메뉴 건너뛰기




Volumn 21, Issue 2, 2011, Pages 89-98

Thinking outside the box: Using metastasis suppressors as molecular tools

Author keywords

Cancer progression; Cell signaling; Metastasis; Metastasis suppressors

Indexed keywords

BETA CATENIN; CASPASE 8; CD82 ANTIGEN; CYCLIC AMP DEPENDENT PROTEIN KINASE ANCHORING PROTEIN; DELETED IN LIVER CANCER 1 PROTEIN; ELONGATION FACTOR 1; ELONGATION FACTOR 1A1; ENDOTHELIN 1; GELSOLIN; GUANOSINE TRIPHOSPHATASE ACTIVATING PROTEIN; HORMONALLY UPREGULATED NEU ASSOCIATED KINASE; HYDROLASE INHIBITOR; KRUPPEL LIKE FACTOR; KRUPPEL LIKE FACTOR 17; METASTIN; MITOGEN ACTIVATED PROTEIN KINASE KINASE 4; MITOGEN ACTIVATED PROTEIN KINASE KINASE 6; MITOGEN ACTIVATED PROTEIN KINASE KINASE 7; NERVE CELL ADHESION MOLECULE; NUCLEOSIDE DIPHOSPHATE KINASE NM23; PHOSPHATIDYLETHANOLAMINE BINDING PROTEIN; PROTEIN CDC42; PROTEIN SERINE THREONINE KINASE; RAC1 PROTEIN; RHO GUANOSINE DIPHOSPHATE DISSOCIATED INHIBITOR 2; RHOA GUANINE NUCLEOTIDE BINDING PROTEIN; SRC SUPPRESSED C KINASE SUBSTRATE; UNCLASSIFIED DRUG; UVOMORULIN;

EID: 79952069716     PISSN: 1044579X     EISSN: None     Source Type: Journal    
DOI: 10.1016/j.semcancer.2010.12.008     Document Type: Review
Times cited : (20)

References (116)
  • 1
    • 0025256084 scopus 로고
    • Metastatic inefficiency
    • Weiss L. Metastatic inefficiency. Adv Cancer Res 1990, 54:159-211.
    • (1990) Adv Cancer Res , vol.54 , pp. 159-211
    • Weiss, L.1
  • 4
    • 33746088024 scopus 로고    scopus 로고
    • Metastasis suppressor proteins: discovery, molecular mechanisms, and clinical application
    • Rinker-Schaeffer C.W., O'Keefe J.P., Welch D.R., Theodorescu D. Metastasis suppressor proteins: discovery, molecular mechanisms, and clinical application. Clin Cancer Res 2006, 12:3882-3889.
    • (2006) Clin Cancer Res , vol.12 , pp. 3882-3889
    • Rinker-Schaeffer, C.W.1    O'Keefe, J.P.2    Welch, D.R.3    Theodorescu, D.4
  • 6
    • 0027193360 scopus 로고
    • Inhibition of cell motility after nm23 transfection of human and murine tumor cells
    • Kantor J.D., McCormick B., Steeg P.S., Zetter B.R. Inhibition of cell motility after nm23 transfection of human and murine tumor cells. Cancer Res 1993, 53:1971-1973.
    • (1993) Cancer Res , vol.53 , pp. 1971-1973
    • Kantor, J.D.1    McCormick, B.2    Steeg, P.S.3    Zetter, B.R.4
  • 7
    • 32144460349 scopus 로고    scopus 로고
    • Expression of Nm23 in gliomas and its effect on migration and invasion in vitro
    • Jung S., Paek Y.W., Moon K.S., Wee S.C., Ryu H.H., Jeong Y.I., et al. Expression of Nm23 in gliomas and its effect on migration and invasion in vitro. Anticancer Res 2006, 26:249-258.
    • (2006) Anticancer Res , vol.26 , pp. 249-258
    • Jung, S.1    Paek, Y.W.2    Moon, K.S.3    Wee, S.C.4    Ryu, H.H.5    Jeong, Y.I.6
  • 8
    • 0032887607 scopus 로고    scopus 로고
    • Inhibitory activity of nm23-H1 on invasion and colonization of human prostate carcinoma cells is not mediated by its NDP kinase activity
    • Lee H.Y., Lee H. Inhibitory activity of nm23-H1 on invasion and colonization of human prostate carcinoma cells is not mediated by its NDP kinase activity. Cancer Lett 1999, 145:93-99.
    • (1999) Cancer Lett , vol.145 , pp. 93-99
    • Lee, H.Y.1    Lee, H.2
  • 9
    • 0347382319 scopus 로고    scopus 로고
    • Nm23-H1 reduces in vitro cell migration and the liver metastatic potential of colon cancer cells by regulating myosin light chain phosphorylation
    • Suzuki E., Ota T., Tsukuda K., Okita A., Matsuoka K., Murakami M., et al. nm23-H1 reduces in vitro cell migration and the liver metastatic potential of colon cancer cells by regulating myosin light chain phosphorylation. Int J Cancer 2004, 108:207-211.
    • (2004) Int J Cancer , vol.108 , pp. 207-211
    • Suzuki, E.1    Ota, T.2    Tsukuda, K.3    Okita, A.4    Matsuoka, K.5    Murakami, M.6
  • 10
    • 0029168020 scopus 로고
    • Colon carcinoma cells with inactive nm23 show increased motility and response to motility factors
    • Hsu S., Huang F., Ossowski L., Friedman E. Colon carcinoma cells with inactive nm23 show increased motility and response to motility factors. Carcinogenesis 1995, 16:2259-2262.
    • (1995) Carcinogenesis , vol.16 , pp. 2259-2262
    • Hsu, S.1    Huang, F.2    Ossowski, L.3    Friedman, E.4
  • 11
    • 7344257797 scopus 로고    scopus 로고
    • Relationship of nm23 to proteolytic factors, proliferation and motility in breast cancer tissues and cell lines
    • Russell R.L., Pedersen A.N., Kantor J., Geisinger K., Long R., Zbieranski N., et al. Relationship of nm23 to proteolytic factors, proliferation and motility in breast cancer tissues and cell lines. Br J Cancer 1998, 78:710-717.
    • (1998) Br J Cancer , vol.78 , pp. 710-717
    • Russell, R.L.1    Pedersen, A.N.2    Kantor, J.3    Geisinger, K.4    Long, R.5    Zbieranski, N.6
  • 12
    • 0035023681 scopus 로고    scopus 로고
    • Nm23-H1 suppresses invasion of oral squamous cell carcinoma-derived cell lines without modifying matrix metalloproteinase-2 and matrix metalloproteinase-9 expression
    • Khan M.H., Yasuda M., Higashino F., Haque S., Kohgo T., Nakamura M., et al. nm23-H1 suppresses invasion of oral squamous cell carcinoma-derived cell lines without modifying matrix metalloproteinase-2 and matrix metalloproteinase-9 expression. Am J Pathol 2001, 158:1785-1791.
    • (2001) Am J Pathol , vol.158 , pp. 1785-1791
    • Khan, M.H.1    Yasuda, M.2    Higashino, F.3    Haque, S.4    Kohgo, T.5    Nakamura, M.6
  • 13
    • 33746956118 scopus 로고    scopus 로고
    • Reduction of invasion in human fibrosarcoma cells by ribosomal protein S3 in conjunction with Nm23-H1 and ERK
    • Kim S.H., Kim J. Reduction of invasion in human fibrosarcoma cells by ribosomal protein S3 in conjunction with Nm23-H1 and ERK. Biochim Biophys Acta 2006, 1763:823-832.
    • (2006) Biochim Biophys Acta , vol.1763 , pp. 823-832
    • Kim, S.H.1    Kim, J.2
  • 14
    • 34547616645 scopus 로고    scopus 로고
    • Nm23-H1 suppresses tumor cell motility by down-regulating the lysophosphatidic acid receptor EDG2
    • Horak C.E., Lee J.H., Elkahloun A.G., Boissan M., Dumont S., Maga T.K., et al. Nm23-H1 suppresses tumor cell motility by down-regulating the lysophosphatidic acid receptor EDG2. Cancer Res 2007, 67:7238-7246.
    • (2007) Cancer Res , vol.67 , pp. 7238-7246
    • Horak, C.E.1    Lee, J.H.2    Elkahloun, A.G.3    Boissan, M.4    Dumont, S.5    Maga, T.K.6
  • 15
    • 37549041384 scopus 로고    scopus 로고
    • Nm23-H1 suppresses metastasis by inhibiting expression of the lysophosphatidic acid receptor EDG2
    • Horak C.E., Mendoza A., Vega-Valle E., Albaugh M., Graff-Cherry C., McDermott W.G., et al. Nm23-H1 suppresses metastasis by inhibiting expression of the lysophosphatidic acid receptor EDG2. Cancer Res 2007, 67:11751-11759.
    • (2007) Cancer Res , vol.67 , pp. 11751-11759
    • Horak, C.E.1    Mendoza, A.2    Vega-Valle, E.3    Albaugh, M.4    Graff-Cherry, C.5    McDermott, W.G.6
  • 17
    • 0001027292 scopus 로고    scopus 로고
    • Gelsolin, a multifunctional actin regulatory protein
    • Sun H.Q., Yamamoto M., Mejillano M., Yin H.L. Gelsolin, a multifunctional actin regulatory protein. J Biol Chem 1999, 274:33179-33182.
    • (1999) J Biol Chem , vol.274 , pp. 33179-33182
    • Sun, H.Q.1    Yamamoto, M.2    Mejillano, M.3    Yin, H.L.4
  • 18
    • 0021356621 scopus 로고
    • Unphosphorylated gelsolin is localized in regions of cell-substratum contact or attachment in Rous sarcoma virus-transformed rat cells
    • Wang E., Yin H.L., Krueger J.G., Caliguiri L.A., Tamm I. Unphosphorylated gelsolin is localized in regions of cell-substratum contact or attachment in Rous sarcoma virus-transformed rat cells. J Cell Biol 1984, 98:761-771.
    • (1984) J Cell Biol , vol.98 , pp. 761-771
    • Wang, E.1    Yin, H.L.2    Krueger, J.G.3    Caliguiri, L.A.4    Tamm, I.5
  • 19
    • 0025782925 scopus 로고
    • Enhanced motility in NIH 3T3 fibroblasts that overexpress gelsolin
    • Cunningham C.C., Stossel T.P., Kwiatkowski D.J. Enhanced motility in NIH 3T3 fibroblasts that overexpress gelsolin. Science 1991, 251:1233-1236.
    • (1991) Science , vol.251 , pp. 1233-1236
    • Cunningham, C.C.1    Stossel, T.P.2    Kwiatkowski, D.J.3
  • 20
    • 0032473498 scopus 로고    scopus 로고
    • Gelsolin is a downstream effector of rac for fibroblast motility
    • Azuma T., Witke W., Stossel T.P., Hartwig J.H., Kwiatkowski D.J. Gelsolin is a downstream effector of rac for fibroblast motility. EMBO J 1998, 17:1362-1370.
    • (1998) EMBO J , vol.17 , pp. 1362-1370
    • Azuma, T.1    Witke, W.2    Stossel, T.P.3    Hartwig, J.H.4    Kwiatkowski, D.J.5
  • 22
    • 0032893518 scopus 로고    scopus 로고
    • Cell motility as a prognostic factor in Stage I nonsmall cell lung carcinoma: the role of gelsolin expression
    • Shieh D.B., Godleski J., Herndon J.E., Azuma T., Mercer H., Sugarbaker D.J., et al. Cell motility as a prognostic factor in Stage I nonsmall cell lung carcinoma: the role of gelsolin expression. Cancer 1999, 85:47-57.
    • (1999) Cancer , vol.85 , pp. 47-57
    • Shieh, D.B.1    Godleski, J.2    Herndon, J.E.3    Azuma, T.4    Mercer, H.5    Sugarbaker, D.J.6
  • 23
    • 0034892806 scopus 로고    scopus 로고
    • Gelsolin as a negative prognostic factor and effector of motility in erbB-2-positive epidermal growth factor receptor-positive breast cancers
    • Thor A.D., Edgerton S.M., Liu S., Moore D.H., Kwiatkowski D.J. Gelsolin as a negative prognostic factor and effector of motility in erbB-2-positive epidermal growth factor receptor-positive breast cancers. Clin Cancer Res 2001, 7:2415-2424.
    • (2001) Clin Cancer Res , vol.7 , pp. 2415-2424
    • Thor, A.D.1    Edgerton, S.M.2    Liu, S.3    Moore, D.H.4    Kwiatkowski, D.J.5
  • 25
    • 33645343819 scopus 로고    scopus 로고
    • Murine androgen-independent neuroendocrine carcinoma promotes metastasis of human prostate cancer cell line LNCaP
    • Uchida K., Masumori N., Takahashi A., Itoh N., Kato K., Matusik R.J., et al. Murine androgen-independent neuroendocrine carcinoma promotes metastasis of human prostate cancer cell line LNCaP. Prostate 2006, 66:536-545.
    • (2006) Prostate , vol.66 , pp. 536-545
    • Uchida, K.1    Masumori, N.2    Takahashi, A.3    Itoh, N.4    Kato, K.5    Matusik, R.J.6
  • 26
    • 77950531312 scopus 로고    scopus 로고
    • BCL2 inhibits cell adhesion, spreading, and motility by enhancing actin polymerization
    • Ke H., Parron V.I., Reece J., Zhang J.Y., Akiyama S.K., French J.E. BCL2 inhibits cell adhesion, spreading, and motility by enhancing actin polymerization. Cell Res 2010, 20:458-469.
    • (2010) Cell Res , vol.20 , pp. 458-469
    • Ke, H.1    Parron, V.I.2    Reece, J.3    Zhang, J.Y.4    Akiyama, S.K.5    French, J.E.6
  • 27
    • 0036368684 scopus 로고    scopus 로고
    • Targeting actin remodeling profiles for the detection and management of urothelial cancers - a perspective for bladder cancer research
    • Rao J. Targeting actin remodeling profiles for the detection and management of urothelial cancers - a perspective for bladder cancer research. Front Biosci 2002, 7:e1-e8.
    • (2002) Front Biosci , vol.7
    • Rao, J.1
  • 28
    • 21744432683 scopus 로고    scopus 로고
    • GDIs: central regulatory molecules in Rho GTPase activation
    • DerMardirossian C., Bokoch G.M. GDIs: central regulatory molecules in Rho GTPase activation. Trends Cell Biol 2005, 15:356-363.
    • (2005) Trends Cell Biol , vol.15 , pp. 356-363
    • DerMardirossian, C.1    Bokoch, G.M.2
  • 29
    • 0035808306 scopus 로고    scopus 로고
    • Structure-activity relationships in flexible protein domains: regulation of rho GTPases by RhoGDI and D4 GDI
    • Golovanov A.P., Chuang T.H., DerMardirossian C., Barsukov I., Hawkins D., Badii R., et al. Structure-activity relationships in flexible protein domains: regulation of rho GTPases by RhoGDI and D4 GDI. J Mol Biol 2001, 305:121-135.
    • (2001) J Mol Biol , vol.305 , pp. 121-135
    • Golovanov, A.P.1    Chuang, T.H.2    DerMardirossian, C.3    Barsukov, I.4    Hawkins, D.5    Badii, R.6
  • 30
    • 0343750716 scopus 로고    scopus 로고
    • The Rac-RhoGDI complex and the structural basis for the regulation of Rho proteins by RhoGDI
    • Scheffzek K., Stephan I., Jensen O.N., Illenberger D., Gierschik P. The Rac-RhoGDI complex and the structural basis for the regulation of Rho proteins by RhoGDI. Nat Struct Biol 2000, 7:122-126.
    • (2000) Nat Struct Biol , vol.7 , pp. 122-126
    • Scheffzek, K.1    Stephan, I.2    Jensen, O.N.3    Illenberger, D.4    Gierschik, P.5
  • 31
    • 0141954162 scopus 로고    scopus 로고
    • The role of Ras superfamily proteins in bladder cancer progression
    • Oxford G., Theodorescu D. The role of Ras superfamily proteins in bladder cancer progression. J Urol 2003, 170:1987-1993.
    • (2003) J Urol , vol.170 , pp. 1987-1993
    • Oxford, G.1    Theodorescu, D.2
  • 33
    • 34548052025 scopus 로고    scopus 로고
    • Biphasic expression of RhoGDI2 in the progression of breast cancer and its negative relation with lymph node metastasis
    • Hu L.D., Zou H.F., Zhan S.X., Cao K.M. Biphasic expression of RhoGDI2 in the progression of breast cancer and its negative relation with lymph node metastasis. Oncol Rep 2007, 17:1383-1389.
    • (2007) Oncol Rep , vol.17 , pp. 1383-1389
    • Hu, L.D.1    Zou, H.F.2    Zhan, S.X.3    Cao, K.M.4
  • 34
    • 2542606977 scopus 로고    scopus 로고
    • Reduced expression of metastasis suppressor RhoGDI2 is associated with decreased survival for patients with bladder cancer
    • Theodorescu D., Sapinoso L.M., Conaway M.R., Oxford G., Hampton G.M., Frierson H.F., et al. Reduced expression of metastasis suppressor RhoGDI2 is associated with decreased survival for patients with bladder cancer. Clin Cancer Res 2004, 10:3800-3806.
    • (2004) Clin Cancer Res , vol.10 , pp. 3800-3806
    • Theodorescu, D.1    Sapinoso, L.M.2    Conaway, M.R.3    Oxford, G.4    Hampton, G.M.5    Frierson, H.F.6
  • 35
    • 23844468385 scopus 로고    scopus 로고
    • Endothelin axis is a target of the lung metastasis suppressor gene RhoGDI2
    • Titus B., Frierson H.F., Conaway M., Ching K., Guise T., Chirgwin J., et al. Endothelin axis is a target of the lung metastasis suppressor gene RhoGDI2. Cancer Res 2005, 65:7320-7327.
    • (2005) Cancer Res , vol.65 , pp. 7320-7327
    • Titus, B.1    Frierson, H.F.2    Conaway, M.3    Ching, K.4    Guise, T.5    Chirgwin, J.6
  • 36
    • 0034730781 scopus 로고    scopus 로고
    • Involvement of Rho GTPases in the transcriptional inhibition of preproendothelin-1 gene expression by simvastatin in vascular endothelial cells
    • Hernandez-Perera O., Perez-Sala D., Soria E., Lamas S. Involvement of Rho GTPases in the transcriptional inhibition of preproendothelin-1 gene expression by simvastatin in vascular endothelial cells. Circ Res 2000, 87:616-622.
    • (2000) Circ Res , vol.87 , pp. 616-622
    • Hernandez-Perera, O.1    Perez-Sala, D.2    Soria, E.3    Lamas, S.4
  • 37
    • 1942453322 scopus 로고    scopus 로고
    • Historical review: endothelin
    • Masaki T. Historical review: endothelin. Trends Pharmacol Sci 2004, 25:219-224.
    • (2004) Trends Pharmacol Sci , vol.25 , pp. 219-224
    • Masaki, T.1
  • 38
    • 66149117890 scopus 로고    scopus 로고
    • Rho GDP dissociation inhibitor 2 suppresses metastasis via unconventional regulation of RhoGTPases
    • Moissoglu K., McRoberts K.S., Meier J.A., Theodorescu D., Schwartz M.A. Rho GDP dissociation inhibitor 2 suppresses metastasis via unconventional regulation of RhoGTPases. Cancer Res 2009, 69:2838-2844.
    • (2009) Cancer Res , vol.69 , pp. 2838-2844
    • Moissoglu, K.1    McRoberts, K.S.2    Meier, J.A.3    Theodorescu, D.4    Schwartz, M.A.5
  • 39
    • 26944440432 scopus 로고    scopus 로고
    • RhoGDIs revisited: novel roles in Rho regulation
    • Dransart E., Olofsson B., Cherfils J. RhoGDIs revisited: novel roles in Rho regulation. Traffic 2005, 6:957-966.
    • (2005) Traffic , vol.6 , pp. 957-966
    • Dransart, E.1    Olofsson, B.2    Cherfils, J.3
  • 41
    • 7244251723 scopus 로고    scopus 로고
    • The RacGEF Tiam1 inhibits migration and invasion of metastatic melanoma via a novel adhesive mechanism
    • Uhlenbrock K., Eberth A., Herbrand U., Daryab N., Stege P., Meier F., et al. The RacGEF Tiam1 inhibits migration and invasion of metastatic melanoma via a novel adhesive mechanism. J Cell Sci 2004, 117:4863-4871.
    • (2004) J Cell Sci , vol.117 , pp. 4863-4871
    • Uhlenbrock, K.1    Eberth, A.2    Herbrand, U.3    Daryab, N.4    Stege, P.5    Meier, F.6
  • 42
    • 34248210086 scopus 로고    scopus 로고
    • Rho activation at a glance
    • Buchsbaum R.J. Rho activation at a glance. J Cell Sci 2007, 120:1149-1152.
    • (2007) J Cell Sci , vol.120 , pp. 1149-1152
    • Buchsbaum, R.J.1
  • 43
    • 22244443056 scopus 로고    scopus 로고
    • The RhoGAP protein DLC-1 functions as a metastasis suppressor in breast cancer cells
    • Goodison S., Yuan J., Sloan D., Kim R., Li C., Popescu N.C., et al. The RhoGAP protein DLC-1 functions as a metastasis suppressor in breast cancer cells. Cancer Res 2005, 65:6042-6053.
    • (2005) Cancer Res , vol.65 , pp. 6042-6053
    • Goodison, S.1    Yuan, J.2    Sloan, D.3    Kim, R.4    Li, C.5    Popescu, N.C.6
  • 44
    • 35248890380 scopus 로고    scopus 로고
    • Morphological changes and nuclear translocation of DLC1 tumor suppressor protein precede apoptosis in human non-small cell lung carcinoma cells
    • Yuan B.Z., Jefferson A.M., Millecchia L., Popescu N.C., Reynolds S.H. Morphological changes and nuclear translocation of DLC1 tumor suppressor protein precede apoptosis in human non-small cell lung carcinoma cells. Exp Cell Res 2007, 313:3868-3880.
    • (2007) Exp Cell Res , vol.313 , pp. 3868-3880
    • Yuan, B.Z.1    Jefferson, A.M.2    Millecchia, L.3    Popescu, N.C.4    Reynolds, S.H.5
  • 45
    • 78449267307 scopus 로고    scopus 로고
    • DLC1 negatively regulates angiogenesis in a paracrine fashion
    • Shih Y.P., Liao Y.C., Lin Y., Lo S.H. DLC1 negatively regulates angiogenesis in a paracrine fashion. Cancer Res 2010.
    • (2010) Cancer Res
    • Shih, Y.P.1    Liao, Y.C.2    Lin, Y.3    Lo, S.H.4
  • 46
    • 33847073342 scopus 로고    scopus 로고
    • DLC-1, a GTPase-activating protein for Rho, is associated with cell proliferation, morphology, and migration in human hepatocellular carcinoma
    • Kim T.Y., Lee J.W., Kim H.P., Jong H.S., Jung M., Bang Y.J. 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.
    • (2007) Biochem Biophys Res Commun , vol.355 , pp. 72-77
    • Kim, T.Y.1    Lee, J.W.2    Kim, H.P.3    Jong, H.S.4    Jung, M.5    Bang, Y.J.6
  • 47
    • 40249094998 scopus 로고    scopus 로고
    • Inhibition of DLC-1 gene expression by RNA interference in the colon cancer LoVo cell line
    • Jin Y., Tian X., Shang Y., Huang P. Inhibition of DLC-1 gene expression by RNA interference in the colon cancer LoVo cell line. Oncol Rep 2008, 19:669-674.
    • (2008) Oncol Rep , vol.19 , pp. 669-674
    • Jin, Y.1    Tian, X.2    Shang, Y.3    Huang, P.4
  • 48
    • 0345731237 scopus 로고    scopus 로고
    • Cell migration: Rho GTPases lead the way
    • Raftopoulou M., Hall A. Cell migration: Rho GTPases lead the way. Dev Biol 2004, 265:23-32.
    • (2004) Dev Biol , vol.265 , pp. 23-32
    • Raftopoulou, M.1    Hall, A.2
  • 49
    • 42949100728 scopus 로고    scopus 로고
    • DLC-1 suppresses non-small cell lung cancer growth and invasion by RhoGAP-dependent and independent mechanisms
    • Healy K.D., Hodgson L., Kim T.Y., Shutes A., Maddileti S., Juliano R.L., 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.
    • (2008) Mol Carcinog , vol.47 , pp. 326-337
    • Healy, K.D.1    Hodgson, L.2    Kim, T.Y.3    Shutes, A.4    Maddileti, S.5    Juliano, R.L.6
  • 50
    • 48149111026 scopus 로고    scopus 로고
    • DLC1 suppresses distant dissemination of human hepatocellular carcinoma cells in nude mice through reduction of RhoA GTPase activity, actin cytoskeletal disruption and down-regulation of genes involved in metastasis
    • Zhou X., Zimonjic D.B., Park S.W., Yang X.Y., Durkin M.E., Popescu N.C. DLC1 suppresses distant dissemination of human hepatocellular carcinoma cells in nude mice through reduction of RhoA GTPase activity, actin cytoskeletal disruption and down-regulation of genes involved in metastasis. Int J Oncol 2008, 32:1285-1291.
    • (2008) Int J Oncol , vol.32 , pp. 1285-1291
    • Zhou, X.1    Zimonjic, D.B.2    Park, S.W.3    Yang, X.Y.4    Durkin, M.E.5    Popescu, N.C.6
  • 51
    • 61849091401 scopus 로고    scopus 로고
    • Transcriptional induction of DLC-1 gene through Sp1 sites by histone deacetylase inhibitors in gastric cancer cells
    • Kim T.Y., Kim I.S., Jong H.S., Lee J.W., Jung M., Bang Y.J. Transcriptional induction of DLC-1 gene through Sp1 sites by histone deacetylase inhibitors in gastric cancer cells. Exp Mol Med 2008, 40:639-646.
    • (2008) Exp Mol Med , vol.40 , pp. 639-646
    • Kim, T.Y.1    Kim, I.S.2    Jong, H.S.3    Lee, J.W.4    Jung, M.5    Bang, Y.J.6
  • 52
    • 60849123432 scopus 로고    scopus 로고
    • Focal adhesion-localization of START-GAP1/DLC1 is essential for cell motility and morphology
    • Kawai K., Iwamae Y., Yamaga M., Kiyota M., Ishii H., Hirata H., et al. Focal adhesion-localization of START-GAP1/DLC1 is essential for cell motility and morphology. Genes Cells 2009, 14:227-241.
    • (2009) Genes Cells , vol.14 , pp. 227-241
    • Kawai, K.1    Iwamae, Y.2    Yamaga, M.3    Kiyota, M.4    Ishii, H.5    Hirata, H.6
  • 53
    • 77955294043 scopus 로고    scopus 로고
    • START-GAP1/DLC1 is localized in focal adhesions through interaction with the PTB domain of tensin2
    • Kawai K., Kitamura S.Y., Maehira K., Seike J., Yagisawa H. START-GAP1/DLC1 is localized in focal adhesions through interaction with the PTB domain of tensin2. Adv Enzyme Regul 2010, 50:202-215.
    • (2010) Adv Enzyme Regul , vol.50 , pp. 202-215
    • Kawai, K.1    Kitamura, S.Y.2    Maehira, K.3    Seike, J.4    Yagisawa, H.5
  • 54
    • 65249084000 scopus 로고    scopus 로고
    • The SAM domain of the RhoGAP DLC1 binds EF1A1 to regulate cell migration
    • Zhong D., Zhang J., Yang S., Soh U.J., Buschdorf J.P., Zhou Y.T., et al. The SAM domain of the RhoGAP DLC1 binds EF1A1 to regulate cell migration. J Cell Sci 2009, 122:414-424.
    • (2009) J Cell Sci , vol.122 , pp. 414-424
    • Zhong, D.1    Zhang, J.2    Yang, S.3    Soh, U.J.4    Buschdorf, J.P.5    Zhou, Y.T.6
  • 55
    • 0036674955 scopus 로고    scopus 로고
    • The role of SSeCKS/gravin/AKAP12 scaffolding proteins in the spaciotemporal control of signaling pathways in oncogenesis and development
    • Gelman I.H. The role of SSeCKS/gravin/AKAP12 scaffolding proteins in the spaciotemporal control of signaling pathways in oncogenesis and development. Front Biosci 2002, 7:d1782-d1797.
    • (2002) Front Biosci , vol.7
    • Gelman, I.H.1
  • 56
    • 0029989387 scopus 로고    scopus 로고
    • A novel src- and ras-suppressed protein kinase C substrate associated with cytoskeletal architecture
    • Lin X., Tombler E., Nelson P.J., Ross M., Gelman I.H. A novel src- and ras-suppressed protein kinase C substrate associated with cytoskeletal architecture. J Biol Chem 1996, 271:28430-28438.
    • (1996) J Biol Chem , vol.271 , pp. 28430-28438
    • Lin, X.1    Tombler, E.2    Nelson, P.J.3    Ross, M.4    Gelman, I.H.5
  • 57
    • 0035878927 scopus 로고    scopus 로고
    • The Src-suppressed C kinase substrate, SSeCKS, is a potential metastasis inhibitor in prostate cancer
    • Xia W., Unger P., Miller L., Nelson J., Gelman I.H. The Src-suppressed C kinase substrate, SSeCKS, is a potential metastasis inhibitor in prostate cancer. Cancer Res 2001, 61:5644-5651.
    • (2001) Cancer Res , vol.61 , pp. 5644-5651
    • Xia, W.1    Unger, P.2    Miller, L.3    Nelson, J.4    Gelman, I.H.5
  • 58
    • 77951170093 scopus 로고    scopus 로고
    • SSeCKS/Gravin/AKAP12 inhibits cancer cell invasiveness and chemotaxis by suppressing a protein kinase C- Raf/MEK/ERK pathway
    • Su B., Bu Y., Engelberg D., Gelman I.H. SSeCKS/Gravin/AKAP12 inhibits cancer cell invasiveness and chemotaxis by suppressing a protein kinase C- Raf/MEK/ERK pathway. J Biol Chem 2010, 285:4578-4586.
    • (2010) J Biol Chem , vol.285 , pp. 4578-4586
    • Su, B.1    Bu, Y.2    Engelberg, D.3    Gelman, I.H.4
  • 59
    • 77149179101 scopus 로고    scopus 로고
    • Adherens junction: molecular architecture and regulation
    • Meng W., Takeichi M. Adherens junction: molecular architecture and regulation. Cold Spring Harb Perspect Biol 2009, 1:a002899.
    • (2009) Cold Spring Harb Perspect Biol , vol.1
    • Meng, W.1    Takeichi, M.2
  • 60
    • 68549106083 scopus 로고    scopus 로고
    • E-cadherin, beta-catenin, and ZEB1 in malignant progression of cancer
    • Schmalhofer O., Brabletz S., Brabletz T. E-cadherin, beta-catenin, and ZEB1 in malignant progression of cancer. Cancer Metastasis Rev 2009, 28:151-166.
    • (2009) Cancer Metastasis Rev , vol.28 , pp. 151-166
    • Schmalhofer, O.1    Brabletz, S.2    Brabletz, T.3
  • 61
  • 63
    • 0036781973 scopus 로고    scopus 로고
    • A signaling pathway leading to metastasis is controlled by N-cadherin and the FGF receptor
    • Suyama K., Shapiro I., Guttman M., Hazan R.B. A signaling pathway leading to metastasis is controlled by N-cadherin and the FGF receptor. Cancer Cell 2002, 2:301-314.
    • (2002) Cancer Cell , vol.2 , pp. 301-314
    • Suyama, K.1    Shapiro, I.2    Guttman, M.3    Hazan, R.B.4
  • 64
    • 0037112443 scopus 로고    scopus 로고
    • An alternatively spliced cadherin-11 enhances human breast cancer cell invasion
    • Feltes C.M., Kudo A., Blaschuk O., Byers S.W. An alternatively spliced cadherin-11 enhances human breast cancer cell invasion. Cancer Res 2002, 62:6688-6697.
    • (2002) Cancer Res , vol.62 , pp. 6688-6697
    • Feltes, C.M.1    Kudo, A.2    Blaschuk, O.3    Byers, S.W.4
  • 65
    • 24344467835 scopus 로고    scopus 로고
    • Transendothelial migration of melanoma cells involves N-cadherin-mediated adhesion and activation of the beta-catenin signaling pathway
    • Qi J., Chen N., Wang J., Siu C.H. Transendothelial migration of melanoma cells involves N-cadherin-mediated adhesion and activation of the beta-catenin signaling pathway. Mol Biol Cell 2005, 16:4386-4397.
    • (2005) Mol Biol Cell , vol.16 , pp. 4386-4397
    • Qi, J.1    Chen, N.2    Wang, J.3    Siu, C.H.4
  • 66
    • 33644864094 scopus 로고    scopus 로고
    • Involvement of Src family kinases in N-cadherin phosphorylation and beta-catenin dissociation during transendothelial migration of melanoma cells
    • Qi J., Wang J., Romanyuk O., Siu C.H. Involvement of Src family kinases in N-cadherin phosphorylation and beta-catenin dissociation during transendothelial migration of melanoma cells. Mol Biol Cell 2006, 17:1261-1272.
    • (2006) Mol Biol Cell , vol.17 , pp. 1261-1272
    • Qi, J.1    Wang, J.2    Romanyuk, O.3    Siu, C.H.4
  • 67
    • 0038275924 scopus 로고    scopus 로고
    • Effects of raf kinase inhibitor protein expression on suppression of prostate cancer metastasis
    • Fu Z., Smith P.C., Zhang L., Rubin M.A., Dunn R.L., Yao Z., et al. Effects of raf kinase inhibitor protein expression on suppression of prostate cancer metastasis. J Natl Cancer Inst 2003, 95:878-889.
    • (2003) J Natl Cancer Inst , vol.95 , pp. 878-889
    • Fu, Z.1    Smith, P.C.2    Zhang, L.3    Rubin, M.A.4    Dunn, R.L.5    Yao, Z.6
  • 68
    • 27144473329 scopus 로고    scopus 로고
    • Reduction of Raf-1 kinase inhibitor protein expression correlates with breast cancer metastasis
    • Hagan S., Al-Mulla F., Mallon E., Oien K., Ferrier R., Gusterson B., et al. Reduction of Raf-1 kinase inhibitor protein expression correlates with breast cancer metastasis. Clin Cancer Res 2005, 11:7392-7397.
    • (2005) Clin Cancer Res , vol.11 , pp. 7392-7397
    • Hagan, S.1    Al-Mulla, F.2    Mallon, E.3    Oien, K.4    Ferrier, R.5    Gusterson, B.6
  • 69
    • 50349086790 scopus 로고    scopus 로고
    • Effects of raf kinase inhibitor protein expression on metastasis and progression of human epithelial ovarian cancer
    • Li H.Z., Wang Y., Gao Y., Shao J., Zhao X.L., Deng W.M., et al. Effects of raf kinase inhibitor protein expression on metastasis and progression of human epithelial ovarian cancer. Mol Cancer Res 2008, 6:917-928.
    • (2008) Mol Cancer Res , vol.6 , pp. 917-928
    • Li, H.Z.1    Wang, Y.2    Gao, Y.3    Shao, J.4    Zhao, X.L.5    Deng, W.M.6
  • 70
    • 24344494340 scopus 로고    scopus 로고
    • How microRNAs control cell division, differentiation and death
    • Miska E.A. How microRNAs control cell division, differentiation and death. Curr Opin Genet Dev 2005, 15:563-568.
    • (2005) Curr Opin Genet Dev , vol.15 , pp. 563-568
    • Miska, E.A.1
  • 71
    • 40749121726 scopus 로고    scopus 로고
    • Node-negative colorectal cancer at high risk of distant metastasis identified by combined analysis of lymph node status, vascular invasion, and Raf-1 kinase inhibitor protein expression
    • Zlobec I., Baker K., Minoo P., Jass J.R., Terracciano L., Lugli A. Node-negative colorectal cancer at high risk of distant metastasis identified by combined analysis of lymph node status, vascular invasion, and Raf-1 kinase inhibitor protein expression. Clin Cancer Res 2008, 14:143-148.
    • (2008) Clin Cancer Res , vol.14 , pp. 143-148
    • Zlobec, I.1    Baker, K.2    Minoo, P.3    Jass, J.R.4    Terracciano, L.5    Lugli, A.6
  • 72
    • 3442883359 scopus 로고    scopus 로고
    • Reduction in Raf kinase inhibitor protein expression is associated with increased Ras-extracellular signal-regulated kinase signaling in melanoma cell lines
    • Schuierer M.M., Bataille F., Hagan S., Kolch W., Bosserhoff A.K. Reduction in Raf kinase inhibitor protein expression is associated with increased Ras-extracellular signal-regulated kinase signaling in melanoma cell lines. Cancer Res 2004, 64:5186-5192.
    • (2004) Cancer Res , vol.64 , pp. 5186-5192
    • Schuierer, M.M.1    Bataille, F.2    Hagan, S.3    Kolch, W.4    Bosserhoff, A.K.5
  • 73
    • 61349156154 scopus 로고    scopus 로고
    • Identification of RKIP as an invasion suppressor protein in nasopharyngeal carcinoma by proteomic analysis
    • Chen Y., Ouyang G.L., Yi H., Li M.Y., Zhang P.F., Li C., et al. Identification of RKIP as an invasion suppressor protein in nasopharyngeal carcinoma by proteomic analysis. J Proteome Res 2008, 7:5254-5262.
    • (2008) J Proteome Res , vol.7 , pp. 5254-5262
    • Chen, Y.1    Ouyang, G.L.2    Yi, H.3    Li, M.Y.4    Zhang, P.F.5    Li, C.6
  • 74
    • 67649612845 scopus 로고    scopus 로고
    • Effects of raf kinase inhibitor protein expression on metastasis and progression of human breast cancer
    • Li H.Z., Gao Y., Zhao X.L., Liu Y.X., Sun B.C., Yang J., et al. Effects of raf kinase inhibitor protein expression on metastasis and progression of human breast cancer. Mol Cancer Res 2009, 7:832-840.
    • (2009) Mol Cancer Res , vol.7 , pp. 832-840
    • Li, H.Z.1    Gao, Y.2    Zhao, X.L.3    Liu, Y.X.4    Sun, B.C.5    Yang, J.6
  • 75
    • 70349973076 scopus 로고    scopus 로고
    • Inhibition of epithelial to mesenchymal transition in metastatic prostate cancer cells by the novel proteasome inhibitor, NPI-0052: pivotal roles of Snail repression and RKIP induction
    • Baritaki S., Chapman A., Yeung K., Spandidos D.A., Palladino M., Bonavida B. Inhibition of epithelial to mesenchymal transition in metastatic prostate cancer cells by the novel proteasome inhibitor, NPI-0052: pivotal roles of Snail repression and RKIP induction. Oncogene 2009, 28:3573-3585.
    • (2009) Oncogene , vol.28 , pp. 3573-3585
    • Baritaki, S.1    Chapman, A.2    Yeung, K.3    Spandidos, D.A.4    Palladino, M.5    Bonavida, B.6
  • 76
    • 78049276666 scopus 로고    scopus 로고
    • Raf kinase inhibitor protein suppresses nuclear factor-kappaB-dependent cancer cell invasion through negative regulation of matrix metalloproteinase expression
    • Beshir A.B., Ren G., Magpusao A.N., Barone L.M., Yeung K.C., Fenteany G. Raf kinase inhibitor protein suppresses nuclear factor-kappaB-dependent cancer cell invasion through negative regulation of matrix metalloproteinase expression. Cancer Lett 2010.
    • (2010) Cancer Lett
    • Beshir, A.B.1    Ren, G.2    Magpusao, A.N.3    Barone, L.M.4    Yeung, K.C.5    Fenteany, G.6
  • 77
    • 1942469421 scopus 로고    scopus 로고
    • Snail and SIP1 increase cancer invasion by upregulating MMP family in hepatocellular carcinoma cells
    • Miyoshi A., Kitajima Y., Sumi K., Sato K., Hagiwara A., Koga Y., et al. Snail and SIP1 increase cancer invasion by upregulating MMP family in hepatocellular carcinoma cells. Br J Cancer 2004, 90:1265-1273.
    • (2004) Br J Cancer , vol.90 , pp. 1265-1273
    • Miyoshi, A.1    Kitajima, Y.2    Sumi, K.3    Sato, K.4    Hagiwara, A.5    Koga, Y.6
  • 78
    • 60549104367 scopus 로고    scopus 로고
    • Raf kinase inhibitory protein suppresses a metastasis signalling cascade involving LIN28 and let-7
    • Dangi-Garimella S., Yun J., Eves E.M., Newman M., Erkeland S.J., Hammond S.M., et al. Raf kinase inhibitory protein suppresses a metastasis signalling cascade involving LIN28 and let-7. EMBO J 2009, 28:347-358.
    • (2009) EMBO J , vol.28 , pp. 347-358
    • Dangi-Garimella, S.1    Yun, J.2    Eves, E.M.3    Newman, M.4    Erkeland, S.J.5    Hammond, S.M.6
  • 79
    • 70449529854 scopus 로고    scopus 로고
    • KLF17 is a negative regulator of epithelial-mesenchymal transition and metastasis in breast cancer
    • Gumireddy K., Li A., Gimotty P.A., Klein-Szanto A.J., Showe L.C., Katsaros D., et al. KLF17 is a negative regulator of epithelial-mesenchymal transition and metastasis in breast cancer. Nat Cell Biol 2009, 11:1297-1304.
    • (2009) Nat Cell Biol , vol.11 , pp. 1297-1304
    • Gumireddy, K.1    Li, A.2    Gimotty, P.A.3    Klein-Szanto, A.J.4    Showe, L.C.5    Katsaros, D.6
  • 80
    • 33846684874 scopus 로고    scopus 로고
    • Tubular epithelial cell dedifferentiation is driven by the helix-loop-helix transcriptional inhibitor Id1
    • Li Y., Yang J., Luo J.H., Dedhar S., Liu Y. Tubular epithelial cell dedifferentiation is driven by the helix-loop-helix transcriptional inhibitor Id1. J Am Soc Nephrol 2007, 18:449-460.
    • (2007) J Am Soc Nephrol , vol.18 , pp. 449-460
    • Li, Y.1    Yang, J.2    Luo, J.H.3    Dedhar, S.4    Liu, Y.5
  • 81
    • 0033554723 scopus 로고    scopus 로고
    • Id1 and Id3 are required for neurogenesis, angiogenesis and vascularization of tumour xenografts
    • Lyden D., Young A.Z., Zagzag D., Yan W., Gerald W., O'Reilly R., et al. Id1 and Id3 are required for neurogenesis, angiogenesis and vascularization of tumour xenografts. Nature 1999, 401:670-677.
    • (1999) Nature , vol.401 , pp. 670-677
    • Lyden, D.1    Young, A.Z.2    Zagzag, D.3    Yan, W.4    Gerald, W.5    O'Reilly, R.6
  • 83
    • 44449084242 scopus 로고    scopus 로고
    • Id1 cooperates with oncogenic Ras to induce metastatic mammary carcinoma by subversion of the cellular senescence response
    • Swarbrick A., Roy E., Allen T., Bishop J.M. Id1 cooperates with oncogenic Ras to induce metastatic mammary carcinoma by subversion of the cellular senescence response. Proc Natl Acad Sci U S A 2008, 105:5402-5407.
    • (2008) Proc Natl Acad Sci U S A , vol.105 , pp. 5402-5407
    • Swarbrick, A.1    Roy, E.2    Allen, T.3    Bishop, J.M.4
  • 84
    • 77249126947 scopus 로고    scopus 로고
    • HUNK suppresses metastasis of basal type breast cancers by disrupting the interaction between PP2A and cofilin-1
    • Quintela-Fandino M., Arpaia E., Brenner D., Goh T., Yeung F.A., Blaser H., et al. HUNK suppresses metastasis of basal type breast cancers by disrupting the interaction between PP2A and cofilin-1. Proc Natl Acad Sci U S A 2010, 107:2622-2627.
    • (2010) Proc Natl Acad Sci U S A , vol.107 , pp. 2622-2627
    • Quintela-Fandino, M.1    Arpaia, E.2    Brenner, D.3    Goh, T.4    Yeung, F.A.5    Blaser, H.6
  • 87
    • 0346792725 scopus 로고    scopus 로고
    • TRAIL and apoptosis induction by TNF-family death receptors
    • Wang S., El-Deiry W.S. TRAIL and apoptosis induction by TNF-family death receptors. Oncogene 2003, 22:8628-8633.
    • (2003) Oncogene , vol.22 , pp. 8628-8633
    • Wang, S.1    El-Deiry, W.S.2
  • 90
    • 14844317695 scopus 로고    scopus 로고
    • Unoccupied alpha(v)beta3 integrin regulates osteoclast apoptosis by transmitting a positive death signal
    • Zhao H., Ross F.P., Teitelbaum S.L. Unoccupied alpha(v)beta3 integrin regulates osteoclast apoptosis by transmitting a positive death signal. Mol Endocrinol 2005, 19:771-780.
    • (2005) Mol Endocrinol , vol.19 , pp. 771-780
    • Zhao, H.1    Ross, F.P.2    Teitelbaum, S.L.3
  • 91
    • 0026537225 scopus 로고
    • Identification of membrane antigen C33 recognized by monoclonal antibodies inhibitory to human T-cell leukemia virus type 1 (HTLV-1)-induced syncytium formation: altered glycosylation of C33 antigen in HTLV-1-positive T cells
    • Fukudome K., Furuse M., Imai T., Nishimura M., Takagi S., Hinuma Y., et al. Identification of membrane antigen C33 recognized by monoclonal antibodies inhibitory to human T-cell leukemia virus type 1 (HTLV-1)-induced syncytium formation: altered glycosylation of C33 antigen in HTLV-1-positive T cells. J Virol 1992, 66:1394-1401.
    • (1992) J Virol , vol.66 , pp. 1394-1401
    • Fukudome, K.1    Furuse, M.2    Imai, T.3    Nishimura, M.4    Takagi, S.5    Hinuma, Y.6
  • 92
    • 28244451289 scopus 로고    scopus 로고
    • CD82 metastasis suppressor gene: a potential target for new therapeutics?
    • Tonoli H., Barrett J.C. CD82 metastasis suppressor gene: a potential target for new therapeutics?. Trends Mol Med 2005, 11:563-570.
    • (2005) Trends Mol Med , vol.11 , pp. 563-570
    • Tonoli, H.1    Barrett, J.C.2
  • 93
    • 33746791465 scopus 로고    scopus 로고
    • Interaction of KAI1 on tumor cells with DARC on vascular endothelium leads to metastasis suppression
    • Bandyopadhyay S., Zhan R., Chaudhuri A., Watabe M., Pai S.K., Hirota S., et al. Interaction of KAI1 on tumor cells with DARC on vascular endothelium leads to metastasis suppression. Nat Med 2006, 12:933-938.
    • (2006) Nat Med , vol.12 , pp. 933-938
    • Bandyopadhyay, S.1    Zhan, R.2    Chaudhuri, A.3    Watabe, M.4    Pai, S.K.5    Hirota, S.6
  • 95
    • 0034842795 scopus 로고    scopus 로고
    • The relationship of BRMS1 and RhoGDI2 gene expression to metastatic potential in lineage related human bladder cancer cell lines
    • Seraj M.J., Harding M.A., Gildea J.J., Welch D.R., Theodorescu D. The relationship of BRMS1 and RhoGDI2 gene expression to metastatic potential in lineage related human bladder cancer cell lines. Clin Exp Metastasis 2000, 18:519-525.
    • (2000) Clin Exp Metastasis , vol.18 , pp. 519-525
    • Seraj, M.J.1    Harding, M.A.2    Gildea, J.J.3    Welch, D.R.4    Theodorescu, D.5
  • 96
    • 33646023398 scopus 로고    scopus 로고
    • Suppression of murine mammary carcinoma metastasis by the murine ortholog of breast cancer metastasis suppressor 1 (Brms1)
    • Samant R.S., Debies M.T., Hurst D.R., Moore B.P., Shevde L.A., Welch D.R. Suppression of murine mammary carcinoma metastasis by the murine ortholog of breast cancer metastasis suppressor 1 (Brms1). Cancer Lett 2006, 235:260-265.
    • (2006) Cancer Lett , vol.235 , pp. 260-265
    • Samant, R.S.1    Debies, M.T.2    Hurst, D.R.3    Moore, B.P.4    Shevde, L.A.5    Welch, D.R.6
  • 97
    • 0036006477 scopus 로고    scopus 로고
    • Identification and characterization of the murine ortholog (brms1) of breast-cancer metastasis suppressor 1 (BRMS1)
    • Samant R.S., Debies M.T., Shevde L.A., Verderame M.F., Welch D.R. Identification and characterization of the murine ortholog (brms1) of breast-cancer metastasis suppressor 1 (BRMS1). Int J Cancer 2002, 97:15-20.
    • (2002) Int J Cancer , vol.97 , pp. 15-20
    • Samant, R.S.1    Debies, M.T.2    Shevde, L.A.3    Verderame, M.F.4    Welch, D.R.5
  • 98
    • 40449094341 scopus 로고    scopus 로고
    • BRMS1 suppresses breast cancer experimental metastasis to multiple organs by inhibiting several steps of the metastatic process
    • Phadke P.A., Vaidya K.S., Nash K.T., Hurst D.R., Welch D.R. BRMS1 suppresses breast cancer experimental metastasis to multiple organs by inhibiting several steps of the metastatic process. Am J Pathol 2008, 172:809-817.
    • (2008) Am J Pathol , vol.172 , pp. 809-817
    • Phadke, P.A.1    Vaidya, K.S.2    Nash, K.T.3    Hurst, D.R.4    Welch, D.R.5
  • 99
    • 33646037353 scopus 로고    scopus 로고
    • Suppression of human ovarian carcinoma metastasis by the metastasis-suppressor gene, BRMS1
    • Zhang S., Lin Q.D., Di W. Suppression of human ovarian carcinoma metastasis by the metastasis-suppressor gene, BRMS1. Int J Gynecol Cancer 2006, 16:522-531.
    • (2006) Int J Gynecol Cancer , vol.16 , pp. 522-531
    • Zhang, S.1    Lin, Q.D.2    Di, W.3
  • 100
    • 61349201417 scopus 로고    scopus 로고
    • Breast cancer metastasis suppressor 1 (BRMS1) suppresses metastasis and correlates with improved patient survival in non-small cell lung cancer
    • Smith P.W., Liu Y., Siefert S.A., Moskaluk C.A., Petroni G.R., Jones D.R. Breast cancer metastasis suppressor 1 (BRMS1) suppresses metastasis and correlates with improved patient survival in non-small cell lung cancer. Cancer Lett 2009, 276:196-203.
    • (2009) Cancer Lett , vol.276 , pp. 196-203
    • Smith, P.W.1    Liu, Y.2    Siefert, S.A.3    Moskaluk, C.A.4    Petroni, G.R.5    Jones, D.R.6
  • 101
    • 43149092915 scopus 로고    scopus 로고
    • Alterations of BRMS1-ARID4A interaction modify gene expression but still suppress metastasis in human breast cancer cells
    • Hurst D.R., Xie Y., Vaidya K.S., Mehta A., Moore B.P., Accavitti-Loper M.A., et al. Alterations of BRMS1-ARID4A interaction modify gene expression but still suppress metastasis in human breast cancer cells. J Biol Chem 2008, 283:7438-7444.
    • (2008) J Biol Chem , vol.283 , pp. 7438-7444
    • Hurst, D.R.1    Xie, Y.2    Vaidya, K.S.3    Mehta, A.4    Moore, B.P.5    Accavitti-Loper, M.A.6
  • 102
    • 0347723868 scopus 로고    scopus 로고
    • Breast cancer metastasis suppressor 1 (BRMS1) forms complexes with retinoblastoma-binding protein 1 (RBP1) and the mSin3 histone deacetylase complex and represses transcription
    • Meehan W.J., Samant R.S., Hopper J.E., Carrozza M.J., Shevde L.A., Workman J.L., et al. Breast cancer metastasis suppressor 1 (BRMS1) forms complexes with retinoblastoma-binding protein 1 (RBP1) and the mSin3 histone deacetylase complex and represses transcription. J Biol Chem 2004, 279:1562-1569.
    • (2004) J Biol Chem , vol.279 , pp. 1562-1569
    • Meehan, W.J.1    Samant, R.S.2    Hopper, J.E.3    Carrozza, M.J.4    Shevde, L.A.5    Workman, J.L.6
  • 103
    • 79952067484 scopus 로고    scopus 로고
    • Metastasis suppressor genes: At the interface between the environment and tumor cell growth. International Review of Cell and Molecular Biology in press.
    • Hurst DR, Welch DR. (2010) Metastasis suppressor genes: At the interface between the environment and tumor cell growth. International Review of Cell and Molecular Biology in press.
    • (2010)
    • Hurst, D.R.1    Welch, D.R.2
  • 104
    • 0028029129 scopus 로고
    • Microcell-mediated transfer of chromosome 6 into metastatic human C8161 melanoma cells suppresses metastasis but does not inhibit tumorigenicity
    • Welch D.R., Chen P., Miele M.E., McGary C.T., Bower J.M., Stanbridge E.J., et al. Microcell-mediated transfer of chromosome 6 into metastatic human C8161 melanoma cells suppresses metastasis but does not inhibit tumorigenicity. Oncogene 1994, 9:255-262.
    • (1994) Oncogene , vol.9 , pp. 255-262
    • Welch, D.R.1    Chen, P.2    Miele, M.E.3    McGary, C.T.4    Bower, J.M.5    Stanbridge, E.J.6
  • 105
    • 0030956852 scopus 로고    scopus 로고
    • Suppression of metastasis in human breast carcinoma MDA-MB-435 cells after transfection with the metastasis suppressor gene, KiSS-1
    • Lee J.H., Welch D.R. Suppression of metastasis in human breast carcinoma MDA-MB-435 cells after transfection with the metastasis suppressor gene, KiSS-1. Cancer Res 1997, 57:2384-2387.
    • (1997) Cancer Res , vol.57 , pp. 2384-2387
    • Lee, J.H.1    Welch, D.R.2
  • 106
    • 77950580049 scopus 로고    scopus 로고
    • The KISS1 metastasis suppressor: a good night kiss for disseminated cancer cells
    • Beck B.H., Welch D.R. The KISS1 metastasis suppressor: a good night kiss for disseminated cancer cells. Eur J Cancer 2010, 46:1283-1289.
    • (2010) Eur J Cancer , vol.46 , pp. 1283-1289
    • Beck, B.H.1    Welch, D.R.2
  • 107
    • 0035860778 scopus 로고    scopus 로고
    • The metastasis suppressor gene KiSS-1 encodes kisspeptins, the natural ligands of the orphan G protein-coupled receptor GPR54
    • Kotani M., Detheux M., Vandenbogaerde A., Communi D., Vanderwinden J.M., Le Poul E., et al. The metastasis suppressor gene KiSS-1 encodes kisspeptins, the natural ligands of the orphan G protein-coupled receptor GPR54. J Biol Chem 2001, 276:34631-34636.
    • (2001) J Biol Chem , vol.276 , pp. 34631-34636
    • Kotani, M.1    Detheux, M.2    Vandenbogaerde, A.3    Communi, D.4    Vanderwinden, J.M.5    Le Poul, E.6
  • 108
    • 0035978639 scopus 로고    scopus 로고
    • Metastasis suppressor gene KiSS-1 encodes peptide ligand of a G-protein-coupled receptor
    • Ohtaki T., Shintani Y., Honda S., Matsumoto H., Hori A., Kanehashi K., et al. Metastasis suppressor gene KiSS-1 encodes peptide ligand of a G-protein-coupled receptor. Nature 2001, 411:613-617.
    • (2001) Nature , vol.411 , pp. 613-617
    • Ohtaki, T.1    Shintani, Y.2    Honda, S.3    Matsumoto, H.4    Hori, A.5    Kanehashi, K.6
  • 110
    • 33847784034 scopus 로고    scopus 로고
    • Requirement of KISS1 secretion for multiple organ metastasis suppression and maintenance of tumor dormancy
    • Nash K.T., Phadke P.A., Navenot J.M., Hurst D.R., Accavitti-Loper M.A., Sztul E., et al. Requirement of KISS1 secretion for multiple organ metastasis suppression and maintenance of tumor dormancy. J Natl Cancer Inst 2007, 99:309-321.
    • (2007) J Natl Cancer Inst , vol.99 , pp. 309-321
    • Nash, K.T.1    Phadke, P.A.2    Navenot, J.M.3    Hurst, D.R.4    Accavitti-Loper, M.A.5    Sztul, E.6
  • 111
    • 32944469603 scopus 로고    scopus 로고
    • The KISS1 metastasis suppressor: mechanistic insights and clinical utility
    • Nash K.T., Welch D.R. The KISS1 metastasis suppressor: mechanistic insights and clinical utility. Front Biosci 2006, 11:647-659.
    • (2006) Front Biosci , vol.11 , pp. 647-659
    • Nash, K.T.1    Welch, D.R.2
  • 112
    • 0033229853 scopus 로고    scopus 로고
    • Mitogen-activated protein kinase kinase 4/stress-activated protein/Erk kinase 1 (MKK4/SEK1), a prostate cancer metastasis suppressor gene encoded by human chromosome 17
    • Yoshida B.A., Dubauskas Z., Chekmareva M.A., Christiano T.R., Stadler W.M., Rinker-Schaeffer C.W. Mitogen-activated protein kinase kinase 4/stress-activated protein/Erk kinase 1 (MKK4/SEK1), a prostate cancer metastasis suppressor gene encoded by human chromosome 17. Cancer Res 1999, 59:5483-5487.
    • (1999) Cancer Res , vol.59 , pp. 5483-5487
    • Yoshida, B.A.1    Dubauskas, Z.2    Chekmareva, M.A.3    Christiano, T.R.4    Stadler, W.M.5    Rinker-Schaeffer, C.W.6
  • 113
    • 53949093245 scopus 로고    scopus 로고
    • New paradigms for the function of JNKK1/MKK4 in controlling growth of disseminated cancer cells
    • Taylor J.L., Szmulewitz R.Z., Lotan T., Hickson J., Griend D.V., Yamada S.D., et al. New paradigms for the function of JNKK1/MKK4 in controlling growth of disseminated cancer cells. Cancer Lett 2008, 272:12-22.
    • (2008) Cancer Lett , vol.272 , pp. 12-22
    • Taylor, J.L.1    Szmulewitz, R.Z.2    Lotan, T.3    Hickson, J.4    Griend, D.V.5    Yamada, S.D.6
  • 114
    • 28244493042 scopus 로고    scopus 로고
    • Suppression of metastatic colonization by the context-dependent activation of the c-Jun NH2-terminal kinase kinases JNKK1/MKK4 and MKK7
    • Vander Griend D.J., Kocherginsky M., Hickson J.A., Stadler W.M., Lin A., Rinker-Schaeffer C.W. Suppression of metastatic colonization by the context-dependent activation of the c-Jun NH2-terminal kinase kinases JNKK1/MKK4 and MKK7. Cancer Res 2005, 65:10984-10991.
    • (2005) Cancer Res , vol.65 , pp. 10984-10991
    • Vander Griend, D.J.1    Kocherginsky, M.2    Hickson, J.A.3    Stadler, W.M.4    Lin, A.5    Rinker-Schaeffer, C.W.6
  • 116
    • 42049120920 scopus 로고    scopus 로고
    • C-Jun NH2-terminal kinase activating kinase 1/mitogen-activated protein kinase kinase 4-mediated inhibition of SKOV3ip.1 ovarian cancer metastasis involves growth arrest and p21 up-regulation
    • Lotan T., Hickson J., Souris J., Huo D., Taylor J., Li T., et al. c-Jun NH2-terminal kinase activating kinase 1/mitogen-activated protein kinase kinase 4-mediated inhibition of SKOV3ip.1 ovarian cancer metastasis involves growth arrest and p21 up-regulation. Cancer Res 2008, 68:2166-2175.
    • (2008) Cancer Res , vol.68 , pp. 2166-2175
    • Lotan, T.1    Hickson, J.2    Souris, J.3    Huo, D.4    Taylor, J.5    Li, T.6


* 이 정보는 Elsevier사의 SCOPUS DB에서 KISTI가 분석하여 추출한 것입니다.