Expression and function of PIM kinases in osteosarcoma

Retraction in: /10.3892/ijo.2023.5577

  • Authors:
    • Shuai Mou
    • Guangbin Wang
    • Ding Ding
    • Dongdong Yu
    • Yi Pei
    • Songling Teng
    • Qin Fu
  • View Affiliations

  • Published online on: September 26, 2016     https://doi.org/10.3892/ijo.2016.3708
  • Pages: 2116-2126
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The provirus integrating site Moloney murine leukemia virus (PIM) family of serine/threonine protein kinases is composed of three members, PIM1, PIM2 and PIM3, which have been identified as oncoproteins in various malignancies. However, their role in osteosarcoma (OS) remains largely unknown. This study aimed to examine the expression patterns and the clinical significance of PIM kinases in human OS and their biological effects in human OS cell lines. Immunohistochemical staining was used to detect PIM kinases in archived pathologic material from 43 patients with primary OS; in addition, the effects of PIM knockdown and overexpression on the proliferation, migration and invasion of OS cell lines were determined. We observed that all three PIM kinases were frequently expressed in OS, but only PIM1 positive expression was associated with poorer prognosis regarding overall survival of OS patients. In addition, knockdown of PIM kinases notably inhibited OS cell proliferation, migration and invasiveness, whereas overexpression of PIM kinases resulted in increased OS cell growth and motility. This study suggests that PIM1 could be a valuable prognostic marker in patients with OS, and the biological functions of PIM kinase family in the osteosarcoma cell lines indicate that they could serve as potential therapeutic targets for OS.

Introduction

Osteosarcoma (OS) is the most common primary malignant bone tumor, and affects all ages, but especially two age-groups, namely adolescents and, to a lesser extent, the elderly (7th and 8th decades) (1,2). OS can arise in any bone but preferentially affects the metaphyses of long bones, such as the distal femur, proximal tibia, and proximal humerus (3). OS is highly aggressive with rapid growth, local invasion and early metastasis. In the past, OS was rarely curable, but it now has a 5-year overall survival rate of 70–80%, due to modern multimodal therapeutic strategies (4,5); however, in the last two decades, there has been no substantial improvement in the prognosis of OS patients with metastases or relapse, and pulmonary metastasis remains the main cause of death in OS patients (6,7). Therefore, it is essential to find new valuable pathogenic factors associated with OS, which can deepen the understanding of this neoplasm, help early diagnosis and improve prognosis.

Provirus integrating site Moloney murine leukemia virus (PIM) proteins are a family of short-lived, highly conserved serine/threonine kinases with three members: PIM1, PIM2 and PIM3. These kinases are highly homologous at the amino acid level, and many of their functions overlap with each other. PIM kinases have unique structural properties and are mainly regulated by transcription and translation (8,9). The Janus-activated kinase/signal transducers and activators of transcription (JAK/STAT) pathway, which is activated by various cytokines and hormones, plays an important role in regulating the expression of PIM proteins (8). Using transgenic mouse models, PIM kinases have been shown to have carcinogenic potential, especially when they collaborate with other oncogenes such as c-Myc (avian myelocytomatosis viral oncogene homolog), N-Myc, Bcl2 (B-cell CLL/lymphoma 2), GFI1 (growth factor independent 1 transcriptional repressor), and Frat1 (frequently rearranged in advanced T-cell lymphomas-1) in tumorigenesis (10). Moreover, expression of the three PIM kinases is found to be increased in various tumors, mainly in hematopoietic malignancies (11), and have recently been shown to be over-expressed in numerous solid tumors (e.g., prostate cancers, gastric carcinoma, pancreatic cancers, bladder cancer, Ewing's sarcoma and liposarcoma) (1218), their expression patterns correlate with the diagnosis and prognosis of the various pathologies (15,19).

PIM kinases mediate their oncogenic activity in tumor cells by modifying a variety of cellular substrates, and the responses are either isoform-specific or common to the three kinases (8). PIM kinases regulate cell cycle progression by directly phosphorylating p21 (cyclin-dependent kinase inhibitor 1A), p27 (cyclin-dependent kinase inhibitor 1B), Cdc25A (Cell division cycle 25A), and Cdc25C (Cell division cycle 25C) (2023). They can block cell apoptosis through the regulation of BAD (Bcl-2-associated agonist of cell death) (24,25), and can also affect cell motility by modifying NFATc1 (nuclear factor of activated T-cells, cytoplasmic 1), CXCR4 (C-X-C chemokine receptor type 4), GSK3B (glycogen synthase kinase 3β) and FOXP3 (forkhead box P3) (2628). Additionally, PIM kinases may be involved in early tumorigenesis, primarily by interaction of PIM1 with nuclear mitotic apparatus protein (NUMA), which has been shown to result in genomic instability (29).

Inhibition of PIM kinases has become a promising approach for cancer therapy (30,31). This is because they have unique structural features that enable the design of highly selective inhibitors (32), which do not cause serious side effects, supported by the fact that mice deficient for all three PIM family members are viable and fertile (33). Furthermore, inhibitors of PIM kinases can sensitize cancer cells to chemotherapy and may synergize with other antitumor agents (34).

However, the roles of PIM kinases in OS remain largely unknown. In this study, we examined all three PIM kinases in 43 paraffin-embedded OS samples and analyzed their expression patterns associated with clinicopathological features to elucidate their clinical significance. Furthermore, we performed in vitro experiments to evaluate their biological effects on OS cells.

Materials and methods

Clinical specimens

This study was approved by the Research Ethics Committee of Shengjing Hospital (IRB number, 2015PS203K). A total of 45 cases of paraffin-embedded OS tumor specimens were obtained from the department of pathology, Shengjing Hospital of China Medical University, between 2007 and 2015. Among them, one case of extra-skeletal OS and one case of sclerotic OS were excluded. Thus, 43 patients were included in this study. All 43 patients had primary OS, and all specimens were stained with hematoxylin and eosin (H&E) to verify the targeted tissues. Clinical stages of these OS patients were classified according to the guidelines of the American Joint Committee on Cancer. Clinical information was obtained by reviewing medical records. A total of 21 patients received adjuvant chemotherapy, four patients received postoperative radiation therapy, two patients received both adjuvant chemotherapy and radiation therapy, seven patients received no adjuvant treatment, and the adjuvant treatment for nine patients was unknown. Alkaline phosphatase results of five patients were not available. Follow-up was terminated April 25, 2016. The median time of follow-up was 37 months (6–86 months). Overall survival was defined as the interval between surgery and death or the last observation taken.

Immunohistochemistry and evaluation of staining

The immunohistochemical analysis steps were as follows: after heat-induced antigen retrieval, tissue sections were deparaffinized in xylene and rehydrated in graded alcohols. Antigens were retrieved by boiling the tissue sections in citrate buffer (pH 6.0) for 3 min, followed by successive rinses in phosphate-buffered saline (PBS). Endogenous peroxidase was inhibited by incubating the sections in 3% hydrogen peroxide for 30 min. Non-specific staining was blocked by incubation with 5–10% bovine serum albumin at room temperature for 20 min. Slides were incubated overnight with anti-PIM1 goat polyclonal antibody (1:250; Santa Cruz, Dallas, TX, USA), anti-PIM2 (1:400; GeneTex, Irvine, CA, USA) and anti-PIM3 (1:400; Abcam, Cambridge, MA, USA) rabbit polyclonal antibodies at 4°C. After washing with PBS, slides were incubated with the corresponding secondary antibody for 30 min at room temperature. Slides were then incubated with avidin horseradish peroxidase and the DAB (Diaminobenzidine, JSGB-BIO, Beijing, China) substrate. Finally, sections were counterstained with hematoxylin. PBS was used to replace anti-PIM antibodies in negative controls.

Immunostaining results were independently evaluated by two observers (Shuai Mou and Ding Ding) who were blinded to the clinicopathological features. The extent of immunoreactivity for PIM kinases was considered by both staining intensity and staining extent, as previously described (35); the staining intensity was scored as 0 (negative), 1 (mild), 2 (moderate), or 3 (strong). The percentage of positive cells in the whole tumor area was scored as 0 (0%), 1 (1–25%), 2 (26–50%), 3 (51–75%), or 4 (76–100%). The sum of the staining intensity and extent scores was used as the final staining scores (0–7), and we considered samples having a final score of ≥3 as positive.

Cell lines and cell culture

Three OS cell lines, including MG-63, U2OS and MNNG/HOS were obtained from the cell bank of Shanghai Biology Institute, Chinese Academy of Science. MG-63 and MNNG cells were cultured in DMEM (Hyclone, UT, USA) with 10% fetal bovine serum (Biological Industries) and 1% penicillin and streptomycin (Hyclone). U2OS cells were grown in RPMI 1640 medium (Hyclone) with 10% fetal bovine serum (Biological Industries, Haemek, Israel) and 1% penicillin and streptomycin (Hyclone).

Cell proliferation assay

Human OS cells (1×104/well) were plated in 96-well plates in DMEM containing 10% FBS at a density of 1×105 cells/ml and incubated for 24, 36, 48 or 60 h. At the end of the incubations, cellular proliferation was measured by the 3-(4,5)-(dimethylthiazol-zyl)-3,5-diphenyltetrazolium bromide (MTT) assay. MTT crystals were dissolved in 150 μl of DMSO. After a 4-h incubation at 37°C, the optical densities at 490 nm were measured using a Microplate Reader (Bio-Rad, CA, USA).

Cell transfection

To knock down the expression of PIM kinases, three siRNA, which specifically targeted PIM1, PIM2 and PIM3 (PIM1: 5′-GGUGUGUGGAGAUAUUCCUTT-3′, PIM2: 5′-CUGCUUGACUGGUUUGAGATT-3′ and PIM3: 5′-UCGUGCACCGCGACAUUAATT-3′), and negative control siRNA (siCont) were designed and synthesized by Genepharma (Shanghai, China). For overexpression, three pcDNA3.1/V5-HisC plasmids for expressing human PIM kinase genes were kindly provided by Professor Päivi M. Ojala (University of Helsinki, Helsinki, Finland), as previously described (36). Cells were transfected with siRNA or plasmids using Lipofectamine 2000 (Invitrogen, CA, USA), according to the manufacturer's protocol. For RNA extraction and western blot assays, cells were used at 24 or 48 h after transfection.

Cell migration and invasion assay

Assays were performed using modified Boyden chambers with polycarbonate nucleopore membranes. Pre-coated filters (6.5 mm in diameter, 8-μm pore size, Matrigel 100 μg/cm2) were rehydrated with 100 μl medium. Then, 1×105 cells in 100 μl serum-free DMEM supplemented with 0.1% bovine serum albumin were placed in the upper part of each chamber, whereas the lower compartments were filled with 600 μl DMEM containing 10% serum. After incubation for 24 h at 37°C, non-invaded cells were removed from the upper surface of the filter using a cotton swab, and the invaded cells on the lower surface of the filter were fixed, stained, photographed and counted under high-power magnification.

Real-time PCR analysis

Total RNA was extracted from the cells using TRIzol reagent (Invitrogen). The concentration and purity of the total-RNA were calculated by absorbance at 260 and 280 nm. Single-strand cDNA synthesis was performed using the PrimeScript RT Reagent kit (Takara Bio, Shiga, Japan). Real-time PCR was performed using SYBR Premix Ex Taq (Takara Bio) on an Mx 3000P real-time PCR system (Applied Biosystems, MA, USA) using the following conditions: 50 cycles of 95°C for 10 sec and 60°C for 30 sec. All the reactions were repeated at least three times. PCR primers were designed using Primer 5.0 software and the sequences are listed in Table I.

Table I

Primer sequences for detection of mRNA expression.

Table I

Primer sequences for detection of mRNA expression.

GenesPrimersPrimer sequence (5′-3′)
PIM1Forward GCTCGGTCTACTCAGGCATC
Reverse GCTCCCCTTTCCGTGATGAA
PIM2Forward CGTGGAGTTGTCCATCGTG
Reverse AAGGGAATGTCCCCACACAC
PIM3Forward GTACAGTCTGCTTGTGGGCT
Reverse GAAAGAACCCCCATCTGCGA
Cyclin D1Forward CCGAGGAGCTGCTGCAAATGGAGCT
Reverse TGAAATCGTGCGGGGTCATTGCGGC
MMP2Forward CGCATCTGGGGCTTTAAACAT
Reverse TCAGCACAAACAGGTTGCAG
GAPDHForward CTCTGCTCCTCCTGTTCGAC
Reverse GCGCCCAATACGACCAAATC
Western blot analysis

Total protein was extracted in RIPA lysis buffer (Beyotime, Shanghai, China), quantified by the BCA protein assay (Beyotime), and equal amounts of protein were separated by SDS-PAGE (sodium dodecyl sulfate-polyacrylamide gel electrophoresis) and transferred to PVDF (polyvinylidene fluoride) membranes (Millipore, MA, USA). Membranes were blocked with 5% nonfat milk in PBS for 1 h at room temperature and then incubated with PIM1, PIM2, PIM3, Cyclin D1 (Santa Cruz) and MMP2 (Santa Cruz) antibodies overnight at 4°C. After extensive washing, membranes were re-probed with horseradish peroxidase-conjugated secondary antibodies (Boster Wuhan, China). Membranes were developed using ECL or ECL plus (Thermo Fisher Scientific, MA, USA), according to the manufacturer's instructions.

Statistical analysis

All statistical analyses were performed using SPSS19.0. The Chi-square test was applied to analyze the correlation between the expression of PIM kinases and the clinicopathological features. Immunohistochemistry (IHC) scores of PIM kinases was calculated by multiplying the percentage and intensity scores, and the cut-off point was considered by a final score of ≥3 as positive. The Kaplan-Meier method was used for survival analysis, and differences in survival were estimated using the log-rank test. Both univariate and multivariate analyses were carried out to identify independent prognostic factors for survival using the Cox proportional hazards model. The two-tailed, unpaired Student's t-test was used to compare differences between experimental and control groups in the in vitro assays. Results are shown as mean values with 95% confidence intervals. Error bars represent standard deviations. A value of P<0.05 was considered statistically significant.

Results

All three PIM kinases are expressed in the majority of OS tumor samples

To explore whether PIM kinases were expressed in OS, we performed immunohistochemistry to detect the protein expression of the three kinase proteins in 43 paraffin-embedded OS samples. In the OS tissues, expression of PIM1, PIM2 and PIM3 was observed as a brown-yellow color, localized both in the nuclei and cytoplasm. The expression intensities of PIM kinases varied from negative- to-weak, medium and strong staining (Fig. 1). According to our IHC score (see Materials and methods), 76.7% (33/43), 74.4% (32/43) and 69.7% (30/43) of cases were PIM1-positive, PIM2-positive and PIM3-positive, respectively (Fig. 2C). Of the 43 cases, 21 cases expressed all three PIM kinases, and 3 cases did not express the PIM kinases. Of the remaining cases, 11 cases were positive for either PIM1 alone or co-expressed with PIM2 or PIM3, 11 cases were positive for PIM2 alone or in combination with PIM1 or PIM3, and 9 cases were positive for PIM3 alone or together with PIM1 or PIM2 (Fig. 2D).

Investigation of the expression patterns of PIM kinases and the clinical significance in OS

In order to elucidate the clinical significance of PIM kinase expression, we evaluated the association between the positive expression of the three PIM kinases and the clinicopathological characteristics, including sex, age, location, pathologic subtype, tumor size, tumor stage and alkaline phosphatase (ALP). However, there was no significant correlation with these clinicopathological parameters and positive expression of the three PIM kinases (Table II). We also observed the association of the expression of the three PIM kinases and the clinical outcome of OS patients. Kaplan-Meier analysis showed that only positive expression of PIM1 showed a poorer prognosis than patients with negative expression of PIM1 (log-rank test, P=0.0432 in overall survival), but there were no significant differences between positive and negative expression of PIM2 and PIM3 (Fig. 2A). Kaplan-Meier analysis by AJCC tumor stage is shown in Fig. 2E. We then performed univariate and multivariate Cox regression survival analysis, with the variables including sex, age, pathologic subtype, tumor size, tumor stage, and positive expression of ALP, PIM1, PIM2 and PIM3. In the univariate analysis, only the tumor stage of OS was a significant predictor of poor prognosis, while the other indicators, including PIM1, PIM2 and PIM3 expression were not good predictors for overall survival of OS patients, according to both univariate and multivariate analysis (data not shown).

Table II

Correlation between the Pim kinase expression and different clinicopathological features in 43 osteosarcoma patients.a

Table II

Correlation between the Pim kinase expression and different clinicopathological features in 43 osteosarcoma patients.a

PIM1PIM2PIM3



CharacteristicsNo. of patientsPositiveP-valuePositiveP-valuePositiveP-value
Gender
 Male16130.590110.512100.424
 Female27202120
Age (years)
 <3029210.333220.755220.210
 ≥301412108
Site of primary disease
 Femur17130.957130.93190.097
 Tibia10877
 Other16121214
AJCC Stage
 I–II35250.165270.392240.721
 III–IV8856
Metastasis
 Absence35250.165270.392240.721
 Presence8856
Tumor size, cm
 ≤829230.566210.665200.869
 >814101110
Histologic type
 Conventional37290.529260.312240.155
 Special6466
ALP
 Normal range24180.809170.601180.482
 Beyond normal range1410119

a ALP, alkaline phosphatase.

{ label (or @symbol) needed for fn[@id='tfn2-ijo-49-05-2116'] } P<0.05 was considered to be statistically significant.

Subcellular distribution of PIM kinases was found to be related to their biological function and disease stage (37,38), we therefore investigated the nuclear and cytoplasmic expression patterns of the three PIM kinases in our OS samples (Fig. 2B). Nuclear staining of positive expression of PIM1, PIM2 and PIM3 was 36.4% (12/33), 25% (8/32) and 43.3% (13/30), respectively (Fig. 2C). However, we observed that neither nuclear nor cytoplasmic expression of the three PIM kinases was significantly correlated with OS tumor stage or prognosis (data not shown).

PIM kinases promote proliferation of OS cells through cyclin D1 regulation

We then examined the biological effects of PIM kinases on OS cells. Firstly, we measured the basal protein levels of the three PIM kinases in the three OS cell lines by western blotting. We found that the protein levels of PIM1, PIM2 and PIM3 were relatively higher in MG-63 cells than those in MNNG/HOS cells or U2OS cells (Fig. 3A). Thus, we chose the MG-63 cell line for use in RNA interference studies and the U2OS cell line for use in plasmid overexpression studies. Downregulation and increased expression of PIM1, PIM2 and PIM3 was verified at both the mRNA and protein expression levels (Fig. 4). Cell proliferation rates were measured using the MTT assay and results were determined at 0, 12, 24, 36 and 48 h post-transfection. Results showed that PIM1 knockdown inhibited MG-63 cell growth to 69 and 65% of the controls at 36 and 48 h, respectively; PIM2 knockdown inhibited MG-63 cell growth to 64 and 61% of the controls at 36 and 48 h, respectively, and PIM3 knockdown inhibited MG-63 cell growth to 55 and 54% of the controls at 36 and 48 h, respectively (Fig. 3B). Conversely, PIM1, PIM2 and PIM3 overexpression in U2OS cells exhibited a significantly higher rate of proliferation at 48 h post-transfection (Fig. 3D).

It has been recently reported that the association between the three PIM kinases can mediate tumor cell proliferation and cyclin D1 expression (3941). To further investigate these underlying mechanisms, we examined the protein and mRNA levels of cyclin D1 after PIM knockdown or overexpression. PIM1 depletion decreased cyclin D1 expression in MG-63 cells (Fig. 3C); accordingly PIM1 overexpression increased cyclin D1 expression in U2OS cells. Similar results were also observed in the case of PIM2 and PIM3 (Fig. 3E). Taken together, these results demonstrated that all three PIM kinases affected the proliferation of OS cells, possibly via cyclin D1 regulation.

PIM kinases promote the migratory and invasive potential of OS cells by modulating MMP2

We also investigated the effects of depletion of PIM kinases on OS cell motility; cell migration and invasion were examined by Transwell assays. Results showed that, compared with the control cells, PIM1, PIM2 and PIM3 knockdown significantly inhibited MG-63 cell migration by 69, 63 and 70%, respectively (Fig. 5A and B). Moreover, deficiency of the three kinases markedly inhibited the invasiveness of MG-63 cells by 59, 85 and 83%, respectively (Fig. 5A and B). Accordingly, upregulation of PIM1, PIM2 and PIM3 expression promoted the migration and invasion of U2OS cells (Fig. 5D and E).

It is well known that matrix metalloproteinases 2 (MMP2) can facilitate enhanced tumor cell invasion and metastasis potential (42). Therefore, we investigated the expression levels of MMP2 in PIM kinase-knockdown MG63 cells and in PIM kinase-overexpressing U2OS cells by western blotting and real-time PCR analysis. Results revealed that MMP-2 protein and mRNA levels decreased in PIM kinase-knockdown cells compared with the negative controls, whereas the cells over-expressing PIM kinase showed an increase in MMP2 levels (Fig. 5C and F). These results suggest that all three PIM kinases can enhance the motility of OS cells, at least in part, by modulating MMP-2.

Discussion

PIM kinases have been identified as proto-oncogenes implicated in tumorigenesis, and their expression and functions have been demonstrated in various tumors (8,10). However, their role in OS has not been clearly defined. In this study, we found that all three PIM kinases were frequently expressed in OS tissues. We also showed that positive expression of PIM1 was associated with poorer prognosis for overall survival of OS patients, while the positive expression of PIM2 and PIM3 was not found to predict prognosis. The subcellular distributions of the three PIM kinases were not correlated with the disease stage or clinical outcome. In addition, our in vitro study demonstrated that knockdown of PIM kinases effectively inhibited OS cell proliferation, migration and invasion, whereas overexpression of PIM kinases resulted in increased OS cell growth and motility.

The literature on expression of PIM kinases in OS is extremely limited, Lu et al reported that the PIM1 gene locus (6p12–21) is a common amplification event in OS (43). More recently, during the course of this work, Liao et al reported five out of seven OS tissue samples had high expression of PIM1 protein, and 77.2% (88/144) OS specimens from 70 patients exhibited PIM1 immunostaining on the OS cell nucleus (44). Both of these studies indicated that PIM1 is expressed in OS and may therefore play an important role. Nevertheless, in this present study, we demonstrated that all three PIM kinases were expressed in a majority of OS samples, but only positive expression of PIM1 was associated with poorer prognosis for overall survival of OS patients. The prognostic value of PIM1 in OS patients was also reported in the previous study (44). However, our univariate and multivariate Cox regression analyses indicated the three PIM kinases are not good predictors for overall survival of OS patients. This conflict may be due to our small sample size, as OS is relative rare (1,45,46). Therefore, an additional study with a larger number of cases is needed to confirm the clinical significance of the expression of the three PIM kinases, particularly PIM2 and PIM3, in OS patients.

Some studies have reported that nuclear accumulation of PIM1 is necessary for its biologic effects and may correlate with tumor progression and disease stage (37,38). In the previous study, Liao et al reported that the cellular 34 kDa PIM1 isoform staining in OS tissues was predominantly nuclear (44). However, in our study, nuclear PIM1 localization was not dominant in our OS samples, and PIM1 nuclear staining accounted for 36.4% in the PIM1 positive group. Furthermore, we observed all three PIM kinases are distributed to both the nucleus and cytoplasm, and their expression patterns are uncorrelated with OS tumor stage or prognosis (data not shown). Notably, the shuttling of subcellular distribution of PIM kinases between the nucleus and the cytoplasm is complicated and not well understood (38). Cytoplasmic sequestration of PIM1 may also be relevant and can promote cancer cell survival (47). Thus, further investigations are needed to clarify the functions of different subcellular distributions of PIM kinases and the underlying mechanisms, particularly in OS.

PIM kinases are associated with cell proliferation as they prevent apoptosis and promote cell survival (8). Consistently, our in vitro study revealed that all three PIM kinases can promote OS cell proliferation, while silencing PIM kinases can decrease cell proliferation rates. The underlying mechanism may be related to the association of PIM kinase with cyclin D1. Previous studies have found that when PIM kinases were inhibited, the expression of cyclin D1 was reduced and the change in cyclin D1 expression could influence cell cycle progression and cancer cell proliferation (3941). Furthermore, we found that knock-down of any of the three PIM kinases resulted in decreased cyclin D1 expression in OS cells, whereas overexpression of PIM kinases in OS cells increased cyclin D1 expression. Together, these results indicate that all three PIM kinases may have a role in the growth of OS cells.

We also investigated the association of the three PIM kinases with the motility of OS cells. Our results showed that OS cells in which the three PIM kinases had been knocked down had lower migration and invasion. In contrast, overexpression of any PIM kinase family member had the opposite effect, displaying increased migration and invasion of OS cells. These results suggest that PIM kinases may affect OS metastasis. In agreement with this, previous studies have suggested a role for PIM kinases in cell motility and metastasis, for example, PIM1 was able to promote migration in leukemia and prostate cancer cells (48), PIM2 was shown to induce human liver cell malignant transformation and increase the migration rate (49), and PIM3 can promote ovarian cancer cell migration (50). In addition, absence of PIM kinases blocks the process of bone invasion induced by the 3-methylcholanthrene-induced sarcoma in vivo (51). However, the regulatory mechanisms involved in controlling cell motility by the three PIM kinases are not well known; in this study, we found that PIM kinase depletion downregulated MMP2 expression, whereas cells overexpressing PIM kinase showed an increase in MMP2 levels, suggesting that MMP2 may play a role in regulating OS cell motility by PIM kinases.

Recently, multiple substrates of PIM kinases that promoted cancer cell metastasis were identified, including CXCR4, NFATc1, GSK3B and FOXP3 (26). Of these, CXCR4 was the first to be confirmed and the most studied. PIM1, but not PIM2 and PIM3, was shown to regulate homing and migration of leukemia cells via modulation of stromal-derived factor-1alpha (SDF-1/CXCL12)-CXCR4 signaling (27,52). Subsequent research has demonstrated that PIM1 and PIM3, but not PIM2, can phosphorylate CXCR4 and promote metastatic properties of prostate cancer. Importantly, overexpressing PIM1 or PIM3 may take advantage of the CXCL12/CXCR4 chemokine pathway and stimulate the formation of lung metastases from orthotopically-induced prostate tumors in mice (53). It is known that CXCR4 expression plays an important role in the pulmonary metastatic process in OS (54,55). Therefore, the association between PIM kinases and CXCR4 suggests that PIM kinases may also be involved in the lung metastasis of OS, but this remains to be elucidated. Unfortunately, in this study, we were unable to verify the expression of PIM kinases in OS metastasis, especially lung metastasis, which are the main cause of death in OS patients. Future studies are needed to substantiate this intriguing possibility.

In conclusion, in this study, we show for the first time, that all three PIM kinases are frequently expressed in OS. Although the expression patterns and the functions of PIM kinases in OS are not completely understood, we found that positive expression of PIM1 was associated with poorer prognosis, which supports the use of PIM1 as a potential prognostic biomarker of OS. Moreover, in our in vitro study, we found that all three PIM kinases can significantly affect OS cell proliferation, migration and invasion, which suggests that PIM kinases have multiple biological functions on OS cells, and could therefore serve as potential therapeutic targets in OS. However, more studies are needed to elucidate the oncogenic roles of PIM kinases and their clinical significance in OS.

Acknowledgements

This study was supported by Outstanding Scientific Fund of Shengjing Hospital and two grants from the National Natural Science Foundation of China (81370981 and 81300714). We acknowledge Professor Päivi M. Ojala (University of Helsinki, Helsinki, Finland) for providing us with the PIM pcDNA3.1/V5-HisC plasmids.

References

1 

Mirabello L, Troisi RJ and Savage SA: Osteosarcoma incidence and survival rates from 1973 to 2004: Data from the Surveillance, Epidemiology, and End Results Program. Cancer. 115:1531–1543. 2009. View Article : Google Scholar : PubMed/NCBI

2 

Jawad MU, Cheung MC, Clarke J, Koniaris LG and Scully SP: Osteosarcoma: Improvement in survival limited to high-grade patients only. J Cancer Res Clin Oncol. 137:597–607. 2011. View Article : Google Scholar

3 

Bielack SS, Kempf-Bielack B, Delling G, Exner GU, Flege S, Helmke K, Kotz R, Salzer-Kuntschik M, Werner M, Winkelmann W, et al: Prognostic factors in high-grade osteosarcoma of the extremities or trunk: An analysis of 1,702 patients treated on neoadjuvant cooperative osteosarcoma study group protocols. J Clin Oncol. 20:776–790. 2002. View Article : Google Scholar : PubMed/NCBI

4 

Hegyi M, Semsei AF, Jakab Z, Antal I, Kiss J, Szendroi M, Csoka M and Kovacs G: Good prognosis of localized osteosarcoma in young patients treated with limb-salvage surgery and chemotherapy. Pediatr Blood Cancer. 57:415–422. 2011. View Article : Google Scholar : PubMed/NCBI

5 

Anderson ME: Update on survival in osteosarcoma. Orthop Clin North Am. 47:283–292. 2016. View Article : Google Scholar

6 

Zwaga T, Bovee JV and Kroon HM: Osteosarcoma of the femur with skip, lymph node, and lung metastases. Radiographics: A review publication of the Radiological Society of North America Inc. 28:277–283. 2008. View Article : Google Scholar

7 

Miller BJ, Cram P, Lynch CF and Buckwalter JA: Risk factors for metastatic disease at presentation with osteosarcoma: An analysis of the SEER database. J Bone Joint Surg Am. 95:e892013. View Article : Google Scholar : PubMed/NCBI

8 

Narlik-Grassow M, Blanco-Aparicio C and Carnero A: The PIM family of serine/threonine kinases in cancer. Med Res Rev. 34:136–159. 2014. View Article : Google Scholar

9 

Nawijn MC, Alendar A and Berns A: For better or for worse: The role of Pim oncogenes in tumorigenesis. Nat Rev Cancer. 11:23–34. 2011. View Article : Google Scholar

10 

Aguirre E, Renner O, Narlik-Grassow M and Blanco-Aparicio C: Genetic modeling of PIM proteins in cancer: Proviral tagging and cooperation with oncogenes, tumor suppressor genes, and carcinogens. Front Oncol. 4:1092014. View Article : Google Scholar : PubMed/NCBI

11 

Alvarado Y, Giles FJ and Swords RT: The PIM kinases in hematological cancers. Expert Rev Hematol. 5:81–96. 2012. View Article : Google Scholar : PubMed/NCBI

12 

Guo S, Mao X, Chen J, Huang B, Jin C, Xu Z and Qiu S: Overexpression of Pim-1 in bladder cancer. J Exp Clin Cancer Res. 29:1612010. View Article : Google Scholar : PubMed/NCBI

13 

Reiser-Erkan C, Erkan M, Pan Z, Bekasi S, Giese NA, Streit S, Michalski CW, Friess H and Kleeff J: Hypoxia-inducible proto-oncogene Pim-1 is a prognostic marker in pancreatic ductal adenocarcinoma. Cancer Biol Ther. 7:1352–1359. 2008. View Article : Google Scholar : PubMed/NCBI

14 

Brault L, Gasser C, Bracher F, Huber K, Knapp S and Schwaller J: PIM serine/threonine kinases in the pathogenesis and therapy of hematologic malignancies and solid cancers. Haematologica. 95:1004–1015. 2010. View Article : Google Scholar : PubMed/NCBI

15 

Nga ME, Swe NN, Chen KT, Shen L, Lilly MB, Chan SP, Salto-Tellez M and Das K: PIM-1 kinase expression in adipocytic neoplasms: Diagnostic and biological implications. Int J Exp Pathol. 91:34–43. 2010. View Article : Google Scholar

16 

Holder SL and Abdulkadir SA: PIM1 kinase as a target in prostate cancer: Roles in tumorigenesis, castration resistance, and docetaxel resistance. Curr Cancer Drug Targets. 14:105–114. 2014. View Article : Google Scholar

17 

Warnecke-Eberz U, Bollschweiler E, Drebber U, Metzger R, Baldus SE, Hölscher AH and Mönig S: Prognostic impact of protein overexpression of the proto-oncogene PIM-1 in gastric cancer. Anticancer Res. 29:4451–4455. 2009.PubMed/NCBI

18 

Albertson DJ, Schmidt RL, Bearss JJ, Tripp SR, Bearss DJ and Liu T: Patterns and significance of PIM kinases in urothelial carcinoma. Appl Immunohistochem Mol Morphol. 23:717–723. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Peng YH, Li JJ, Xie FW, Chen JF, Yu YH, Ouyang XN and Liang HJ: Expression of pim-1 in tumors, tumor stroma and tumor-adjacent mucosa co-determines the prognosis of colon cancer patients. PLoS One. 8:e766932013. View Article : Google Scholar : PubMed/NCBI

20 

Mochizuki T, Kitanaka C, Noguchi K, Muramatsu T, Asai A and Kuchino Y: Physical and functional interactions between Pim-1 kinase and Cdc25A phosphatase. Implications for the Pim-1-mediated activation of the c-Myc signaling pathway. J Biol Chem. 274:18659–18666. 1999. View Article : Google Scholar : PubMed/NCBI

21 

Morishita D, Katayama R, Sekimizu K, Tsuruo T and Fujita N: Pim kinases promote cell cycle progression by phosphorylating and down-regulating p27Kip1 at the transcriptional and posttranscriptional levels. Cancer Res. 68:5076–5085. 2008. View Article : Google Scholar : PubMed/NCBI

22 

Zhang Y, Wang Z and Magnuson NS: Pim-1 kinase-dependent phosphorylation of p21Cip1/WAF1 regulates its stability and cellular localization in H1299 cells. Mol Cancer Res. 5:909–922. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Bachmann M, Kosan C, Xing PX, Montenarh M, Hoffmann I and Möröy T: The oncogenic serine/threonine kinase Pim-1 directly phosphorylates and activates the G2/M specific phosphatase Cdc25C. Int J Biochem Cell Biol. 38:430–443. 2006. View Article : Google Scholar

24 

Yan B, Zemskova M, Holder S, Chin V, Kraft A, Koskinen PJ and Lilly M: The PIM-2 kinase phosphorylates BAD on serine 112 and reverses BAD-induced cell death. J Biol Chem. 278:45358–45367. 2003. View Article : Google Scholar : PubMed/NCBI

25 

Aho TL, Sandholm J, Peltola KJ, Mankonen HP, Lilly M and Koskinen PJ: Pim-1 kinase promotes inactivation of the pro-apoptotic Bad protein by phosphorylating it on the Ser112 gatekeeper site. FEBS Lett. 571:43–49. 2004. View Article : Google Scholar : PubMed/NCBI

26 

Santio NM, Salmela M, Arola H, Eerola SK, Heino J, Rainio EM and Koskinen PJ: The PIM1 kinase promotes prostate cancer cell migration and adhesion via multiple signalling pathways. Exp Cell Res. 342:113–124. 2016. View Article : Google Scholar : PubMed/NCBI

27 

Grundler R, Brault L, Gasser C, Bullock AN, Dechow T, Woetzel S, Pogacic V, Villa A, Ehret S, Berridge G, et al: Dissection of PIM serine/threonine kinases in FLT3-ITD-induced leukemogenesis reveals PIM1 as regulator of CXCL12-CXCR4-mediated homing and migration. J Exp Med. 206:1957–1970. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Rainio EM, Sandholm J and Koskinen PJ: Cutting edge: Transcriptional activity of NFATc1 is enhanced by the Pim-1 kinase. J Immunol. 168:1524–1527. 2002. View Article : Google Scholar : PubMed/NCBI

29 

Bhattacharya N, Wang Z, Davitt C, McKenzie IF, Xing PX and Magnuson NS: Pim-1 associates with protein complexes necessary for mitosis. Chromosoma. 111:80–95. 2002. View Article : Google Scholar : PubMed/NCBI

30 

Magnuson NS, Wang Z, Ding G and Reeves R: Why target PIM1 for cancer diagnosis and treatment? Future Oncol. 6:1461–1478. 2010. View Article : Google Scholar : PubMed/NCBI

31 

Arunesh GM, Shanthi E, Krishna MH, Sooriya Kumar J and Viswanadhan VN: Small molecule inhibitors of PIM1 kinase: July 2009 to February 2013 patent update. Expert Opin Ther Pat. 24:5–17. 2014. View Article : Google Scholar

32 

Qian KC, Wang L, Hickey ER, Studts J, Barringer K, Peng C, Kronkaitis A, Li J, White A, Mische S, et al: Structural basis of constitutive activity and a unique nucleotide binding mode of human Pim-1 kinase. J Biol Chem. 280:6130–6137. 2005. View Article : Google Scholar

33 

Mikkers H, Nawijn M, Allen J, Brouwers C, Verhoeven E, Jonkers J and Berns A: Mice deficient for all PIM kinases display reduced body size and impaired responses to hematopoietic growth factors. Mol Cell Biol. 24:6104–6115. 2004. View Article : Google Scholar : PubMed/NCBI

34 

Xie Y, Xu K, Linn DE, Yang X, Guo Z, Shimelis H, Nakanishi T, Ross DD, Chen H, Fazli L, et al: The 44-kDa Pim-1 kinase phosphorylates BCRP/ABCG2 and thereby promotes its multimerization and drug-resistant activity in human prostate cancer cells. J Biol Chem. 283:3349–3356. 2008. View Article : Google Scholar

35 

Zemskova MY, Song JH, Cen B, Cerda-Infante J, Montecinos VP and Kraft AS: Regulation of prostate stromal fibroblasts by the PIM1 protein kinase. Cell Signal. 27:135–146. 2015. View Article : Google Scholar :

36 

Cheng F, Weidner-Glunde M, Varjosalo M, Rainio EM, Lehtonen A, Schulz TF, Koskinen PJ, Taipale J and Ojala PM: KSHV reactivation from latency requires Pim-1 and Pim-3 kinases to inactivate the latency-associated nuclear antigen LANA. PLoS Pathog. 5:e10003242009. View Article : Google Scholar : PubMed/NCBI

37 

Ionov Y, Le X, Tunquist BJ, Sweetenham J, Sachs T, Ryder J, Johnson T, Lilly MB and Kraft AS: Pim-1 protein kinase is nuclear in Burkitt's lymphoma: Nuclear localization is necessary for its biologic effects. Anticancer Res. 23A:167–178. 2003.

38 

Brault L, Menter T, Obermann EC, Knapp S, Thommen S, Schwaller J and Tzankov A: PIM kinases are progression markers and emerging therapeutic targets in diffuse large B-cell lymphoma. Br J Cancer. 107:491–500. 2012. View Article : Google Scholar : PubMed/NCBI

39 

Zhu X, Xu JJ, Hu SS, Feng JG, Jiang LH, Hou XX, Cao J, Han J, Ling ZQ and Ge MH: Pim-1 acts as an oncogene in human salivary gland adenoid cystic carcinoma. J Exp Clin Cancer Res. 33:1142014. View Article : Google Scholar

40 

Yang Q, Chen LS, Neelapu SS, Miranda RN, Medeiros LJ and Gandhi V: Transcription and translation are primary targets of Pim kinase inhibitor SGI-1776 in mantle cell lymphoma. Blood. 120:3491–3500. 2012. View Article : Google Scholar : PubMed/NCBI

41 

Quan J, Zhou L and Qu J: Knockdown of Pim-3 suppresses the tumorigenicity of glioblastoma by regulating cell cycle and apoptosis. Cell Mol Biol. 61:42–50. 2015.PubMed/NCBI

42 

Wang Z, Cao CJ, Huang LL, Ke ZF, Luo CJ, Lin ZW, Wang F, Zhang YQ and Wang LT: EFEMP1 promotes the migration and invasion of osteosarcoma via MMP-2 with induction by AEG-1 via NF-κB signaling pathway. Oncotarget. 6:14191–14208. 2015. View Article : Google Scholar : PubMed/NCBI

43 

Lu XY, Lu Y, Zhao YJ, Jaeweon K, Kang J, Xiao-Nan L, Ge G, Meyer R, Perlaky L, Hicks J, et al: Cell cycle regulator gene CDC5L, a potential target for 6p12-p21 amplicon in osteosarcoma. Mol Cancer Res. 6:937–946. 2008. View Article : Google Scholar : PubMed/NCBI

44 

Liao Y, Feng Y, Shen J, Gao Y, Cote G, Choy E, Harmon D, Mankin H1, Hornicek F1 and Duan Z1: Clinical and biological significance of PIM1 kinase in osteosarcoma. J Orthop Res. 34:1185–1194. 2016. View Article : Google Scholar

45 

Gurney J, Swensen A and Bulterys M: Malignant bone tumors. Cancer Incidence and Survival Among Children and Adolescents: United States SEER Program 1975–1995. National Cancer Institute, SEER Program; Ries L, Smith M, Gurney J, Linet M, Tamra T, Young J and Bunin G: Bethesda, MD: NIH Pub. No. 99-4649; pp. 99–110. 1999

46 

Wagle S, Park SH, Kim KM, Moon YJ, Bae JS, Kwon KS, Park HS, Lee H, Moon WS, Kim JR, et al: DBC1/CCAR2 is involved in the stabilization of androgen receptor and the progression of osteosarcoma. Sci Rep. 5:131442015. View Article : Google Scholar : PubMed/NCBI

47 

Kim JH, Kim WS, Yun Y and Park C: Epstein-Barr virus latent membrane protein 1 increases chemo-resistance of cancer cells via cytoplasmic sequestration of Pim-1. Cell Signal. 22:1858–1863. 2010. View Article : Google Scholar : PubMed/NCBI

48 

Santio NM, Vahakoski RL, Rainio EM, Sandholm JA, Virtanen SS, Prudhomme M, Anizon F, Moreau P and Koskinen PJ: Pim-selective inhibitor DHPCC-9 reveals Pim kinases as potent stimulators of cancer cell migration and invasion. Mol Cancer. 9:2792010. View Article : Google Scholar : PubMed/NCBI

49 

Ren K, Duan W, Shi Y, Li B, Liu Z and Gong J: Ectopic over-expression of oncogene Pim-2 induce malignant transformation of nontumorous human liver cell line L02. J Korean Med Sci. 25:1017–1023. 2010. View Article : Google Scholar : PubMed/NCBI

50 

Zhuang H, Zhao MY, Hei KW, Yang BC, Sun L, Du X and Li YM: Aberrant expression of PIM-3 promotes proliferation and migration of ovarian cancer cells. Asian Pac J Cancer Prev. 16:3325–3331. 2015. View Article : Google Scholar : PubMed/NCBI

51 

Narlik-Grassow M, Blanco-Aparicio C, Cecilia Y, Peregrina S, Garcia-Serelde B, Muñoz-Galvan S, Cañamero M and Carnero A: The essential role of PIM kinases in sarcoma growth and bone invasion. Carcinogenesis. 33:1479–1486. 2012. View Article : Google Scholar : PubMed/NCBI

52 

Decker S, Finter J, Forde AJ, Kissel S, Schwaller J, Mack TS, Kuhn A, Gray N, Follo M, Jumaa H, et al: PIM kinases are essential for chronic lymphocytic leukemia cell survival (PIM2/3) and CXCR4-mediated microenvironmental interactions (PIM1). Mol Cancer Ther. 13:1231–1245. 2014. View Article : Google Scholar : PubMed/NCBI

53 

Santio NM, Eerola SK, Paatero I, Yli-Kauhaluoma J, Anizon F, Moreau P, Tuomela J, Härkönen P and Koskinen PJ: Pim kinases promote migration and metastatic growth of prostate cancer xenografts. PLoS One. 10:e01303402015. View Article : Google Scholar : PubMed/NCBI

54 

Oda Y, Yamamoto H, Tamiya S, Matsuda S, Tanaka K, Yokoyama R, Iwamoto Y and Tsuneyoshi M: CXCR4 and VEGF expression in the primary site and the metastatic site of human osteosarcoma: analysis within a group of patients, all of whom developed lung metastasis. Mod Pathol. 19:738–745. 2006. View Article : Google Scholar : PubMed/NCBI

55 

Laverdiere C, Hoang BH, Yang R, Sowers R, Qin J, Meyers PA, Huvos AG, Healey JH and Gorlick R: Messenger RNA expression levels of CXCR4 correlate with metastatic behavior and outcome in patients with osteosarcoma. Clin Cancer Res. 11:2561–2567. 2005. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

November-2016
Volume 49 Issue 5

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Mou S, Wang G, Ding D, Yu D, Pei Y, Teng S and Fu Q: Expression and function of PIM kinases in osteosarcoma Retraction in /10.3892/ijo.2023.5577. Int J Oncol 49: 2116-2126, 2016
APA
Mou, S., Wang, G., Ding, D., Yu, D., Pei, Y., Teng, S., & Fu, Q. (2016). Expression and function of PIM kinases in osteosarcoma Retraction in /10.3892/ijo.2023.5577. International Journal of Oncology, 49, 2116-2126. https://doi.org/10.3892/ijo.2016.3708
MLA
Mou, S., Wang, G., Ding, D., Yu, D., Pei, Y., Teng, S., Fu, Q."Expression and function of PIM kinases in osteosarcoma Retraction in /10.3892/ijo.2023.5577". International Journal of Oncology 49.5 (2016): 2116-2126.
Chicago
Mou, S., Wang, G., Ding, D., Yu, D., Pei, Y., Teng, S., Fu, Q."Expression and function of PIM kinases in osteosarcoma Retraction in /10.3892/ijo.2023.5577". International Journal of Oncology 49, no. 5 (2016): 2116-2126. https://doi.org/10.3892/ijo.2016.3708