Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2023 Jul 4;24(13):11059.
doi: 10.3390/ijms241311059.

Bone Morphogenetic Protein 13 Has Protumorigenic Effects on Hepatocellular Carcinoma Cells In Vitro

Affiliations

Bone Morphogenetic Protein 13 Has Protumorigenic Effects on Hepatocellular Carcinoma Cells In Vitro

Vanessa Kersten et al. Int J Mol Sci. .

Abstract

Activated hepatic stellate cells (HSCs) play a key role in hepatic fibrosis and, thus, build the "soil" for hepatocarcinogenesis. Furthermore, HSCs are known to promote the progression of hepatocellular carcinoma (HCC), but the molecular mechanisms are only incompletely understood. Recently, we newly described the expression of bone morphogenetic protein 13 (BMP13) by HSCs in fibrotic liver tissue. In addition, BMP13 has mostly been studied in the context of cartilage and bone repair, but not in liver disease or cancer. Thus, we aimed to analyze the expression and function of BMP13 in HCC. Expression analyses revealed high BMP13-expression in activated human HSCs, but not in human HCC-cell-lines. Furthermore, analysis of human HCC tissues showed a significant correlation between BMP13 and α-smooth muscle actin (α-SMA), and immunofluorescence staining confirmed the co-localization of BMP13 and α-SMA, indicating activated HSCs as the cellular source of BMP13 in HCC. Stimulation of HCC cells with recombinant BMP13 increased the expression of the inhibitors of differentiation 1 (ID1) and 2 (ID2), which are known targets of BMP-signaling and cell-cycle promotors. In line with this, BMP13-stimulation caused an induced SMAD 1/5/9 and extracellular signal-regulated kinase (ERK) phosphorylation, as well as reduced expression of cyclin-dependent kinase inhibitors 1A (CDKN1A) and 2A (CDKN2A). Furthermore, stimulation with recombinant BMP13 led to increased proliferation and colony size formation of HCC cells in clonogenicity assays. The protumorigenic effects of BMP13 on HCC cells were almost completely abrogated by the small molecule dorsomorphin 1 (DMH1), which selectively blocks the intracellular kinase domain of ALK2 and ALK3, indicating that BMP13 acts via these BMP type I receptors on HCC cells. In summary, this study newly identifies stroma-derived BMP13 as a potential new tumor promotor in HCC and indicates this secreted growth-factor as a possible novel therapeutic target in HCC.

Keywords: BMP13; hepatic stellate cells; hepatocellular carcinoma.

PubMed Disclaimer

Conflict of interest statement

The Human Tissue and Cell Research-Services GmbH is the part of the HTCR Foundation that is in charge for the technical isolation of primary cells out of human specimens. The primary liver cells used in the present study have been provided by the HTCR Foundation for free/without any charges. The authors declare no conflict of interest.

Figures

Figure 1
Figure 1
Expression of BMP13 in hepatocellular carcinoma (HCC). (A) Analysis of mRNA expression of BMP13 in activated hepatic stellate cells (HSCs) and four human HCC cell lines. (B) Western blot analysis of BMP13 protein expression in HSCs and four human HCC cell lines. GAPDH was used as a housekeeper. Densitometric analysis (BMP13/GAPDH) revealed that, compared to the BMP13 signal in HSCs, the immunosignal in HCC cells was 20 to 210-fold lower (20-fold in Hep3B cells, 23-fold in HepG2 cells, 210-fold in PLC/PRF/5 cells, and 30-fold in Huh7 cells). (C) Correlation of BMP13 and α-smooth muscle actin (α-SMA) RNA expression levels (log2(transcript per million)) in 361 human HCC tissues. The cancer genome atlas (TCGA)-derived data were used, applying the gene expression profiling interactive analysis (GEPIA) database. (D) Representative image of immunofluorescence staining for BMP13 (red) and α-SMA (green) in human HCC tissue section. (E) Hematoxylin and eosin staining of the same tissue section area as shown in panel (D) (*: p  <  0.05).
Figure 2
Figure 2
Effect of BMP13 on tumorigenicity of HCC cells in vitro. (A) Analysis of the proliferation of HCC cell lines Hep3B, HepG2, and PLC/PRF/5 after 24 h stimulation with rBMP13. The proliferation rate of control cells has been set as 1 and the effect of BMP13 is shown as the relative induction of proliferation in stimulated cells, compared to control cells. (B) Representative images of Hep3B cells stimulated without (ctr) and with recombinant BMP13 for 24 h (40-fold magnification). Quantification of (C) colony size, (D) colony number, and (E) representative images (1-fold magnification) in anchorage-dependent clonogenic assays with Hep3B treated without (ctr) or with rBMP13 for 9 days. The size and number of colonies of control cells have been set as 1 and the effect of BMP13 is shown as relative induction compared to control cells (*: p  <  0.05).
Figure 3
Figure 3
Effect of BMP13 on ID1 and cell-cycle regulators in HCC cells. (A) Analysis of ID1 mRNA expression in Hep3B cells treated with different doses of recombinant BMP13 (rBMP13) for 2 h. (B) Analysis of mRNA expression of ID1 in Hep3B cells after treatment with rBMP13 (200 ng/mL) for different time points. (C) Western blot analysis of ID1 expression in rBMP13 (200 ng/mL) treated and Hep3B control cells (ctr) after 24 h. GAPDH was used as a housekeeper. Densitometric analysis (ID1/GAPDH) confirmed an increased (1.9-fold) staining signal of ID1 in BMP13-treated cells. (D) Analysis of CDKN2A mRNA expression in Hep3B stimulated without (ctr) or with rBMP13 (200 ng/mL) for 24 h. (E) Western blot analysis of p16 protein expression in Hep3B cells stimulated with rBMP13 for 24 h. GAPDH was used as a housekeeper. Densitometric analysis (p16/GAPDH) showed a decreased (0.8-fold) staining signal of p16 in BMP13 treated cells. (F) Analysis of CDKN1A mRNA expression in Hep3B stimulated without (ctr) or with rBMP13 (200 ng/mL) for 24 h. (G) Western blot analysis of p21 protein expression in Hep3B cells stimulated with rBMP13 for 24 h. GAPDH was used as a housekeeper. Densitometric analysis (p21/GAPDH) showed a decreased (0.4-fold) staining signal of p21 in BMP13-treated cells (*: p  <  0.05).
Figure 4
Figure 4
Effect of BMP receptor inhibitor dorsomorphin 1 (DMH1), which inhibits ALK2 and ALK3 in HCC cells. (A) Western blot analysis of phosphorylated SMAD 1/5/9 and phosphorylated ERK in HCC cells after stimulation with rBMP13 (200 ng/mL) for 20 min and control cells (ctr). GAPDH was used as a housekeeper. Densitometric analysis (p-SMAD 1/5/9 /GAPDH) confirmed an increased staining signal of p-SMAD 1/5/9 in Hep3B cells (18.7-fold), HepG2 cells (5.9-fold), and PLC/PRF/5 cells (4.8-fold) treated with rBMP13. Densitometric analysis (p-ERK/GAPDH) confirmed an increased staining signal of p-ERK in Hep3B cells (4.4-fold), HepG2 cells (2.9-fold), and PLC/PRF/5 cells (2.0-fold) treated with rBMP13. (B) Effect of BMP receptor inhibitor DMH1 (10 nM) in Hep3B cells on rBMP13 (200 ng/mL)-induced phosphorylation of SMAD 1/5/9. Cells were preincubated with DMH1 for 15 min before 15 min stimulation with rBMP13. Densitometric analysis (p-SMAD 1/5/9 /GAPDH) confirmed that the p-SMAD 1/5/9 signal induced by BMP13 (2.1-fold compared to control) was completely abolished (0.45 compared to control). (C) Effect of DMH1 (10 nM) on rBMP13 (200 ng/mL)-induced ID1 mRNA expression. For mRNA expression analysis, cells were treated with DMH1 for 20 min and then stimulated with rBMP13 for 8 h. Effect of BMP receptor inhibitor DMH1 (10 nM) in Hep3B cells on rBMP13 (200 ng/mL)-induced (D) proliferation of Hep3B (E) and representative images (40-fold magnification) of Hep3B cells. Proliferation was assessed after 24 h stimulation with rBMP13. Proliferation of control cells was set as 1 and the effect of BMP13 is shown as relative induction of the proliferation rate compared to ctr-cells (*: p  <  0.05).

Similar articles

References

    1. Sung H., Ferlay J., Siegel R.L., Laversanne M., Soerjomataram I., Jemal A., Bray F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021;71:209–249. doi: 10.3322/caac.21660. - DOI - PubMed
    1. Llovet J.M., Zucman-Rossi J., Pikarsky E., Sangro B., Schwartz M., Sherman M., Gores G. Hepatocellular carcinoma. Nat. Rev. Dis. Prim. 2016;2:16018. doi: 10.1038/nrdp.2016.18. - DOI - PubMed
    1. Kim D.W., Talati C., Kim R. Hepatocellular carcinoma (HCC): Beyond sorafenib-chemotherapy. J. Gastrointest. Oncol. 2017;8:256–265. doi: 10.21037/jgo.2016.09.07. - DOI - PMC - PubMed
    1. Ahmed S., Gordon L., Dueck D.A., Souied O., Haider K. Current status of systemic therapy in hepatocellular cancer. Dig. Liver. Dis. 2020;53:397–402. doi: 10.1016/j.dld.2020.10.005. - DOI - PubMed
    1. El-Serag H.B. Hepatocellular carcinoma. N. Engl. J. Med. 2011;365:1118–1127. doi: 10.1056/NEJMra1001683. - DOI - PubMed

MeSH terms

Substances

Grants and funding

-