American Association for Cancer Research
Browse

sorry, we can't preview this file

00085472can173844-sup-193888_3_supp_4834213_pfffvy.png (182.04 kB)

Supplementary Figure 6 from CTGF Mediates Tumor–Stroma Interactions between Hepatoma Cells and Hepatic Stellate Cells to Accelerate HCC Progression

Download (182.04 kB)
figure
posted on 2023-03-31, 03:45 authored by Yuki Makino, Hayato Hikita, Takahiro Kodama, Minoru Shigekawa, Ryoko Yamada, Ryotaro Sakamori, Hidetoshi Eguchi, Eiichi Morii, Hideki Yokoi, Masashi Mukoyama, Suemizu Hiroshi, Tomohide Tatsumi, Tetsuo Takehara

Supplementary Fig. 6. Protein expression of p-STAT-3 in liver cancer is decreased by the hepatocyte-specific CTGF knockout in KrasG12D mice. Protein expression of p-STAT-3 in liver tumors in KrasG12D CTGF+/+, KrasG12D CTGF+/- and KrasG12D CTGF-/- mice at 8 months of age.

Funding

Grant-in-Aid for Scientific Research

Grant-in-Aid for Young Scientists

Japan Agency for Medical Research and Development

History

ARTICLE ABSTRACT

Connective tissue growth factor (CTGF) is a matricellular protein related to hepatic fibrosis. This study aims to clarify the roles of CTGF in hepatocellular carcinoma (HCC), which usually develops from fibrotic liver. CTGF was overexpressed in 93 human HCC compared with nontumorous tissues, primarily in tumor cells. Increased CTGF expression was associated with clinicopathologic malignancy of HCC. CTGF was upregulated in hepatoma cells in hepatocyte-specific Kras-mutated mice (Alb-Cre KrasLSL-G12D/+). Hepatocyte-specific knockout of CTGF in these mice (Alb-Cre KrasLSL-G12D/+ CTGFfl/fl) decreased liver tumor number and size. Hepatic stellate cells (HSC) were present in both human and murine liver tumors, and α-SMA expression, a marker of HSC activation, positively correlated with CTGF expression. Forced expression of CTGF did not affect growth of PLC/PRF/5 cells, a hepatoma cell line with little CTGF expression, but facilitated their growth in the presence of LX-2 cells, an HSC line. The growth of HepG2 cells, which express high levels of CTGF, was promoted by coculture with LX-2 cells compared with monoculture. Growth promotion by LX-2 cells was negated by an anti-CTGF antibody in both culture and xenografts. Coculturing LX-2 cells with HepG2 cells drove LX-2-derived production of IL6, which led to STAT-3 activation and proliferation of HepG2 cells. An anti-CTGF antibody reduced IL6 production in LX-2 cells and suppressed STAT-3 activation in HepG2 cells. In conclusion, our data identify tumor cell–derived CTGF as a keystone in the HCC microenvironment, activating nearby HSC that transmit progrowth signals to HCC cells, and this interaction is susceptible to inhibition by an anti-CTGF antibody.Significance: Protumor cross-talk between cancer cells and hepatic stellate cells presents an opportunity for therapeutic intervention against HCC.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/78/17/4902/F1.large.jpg. Cancer Res; 78(17); 4902–14. ©2018 AACR.

Usage metrics

    Cancer Research

    Licence

    Exports

    RefWorks
    BibTeX
    Ref. manager
    Endnote
    DataCite
    NLM
    DC