction (Fig. 4A). In addition, PMP22 knockdown in MGC-803 cells significantly inhibited chemoresistance to DDP (Fig. 4B). SGC-7901/DDP was an existing and approved DDP-resistant gastric cancer cell line, so we chose SGC-7901 as well as corresponding DDP-resistant cell lines SGC-7901/DDP to carry out related experiment. Increased levels of PMP22+ were also detected in SGC-7901/DDP cells compared with SGC-7901 cells (Fig. 4C). Consistently, qPCR results showed that PMP22 mRNA expression was significantly increased in SGC-7901/DDP cells compared with SGC-7901 cells. Next, we knocked down PMP22 expression in SGC-7901/DDP cells to assess chemotherapeutic sensitivity (Fig. 4D). As shown in Fig. 4E, SGC-7901/DDP cells naturally exhibited increased resistance to DDP compared with SGC-7901 cells; however, PMP22 knockdown significantly increased chemotherapeutic sensitivity to DDP. Finally, we assessed the effect of PMP22 on the sphere formation ability of DDP-resistant cells. As expected, SGC-7901/DDP cells formed significantly more and larger spheres than SGC-7901 cells, but PMP22 knockdown in SGC-7901/DDP cells significantly inhibited the sphere formation ability (Fig. 4F). Together, our findings show that reduced PMP22 could inhibit chemoresistance to DDP in gastric cancer cells and that this effect may be associated with self-renewal phenotypes.

Figure 4.

PMP22 expression is critical for cisplatin resistance of gastric cancer cells. A, Viability of PMP22+ and PMP22 MGC-803 cells after treatment with varying concentrations of cisplatin for 36 hours. B, Viability of PMP22-depleted MGC-803 cells after treatment with varying concentrations of cisplatin for 36 hours. C, FACS analysis of PMP22+ levels in SGC-7901 and SGC-7901/DDP cells. D, qPCR analysis of the PMP22 mRNA levels in SGC-7901, SGC-7901/DDP, and PMP22-depleted SGC-7901/DDP cells. E, Viability of SGC-7901, SGC-7901/DDP, and PMP22-depleted SGC-7901/DDP cells after treatment with varying concentrations of cisplatin for 36 hours. F, Analysis of sphere formation ability of SGC-7901, SGC-7901/DDP, and PMP22-depleted SGC-7901/DDP cells. The number (left) and size (right) of spheres were quantified on day 14. The data are expressed as the mean ± SD of four independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 4.

PMP22 expression is critical for cisplatin resistance of gastric cancer cells. A, Viability of PMP22+ and PMP22 MGC-803 cells after treatment with varying concentrations of cisplatin for 36 hours. B, Viability of PMP22-depleted MGC-803 cells after treatment with varying concentrations of cisplatin for 36 hours. C, FACS analysis of PMP22+ levels in SGC-7901 and SGC-7901/DDP cells. D, qPCR analysis of the PMP22 mRNA levels in SGC-7901, SGC-7901/DDP, and PMP22-depleted SGC-7901/DDP cells. E, Viability of SGC-7901, SGC-7901/DDP, and PMP22-depleted SGC-7901/DDP cells after treatment with varying concentrations of cisplatin for 36 hours. F, Analysis of sphere formation ability of SGC-7901, SGC-7901/DDP, and PMP22-depleted SGC-7901/DDP cells. The number (left) and size (right) of spheres were quantified on day 14. The data are expressed as the mean ± SD of four independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal

Gastric cancer chemotherapy combined with PMP22 targeting in vitro and in vivo

Standard chemotherapy approaches for gastric cancer are still associated with a high rate of recurrence and mortality, as resistance is rapidly acquired (37). Therefore, we hypothesized that standard chemotherapy combined with PMP22 targeting may yield better therapeutic effects than either used alone. Previous studies have reported that bortezomib was used as a redifferentiation agent for the treatment of multiple myeloma patients and significantly inhibited PMP22 expression levels (30, 32). However, bortezomib has not been used for the treatment of gastric cancer. Therefore, we used bortezomib as a PMP22 inhibitor in combination with DDP for chemoresistant gastric cancer therapy. The MTS results showed that the survival rate of SGC-7901/DDP cells was greater than 60% after treatment with 8 μg/mL DDP or 40 μmol/L bortezomib, separately, for 36 hours; however, treatment with 4 μg/mL DDP combined with 20 μmol/L bortezomib for 36 hours resulted in a cell survival rate of less than 25% (Fig. 5A). Moreover, this combined chemotherapy regimen dramatically induced the activation of PARP cleavage (Fig. 5A). Consistently, when we used flow cytometry to determine the percentage of apoptotic cells after exposure to different chemotherapies for 24 hours, the results indicated that cell apoptosis in the combined chemotherapy group was greatly increased compared with that resulting from either agent alone (Fig. 5B). Interestingly, the qPCR results revealed that PMP22 mRNA levels were significantly increased by DDP treatment and were significantly reduced by bortezomib treatment. However, there were no significant differences in PMP22 mRNA levels between the combined chemotherapy and PBS treatment groups (Fig. 5C). Collectively, the above in vitro results verify the hypothesis that standard cisplatin chemotherapy combined with PMP22 inhibition by bortezomib could increase chemotherapeutic sensitivity by inducing cell apoptosis.

Figure 5.

PMP22 inhibition using bortezomib increases cisplatin chemotherapeutic sensitivity of gastric cancer cells by inducing cell apoptosis. A, MTS analysis of the survival rate of SGC-7901/DDP cells treated with 8 μg/mL DDP, 40 μmol/L bortezomib, or 4 μg/mL DDP combined with 20 μmol/L bortezomib for 36 hours (left, top). Representative photographs of cells were obtained during the 36-hour treatment period (right). Scale bar, 100 μm. Representative immunoblot for PARP cleavage in SGC-7901/DDP cells treated as indicated for 36 hours (left, bottom). B, FACS analysis of cell apoptosis was performed with Annexin-V/PI staining in SGC-7901/DDP cells treated as indicated for 36 hours. The combined chemotherapy enhanced the early apoptosis (Annexin-V+PI) and late apoptosis (Annexin-V+PI+) of SGC-7901/DDP cells. C, qPCR analysis of the PMP22 mRNA levels in SGC-7901/DDP cells treated with 8 μg/mL DDP, 40 μmol/L bortezomib, or 4 μg/mL DDP combined with 20 μmol/L bortezomib for 36 hours. In A and C, the data are expressed as the mean ± SD of four independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Figure 5.

PMP22 inhibition using bortezomib increases cisplatin chemotherapeutic sensitivity of gastric cancer cells by inducing cell apoptosis. A, MTS analysis of the survival rate of SGC-7901/DDP cells treated with 8 μg/mL DDP, 40 μmol/L bortezomib, or 4 μg/mL DDP combined with 20 μmol/L bortezomib for 36 hours (left, top). Representative photographs of cells were obtained during the 36-hour treatment period (right). Scale bar, 100 μm. Representative immunoblot for PARP cleavage in SGC-7901/DDP cells treated as indicated for 36 hours (left, bottom). B, FACS analysis of cell apoptosis was performed with Annexin-V/PI staining in SGC-7901/DDP cells treated as indicated for 36 hours. The combined chemotherapy enhanced the early apoptosis (Annexin-V+PI) and late apoptosis (Annexin-V+PI+) of SGC-7901/DDP cells. C, qPCR analysis of the PMP22 mRNA levels in SGC-7901/DDP cells treated with 8 μg/mL DDP, 40 μmol/L bortezomib, or 4 μg/mL DDP combined with 20 μmol/L bortezomib for 36 hours. In A and C, the data are expressed as the mean ± SD of four independent experiments. *, P < 0.05; **, P < 0.01; ***, P < 0.001.

Close modal

The treatment effects of PMP22 inhibition and DDP chemotherapy were next examined in vivo. SGC-7901/DDP cells were subcutaneously injected into the back of mice to form xenografts. After tumors reached 50 to 100 mm3 in size, mice were treated with PBS or different chemotherapies 3 days for 15 days. Control tumors treated with PBS grew to over 800 mm3 by 15 days following treatment, whereas the tumors treated separately with 5 mg/kg DDP or 0.4 mg/kg bortezomib grew to over 500 mm3. However, the combination of 2.5 mg/kg DDP and 0.25 mg/kg bortezomib dramatically reduced tumor growth to 200 mm3 (Fig. 6A and B). Similar results were achieved when we measured tumor weight (Fig. 6C). Notably, the relative body weight of mice pre- and postchemotherapy (d8/d23) was significantly increased with the combined chemotherapy compared with 5 mg/kg DDP alone (Fig. 6D). FACS and qPCR were performed to determine the PMP22+ cell levels and PMP22 mRNA levels in gastric cancer xenograft cells after treatment with different chemotherapies. PMP22+ cell levels and PMP22 mRNA levels were significantly increased after 5 mg/kg DDP chemotherapy for 15 days, whereas 0.4 mg/kg bortezomib treatment significantly inhibited PMP22 expression. As expected, PMP22+ cell levels and PMP22 mRNA levels after the combined chemotherapy were similar to those in the PBS treatment group (Fig. 6E and F).

Figure 6.

Effects of standard cisplatin chemotherapy combined with bortezomib on gastric primary tumor models and the clinical significance of PMP22 levels in patients with gastric cancer. A–E, Nude mice were subcutaneously inoculated with SGC-7901/DDP cells (2 × 106 cells per mouse). After 8 days, tumor-bearing mice were treated with 5 mg/kg DDP, 0.4 mg/kg bortezomib, or 2.5 mg/kg DDP combined with 0.25 mg/kg bortezomib for 3 days of a 15-day period (n = 6). *, P < 0.05; **, P < 0.01; ***, P < 0.001. A, Photographs of the dissected xenograft tumors from different groups of nude mice treated as indicated. B, Growth curve of tumors from different groups of nude mice treated as indicated. C, Diagram of the average weight of the dissected xenograft tumors from different groups of nude mice treated as indicated. D, Diagram of the relative body weight of mice pre- and postchemotherapy (d8/d23). E, FACS analysis of PMP22+ levels in the dissected xenograft tumor cells from different groups of nude mice treated as indicated. F, qPCR analysis of the PMP22 mRNA levels in the dissected xenograft tumors from different groups of nude mice treated as indicated. G, qPCR analysis of the PMP22 mRNA levels in the tumor samples obtained by surgery after 6 cycles of perioperative ECF chemotherapy. These patients were divided into two groups based on whether the cancer recurred within 2 years (log2 median-centered ratio, P = 0.0008, unpaired t test).

Figure 6.

Effects of standard cisplatin chemotherapy combined with bortezomib on gastric primary tumor models and the clinical significance of PMP22 levels in patients with gastric cancer. A–E, Nude mice were subcutaneously inoculated with SGC-7901/DDP cells (2 × 106 cells per mouse). After 8 days, tumor-bearing mice were treated with 5 mg/kg DDP, 0.4 mg/kg bortezomib, or 2.5 mg/kg DDP combined with 0.25 mg/kg bortezomib for 3 days of a 15-day period (n = 6). *, P < 0.05; **, P < 0.01; ***, P < 0.001. A, Photographs of the dissected xenograft tumors from different groups of nude mice treated as indicated. B, Growth curve of tumors from different groups of nude mice treated as indicated. C, Diagram of the average weight of the dissected xenograft tumors from different groups of nude mice treated as indicated. D, Diagram of the relative body weight of mice pre- and postchemotherapy (d8/d23). E, FACS analysis of PMP22+ levels in the dissected xenograft tumor cells from different groups of nude mice treated as indicated. F, qPCR analysis of the PMP22 mRNA levels in the dissected xenograft tumors from different groups of nude mice treated as indicated. G, qPCR analysis of the PMP22 mRNA levels in the tumor samples obtained by surgery after 6 cycles of perioperative ECF chemotherapy. These patients were divided into two groups based on whether the cancer recurred within 2 years (log2 median-centered ratio, P = 0.0008, unpaired t test).

Close modal

To investigate the association of PMP22 with clinical chemoresistance in gastric cancer patients, we selected 38 gastric cancer specimens from patients who received 6 cycles of perioperative chemotherapy to assess PMP22 mRNA levels by qPCR. The patients were divided into two groups according to clinical outcome as follows: group 1, patients with recurrence; group 2, patients without recurrence 2 years after surgery. The results showed that the PMP22 levels were significantly increased in gastric cancer tissues from patients with recurrence compared with the group without recurrence (Fig. 6G). Together, these results strongly indicate that PMP22 activation is involved in chemoresistance in gastric cancer patients and that standard cisplatin chemotherapy combined with PMP22 inhibition using bortezomib may yield better therapeutic effects.

Chemotherapy-induced drug resistance remains a major obstacle to the clinical management of gastric cancer, resulting in relapse and metastasis. Thus, novel strategies to overcome drug resistance are currently under intense investigation. GCSCs are a major factor in gastric cancer resistance to radiation and chemotherapy (11). CSCs isolated from gastric cancer cell lines using the SP method exhibit drug tolerance for chemotherapy (12, 13). However, this is not a direct method for the simultaneous enrichment of GCSCs and chemoresistant gastric cancer cells. As resistance to chemotherapy distinguishes CSCs from other cancer cells (10), the in vivo consecutive passage of cancer cells in NOD/SCID mice treated with chemotherapy is an effective method for the enrichment of CSCs, as demonstrated in previous studies and our study (38). A long period of chemotherapy also simulates the induction of drug resistance in tumors in patients undergoing clinical treatment. Therefore, this method is suitable for the identification of diagnostic or therapeutic targets for both CSCs and chemoresistance.

In a search for cell surface protein(s) that are markers of self-renewing and chemoresistant gastric cancer cells using the Explorer Antibody Microarray, we identified PMP22, a target gene of the Wnt/β-catenin pathway (39, 40), as the most upregulated cell surface protein in the xenografts of the consecutive chemotherapy group. The biological functions of PMP22 proteins in mammalian cells are complex. PMP22 comprises an estimated 2% to 5% of the total myelin proteins in the peripheral nervous system (41). Its functional importance is highlighted by the fact that mutations in the PMP22 gene are the most common causes of inherited peripheral nerve disorders (20). In addition to its expression in neural cells, PMP22 is also observed in non-neural cells and tissues during development, such as epithelial cells of the lung, intestines, and stomach (42–44). The precise expression and function of PMP22 in tumors remain to be clarified. Several studies indicate a potential tumor suppressor function of PMP22. For example, PMP22 depletion results in increased proliferation in breast cancer cells (21). High expression of PMP22 results in growth arrest and apoptosis in fibroblasts (22). In contrast, some observations suggest a potential oncogenic function of PMP22 in tumors. PMP22 participates in the oncogenesis, progression, and metastasis of osteosarcoma (23–26). Furthermore, high-level PMP22 amplification was observed in high-grade glioma (27, 28). In the current study, we revealed for the first time the role of PMP22 in gastric cancer in potentially mediating self-renewal and chemoresistance in gastric cancer cells. This role is strongly supported by the following findings: (i) PMP22 expression levels and PMP22+ cell levels were upregulated in both tumor spheres and a DDP-resistant gastric cancer cell line; (ii) PMP22+ gastric cancer cells exhibited enhanced self-renewing ability and drug tolerance for chemotherapy compared with PMP22 cells; (iii) knockdown of PMP22-inhibited self-renewal and chemoresistance in gastric cancer cells was found in vitro and in vivo; and (iv) the expression pattern and functional relevance of PMP22 in tumor model and cancer samples were identified. Because CSCs possess chemoresistant features, we hypothesized that chemoresistant cancer cells acquire more stem cells. Indeed, we observed an expanded stem cell population in SGC-7901/DDP cells compared with SGC-7901 cells and revealed a reciprocal link between stem cell properties and chemoresistance in cancer cells. Moreover, the inhibition of PMP22 expression further reduced the GCSC population in the resistant gastric cancer cells. Above all, these results indicated that PMP22 is more than just a marker of the self-renewing capacity and chemoresistance of gastric cancer cells. PMP22 functionally contributes to the self-renewal, tumorigenicity, and chemoresistance of gastric cancer cells. We also predict that when combined with other GCSC surface markers, such as CD44, PMP22 can precisely recognize the GCSC population.

Conventional chemotherapy only kills differentiated tumor cells, resulting in tumor size reduction. However, the tumors relapse after some time, possibly due to the presence of residual and selected CSCs (11). Recent advanced strategies have been suggested for targeting GCSCs, including a combination of chemotherapy-associated apoptosis, tumor stem cell differentiation induction, therapies targeting GCSC surface molecules, and inhibition of GCSC-related pathways (11–13, 45, 46). However, these strategies are difficult to implement directly in clinical treatment. In our study, which aimed to investigate an existing clinical drug to target PMP22 in gastric cancer cells, we identified bortezomib, which has been used for the clinical treatment of multiple myeloma (30, 31). More importantly, bortezomib markedly reduces endogenous PMP22 mRNA and protein (32). Indeed, we observed effective inhibition of PMP22 by bortezomib in chemoresistant gastric cancer cells. When combined with DDP, bortezomib treatment dramatically increased chemotherapeutic sensitivity by inducing cell apoptosis.

In summary, we have shown that cisplatin chemotherapy selectively enriches self-renewing gastric cancer cells and that standard cisplatin chemotherapy combined with bortezomib enhances the effects of chemotherapy. We identified that PMP22 not only acts as a marker for gastric CSCs but may also have an essential role in regulating the self-renewal and chemoresistance of gastric cancer. Our findings suggest that PMP22 has clinical value for the prognosis and treatment of chemoresistant gastric cancer.

No potential conflicts of interest were disclosed.

Conception and design: W. Cai, Q. Luo, B. Li, J. Cai

Development of methodology: W. Cai, G. Chen, Q. Luo, J. Liu, X. Guo

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): G. Chen, T. Zhang, F. Ma, L. Yuan

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): W. Cai, G. Chen, J. Liu, F. Ma

Writing, review, and/or revision of the manuscript: W. Cai, B. Li, J. Cai

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): W. Cai, G. Chen

Study supervision: B. Li

This work was supported by grants from the National Natural Science Foundation of China: grant numbers 81372616 (to J. Cai), 81272384 (to B. Li), and 81602148 (to W. Cai); and the Natural Science Foundation of Fujian Province: grant number 2014D018 (to J. Cai).

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Chen
W
,
Zheng
R
,
Baade
PD
,
Zhang
S
,
Zeng
H
,
Bray
F
, et al
Cancer statistics in China, 2015
.
CA Cancer J Clin
2016
;
66
:
115
32
.
2.
Szakacs
G
,
Paterson
JK
,
Ludwig
JA
,
Booth-Genthe
C
,
Gottesman
MM
. 
Targeting multidrug resistance in cancer
.
Nat Rev Drug Discov
2006
;
5
:
219
34
.
3.
Pasechnikov
V
,
Chukov
S
,
Fedorov
E
,
Kikuste
I
,
Leja
M
. 
Gastric cancer: prevention, screening and early diagnosis
.
World J Gastroenterol
2014
;
20
:
13842
62
.
4.
Hambardzumyan
D
,
Becher
OJ
,
Holland
EC
. 
Cancer stem cells and survival pathways
.
Cell Cycle
2008
;
7
:
1371
8
.
5.
Visvader
JE
,
Lindeman
GJ
. 
Cancer stem cells: current status and evolving complexities
.
Cell Stem Cell
2012
;
10
:
717
28
.
6.
Clevers
H
. 
The cancer stem cell: premises, promises and challenges
.
Nat Med
2011
;
17
:
313
9
.
7.
Dick
JE
. 
Stem cell concepts renew cancer research
.
Blood
2008
;
112
:
4793
807
.
8.
Dalerba
P
,
Clarke
MF
. 
Cancer stem cells and tumor metastasis: first steps into uncharted territory
.
Cell Stem Cell
2007
;
1
:
241
2
.
9.
Wicha
MS
. 
Cancer stem cells and metastasis: lethal seeds - commentary
.
Clin Cancer Res
2006
;
12
:
5606
7
.
10.
Al-Hajj
M
. 
Cancer stem cells and oncology therapeutics
.
Curr Opinion Oncol
2007
;
19
:
61
4
.
11.
Singh
SR
. 
Gastric cancer stem cells: a novel therapeutic target
.
Cancer Lett
2013
;
338
:
110
9
.
12.
Takaishi
S
,
Okumura
T
,
Tu
S
,
Wang
SS
,
Shibata
W
,
Vigneshwaran
R
, et al
Identification of gastric cancer stem cells using the cell surface marker CD44
.
Stem Cells
2009
;
27
:
1006
20
.
13.
Jiang
J
,
Zhang
Y
,
Chuai
S
,
Wang
Z
,
Zheng
D
,
Xu
F
, et al
Trastuzumab (herceptin) targets gastric cancer stem cells characterized by CD90 phenotype
.
Oncogene
2012
;
31
:
671
82
.
14.
Ohkuma
M
,
Haraguchi
N
,
Ishii
H
,
Mimori
K
,
Tanaka
F
,
Kim
HM
, et al
Absence of CD71 transferrin receptor characterizes human gastric adenosquamous carcinoma stem cells
.
Ann Surg Oncol
2012
;
19
:
1357
64
.
15.
Jiang
Y
,
He
Y
,
Li
H
,
Li
HN
,
Zhang
L
,
Hu
W
, et al
Expressions of putative cancer stem cell markers ABCB1, ABCG2, and CD133 are correlated with the degree of differentiation of gastric cancer
.
Gastric Cancer
2012
;
15
:
440
50
.
16.
Zhang
C
,
Li
C
,
He
F
,
Cai
Y
,
Yang
H
. 
Identification of CD44+CD24+ gastric cancer stem cells
.
J Cancer Res Clin Oncol
2011
;
137
:
1679
86
.
17.
Han
ME
,
Jeon
TY
,
Hwang
SH
,
Lee
YS
,
Kim
HJ
,
Shim
HE
, et al
Cancer spheres from gastric cancer patients provide an ideal model system for cancer stem cell research
.
Cell Mol Life Sci
2011
;
68
:
3589
605
.
18.
Chen
T
,
Yang
K
,
Yu
J
,
Meng
W
,
Yuan
D
,
Bi
F
, et al
Identification and expansion of cancer stem cells in tumor tissues and peripheral blood derived from gastric adenocarcinoma patients
.
Cell Res
2012
;
22
:
248
58
.
19.
Snipes
GJ
,
Suter
U
,
Welcher
AA
,
Shooter
EM
. 
Characterization of a novel peripheral nervous system myelin protein (PMP-22/SR13)
.
J Cell Biol
1992
;
117
:
225
38
.
20.
Li
J
,
Parker
B
,
Martyn
C
,
Natarajan
C
,
Guo
J
. 
The PMP22 gene and its related diseases
.
Mol Neurobiol
2013
;
47
:
673
98
.
21.
Winslow
S
,
Leandersson
K
,
Larsson
C
. 
Regulation of PMP22 mRNA by G3BP1 affects cell proliferation in breast cancer cells
.
Mol Cancer
2013
;
12
:
156
.
22.
Karlsson
C
,
Afrakhte
M
,
Westermark
B
,
Paulsson
Y
. 
Elevated level of gas3 gene expression is correlated with G0 growth arrest in human fibroblasts
.
Cell Biol Intl
1999
;
23
:
351
8
.
23.
Liu
S
,
Chen
Z
. 
The functional role of PMP22 gene in the proliferation and invasion of osteosarcoma
.
Med Sci Monitor
2015
;
21
:
1976
82
.
24.
van Dartel
M
,
Hulsebos
TJ
. 
Characterization of PMP22 expression in osteosarcoma
.
Cancer Genet Cytogenet
2004
;
152
:
113
8
.
25.
van Dartel
M
,
Cornelissen
PWA
,
Redeker
S
,
Tarkkanen
M
,
Knuutila
S
,
Hogendoorn
PCW
, et al
Amplification of 17p11.2 similar to p12, including PMP22, TOP3A, and MAPK7, in high-grade osteosarcoma
.
Cancer Genet Cytogenet
2002
;
139
:
91
6
.
26.
van Dartel
M
,
Redeker
S
,
Bras
J
,
Kool
M
,
Hulsebos
TJM
. 
Overexpression through amplification of genes in chromosome region 17p11.2 similar to p12 in high-grade osteosarcoma
.
Cancer Genet Cytogenet
2004
;
152
:
8
14
.
27.
Huhne
K
,
Park
O
,
Liehr
T
,
Rautenstrauss
B
. 
Expression analysis of the PMP22 gene in glioma and osteogenic sarcoma cell lines
.
J Neurosci Res
1999
;
58
:
624
31
.
28.
van Dartel
M
,
Leenstra
S
,
Troost
D
,
Hulsebos
TJM
. 
Infrequent but high-level amplification of 17p11.2 similar to p12 in human glioma
.
Cancer Genet Cytogenet
2003
;
140
:
162
6
.
29.
Wang
HJ
,
Yao
JM
,
Zhang
ZW
,
Zhao
JY
,
Shang
HX
,
Liao
L
, et al
Expression of Pax8 is decreased and bortezomib does not increase the iodine uptake in thyroid carcinoma cells
.
Thoracic Cancer
2015
;
6
:
792
6
.
30.
Richardson
PG
,
Mitsiades
C
,
Hideshima
T
,
Anderson
KC
. 
Bortezomib: proteasome inhibition as an effective anticancer therapy
.
Annu Rev Med
2006
;
57
:
33
47
.
31.
Abd-Aziz
N
,
Stanbridge
EJ
,
Shafee
N
. 
Bortezomib attenuates HIF-1-but not HIF-2-mediated transcriptional activation
.
Oncol Lett
2015
;
10
:
2192
6
.
32.
Jang
SW
,
Lopez-Anido
C
,
MacArthur
R
,
Svaren
J
,
Inglese
J
. 
Identification of drug modulators targeting gene-dosage disease CMT1A
.
ACS Chem Biol
2012
;
7
:
1205
13
.
33.
Dontu
G
,
Abdallah
WM
,
Foley
JM
,
Jackson
KW
,
Clarke
MF
,
Kawamura
MJ
, et al
In vitro propagation and transcriptional profiling of human mammary stem/progenitor cells
.
Gene Dev
2003
;
17
:
1253
70
.
34.
Dontu
G
,
Jackson
KW
,
McNicholas
E
,
Kawamura
MJ
,
Abdallah
WM
,
Wicha
MS
. 
Role of Notch signaling in cell-fate determination of human mammary stem/progenitor cells
.
Breast Cancer Res
2004
;
6
:
R605
15
.
35.
Lobo
NA
,
Shimono
Y
,
Qian
D
,
Clarke
MF
. 
The biology of cancer stem cells
.
Annu Rev Cell Dev Biol
2007
;
23
:
675
99
.
36.
Vermeulen
L
,
Sprick
MR
,
Kemper
K
,
Stassi
G
,
Medema
JP
. 
Cancer stem cells—old concepts, new insights
.
Cell Death Differentiation
2008
;
15
:
947
58
.
37.
Andrews
PA
,
Jones
JA
,
Varki
NM
,
Howell
SB
. 
Rapid emergence of acquired cis-diamminedichloroplatinum(II) resistance in an in vivo model of human ovarian carcinoma
.
Cancer Commun
1990
;
2
:
93
100
.
38.
Yu
F
,
Yao
H
,
Zhu
P
,
Zhang
X
,
Pan
Q
,
Gong
C
, et al
let-7 regulates self renewal and tumorigenicity of breast cancer cells
.
Cell
2007
;
131
:
1109
23
.
39.
Tawk
M
,
Makoukji
J
,
Belle
M
,
Fonte
C
,
Trousson
A
,
Hawkins
T
, et al
Wnt/beta-catenin signaling is an essential and direct driver of myelin gene expression and myelinogenesis
.
J Neurosci
2011
;
31
:
3729
42
.
40.
Makoukji
J
,
Shackleford
G
,
Meffre
D
,
Grenier
J
,
Liere
P
,
Lobaccaro
JM
, et al
Interplay between LXR and Wnt/beta-catenin signaling in the negative regulation of peripheral myelin genes by oxysterols
.
J Neurosci
2011
;
31
:
9620
9
.
41.
Watila
MM
,
Balarabe
SA
. 
Molecular and clinical features of inherited neuropathies due to PMP22 duplication
.
J Neurol Sci
2015
;
355
:
18
24
.
42.
Taylor
ST
,
Menzel
R
. 
The creation of a camptothecin-sensitive Escherichia coli based on the expression of the human topoisomerase I
.
Gene
1995
;
167
:
69
74
.
43.
Baechner
D
,
Liehr
T
,
Hameister
H
,
Altenberger
H
,
Grehl
H
,
Suter
U
, et al
Widespread expression of the peripheral myelin protein-22 gene (PMP22) in neural and non-neural tissues during murine development
.
J Neurosci Res
1995
;
42
:
733
41
.
44.
Wulf
P
,
Bernhardt
RR
,
Suter
U
. 
Characterization of peripheral myelin protein 22 in zebrafish (zPMP22) suggests an early role in the development of the peripheral nervous system
.
J Neurosci Res
1999
;
57
:
467
78
.
45.
Li
K
,
Dan
Z
,
Nie
YQ
. 
Gastric cancer stem cells in gastric carcinogenesis, progression, prevention and treatment
.
World J Gastroenterol
2014
;
20
:
5420
6
.
46.
Mao
J
,
Fan
S
,
Ma
W
,
Fan
P
,
Wang
B
,
Zhang
J
, et al
Roles of Wnt/beta-catenin signaling in the gastric cancer stem cells proliferation and salinomycin treatment
.
Cell Death Dis
2014
;
5
:
e1039
.