Prostate cancer cell‑derived spondin 2 boosts osteogenic factor levels in osteoblasts via the PI3K/AKT/mTOR pathway

  • Authors:
    • Hongbo Wang
    • Ming Zhang
    • Wencheng Lu
    • Chao Yuan
  • View Affiliations

  • Published online on: December 9, 2022     https://doi.org/10.3892/or.2022.8460
  • Article Number: 23
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Prostate cancer is the leading cause of cancer death among men worldwide. Bone metastasis is one of the main problems arising from prostate cancer. Spondin 2 is a diagnostic marker specific for prostate cancer; however, the role of spondin 2 in prostate cancer‑driven osteogenesis remains unclear. The present study was carried out to explore the role of spondin 2 on prostate cancer cell‑induced osteogenesis. In the present study, the expression of spondin 2 was analyzed in prostate cancer samples obtained from Gene Expression Omnibus. The supernatant of prostate cancer cells was used to treat the osteoblast precursor MC3T3‑E1 cell line to determine the effect of spondin 2 on osteoblasts. The effect of spondin 2 on osteogenic factor production was also examined after neutralization with a spondin 2 antibody in vitro via reverse transcription‑quantitative PCR. Furthermore, the effect of spondin 2 on the PI3K/AKT/mTOR pathway was assessed using a patient dataset from The Cancer Genome Atlas and in vitro via western blot analysis. In addition, an inhibitor of spondin 2 receptor (ATN‑161) was used to explore the inhibition effect of spondin 2 receptor in MC3T3‑E1 cells. The results showed that spondin 2 promoted Osterix and Runx2 expression in osteoblasts, and this process was tightly associated with the activation of the PI3K/AKT/mTOR pathway. Moreover, it was demonstrated that the function of spondin 2 on prostate cancer‑driven osteogenesis at least partly relied on the integrin receptor α5β1. These results demonstrated that spondin 2 boosts osteogenesis via the PI3K/AKT/mTOR pathway under conditions of prostate tumor progression.

Introduction

Prostate cancer (PCa) is the most frequently diagnosed malignant tumor and a major cause of cancer mortality (6.8%) in men worldwide (1). A main problem arising from PCa is bone tumor metastasis. A total of ~80% of patients with advanced PCa develop bone metastases and are treated with androgen deprivation therapy (2). Androgen receptor (AR) is a leading factor for the development of bone metastasis, and recent advances in therapeutic options for PCa highlight the necessity to block AR signaling (3). However, the role of AR in osteogenesis in PCa remains controversial and unclear. Therefore, for future therapeutic developments, it is essential to determine the underlying mechanism of PCa-driven osteogenesis.

Tumor-induced osteogenesis is a complex process that involves cell disengagement from the microenvironment in situ, degradation of the surrounding extracellular matrix, tumor cell dissemination and final proliferation of distant secondary bone tumors (4). Emerging evidence suggests that cytokines involved in the process mentioned above can also act as chemoattractants on pre-osteoblastic MC3T3-E1 cells and promote the secretion of osteogenic factors (5). Almost all osteogenic factors are activated via two important osteogenic transcription factors, which are runt-related transcription factor 2 (Runx2) and osteoblast-specific transcription factor Osterix (68). However, the upstream factors and signaling pathways regulating these two osteogenic factors are still poorly understood.

As a member of the F-spondin family of secreted extracellular matrix proteins, spondin 2 is encoded by the SPON2 gene (9). Initially, spondin 2 was reported as a diagnostic marker specific for PCa (10,11). However, previous studies have shown that spondin 2 is overexpressed in the serum or tissue samples of malignant tumors, such as colorectal cancer and hepatocellular carcinoma (12,13). High levels of spondin 2 in colorectal cancer cells have been indicated to increase cell motility, thereby resulting in colorectal cancer metastasis in mice (14). Integrins are transmembrane heterodimers with α and β subunits that are considered to be major candidates as receptors for spondin 2 (15). Yang et al (16) analyzed the expression of integrins in MC3T3-E1 cells by flow cytometry and found high expression of integrin α5β1. Therefore, it was hypothesized that spondin 2 may play an important role in osteogenesis caused by PCa through integrin α5β1.

The aim of the present study was to elucidate the function as well as the underlying mechanism of PCa cell-derived spondin 2 during PCa-driven osteogenesis. The detailed mechanisms of spondin 2 function in PCa-induced bone metastasis need to be further clarified in future studies.

Materials and methods

Cell culture and treatment

The human RWPE-1 cell line, PCa cell lines (LNCaP and C4-2 cells) and the osteoblastic cell line MC3T3-E1 were all purchased from American Type Culture Collection. RWPE-1 cells were cultured in keratinocyte serum-free medium supplemented with 25 µg/ml bovine pituitary extract, 5 ng/ml human recombinant epidermal growth factor, 100 U/ml penicillin and 100 µg/ml streptomycin (all from Invitrogen; Thermo Fisher Scientific, Inc.). LNCaP and C4-2 cells were cultured in DMEM (Invitrogen; Thermo Fisher Scientific, Inc.), while MC3T3-E1 cells were cultured in α-MEM (Invitrogen; Thermo Fisher Scientific, Inc.). The culture media were supplemented with 10% fetal bovine serum (FBS; Invitrogen; Thermo Fisher Scientific, Inc.), 100 U/ml penicillin and 100 µg/ml streptomycin with (or without) 0.5 µg/ml spondin 2 neutralizing antibody (cat. no. SP2021041B; Wuhan Dian Biotechnology Co., Ltd.), and cells were maintained in a humidified incubator at 37°C with 5% CO2. MC3T3-E1 cells were treated with 0.1 or 1.0 µg/ml spondin 2 recombinant protein (rSpondin 2; cat. no. RPF396Mu01; Cloud-Clone Corp.) for 24 h in a humidified incubator at 37°C with 5% CO2. Control group cells were treated with 1X PBS for 24 h in a humidified incubator at 37°C with 5% CO2. In the integrin α5β1 inhibitor assay, MC3T3-E1 cells were treated with 100 µM ATN-161 (MedChemExpress) for 24 h in a humidified incubator at 37°C with 5% CO2.

Collection of conditioned media (CM)

LNCaP and C4-2 cells (2×106) were grown overnight in 100 mm culture dishes. After two washes with PBS, the cells were cultured in DMEM with 1% FBS for 48 h prior to collection of CM.

ELISA

The supernatants of normal prostate epithelial cells (RWPE-1) and PCa cells (LNCaP and C4-2) cultures were centrifuged at 1,000 × g for 15 min at 4°C before the assay. Proteins were assessed using a spondin-2 ELISA kit (cat. no. JCSJ2862; Shanghai Jichun Industrial Co., Ltd.) according to the manufacturer's instructions.

Reverse transcription-quantitative PCR (RT-qPCR)

Total RNA of MC3T3-E1 cells was extracted using TRIzol® reagent (Invitrogen; Thermo Fisher Scientific, Inc.). according to the protocol provided by the manufacturer. cDNA was synthesized using Prime Script RT Master Mix Kit (Takara Bio, Inc.) according to the manufacturer's instructions. RT-qPCR was performed in duplicate with a SYBR Premix Ex Taq™ kit (Takara Bio, Inc.) according to the manufacturer's instructions. PCR amplification conditions were as follows: Pre-denaturation at 95°C for 15 sec; 45 cycles of denaturation at 95°C for 5 sec and annealing/extension at 62°C for 30 sec. The following primers were used in the present study: Osterix forward, 5′-GATGGCGTCCTCTCTGCTTG-3′ and reverse, 5′-TCTTTGTGCCTCCTTTCCCC-3′; Runx2 forward, 5′-GACGAGGCAAGAGTTTCACC-3′ and reverse, 5′-GGACCGTCCACTGTCACTTT-3′; Gapdh forward, 5′-TCCACCACCCTGTTGCTGTA-3′ and reverse, 5′-ACCACAGTCCATGCCATCAC-3′. Relative expression of the targeted genes was calculated using the 2ΔΔCq method (17).

Western blotting

MC3T3-E1 cells were collected and lysed using RIPA buffer (Boster Biological Technology) containing protease inhibitor cocktail and PMSF (Boster Biological Technology). For determining the protein concentration, a BCA method was used, and equal amounts (30 µg) of proteins were separated under 90 V via 10% SDS-PAGE and subsequently transferred onto PVDF membranes (MilliporeSigma). After blocking the membranes with 1X TBS-Tween (TBST; 0.05% Tween-20) containing 5% skimmed milk for 2 h at room temperature, the membranes were incubated with primary anti-phosphorylated (p)-PI3K (cat. no. 4228), anti-PI3K (cat. no. 4257), anti-p-AKT (cat. no. 4060), anti-AKT (cat. no. 9272), anti-p-mTOR (cat. no. 5536), anti-mTOR (cat. no. 2972) (all 1:1,000 dilution; Cell Signaling Technology, Inc.) and anti-GAPDH (1:1,000 dilution; cat. no. 60004-1-Ig; ProteinTech Group, Inc.) antibodies overnight at 4°C. GAPDH was used as a normalization control. Membranes were rinsed in TBST, incubated with secondary anti-mouse IgG, AP-linked antibody (cat. no. 7056; 1:4,000 dilution; Cell Signaling Technology, Inc.) and anti-rabbit IgG, HRP-linked antibody (cat. no. 7074; 1:3,000 dilution; Cell Signaling Technology, Inc.) for 1 h at room temperature and then washed in 1X TBST. After incubation with the ECL Plus system (Amersham; Cytiva), signals were detected using the ImageQuant LAS 4000 mini system (GE Healthcare Bio-Sciences). The signals were detected using Adobe Photoshop CS3 software (Adobe Systems, Inc.).

Public database analysis

Gene expression data (GSE101607 dataset) were downloaded as raw signals from Gene Expression Omnibus (http://www.ncbi.nlm.nih.gov/geo) (18), and analyzed using the Geo2R tool from NCBI (https://www.ncbi.nlm.nih.gov/geo/geo2r). The differentially expressed genes were filtered by |log2FoldChange|>1 and FDR<0.05. Accordingly, a heatmap was generated using the ‘pheatmap’ package in R 3.6.1 (https://mirrors.tuna.tsinghua.edu.cn/CRAN/). Published gene expression profiles and clinical data of PCa patients were obtained from The Cancer Genome Atlas (TCGA) database (https://portal.gdc.cancer.gov). The data type was selected as ‘count’ and transformed into the transcript per million format. All patients with PCa were divided into two subgroups according to the median SPON2 expression, namely SPON2 low group (n=246) and SPON2 high group (n=246). This part of data was analyzed via Gene Set Enrichment Analysis (GSEA v3.0; http://www.gsea-msigdb.org/gsea/index.jsp). GSEA is a computational method that determines whether an a priori defined set of genes shows statistically significant, concordant differences between two biological states (19).

Statistical analysis

Data are presented as the mean ± SD and were analyzed using GraphPad Prism software v6.01 (GraphPad Software, Inc.). Differences between two groups were assessed using a two-tailed unpaired Student's t-test. One-way analysis of variance tests with Bonferroni's post hoc test was used for multiple comparisons. The association between SPON2 and the survival rate of patients with PCa was obtained using Kaplan-Meier analysis (the log-rank test was used to obtain the P-value) in GraphPad Prism software. P<0.05 was considered to indicate a statistically significant difference. All experiments were repeated at least three times.

Results

SPON2 expression is increased in patients with AR-positive PCa

Patients with castration-resistant PCa (GSE101607 dataset) were divided into two subgroups according to AR positivity, namely the AR-positive group (n=32) and the non-AR-positive group (n=8). By setting log2 (fold change) at ±1 and P<0.05, 687 differentially expressed genes (DEGs) between the AR-positive and the non-AR-positive group were identified with 188 upregulated DEGs and 499 downregulated DEGs. According to the heat map of the top five most up- and downregulated DEGs (Fig. 1A and Table SI), SPON2 was indicated to be highly expressed in AR-positive group (Fig. 1B). Consistent with this, ELISA results also showed that spondin 2 protein was secreted from PCa cells. Spondin 2 protein in the supernatants of PCa cells was significantly increased compared with normal prostate epithelial cells (235.13±61.82 pg/ml in LNCaP and 280.02±90.50 pg/ml in C4-2 cells vs. 34.97±12.60 pg/ml in RWPE-1 cells) (Fig. S1). In the present study, the association of SPON2 with the survival of patients with PCa based on TCGA data was also analyzed. The data showed that high level of SPON2 was associated with poor prognosis, but there was no significant difference between the two groups (Fig. S2).

PCa cell-derived spondin 2 promotes osteogenic factor production in osteoblasts

Next, the osteoblastic classification of PCa bone metastasis was sought to be determined, based on the evidence of tumor cell-derived osteogenic factors from two AR-positive PCa cell lines (LNCaP and C4-2), leading to increased bone formation. Compared with untreated MC3T3-E1 cells, it was found that CM from the LNCaP and C4-2 cells enhanced Osterix and Runx2 mRNA expression in osteoblasts (Fig. 2A and B). To examine whether spondin 2 was a critical factor in PCa cells, CM was treated with spondin 2 antibody. The results showed that spondin 2 antibody effectively reduced Osterix and Runx2 mRNA synthesis after treatment with PCa cell CM (Fig. 2C and D), indicating that PCa cell-derived spondin 2 promotes the production of osteogenic factors in osteoblasts.

SPON2 is positively associated with the PI3K/AKT/mTOR pathway in patients

The expression levels of SPON2 were further analyzed using TCGA PCa dataset, and it was found that the expression levels of SPON2 were elevated in tumor compared with normal tissues (Fig. 3A). Furthermore, patients with PCa were divided into two subgroups according to the median SPON2 expression, namely SPON2 low group (n=246) and SPON2 high group (n=246). As a previous study indicated that the PI3K/AKT/mTOR pathway could regulate the migration and invasion of PCa (20,21), it was then explored whether SPON2 expression was associated with the PI3K/AKT/mTOR pathway. According to GSEA using TCGA PCa dataset, high expression of SPON2 was positively associated with the enrichment of the PI3K/AKT/mTOR signaling pathway (Fig. 3B).

Spondin 2 activates the PI3K/AKT/mTOR pathway in osteoblasts

To investigate the specific effect of spondin 2 on osteogenic factor production in osteoblasts, MC3T3-E1 cells were treated with rSpondin 2. Different concentrations of rSpondin 2 were used according to a previous study (22), and in the present study it was observed that two concentrations of rSpondin2 (0.1 and 1.0 µg/ml) showed the most highly promoting effect on the transcription of Osterix and Runx2 as well as the phosphorylation of PI3K, AKT and mTOR. RT-qPCR analysis showed that rSpondin 2-induced MC3T3-E1 cells had higher transcriptional levels of Osterix and Runx2 compared with control cells in a concentration-dependent manner (Fig. 4A and B). To further investigate the underlying mechanism, the activity of the PI3K/AKT/mTOR pathway was further explored. The protein levels of PI3K, p-PI3K, AKT, p-AKT, p-mTOR and mTOR were measured in osteoblasts cultured with various concentrations of rSpondin 2. According to the results, the phosphorylation of PI3K, AKT and mTOR were significantly increased after treatment with rSpondin 2 in a concentration-dependent manner, compared with control cells. On the other hand, the total PI3K, AKT and mTOR levels remain unchanged (Fig. 4C).

Inhibition of integrin α5β1 suppresses the PI3K/AKT/mTOR pathway in osteoblasts

Spondin 2 is known to bind to integrin receptors (9). Integrins α5 and β1 are known to be expressed in osteoblasts on the bone surface (23). In order to determine whether spondin 2 activated the PI3K/AKT/mTOR pathway via integrin α5β1, an integrin α5β1 inhibitor (ATN-161) was used to determine its effects on spondin 2-mediated osteogenic factor production. According to RT-qPCR analysis, ATN-161 significantly inhibited spondin 2-induced mRNA expression of Osterix and Runx2 (Fig. 5A and B). Furthermore, western blot analysis indicated that ATN-161 significantly inhibited spondin 2-mediated PI3K, AKT and mTOR phosphorylation (Figs. 5C and S3).

Discussion

A previous study has shown that SPON2 is a new serum and histological diagnostic biomarker for PCa (11). Similarly, the present study found that spondin 2 facilitated osteogenic factor production induced by PCa cells, and SPON2 was upregulated in AR-positive tumors of patients with PCa. Moreover, it was identified that PCa cell-derived spondin 2 promoted the osteogenic activity of osteoblasts by activating the PI3K/AKT/mTOR pathway.

The disruption of homeostasis between osteoblasts is involved in PCa-induced osteogenesis (2,24). These processes are regulated by tumor cell-derived cytokines, such as bone morphogenetic protein, platelet-derived growth factor, insulin-like growth factor and extracellular calcium (2528). As an extracellular matrix protein, increased levels of spondin 2 in serum are correlated with high incidence of osteogenesis induced by prostate tumor cells (29). According to previous studies, spondin 2 is a critical regulator of cancer progression; however, its underlying mechanism in osteoblast activity remains to be elucidated (1113). On the other hand, Runx2 can promote the differentiation of mesenchymal stem cells towards osteoblasts, while Osterix plays an important role in osteoblast differentiation (30,31). On this basis, it was identified that spondin 2 derived from PCa cells significantly enhanced the expression of the osteogenic genes Runx2 and Osterix, indicating an increased activity of osteoblasts.

The PI3K/AKT/mTOR signaling pathway has been indicated to participate in cell proliferation, inflammation, immunity and tumorigenesis (3234). Inhibition of the PI3K/AKT/mTOR signaling pathway can suppress tumor growth and tumor-induced osteogenesis (35,36). Moreover, the PI3K/AKT/mTOR pathway has been reported to be a potential target in castration resistant PCa (37,38). However, whether spondin 2 upregulates the PI3K/AKT/mTOR signaling during osteogenesis driven by PCa progression requires further investigation. In the present study, it was indicated that spondin 2 secreted by AR-positive PCa cells could promote the differentiation of osteoblast precursors to mature osteoblasts (showed by the increased expression of Runx2 and Osterix) through the PI3K/AKT/mTOR signaling pathway. As a well-known receptor of spondin 2, integrin α5β1 plays a significant role in bone formation (23), and the present study showed that spondin 2 receptor inhibitor ATN-161 could inhibit spondin 2-mediated PI3K, AKT and mTOR phosphorylation in osteoblast precursor MC3T3-E1 cells.

The present study explored the role of spondin 2 on osteogenesis caused by PCa cells in vitro, while the association between spondin 2 and bone metastasis in animal models as well as patients with PCa requires further investigation.

In summary, the current study demonstrated that spondin 2 derived from AR-positive PCa cells could effectively enhance PCa-induced osteogenesis through activation of the PI3K/AKT/mTOR signaling cascade (Fig. 6). Furthermore, to the best of our knowledge, the present study was the first to demonstrate that integrin α5β1 is involved in spondin 2-regulated osteogenesis in PCa cells in vitro.

Supplementary Material

Supporting Data
Supporting Data

Acknowledgements

Not applicable.

Funding

Funding: No funding was received.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

Experiments were conceived and designed by CY and HW. HW and MZ performed the experiments and data analysis, prepared the figures and wrote the manuscript. WL helped with the experimental operation and data analysis. CY participated in manuscript writing. CY and HW confirm the authenticity of all the raw data. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Guise TA, Mohammad KS, Clines G, Stebbins EG, Wong DH, Higgins LS, Vessella R, Corey E, Padalecki S, Suva L and Chirgwin JM: Basic mechanisms responsible for osteolytic and osteoblastic bone metastases. Clin Cancer Res. 12:6213s–6216s. 2006. View Article : Google Scholar : PubMed/NCBI

3 

Cornford P, van den Bergh RCN, Briers E, Van den Broeck T, Cumberbatch MG, De Santis M, Fanti S, Fossati N, Gandaglia G, Gillessen S, et al: EAU-EANM-ESTRO-ESUR-SIOG guidelines on prostate cancer. Part II-2020 update: Treatment of relapsing and metastatic prostate cancer. Eur Urol. 79:263–282. 2021. View Article : Google Scholar : PubMed/NCBI

4 

Clezardin P, Coleman R, Puppo M, Ottewell P, Bonnelye E, Paycha F, Confavreux CB and Holen I: Bone metastasis: Mechanisms, therapies, and biomarkers. Physiol Rev. 101:797–855. 2021. View Article : Google Scholar : PubMed/NCBI

5 

Dayyani F, Varkaris A, Araujo JC, Song JH, Chatterji T, Trudel GC, Logothetis CJ and Gallick GE: Increased serum insulin-like growth factor-1 levels are associated with prolonged response to dasatinib-based regimens in metastatic prostate cancer. Prostate. 73:979–985. 2013. View Article : Google Scholar : PubMed/NCBI

6 

Zhang C, Long F, Wan J, Hu Y and He H: MicroRNA-205 acts as a tumor suppressor in osteosarcoma via targeting RUNX2. Oncol Rep. 35:3275–3284. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Jagga S, Sharma AR, Kim EJ and Nam JS: Isoflavone-enriched whole soy milk powder stimulates osteoblast differentiation. J Food Sci Technol. 58:595–603. 2021. View Article : Google Scholar : PubMed/NCBI

8 

Choi YH, Han Y, Jin SW, Lee GH, Kim GS, Lee DY, Chung YC, Lee KY and Jeong HG: Pseudoshikonin I enhances osteoblast differentiation by stimulating Runx2 and Osterix. J Cell Biochem. 119:748–757. 2018. View Article : Google Scholar : PubMed/NCBI

9 

Li Y, Cao C, Jia W, Yu L, Mo M, Wang Q, Huang Y, Lim JM, Ishihara M, Wells L, et al: Structure of the F-spondin domain of mindin, an integrin ligand and pattern recognition molecule. EMBO J. 28:286–297. 2009. View Article : Google Scholar : PubMed/NCBI

10 

Parry R, Schneider D, Hudson D, Parkes D, Xuan JA, Newton A, Toy P, Lin R, Harkins R, Alicke B, et al: Identification of a novel prostate tumor target, mindin/RG-1, for antibody-based radiotherapy of prostate cancer. Cancer Res. 65:8397–8405. 2005. View Article : Google Scholar : PubMed/NCBI

11 

Qian X, Li C, Pang B, Xue M, Wang J and Zhou J: Spondin-2 (SPON2), a more prostate-cancer-specific diagnostic biomarker. PLoS One. 7:e372252012. View Article : Google Scholar : PubMed/NCBI

12 

Zhang Q, Wang XQ, Wang J, Cui SJ, Lou XM, Yan B, Qiao J, Jiang YH, Zhang LJ, Yang PY and Liu F: Upregulation of spondin-2 predicts poor survival of colorectal carcinoma patients. Oncotarget. 6:15095–15110. 2015. View Article : Google Scholar : PubMed/NCBI

13 

Feng Y, Hu Y, Mao Q, Guo Y, Liu Y, Xue W and Cheng S: Upregulation of spondin-2 protein expression correlates with poor prognosis in hepatocellular carcinoma. J Int Med Res. 47:569–579. 2019. View Article : Google Scholar : PubMed/NCBI

14 

Schmid F, Wang Q, Huska MR, Andrade-Navarro MA, Lemm M, Fichtner I, Dahlmann M, Kobelt D, Walther W, Smith J, et al: SPON2, a newly identified target gene of MACC1, drives colorectal cancer metastasis in mice and is prognostic for colorectal cancer patient survival. Oncogene. 35:5942–5952. 2016. View Article : Google Scholar : PubMed/NCBI

15 

Huang C, Ou R, Chen X, Zhang Y, Li J, Liang Y, Zhu X, Liu L, Li M, Lin D, et al: Tumor cell-derived SPON2 promotes M2-polarized tumor-associated macrophage infiltration and cancer progression by activating PYK2 in CRC. J Exp Clin Cancer Res. 40:3042021. View Article : Google Scholar : PubMed/NCBI

16 

Yang RS, Lin WL, Chen YZ, Tang CH, Huang TH, Lu BY and Fu WM: Regulation by ultrasound treatment on the integrin expression and differentiation of osteoblasts. Bone. 36:276–283. 2005. View Article : Google Scholar : PubMed/NCBI

17 

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

18 

Ylitalo EB, Thysell E, Jernberg E, Lundholm M, Crnalic S, Egevad L, Stattin P, Widmark A, Bergh A and Wikström P: Subgroups of castration-resistant prostate cancer bone metastases defined through an inverse relationship between androgen receptor activity and immune response. Eur Urol. 71:776–787. 2017. View Article : Google Scholar : PubMed/NCBI

19 

Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES and Mesirov JP: Gene set enrichment analysis: A knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 102:15545–15550. 2005. View Article : Google Scholar : PubMed/NCBI

20 

Vo BT, Morton D Jr, Komaragiri S, Millena AC, Leath C and Khan SA: TGF-β effects on prostate cancer cell migration and invasion are mediated by PGE2 through activation of PI3K/AKT/mTOR pathway. Endocrinology. 154:1768–1779. 2013. View Article : Google Scholar : PubMed/NCBI

21 

Tan M, Xu J, Siddiqui J, Feng F and Sun Y: Depletion of SAG/RBX2 E3 ubiquitin ligase suppresses prostate tumorigenesis via inactivation of the PI3K/AKT/mTOR axis. Mol Cancer. 15:812016. View Article : Google Scholar : PubMed/NCBI

22 

Zhang YL, Li Q, Yang XM, Fang F, Li J, Wang YH, Yang Q, Zhu L, Nie HZ, Zhang XL, et al: SPON2 Promotes M1-like macrophage recruitment and inhibits hepatocellular carcinoma metastasis by distinct integrin-Rho GTPase-hippo pathways. Cancer Res. 78:2305–2317. 2018. View Article : Google Scholar : PubMed/NCBI

23 

Jiang W, Takeshita N, Maeda T, Sogi C, Oyanagi T, Kimura S, Yoshida M, Sasaki K, Ito A and Takano-Yamamoto T: Connective tissue growth factor promotes chemotaxis of preosteoblasts through integrin α5 and Ras during tensile force-induced intramembranous osteogenesis. Sci Rep. 11:23682021. View Article : Google Scholar : PubMed/NCBI

24 

Bai G, Cai Z, Zhai X, Xiong J, Zhang F and Li H: A new nomogram for the prediction of bone metastasis in patients with prostate cancer. J Int Med Res. 49:30006052110583642021. View Article : Google Scholar : PubMed/NCBI

25 

Lee Y, Schwarz E, Davies M, Jo M, Gates J, Wu J, Zhang X and Lieberman JR: Differences in the cytokine profiles associated with prostate cancer cell induced osteoblastic and osteolytic lesions in bone. J Orthop Res. 21:62–72. 2003. View Article : Google Scholar : PubMed/NCBI

26 

Boucher J, Balandre AC, Debant M, Vix J, Harnois T, Bourmeyster N, Péraudeau E, Chépied A, Clarhaut J, Debiais F, et al: Cx43 present at the leading edge membrane governs promigratory effects of osteoblast-conditioned medium on human prostate cancer cells in the context of bone metastasis. Cancers (Basel). 12:30132020. View Article : Google Scholar : PubMed/NCBI

27 

Rubin J, Fan X, Rahnert J, Sen B, Hsieh CL, Murphy TC, Nanes MS, Horton LG, Beamer WG and Rosen CJ: IGF-I secretion by prostate carcinoma cells does not alter tumor-bone cell interactions in vitro or in vivo. Prostate. 66:789–800. 2006. View Article : Google Scholar : PubMed/NCBI

28 

Tucci M, Mosca A, Lamanna G, Porpiglia F, Terzolo M, Vana F, Cracco C, Russo L, Gorzegno G, Tampellini M, et al: Prognostic significance of disordered calcium metabolism in hormone-refractory prostate cancer patients with metastatic bone disease. Prostate Cancer Prostatic Dis. 12:94–99. 2009. View Article : Google Scholar : PubMed/NCBI

29 

Marie PJ: Targeting integrins to promote bone formation and repair. Nat Rev Endocrinol. 9:288–295. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Xu X, Zhang C, Trotter TN, Gowda PS, Lu Y, Ponnazhagan S, Javed A, Li J and Yang Y: Runx2 deficiency in osteoblasts promotes myeloma progression by altering the bone microenvironment at new bone sites. Cancer Res. 80:1036–1048. 2020. View Article : Google Scholar : PubMed/NCBI

31 

Liu Q, Li M, Wang S, Xiao Z, Xiong Y and Wang G: Recent advances of osterix transcription factor in osteoblast differentiation and bone formation. Front Cell Dev Biol. 8:6012242020. View Article : Google Scholar : PubMed/NCBI

32 

Yuan G, Lian Z, Liu Q, Lin X, Xie D, Song F, Wang X, Shao S, Zhou B, Li C, et al: Phosphatidyl inositol 3-kinase (PI3K)-mTOR inhibitor PKI-402 inhibits breast cancer induced osteolysis. Cancer Lett. 443:135–144. 2019. View Article : Google Scholar : PubMed/NCBI

33 

Zhu WB, Xiao N and Liu XJ: Dietary flavonoid tangeretin induces reprogramming of epithelial to mesenchymal transition in prostate cancer cells by targeting the PI3K/Akt/mTOR signaling pathway. Oncol Lett. 15:433–440. 2018.PubMed/NCBI

34 

Le B, Powers GL, Tam YT, Schumacher N, Malinowski RL, Steinke L, Kwon G and Marker PC: Multi-drug loaded micelles delivering chemotherapy and targeted therapies directed against HSP90 and the PI3K/AKT/mTOR pathway in prostate cancer. PLoS One. 12:e01746582017. View Article : Google Scholar : PubMed/NCBI

35 

LoRusso PM: Inhibition of the PI3K/AKT/mTOR pathway in solid tumors. J Clin Oncol. 34:3803–3815. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Liu H, Li X, Lin J and Lin M: Morroniside promotes the osteogenesis by activating PI3K/Akt/mTOR signaling. Biosci Biotechnol Biochem. 85:332–339. 2021. View Article : Google Scholar : PubMed/NCBI

37 

Chang L, Graham PH, Hao J, Ni J, Bucci J, Cozzi PJ, Kearsley JH and Li Y: PI3K/Akt/mTOR pathway inhibitors enhance radiosensitivity in radioresistant prostate cancer cells through inducing apoptosis, reducing autophagy, suppressing NHEJ and HR repair pathways. Cell Death Dis. 5:e14372014. View Article : Google Scholar : PubMed/NCBI

38 

Berrak O, Arisan ED, Obakan-Yerlikaya P, Coker-Gürkan A and Palavan-Unsal N: mTOR is a fine tuning molecule in CDK inhibitors-induced distinct cell death mechanisms via PI3K/AKT/mTOR signaling axis in prostate cancer cells. Apoptosis. 21:1158–1178. 2016. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2023
Volume 49 Issue 1

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
Wang H, Zhang M, Lu W and Yuan C: Prostate cancer cell‑derived spondin 2 boosts osteogenic factor levels in osteoblasts via the PI3K/AKT/mTOR pathway. Oncol Rep 49: 23, 2023
APA
Wang, H., Zhang, M., Lu, W., & Yuan, C. (2023). Prostate cancer cell‑derived spondin 2 boosts osteogenic factor levels in osteoblasts via the PI3K/AKT/mTOR pathway. Oncology Reports, 49, 23. https://doi.org/10.3892/or.2022.8460
MLA
Wang, H., Zhang, M., Lu, W., Yuan, C."Prostate cancer cell‑derived spondin 2 boosts osteogenic factor levels in osteoblasts via the PI3K/AKT/mTOR pathway". Oncology Reports 49.1 (2023): 23.
Chicago
Wang, H., Zhang, M., Lu, W., Yuan, C."Prostate cancer cell‑derived spondin 2 boosts osteogenic factor levels in osteoblasts via the PI3K/AKT/mTOR pathway". Oncology Reports 49, no. 1 (2023): 23. https://doi.org/10.3892/or.2022.8460