Deleted in liver cancer 1 (DLC1) negatively regulates Rho/ROCK/MLC pathway in hepatocellular carcinoma

Carmen Chak-Lui Wong, Chun-Ming Wong, Frankie Chi-Fat Ko, Lo-Kong Chan, Yick-Pang Ching, Judy Wai-Ping Yam, Irene Oi-lin Ng, Carmen Chak-Lui Wong, Chun-Ming Wong, Frankie Chi-Fat Ko, Lo-Kong Chan, Yick-Pang Ching, Judy Wai-Ping Yam, Irene Oi-lin Ng

Abstract

Aims: Deleted in liver cancer 1 (DLC1), a member of RhoGTPase activating protein (GAP) family, is known to have suppressive activities in tumorigenicity and cancer metastasis. However, the underlying molecular mechanisms of how DLC1 suppresses cell motility have not been fully elucidated. Rho-kinase (ROCK) is an immediate down-stream effector of RhoA in mediating cellular cytoskeletal events and cell motility. In the present study, we aimed to investigate the effects of DLC1 on Rho/ROCK signaling pathway in hepatocellular carcinoma (HCC).

Methodology/principal findings: We demonstrated that DLC1 negatively regulated ROCK-dependent actomyosin contractility. From immunofluorescence study, we found that ectopic expression of DLC1 abrogated Rho/ROCK-mediated cytoskeletal reorganization including formation of stress fibers and focal adhesions. It also downregulated cortical phosphorylation of myosin light chain 2 (MLC2). These inhibitory events by DLC1 were RhoGAP-dependent, as RhoGAP-deficient mutant of DLC1 (DLC1 K714E) abolished these inhibitory events. In addition, from western study, DLC1 inhibited ROCK-related myosin light chain phosphatase targeting unit 1 (MYPT1) phosphorylation at Threonine 853. By examining cell morphology under microscope, we found that ectopic expression of dominant-active ROCK released cells from DLC1-induced cytoskeletal collapse and cell shrinkage.

Conclusion: Our data suggest that DLC1 negatively regulates Rho/ROCK/MLC2. This implicates a ROCK-mediated pathway of DLC1 in suppressing metastasis of HCC cells and enriches our understanding in the molecular mechanisms involved in the progression of hepatocellular carcinoma.

Conflict of interest statement

Competing Interests: The authors have declared that no competing interests exist.

Figures

Figure 1. DLC1 abolished ROCK-mediated stress fiber…
Figure 1. DLC1 abolished ROCK-mediated stress fiber and focal adhesion formation.
(A) ROCK inhibitor suppressed stress fiber and focal adhesion formation in HCC cells. SMMC-7721 cells were seeded onto cover-slip one day before treatment. Stress fibers were stained with phalloidin stain (red) and focal adhesions with paxillin stain (green). Stress fibers could be clearly observed as bundles stretching across the cells and focal adhesions were attached to the stress fiber bundling arrays in SMMC-7721 mock treated control. Treatment with ROCK inhibitor Y27632 at 10 µM for 60 minutes suppressed formation of stress fiber and focal adhesion network in SMMC-7721. (B) and (C) DLC1 suppressed ROCK-mediated stress fiber and focal adhesion formation in HCC cells. SMMC-7721 cells (i) and BEL7402 (ii) were transfected with myc-tagged expression plasmids, pCS2+MT, pCS2+MT/DLC1, and pCS2+MT/DLC1 K714E, and recognized by anti-myc antibodies (9E10). Stress fibers and focal adhesions were stained with phalloidin-TRITC and anti-paxillin antibody, respectively. Wild-type DLC1, but not RhoGAP-deficient mutant (DLC1 K714E), suppressed stress fibers formation and reduced the number of stress fibers-linked focal adhesions formed in SMMC-7721 cells. Percentage of different DLC1 constructs transfected cells exhibiting stress fibers or focal adhesions were presented in a bar graph accordingly. For each construct, total 67–170 transfected cells were counted under microscope. Error bars represent standard deviation (SD) of data obtained from two independent experiments.
Figure 2. ROCK-mediated cortical phosphorylation of MLC2.
Figure 2. ROCK-mediated cortical phosphorylation of MLC2.
(A) BEL4702 HCC cell line treated with mock control or ROCK inhibitor (Y27632) was probed with mouse monoclonal antibody against phospho-MLC2 (Ser 19). Phospho-MLC2 (green) was mainly detected at the cell cortex of BEL7402 cells (as indicated by the arrows) and this cortical staining pattern was abolished by treatment with ROCK inhibitor Y27632. (B) BEL7402 cells were transfected with myc-tagged dominant negative ROCK or dominant active ROCK constructs and detected with rabbit polyclonal antibody against myc-epitope (A14) (Red). Dominant active (DA) ROCK caused intense phosphorylation of MLC2 (Ser 19) at the actin bundles (Precentage of cells exhibiting this phenomenon: 94.0% of the DA ROCK-transfected cells compared to 0% of the vector transfected cells). Arrows point at the actin bundles of the dominant active ROCK transfected cell where phosphorylation of MLC2 (Ser 19) was enhanced. Dominant negative (DN) ROCK suppressed cortical phosphorylation of MLC2 (Ser 19) (Percentage of cells exhibiting this phenomenon: 87.5% of the DN ROCK-transfected cells compared to 7.3% of the vector transfected cells). Arrows point at the cell cortex of the dominant negative ROCK transfected cell where phosphorylation of MLC2 (Ser 19) was lost. As shown, a tight regulation and an optimal level of ROCK activity were required to maintain cortical phosphorylation of MLC2 (Ser 19) in BEL7402 cells.
Figure 3. DLC1 expression was associated with…
Figure 3. DLC1 expression was associated with cortical phosphorylation of MLC2.
(A) DLC1 mRNA and protein expression in HCC cell lines. (B) and (C) DLC1 expression was associated with phospho-MLC2 staining pattern. Representative pictures from DLC1-positive and DLC1-negative cells, respectively. DLC1-positive Hep3B and HepG2, displayed diffuse phosphorylation of MLC2, whereas DLC1 non-expressing BEL7402 and HeLa, displayed pronounced cortical phosphorylation of MLC2 at cell cortex.
Figure 4. DLC1 RhoGAP was responsible for…
Figure 4. DLC1 RhoGAP was responsible for regulating cortical phosphorylation of MLC2.
(A) pCS2+MT vector alone, pCS2+MT DLC1, and pCS2+MT DLC1 K714E, respectively, were transfected into BEL7402 cells. Recognition of transfected cells was done by probing cells with c-myc (A14) rabbit antibody followed by staining with anti-rabbit antibody conjugated with Texas Red. Cells transfected with pCS2+MT vector alone displayed pronounced cortical phosphorylation of MLC2 as indicated by the arrows. Cells transfected with DLC1 displayed loss of cortical phosphorylation of MLC2. Cells transfected with DLC1 K714E, the RhoGAP-deficient mutant, still displayed pronounced cortical phosphorylation of MLC2 as indicated by the arrows. (B) Percentage of different DLC1 constructs exhibiting cortical phosphorylation of MLC2 at Ser 19 was calculated and presented in a bar graph. For each construct, 80–100 transfected cells were counted and cortical MLC2 phosphorylation was recorded. Error bars represent standard deviation (SD) of data obtained from three independent experiments.
Figure 5. DLC1 and ROCK regulated MYPT…
Figure 5. DLC1 and ROCK regulated MYPT phosphorylation.
(A) ROCK inhibitor Y27632 suppressed MYPT phosphorylation at Thr 853 in all HCC cell lines. (B) DLC1 suppressed MYPT phosphorylation at Thr 853 in 293 T cells. Band intensity was analyzed by AlphaEaseFC™ and percentage was calculated from comparison with its according control.
Figure 6. ROCK inhibitor suppressed HCC cell…
Figure 6. ROCK inhibitor suppressed HCC cell migration.
ROCK inhibitor Y27632 suppressed HCC cell migration. Y27632 decreased number of migrated (A) SMMC-7721 cells (P<0.0001, t-test) and (B) BEL7402 cells (P<0.0001, t-test). Error bars represent standard deviation (SD) of data obtained from three microscopic fields. Experiments have been repeated three times. Cell proliferation assay was shown on the right to compare growth rate of mock control cells and Y27632 treated cells.
Figure 7. ROCK reversed the cell morphological…
Figure 7. ROCK reversed the cell morphological alteration of DLC1.
COS7 cells were co-transfected with (A) DLC1 and ROCK or (B) DLC1ΔSAM and ROCK, respectively. Cells transfected with pCS2MT DLC1 or pCS2MT DLC1ΔSAM appeared red, while cells transfected with pEGFP or pEGFP ROCK appeared green. DLC1-induced cell cytoskeletal collapse or cell shrinkage (A-i), which was further enhanced and clearly demonstrated by SAM-deleted construct, pCS2+MT DLC1ΔSAM (B-i). ROCK was able to restore cells from DLC1-induced cell shrinkage (A-ii) and even from DLC1ΔSAM-induced intense cell shrinkage (B-ii). (C) Co-transfected cells were counted and their morphologies recorded. The number of co-transfected cells displaying observable cell shrinkage (cell collapse) was divided by total number of co-transfected cells counted, to calculate the percentage of cells in displaying shrinkage as shown in the bar graph. For each column, 150–200 co-transfected cells were counted. Error bars represent standard deviation (SD) of data obtained from three independent experiments.
Figure 8. Model of DLC1 induced cytoskeletal…
Figure 8. Model of DLC1 induced cytoskeletal collapse.
(A) Stress fibers were connected to focal adhesions forming a network. Stress fibers stretched across the cell to provide the cell an intact morphology. (B) DLC1 RhoGAP or loss of ROCK activity induced the loss of stress fibers and focal adhesions. A few focal adhesions and bundles of stress fibers remained. (C) A prolonged or severe loss of stress fiber and focal adhesion network caused by DLC1 RhoGAP or suppression of ROCK activity would result in intensive cytoskeletal collapse. All the stress fibers and focal adhesions were abolished.

References

    1. Ananthakrishnan R, Ehrlicher A. The forces behind cell movement. Int J Biol Sci. 2007;3:303–317.
    1. Yamazaki D, Kurisu S, Takenawa T. Regulation of cancer cell motility through actin reorganization. Cancer Sci. 2005;96:379–386.
    1. Yuan BZ, Miller MJ, Keck CL, Zimonjic DB, Thorgeirsson SS, et al. Cloning, characterization, and chromosomal localization of a gene frequently deleted in human liver cancer (DLC-1) homologous to rat RhoGAP. Cancer Res. 1998;58:2196–2199.
    1. Wong CM, Yam JW, Ching YP, Yau TO, Leung TH, et al. Rho GTPase-activating protein deleted in liver cancer suppresses cell proliferation and invasion in hepatocellular carcinoma. Cancer Res. 2005;65:8861–8868.
    1. Xue W, Krasnitz A, Lucito R, Sordella R, Vanaelst L, et al. DLC1 is a chromosome 8p tumor suppressor whose loss promotes hepatocellular carcinoma. Genes Dev. 2008;22:1439–1444.
    1. Yuan BZ, Zhou X, Durkin ME, Zimonjic DB, Gumundsdottir K, et al. DLC-1 gene inhibits human breast cancer cell growth and in vivo tumorigenicity. Oncogene. 2003;22:445–450.
    1. Ng IO, Liang ZD, Cao L, Lee TK. DLC-1 is deleted in primary hepatocellular carcinoma and exerts inhibitory effects on the proliferation of hepatoma cell lines with deleted DLC-1. Cancer Res. 2000;60:6581–6584.
    1. Wong CM, Lee JM, Ching YP, Jin DY, Ng IO. Genetic and epigenetic alterations of DLC-1 gene in hepatocellular carcinoma. Cancer Res. 2003;63:7646–7651.
    1. Yuan BZ, Durkin ME, Popescu NC. Promoter hypermethylation of DLC-1, a candidate tumor suppressor gene, in several common human cancers. Cancer Genet Cytogenet. 2003;140:113–117.
    1. Guan M, Zhou X, Soulitzis N, Spandidos DA, Popescu NC. Aberrant methylation and deacetylation of deleted in liver cancer-1 gene in prostate cancer: potential clinical applications. Clin Cancer Res. 2006;12:1412–1419.
    1. Plaumann M, Seitz S, Frege R, Estevez-Schwarz L, Scherneck S. Analysis of DLC-1 expression in human breast cancer. J Cancer Res Clin Oncol. 2003;129:349–354.
    1. Seng TJ, Low JS, Li H, Cui Y, Goh HK, et al. The major 8p22 tumor suppressor DLC1 is frequently silenced by methylation in both endemic and sporadic nasopharyngeal, esophageal, and cervical carcinomas, and inhibits tumor cell colony formation. Oncogene. 2007;26:934–944.
    1. Ullmannova V, Popescu NC. Expression profile of the tumor suppressor genes DLC-1 and DLC-2 in solid tumors. Int J Oncol. 2006;29:1127–1132.
    1. Yuan BZ, Jefferson AM, Baldwin KT, Thorgeirsson SS, Popescu NC, et al. DLC-1 operates as a tumor suppressor gene in human non-small cell lung carcinomas. Oncogene. 2004;23:1405–1411.
    1. Goodison S, Yuan J, Sloan D, Kim R, Li C, et al. The RhoGAP protein DLC-1 functions as a metastasis suppressor in breast cancer cells. Cancer Res. 2005;65:6042–6053.
    1. Song LJ, Ye SL, Wang KF, Weng YQ, Liang CM, et al. [Relationship between DLC-1 expressions and metastasis in hepatocellular carcinoma]. Zhonghua Gan Zang Bing Za Zhi. 2005;13:428–431.
    1. Kim TY, Lee JW, Kim HP, Jong HS, Jung M, et al. DLC-1, a GTPase-activating protein for Rho, is associated with cell proliferation, morphology, and migration in human hepatocellular carcinoma. Biochem Biophys Res Commun. 2007;355:72–77.
    1. Qian X, Li G, Asmussen HK, Asnaghi L, Vass WC, et al. Oncogenic inhibition by a deleted in liver cancer gene requires cooperation between tensin binding and Rho-specific GTPase-activating protein activities. Proc Natl Acad Sci U S A. 2007;104:9012–9017.
    1. Syed V, Mukherjee K, Lyons-Weiler J, Lau KM, Mashima T, et al. Identification of ATF-3, caveolin-1, DLC-1, and NM23-H2 as putative antitumorigenic, progesterone-regulated genes for ovarian cancer cells by gene profiling. Oncogene. 2005;24:1774–1787.
    1. Zhou X, Thorgeirsson SS, Popescu NC. Restoration of DLC-1 gene expression induces apoptosis and inhibits both cell growth and tumorigenicity in human hepatocellular carcinoma cells. Oncogene. 2004;23:1308–1313.
    1. Healy KD, Hodgson L, Kim TY, Shutes A, Maddileti S, et al. DLC-1 suppresses non-small cell lung cancer growth and invasion by RhoGAP-dependent and independent mechanisms. Mol Carcinog. 2008;47:326–337.
    1. Hall A. Signal transduction through small GTPases–a tale of two GAPs. Cell. 1992;69:389–391.
    1. Leung T, Chen XQ, Manser E, Lim L. The p160 RhoA-binding kinase ROK alpha is a member of a kinase family and is involved in the reorganization of the cytoskeleton. Mol Cell Biol. 1996;16:5313–5327.
    1. Matsui T, Amano M, Yamamoto T, Chihara K, Nakafuku M, et al. Rho-associated kinase, a novel serine/threonine kinase, as a putative target for small GTP binding protein Rho. Embo J. 1996;15:2208–2216.
    1. Chen XQ, Tan I, Ng CH, Hall C, Lim L, et al. Characterization of RhoA-binding kinase ROKalpha implication of the pleckstrin homology domain in ROKalpha function using region-specific antibodies. J Biol Chem. 2002;277:12680–12688.
    1. Amano M, Chihara K, Nakamura N, Kaneko T, Matsuura Y, et al. The COOH terminus of Rho-kinase negatively regulates rho-kinase activity. J Biol Chem. 1999;274:32418–32424.
    1. Ishizaki T, Naito M, Fujisawa K, Maekawa M, Watanabe N, et al. p160ROCK, a Rho-associated coiled-coil forming protein kinase, works downstream of Rho and induces focal adhesions. FEBS Lett. 1997;404:118–124.
    1. Li Z, Dong X, Wang Z, Liu W, Deng N, et al. Regulation of PTEN by Rho small GTPases. Nat Cell Biol. 2005;7:399–404.
    1. Alblas J, Ulfman L, Hordijk P, Koenderman L. Activation of Rhoa and ROCK are essential for detachment of migrating leukocytes. Mol Biol Cell. 2001;12:2137–2145.
    1. Worthylake RA, Lemoine S, Watson JM, Burridge K. RhoA is required for monocyte tail retraction during transendothelial migration. J Cell Biol. 2001;154:147–160.
    1. Somlyo AV, Bradshaw D, Ramos S, Murphy C, Myers CE, et al. Rho-kinase inhibitor retards migration and in vivo dissemination of human prostate cancer cells. Biochem Biophys Res Commun. 2000;269:652–659.
    1. Pellegrin S, Mellor H. Actin stress fibres. J Cell Sci. 2007;120:3491–3499.
    1. Riento K, Ridley AJ. Rocks: multifunctional kinases in cell behaviour. Nat Rev Mol Cell Biol. 2003;4:446–456.
    1. Hagerty L, Weitzel DH, Chambers J, Fortner CN, Brush MH, et al. ROCK1 phosphorylates and activates zipper-interacting protein kinase. J Biol Chem. 2007;282:4884–4893.
    1. Takamura M, Sakamoto M, Genda T, Ichida T, Asakura H, et al. Inhibition of intrahepatic metastasis of human hepatocellular carcinoma by Rho-associated protein kinase inhibitor Y-27632. Hepatology. 2001;33:577–581.
    1. Wilkinson S, Paterson HF, Marshall CJ. Cdc42-MRCK and Rho-ROCK signalling cooperate in myosin phosphorylation and cell invasion. Nat Cell Biol. 2005;7:255–261.
    1. Yoshioka K, Matsumura F, Akedo H, Itoh K. Small GTP-binding protein Rho stimulates the actomyosin system, leading to invasion of tumor cells. J Biol Chem. 1998;273:5146–5154.
    1. Yoshioka K, Nakamori S, Itoh K. Overexpression of small GTP-binding protein RhoA promotes invasion of tumor cells. Cancer Res. 1999;59:2004–2010.
    1. Itoh K, Yoshioka K, Akedo H, Uehata M, Ishizaki T, et al. An essential part for Rho-associated kinase in the transcellular invasion of tumor cells. Nat Med. 1999;5:221–225.
    1. Genda T, Sakamoto M, Ichida T, Asakura H, Kojiro M, et al. Cell motility mediated by rho and Rho-associated protein kinase plays a critical role in intrahepatic metastasis of human hepatocellular carcinoma. Hepatology. 1999;30:1027–1036.
    1. Kamai T, Tsujii T, Arai K, Takagi K, Asami H, et al. Significant association of Rho/ROCK pathway with invasion and metastasis of bladder cancer. Clin Cancer Res. 2003;9:2632–2641.
    1. Ueda K, Murata-Hori M, Tatsuka M, Hosoya H. Rho-kinase contributes to diphosphorylation of myosin II regulatory light chain in nonmuscle cells. Oncogene. 2002;21:5852–5860.
    1. Amano M, Ito M, Kimura K, Fukata Y, Chihara K, et al. Phosphorylation and activation of myosin by Rho-associated kinase (Rho-kinase). J Biol Chem. 1996;271:20246–20249.
    1. Totsukawa G, Yamakita Y, Yamashiro S, Hartshorne DJ, Sasaki Y, et al. Distinct roles of ROCK (Rho-kinase) and MLCK in spatial regulation of MLC phosphorylation for assembly of stress fibers and focal adhesions in 3T3 fibroblasts. J Cell Biol. 2000;150:797–806.
    1. Sturge J, Wienke D, Isacke CM. Endosomes generate localized Rho-ROCK-MLC2-based contractile signals via Endo180 to promote adhesion disassembly. J Cell Biol. 2006;175:337–347.
    1. Katoh K, Kano Y, Amano M, Onishi H, Kaibuchi K, et al. Rho-kinase–mediated contraction of isolated stress fibers. J Cell Biol. 2001;153:569–584.
    1. Lavelin I, Geiger B. Characterization of a novel GTPase-activating protein associated with focal adhesions and the actin cytoskeleton. J Biol Chem. 2005;280:7178–7185.
    1. Barberis D, Casazza A, Sordella R, Corso S, Artigiani S, et al. p190 Rho-GTPase activating protein associates with plexins and it is required for semaphorin signalling. J Cell Sci. 2005;118:4689–4700.
    1. Kawai K, Yamaga M, Iwamae Y, Kiyota M, Kamata H, et al. A PLCdelta1-binding protein, p122RhoGAP, is localized in focal adhesions. Biochem Soc Trans. 2004;32:1107–1109.
    1. Kaverina I, Krylyshkina O, Small JV. Regulation of substrate adhesion dynamics during cell motility. Int J Biochem Cell Biol. 2002;34:746–761.
    1. Hall A. Rho GTPases and the actin cytoskeleton. Science. 1998;279:509–514.
    1. Peacock JG, Miller AL, Bradley WD, Rodriguez OC, Webb DJ, et al. The Abl-related gene tyrosine kinase acts through p190RhoGAP to inhibit actomyosin contractility and regulate focal adhesion dynamics upon adhesion to fibronectin. Mol Biol Cell. 2007;18:3860–3872.
    1. Yam JW, Ko FC, Chan CY, Jin DY, Ng IO. Interaction of deleted in liver cancer 1 with tensin2 in caveolae and implications in tumor suppression. Cancer Res. 2006;66:8367–8372.
    1. Liao YC, Si L, deVere White RW, Lo SH. The phosphotyrosine-independent interaction of DLC-1 and the SH2 domain of cten regulates focal adhesion localization and growth suppression activity of DLC-1. J Cell Biol. 2007;176:43–49.
    1. Sahai E, Marshall CJ. Differing modes of tumour cell invasion have distinct requirements for Rho/ROCK signalling and extracellular proteolysis. Nat Cell Biol. 2003;5:711–719.
    1. Nakagawa H, Yoshioka K, Miyahara E, Fukushima Y, Tamura M, et al. Intrathecal administration of Y-27632, a specific rho-associated kinase inhibitor, for rat neoplastic meningitis. Mol Cancer Res. 2005;3:425–433.
    1. Vicari RM, Chaitman B, Keefe D, Smith WB, Chrysant SG, et al. Efficacy and safety of fasudil in patients with stable angina: a double-blind, placebo-controlled, phase 2 trial. J Am Coll Cardiol. 2005;46:1803–1811.

Source: PubMed

3
Se inscrever