Silencing XIAP suppresses osteosarcoma cell growth, and enhances the sensitivity of osteosarcoma cells to doxorubicin and cisplatin

Retraction in: /10.3892/or.2021.8213

  • Authors:
    • Yang Qu
    • Peng Xia
    • Shanyong Zhang
    • Su Pan
    • Jianwu Zhao
  • View Affiliations

  • Published online on: December 23, 2014     https://doi.org/10.3892/or.2014.3698
  • Pages: 1177-1184
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

X-chromosome-linked inhibitor of apoptosis protein (XIAP) is an important member of the inhibitors of apoptosis (IAP) family. It has been shown that XIAP promotes the invasion, metastasis, growth and survival of malignant cells, and confers resistance to some chemotherapeutic drugs in various types of cancer. However, little is known regarding its detailed role in osteosarcoma (OS). In the present study, we first investigated the expression of XIAP in OS tissues, and an increased expression of XIAP in OS tissues compared to adjacent non-tumor tissue was identified. Additionally, its expression level correlated with key pathological characteristics including clinical stage, tumor size and metastasis. Subsequently, small interfering RNA (siRNA) was used to block XIAP expression to evaluate the effect of XIAP siRNA on cell proliferation, colony formation, cell cycle, apoptosis, tumorigenicity, and the combined effects of doxorubicin or cisplatin in OS cell lines (MG63 cells). Downregulation of XIAP expression using the RNA silencing approach efficiently decreased cell proliferation and colony formation, and induced cell apoptosis and cell cycle in the G0/G1 stage. In addition, this downregulation inhibited tumor growth in a nude murine model. The resuls also showed that treatment with XIAP-shRNA in combination with doxorubicin or cisplatin enhanced chemosensitivity. These results suggested that XIAP may aid the diagnosis of OS and may be an effective strategy for gene therapy of this disease.

Introduction

Osteosarcoma (OS) is the most common primary bone malignancy in children and adolescents (1,2). Although neoadjuvant chemotherapy followed by surgical excision has ameliorated the long-term survival of OS patients, most patients that do not respond to commonly used drugs, such as cisplatin and doxorubicin, have a poor prognosis due to intrinsic or acquired drug resistance (3,4). Identification of the critical molecules and/or signal transduction pathways responsible for regulating the development to drug resistance is therefore required to optimize therapeutic options or develop novel effective therapy for OS (5,6).

At the molecular level, a number of aberrations have been identified that contribute to this chemoresistance. The deregulated expression of members of the inhibitor of apoptosis protein (IAP) family, such as X-chromosome-linked inhibitor of apoptosis protein (XIAP) and survivin (7,8), have been shown to influence the sensitivity towards chemotherapeutic agents. XIAP, an important member of the IAP family proteins, binds to and inhibits the activities of caspase-3, −7 or −9, leading to apoptosis inhibition (9). XIAP is extensively expressed in many types of cancer and is not expressed or expressed at a substantially lower level in normal tissue counterparts (1012). Accumulating evidence showed that inhibition of XIAP expression using antisense oligonucleotides or small-interfering RNA (siRNA) suppressed tumor cell proliferation and invasion, induced cell apoptosis, and suppressed tumor growth (1318). In addition, Harlin et al reported that XIAP knockout mice are phenotypically normal with no significant pathological characteristics (19). XIAP is considered one of the most important factors involved in resistance to the apoptotic effects of drugs and radiation in tumor cells (20). It has been shown to be one of the important regulators in cisplatin- and doxorubicin-induced apoptosis in some cancer cells (21,22) and downregulation of XIAP sensitizes cells to cisplatin and doxorubicin (13,23). Therefore, XIAP is considered to be an attractive target with respect to the molecular therapy of cancer. However, for XIAP, the possible roles and whether increase resistance of doxorubicin and cisplatin in OS remain unclear.

The present study was undertaken to evaluate the expression and its clinical diagnostic significance in OS patients and to analyze its functional role, especially with respect to chemoresistance. Our results showed that the downregulation of XIAP expression by short hairpin RNA (shRNA) significantly inhibited MG63 cell proliferation and colony formation, induced cell arrest in the G0/G1 and cell apoptosis, and suppressed OS tumor growth in a nude murine model. Furthermore, downregulation of XIAP significantly enhanced the chemosensitivity of doxorubicin and cisplatin in OS cells. These experimental data suggested that XIAP was associated with the development of human OS and silencing XIAP is a potential strategy therapy for human OS.

Materials and methods

Patients and tissue samples

This study was approved by the Research Ethics Committee of the Second Hospital of Jilin University (Changchun, China). Written informed consent was obtained from all the patients.

Sixty OS patients (36 male and 24 female) treated in the Department of Orthopedics, The Second Hospital of Jilin University, Changchun, China, between August 2008 and August 2013, were enrolled retrospectively in this study. The human OS biopsy specimens were obtained from primary lesions. The biopsies were performed prior to chemotherapy or radiotherapy for diagnostic purposes. Clinicopathological characteristics were recorded from the subjects, such as personal information, diagnosis, disease stage, metastasis and tumor characteristics. The selection criteria were selected as clear pathological diagnosis, full medical records, and completed follow-up data. Patients were excluded when a previous or secondary malignancy was identified. Normal tissue samples adjacent to the tumor were removed 5 cm away from the peripheral tumor cells, and the lack of tumor cell infiltration was verified by pathological examination.

Immunohistochemistry

Samples were divided into the OS group and tissue adjacent to tumor, which was stained against XIAP, in the two treatment groups. Briefly, the sections were dewaxed in xylene, rehydrated in descending alcohols, and blocked for endogenous peroxidase and avidin/biotin activities. After blocking with 3% BSA in 0.01 M PBS, the sections were incubated with mouse monoclonal antibody against human XIAP (Santa Cruz Biotechnology, Inc., Santa Cruz, CA, USA) at a dilution of 1:1,000 overnight at 4°C. The samples incubated with PBS instead of the primary antibody were used as the negative control. The samples were then incubated with horseradish peroxidase (HRP)-labeled goat anti-rabbit secondary antibody for 2 h at room temperature. After washing with PBS three times, immunostaining was visualized using a streptavidin-peroxidase reaction system, and developed with diaminobenzidine (DAB)-H2O2 (Wuhan Boster Biological Technology Ltd., China) and then observed under an optical microscope.

Immunoreactivity was graded according to the percentage of positive tumor cells (0, 0%; 1<10%; 2, 10–50%; 3, 51–80%; 4, 80–100%), and the intensity of staining (0, no staining; 1, weak; 2, moderate; and 3, strong). These scores were calculated by multiplying the intensity score to the percentage score. The section with a final score of <4 being classified as negative, and a score of ≥4 as positive.

shRNA design and plasmid construction

XIAP-shRNA (GenBank accession no. NM_001167) and non-specific shRNA sequences were designed using the siRNA target design tools (Ambion Inc., Austin, TX, USA), XIAP-shRNA and non-specific shRNA were synthesized as follows: XIAP-shRNA sense: 5′-GATCCACTGGACAGAGAAAGAGCCTTCAAGAGAGGCTCTTTCTCTGTCCAGTTTTTTTGGAAA-3′ and antisense: 5′-AGCTTTTCCAAAAAAACGGACAGAGAAAGAGCCTCTCTTGAAGGCTCTTTCTCTGTCCAGTG-3′; non-specific-shRNA sense: 5′-GATCCTTCTCCGAACGTGTCACGTTCAAGAGAAAACTACTTCTTTTACCTTAGA-3′ and antisense: 5′-AGCTTCTTCTTTCTCCGAACGTGTCACGTCTCTTGAAAAACTACTTCTTTTACCTTG-3′, where the sequence did not target any gene product and where there was no significant sequence similarity to human gene sequences. The oligonucleotides were annealed and then ligated into the BamHI and HindIII sites of pSilencer 6.1-CMV neo expression vector according to the manufacturer’s instructions (Ambion). The two recombinant plasmids were verified by DNA sequencing. The plasmid encoding XIAP-shRNA was referred to as pSi-XIAP, and the plasmid encoding non-specific shRNA, which was used as a negative control, was designated as pSi-NC.

Cell culture and transfection

Human MG63 OS cells were obtained from the Cell Bank of the Type Culture Collection of Chinese Academy of Sciences, Shanghai Institute of Cell Biology (Shanghai, China). MG63 cells were grown in DMEM medium (HyClone, Logan, UT, USA) supplemented with 10% fetal bovine serum (FBS, Gibco, Carlsbad, CA, USA), 2 mM glutamine, 100 U/ml penicillin and 100 mg/ml streptomycin at 37°C in a humidified atmosphere containing 5% CO2.

MG63 cells were seeded in 6-well plates at 2.0×104 cells/well, respectively, and cultured overnight to 80% confluence prior to transfection. The cells were then transfected with plasmid pSi-XIAP or plasmid pSi-NC using Lipofectamine™ 2000 (Invitrogen, Carlsbad, CA, USA) in medium according to the manufacturer’s instructions. G418 (800 μg/ml, Sigma, St. Louis, MO, USA) was used to screen stably transfected clones. Stable transfectants were designated to as MG63/pSi-XIAP and MG63/pSi-NC. After 48 h, quantitative real-time polymerase chain reaction (RT-qPCR) and western blot analysis were performed to evaluate the effect of gene silencing.

Quantification by real-time polymerase chain reaction

Total RNA of OS tissue and cells was isolated using TRIzol reagent following the manufacturer’s instructions (Invitrogen), and quantified with the Nanodrop 2000 (Thermo, Japan). First-strand cDNA synthesis and amplification were performed using reverse transcription reagents (Takara, China) according to the manufacturer’s instructions. RT-qPCR assays were carried out using SYBR-TAQ real-time kits (Takara Biotechnology, Otsu, Japan) and amplification equipment ABI Prism 7900 HT (Applied Biosystems, Foster City, CA, USA). GADPH was used as the endogenous control for quantifying mRNA levels. Primers used for of XIAP and GAPDH were: XIAP sense: 5′-GACAGTATGCAAGATGAGTCAAGTCA-3′ and antisense: 5′-GCAAAGCTTCTCCTCTTGCAG-3′; GAPDH sense: 5′-TGTGGGCATCAATGGATTTGG-3′ and antisense: 5′-ACACCATGTATTCCGGGTCAAT-3′. The qPCR conditions were as follows: a pre-denaturing at 95°C for 5 min, followed by 40 cycles of denaturation at 95°C for 10 sec, annealing/extension at 60°C for 20 sec, within final extension of 72°C for 5 min. The amplification specificity was assessed by the melting curve analysis. The 2−ΔΔCt method was used to calculate the relative abundance of target gene expression generated using the Rotor-Gene 6000 Series Software 1.7 (Qiagen, Hilden, Germany).

Western blotting

After 48 h of transfection, MG63 cells were trypsinized, lysed in RIPA lysis buffer (pH 7.4, 150 mM NaCl, 0.1% SDS, 0.5% sodium deoxycholate, 1% NP-40 in PBS, protease complete inhibitor; Roche Diagnostics GmbH, Mannheim, Germany), frozen and thawed three times, centrifuged at 13,000 × g for 20 min at 4°C to remove insoluble material, and then the supernatant was harvested as total proteins for experiments. Total protein concentration was determined using the BCA assay kit (Sigma). Cell extracts (50 μg of protein) were separated by 8–15% SDS-PAGE gel and transferred to nitrocellulose membranes. After blocking non-specific binding sites with 5% dried milk in PBST, the membranes were incubated with anti-cleaved caspase-3 (1:1,500; Santa Cruz Biotechnology, Inc.), anti-β-actin (1:5,000; Sigma-Aldrich, St. Louis, MO, USA) anti-cleaved PARP (1:2,000) and anti-XIAP (1:2,000) (both from Santa Cruz Biotechnology, Inc.) antibody in PBST containing 3% BSA overnight at 4°C, followed by incubation for 2 h at room temperature with the appropriate HRP secondary antibody diluted (1:3,000) in PBST containing 3% BSA. Protein bands were visualized with enhanced chemiluminescence reagent (ECL; Amersham, GE Healthcare, Velizy-Villacoublay, France). Densitometry was performed by Quantity One image analysis software.

Cell proliferation and colony formation

The MTT assay was used to determine the effect of downregulated XIAP on cell proliferation. Briefly, MG63 cells transfected with pSi-XIAP and pSi-NC, along with untreated cells were seeded in 96-well plates at a density of 5×103 cells/well. At indicated time points, 20 μl methyl thiazol tetrazolium (MTT) solution (5 mg/ml) was added to each well and cultured for 4 h. Centrifugation was performed to remove the supernatant, and 200 μl of DMSO was added to each well followed by agitation for 10 min to dissolve the crystals. Absorbance was measured at 570 nm with a microplate reader (Molecular Devices Corp., Sunnyvale, CA, USA), and growth inhibition was calculated. All the experiments were performed in triplicate.

For the colony formation assay, MG63 cells transfected with plasmid pSi-XIAP or pSi-NC were seeded in a 6-well plate at a low density (1×103 cells/well), and then cultured for 7 days. The cells were fixed with 4% paraformaldehyde for 20 min and counted after staining with 1% crystal violet. The experiments were carried out in triplicate wells at least three times.

Cell cycle analysis

MG63 cells were plated in 10-cm dishes and treated with plasmid pSi-XIAP or pSi-NC for 48 h. The cells were trypsinized, fixed with 70% ethanol, and incubated overnight at −20°C. The cells were pelleted and resuspended in 200 μl of PBS with 50 μl DNAase-free, RNase A, and incubated at 37°C for 1 h. Propidium iodide (PI, 750 μl) was added and incubated at room temperature for 15 min. The DNA contents of samples were measured using a FACS-Calibur™ flow cytometer (BD Biosciences, San Jose, CA, USA). The flow data obtained from the samples were then analysed by CellQuest software (BD Biosciences).

Cell apoptosis

Cell apoptosis was detected using flow cytometry. Briefly, at 24 h after transfection, a total of 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 then 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., Brea, CA, USA), and the percentage of apoptotic cells was calculated using CellQuest software (BD Biosciences). Experiments were performed in triplicate. In addition, cleaved caspase-3 and cleaved PARP protein expression were determined by western blotting using specific antibody as an additional indicator of apoptosis.

In vitro drug treatment

MG63 cells transfected with plasmid pSi-XIAP or pSi-NC were cultured for 24 h, and the medium was changed to DMEM containing indicated concentrations of doxorubicin and cisplatin. After 72 h of treatment, cell proliferation was assessed using an MTT assay, and cell apoptosis was measured by flow cytometry. In addition, caspase-3 activity was determined by the caspase colorimetric protease assay kits (Millipore Corporation, Billerica, MA, USA), following the manufacturer’s instructions, as an additional indicator of apoptosis.

Tumor growth in vivo

To investigate the effects of downregulated XIAP by RNAi on the tumorigenicity of xenograft and the influence on survival of tumor-burdened animals, 30 female BALB/nude mice (aged 4–6 weeks) were purchased from the Jilin Institute of Experimental Animals. The study protocol was approved and mice were maintained in accordance with the Institutional Guidelines of the Experimental Animals of Jilin University.

Stable MG63/pSi-XIAP and MG63/pSi-NC clone cells and untreated MG63 cells (1×108), respectively, were subcutaneously injected into the right flank of mice. The animals were sacrificed on the 21st day after injection, tumor tissues were resected and the volume and weight were measured. In addition, spleen tissue was collected and cultured for a splenocyte surveillance study using the MTT assay as previously described (30).

Statistical analysis

Data are presented as mean ± SD (standard deviation). Statistical analysis between two samples was performed using the Student’s t-test. Statistical comparison of >2 groups was performed using one-way ANOVA followed by a Tukey post-hoc test. The relationship between XIAP expression levels and clinical and pathological variables was analysed using Pearson’s Chi-square test. Graphpad Prism 5.01 software (GraphPad Software, San Diego, CA, USA) and SPSS® 16.0 (SPSS Inc., Chicago, IL, USA) for Windows® were used for statistical analyses. P<0.05 was considered to indicate a statistically significant difference.

Results

XIAP is upregulated in OS patients and correlates with clinical characteristics of OS patients

To identify the potential roles of XIAP in the development and progression of OS, we detected XIAP expression levels in tumor tissues and adjacent non-tumoral tissues from 60 patients with OS by RT-qPCR and immunohistochemistry. As revealed by the RT-qPCR analysis, the mRNA expression level of XIAP was significantly upregulated in tumoral tissues compared to expression in the matched adjacent non-tumoral tissues (P<0.05; Fig. 1A). In addition, elevated levels of XIAP protein were found in OS tumoral tissues compared with the paired adjacent non-tumoral tissues from the same patients as shown by immunohistochemical staining (Fig. 1B).

The associations of XIAP protein expression with various clinicopathological parameters of OS tissues was analyzed. XIAP protein was significantly upregulated in OS patients with advanced clinical stage, larger tumour size (>8 cm) and positive distant metastasis as compared to those with low clinical stage, small tumor size (<8 cm) and without distant metastasis (all P<0.05) (Table I). No correlations occurred between XIAP protein levels and patient age, gender and anatomic location.

Table I

The correlation of XIAP expression with clinicopathological characteristics of osteosarcoma.

Table I

The correlation of XIAP expression with clinicopathological characteristics of osteosarcoma.

Clinical factorPositiveNegativeP-value
Age, years
 <20 (n=39)309>0.05
 ≥60 (n=21)156
Gender
 Male (n=36)288>0.05
 Female (n=24)177
Clinical stage
 I–IIA (n=31)2011<0.01
 IIB–III (n=29)254
Distant metastasis
 Absent (n=41)2714<0.01
 Present (n=19)181
Anatomic location
 Tibia/femur (n=46)3610>0.05
 Other location (n=14)95
Tumor size, cm
 <8 (n=38)2513<0.01
 ≥8 (n=22)202
Silencing XIAP causes effective and specific downregulation of XIAP expression in MG63 cells

We constructed a plasmid (pSi-XIAP) that is capable of expressing a shRNA that targets the XIAP, and then transfected this plasmid into MG63 cells. The mRNA and protein expression intensities of XIAP were analyzed by RT-qPCR and western blotting which showed that plasmid pSi-XIAP markedly decreased the expression of XIAP compared with the control and pSi-NC groups (Fig. 2A and B). The above results indicated that the expression of XIAP gene in MG63 cells was downregulated specifically and effectively by plasmid pSi-XIAP.

Silencing XIAP inhibited cell proliferation and colony formation in MG63 cells

The anti-proliferative effect of XIAP silencing on MG63 tumor cells was examined using the MTT assay following MG63 cell transfectection with individual plasmid. Downregulation of XIAP by pSi-XIAP could significantly inhibited the proliferation of MG63 tumor cells compared with the control and pSi-NC groups (P<0.01; Fig. 3A).

The effects of silencing XIAP on tumor cell colony formation were determined in MG63 cell lines by analyzing cell colony formation at 14 days after transfection with the indicated plasmid. Cell colony formation in the pSi-XIAP groups was significantly reduced compared to the control and pSi-NC groups (P<0.05; Fig. 3B). No significant difference was identified between the pSi-NC and control groups (P>0.05).

Silencing XIAP induces cell arrest and apoptosis in MG63 cells

To determine the effects of silencing XIAP on the cell cycle, FACScan flow cytometry assays were performed. A flow cytometric analysis revealed that G1-phase cell population was increased in the pSi-XIAP group compared to the control and pSi-NC groups (P<0.05; Fig. 4A and B). In addition, silencing XIAP resulted in a much lower percentage of cells in S phase compared with those of the control and pSi-NC groups (P<0.05; Fig. 4A and B).

In order to investigate the effect of silencing XIAP on cell apoptosis in MG63 cells, flow cytometry assays were performed. Flow cytometric analysis showed that cells transfected with plasmid pSi-XIAP significantly induced cell apoptosis compared to untreated cells and cells transfected with plasmid pSi-NC (P<0.05; Fig. 4C).

To determine the potential mechanism of cell apoptosis in vitro, cleaved PARP and cleaved caspase-3 protein expression were detected using western blotting. It was found that cleaved caspase-3 and cleaved PARP protein expression was significantly increased in pSi-XIAP treatment groups compared to the control and pSi-NC groups (Fig. 4D).

Downregulation of XIAP sensitizes OS cells to doxorubicin and cisplatin

To investigate whether downregulation of XIAP expression has the potential to sensitize OS cells to chemotherapy, a combination treatment of XIAP-specific siRNA with anticancer drugs was performed. Twenty-four hours after transfection with plasmid pSi-XIAP, cells were treated with doxorubicin and cisplatin at indicated concentrations for 72 h, respectively. To evaluate the growth inhibitory effects of pSi-XIAP in combination with doxorubicin or cisplatin in MG63 cells, MTT assays were performed. As shown in Fig. 5A, pSi-XIAP treatment significantly enhanced the growth inhibitory effect of doxorubicin and cisplatin in MG63 cells. The pSi-NC had either no effect or only a minimal effect.

We also evaluated the effect of pSi-XIAP combination with doxorubicin or cisplatin on cell apoptosis in MG63 cells. It was found that pSi-XIAP combination with doxorubicin or cisplatin significantly increased cell apoptosis compared to monotherapy or combination with pSi-NC (P<0.05; Fig. 5B). In addition, we evaluated the effect of pSi-XIAP combination with doxorubicin or cisplatin on caspase-3 activity, and found that pSi-XIAP combination with doxorubicin or cisplatin could increase caspase-3 activity compared to single drug treatment or combination with pSi-NC (P<0.05; Fig. 5C). These results suggested that downregulation of XIAP sensitizes OS cells to doxorubicin and cisplatin.

Silencing XIAP suppressed tumor growth in vivo

We also investigated the effect of silencing XIAP on tumor growth in nude mice. Tumor growth was monitored for 21 days. On day 21, the animals were sacrificed and tumor weight and volume were measured. Our results showed that tumor weight and volume of mice treated with pSi-XIAP were significantly reduced when compared to the control and pSi-NC groups (P<0.05; Fig. 6A–C). We employed MTT assay in modulating splenocyte proliferation to demonstrate the antitumor activities. We found that splenocyte cell proliferation of pSi-XIAP group was significantly decreased compared to the control and pSi-NC groups (P<0.01; Fig. 6D). These results indicated that suppression of XIAP expression in OS cells markedly inhibited their tumorigenicity in nude mice.

Discussion

It is well known that inhibition of apoptosis contributes to tumorigenesis by aberrantly prolonging cell life span, permitting resistance to immune-based cytotoxicity, and allowing diobeyance of cell cycle check points that usually induce apoptosis (24). Several critical genes in the regulation of apoptosis have been identified, such as the anti-apoptotic gene family IAPs and Bcl-2 family (9,25). The IAPs have been identified as acting downstream of Bcl-2 by inhibiting caspases (2527). Eight members of IAPs have been identified in humans, and XIAP is the most potent one (28). A larger number of studies in various types of cancer cells have suggested that the expression of XIAP is elevated in various cancers and that it is important in oncogenesis (1012,29). It has been shown that the downregulated expression of XIAP inhibited cell proliferation and induced the apoptosis of many types of tumor, and sensitized cancer cells to chemotherapeutic drugs (1324). However, the expression and detailed role of XIAP remains to be thoroughly investigated in OS. Therefore, this study mainly focuses on XIAP expression and its detailed role in OS. We first found that XIAP was elevated in most OS tissue compared to adjacent non-tumoral tissue, and its expression level correlated with key pathological characteristics, including clinical stage and metastasis. In addition, our findings show that downregulation of XIAP results in the inhibition of proliferation of MG63 cells in vitro, and suppression of solid tumor growth in a nude murine model. These results provide evidence that XIAP is required for tumor growth and that it is important in tumorigenicity for OS.

Cisplatin (cis-diamminedichloroplatinum or cis-DDP/CDDP) is an anticancer drug widely used in the treatment of various types of cancer such as ovarian cancer, bladder, cervical, head and neck, esophageal and small cell lung cancer (SCLC) and OS (13,21,23,30). Cisplatin-based treatment has been successful in treating some patients with OS to either prolong survival or ameliorate symptoms (30,31). Other OS patients remain inherently resistant to cisplatin, which limits the therapeutic efficacy of cisplatin (32). It has been shown that the overexpression of XIAP by adenoviral sense XIAP complementary DNA attenuated the ability of cisplatin to induce apoptosis (33). Extensive studies have shown that downregulation of XIAP using adenoviral antisense XIAP infection or siRNA increased sensitivity to cisplatin in resistant various cancer cells (13,34,35). Consistent with those results, our results first showed that downregulation of XIAP by plasmid pSi-XIAP increased sensitivity to cisplatin in resistant OS cancer cells. These findings support that XIAP is an adjuvant gene therapy target to the chemotherapy of OS.

Doxorubicin, an anthracycline antibiotic, was isolated from Streptomyces peucetius which has been widely used for the treatment of various types of cancer including lymphomas, leukemias, lung, breast, OS and ovarian cancers (36). In fact, doxorubicin is the most widely used anticancer drug with FDA approval (37). However, studies have shown that some cancer cells, including OS, are resistant to the apoptotic effects of doxorubicin (38). It has been shown that downregulation of XIAP by antisense oligonucleotide (ASO) induced cell apoptosis and increased sensitivity to doxorubicin in resistant OS cells (23). Consistent with that result, our study showed that treatment with XIAP-shRNA in combination with doxorubicin significantly induced cell apoptosis and increased caspase-3 activity, and enhanced sensitivity to doxorubicin in resistant OS cancer cells.

In conclusion, to the best of our knowledge, this is the first full-scale report concerning the association of XIAP with OS. The present study has demonstrated that XIAP was elevated in most OS patients and its expression level was correlated with key pathological characteristics, including clinical stage and metastasis. Additionally, downregulation of XIAP suppressed tumor growth in vitro and in vivo and increased sensitivity of OS cells to doxorubicin and cisplatin. Based on the multiple functions of XIAP in OS tumor growth, it may be considered a diagnostic marker and a potential anticancer therapeutic target for OS.

References

1 

Ottaviani G, Robert RS, Huh WW, Palla S and Jaffe N: Sociooccupational and physical outcomes more than 20 years after the diagnosis of osteosarcoma in children and adolescents: limb salvage versus amputation. Cancer. 119:3727–3736. 2013.PubMed/NCBI

2 

Ottaviani G and Jaffe N: The epidemiology of osteosarcoma. Cancer Treat Res. 152:3–13. 2009. View Article : Google Scholar

3 

Wittig JC, Bickels J, Priebat D, et al: Osteosarcoma: a multidisciplinary approach to diagnosis and treatment. Am Fam Physician. 65:1123–1132. 2002.PubMed/NCBI

4 

Meyers PA, Schwartz CL, Krailo M, et al: Osteosarcoma: a randomized, prospective trial of the addition of ifosfamide and/or muramyl tripeptide to cisplatin, doxorubicin, and high-dose methotrexate. J Clin Oncol. 23:2004–2011. 2005. View Article : Google Scholar : PubMed/NCBI

5 

Yang J and Zhang W: New molecular insights into osteosarcoma targeted therapy. Curr Opin Oncol. 25:398–406. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Zhao Z, Tao L, Shen C, Liu B, Yang Z and Tao H: Silencing of Barkor/ATG14 sensitizes osteosarcoma cells to cisplatin-induced apoptosis. Int J Mol Med. 33:271–276. 2014.

7 

Mobahat M, Narendran A and Riabowol K: Survivin as a preferential target for cancer therapy. Int J Mol Sci. 15:2494–2516. 2014. View Article : Google Scholar : PubMed/NCBI

8 

Wang S, Bai L, Lu J, Liu L, Yang CY and Sun H: Targeting inhibitors of apoptosis proteins (IAPs) for new breast cancer therapeutics. J Mammary Gland Biol Neoplasia. 17:217–228. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Salvesen GS and Duckett CS: IAP proteins: blocking the road to death’s door. Nat Rev Mol Cell Biol. 3:401–410. 2002. View Article : Google Scholar : PubMed/NCBI

10 

Devi GR: XIAP as target for therapeutic apoptosis in prostate cancer. Drug News Perspect. 17:127–134. 2004. View Article : Google Scholar : PubMed/NCBI

11 

Stennicke HR, Ryan CA and Salvesen GS: Reprieval from execution: the molecular basis of caspase inhibition. Trends Biochem Sci. 27:94–101. 2002. View Article : Google Scholar : PubMed/NCBI

12 

Holcik M, Gibson H and Korneluk RG: XIAP: apoptotic brake and promising therapeutic target. Apoptosis. 6:253–261. 2001. View Article : Google Scholar : PubMed/NCBI

13 

Ma JJ, Chen BL and Xin XY: XIAP gene downregulation by small interfering RNA inhibits proliferation, induces apoptosis, and reverses the cisplatin resistance of ovarian carcinoma. Eur J Obstet Gynecol Reprod Biol. 146:222–226. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Yamaguchi Y, Shiraki K, Fuke H, et al: Targeting of X-linked inhibitor of apoptosis protein or survivin by short interfering RNAs sensitize hepatoma cells to TNF-related apoptosis-inducing ligand- and chemotherapeutic agent-induced cell death. Oncol Rep. 14:1311–1316. 2005.PubMed/NCBI

15 

Jiang C, Yi XP, Shen H and Li YX: Targeting X-linked inhibitor of apoptosis protein inhibits pancreatic cancer cell growth through p-Akt depletion. World J Gastroenterol. 18:2956–2965. 2012. View Article : Google Scholar : PubMed/NCBI

16 

Kwatra SG: Targeting x-linked inhibitor of apoptosis protein for melanoma therapy: the need for more homogeneous samples and the importance of cell lines. J Invest Dermatol. 131:7972011. View Article : Google Scholar

17 

Hiscutt EL, Hill DS, Martin S, et al: Targeting X-linked inhibitor of apoptosis protein to increase the efficacy of endoplasmic reticulum stress-induced apoptosis for melanoma therapy. J Invest Dermatol. 130:2250–2258. 2010. View Article : Google Scholar : PubMed/NCBI

18 

Mastrangelo E, Cossu F, Milani M, et al: Targeting the X-linked inhibitor of apoptosis protein through 4-substituted azabicyclo[5.3.0]alkane smac mimetics. Structure, activity, and recognition principles. J Mol Biol. 384:673–689. 2008. View Article : Google Scholar : PubMed/NCBI

19 

Harlin H, Reffey SB, Duckett CS, Lindsten T and Thompson CB: Characterization of XIAP-deficient mice. Mol Cell Biol. 21:3604–3608. 2001. View Article : Google Scholar : PubMed/NCBI

20 

Deveraux QL and Reed JC: IAP family proteins - suppressors of apoptosis. Genes Dev. 13:239–252. 1999. View Article : Google Scholar : PubMed/NCBI

21 

Cheng JQ, Jiang X, Fraser M, et al: Role of X-linked inhibitor of apoptosis protein in chemoresistance in ovarian cancer: possible involvement of the phosphoinositide-3 kinase/Akt pathway. Drug Resist Updat. 5:131–146. 2002. View Article : Google Scholar : PubMed/NCBI

22 

Gagnon V, Van Themsche C, Turner S, Leblanc V and Asselin E: Akt and XIAP regulate the sensitivity of human uterine cancer cells to cisplatin, doxorubicin and taxol. Apoptosis. 13:259–271. 2008. View Article : Google Scholar

23 

Holt SV, Brookes KE, Dive C and Makin GW: Down-regulation of XIAP by AEG35156 in paediatric tumour cells induces apoptosis and sensitises cells to cytotoxic agents. Oncol Rep. 25:1177–1181. 2011.PubMed/NCBI

24 

Zhang Y, Wang Y, Gao W, et al: Transfer of siRNA against XIAP induces apoptosis and reduces tumor cells growth potential in human breast cancer in vitro and in vivo. Breast Cancer Res Treat. 96:267–277. 2006. View Article : Google Scholar

25 

Oberoi-Khanuja TK, Murali A and Rajalingam K: IAPs on the move: role of inhibitors of apoptosis proteins in cell migration. Cell Death Dis. 4:e7842013. View Article : Google Scholar : PubMed/NCBI

26 

Hunter AM, LaCasse EC and Korneluk RG: The inhibitors of apoptosis (IAPs) as cancer targets. Apoptosis. 12:1543–1568. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Berthelet J and Dubrez L: Regulation of apoptosis by inhibitors of apoptosis (IAPs). Cells. 2:163–187. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Schimmer AD: Inhibitor of apoptosis proteins: translating basic knowledge into clinical practice. Cancer Res. 64:7183–7190. 2004. View Article : Google Scholar : PubMed/NCBI

29 

Carter BZ, Kornblau SM, Tsao T, et al: Caspase-independent cell death in AML: caspase inhibition in vitro with pan-caspase inhibitors or in vivo by XIAP or Survivin does not affect cell survival or prognosis. Blood. 102:4179–4186. 2003. View Article : Google Scholar : PubMed/NCBI

30 

Abe S, Nishimoto Y, Isu K, Ishii T and Goto T; Japanese Musculoskeletal Oncology G. 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

31 

Anninga JK, Gelderblom H, Fiocco M, et al: Chemotherapeutic adjuvant treatment for osteosarcoma: where do we stand? Eur J Cancer. 47:2431–2445. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Chou AJ and Gorlick R: Chemotherapy resistance in osteosarcoma: current challenges and future directions. Expert Rev Anticancer Ther. 6:1075–1085. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Li J, Feng Q, Kim JM, et al: Human ovarian cancer and cisplatin resistance: possible role of inhibitor of apoptosis proteins. Endocrinology. 142:370–380. 2001.PubMed/NCBI

34 

Miyamoto M, Takano M, Iwaya K, et al: X-chromosome-linked inhibitor of apoptosis as a key factor for chemoresistance in clear cell carcinoma of the ovary. Br J Cancer. 110:2881–2886. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Cheng YJ, Jiang HS, Hsu SL, et al: XIAP-mediated protection of H460 lung cancer cells against cisplatin. Eur J Pharmacol. 627:75–84. 2010. View Article : Google Scholar

36 

Weiss RB: The anthracyclines: will we ever find a better doxorubicin? Semin Oncol. 19:670–686. 1992.PubMed/NCBI

37 

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

38 

Nobuto H, Sugita T, Kubo T, et al: Evaluation of systemic chemotherapy with magnetic liposomal doxorubicin and a dipole external electromagnet. Int J Cancer. 109:627–635. 2004. 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
Qu Y, Xia P, Zhang S, Pan S and Zhao J: Silencing XIAP suppresses osteosarcoma cell growth, and enhances the sensitivity of osteosarcoma cells to doxorubicin and cisplatin Retraction in /10.3892/or.2021.8213. Oncol Rep 33: 1177-1184, 2015
APA
Qu, Y., Xia, P., Zhang, S., Pan, S., & Zhao, J. (2015). Silencing XIAP suppresses osteosarcoma cell growth, and enhances the sensitivity of osteosarcoma cells to doxorubicin and cisplatin Retraction in /10.3892/or.2021.8213. Oncology Reports, 33, 1177-1184. https://doi.org/10.3892/or.2014.3698
MLA
Qu, Y., Xia, P., Zhang, S., Pan, S., Zhao, J."Silencing XIAP suppresses osteosarcoma cell growth, and enhances the sensitivity of osteosarcoma cells to doxorubicin and cisplatin Retraction in /10.3892/or.2021.8213". Oncology Reports 33.3 (2015): 1177-1184.
Chicago
Qu, Y., Xia, P., Zhang, S., Pan, S., Zhao, J."Silencing XIAP suppresses osteosarcoma cell growth, and enhances the sensitivity of osteosarcoma cells to doxorubicin and cisplatin Retraction in /10.3892/or.2021.8213". Oncology Reports 33, no. 3 (2015): 1177-1184. https://doi.org/10.3892/or.2014.3698