Silencing NOB1 enhances doxorubicin antitumor activity of the papillary thyroid carcinoma in vitro and in vivo

Retraction in: /10.3892/or.2022.8297

  • Authors:
    • Jia Liu
    • Bing-Fei Dong
    • Pei-Song Wang
    • Pei-You Ren
    • Shuai Xue
    • Xiao-Νan Zhang
    • Zhe Han
    • Guang Chen
  • View Affiliations

  • Published online on: January 15, 2015     https://doi.org/10.3892/or.2015.3730
  • Pages: 1551-1559
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Doxorubicin (DOX), a broad‑spectrum anthra­cyclin, is in wide clinical use for the treatment and prevention of thyroid cancer. However, the effectiveness of the treatment remains limited due to inherent tumor resistance to DOX. Results of a previous study demonstrated that downregulation of NIN1/RPN12 binding protein 1 homolog (NOB1) expression via adenovirus expression vector carrying NOB1 siRNA (Ad/sh-NOB1) induced cancer apoptosis and increased the radiosensitivity of papillary thyroid carcinoma (PTC) cells. However, whether knockout NOB1 can decrease DOX resistance remains unclear. Therefore, in the present study, the effect of Ad/sh-NOB1 infection, independently or in combination with DOX, was determined in a PTC cell line to identify more effective therapeutics against PTC cancer. Furthermore, tumor growth ability in nude mice was determined to identify the combination treatment effect in tumorigenesis in vivo. The results showed that Ad/sh-NOB1 combined with DOX treatment in PTC cells significantly suppressed proliferation, colony formation, migration and invasion, and induced cell apoptosis and arrest in the G0/G1 stage as compared to Ad/sh-NOB1 or DOX monotherapy. We also found that this combination suppressed the tumor growth of a nude mouse model as compared to Ad/sh-NOB1 or DOX monotherapy. In addition, Ad/sh-NOB1 combined with DOX treatment significantly increased activation of the p38 MAPK pathway, which may contribute to inhibition of PTC cell growth and decreased DOX resistance. Taken together, the experimental results indicate that Ad/sh-NOB1 combined with DOX treatment is a potential drug candidate for the treatment of papillary thyroid carcinoma.

Introduction

Thyroid cancer is the most common malignancy of the endocrine system and accounts for ~1% of all malignant tumors (1). In 2013, an estimated 60,220 Americans may be diagnosed with thyroid cancer, and the disease is likely to account for ~1,850 mortalities (2). Papillary thyroid cancer (PTC) is the most common subtype of this disease, accounting for >83% of all such malignancies (3,4). Although the majority of PTC is effectively managed by surgical resection followed by radioactive iodine therapy, a subset of PTC is aggressive and refractory to current therapeutic approaches. Doxorubicin (DOX), a broad-spectrum anthracyclin, is the only FDA-approved chemotherapeutic agent for patients with non-radioiodine avid thyroid cancer (5). However, the clinical impact of DOX on thyroid cancer remains poor due to a high degree of chemoresistance (5,6). In particular, in heart tissues, DOX causes dilated cardiomyopathy, congestive heart failure and even cardiac sudden death (7,8), which limits its clinical applications. Therefore, improved therapeutic regimens that potentiate DOX effects, allowing dose reduction are required to improve the treatment of patients with the aggressive subtype of papillary thyroid cancer.

NIN1/RPN12 binding protein 1 homolog (NOB1) encodes a chaperone protein that joins the 20S proteasome with the 19S regulatory particle in the nucleus and is required for the biogenesis and function of the 26S proteasome, which plays a role in maintaining cellular homeostasis by controlling protein degradation. The human ortholog of the NOB1 gene was previously cloned (9). The NOB1 gene, located on chromosome 16q22.1, encoding a 50-kDa protein consisting of a PilT amino terminus (PIN) domain and a zinc ribbon domain, is mainly expressed in the liver, lung and spleen (9,10). NOB1 has been found to be upregulated in various types of cancer, including ovarian cancer, prostate and breast-infiltrating ductal carcinoma and non-small cell lung cancer (1114). In addition, the downregulation of endogenous NOB1 inhibited tumor cell proliferation and colony formation, and induced cell apoptosis and arrest in the G0/G1 stage in various types of cancer (11,1416). For PTC, results of a previous study showed that the expression level of NOB1 protein was increased in PTC tissue compared to normal thyroid tissue and benign thyroid tumor tissue and its expression was associated with UICC stage, tumor size and lymph node metastasis (17). Findings of a recent study demonstrated that adenovirus-mediated siRNA targeting NOB1 (Ad/sh-NOB1) in human PTC cells inhibited proliferation, migration and invasion in vitro, and suppressed tumor growth in a mouse model, and enhanced the in vitro and in vivo radiosensitivity of PTC cells (18). However, to the best of our knowledge, no previous studies have elucidated the role of NOB1 in thyroid cancer chemoresistance.

The aim of the present study was to evaluate the potency of Ad/sh-NOB1 in combination with DOX in inhibiting PTC cell growth, proliferation and migration and invasion, and inducing cell apoptosis and cell cycle arrest. In addition, tumor growth ability in nude mice was detected to determine the combination treatment effect on tumorigenesis in a nude mouse model.

Materials and methods

Reagents and antibody

Adenovirus expression vector carrying NOB1 siRNA (Ad/sh-NOB1) and adenovirus expression vector carrying scramble siRNA (Ad/sh-Scramble) were constructed and stored, as previously described (18). Doxorubicin (DOX) was obtained from Pfizer, Inc. (New York, NY, USA). Nonidet P-40 lysis buffer, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) was purchased from Sigma-Aldrich, St. Louis, MO, USA. Stock solutions of propidium iodide (PI) and MTT were prepared by dissolving 1 mg of each compound in 1 ml of phosphate-buffered saline (PBS). The solution was protected from light, stored at 4°C and used within one month.

For the western blot analysis, the antibodies used were: mouse monoclonal anti-human NOB1 (Abcam, Cambridge, UK), mouse monoclonal anti-human phosphorylated(p-) p38MAPK and mouse monoclonal anti-human cyclin D1 (Cell Signaling Technology, Danvers, MA, USA), mouse monoclonal anti-human p38MAPK, mouse monoclonal anti-human p-ERK1/2, mouse monoclonal anti-human ERK1/2, mouse monoclonal anti-human JNK, mouse monoclonal anti-human p-JNK, mouse monoclonal anti-human P21, mouse monoclonal anti-human cyclin D3 and mouse monoclonal anti-human GAPDH (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA). Secondary antibodies HRP-conjugated goat anti-mouse IgG was purchased from Amersham Biosciences (Uppsala, Sweden).

Cell culture

The TPC-1 human PTC cell line, provided by the Chinese Cell Bank of the Chinese Academy of Sciences (Shanghai, China), was cultured in RPMI-1640 containing 10% fetal bovine serum (FBS) (both from Gibco, Carlsbad, CA, USA), 100 U/ml penicillin and 100 mg/ml streptomycin (both from Sigma-Aldrich) at 37°C in a humidified atmosphere of 5% CO2.

In vitro assay of chemosensitivity to DOX

TPC-1 cells infected with Ad/sh-NOB1 or Ad/sh-Scramble (MOI of 50 each), along with untreated cells were seeded in a 96-well plates at a density of 5×103 cells/well and were cultured for 24 h. DOX was freshly prepared and added to cells with final concentration series from 0 to 10,000 nM. After incubation for 72 h, cell viability was determined by MTT assay as described below. The IC50 values were calculated from three independent experiments.

Proliferation assays

To measure the effect of Ad/sh-NOB1 combined with DOX or as monotherapy on cell proliferation, MTT assay was performed. Briefly, TPC-1 cells grown in monolayers were collected and dispensed in 96-well culture plates in 100 μl of DMEM at a concentration of 5×103 cells/well. After being cultured for 24 h, the cells were treated with Ad/sh-NOB1 (MOI of 50 each), Ad/sh-Scramble (MOI of 50 each), DOX (2X IC50) or Ad/sh-NOB1 (MOI of 50 each) in combination with DOX (1X IC50), respectively. After 48 h, the cells were washed with PBS (pH 7.4) and incubated in 50 μl of 0.5 mg/ml MTT in culture medium at 37°C for 4 h. Then 150 μl dimethyl sulfoxide (DMSO) was added to dissolve the crystals. After 10 min at room temperature, the absorbance was recorded at 570 nm in an ELISA plate reader (Molecular Devices Corporation, Sunnyvale, CA, USA). The mean proliferation of cells without any treatment was expressed as 100%.

Colony formation assay

TPC-1 cells were seeded in 6-well culture plates at 1×104 cells/well. After being cultured for 24 h, the cells were treated with Ad/sh-NOB1 (MOI of 50 each), Ad/sh-Scramble (MOI of 50 each), DOX (2X IC50) or Ad/sh-NOB1 (MOI of 50 each) in combination with DOX (1X IC50), respectively. After 14 days, the cells were washed, fixed in paraformaldehyde, and stained with Giemsa for 10 min. Extra Giemsa was washed three times by ddH2O, and the colonies were photographed using a digital camera. The visible colonies in each group were counted.

Cell cycle analysis

TPC-1 cells (5×105) were plated in 60-mm dishes and treated with Ad/sh-NOB1 (MOI of 50 each), Ad/sh-Scramble (MOI of 50 each), DOX (2X IC50) or Ad/sh-NOB1 (MOI of 50 each) in combination with DOX (1X IC50), respectively. Forty-eight hours after treatment, the cells were collected by trypsinization, fixed in 70% ethanol, and kept at −20°C overnight for fixation. The cells were washed in PBS, resuspended in 1 ml of PBS containing 100 μg/ml RNase and 40 μg/ml PI and incubated in the dark at room temperature for 30 min. Cell distribution in the cell cycle phases was analyzed from the DNA histogram with a FACSCalibur flow cytometer and CellQuest software (both from Becton-Dickinson, San Jose, CA, USA).

Cell apoptosis

Cell apoptosis was detected using flow cytometry. In briefly, TPC-1 cells were treated with Ad/sh-NOB1 (MOI of 50 each), Ad/sh-Scramble (MOI of 50 each), DOX (2X IC50) or Ad/sh-NOB1 (MOI of 50 each) in combination with DOX (1X IC50), respectively. After 48 h, 1×106 cells were digested with 10 μg/ml RNase for 30 min at 37°C. Annexin V-fluorescein isothiocyanate (0.5 μg/ml) and PI (0.6 μg/ml) were added to a 250 μl aliquot of this cell suspension. After a 15-min incubation in the dark at room temperature, the sample was read on a Coulter EPICS XL flow cytometer (Beckman Coulter, Inc., Fullerton, CA, USA), and the data were analyzed using CellQuest software (BD Biosciences, San Jose, CA, USA). Experiments were performed in triplicate. In addition, caspase-3, −8 and −9 activity were detected as an additional indicator of apoptosis.

Caspase activity

Caspase-3, −8 and −9 activity was measured using caspase colorimetric protease assay kits (Millipore Corporation, Billerica, MA, USA) according to the manufacturer’s instructions.

Transwell migration and invasion assays

To assess the effect of DOX and Ad/sh-NOB1 on cell migration and invasion, the migration and invasion assays were performed using Transwell insert chambers (Corning, Costar, Cambridge, MA, USA). For the migration assay, TPC-1 cells were treated with Ad/sh-NOB1 (MOI of 50 each), Ad/sh-Scramble (MOI of 50 each), DOX (2X IC50) or Ad/sh-NOB1 (MOI of 50 each) in combination with DOX (1X IC50), respectively. Forty-eight hours post-treatment, 1×105 cells were plated into the upper chamber in serum-free RPMI-1640 medium. Medium containing 20% FBS in the lower chamber served as the chemoattractant. After being cultured for 24 h, the medium was removed from the upper chamber by wiping with a cotton swab and cells that migrated to the lower surface of filter were fixed in 70% ethanol for 30 min, followed by staining with 0.2% crystal violet for 10 min. Cell migration was determined by counting five random fields/filter under a light microscope (Olympus, Tokyo, Japan).

For the invasion assay, after treatment, 3×105 cells were seeded in upper chambers precoated with Matrigel (BD Biosciences, Bedford, MA USA) in serum-free medium in triplicate, and the subsequent steps were similar to those of the migration assay. The number of cells invading the Matrigel was counted in five randomly selected fields using an inverted microscope (Olympus). All the experiments were performed in triplicate.

Measurement of matrix metalloproteinase-9 (MMP-9) and vascular endothelial growth factor (VEGF) level

MMP-9 and VEGF levels were determined by ELISA. Briefly, TPC-1 cells were treated with Ad/sh-NOB1 (MOI of 50 each), Ad/sh-Scramble (MOI of 50 each), DOX (2X IC50) or Ad/sh-NOB1 (MOI of 50 each) in combination with DOX (1X IC50) for 48 h in 24-well plates, and the culture media were centrifuged to remove cell debris. PGE2 levels in the cell supernatant were then measured by human MMP-9 ELISA kits (R&D Systems, Shanghai, China) according to the manufacturer’s instructions. VEGF levels in the cell supernatant were determined by a human VEGF ELISA kit (Yanyu, Shanghai, China) according to the manufacturer’s instructions.

Western blotting

Proteins were extracted from TPC-1 cells, and were characterized using western blot analysis. Briefly, the cells were lysed in lysis buffer. The homogenates were then centrifuged at 14,000 rpm at 4°C for 30 min to remove insoluble material, and the supernatants were collected for protein concentration determination using the BCA assay kit (Sigma-Aldrich). Cell extracts (20 μg of protein) were separated on a sodium dodecyl sulfate-polyacrylamide electrophoretic gel (SDS-PAGE) and transferred to nitrocellulose membranes, which were blocked in 3% bovine serum albumin (BSA) in PBST for 1 h at room temperature. The membrane was probed with primary antibody overnight at 4°C. After extensive washing, the membrane was incubated with secondary antibody for 1 h at room temperature. The proteins detected by the enhanced protein bands were visualized with enhanced chemiluminescent reagent (ECL; Sigma-Aldrich). The integrated densities value (IDV) was analyzed with computerized image analysis system (Fluor Chen 2.0) and normalized with that of GAPDH.

Tumor xenograft in nude mice

Female BALB/c nude mice at 6–7 weeks old were obtained from the Experimental Animal Center of the Jilin University (Changchun, China) and maintained under specific pathogen-free conditions and provided with food and water ad libitum.

Exponentially growing TPC-1 cells were collected and a tumorigenic dose of 2×106 cells was injected intraperitoneally into 4- to 5-week-old female BALB mice. The tumor volume was calculated using the formula: Volume = (length × width2)/2. When tumors grew to an average volume of 100 mm3, the mice were divided randomly into five groups (n=10 mice/group). The control group received 1% PBS in deionized water. The remaining four groups were treated with Ad/sh-Scramble (1×108 PFU/dose), DOX (4 mg/kg body weight), Ad/sh-NOB1 (1×108 PFU/dose), or DOX plus Ad/sh-NOB1 (DOX, 2 mg/kg body weight; Ad/sh-NOB1, 1×108 PFU/dose, respectively) intraperitoneally on alternative days for 3 weeks. Tumor size was measured using calipers prior to administration of the treatment injections and on day 7, 14 and 21 of treatment. On day 21, the animals were euthanized using chloroform, and tumor tissues were isolated and weighed. In addition, spleen tissues of the mice were collected and cultured for a splenocyte surveillance study, as previously described (19). The present animal study was performed following approval of the protocol by the Jilin University Animal Care and Use Committee (Changchun, Jilin, China)

Statistical analysis

Data from at least three independent experiments were expressed as mean ± standard deviation (SD). One-way ANOVA followed by Tukey’s post-hoc test were utilized to determine the significant difference among multiple groups. Statistical analyses were undertaken using GraphPad Prism version 5.01 (GraphPad Software, San Diego, CA, USA) and the SPSS® statistical package, version 19.0 (SPSS, Inc., Chicago, IL, USA) for Windows®. P<0.05 was considered to indicate a statistically significant result.

Results

Effects of DOX and Ad/sh-NOB1 alone or in combination on cell proliferation and colony formation of TPC-1 cells

TPC-1 cells were infected with Ad/sh-NOB1 or Ad/sh-Scramble, and then incubated with DOX (0–10,000 nM). MTT assays were performed following treatment and the IC50 for DOX was calculated for each of the groups treated. It was found that downregulation of NOB1 by Ad/sh-NOB1 resulted in a 50.56% reduction of the DOX IC50 (75.97±6.12 nM) in TPC-1 cells as compared to cells infected Ad/sh-Scramble (150.25±11.58 nM) (Fig. 1A). Thus, a reduced NOB1 expression sensitized TPC-1 cells to DOX.

To evaluate the effect of DOX combined with Ad/sh-NOB1 or independently on proliferation of PTC cells, an MTT assay was performed 48 h after TPC-1 cells were treated with DOX and Ad/sh-NOB1 alone or in combination. The results showed a significant decrease in cell proliferation as compared to the control and Ad/sh-Scramble treatment group (Fig. 1B, P<0.01). Compared to the monotherapy group, DOX in combination with Ad/sh-NOB1 obviously reduced cell proliferation (Fig. 1B, P<0.05).

The effects of DOX and Ad/sh-NOB1 alone or combination on the TPC-1 cell colony formation cycles of cells were then analyzed. As shown Fig. 1C, colony number tumor cell have significantly reduction in DOX and Ad/sh-NOB1 alone or combination groups compared with control and Ad/sh-Scramble group, (P<0.05, Fig. 1C). The DOX in combination with Ad/sh-NOB1 resulted in a greater reduction of colony number compared to either DOX or Ad/sh-NOB1 (P<0.05, Fig. 1C).

Effects of DOX and Ad/sh-NOB1 alone or in combination on cell cycle of TPC-1 cells

The effects of DOX and Ad/sh-NOB1 alone or in combination on the cell cycles of TPC-1 cells were analyzed using flow cytometry. It was found that TPC-1 cells treated with DOX and Ad/sh-NOB1 alone or in combination had an increased percentage of arrest at the G0/G1 phase, and a decreased percentage of arrest at the S stage compared to the control and Ad/sh-Scramble groups (P<0.05, Fig. 2A and B). The DOX combination with Ad/sh-NOB1 resulted in a greater effect at the G0/G1 phase and S stage (Fig. 2A and B, P<0.01).

We analyzed the effects of DOX and Ad/sh-NOB1 alone or in combination on the expression of cell cycle relevant proteins, such as cyclin D1 and D3 and p21 by western blotting. As shown in Fig. 2C and D, p21 expression was significantly increased, while cyclin D1 and D3 expression was significantly decreased in the DOX and Ad/sh-NOB1 alone or combination group compared to the control and Ad/sh-Scramble groups (P<0.05). Compared to the DOX or Ad/sh-NOB1 groups, the combination treatment group showed a synergistic effect on p21, cyclin D1 and D3 protein expression (P<0.05, Fig. 2C and D).

Effects of DOX and Ad/sh-NOB1 alone or in combination on cell apoptosis of TPC-1 cells

To investigate whether DOX and Ad/sh-NOB1 alone or in combination induced apoptosis, cell apoptosis was analyzed by flow cytometry following treatment. It was found that TPC-1 cells treated with DOX and Ad/sh-NOB1 alone or in combination significantly induced cell apoptosis compared with the control and Ad/sh-Scramble groups (P<0.05, Fig. 3A). Treatment with a combination of DOX and Ad/sh-NOB1 led to a marked increase in apoptotic cells compared to single DOX or Ad/sh-NOB1 treatment groups (P<0.05, Fig. 3A).

To examine the possible mechanism of induction of cell apoptosis of the combination of DOX and Ad/sh-NOB1, caspase-3, −8 and −9 activity was determined by ELISA. The results showed that DOX and Ad/sh-NOB1 alone or combined significantly increased caspase-3, −8 and −9 activity in TPC-1 cells, compared to the control and Ad/sh-Scramble groups (P<0.05, Fig. 3B and D). Compared to DOX or Ad/sh-NOB1 alone, caspase-3, −8 and −9 activity was significantly increased in the combination treatment group (P<0.05, Fig. 3B and D).

Effects of DOX and Ad/sh-NOB1 alone or in combination on cell migration and invasion of TPC-1 cells

To determine the effect of DOX and Ad/sh-NOB1 alone or as combined treatment on PTC cell migration and invasion, a Transwell assay was performed. The Transwell assay (without Matrigel) showed that the migratory speed of TPC-1 cells was markedly slower in the DOX and Ad/sh-NOB1 alone or combined groups than that of the control and Ad/sh-Scramble groups (Fig. 4A, P<0.05). Compared to the DOX and Ad/sh-NOB1 group, migration was significantly reduced in the combination group. Transwell matrix penetration (coated with Matrigel) assay showed that DOX and Ad/sh-NOB1 alone or in combination markedly reduced the invasiveness of TPC-1 cells as compared to the control and Ad/sh-Scramble groups (Fig. 4B, P<0.05). DOX in combination with Ad/sh-NOB1 resulted in the greatest reduction of invation of TPC-1 cells (Fig. 4B, P<0.05).

We also analyzed the effects of DOX and Ad/sh-NOB1 alone or in combination on the level of MMP-9 and VEGF by ELISA. It was found that MMP-9 and VEGF were significantly decreased in the DOX and Ad/sh-NOB1 alone or combinatorial treatment group compared to the control and Ad/sh-Scramble groups (P<0.05, Fig. 4C and D). The combinatorial treatment group showed synergistic inhibition of the MMP-9 and VEGF level.

Effects of DOX and Ad/sh-NOB1 alone or in combination on the p38MAPK pathway

It was previously shown that DOX stimulated the p38MAPK pathway and induced cell apoptosis (20). Therefore, we examined whether this mediated signaling pathway was enhanced by DOX in combination with AD/sh-NOB1. Measurements of the phosphorylation/activation pattern of p38MAPK, ERK1/2 and JNK were performed by western blotting 12 h after treatment. It was shown that DOX and Ad/sh-NOB1 alone or in combination resulted in a marked addition of phosphorylated p38MAPK, ERK1/2 and JNK as compared to the control and Ad/sh-Scramble groups, without altering the total protein levels of p38MAPK, ERK1/2 and JNK in each group (Fig. 5). Compared to the monotherapy group, the DOX and Ad/sh-NOB1 combinatorial treatment group increased phosphorylated p38MAPK, ERK1/2 and JNK protein expression, demonstrating a synergistic effect.

Effects of DOX and Ad/sh-NOB1 alone or in combination on tumor growth in vivo

We assessed the in vivo therapeutic efficacy of DOX and Ad/sh-NOB1 alone or in combination on female BALB mice bearing TPC-1 tumor cells. Tumor growth was monitored for 21 days. On day 21, the mice were anaesthetized and decapitated, and then tumors were excised, weighed and measured. The tumor weight was significantly reduced in the DOX and Ad/sh-NOB1 alone or combinatorial groups compared to that of the control (PBS group) and Ad/sh-Scramble groups (P<0.05; Fig. 6A and B). Compared to other groups, the DOX in combination with Ad/sh-NOB1 group showed maximally reduced weight. The tumor volume was determined at different time points (7, 14 and 21 day). The tumor volume in the DOX and Ad/sh-NOB1 alone or combinatorial groups were significantly (P<0.05) reduced compared to the Ad/sh-Scramble and control groups (PBS group) at the different time points (P<0.05, Fig. 6C). DOX in combination with Ad/sh-NOB1 led to a significant inhibition of tumor volume compared with other treatment groups (P<0.05, Fig. 6C).

In addition, we employed MTT assays to modulate splenocyte proliferation to demonstrate the antitumor activities. We found that splenocyte cell proliferation in the DOX and Ad/sh-NOB1 alone or combinatorial groups were significantly decreased as compared to the control and Ad/sh-Scramble groups (P<0.05, Fig. 6D). The combination treatment led to a significant inhibition of splenocyte cell proliferation compared with other treatment groups (P<0.05, Fig. 6D). These results suggested that the most prominent tumor decrease occurred in the DOX in combination with Ad/sh-Scramble group, demonstrating a synergistic effect in vivo.

Discussion

It is well known that drug resistance is a major hindrance encountered in cancer chemotherapy. During tumorigenesis or the course of chemotherapy, alteration of the gene expression may occur and influence cell sensitivity to chemotherapeutic drugs (21). In the present study, we found that the downregulation of NOB1 expression by Ad/sh-NOB1 sensitized TPC-1 cells to doxorubicin (DOX). In addition, our in vitro studies demonstrated that Ad/sh-NOB1 in combination with a reduced dose of DOX significantly suppressed cell proliferation, migration and invasion, and induced cell apoptosis and cell cycle arrest at G0/G1 stage compared to an increased dose of DOX. Additionally, further studies with in vivo mouse models confirmed that this combination significantly the suppressed tumor growth of PTC compared to a high dose of DOX. Therefore, combination of Ad/sh-NOB1 and DOX may serve as a novel therapeutic strategy for further evaluation in clinical trials for the treatment of PTC.

NOB1 is an important component of 26S proteasome, and favors 26S proteasome biogenesis (22). The 26S proteasome involved in the ubiquitin-proteasome pathway (UPP), required for the degradation of cyclic proteins and regulates multiple aspects of the cell cycle progression in eukaryotes (23,24). Previous studies have demonstrated that inhibition of 26S proteasomes is an effective anticancer therapeutic approach in vitro and in vivo. For example, bortezomib, one of the first proteasome inhibitors, which was designed to inhibit the activity of the 26S proteasome by binding to the N-terminal threonine residues at the active site of the catalytic region (25), has been shown to act as an effective drug against various human cancers and solid tumor types (2527). Therefore, NOB1, a key factor in the UPP and proteasome complex, has received attention, and has been found to play role in tumor development and progression (1118). In a recent study (18), we showed that blockade of NOB1 using Ad/sh-NOB1 via direct intratumoral injections significantly reduced PTC tumor growth in nude mice. However, complete suppression of tumor growth was not observed. RNA interference does not completely block gene expression, particularly when the target mRNA is expressed at abnormally high levels (28). Findings of our recent study showed that Ad/sh-NOB1 combined with irradiation treatment led to a significant inhibition of tumor growth in nude mice compated to Ad/sh-NOB1 treatment, which suggested that Ad/sh-NOB1 combined with other drugs or irradiation may lead to a stronger antitumor effect for human thyroid cancer. Therefore, in the present study, we used Ad/sh-NOB1 combined with DOX for this purpose. Our results show that the most prominent tumor decrease occurred in DOX combined with the Ad/sh-Scramble group as compared to Ad/sh-NOB1 and DOX as monotherapy, demonstrating a synergistic effect in vivo. These findings together with those of our previous study (17,18) have shown that the downregulation of NOB1 expression by Ad/sh-NOB1 suppressed the tumor growth of PTC in vitro and in vivo, and enhanced the radio- and chemosensitivity of PTC cells.

The MAPK pathway plays a crucial role in signal transduction and mediates cell proliferation, differentiation, inflammation and apoptosis, which consists of extracellular signal-regulated kinase (ERK), p38 and Jun N-terminal kinase (JNK) (29). It has been demonstrated that DOX suppressed tumor growth via activation of the MAPK pathway (19). In particular, it has been shown that knockdown of NOB1 expression inhibited tumor growth, to some extent, by activating the MAPK pathway (30,31). Our recently study also showed that adenovirus-mediated siRNA targeting NOB1 could active MAPK pathway (18). Therefore, whether mediated MAPK pathway was enhanced by DOX in combination with Ad/sh-NOB1 was determined. In the present study, we found that Ad/sh-NOB1 in combination with DOX treatment resulted in a marked increase of phosphorylated f p38MAPK, ERK1/2 and JNK relative to DOX and Ad/sh-NOB1 group, without altering the total protein levels of f p38MAPK, ERK1/2 and JNK in each group. These findings suggest that Ad/sh-NOB1 in combination with DOX demonstrated the synergistic effect on inhibiting the tumor growth of PTC in vitro and in vivo, at least in part through activation of the MAPK pathway.

Taken together, the findings reported herein present evidence that Ad/sh-NOB1 in combination with DOX significantly inhibited cell proliferation, migration and invasion, induced cell apoptosis in vitro, and suppressed tumor growth in vivo, compared to either Ad/sh-NOB1 or DOX, demonstrating a synergistic effect. In addition, this combination also activated the p38MAPK signaling pathway, which contributes to inhibition of tumor growth. These findings suggest that the combination of DOX and Ad/sh-NOB1 is a promising drug candidate for the treatment of PTC.

Acknowledgements

This study was supported by the Science and Technology Research and Innovation Team funded of Jilin Provincial (JL20130518).

References

1 

Brown RL, de Souza JA and Cohen EE: Thyroid cancer: burden of illness and management of disease. J Cancer. 2:193–199. 2011. View Article : Google Scholar : PubMed/NCBI

2 

Pitoia F, Bueno F, Urciuoli C, Abelleira E, Cross G and Tuttle RM: Outcomes of patients with differentiated thyroid cancer risk-stratified according to the American thyroid association and Latin American thyroid society risk of recurrence classification systems. Thyroid. 23:1401–1407. 2013. View Article : Google Scholar : PubMed/NCBI

3 

Thompson L: World Health Organization classification of tumours: pathology and genetics of head and neck tumours. Ear Nose Throat J. 85:742006.PubMed/NCBI

4 

Nikiforova MN and Nikiforov YE: Molecular genetics of thyroid cancer: implications for diagnosis, treatment and prognosis. Expert Rev Mol Diagn. 8:83–95. 2008. View Article : Google Scholar

5 

Carvalho C, Santos RX, Cardoso S, et al: Doxorubicin: the good, the bad and the ugly effect. Curr Med Chem. 16:3267–3285. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Gottesman MM, Fojo T and Bates SE: Multidrug resistance in cancer: role of ATP-dependent transporters. Nat Rev Cancer. 2:48–58. 2002. View Article : Google Scholar : PubMed/NCBI

7 

Silber JH and Barber G: Doxorubicin-induced cardiotoxicity. N Engl J Med. 333:1359–1360. 1995. View Article : Google Scholar : PubMed/NCBI

8 

King PD and Perry MC: Hepatotoxicity of chemotherapy. Oncologist. 6:162–176. 2001. View Article : Google Scholar : PubMed/NCBI

9 

Zhang Y, Ni J, Zhou G, et al: Cloning, expression and characterization of the human NOB1 gene. Mol Biol Rep. 32:185–189. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Arcus VL, Bäckbro K, Roos A, Daniel EL and Baker EN: Distant structural homology leads to the functional characterization of an archaeal PIN domain as an exonuclease. J Biol Chem. 279:16471–16478. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Lin Y, Peng S, Yu H, et al: RNAi-mediated downregulation of NOB1 suppresses the growth and colony-formation ability of human ovarian cancer cells. Med Oncol. 29:311–317. 2012. View Article : Google Scholar

12 

Che JP, Li W, Yan Y, et al: Expression and clinical significance of the nin one binding protein and p38 MAPK in prostate carcinoma. Int J Clin Exp Pathol. 6:2300–2311. 2013.PubMed/NCBI

13 

Li XY, Luo QF, Li J, et al: Clinical significance of NOB1 expression in breast infiltrating ductal carcinoma. Int J Clin Exp Pathol. 6:2137–2144. 2013.PubMed/NCBI

14 

Li Y, Ma C, Qian M, Wen Z, Jing H and Qian D: Downregulation of NOB1 suppresses the proliferation and tumor growth of non-small cell lung cancer in vitro and in vivo. Oncol Rep. 31:1271–1276. 2014.PubMed/NCBI

15 

Huang WY, Chen DH, Ning L and Wang LW: siRNA mediated silencing of NIN1/RPN12 binding protein 1 homolog inhibits proliferation and growth of breast cancer cells. Asian Pac J Cancer Prev. 13:1823–1827. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Lu Z, Guo Q, Shi A, et al: Downregulation of NIN/RPN12 binding protein inhibit the growth of human hepatocellular carcinoma cells. Mol Biol Rep. 39:501–507. 2012. View Article : Google Scholar

17 

Lin S, Meng W, Zhang W, et al: Expression of the NOB1 gene and its clinical significance in papillary thyroid carcinoma. J Int Med Res. 41:568–572. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Meng W, Wang PS, Liu J, et al: Adenovirus-mediated siRNA targeting NOB1 inhibits tumor growth and enhances radiosensitivity of human papillary thyroid carcinoma in vitro and in vivo. Oncol Rep. 32:2411–2420. 2014.PubMed/NCBI

19 

Abe S, Nishimoto Y, Isu K, Ishii T and Goto T; Japanese Musculoskeletal Oncology Group. Preoperative cisplatin for initial treatment of limb osteosarcoma: its local effect and impact on prognosis. Cancer Chemother Pharmacol. 50:320–324. 2002. View Article : Google Scholar : PubMed/NCBI

20 

Zhao BX, Sun YB, Wang SQ, et al: Grape seed procyanidin reversal of P-glycoprotein associated multi-drug resistance via down-regulation of NF-κB and MAPK/ERK mediated YB-1 activity in A2780/T cells. PLoS One. 8:e710712013. View Article : Google Scholar

21 

el-Deiry WS: Role of oncogenes in resistance and killing by cancer therapeutic agents. Curr Opin Oncol. 9:79–87. 1997. View Article : Google Scholar : PubMed/NCBI

22 

Tone Y and Toh-E A: Nob1p is required for biogenesis of the 26S proteasome and degraded upon its maturation in Saccharomyces cerevisiae. Genes Dev. 16:3142–3157. 2002. View Article : Google Scholar : PubMed/NCBI

23 

Tone Y, Tanahashi N, Tanaka K, Fujimuro M, Yokosawa H and Toh-E A: Nob1p, a new essential protein, associates with the 26S proteasome of growing Saccharomyces cerevisiae cells. Gene. 243:37–45. 2000. View Article : Google Scholar : PubMed/NCBI

24 

Xu G, Bernaudo S, Fu G, et al: Cyclin G2 is degraded through the ubiquitin-proteasome pathway and mediates the antiproliferative effect of activin receptor-like kinase 7. Mol Biol Cell. 19:4968–4979. 2008. View Article : Google Scholar : PubMed/NCBI

25 

Pandit B and Gartel AL: Thiazole antibiotic thiostrepton synergize with bortezomib to induce apoptosis in cancer cells. PLoS One. 6:e171102011. View Article : Google Scholar : PubMed/NCBI

26 

Piperdi B, Ling YH, Liebes L, Muggia F and Perez-Soler R: Bortezomib: understanding the mechanism of action. Mol Cancer Ther. 10:2029–2030. 2011. View Article : Google Scholar : PubMed/NCBI

27 

Dick LR and Fleming PE: Building on bortezomib: second-generation proteasome inhibitors as anti-cancer therapy. Drug Discov Today. 15:243–249. 2010. View Article : Google Scholar : PubMed/NCBI

28 

Elbashir SM, Harborth J, Weber K and Tuschl T: Analysis of gene function in somatic mammalian cells using small interfering RNAs. Methods. 26:199–213. 2002. View Article : Google Scholar : PubMed/NCBI

29 

Dhillon AS, Hagan S, Rath O and Kolch W: MAP kinase signalling pathways in cancer. Oncogene. 26:3279–3290. 2007. View Article : Google Scholar : PubMed/NCBI

30 

Zhou J, Xu T, Yan Y, et al: MicroRNA-326 functions as a tumor suppressor in glioma by targeting the Nin one binding protein (NOB1). PLoS One. 8:e684692013. View Article : Google Scholar : PubMed/NCBI

31 

Li W, Liu M, Feng Y, et al: Downregulated miR-646 in clear cell renal carcinoma correlated with tumour metastasis by targeting the nin one binding protein (NOB1). Br J Cancer. 111:1188–1200. 2014. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

March-2015
Volume 33 Issue 3

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
Liu J, Dong B, Wang P, Ren P, Xue S, Zhang X, Han Z and Chen G: Silencing NOB1 enhances doxorubicin antitumor activity of the papillary thyroid carcinoma in vitro and in vivo Retraction in /10.3892/or.2022.8297. Oncol Rep 33: 1551-1559, 2015
APA
Liu, J., Dong, B., Wang, P., Ren, P., Xue, S., Zhang, X. ... Chen, G. (2015). Silencing NOB1 enhances doxorubicin antitumor activity of the papillary thyroid carcinoma in vitro and in vivo Retraction in /10.3892/or.2022.8297. Oncology Reports, 33, 1551-1559. https://doi.org/10.3892/or.2015.3730
MLA
Liu, J., Dong, B., Wang, P., Ren, P., Xue, S., Zhang, X., Han, Z., Chen, G."Silencing NOB1 enhances doxorubicin antitumor activity of the papillary thyroid carcinoma in vitro and in vivo Retraction in /10.3892/or.2022.8297". Oncology Reports 33.3 (2015): 1551-1559.
Chicago
Liu, J., Dong, B., Wang, P., Ren, P., Xue, S., Zhang, X., Han, Z., Chen, G."Silencing NOB1 enhances doxorubicin antitumor activity of the papillary thyroid carcinoma in vitro and in vivo Retraction in /10.3892/or.2022.8297". Oncology Reports 33, no. 3 (2015): 1551-1559. https://doi.org/10.3892/or.2015.3730