We therefore performed flow-cytometric analyses for putative canc

We therefore performed flow-cytometric analyses for putative cancer stem cell markers in HCC cells cultured on soft (1 kPa) and stiff (12 kPa) supports, both without and following cisplatin treatment (Fig. 8A). Culture on soft versus stiff supports was associated with an enrichment for the cell surface markers CD133 (1.5-fold, P < 0.001), c-kit (1.3-fold, ATM inhibitor cancer P = 0.78), CD44 (6.4-fold, P < 0.001), and CXCR-4

(2.9-fold, P < 0.01). Following cisplatin treatment, there was statistically significant up-regulation of CD44 (1.7-fold, P < 0.01), CD133 (1.6-fold, P < 0.01) and c-kit (15.8-fold, P < 0.01) for cells maintained on soft but not stiff supports. Additionally, real-time PCR demonstrated up-regulation of selleck chemical stem cell-associated

transcription factors OCT4 and NANOG in HepG2 cells cultured on soft versus stiff supports, both in untreated controls (OCT4 2.0-fold increase, P < 0.05; NANOG 2.7-fold increase, P < 0.05) and following cisplatin treatment (OCT4 2.0-fold increase, P < 0.05; NANOG 3.4-fold increase, P < 0.05) (Fig. 8B). In this study, we demonstrated that the stiffness of the subcellular matrix profoundly alters the phenotype and behavior of HCC cells in vitro. Pathophysiological increases in matrix stiffness, as encountered in fibrotic and cirrhotic livers,19 Phloretin promote the proliferation of HCC cells. Our work defines novel mechanisms linking the physical properties of the fibrotic liver and the malignant behavior of HCC. Our data is consistent with in vivo evidence, not only of de novo HCC development and progression against a background of

cirrhosis, but also animal studies showing that the induction of liver fibrosis is associated with accelerated tumor growth following orthotopic HCC implantation.20, 21 Furthermore, histological examination of human HCC specimens demonstrates a significant association between the presence of hepatic fibrosis and enhanced tumor cell proliferation.22 Critically, our findings suggest that a reduction in the stiffness of the cancer cell niche, as would be encountered by a disseminated tumor cell entering an unaffected secondary site, would be sufficient to promote reversible cellular quiescence and cancer cell dormancy. It has previously been demonstrated that matrix stiffness can regulate proliferation in nontransformed cells. More recently increased matrix stiffness has been shown to promote cellular proliferation in glioma cells.23 We have extended these findings to a range of epithelial malignancies, including HCC (Supporting Fig. 9). Furthermore, we have shown that β1-integrin and FAK (the canonical mediator of integrin-related signaling) regulate stiffness-dependent proliferation in HCC cells.

Leave a Reply

Your email address will not be published. Required fields are marked *

*

You may use these HTML tags and attributes: <a href="" title=""> <abbr title=""> <acronym title=""> <b> <blockquote cite=""> <cite> <code> <del datetime=""> <em> <i> <q cite=""> <strike> <strong>