SOX18 Is a Novel Target Gene of Hedgehog Signaling in Cervical Carcinoma Cell Lines

Isidora Petrovic, Milena Milivojevic, Jelena Popovic, Marija Schwirtlich, Branislava Rankovic, Milena Stevanovic, Isidora Petrovic, Milena Milivojevic, Jelena Popovic, Marija Schwirtlich, Branislava Rankovic, Milena Stevanovic

Abstract

Although there is much evidence showing functional relationship between Hedgehog pathway, in particular Sonic hedgehog, and SOX transcription factors during embryonic development, scarce data are available regarding their crosstalk in cancer cells. SOX18 protein plays an important role in promoting tumor angiogenesis and therefore emerged as a promising potential target in antiangiogenic tumor therapy. Recently it became evident that expression of SOX18 gene in tumors is not restricted to endothelium of accompanying blood and lymphatic vessels, but in tumor cells as well.In this paper we have identified human SOX18 gene as a novel target gene of Hedgehog signaling in cervical carcinoma cell lines. We have presented data showing that expression of SOX18 gene is regulated by GLI1 and GLI2 transcription factors, final effectors of Hedgehog signaling, and that modulation of Hedgehog signaling activity in considerably influence SOX18 expression. We consider important that Hedgehog pathway inhibitors reduced SOX18 expression, thus showing, for the first time, possibility for manipulationwith SOX18 gene expression. In addition, we analyzed the role of SOX18 in malignant potential of cervical carcinoma cell line, and showed that its overexpression has no influence on cells proliferation and viability, but substantially promotes migration and invasion of cells in vitro. Pro-migratory effect of SOX18 suggests its role in promoting malignant spreading, possibly in response to Hedgehog activation.

Conflict of interest statement

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

Figures

Fig 1. The role of GLI transcription…
Fig 1. The role of GLI transcription factors in the regulation of SOX18 promoter activity and SOX18 endogenous expression.
a) Expression analysis of selected HH signaling components in HeLa, SiHa and Ca Ski carcinoma cell lines.M-DNA ladder.b) Functional analysis of each GLI transcription factor overexpression on SOX18 promoter activity. Schematic illustration of putative biding sites for GLI transcription factors within SOX18 optimal promoter region represented by promoter construct 892pCAT6 is presented on upper panel. Normalized CAT activities were calculated as percentages of the corresponding reporter construct activity in cells co-transfected with empty pcDNA3.1 vector (which was set as 100%). Relative CAT activities were presented as the means ± SEM of at least four independent experiments. P values were calculated using Student’s t-test, *p ≤ 0.05, **p ≤ 0.01c)The effect of GLI’s overexpression on SOX18 gene expression detected by qRT-PCR. Relative SOX18 expression was presented as percentage of SOX18 expression in mock transfected cells that was set as 100%. Results were presented as the means ± SEM of at least three independent experiments performed in triplicates. P values were calculated using Student’s t-test, *p ≤ 0.05, **p ≤ 0.01.
Fig 2. The effect of GLI’s overexpression…
Fig 2. The effect of GLI’s overexpression on SOX18 protein level in cervical carcinoma cell lines.
a) The effect of GLI’s overexpression on SOX18 protein leveldetected by Western blot. One representative blot was presented, and quantification of protein level was presented as histogram chart.α-tubuline was used as a loading control. The relative SOX18 protein level in HeLa, SiHa and Ca Ski cells upon transfection with GLI1-3 was calculated as a percentage of SOX18 level in mock transfected cells which was set as 100%. Data of three independent experiments are presented at histograms as the means ± SEM. Values of p≤0.05 are marked by *.b)The effect of GLI’s overexpression on SOX18 protein leveldetected by immunocytochemistry. Cells were cotransfected with EGFP-C1 (that was used as a marker of transfected cells) and eiherpcDNA-mock transfection (A-C), GLI1(D-F), GLI2 (G-I), or GLI3(J-L). Boxed regions in A-L are enlarged in the same figures. Cell nuclei were counterstained with DAPI (A, D, G and J). Scale bars: 50 μm.
Fig 3. EMSA with six different DNA…
Fig 3. EMSA with six different DNA probes deriving from SOX18 optimal promoter.
Upper panel represents schematic illustration of putative GLI binding sites. Positions relative to tsp are indicated above scheme, and relative positions of corresponding probes are presented by lines and names below. Lower panel represents EMSA reactions; NE HeLa-HeLa nuclear extracts; Unlab.probe- unlabeled corresponding probe in 100-fold molar excess; Cons.GLI- unlabeled oligonucleotide probe with GLI consensus binding site in 100-fold molar excess. Completely faded complexes in competition reaction with GLI consensus probe are marked with black arrows, and partially faded with red arrows.
Fig 4. EMSA with “enriched” proteins and…
Fig 4. EMSA with “enriched” proteins and supershift assay.
a) EMSA with whole cell lysates prepared from HeLa cells transfected with pcDNA3.1 empty vector (WCL HeLa) or cells transfected with either GLI1 (WCL HeLa+GLI1) or GLI2 expression vector construct (WCL HeLa+GLI2). b)Supershift assay with anti-GLI1 antibody (GLI1Ab). “Enriched” complexes are marked by asterisks,supershifts i.e. fading of complexes are marked by arrows and ss.
Fig 5. The effect of HH pathway…
Fig 5. The effect of HH pathway inhibition on proliferation, viability, migration and SOX18 expression in HeLa cells.
a) The inhibitory effect of cyclopamine on GLI1 and PTCH expression. b) Proliferation curve of HeLa cells. Cells were treated with 10μM cyclopamine or tomatidine as a negative control, and counted after 1, 3 and 5 days of continuous treatment. Results were presented as the means ± SEM of at least three independent experiments. c) MTT viability assay performed after 1, 3 and 4 days of treatment with 10 μM cyclopamine or tomatidine. Relative cell viability was calculated as a percentage of HeLa cells viability after tomatidine treatment that was set as 100%. Results were presented as the means ± SEM of at least three independent experiments. P values were calculated using Student’s t-test, *p ≤ 0.05, **p ≤ 0.01.d) The effect of cyclopamine on cell’s migration, wound-scratch migration assay. Cells migration was quantified 24 h after scratching in constant presence of cyclopamine or tomatidine,by measuring the difference in gap closure where gap wide at 0 h was set as 100%. Results were presented as the means ± SEM of at least three independent experiments. e) Relative SOX18 expression after cyclopamine treatment detected by qRT-PCR. Relative SOX18 expression was presented as percentage of SOX18 expression in cells treated with tomatidine that was set as 100%. Results were presented as the means ± SEM of at least three independent experiments performed in triplicates. P values were calculated using Student’s t-test, *p ≤ 0.05, **p ≤ 0.01.f) The effect of cyclopamine on SOX18 protein level. Proteins were isolated after three independent treatments together with adequate controls, followed by Western blot. One representative blot was presented. α-tubuline was used as a loading control. The relative SOX18 protein level in HeLa cells upon treatment with cyclopamine was calculated as a percentage of SOX18 level in cells trated with tomatidine which was set as 100%. Data of three independent experiments are presented at histograms as the means ± SEM. Values of p≤0.01 are marked by **.g) The effect of GANT61 on SOX18 protein level. Proteins were isolated after three independent treatments together with adequate controls, followed by Western blot. One representative blot was presented. α-tubuline was used as a loading control.
Fig 6. The effect of HH pathway…
Fig 6. The effect of HH pathway activation on proliferation, viability, migration and SOX18 expression in HeLa cells.
a) Stimulatory effect of purmorphamine on GLI1 and PTCH expression. b) Proliferation curve of HeLa cells. Cells were treated with 10μM purmorphamine or DMSO as a control, and counted after 1 and 3 days. Results were presented as the means ± SEM of at least three independent experiments. c) MTT viability assay performed after 1 and 3 days of treatment with 10μM purmorphamine or DMSO. Relative cell viability was calculated as a percentage of HeLa cells viability after DMSO treatment that was set as 100%. Results were presented as the means ± SEM of at least five independent experiments. P values were calculated using Student’s t-test, *p ≤ 0.05, **p ≤ 0.01.d) The effect of purmorphamine treatment on cell migration, wound-scratch migration assay. Cells migration was quantified 24 h after scratching in constant presence of purmorphamine or DMSO, by measuring the difference in gap closure where gap wide at 0 h was set as 100%. Results were presented as the means ± SEM of at least three independent experiments. e) Relative SOX18 expression after purmorphamine treatment detected by qRT-PCR. Relative SOX18 expression was presented as percentage of SOX18 expression in cells treated with DMSO that was set as 100%. Results were presented as the means ± SEM of at least three independent experiments performed in triplicates. P values were calculated using Student’s t-test, *p ≤ 0.05, **p ≤ 0.01.f) The effect of purmorphamine treatment on SOX18 protein leveldetected by immunocytochemistry. Cell nuclei were counterstained with DAPI (A, and C). Scale bars: 50 μm.
Fig 7. The role of SOX18 in…
Fig 7. The role of SOX18 in the regulation of HeLa cells proliferation, viability and migration.
a) Proliferation chart. HeLa cells were seeded day prior to transient transfection with empty pCI vector, wt or DN SOX18 expression constructs and counted 24 h after transfection. Results were presented as the means ± SEM of at least four independent experiments. b) MTT viability assay. HeLa cells were transiently transfected while seeded in microplate and MTT test was performed 24 h later. Relative cell viability was calculated as a percentage of HeLa cells viability without transfection that was set as 100% Results were presented as the means ± SEM of at least four independent experiments. c)Semiquantitative RT-PCR. Cells were transiently transfected with empty pCI vector, wt or DN SOX18 expression constructs followed by RNA isolation and RT-PCR analysis for the expression level of cyclin D1. The level of wt or DN SOX18 transcription upon transfection was also evaluated. d) The effect of wt or DN SOX18 overexpression on HeLa cell’s migration, wound-scratch migration assay. Representative images of cells migration were presented at left panel. Graphs presented at right panels quantify the migration of transfected cells 24 h after scratching. The changes in migration distance were quantified by measuring the difference in gap closure where gap wide at 0 h was set as 100%. Results were presented as the means ± SEM of at least three independent experiments. The differences in number of single cells that migrated into the empty area was measured by counting the number of single cells in empty scratched area in two different fields in at least three independent experiments and presented as scatter chart. P values were calculated using Student’s t-test, *p ≤ 0.05, **p ≤ 0.01.e) Transwell migration and invasion assays on HeLa cells transfected with empty pCI vector, wt SOX18 or DN SOX18. Representative images of transwell migration/invasion assays were presented.The relative change in cells migration/invasion was calculated as a percentage of HeLa cells migration/invasion in mock transfection that was set as 100%. Cells were counted from five fields and averages were calculated. Results were presented as the means ± SEM of at least three independent experiments performed in duplicates and P values were calculated using Student’s t-test, *p ≤ 0.05, **p ≤ 0.01.f) The influence of DN SOX18 overexpression on GLI1/GLI2-mediated HeLa cell’s migration, wound-scratch migration assay. Graph quantifies the migration of transfected cells 24 h after scratching. Table describes combination of expression vectors used in each experiment. The changes in migration distance were quantified by measuring the difference in gap closure where gap wide at 0 h was set as 100%. Results were presented as the means ± SEM of at least three independent experiments. P values were calculated using Student’s t-test comparing group 1 (cells transfected with GLI1 or GLI2) with group 2 (cells co-transfected with GLI1 or GLI2 together with DN SOX18);*p ≤ 0.05, **p ≤ 0.01.
Fig 8. The role of SOX18 in…
Fig 8. The role of SOX18 in the regulation of GLI1-3 and PTCHexpression.
HeLa cells were transiently transfected with either empty pCI vector or wtSOX18 or DNSOX18 and the effect of their overexpression on GLI1-3 and PTCH genes expression was analyzed by qRT-PCR. Relative gene expression was presented as percentage of gene expression in cells transfected with empty, pCI vector that was set as 100%. Results were presented as the means ± SEM of at least three independent experiments performed in triplicates.P values were calculated using Student’s t-test, *p ≤ 0.05, **p ≤ 0.01.

References

    1. Ingham PW, McMahon AP (2001) Hedgehog signaling in animal development: paradigms and principles. Genes Dev. 15: 3059–87.
    1. Stone DM, Hynes M, Armanini M, Swanson TA, Gu Q, Johnson RL, et al. (1996) The tumour-suppressor gene patched encodes a candidate receptor for Sonic hedgehog. Nature. 384: 129–34.
    1. Villavicencio EH, Walterhouse DO, Iannaccone PM (2000) The sonic hedgehog-patched-gli pathway in human development and disease. Am J Hum Genet. 67: 1047–54.
    1. Hooper JE, Scott MP (2005) Communicating with Hedgehogs. Nat Rev Mol Cell Biol. 6: 306–17.
    1. Jiang J, Hui CC (2008) Hedgehog signaling in development and cancer. Dev Cell. 15: 801–12.
    1. Mullor JL, Sanchez P, Ruiz i Altaba A (2002) Pathways and consequences: Hedgehog signaling in human disease. Trends Cell Biol. 12: 562–9.
    1. Pasca di Magliano M, Hebrok M (2003) Hedgehog signalling in cancer formation and maintenance. Nat Rev Cancer. 3: 903–11.
    1. Lauth M, Toftgard R (2007) Non-canonical activation of GLI transcription factors: implications for targeted anti-cancer therapy. Cell Cycle. 6: 2458–63.
    1. Scales SJ, de Sauvage FJ (2009) Mechanisms of Hedgehog pathway activation in cancer and implications for therapy. Trends Pharmacol Sci. 30: 303–12.
    1. Domingo-Domenech J, Vidal SJ, Rodriguez-Bravo V, Castillo-Martin M, Quinn SA, Rodriguez-Barrueco R, et al. (2012) Suppression of acquired docetaxel resistance in prostate cancer through depletion of notch- and hedgehog-dependent tumor-initiating cells. Cancer Cell. 22: 373–88.
    1. Gu D, Liu H, Su GH, Zhang X, Chin-Sinex H, Hanenberg H, et al. (2013) Combining hedgehog signaling inhibition with focal irradiation on reduction of pancreatic cancer metastasis. Mol Cancer Ther. 12: 1038–48.
    1. Lin SH, George TJ, Ben-Josef E, Bradley J, Choe KS, Edelman MJ, et al. (2013) Opportunities and challenges in the era of molecularly targeted agents and radiation therapy. J Natl Cancer Inst. 105: 686–93.
    1. Ramaswamy B, Lu Y, Teng KY, Nuovo G, Li X, Shapiro CL, et al. (2012) Hedgehog signaling is a novel therapeutic target in tamoxifen-resistant breast cancer aberrantly activated by PI3K/AKT pathway. Cancer Res. 72: 5048–59.
    1. Pevny LH, Lovell-Badge R (1997) Sox genes find their feet. Curr Opin Genet Dev. 7: 338–44.
    1. Wegner M (1999) From head to toes: the multiple facets of Sox proteins. Nucleic Acids Res. 27: 1409–20.
    1. Cermenati S, Moleri S, Cimbro S, Corti P, Del Giacco L, Amodeo R, et al. (2008) Sox18 and Sox7 play redundant roles in vascular development. Blood. 111: 2657–66.
    1. Matsui T, Kanai-Azuma M, Hara K, Matoba S, Hiramatsu R, Kawakami H, et al. (2006) Redundant roles of Sox17 and Sox18 in postnatal angiogenesis in mice. J Cell Sci. 119: 3513–26.
    1. Darby IA, Bisucci T, Raghoenath S, Olsson J, Muscat GE, Koopman P (2001) Sox18 is transiently expressed during angiogenesis in granulation tissue of skin wounds with an identical expression pattern to Flk-1 mRNA. Lab Invest. 81: 937–43.
    1. Young N, Hahn CN, Poh A, Dong C, Wilhelm D, Olsson J, et al. (2006) Effect of disrupted SOX18 transcription factor function on tumor growth, vascularization, and endothelial development. J Natl Cancer Inst. 98: 1060–7.
    1. Pula B, Olbromski M, Wojnar A, Gomulkiewicz A, Witkiewicz W, Ugorski M, et al. (2013) Impact of SOX18 expression in cancer cells and vessels on the outcome of invasive ductal breast carcinoma. Cell Oncol (Dordr). 36: 469–83.
    1. Hargrave M, Karunaratne A, Cox L, Wood S, Koopman P, Yamada T (2000) The HMG box transcription factor gene Sox14 marks a novel subset of ventral interneurons and is regulated by sonic hedgehog. Dev Biol. 219: 142–53.
    1. Scotting PJ, Rex M (1996) Transcription factors in early development of the central nervous system. Neuropathol Appl Neurobiol. 22: 469–81.
    1. Milivojevic M, Petrovic I, Kovacevic-Grujicic N, Popovic J, Mojsin M, Stevanovic M (2013) Construction and functional analysis of novel dominant-negative mutant of human SOX18 protein. Biochemistry (Mosc). 78: 1287–92.
    1. Petrovic I, Kovacevic-Grujicic N, Stevanovic M (2010) Early growth response protein 1 acts as an activator of SOX18 promoter. Exp Mol Med. 42: 132–42.
    1. Kinzler KW, Vogelstein B (1990) The GLI gene encodes a nuclear protein which binds specific sequences in the human genome. Mol Cell Biol. 10: 634–42.
    1. Regl G, Neill GW, Eichberger T, Kasper M, Ikram MS, Koller J, et al. (2002) Human GLI2 and GLI1 are part of a positive feedback mechanism in Basal Cell Carcinoma. Oncogene. 21: 5529–39.
    1. Kovacevic Grujicic N, Mojsin M, Krstic A, Stevanovic M (2005) Functional characterization of the human SOX3 promoter: identification of transcription factors implicated in basal promoter activity. Gene. 344: 287–97.
    1. Popovic J, Stanisavljevic D, Schwirtlich M, Klajn A, Marjanovic J, Stevanovic M (2014) Expression analysis of SOX14 during retinoic acid induced neural differentiation of embryonal carcinoma cells and assessment of the effect of its ectopic expression on SOXB members in HeLa cells. PLoS One. 9: e91852.
    1. Dignam JD, Lebovitz RM, Roeder RG (1983) Accurate transcription initiation by RNA polymerase II in a soluble extract from isolated mammalian nuclei. Nucleic Acids Res. 11: 1475–89.
    1. Xuan YH, Jung HS, Choi YL, Shin YK, Kim HJ, Kim KH, et al. (2006) Enhanced expression of hedgehog signaling molecules in squamous cell carcinoma of uterine cervix and its precursor lesions. Mod Pathol. 19: 1139–47.
    1. Ikram MS, Neill GW, Regl G, Eichberger T, Frischauf AM, Aberger F, et al. (2004) GLI2 is expressed in normal human epidermis and BCC and induces GLI1 expression by binding to its promoter. J Invest Dermatol. 122: 1503–9.
    1. Eichberger T, Regl G, Ikram MS, Neill GW, Philpott MP, Aberger F, et al. (2004) FOXE1, a new transcriptional target of GLI2 is expressed in human epidermis and basal cell carcinoma. J Invest Dermatol. 122: 1180–7.
    1. Shevde LA, Samant RS (2014) Nonclassical hedgehog-GLI signaling and its clinical implications. Int J Cancer. 135: 1–6.
    1. Cooper MK, Porter JA, Young KE, Beachy PA (1998) Teratogen-mediated inhibition of target tissue response to Shh signaling. Science. 280: 1603–7.
    1. Incardona JP, Gaffield W, Kapur RP, Roelink H (1998) The teratogenic Veratrum alkaloid cyclopamine inhibits sonic hedgehog signal transduction. Development. 125: 3553–62.
    1. Chen JK, Taipale J, Cooper MK, Beachy PA (2002) Inhibition of Hedgehog signaling by direct binding of cyclopamine to Smoothened. Genes Dev. 16: 2743–8.
    1. Sinha S, Chen JK (2006) Purmorphamine activates the Hedgehog pathway by targeting Smoothened. Nat Chem Biol. 2: 29–30.
    1. Kenney AM, Rowitch DH (2000) Sonic hedgehog promotes G(1) cyclin expression and sustained cell cycle progression in mammalian neuronal precursors. Mol Cell Biol. 20: 9055–67.
    1. Roy S, Ingham PW (2002) Hedgehogs tryst with the cell cycle. J Cell Sci. 115: 4393–7.
    1. Wickstrom M, Dyberg C, Shimokawa T, Milosevic J, Baryawno N, Fuskevag OM, et al. (2013) Targeting the hedgehog signal transduction pathway at the level of GLI inhibits neuroblastoma cell growth in vitro and in vivo. Int J Cancer. 132: 1516–24.
    1. Thu KL, Becker-Santos DD, Radulovich N, Pikor LA, Lam WL, Tsao MS (2014) SOX15 and other SOX family members are important mediators of tumorigenesis in multiple cancer types. Oncoscience. 1: 326–35.
    1. Castillo SD, Sanchez-Cespedes M (2012) The SOX family of genes in cancer development: biological relevance and opportunities for therapy. Expert Opin Ther Targets. 16: 903–19.
    1. Dong C, Wilhelm D, Koopman P (2004) Sox genes and cancer. Cytogenet Genome Res. 105: 442–7.
    1. Vervoort SJ, van Boxtel R, Coffer PJ (2013) The role of SRY-related HMG box transcription factor 4 (SOX4) in tumorigenesis and metastasis: friend or foe? Oncogene. 32: 3397–409.
    1. Zhu Y, Li Y, Jun Wei JW, Liu X (2012) The role of Sox genes in lung morphogenesis and cancer. Int J Mol Sci. 13: 15767–83.
    1. Vidal VP, Chaboissier MC, Lutzkendorf S, Cotsarelis G, Mill P, Hui CC, et al. (2005) Sox9 is essential for outer root sheath differentiation and the formation of the hair stem cell compartment. Curr Biol. 15: 1340–51.
    1. Lu B, Fang Y, Xu J, Wang L, Xu F, Xu E, et al. (2008) Analysis of SOX9 expression in colorectal cancer. Am J Clin Pathol. 130: 897–904.
    1. Santini R, Pietrobono S, Pandolfi S, Montagnani V, D'Amico M, Penachioni JY, et al. (2014) SOX2 regulates self-renewal and tumorigenicity of human melanoma-initiating cells. Oncogene. 33: 4697–708.
    1. Popovic J, Klajn A, Petrovic I, Stevanovic M (2010) Tissue-specific Forkhead protein FOXA2 up-regulates SOX14 gene expression. Biochim Biophys Acta. 1799: 411–8.
    1. Alvarez JI, Dodelet-Devillers A, Kebir H, Ifergan I, Fabre PJ, Terouz S, et al. (2011) The Hedgehog pathway promotes blood-brain barrier integrity and CNS immune quiescence. Science. 334: 1727–31.
    1. Woo WM, Zhen HH, Oro AE (2012) Shh maintains dermal papilla identity and hair morphogenesis via a Noggin-Shh regulatory loop. Genes Dev. 26: 1235–46.
    1. Bohr Mordhorst L, Ahlin C, Sorbe B (2014) Prognostic impact of the expression of Hedgehog proteins in cervical carcinoma FIGO stages I-IV treated with radiotherapy or chemoradiotherapy. Gynecol Oncol. 135: 305–11.
    1. Chaudary N, Pintilie M, Hedley D, Fyles AW, Milosevic M, Clarke B, et al. (2012) Hedgehog pathway signaling in cervical carcinoma and outcome after chemoradiation. Cancer. 118: 3105–15.
    1. Samarzija I, Beard P (2012) Hedgehog pathway regulators influence cervical cancer cell proliferation, survival and migration. Biochem Biophys Res Commun. 425: 64–9.
    1. Perrot CY, Javelaud D, Mauviel A (2013) Overlapping activities of TGF-beta and Hedgehog signaling in cancer: therapeutic targets for cancer treatment. Pharmacol Ther. 137: 183–99.
    1. Pierrat MJ, Marsaud V, Mauviel A, Javelaud D (2012) Expression of microphthalmia-associated transcription factor (MITF), which is critical for melanoma progression, is inhibited by both transcription factor GLI2 and transforming growth factor-beta. J Biol Chem. 287: 17996–8004.
    1. Pula B, Kobierzycki C, Solinski D, Olbromski M, Nowak-Markwitz E, Spaczynski M, et al. (2014) SOX18 expression predicts response to platinum-based chemotherapy in ovarian cancer. Anticancer Res. 34: 4029–37.
    1. Duman-Scheel M, Weng L, Xin S, Du W (2002) Hedgehog regulates cell growth and proliferation by inducing Cyclin D and Cyclin E. Nature. 417: 299–304.
    1. Giese A, Loo MA, Tran N, Haskett D, Coons SW, Berens ME (1996) Dichotomy of astrocytoma migration and proliferation. Int J Cancer. 67: 275–82.
    1. Khoshyomn S, Lew S, DeMattia J, Singer EB, Penar PL (1999) Brain tumor invasion rate measured in vitro does not correlate with Ki-67 expression. J Neurooncol. 45: 111–6.
    1. Merzak A, McCrea S, Koocheckpour S, Pilkington GJ (1994) Control of human glioma cell growth, migration and invasion in vitro by transforming growth factor beta 1. Br J Cancer. 70: 199–203.
    1. Friedl P, Wolf K (2003) Tumour-cell invasion and migration: diversity and escape mechanisms. Nat Rev Cancer. 3: 362–74.
    1. Alexandrova AY (2008) Evolution of cell interactions with extracellular matrix during carcinogenesis. Biochemistry (Mosc). 73: 733–41.
    1. Giampieri S, Manning C, Hooper S, Jones L, Hill CS, Sahai E (2009) Localized and reversible TGFbeta signalling switches breast cancer cells from cohesive to single cell motility. Nat Cell Biol. 11: 1287–96.
    1. Alexaki VI, Javelaud D, Van Kempen LC, Mohammad KS, Dennler S, Luciani F, et al. (2010) GLI2-mediated melanoma invasion and metastasis. J Natl Cancer Inst. 102: 1148–59.
    1. Karhadkar SS, Bova GS, Abdallah N, Dhara S, Gardner D, Maitra A, et al. (2004) Hedgehog signalling in prostate regeneration, neoplasia and metastasis. Nature. 431: 707–12.
    1. Kwon YJ, Hurst DR, Steg AD, Yuan K, Vaidya KS, Welch DR, et al. (2011) Gli1 enhances migration and invasion via up-regulation of MMP-11 and promotes metastasis in ERalpha negative breast cancer cell lines. Clin Exp Metastasis. 28: 437–49.
    1. Nagai S, Nakamura M, Yanai K, Wada J, Akiyoshi T, Nakashima H, et al. (2008) Gli1 contributes to the invasiveness of pancreatic cancer through matrix metalloproteinase-9 activation. Cancer Sci. 99: 1377–84.
    1. Hoeth M, Niederleithner H, Hofer-Warbinek R, Bilban M, Mayer H, Resch U, et al. (2012) The transcription factor SOX18 regulates the expression of matrix metalloproteinase 7 and guidance molecules in human endothelial cells. PLoS One. 7: e30982.
    1. Luo M, Guo XT, Yang W, Liu LQ, Li LW, Xin XY (2008) Inhibition of tumor angiogenesis by cell-permeable dominant negative SOX18 mutants. Med Hypotheses. 70: 880–2.

Source: PubMed

3
Iratkozz fel