Bone morphogenetic protein 2 inhibits the proliferation and growth of human colorectal cancer cells

  • Authors:
    • Yunyuan Zhang
    • Xian Chen
    • Min Qiao
    • Bing-Qiang Zhang
    • Ning Wang
    • Zhonglin Zhang
    • Zhan Liao
    • Liyi Zeng
    • Youlin Deng
    • Fang Deng
    • Junhui Zhang
    • Liangjun Yin
    • Wei Liu
    • Qian Zhang
    • Zhengjian Ya
    • Jixing Ye
    • Zhongliang Wang
    • Lan Zhou
    • Hue H. Luu
    • Rex C. Haydon
    • Tong-Chuan He
    • Hongyu Zhang
  • View Affiliations

  • Published online on: July 3, 2014     https://doi.org/10.3892/or.2014.3308
  • Pages: 1013-1020
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Colorectal cancer (CRC) is one of the most deadly cancers worldwide. Significant progress has been made in understanding the molecular pathogenesis of CRC, which has led to successful early diagnosis, surgical intervention and combination chemotherapy. However, limited therapeutic options are available for metastatic and/or drug-resistant CRC. While the aberrantly activated Wnt/β-catenin pathway plays a critical initiating role in CRC development, disruption of the bone morphogenetic protein (BMP) pathway causes juvenile polyposis syndrome, suggesting that BMP signaling may play a role in CRC development. However, conflicting results have been reported concerning the possible roles of BMP signaling in sporadic colon cancer. Here, we investigated the effect of BMP2 on the proliferation, migration, invasiveness and tumor growth capability of human CRC cells. Using an adenovirus vector that overexpresses BMP2 and the piggyBac transposon-mediated stable BMP2 overexpression CRC line, we found that exogenous BMP2 effectively inhibited HCT116 cell proliferation and colony formation. BMP2 was shown to suppress colon cancer cell migration and invasiveness. Under a low serum culture condition, forced expression of BMP2 induced a significantly increased level of apoptosis in HCT116 cells. Using a xenograft tumor model, we found that forced expression of BMP2 in HCT116 cells suppressed tumor growth, accompanied by decreased cell proliferation activity. Taken together, our results strongly suggest that BMP2 plays an important inhibitory role in governing the proliferation and aggressive features of human CRC cells.

Introduction

Colorectal cancer (CRC) causes an average of 50,000 deaths per year in the US and has emerged as the second leading cause of cancer-related mortality in the US and worldwide (1,2). While early diagnosis and surgical intervention, along with combination chemotherapy, has led to improved outcomes, few effective strategies have emerged with which to treat colon cancer once first-line approaches have been exhausted (1,2). The initiation and progression of CRC development are characterized by the accumulation of growing numbers of genetic and epigenetic changes (3,4), while the aberrant activation of the Wnt pathway, either by inactivation of tumor-suppressor adenomatous polyposis coli (APC) or oncogenic activation of β-catenin, has been demonstrated as the essential initial step of tumorigenesis (3,5). Nonetheless, other alternative pathways have been implicated in CRC development. One involves the formation of serrated adenomas that are associated with mutations in BRAF (6). Another alternative pathway involves the formation of a hamartoma as a precursor lesion, which is in this last rare pathway to CRC that mutations in the bone morphogenetic protein (BMP) pathway were identified (7).

BMPs belong to the transforming growth factor β (TGFβ) superfamily (8). BMPs bind to a heterodimeric complex of transmembrane serine threonine kinase receptors type 1 and 2, triggering the phosphorylation and activation of the type 1 receptor by the type 2 receptor kinase. The activated type 1 receptor phosphorylates a receptor-associated SMAD which subsequently complexes with SMAD4 and translocates to the nucleus to regulate gene transcription (9). The importance of BMP signaling in colon cancer development has been highlighted by the identification of mutations in the BMP pathway in colorectal carcinogenesis (7). SMAD4 was identified as being frequently deleted in CRC, although the biological significance of this genetic change has always been attributed to loss of TGFβ signaling rather than BMP signaling (10). Mutations in BMP receptor 1A (BMPR1A) were found in patients with juvenile polyposis (JP), a rare autosomal dominant hamartomatous polyposis syndrome with an increased risk for the development of CRC (11). Mutations in SMAD4 and BMPR1A account for approximately half of all cases of JP (1214). Moreover, forced expression of the BMP antagonist noggin in the mouse intestine results in the formation of intestinal hamartomatous polyps (15).

However, conflicting results have been reported concerning the possible roles of BMPs in sporadic colon cancer. For example, several BMPs were found to be growth suppressive and may have their promoters methylated in colon cancer, compatible with a tumor-suppressor role for BMPs in CRC (1618). However, the expression of BMP4 and BMP7 was found to increase with progression through the adenoma-carcinoma sequence and to correlate with a worse prognosis (19,20). A more recent report showed that BMP signaling promotes the growth of primary human colon cancer in vivo (21). Therefore, the biological effects of BMPs on colon cancer development and progression remain to be fully elucidated.

In the present study, we investigated the effect of BMP2 on the proliferation, migration, invasiveness and tumor growth capabilities of human colon cancer cells. To achieve high levels of exogenous BMP2 expression, we constructed an adenovirus vector that overexpresses BMP2 and also generated the piggyBac transposon-mediated stable BMP2 overexpression cell line using the commonly used human colon cancer line HCT116. We found that exogenous BMP2 effectively inhibited HCT116 cell proliferation and colony formation. BMP2 was shown to suppress colon cancer cell migration and invasiveness as assessed by cell wound healing assay and Boyden chamber Transwell assay. Under a low serum condition, forced expression of BMP2 induced a significantly higher percentage of apoptosis in HCT116 cells than that in the controls. Using a xenograft tumor model, we found that forced expression of BMP2 in HCT116 cells suppressed tumor growth, accompanied by decreased proliferative activity. Thus, our results strongly suggest that BMP2 may play an important inhibitory role in controlling the proliferation and aggressive features of colon cancer cells.

Materials and methods

Cell culture and chemicals

Human colon cancer cell lines HCT116 and HEK-293 were obtained from the American Type Culture Collection (ATCC; Manassas, VA, USA). The cells were maintained in complete DMEM containing 10% fetal bovine serum (FBS; Hyclone, Logan, UT), 100 units of penicillin and 100 μg of streptomycin at 37°C in 5% CO2 as previously reported (2227). Unless otherwise indicated, all chemicals were purchased from Sigma-Aldrich (St. Louis, MO, USA) or Thermo Fisher (Pittsburgh, PA, USA).

Recombinant adenoviral vectors expressing BMP2 or GFP

Recombinant adenoviruses were generated using AdEasy technology (2832). Briefly, the coding regions of human BMP2 and green fluorescent protein (GFP) were PCR amplified and cloned into adenoviral shuttle vectors, which were subsequently used to generate recombinant adenoviruses in HEK-293 cells as previously described (29,32). The resultant recombinant adenoviruses were designated as AdGFP and AdBMP2, respectively. The amplified adenoviruses were titrated and stored at −80°C.

Establishment of BMP2/FLuc and FLuc expression stable cell lines

In order to construct BMP2 and/or firefly luciferase (FLuc) stable expression cell lines, the coding regions of human BMP2 and/or FLuc were PCR amplified and subcloned into a homemade piggyBac vector pMPB5, resulting in pMPB-BMP2/FLuc and pMPB-FLuc, respectively. The PCR amplified sequences were verified by DNA sequencing. To construct stable cell lines, exponentially growing HCT116 cells were co-transfected with pMPB-BMP2/FLuc or pMPB-FLuc and the Super piggyBac transposase expression vector (System Biosciences, Mountain View, CA, USA) using Lipofectamine transfection reagents by following the manufacturer’s instructions (Life Technologies, Grand Island, NY, USA). At 24 h after transfection, stable clones were selected in the presence of blasticidin S (10 μg/ml) for 5 days. The resultant stable cell lines were designated as HCT116-BMP2/FLuc and HCT116-FLuc, respectively. The stable cell lines were verified by RT-PCR for BMP2 expression and/or firefly luciferase activity assay.

Colony formation assay

Exponentially growing HCT116 cells were seeded in 6-well plates at a low density (300 cells/well) and infected with AdGFP or AdBMP2 (MOI=20) for 2 weeks to form colonies. The medium was replaced every 3–4 days. The uninfected cells were also included as a control. The colonies were stained with crystal violet. Each assay condition was conducted in triplicate and repeated in at least three batches of independent experiments. The average colony number for each group was calculated and expressed as the colony formation rate (colony number/seeded cell number) × 100%.

Cell proliferation (MTT) assay

In order to assess cell proliferation and viability, the MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay was performed as previously described (3339). Briefly, subconfluent HCT116 cells were infected with AdBMP2 or AdGFP (MOI=20) for 16 h and seeded in 96-well plates (1,000 cells/well). The plated cells were incubated in DMEM supplied with 1% FBS. At the indicated time points, the cells were incubated with 10 μl of the CellTiter 96® Non-Radioactive Cell Proliferation Assay (MTT) reagent (Promega, Madison, WI, USA) at 37°C for 4 h, followed by addition of 100 μl DMSO to dissolve the formazan products for 10 min at room temperature with gentle agitation. The absorbance was measured at 492 nm using a microtiter plate reader. Each assay condition was carried out in five replicates. The overall experiments were repeated at least in three batches of independent experiments.

Cell migration/wound healing assay

Subconfluent HCT116 cells were infected with AdGFP or AdBMP2 for 16 h and reseeded in 6-well plates at ~90% confluency. Upon cell attachment, scratches were made with pipette micro-tips. Floating cells were removed and the attached cells were maintained in DMEM supplemented with 1% FBS. The width of the scratched cell gaps were monitored and recorded at different time points. The scratch assay was carried out in triplicate and at least three scratch sites were monitored and recorded in each well. Percentage of the wound area closure was measured using ImageJ software.

Boyden chamber invasion/migration assays

The Matrigel cell invasion assay was performed as previously described (33,34,40). Briefly, subconfluent HCT116 cells were infected with AdGFP or AdBMP2 for 24 h. Polycarbonate membranes with 8-μm pores were coated with Matrigel (BD Biosciences). The membranes were rehydrated, and 5×105 of the transduced cells were placed onto each upper chamber of the Transwell unit. Medium with 10% FBS was used as a chemoattractant in the bottom chamber. The cells were allowed to invade at 37°C in 5% CO2 for 24 h. Cells were fixed in 10% formalin and washed with PBS. The cells were stained with hematoxylin and rinsed with water. Cells on the unmigrated side were gently wiped off with a wet cotton tip applicator, and the membrane was rinsed with water. The membranes containing the migrated cells were dried and mounted onto slides with Permount. The number of migrated cells per high power field (HPF) was determined by averaging 20 randomly counted HPFs. The assays were performed in triplicate and repeated in at least three batches of independent experiments.

Apoptosis and flow cytometric analysis

Subconfluent HCT116-FLuc and HCT116-BMP2/FLuc cells were cultured in DMEM containing 1% FBS for 72 h. Both floating and attached cells were collected, stained with Annexin V-FITC and propidium iodide (PI) using the Annexin V-FITC apoptosis detection kit (BD Pharmingen™, BD Biosciences). The stained cells were subjected to FACS analysis using the BD™ LSR II flow cytometer and FlowJo software. Each assay was performed in triplicate and repeated at least three times.

Xenograft tumor growth and xenogen whole body bioluminescence imaging

All animal experiments reported in this study were carried out in strict accordance with the recommendations established in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. The protocol was approved by the Institutional Animal Care and Use Committee (IACUC). For subcutaneous xenograft tumor formation, 4–6 week old male athymic nude (nu/nu) mice were purchased from Harlan Sprague Dawley (Indianapolis, IN, USA). Exponentially growing HCT116-BMP2/FLuc and HCT116-FLuc cells were harvested and resuspended in PBS. Cells (2×106 in 100 μl of PBS) were injected subcutaneously into the flanks of athymic mice (n=6/group). All animals were sacrificed 4 weeks after injection.

For weekly whole body bioluminescence imaging, the animals were anesthetized with isoflurane attached to a nose-cone mask within the Xenogen IVIS 200 imaging system. Mice were injected with D-Luciferin sodium salt (Gold BioTechnology, St. Louis, MO, USA) at 100 mg/kg in 0.1 ml sterile PBS. The pseudo-images were obtained by superimposing the emitted light over the grayscale images of the animal. The average signals in photons/sec/cm2/steradian were calculated. Quantitative analysis was carried out using the Xenogen’s Living Image software as previously described (22,35,38,41,42).

Histologic evaluation and immunohistochemical staining

The retrieved tissues were fixed in 10% buffered formalin and embedded in paraffin. The 5-μm sections were subjected to H&E staining. Immunohistochemistry was carried out as previously described (22,23,30,31,40,4346). For immunohistochemical staining, sections were deparaffinized, rehydrated, subjected to antigen retrieval and probed with a PNCA antibody (Santa Cruz Biotechnology), followed by incubation with biotin-secondary antibodies and streptavidin-HRP. PCNA protein was visualized by 3,3′-diaminobenzidine staining. Control IgG and minus-primary antibody staining were used as negative controls.

Statistical analysis

All quantitative data were calculated and are expressed as means ± standard deviation. The differences between groups were analyzed using one-way ANOVA followed by the Student-Newman-Keuls test using GraphPad Prism software. A P<0.05 was considered to indicate a statistically significant result.

Results and Discussion

Exogenous BMP2 inhibits the proliferative activity of human colon cancer cells

As the effects of BMP2 on colon cancer cells remain to be fully understood, we used an adenoviral vector overexpressing BMP2 and investigated its effects on the cell proliferation of colon cancer HCT116 cells. Using MTT assay, we found that AdBMP2-infected HCT116 cells exhibited lower proliferative activity at all tested time points when compared with that of the AdGFP-transduced cells, although only the differences on days 3 and 4 exhibited statistical significance (P<0.001) (Fig. 1A). When the AdBMP2, AdGFP or uninfected HCT116 cells were seeded at a very low density and allowed to form colonies, the BMP2-expressing HCT116 group formed significantly fewer colonies (Fig. 1B). Quantitatively, the BMP2-expressing HCT116 group formed approximately one third of the number of colonies when compared with the number in the GFP or uninfected control groups (Fig. 1C). These results suggest that BMP2 inhibits the proliferative activity of human colon cancer cells.

BMP2 inhibits the cell migration capability and invasiveness of human colon cancer cells

We further examined whether BMP2 affects the migration capability and invasiveness of colon cancer cells. We performed the commonly used cell wound healing assay to assess the effect on cell migration. The AdBMP2-transduced HCT116 cells were shown to close the scratched gaps on monolayer culture at a much slower pace than that of the GFP control groups (Fig. 2A). The percentage of wound closure was significantly higher in the GFP-transduced control cells at all tested time points (P<0.001) (Fig. 2B). Thus, these results suggest that exogenous BMP2 expression may significantly inhibit the migratory capability of colon cancer cells.

Using the Boyden Transwell extracellular matrix invasion assay, we analyzed the effect of BMP2 on the invasiveness of colon cancer cells. Consistent with our previous reports, GFP-treated HCT116 control cells were fairly aggressive and invaded the Matrigel-coated Transwell membrane with high efficiency, which was inhibited by exogenous BMP2 (Fig. 3A). Quantitatively, the BMP2-transduced HCT116 cells exhibited approximately <50% of the number of invaded cells in the GFP control group (P<0.001) (Fig. 3B), suggesting that BMP2 exerts an inhibitory effect on the invasiveness of colon cancer cells.

BMP2 effectively induces apoptosis in human colon cancer cells

We next investigated whether BMP2 induces apoptosis in colon cancer cells. We established a stable cell line HCT116-BMP2/FLuc that co-expressed human BMP2 and firefly luciferase (FLuc), while a control stable cell line HCT116-FLuc that only expresses FLuc was established in the same fashion. The exogenous expression of BMP2 was verified by RT-PCR analysis while the FLuc activity was determined using luciferase assay kits. We observed that HCT116-BMP2/FLuc cells grew normally in complete DMEM (with 10% FBS), compared with the parental HCT116 or HCT116-FLuc cells (data not shown). However, when the BMP2-expressing HCT116 cells were grown under no (0%) or low (1%) FBS condition, a significant increase in apoptosis was detected using the Annexin V labeling assay (Fig. 4A, panel a vs. b). When cultured in 1% FBS/DMEM for 72 h, the BMP2-expressing HCT116 cells underwent significant apoptosis (~42%), compared to ~8% in the control group (P<0.001) (Fig. 4B). Thus, these results suggest that BMP2 inhibits colon cancer cell proliferation at least in part through induction of apoptosis.

BMP2 effectively inhibits the growth of xenograft tumors derived from human colon cancer

Although the above in vitro data strongly suggest that BMP2 exhibits an inhibitory effect on colon cancer cell proliferation, we aimed to verify whether the inhibitory effect could be extended to in vivo tumor models. We previously demonstrated that HCT116 cells can reproducibly form subcutaneous tumors in athymic nude mice (35,36). We used the FLuc-tagged stable lines, HCT116-FLuc and HCT116-BMP2/FLuc. Subconfluent HCT116-FLuc and HCT116-BMP2/FLuc (BMP2) cells were subcutaneously injected into athymic nude mice, and the tumor growth was monitored at weeks 2 and 4 using whole body xenogen bioluminescence. We found that the BMP2-expressing HCT116 group formed significantly smaller tumor masses at each time point (Fig. 5A). Quantitative analysis revealed that the tumor growth in the BMP2-expressing HCT116 tumors was ~26% when compared with the control group (P<0.03) (Fig. 5B). When the retrieved tumor samples were fixed, embedded and sectioned for H&E staining, the samples from the BMP2/FLuc group exhibited significant necrosis and low cell proliferation, compared with these parameters in the FLuc control group (Fig. 5C, panel a vs. b). The embedded samples were also sectioned and subjected to immunohistochemical staining with a PCNA antibody. We found that the tumor samples formed by HCT116 cells expressing BMP2 exhibited a significantly diminished staining of PCNA expression in the tumor cells (Fig. 5C, panel c vs. d), suggesting that the BMP2-expressing colon cancer cells have a decreased proliferative activity. Collectively, these in vivo results further confirm that BMP2 exhibits strong inhibitory effects on colon cancer cells, possibly through inhibiting their proliferation and migration and inducing apoptosis.

BMP signaling may play an important role in modulating colorectal tumorigenesis

The findings from our in vitro and in vivo studies demonstrated that exogenous BMP2 inhibits cell proliferation, migration and invasion, induces apoptosis and suppresses in vivo xenograft tumor growth of human colon cancer cells. The importance of BMP signaling in colorectal tumorigenesis has been highlighted by the identification of frequent mutations of SMAD4 in CRC (10) and mutations in BMP receptor 1A (BMPR1A) in patients with juvenile polyposis (JP) (1214), which is associated with an increased risk for the development of CRC (11). Moreover, forced expression of the BMP antagonist noggin in mouse intestine was found to result in the formation of intestinal hamartomatous polyps (15).

Consistent with our findings are previous reports in which BMP2, BMP3 or BMP7 were shown to have growth-suppressive activities in colon cancer cells (1618), although the expression of BMP4 and BMP7 was found to correlate with a worse prognosis (19,20). Notably, a more recent report showed that BMP signaling promotes the growth of primary human colon cancer in vivo and the investigators proposed that blockade of BMP signaling may have beneficial effects against at least a subset of advanced colon cancers (21). Nonetheless, studies have revealed that genetic variations in the BMP signaling pathway may be associated with the etiology, survival and/or prognosis of colon and rectal cancer (18,4749).

Mechanistically, an early study suggested that BMP2 may act as a tumor suppressor promoting apoptosis in mature colonic epithelial cells (16), although it was suggested that BMP may also utilize SMAD4-independent pathways for growth suppression in colon cancers (18). Notably, it was reported that statins, acting as DNMT inhibitors can demethylate the BMP2 promoter, activate BMP signaling, induce differentiation of colon cancer stem cells and reduce their ‘stemness’ (50). Moreover, BMP-induced growth suppression may be mediated in part by p21WAF1, which is inhibited by RAS/ERK, as in colon cancer cells where BMP-SMAD signaling and growth suppression are facilitated by p21WAF1 but diminished by oncogenic K-RAS (51). It has been reported that suppression of the PI3 kinase/Akt pathway may be correlated with the development of BMP2 resistance and invasion in BMP2-induced epithelial-to-mesenchymal transformation (EMT) in colon cancer (52). It has been reported that the anti-mitogenic effect of proteasome inhibitors on colon cancer cells may require BMP signaling (53).

In summary, we investigated the effect of BMP2 on the proliferation, migration, invasiveness and tumor growth capabilities of human colon cancer cells. We found that exogenous BMP2 effectively inhibited HCT116 cell proliferation and colony formation. BMP2 also suppressed colon cancer cell migration and invasiveness. Forced expression of BMP2 induced significant apoptosis in HCT116 cells. Using an xenograft tumor model, we found that forced expression of BMP2 in HCT116 cells suppressed tumor growth, accompanied by decreased proliferative activity. Collectively, our results strongly suggest that BMP2 plays an inhibitory role in controlling the proliferation and aggressive features associated with colon cancer cells.

Acknowledgements

The present study was supported in part by research grants from the National Institutes of Health (CA106569, AT004418, AR50142 and AR054381 to T-C.H, R.C.H. and H.H.L.) and the 973 Program of Ministry of Science and Technology (MOST) of China (#2011CB707900 to T-C.H.). This study was also supported in part by The University of Chicago Core Facility Subsidy grant from the National Center for Advancing Translational Sciences (NCATS) of the National Institutes of Health through grant no. UL1 TR000430.

References

1 

Bertrand FE, Angus CW, Partis WJ and Sigounas G: Developmental pathways in colon cancer: crosstalk between WNT, BMP, Hedgehog and Notch. Cell Cycle. 11:4344–4351. 2012. View Article : Google Scholar : PubMed/NCBI

2 

Siegel R, Ma J, Zou Z and Jemal A: Cancer statistics, 2014. CA Cancer J Clin. 64:9–29. 2014. View Article : Google Scholar

3 

Kinzler KW and Vogelstein B: Lessons from hereditary colorectal cancer. Cell. 87:159–170. 1996. View Article : Google Scholar : PubMed/NCBI

4 

Vogelstein B, Papadopoulos N, Velculescu VE, Zhou S, Diaz LA Jr and Kinzler KW: Cancer genome landscapes. Science. 339:1546–1558. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Kinzler KW, Nilbert MC, Su LK, et al: Identification of FAP locus genes from chromosome 5q21. Science. 253:661–665. 1991. View Article : Google Scholar : PubMed/NCBI

6 

Chan TL, Zhao W, Leung SY and Yuen ST: BRAF and KRAS mutations in colorectal hyperplastic polyps and serrated adenomas. Cancer Res. 63:4878–4881. 2003.PubMed/NCBI

7 

Hardwick JC, Kodach LL, Offerhaus GJ and van den Brink GR: Bone morphogenetic protein signalling in colorectal cancer. Nat Rev Cancer. 8:806–812. 2008. View Article : Google Scholar : PubMed/NCBI

8 

Massague J: TGF-beta signal transduction. Annu Rev Biochem. 67:753–791. 1998. View Article : Google Scholar

9 

Massague J: How cells read TGF-beta signals. Nat Rev Mol Cell Biol. 1:169–178. 2000. View Article : Google Scholar : PubMed/NCBI

10 

Thiagalingam S, Lengauer C, Leach FS, et al: Evaluation of candidate tumour suppressor genes on chromosome 18 in colorectal cancers. Nat Genet. 13:343–346. 1996. View Article : Google Scholar : PubMed/NCBI

11 

Howe JR, Bair JL, Sayed MG, et al: Germline mutations of the gene encoding bone morphogenetic protein receptor 1A in juvenile polyposis. Nat Genet. 28:184–187. 2001. View Article : Google Scholar : PubMed/NCBI

12 

Howe JR, Roth S, Ringold JC, et al: Mutations in the SMAD4/DPC4 gene in juvenile polyposis. Science. 280:1086–1088. 1998. View Article : Google Scholar : PubMed/NCBI

13 

Howe JR, Sayed MG, Ahmed AF, et al: The prevalence of MADH4 and BMPR1A mutations in juvenile polyposis and absence of BMPR2, BMPR1B, and ACVR1 mutations. J Med Genet. 41:484–491. 2004.PubMed/NCBI

14 

Sayed MG, Ahmed AF, Ringold JR, et al: Germline SMAD4 or BMPR1A mutations and phenotype of juvenile polyposis. Ann Surg Oncol. 9:901–906. 2002.PubMed/NCBI

15 

Haramis AP, Begthel H, van den Born M, et al: De novo crypt formation and juvenile polyposis on BMP inhibition in mouse intestine. Science. 303:1684–1686. 2004. View Article : Google Scholar : PubMed/NCBI

16 

Hardwick JC, Van Den Brink GR, Bleuming SA, et al: Bone morphogenetic protein 2 is expressed by, and acts upon, mature epithelial cells in the colon. Gastroenterology. 126:111–121. 2004. View Article : Google Scholar : PubMed/NCBI

17 

Loh K, Chia JA, Greco S, et al: Bone morphogenic protein 3 inactivation is an early and frequent event in colorectal cancer development. Genes Chromosomes Cancer. 47:449–460. 2008. View Article : Google Scholar : PubMed/NCBI

18 

Beck SE, Jung BH, Fiorino A, et al: Bone morphogenetic protein signaling and growth suppression in colon cancer. Am J Physiol Gastrointest Liver Physiol. 291:G135–G145. 2006. View Article : Google Scholar : PubMed/NCBI

19 

Deng H, Makizumi R, Ravikumar TS, Dong H, Yang W and Yang WL: Bone morphogenetic protein-4 is overexpressed in colonic adenocarcinomas and promotes migration and invasion of HCT116 cells. Exp Cell Res. 313:1033–1044. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Motoyama K, Tanaka F, Kosaka Y, et al: Clinical significance of BMP7 in human colorectal cancer. Ann Surg Oncol. 15:1530–1537. 2008. View Article : Google Scholar : PubMed/NCBI

21 

Lorente-Trigos A, Varnat F, Melotti A and Ruiz i Altaba A: BMP signaling promotes the growth of primary human colon carcinomas in vivo. J Mol Cell Biol. 2:318–332. 2010. View Article : Google Scholar : PubMed/NCBI

22 

Luo X, Chen J, Song WX, et al: Osteogenic BMPs promote tumor growth of human osteosarcomas that harbor differentiation defects. Lab Invest. 88:1264–1277. 2008. View Article : Google Scholar : PubMed/NCBI

23 

Tang N, Song WX, Luo J, et al: BMP-9-induced osteogenic differentiation of mesenchymal progenitors requires functional canonical Wnt/beta-catenin signaling. J Cell Mol Med. 13:2448–2464. 2009. View Article : Google Scholar : PubMed/NCBI

24 

Gao Y, Huang E, Zhang H, et al: Crosstalk between Wnt/β-catenin and estrogen receptor signaling synergistically promotes osteogenic differentiation of mesenchymal progenitor cells. PloS One. 8:e824362013.

25 

Kong Y, Zhang H, Chen X, et al: Destabilization of heterologous proteins mediated by the GSK3β phosphorylation domain of the β-catenin protein. Cell Physiol Biochem. 32:1187–1199. 2013.PubMed/NCBI

26 

Zhang W, Zhang H, Wang N, et al: Modulation of β-catenin signaling by the inhibitors of MAP kinase, tyrosine kinase, and PI3-kinase pathways. Int J Med Sci. 10:1888–1898. 2013.

27 

Chen X, Luther G, Zhang W, et al: The E–F hand calcium-binding protein S100A4 regulates the proliferation, survival and differentiation potential of human osteosarcoma Cells. Cell Physiol Biochem. 32:1083–1096. 2013.

28 

Cheng H, Jiang W, Phillips FM, et al: Osteogenic activity of the fourteen types of human bone morphogenetic proteins (BMPs. J Bone Joint Surg Am. 85-A:1544–1552. 2003.PubMed/NCBI

29 

He TC, Zhou S, da Costa LT, Yu J, Kinzler KW and Vogelstein B: A simplified system for generating recombinant adenoviruses. Proc Natl Acad Sci USA. 95:2509–2514. 1998. View Article : Google Scholar : PubMed/NCBI

30 

Kang Q, Song WX, Luo Q, et al: A comprehensive analysis of the dual roles of BMPs in regulating adipogenic and osteogenic differentiation of mesenchymal progenitor cells. Stem Cells Dev. 18:545–559. 2009. View Article : Google Scholar : PubMed/NCBI

31 

Kang Q, Sun MH, Cheng H, et al: Characterization of the distinct orthotopic bone-forming activity of 14 BMPs using recombinant adenovirus-mediated gene delivery. Gene Ther. 11:1312–1320. 2004. View Article : Google Scholar : PubMed/NCBI

32 

Luo J, Deng ZL, Luo X, et al: A protocol for rapid generation of recombinant adenoviruses using the AdEasy system. Nat Protoc. 2:1236–1247. 2007. View Article : Google Scholar : PubMed/NCBI

33 

Luu HH, Kang Q, Park JK, et al: An orthotopic model of human osteosarcoma growth and spontaneous pulmonary metastasis. Clin Exp Metastasis. 22:319–329. 2005. View Article : Google Scholar : PubMed/NCBI

34 

Luu HH, Zhou L, Haydon RC, et al: Increased expression of S100A6 is associated with decreased metastasis and inhibition of cell migration and anchorage independent growth in human osteosarcoma. Cancer Lett. 229:135–148. 2005. View Article : Google Scholar : PubMed/NCBI

35 

He BC, Gao JL, Luo X, et al: Ginsenoside Rg3 inhibits colorectal tumor growth through the down-regulation of Wnt/β-catenin signaling. Int J Oncol. 38:437–445. 2011.PubMed/NCBI

36 

He BC, Gao JL, Zhang BQ, et al: Tetrandrine inhibits Wnt/β-catenin signaling and suppresses tumor growth of human colorectal cancer. Mol Pharmacol. 79:211–219. 2011.

37 

Su Y, Luo X, He BC, et al: Establishment and characterization of a new highly metastatic human osteosarcoma cell line. Clin Exp Metastasis. 26:599–610. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Su Y, Wagner ER, Luo Q, et al: Insulin-like growth factor binding protein 5 suppresses tumor growth and metastasis of human osteosarcoma. Oncogene. 30:3907–3917. 2011. View Article : Google Scholar : PubMed/NCBI

39 

Wang J, Zhang H, Zhang W, et al: Bone morphogenetic protein-9 (BMP9) effectively induces osteo/odontoblastic differentiation of the reversibly immortalized stem cells of dental apical papilla. Stem Cells Dev. Mar 21–2014.(Epub ahead of print).

40 

Luo Q, Kang Q, Si W, et al: Connective tissue growth factor (CTGF) is regulated by Wnt and bone morphogenetic proteins signaling in osteoblast differentiation of mesenchymal stem cells. J Biol Chem. 279:55958–55968. 2004. View Article : Google Scholar : PubMed/NCBI

41 

Luther GA, Lamplot J, Chen X, et al: IGFBP5 domains exert distinct inhibitory effects on the tumorigenicity and metastasis of human osteosarcoma. Cancer Lett. 336:222–230. 2013. View Article : Google Scholar : PubMed/NCBI

42 

He BC, Chen L, Zuo GW, et al: Synergistic antitumor effect of the activated PPARgamma and retinoid receptors on human osteosarcoma. Clin Cancer Res. 16:2235–2245. 2010. View Article : Google Scholar : PubMed/NCBI

43 

Sharff KA, Song WX, Luo X, et al: Hey1 basic helix-loop-helix protein plays an important role in mediating BMP9-induced osteogenic differentiation of mesenchymal progenitor cells. J Biol Chem. 284:649–659. 2009. View Article : Google Scholar : PubMed/NCBI

44 

Chen L, Jiang W, Huang J, et al: Insulin-like growth factor 2 (IGF-2) potentiates BMP-9-induced osteogenic differentiation and bone formation. J Bone Miner Res. 25:2447–2459. 2010. View Article : Google Scholar : PubMed/NCBI

45 

Zhang W, Deng ZL, Chen L, et al: Retinoic acids potentiate BMP9-induced osteogenic differentiation of mesenchymal progenitor cells. PloS One. 5:e119172010. View Article : Google Scholar : PubMed/NCBI

46 

Luo J, Tang M, Huang J, et al: TGFbeta/BMP type I receptors ALK1 and ALK2 are essential for BMP9-induced osteogenic signaling in mesenchymal stem cells. J Biol Chem. 285:29588–29598. 2010. View Article : Google Scholar : PubMed/NCBI

47 

Slattery ML, Lundgreen A, Herrick JS, Wolff RK and Caan BJ: Genetic variation in the transforming growth factor-β signaling pathway and survival after diagnosis with colon and rectal cancer. Cancer. 117:4175–4183. 2011.

48 

Slattery ML, Lundgreen A, Herrick JS, et al: Genetic variation in bone morphogenetic protein and colon and rectal cancer. Int J Cancer. 130:653–664. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Xiang L, Wang S, Jin X, Duan W, Ding X and Zheng C: Expression of BMP2, TLR3, TLR4 and COX2 in colorectal polyps, adenoma and adenocarcinoma. Mol Med Rep. 6:973–976. 2012.PubMed/NCBI

50 

Kodach LL, Jacobs RJ, Voorneveld PW, et al: Statins augment the chemosensitivity of colorectal cancer cells inducing epigenetic reprogramming and reducing colorectal cancer cell ‘stemness’ via the bone morphogenetic protein pathway. Gut. 60:1544–1553. 2011.PubMed/NCBI

51 

Beck SE, Jung BH, Del Rosario E, Gomez J and Carethers JM: BMP-induced growth suppression in colon cancer cells is mediated by p21WAF1stabilization and modulated by RAS/ERK. Cell Signal. 19:1465–1472. 2007. View Article : Google Scholar : PubMed/NCBI

52 

Kang MH, Kang HN, Kim JL, Kim JS, Oh SC and Yoo YA: Inhibition of PI3 kinase/Akt pathway is required for BMP2-induced EMT and invasion. Oncol Rep. 22:525–534. 2009.PubMed/NCBI

53 

Wu WK, Sung JJ, Wu YC, Li ZJ, Yu L and Cho CH: Bone morphogenetic protein signalling is required for the anti-mitogenic effect of the proteasome inhibitor MG-132 on colon cancer cells. Br J Pharmacol. 154:632–638. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2014
Volume 32 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
Zhang Y, Chen X, Qiao M, Zhang B, Wang N, Zhang Z, Liao Z, Zeng L, Deng Y, Deng F, Deng F, et al: Bone morphogenetic protein 2 inhibits the proliferation and growth of human colorectal cancer cells. Oncol Rep 32: 1013-1020, 2014
APA
Zhang, Y., Chen, X., Qiao, M., Zhang, B., Wang, N., Zhang, Z. ... Zhang, H. (2014). Bone morphogenetic protein 2 inhibits the proliferation and growth of human colorectal cancer cells. Oncology Reports, 32, 1013-1020. https://doi.org/10.3892/or.2014.3308
MLA
Zhang, Y., Chen, X., Qiao, M., Zhang, B., Wang, N., Zhang, Z., Liao, Z., Zeng, L., Deng, Y., Deng, F., Zhang, J., Yin, L., Liu, W., Zhang, Q., Ya, Z., Ye, J., Wang, Z., Zhou, L., Luu, H. H., Haydon, R. C., He, T., Zhang, H."Bone morphogenetic protein 2 inhibits the proliferation and growth of human colorectal cancer cells". Oncology Reports 32.3 (2014): 1013-1020.
Chicago
Zhang, Y., Chen, X., Qiao, M., Zhang, B., Wang, N., Zhang, Z., Liao, Z., Zeng, L., Deng, Y., Deng, F., Zhang, J., Yin, L., Liu, W., Zhang, Q., Ya, Z., Ye, J., Wang, Z., Zhou, L., Luu, H. H., Haydon, R. C., He, T., Zhang, H."Bone morphogenetic protein 2 inhibits the proliferation and growth of human colorectal cancer cells". Oncology Reports 32, no. 3 (2014): 1013-1020. https://doi.org/10.3892/or.2014.3308