Share this post on:

A, we also revealed that bKlotho could also reduce tumor genesis ability in vivo. These results demonstrated bKlotho has an anti-tumorigenic role in HCC. Moreover, bKlotho interacts with FGFR4 to form a complex and the bKlotho-FGFR4 partnership mediates some biological functions[4]. Several studies showed that FGFR4 played no positive role in liver regeneration and limited hepatocarcinogenesis using FGFR4 knockout mice, suggesting a negative role of FGFR4 in tumorigenesis[27,28]. These data are consistent with the conclusion that bKlotho could suppress tumor growth. Cell cycle governs the transition from quiescence to cell proliferation, and is typically divided into four phases. The periods associated with DNA synthesis (S phase) and mitosis (M phase) are separated by gaps of varying length called G1 and G2 phase. The majority of human cancers have been MedChemExpress Alprenolol reported to have alterations in the function of cell cycle regulatory proteins[11?3]. cyclin D1 is one of the key regulatory proteins controlling the transition from G1 to S phase. We found that bKlotho could induce cell cycle arrest at the G1 to S phase transition, in association with downregulation of cyclin D1. Given that disruption of the regulatory system controlling G1 phase progression is a common event in human hepatocarcinogenesis and cyclin D1 overexpression plays a carcinogenic role in HCC[29], our data SIS-3 suggested bKlotho inhibited hepatoma cells growth by down-regulation of cyclin D1. bKlotho acts as a co-receptor and facilitates metabolic signaling by FGFs. The bKlotho-FGFR4 partnership causes a depression of Akt signaling[4]. Consistent with this, we showed that bKlotho overexpression reduced the phosphorylation of Akt and subsequent phosphorylation of GSK-3b, indicating Akt inactivation and GSK-3b activation respectively. This might contribute to cyclin D1 degradation because GSK-3b is a critical regulator of cyclin D1 expression[19?1]. Moreover, the Akt/GSK-3b signaling also plays an important role in HCC[30?2]. Thus, our data suggested the Akt/GSK-3b/cyclin D1 signaling pathway mediated the function of bKlotho in hepatoma cells proliferation and hepatocarcinogenesis. In summary, we identified that bKlotho could suppress tumor growth in HCC, and our investigation suggested that restoration^2Klotho Suppresses Tumor Growth in HCC IFigure 5. Overexpression of bKlotho suppressed tumor formation. (A, B) Subcutaneous tumor growth curve of 18325633 bKlotho-transfected Hep3B or SMMC-7721 cells in nude mice was compared with vector transfected cells. The bKlotho group showed a retarded tumor growth compared to the vector group. (C) A representative picture of tumor growth in nude mice subcutaneously inoculated with vector or bKlotho transfected hepatoma cells. The bKlotho group showed a retarded tumor growth compared to the vector group. (D) The mean tumor weights in nude mice subcutaneously inoculated with vector or bKlotho transfected hepatoma cells. (E) Western blotting analysis of bKlotho, cyclin D1, phosphorylated Akt (p-Akt), Akt, phosphorylated GSK-3b (p-GSK-3b), GSK-3b and tubulin levels in the subcutaneous Hep3B cells tumor samples. (F) Subcutaneous tumor growth curve of bKlotho-transfected Hep3B cells in nude mice was compared with bKlotho and myr-Akt co-transfected cells. The bKlotho plus myr-Akt group showed a retarded tumor growth compared to the bKlotho alone group. The data were means 6 SD of three separate experiments. * indicates p , 0.05. doi:10.1371/journal.pone.0055615.A, we also revealed that bKlotho could also reduce tumor genesis ability in vivo. These results demonstrated bKlotho has an anti-tumorigenic role in HCC. Moreover, bKlotho interacts with FGFR4 to form a complex and the bKlotho-FGFR4 partnership mediates some biological functions[4]. Several studies showed that FGFR4 played no positive role in liver regeneration and limited hepatocarcinogenesis using FGFR4 knockout mice, suggesting a negative role of FGFR4 in tumorigenesis[27,28]. These data are consistent with the conclusion that bKlotho could suppress tumor growth. Cell cycle governs the transition from quiescence to cell proliferation, and is typically divided into four phases. The periods associated with DNA synthesis (S phase) and mitosis (M phase) are separated by gaps of varying length called G1 and G2 phase. The majority of human cancers have been reported to have alterations in the function of cell cycle regulatory proteins[11?3]. cyclin D1 is one of the key regulatory proteins controlling the transition from G1 to S phase. We found that bKlotho could induce cell cycle arrest at the G1 to S phase transition, in association with downregulation of cyclin D1. Given that disruption of the regulatory system controlling G1 phase progression is a common event in human hepatocarcinogenesis and cyclin D1 overexpression plays a carcinogenic role in HCC[29], our data suggested bKlotho inhibited hepatoma cells growth by down-regulation of cyclin D1. bKlotho acts as a co-receptor and facilitates metabolic signaling by FGFs. The bKlotho-FGFR4 partnership causes a depression of Akt signaling[4]. Consistent with this, we showed that bKlotho overexpression reduced the phosphorylation of Akt and subsequent phosphorylation of GSK-3b, indicating Akt inactivation and GSK-3b activation respectively. This might contribute to cyclin D1 degradation because GSK-3b is a critical regulator of cyclin D1 expression[19?1]. Moreover, the Akt/GSK-3b signaling also plays an important role in HCC[30?2]. Thus, our data suggested the Akt/GSK-3b/cyclin D1 signaling pathway mediated the function of bKlotho in hepatoma cells proliferation and hepatocarcinogenesis. In summary, we identified that bKlotho could suppress tumor growth in HCC, and our investigation suggested that restoration^2Klotho Suppresses Tumor Growth in HCC IFigure 5. Overexpression of bKlotho suppressed tumor formation. (A, B) Subcutaneous tumor growth curve of 18325633 bKlotho-transfected Hep3B or SMMC-7721 cells in nude mice was compared with vector transfected cells. The bKlotho group showed a retarded tumor growth compared to the vector group. (C) A representative picture of tumor growth in nude mice subcutaneously inoculated with vector or bKlotho transfected hepatoma cells. The bKlotho group showed a retarded tumor growth compared to the vector group. (D) The mean tumor weights in nude mice subcutaneously inoculated with vector or bKlotho transfected hepatoma cells. (E) Western blotting analysis of bKlotho, cyclin D1, phosphorylated Akt (p-Akt), Akt, phosphorylated GSK-3b (p-GSK-3b), GSK-3b and tubulin levels in the subcutaneous Hep3B cells tumor samples. (F) Subcutaneous tumor growth curve of bKlotho-transfected Hep3B cells in nude mice was compared with bKlotho and myr-Akt co-transfected cells. The bKlotho plus myr-Akt group showed a retarded tumor growth compared to the bKlotho alone group. The data were means 6 SD of three separate experiments. * indicates p , 0.05. doi:10.1371/journal.pone.0055615.

Share this post on:

Author: DNA_ Alkylatingdna